Ebook Handbook of Lipid Membranes Molecular Functional and Materials Aspects 1St Edition Cyrus R Safinya Editor Online PDF All Chapter

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 70

Handbook of Lipid Membranes:

Molecular, Functional, and Materials


Aspects 1st Edition Cyrus R. Safinya
(Editor)
Visit to download the full and correct content document:
https://ebookmeta.com/product/handbook-of-lipid-membranes-molecular-functional-a
nd-materials-aspects-1st-edition-cyrus-r-safinya-editor/
More products digital (pdf, epub, mobi) instant
download maybe you interests ...

Cambridge IGCSE and O Level History Workbook 2C - Depth


Study: the United States, 1919-41 2nd Edition Benjamin
Harrison

https://ebookmeta.com/product/cambridge-igcse-and-o-level-
history-workbook-2c-depth-study-the-united-states-1919-41-2nd-
edition-benjamin-harrison/

Handbook of Materials Science Volume 1 Optical


Materials 1st Edition R. S. Ningthoujam

https://ebookmeta.com/product/handbook-of-materials-science-
volume-1-optical-materials-1st-edition-r-s-ningthoujam/

Applications of Porphyrinoids as Functional Materials


1st Edition Heinrich Lang

https://ebookmeta.com/product/applications-of-porphyrinoids-as-
functional-materials-1st-edition-heinrich-lang/

Handbook of Magnetism and Magnetic Materials

https://ebookmeta.com/product/handbook-of-magnetism-and-magnetic-
materials/
Cellular and Molecular Aspects of Myeloproliferative
Neoplasms - Part A (Volume 365) (International Review
of Cell and Molecular Biology, Volume 365) 1st Edition
Niccolo Bartalucci (Editor)
https://ebookmeta.com/product/cellular-and-molecular-aspects-of-
myeloproliferative-neoplasms-part-a-volume-365-international-
review-of-cell-and-molecular-biology-volume-365-1st-edition-
niccolo-bartalucci-editor/

Biochemical Physiological and Molecular Aspects of


Human Nutrition 4th ed 4th Edition Martha H. Stipanuk

https://ebookmeta.com/product/biochemical-physiological-and-
molecular-aspects-of-human-nutrition-4th-ed-4th-edition-martha-h-
stipanuk/

Atomic and Molecular Spectroscopy Basic Aspects and


Practical Applications Fifth Edition Sune Svanberg

https://ebookmeta.com/product/atomic-and-molecular-spectroscopy-
basic-aspects-and-practical-applications-fifth-edition-sune-
svanberg/

Primary Mathematics 3A Hoerst

https://ebookmeta.com/product/primary-mathematics-3a-hoerst/

Functional Materials for the Oil and Gas Industry 1st


Edition Deepak Dwivedi

https://ebookmeta.com/product/functional-materials-for-the-oil-
and-gas-industry-1st-edition-deepak-dwivedi/
Handbook of Lipid Membranes
Handbook of Lipid Membranes
Molecular, Functional, and Materials Aspects

Edited by
Cyrus R. Safinya
Joachim O. Rädler
First Edition published 2021
by CRC Press
6000 Broken Sound Parkway NW, Suite 300, Boca Raton, FL 33487-2742

and by CRC Press


2 Park Square, Milton Park, Abingdon, Oxon, OX14 4RN

© 2021 Taylor & Francis Group, LLC

CRC Press is an imprint of Taylor & Francis Group, LLC

Reasonable efforts have been made to publish reliable data and information, but the author and publisher cannot assume responsibility for the
validity of all materials or the consequences of their use. The authors and publishers have attempted to trace the copyright holders of all material
reproduced in this publication and apologize to copyright holders if permission to publish in this form has not been obtained. If any copyright
material has not been acknowledged, please write and let us know so we may rectify in any future reprint.

Except as permitted under U.S. Copyright Law, no part of this book may be reprinted, reproduced, transmitted, or utilized in any form by any
electronic, mechanical, or other means, now known or hereafter invented, including photocopying, microfilming, and recording, or in any infor-
mation storage or retrieval system, without written permission from the publishers.

For permission to photocopy or use material electronically from this work, access www.copyright.com or contact the Copyright Clearance
Center, Inc. (CCC), 222 Rosewood Drive, Danvers, MA 01923, 978-750-8400. For works that are not available on CCC, please contact mpkbook-
spermissions@tandf.co.uk

Trademark notice: Product or corporate names may be trademarks or registered trademarks and are used only for identification and explanation
without intent to infringe.

ISBN: 978-1-466-55572-3 (hbk)


ISBN: 978-1-032-01441-8 (pbk)
ISBN: 978-0-429-19407-8 (ebk)

Typeset in Times
by codeMantra
Contents
Preface���������������������������������������������������������������������������������������������������������������������������������������������������������������������������������vii
Editors�����������������������������������������������������������������������������������������������������������������������������������������������������������������������������������xi
Contributors����������������������������������������������������������������������������������������������������������������������������������������������������������������������� xiii

Chapter 1 A Short History of Membrane Physics..............................................................................................................1


Erich Sackmann and Avinoam Ben-Shaul

Chapter 2 Structures and Interactions in Freely Suspended Multilayer Membranes and Dilute Lamellar Fluid
Membranes from Synchrotron X-Ray Scattering........................................................................................... 33
Gregory S. Smith and Cyrus R. Safinya

Chapter 3 Structures of Lipid Membranes: Cubic and Inverse Hexagonal Phases......................................................... 49


Charlotte E. Conn and John M. Seddon

Chapter 4 Structure of Lipid Membranes by Advanced X-Ray Scattering and Imaging................................................ 65


Tim Salditt

Chapter 5 Adhesion Protein Architecture and Intermembrane Potentials: Force Measurements


and Biological Significance............................................................................................................................. 83
Deborah E. Leckband

Chapter 6 Charged Membranes: Poisson–Boltzmann Theory, the DLVO Paradigm, and Beyond.................................99
Tomer Markovich, David Andelman, and Rudolf Podgornik

Chapter 7 Membrane Shape Evolution In Vitro............................................................................................................. 129


Alexandra Zidovska

Chapter 8 Mechanisms of Membrane Curvature Generation by Peptides and Proteins: A Unified


Perspective on Antimicrobial Peptides......................................................................................................... 141
Michelle W. Lee, Nathan W. Schmidt, and Gerard C. L. Wong

Chapter 9 Lipid Membrane Shape Evolution and the Actin Cytoskeleton.................................................................... 161
David R. Slochower, Yu-Hsiu Wang, Ravi Radhakrishnan, and Paul A. Janmey

Chapter 10 Effects of Osmotic Stress on Topologically Closed Membrane Compartments........................................... 177


James C. S. Ho, Bo Liedberg, and Atul N. Parikh

Chapter 11 Cationic Liposomes as Spatial Organizers of Nucleic Acids in One, Two, and Three Dimensions:
Liquid Crystal Phases with Applications in Delivery and Bionanotechnology............................................ 195
Cyrus R. Safinya, Kai K. Ewert, Youli Li, and Joachim O. Rädler

Chapter 12 Lipids in DNA, RNA, and Peptide Delivery for In Vivo Therapeutic Applications..................................... 211
Tyler Goodwin and Leaf Huang

v
vi Contents

Chapter 13 Electrostatics of Lipid Membranes Interacting with Oppositely Charged Macromolecules........................ 223
Guilherme Volpe Bossa, Klemen Bohinc, and Sylvio May

Chapter 14 Lipid-Based Bioanalytical Sensors............................................................................................................... 241


Marta Bally, Hudson Pace, and Fredrik Höök

Chapter 15 Lipids in Dermal Applications: Cosmetics and Pharmaceutics.................................................................... 271


Jérôme Bibette and Abdou Rachid Thiam

Chapter 16 Supported Lipid Bilayers............................................................................................................................... 293


Theo Lohmüller, Bert Nickel, and Joachim O. Rädler

Chapter 17 Artificial Membranes Composed of Synthetic Copolypeptides.................................................................... 305


Timothy J. Deming

Chapter 18 Synthetic Membranes from Block Copolymers, Recombinant Proteins, and Dendrimers.......................... 323
Daniel A. Hammer, Zhichun Wang, Ellen Reed, Chen Gao, and Kevin B. Vargo

Chapter 19 Amphiphilic Self-Assembly and the Origin of Life in Hydrothermal Conditions....................................... 337
Christos D. Georgiou and David W. Deamer

Index���������������������������������������������������������������������������������������������������������������������������������������������������������������������������������� 349
Preface
Lipid membrane science has evolved from its roots in bio- we now see a flourishing of lipid-based science opening
chemistry and biophysics to become an essential component new approaches to vaccines with the Moderna and Pfizer/
in the emerging interdisciplinary fields of nanobioscience BioNTech lipid-mRNA nanoparticles saving millions of
and nanobiotechnology, with new everyday applications lives in the worldwide pandemic in 2020 and 2021 due to
appearing at an ever-increasing rate. This shift calls for a COVID-19.
new type of introductory-level reading material spanning This handbook brings together contributions from world
multiple disciplines and appropriate for both beginners and experts in lipid and artificial membrane science and appli-
advanced researchers. The Handbook of Lipid Membranes: cations. In the first chapter, Erich Sackmann and Avinoam
Molecular, Functional, and Materials Aspects offers Ben-Shaul provide a historical survey of membrane physics
a multifaceted perspective, leading the reader through from the perspective of two-dimensional self-assembling
membrane-related processes in reconstituted and living
­ systems consisting of interacting lipids and membrane-
matter systems. It teaches how to learn from nature to build associated proteins. The development of certain key novel
artificial and functional membrane-based systems, includ- experimental techniques and theoretical approaches over
ing those not based on lipids. The emphasis is on the sci- the last few decades, which have contributed to our cur-
ence and technology of lipids and artificial membranes rent understanding of membrane physics, is discussed. The
and includes chapters on advanced X-ray and force mea- significance of lipid mobility, membrane defects and mem-
surement techniques. The 19 chapters are simultaneously brane curvature elasticity, to membrane thermomechanical
self-contained yet connected with common themes run- properties, and the impact on biological membrane struc-
ning between the chapters. Included are chapters covering ture and function are described.
the role of lipids as structural components in determining The following two chapters start out by introducing
distinct membrane shapes, topologies and inter-membrane lipid membrane structure and phase behavior. Gregory
interactions. By making a direct connection to cellular sys- Smith and Cyrus Safinya introduce the phase diagram of
tems, membrane curvature generation, mediated by peptide gel-like ordered membranes, consisting of lipids with phos-
and protein binding, and biological signaling lipids in con- phocholine headgroups, in Chapter 2. X-ray scattering
cert with cytoskeletal proteins are presented. Beyond the and reflectometry techniques are described that determine
science in lipid biology and biophysics, the chapters reveal the intra- and intermolecular lipid arrangement and probe
how mastering lipid interactions enables novel biomedical interlayer interactions by analysis of thermal diffuse scat-
and nanotechnological applications with fine-tuned func- tering. The chapter outlines how structural studies of highly
tionality. Subject-based chapters discuss current applica- oriented freely suspended multilayer preparations have
tions in biomedicine, cosmetics and nanotechnology. This revealed that the gel phase is comprised of three phases
includes lipid vectors in nucleic acid and drug delivery, in distinguished by the direction of chain tilt within the two-
dermal applications, and in artificial biointerfaces and lipid- dimensional ordered membrane. Stacks of fluid membranes
based sensors. A final engaging chapter takes the reader to are analyzed within the context of a Landau–De Gennes
the beginning of where it all started and explores the role of type elastic energy displaying characteristics of a low-
lipids in the origin of life. dimensional Landau–Peierls system. Synchrotron X-ray
The discovery of liposomes by A. D. Bangham and R. scattering shows that such very dilute fluid lamellar phases
W. Horne in the 1960s with efficient encapsulation prop- are stabilized by entropically mediated undulation forces
erties and striking similarities in electron micrographs to elucidated by Wolfgang Helfrich.
cell plasma membranes led to their hypothesis that lipids Chapter 3 by Charlotte Conn and John Seddon continues
are the primary structural components responsible for the with a comprehensive description of the structure and prop-
permeability barriers of biological membranes. This major erties of cubic and inverse hexagonal phases. The authors
discovery led the way for a rapid worldwide increase in describe how the stability of these non-lamellar phases is
research on the biophysical properties of lipid vesicles and derived from forces underlying curvature and topological
on the biological function of membrane-associated proteins, transformations in lipid membranes. Further, the effect of
reconstituted in liposomes. The realization that liposomes hydrostatic pressure on phase behavior is discussed and
contain sites for incorporation of hydrophobic, hydrophilic shown to be relevant in marine biology. Numerous illumi-
and amphiphilic molecules, led researchers early on to nating examples are presented that show the occurrence of
investigate, on the one hand, their promise as components non-lamellar phases in living matter and in a wide range
of personal care products and, on the other hand, as car- of basic science studies and bionanotechnological applica-
riers of drugs and genes for therapeutic applications. The tions. This includes cubic and hexagonal phase structures
latter includes over 100 lipid-based currently-ongoing clini- enabling specific functions, in vivo, and, as structured drug
cal trials worldwide targeting a single gene, such as cystic delivery vectors, cubic phases to enable membrane protein
fibrosis, and multigene cancer diseases. Most remarkably, structure determination.

vii
viii Preface

In Chapter 4, Tim Salditt gives a summary of state-of- Michelle Lee, Nathan Schmidt and Gerard Wong take
the-art X-ray analysis of highly oriented lipid membranes the concept of membrane curvature further in Chapter 8
employing X-ray optics, nanoscale focusing, time-resolved and discuss how peptides modulate membrane morphol-
X-ray diffraction, and lensless coherent imaging. ogy. Membrane binding peptides play a vital role as anti-
Illustrations of these powerful new synchrotron X-ray tech- microbial agents, enabling budding and release of viruses
niques are shown as applied to a wide range of systems of and enabling membrane tubulation and fission. The chapter
high scientific interest, including nonequilibrium dynam- provides a survey of membrane curvature generation mech-
ics of membranes, short-lived intermediate structures of anisms and illustrates the case of antimicrobial peptides.
membrane fusion, and the asymmetric structures of synap- The authors explain how certain cationic and hydrophobic
tic vesicles and myelin multilayers. The chapter concludes structural motifs of antimicrobial peptides lead to selec-
by presenting coherent X-ray imaging as a powerful new tive pore-forming activity against bacterial membranes.
method for membrane structure analysis. Thus, as the authors explain, intelligently designed altera-
Deborah Leckband introduces the reader to the surface tions in amino-acid sequence, intended to enhance peptide
forces apparatus (SFA) in Chapter 5, a unique technique membrane-permeating ability, may be implemented in the
that enables direct measurement of forces between surface- development of novel antibiotics to fight persistent bacterial
bound membranes containing membrane-associated pro- strains.
teins, including intrinsically disordered proteins. The SFA In Chapter 9, David Slochower, Yu-Hsiu Wang, Ravi
technique, with biological material contained between sur- Radhakrishnan and Paul Janmey describe how the shape of
faces, naturally mimics the confined environment in vivo. eukaryotic cell membranes is controlled by both the plasma
Thus, measured protein-mediated forces, both attractive membrane lipid bilayer and the underlying cytoskeletal net-
and repulsive, are expected to elucidate membrane–protein work. At a molecular level, the role of phosphoinositides in
function at biological interfaces. Forces mediated by adhe- regulating the membrane–cytoskeletal interface, by bind-
sion proteins, including lectins that bind carbohydrates, ing cytoskeletal proteins and proteins that cause or sense
neural cell adhesion molecules, and cadherin adhesion membrane curvature, is described. In parallel, mechanisms
proteins, ubiquitous in most tissues, are highlighted. The of producing membrane curvature driven by the cytoskel-
chapter further describes cell-binding kinetics employing eton are introduced. Combining this information within
micropipette measurement techniques. the context of recent experimental results and multiscale
Chapter 6 by Tomer Markovich, David Andelman and simulations, the authors present an integrated view of how
Rudolf Podgornik is an entirely self-contained theoretical these fascinating lipids perform their many cellular func-
chapter covering our current understanding of electrostatic tions and help orchestrate the dynamic changes in mem-
forces in charged membrane systems. Poisson–Boltzmann brane curvature.
(PB) theory of charged membranes, in the presence of no The effect of osmotic stress on cell membrane topology
added salt and added salt, is described. Ion profiles near is the focus of Chapter 10 by James Ho, Bo Liedberg and
single and between two membranes are considered. As Atul Parikh. Starting with giant unilamellar vesicles, as
becomes evident, the electrostatic forces described in the model membrane compartments, the permeation of water
chapter are relevant to many of the biological systems and the generation of osmotic gradients by hindered perme-
described in the handbook. The chapter further reviews ation of solutes is introduced. The chapter gives experimen-
van der Waals forces and briefly discusses the limitations tal and theoretical insights into shape changes of flaccid
of PB theory, including the limit of high surface charge and vesicles in hypertonic media and transient pore formation
multivalent counterions, where the PB theory breaks down in response to hypotonic media. Shape deformations due to
due to its mean-field nature. The material is presented in osmotic stress coupling with phase separation of multicom-
a manner suitable for instructors teaching at the first-year ponent vesicles are also discussed. Finally, the potential rel-
graduate or upper-division undergraduate levels. evance of stress relaxation mechanisms is discussed within
The key physical concepts underlying distinct vesicle the context of ubiquitous osmotic challenges that primitive
shapes are lipid molecule shape, membrane composition, protocells most likely experienced in early life.
and intermolecular interactions. In Chapter 7, Alexandra Lipid-nucleic acid complexes are self-assembled systems
Zidovska takes the reader on a broad review of membrane that incorporate key concepts of lipid molecule shape and
shape evolution in vesicles (i.e., liposomes) as discovered in membrane curvature discussed in previous chapters. Their
laboratories worldwide over the last few decades. The chap- scientific perspective and applications in vitro and in vivo
ter includes sections connecting modern elasticity models are described in three consecutive chapters. Chapter 11, by
to shape evolution observed experimentally. This includes Cyrus Safinya, Kai Ewert, Youli Li and Joachim Rädler,
the hugely successful Helfrich curvature elastic model and introduces the self-assembled structures of cationic lipo-
beyond Helfrich theory with models that incorporate the some (CL)-nucleic acid complexes used in gene delivery.
intrinsic area differences between the outer and inner mem- The authors explain how concepts of membrane curvature
brane leaflets due to the finite curvature of liposomes. The and electrostatic interactions explain the formation of liq-
chapter concludes by connecting in vitro to in vivo observa- uid crystalline inverse hexagonal, lamellar and hexagonal
tions where many similar shapes have been observed. phases with DNA residing in one, two, or three dimensions,
Preface ix

respectively. The chapter describes how transfection effi- Chapter 15, by Jérôme Bibette and Abdou Rachid Thiam,
ciency, measuring the expression of DNA transferred by CL consists of a comprehensive overview of the structure and
complexes into cells, depends on the underlying structures function of the skin and novel dermal applications in cos-
of the complexes. The concepts presented in the chapter metics and pharmaceutics. The role of lipids in dermal
are expected to apply to ongoing efforts to understand the health and the downside ­consequences of lipid disorders on
physics of self-assembly in cationic ionizable lipid-mRNA skin function are reviewed. The chapter contains a wealth
formulations used in cancer therapeutics and vaccine appli- of information on the molecular components of cosmetic
cations (e.g., Moderna and Pfizer/BioNTech nanoparticle and pharmaceutical products. The authors describe the state
vaccines). of the art in cosmetic and therapeutic dermal applications,
In Chapter 12, Tyler Goodwin and Leaf Huang cover through rational design of lipid and surfactant-based vec-
lipids as delivery vehicles of DNA, RNA and peptides in tors, optimized for delivery through the skin. Current lipid
in vivo therapeutic applications. The authors describe how nanovectors used worldwide are described. This includes
lipid vectors may be designed to be highly efficient in the liposome formulations, nano- and micro-emulsions, and
delivery of biomacromolecules. Based on rational design solid lipid nanoparticles.
over many years, lipid vectors were developed to overcome Chapter 16 by Theo Lohmüller, Bert Nickel and Joachim
extracellular and intracellular barriers. The chapter reviews Rädler is dedicated to the properties and applications of
the improvements and the remaining challenges of current so-called supported lipid bilayer systems, which represent
lipid vectors in in vivo applications. The survey highlights planar lipid bilayers that form at the solid–fluid interface
promising ionizable lipids and composite nanocore-lipid through vesicle fusion and in-plane spreading. In these
vectors and discusses the progress made in clinical trials. systems, lipids and incorporated proteins retain high lat-
Chapter 13 turns to theory, where electrostatic mod- eral mobility. Hence, charged macromolecules that bind
els underlying self-assembly of charged membranes and to oppositely charged planar-supported bilayer systems
oppositely charged biomacromolecules, including DNA, exhibit ideal two-dimensional properties and can be manip-
proteins and peptides, are reviewed by Guilherme Volpe ulated in-plane through external electric fields. In polymer-
Bossa, Klemen Bohinc and Sylvio May. The influence of supported lipid bilayer systems, the membrane is elevated
lipid mobility, protein-mediated lipid phase separation and from the solid, providing room for larger transmembrane
charge regulation on the interactions stabilizing proteins proteins. The chapter concludes with examples of the use of
adsorbed on membranes is discussed. Theoretical models supported lipid bilayer systems as novel platforms for pre-
of the phase behavior of lipid-DNA complexes are reviewed senting molecules to living cells.
for cationic and zwitterionic lipids. Discussions on impor- In the last two decades, increasing research efforts have
tant local interactions between neutral zwitterionic lipid been directed toward developing artificial membranes that
headgroups and divalent cations and DNA and membranes are not based on lipids.
containing zwitterionic lipids are presented. The chapter Chapter 17 by Timothy Deming summarizes advances
concludes with a general discussion of the stability of mem- in the synthesis of well-defined block and statistical copoly-
brane pores resulting from the interactions between amphi- peptides that can be assembled into stable membrane struc-
philic peptides and membranes. tures. Polymerization chemistries are described that allow
Lipid membranes are powerful in functionalizing or precision copolypeptide synthesis with control over chain
protecting surfaces. Marta Bally, Hudson Pace and Fredrik length, chain length distribution, and chain-end functional-
Höök give an account of the use of planar lipid mem- ity. The author explains how well-defined copolypeptides
branes as bioanalytical sensors in Chapter 14. The chap- of controlled dimensions, including molecular weight,
ter focuses on a broad range of surface-based biosensing sequence, and composition, can be prepared, which sponta-
techniques. Immobilization of lipid assemblies at a sensor neously assemble into vesicles with polypeptide membranes.
surface is achieved by a supported lipid bilayer, tethered Examples of polypeptide membrane vesicles possessing
bilayers, or a free-spanning bilayer of surface-tethered unique properties (due to the amino acid building blocks
vesicles. Moreover, patterned planar membranes are used and ordered conformations of the polypeptide segments)
for high-throughput screening and lab-on-a-chip devices. are presented in the context of biomedical application.
The authors show how membrane-associated proteins may In Chapter 18, Daniel Hammer, Zhichun Wang, Ellen
refine biosensing to detect interactions between membrane Reed, Chen Gao and Kevin Vargo give a comprehensive
proteins and ligands. Discussions are also presented about overview of membranes constructed with synthetic amphi-
how measurements of membrane–membrane interactions philic polymers and biopolymers with novel architectures.
in biosensors have been enhanced by the invention of sin- The focus of the chapter is on the science and technologi-
gle-vesicle assays that determine equilibrium-binding con- cal applications of artificial vesicles made from spontane-
stants from statistics of residence times. An outlook on the ous assembly of block copolymers (polymersomes) and
use of lipid-based nanoreactors and force-driven manipula- copolypeptides, recombinant proteins and amphiphilic
tion of membrane components in supported lipid bilayers dendrimers. The methods for producing self-assembling
for chip-based bioanalytics is presented. amphipathic proteins employing modern recombinant
x Preface

technology are described. The chapter concludes with the formation of life around four billion years ago. Discussions
latest developments in the construction of artificial vesi- are presented about the physical and chemical properties of
cles from glycan-based Janus dendrimers. The novel arti- lipids, which would have been essential in the spontaneous
ficial vesicles described in this chapter promise to open assembly and formation of very simple early membranes
new opportunities for the intelligent design of functional with their required barrier properties in comparison with
nanoparticles with a broad range of applications, including, the plasma membrane of current living organisms contain-
in molecular delivery and sensing. ing a range of associated proteins designed for a diverse
The final Chapter 19, by Christos Georgiou and David set of functions. The authors conclude with a thought-pro-
Deamer, gives a fascinating presentation of some central voking discussion of the positive and negative aspects of
aspects of the origin of life and, in particular, emphasizes hydrothermal fields versus hydrothermal vents as locations
the role played by single-tailed amphiphilic fatty acids and for the onset of the first cellular life on earth.
double-tailed lipids. The authors consider alkaline hydro-
thermal vents and hydrothermal fields as possible geologi- Cyrus R. Safinya
cal sites for the prebiotic environment for the spontaneous Joachim O. Rädler
Editors
Cyrus R. Safinya is a Distinguished Professor at the Joachim O. Rädler is a Professor of Experimental
University of California, Santa Barbara (UCSB). His primary Physics at Ludwig-Maximilians-University (LMU), and
appointment is in the Materials Department in the College of holds the Chair for Soft Condensed Matter. He studied
Engineering and he has joint appointments in the Molecular, Physics at the Friedrich Wilhelms University in Bonn,
Cellular, and Developmental Biology Department, and, by at Cambridge University (UK) and at the Technical
courtesy, in the Physics Department and the Biomolecular University of Munich. In 1993, he received his doctoral
Science and Engineering Program. He received a B.S. in degree in Biophysics for his work on vesicle adhesion
Physics and Mathematics from Bates College in 1975 and a under the guidance of Erich Sackmann. As a post-doc-
PhD in Physics from the Massachusetts Institute of Technology toral fellow during 1993–1996, he studied cationic lipid-
in 1981 for his studies on liquid crystal phase transitions under DNA complexes with Cyrus Safinya at UC Santa Barbara.
the guidance of Robert J. Birgeneau (currently Chancellor Rädler received his habilitation in Experimental Physics
Emeritus and Distinguished Professor of Physics and at Technical University of Munich, where he worked
Materials Science at the University of California, Berkeley). on supported membranes. In 2000, he was appointed
Safinya joined the Exxon Research & Engineering Company as a senior group leader at the Max Planck Institute
immediately after obtaining his PhD and started his research for Polymer Research. He was named full professor at
studies on the structure of complex fluids and biological mem- LMU in 2001 and became a member of the Center for
branes before moving to UCSB in 1992. He was a Rothschild NanoScience in the same year. From 2008 to 2011, he
Fellow and a Visiting Directeur de Recherche at the Curie held a temporary consulting and teaching position for
Institute in 1994 and, between 2009 and 2013, a Distinguished experimental NanoBio physics at the University College
Visiting Professor at the Korean Advanced Institute of Science Dublin. He served as spokesperson of the Center for
& Technology. Safinya is the author or co-author of more than NanoScience at LMU, of the collaborative research cen-
220 publications, many of which have appeared in journals ter “Nanoagents” and of various graduate programs.
on research topics in biophysics, chemical physics, physical He is the author or co-author of over 180 publications
chemistry, bioengineering, and biomedical sciences. His cur- on research topics in soft matter and biophysics, physi-
rent research aims to elucidate structures and interactions in cal chemistry, chemical physics, bioengineering, and
soft and biological matter systems, including, in lipid-nucleic bionanotechnology. His current research focuses on the
acid mixtures and protein assemblies derived from neurons. self-assembly of siRNA and mRNA lipid nanoparticles,
In parallel, his group works on the development of novel lipid the interaction of nanomaterials with living cells, time-
vectors as carriers of nucleic acid (DNA and RNA) and hydro- resolved studies of single-cell gene expression, and the
phobic drugs in gene and cancer therapeutics. physics of cell migration.

xi
Contributors
David Andelman Timothy J. Deming
School of Physics and Astronomy Department of Bioengineering and Department of
Tel Aviv University Chemistry and Biochemistry
Tel Aviv, Israel University of California Los Angeles
Los Angeles, California, USA
Marta Bally
Department of Physics Chalmers University of Technology Kai K. Ewert
Gothenburg, Sweden Materials Department and Materials Research Laboratory
Department of Clinical Microbiology & Wallenberg University of California Santa Barbara
Centre for Molecular Medicine Santa Barbara, California, USA
Umeå University
Umeå, Sweden Chen Gao
Department of Chemical and Biomolecular Engineering
University of Pennsylvania
Avinoam Ben-Shaul Philadelphia, Pennsylvania, USA
Institute of Chemistry and Fritz Haber Research Center
Hebrew University of Jerusalem Christos D. Georgiou
Jerusalem, Israel Department of Biology
University of Patras
Jérôme Bibette Patras, Greece
Laboratoire de Colloïdes et Matériaux Divisés
École Supérieure de Physique et de Chimie Industrielles Tyler Goodwin
de la Ville de Paris Division of Molecular Pharmaceutics, Eshelman School of
Université Paris Sciences et Lettres (PSL) Pharmacy
Paris, France Department of Biomedical Engineering
University of North Carolina
Guilherme Volpe Bossa Chapel Hill, North Carolina, USA
Department of Physics
North Dakota State University Daniel A. Hammer
Fargo, North Dakota, USA Department of Bioengineering
Department of Chemical and Biomolecular Engineering
Klemen Bohinc University of Pennsylvania
Faculty of Health Sciences Philadelphia, Pennsylvania, USA
University of Ljubljana
Ljubljana, Slovenia James C. S. Ho
Centre for Biomimetic Sensor Science
Charlotte E. Conn School of Materials Science & Engineering
School of Science, College of Science Engineering Nanyang Technological University
and Health Singapore
RMIT University
Melbourne, Australia Fredrik Höök
Department of Physics
David W. Deamer Chalmers University of Technology
Department of Biomolecular Engineering Göteborg, Sweden
Baskin School of Engineering
University of California Santa Cruz
Santa Cruz, California, USA

xiii
xiv Contributors

Leaf Huang Hudson Pace


Division of Molecular Pharmaceutics and Department of Integrative Medical Biology
Center for Nanotechnology Umeå University
Eshelman School of Pharmacy Umeå, Sweden
Department of Biomedical Engineering
University of North Carolina Atul N. Parikh
Chapel Hill, North Carolina, USA Department of Biomedical Engineering
Department of Chemical Engineering and
Paul A. Janmey Materials Science
Departments of Physiology and Physics & Astronomy University of California
University of Pennsylvania Davis, California, USA
Philadelphia, Pennsylvania, USA and
Centre for Biomimetic Sensor Science, School of
Michelle W. Lee Materials Science & Engineering
Department of Bioengineering Nanyang Technological University
University of California Los Angeles Singapore
Los Angeles, California, USA
Rudolf Podgornik
Deborah E. Leckband School of Physical Sciences and Kavli Institute for
Department of Chemistry Theoretical Sciences
Department of Chemical and Biomolecular Engineering University of Chinese Academy of Sciences
University of Illinois at Urbana-Champaign Beijing, China
Urbana, Illinois, USA and
Department of Theoretical Physics, J. Stefan Institute and
Youli Li Department of Physics
Materials Research Laboratory Faculty of Mathematics and Physics
University of California Santa Barbara University of Ljubljana
Santa Barbara, California, USA Ljubljana, Slovenia
Bo Liedberg
Centre for Biomimetic Sensor Science, School of Ravi Radhakrishnan
Materials Science & Engineering Department of Bioengineering
Nanyang Technological University University of Pennsylvania
Singapore Philadelphia, Pennsylvania, USA

Theo Lohmüller Ellen Reed


Photonics and Optoelectronics Group, Nano-Institute Department of Chemical and Biomolecular Engineering
Munich University of Pennsylvania
Faculty of Physics, Ludwig-Maximilians-Universität Philadelphia, Pennsylvania, USA
Munich, Germany
Erich Sackmann
Tomer Markovich
Physik-Department
School of Physics and Astronomy
Technical University Munich
Tel Aviv University
Garching, Germany
Tel Aviv, Israel

Sylvio May Tim Salditt


Department of Physics Institut für Röntgenphysik
North Dakota State University Universität Göttingen
Fargo, North Dakota, USA Göttingen, Germany

Bert Nickel Nathan W. Schmidt


Faculty of Physics, Soft Condensed Matter Group Department of Bioengineering
Ludwig-Maximilians-Universität University of California Los Angeles
Munich, Germany Los Angeles, California, USA
and
Ginkgo Bioworks
Boston, Massachusetts, USA
Contributors xv

John M. Seddon Yu-Hsiu Wang


Department of Chemistry Department of Biochemistry and Molecular Biology
Imperial College London The University of Texas Medical Branch
London, UK Galveston, Texas, USA

David R. Slochower Zhichun Wang


Skaggs School of Pharmacy and Pharmaceutical Sciences Department of Bioengineering
University of California San Diego University of Pennsylvania
La Jolla, California, USA Philadelphia, Pennsylvania, USA

Gregory S. Smith (Retired) Gerard C. L. Wong


Neutron Scattering Division Neutron Sciences Directorate Department of Bioengineering
Oak Ridge National Laboratory Department of Chemistry & Biochemistry
Oak Ridge, Tennessee, USA California NanoSystems Institute
University of California Los Angeles
Abdou Rachid Thiam Los Angeles, California, USA
Laboratoire de Physique de l’École Normale
Supérieure, ENS Alexandra Zidovska
Université PSL, CNRS Center for Soft Matter Research, Department of Physics
Sorbonne Université, Université de Paris New York University
Paris, France New York, New York, USA

Kevin B. Vargo
Department of Chemical and Biomolecular Engineering
University of Pennsylvania
Philadelphia, Pennsylvania, USA
1 A Short History of Membrane Physics
Erich Sackmann
Technical University Munich, Garching, Germany

Avinoam Ben-Shaul
Hebrew University of Jerusalem, Jerusalem, Israel

CONTENTS
1.1  Introduction...................................................................................................................................................................2
1.2  Polymorphism of Lipid–Water Systems........................................................................................................................3
1.2.1 Lyotropic Polymorphism...................................................................................................................................4
1.2.2 Thermotropic Polymorphism of Lipid Bilayers.................................................................................................4
1.2.3 The L β → L α and Other Bilayer Transitions......................................................................................................4
1.2.4 Lipid Monolayers...............................................................................................................................................5
1.2.5 Lipid Chain Order in the L α Phase....................................................................................................................6
1.3  The Lipidome: Membranes as Multicomponent Lipid Mixtures.................................................................................. 6
1.4  Lipid–Protein Sorting, Interactions, and Domain Formation.......................................................................................9
1.4.1 Lipid–Protein Interaction..................................................................................................................................9
1.4.2 Lipid and Protein Sorting and the Formation of Functional Microdomains (Rafts)....................................... 11
1.5  Membranes as Electrified Interfaces: Electrostatic Switching of Functional Proteins............................................... 11
1.5.1 Role of Lipid Charge on the Chain-Melting Transition................................................................................... 11
1.5.2 Underlying Thermodynamics and Kinetics..................................................................................................... 14
1.5.3 Macroion Aggregation on the Membrane Surface.......................................................................................... 14
1.5.4 Counterion Release.......................................................................................................................................... 15
1.6  Membrane Dynamics: Lipid Lateral Diffusion and Its Impact................................................................................... 15
1.7  Membrane Defects: Physics and Biological Function................................................................................................. 17
1.8  Biological Membranes as Elastic Shells...................................................................................................................... 18
1.8.1 Morphology and Shape Transitions of Soft Elastic Shells.............................................................................. 18
1.8.2 Shape Transitions of Stratified Soft Shells: The Red Blood Cell.................................................................... 19
1.8.3 Membrane Bending Excitations and Their Physiological Significance...........................................................20
1.8.4 Red Blood Cell Membranes as Actively Driven Semiflexible Statistical Surfaces......................................... 21
1.8.5 The Physiological Role of Membrane Flickering............................................................................................ 21
1.8.6 Molecular Aspects of Membrane Elasticity.................................................................................................... 22
1.9  Cell Adhesion: A Membrane-Based Process............................................................................................................... 22
1.9.1 Molecular Aspects........................................................................................................................................... 23
1.9.2 Insights Gained by Biomimetic Systems......................................................................................................... 23
1.9.3 Synopsis...........................................................................................................................................................24
1.10 A Short Outlook: Where We Stand and Where to Go.................................................................................................25
List of Abbreviations.............................................................................................................................................................25
Lipid Abbreviations and Nomenclature................................................................................................................................25
Types of Lipids.....................................................................................................................................................................25
Examples������������������������������������������������������������������������������������������������������������������������������������������������������������������������������25
Acknowledgements...............................................................................................................................................................25
Bibliography������������������������������������������������������������������������������������������������������������������������������������������������������������������������26

1
2 Handbook of Lipid Membranes

1.1 INTRODUCTION water, and hydrophobic groups to reside in oil-like envi-


ronment, Singer and Nicolson proposed their Fluid Mosaic
A critical step during biological evolution has been the Model (Figure 1.1b; [3]). This model, generally favoured by
enclosure of biochemical reactions inside vesicles made ­biologists and thus often referred to as “the standard pic-
of semipermeable membranes composed of lipid bilay- ture of a biomembrane”, depicts the lipid bilayer as a pas-
ers, or in the case of archaea bacteria of monolayers of sive two-dimensional (2D) fluid, enabling lateral mobility
bipolar bola-lipids. Integrating into the membranes, light- of integral and peripheral proteins, as needed to fulfil their
driven proton pumps with pH-dependent activity, such biological functions. A second model, the Heterogeneous
as Bacteriorhodopsin, enabled the reactions to proceed Shell Model [4], favoured by many physicists, attributed
under controlled acidic conditions. The incorporation of a more active role to the lipid matrix (Figure 1.1c). This
these pumps into lipid membranes by hydrophobic peptide model emphasized the tendency of proteins to assemble
sequences is a striking example of nature’s ability to trans- in functional microdomains of specific composition by
form genetic information into biological nanomachines by lateral phase separation that are driven by specific lipid–
making use of basic physicochemical principles of self- protein interactions in the multicomponent membrane mix-
assembly, such as the hydrophobic effect. Much of our ture. The model was suggested by studies of auxotrophs
present understanding of the principles underlying mem- of Escherichia coli bacteria which can only grow when
brane self-assembly and the correlation between membrane phospholipids are added to the nutrition medium. One can
structure and functions (such as proton pumping) is based thus replace the natural lipids by synthetic lipids with well-
on insights gained by designing sophisticated model sys- defined transition temperatures. The experiments showed
tems such as giant vesicles, black lipid membranes (BLMs), for the first time that biological membranes can undergo
lipid monolayers, and supported membranes. Correlations temperature-driven phase separation below the lipid chain-
between the molecular organization of lipid–protein bilay- melting transitions and, second, that membrane fluidity
ers and their basic biological function were established by enabling lateral diffusion of constituents is indispensable
comparative studies of artificial and natural membranes. for the division and survival of the cells [4].
Our main goal in this brief historical survey of membrane Two groundbreaking developments that have greatly
physics is to point out how our present understanding of motivated the interest of physicists in membrane biophys-
membrane biophysics has evolved through the development ics were the Huxley–Hodgkin model of nerve conduction
of novel experimental tools and theoretical concepts of soft by voltage-dependent ion channels and the discoveries by
matter physics. Luzzati and coworkers in the 1960s and 1970s of the rich
In 1925, comparing the surface area of erythrocytes with and fascinating lyotropic and thermotropic polymorphism
the monolayer area formed by spreading their constituents, of lipid/water systems [5,6]. The complex phase behaviour
Gortel and Grendel arrived at the conclusion that the thick- of these systems, and the notion that membranes are two-
ness of cell envelopes corresponds to that of a lipid bilayer dimensional smectic phases, has naturally attracted the
[1]. Ten years later, largely based on the electrical proper- attention of many physicists from the liquid crystal com-
ties of erythrocyte suspensions discovered by K. Fricke and munity. Soon afterwards, stimulated by Frank’s theory of
other electrophysiologists, Danielli and Davson suggested liquid crystals, Helfrich presented his curvature elasticity
that cell envelopes are trilaminar films composed of a theory of cell shape changes, relating the splay deforma-
lipid bilayer sandwiched between two layers of protein, as tion of smectic liquid crystals to the bending deformation
illustrated in Figure 1.1a, [2]. It took nearly 40 years before of lipid bilayers [7]. Around the same time, many physicists
this picture of the biomembrane has been independently were inspired by Marcel Bessis’ wonderful book The Red
challenged by more realistic models. In the United States, Blood Cells [8]. In parallel, electron microscopy and bio-
based on thermodynamic considerations pertaining to the chemical studies revealed that the red blood cell envelope is
strong preferences of ionic groups to be in contact with

(a) (b) (c)


Protein

Lipid

FIGURE 1.1 Early models of biological membranes. (a) Danielli and Davson model [2]. The membrane-associated proteins adsorbed
on both surfaces were assumed to control ionic permeability from either side of the lipid bilayer. (b) The fluid mosaic model of Singer
and Nicholson [3]. Lipids are assumed to play a passive role, forming a two-dimensional fluid layer embedding the proteins and
enabling their lateral transport. (c) The heterogeneous shell model. Lipids play an active role and serve the formation of local functional
lipid–protein domains (image reproduced from [4]).
A Short History of Membrane Physics 3

FIGURE 1.2 (a) Schematic view of ion translocation by ion carriers (left) and pore-forming amphiphilic peptides, such as gramicidin
(right). (b) Current fluctuations through BLM in the presence of small concentrations of gramicidin. The jumps in conductivity increase
by constant increments, indicating that one or several channels open simultaneously. Image reproduced from the work of D. Haydon
and S. Hladky [11].

a stratified elastic shell made up of the lipid–protein bilayer membranes by the patch clamp technique (reviewed in Ref.
and the associated spectrin–actin network [9] – stimulating [14]). BLMs have also been successfully used to study vari-
the formulation of the composite shell model of cell enve- ous other fundamental membrane processes, among which
lopes [4]. Today, we know that the spectrin–actin network are vesicle fusion [15] and the control of ion conductivity by
coupled to the plasma membrane (PM) is a universal design lateral phase separation [16]. In the last 20 years, the BLM
principle that stabilizes cell envelopes, e.g., of hair cells in techniques have been widely replaced by solid-supported
the auditory systems [10]. membranes [17], fabricated by deposition of lipid–protein
The development of the BLM technique by P. Mueller, bilayers on solid-state devices such as SiO2- or GaAs-based
M. Montal, and coworkers in 1962 (reviewed in Ref. [11]) semiconductors. Similar to BLMs, these model systems
allowed for the first time systematic studies of the physics offer many (still unexplored) advantages for studying fun-
of ion translocation across lipid membranes, mediated by damental membrane properties, such as the fusion of endo-
ion carriers (e.g., valinomycin) or through membrane pores somes with model membranes [16].
formed by the oligomerization of proteins such as grami- In the following sections, we shall mention various
cidin or alamethicin (Figure 1.2). The subsequent develop- aspects of membrane physics, focusing mainly on the early
ment of voltage jump current relaxation methods and ion contributions to each of the topics considered. More recent
current fluctuation spectroscopy provided detailed insights and modern developments will undoubtedly be described in
into the kinetics of diffusion-limited formation of ion chan- more detail in other chapters of this volume. Accordingly,
nels by complex formation of amphiphatic polypeptides we shall briefly discuss some of the early theoretical stud-
[12,13]. ies of membrane phase transitions, lipid chain order, lipid–
Notwithstanding the fading interest in BLM studies, the protein interactions, and membrane elasticity, yet we shall
contribution of this technique to our understanding of the barely mention the many recent large-scale and multiscale
physics of ion translocation through membranes has been molecular dynamics (MD) simulations of such systems
enormous. It provided the first experimental proof of the and phenomena. Obviously, we shall not be able to ­discuss
Huxley–Hodgkin postulate of voltage-dependent ion chan- all the many relevant aspects of membrane physics, nor
nels, paved the way for the interpretation of patch clamp to properly cite all the outstanding studies that contrib-
experiments (introduced by Neher and Bert Sakmann), uted to our current understanding of membrane structure
and showed that simple ion carriers, such as valinomycin, and ­function. We apologize in advance for any unjustified
exhibit stunning ion selectivity. For instance, the channel- ­omission of a relevant reference, as well as for our possible
forming alamethicin oligomers were shown to exhibit cat- personal biases.
ion or anion selectivity, depending on the charge of the
amino acid side groups facing the channel [12,13], thus 1.2 POLYMORPHISM OF LIPID–
providing the first experimental proof that the charged
WATER SYSTEMS
amino acids lining the surface of a membrane channel pore
determine their ion selectivity. Based on Fourier spectros- In 1968, based on sophisticated X-ray scattering ­studies,
copy of current fluctuations and voltage jump relaxation Luzzati and coworkers were the first to reveal the rich
experiments, sophisticated methods for the analysis of ion ­lyotropic and thermotropic polymorphism of phospholipid–
translocation kinetics through integral membrane chan- water systems [5,6,18]. They observed that natural lipids,
nels or those formed by the assembly of amphiphatic pro- such as egg lecithin, exhibit similar lyotropic phases to
teins (e.g., alamethicin) were developed between 1970 and those formed by synthetic surfactants. They also found that
1980. The simple design of BLM-based systems played a lipid bilayers undergo a first-order chain-melting ­transition.
key role in the development of these techniques and the Since then, numerous experimental and theoretical stud-
demonstration of their reliability. It thus paved the way for ies have been reported, extending and enhancing our
the kinetic analysis of single channel conductivity in cell understanding of lipid–water assemblies and their phase
4 Handbook of Lipid Membranes

behaviour. A few of the early findings are briefly outlined the respective phases denoted as L β and Lβ ′ . The Lβ ′ struc-
below. ture is generally observed in bilayers composed of lipids
with large polar head groups, such as phosphatidylcholines
(PCs), whose cross-sectional area is larger than that of their
1.2.1 Lyotropic Polymorphism
(fully stretched) two-chain hydrocarbon tails. The tilting
The lyotropic lipid assemblies of the greatest biological rel- enables tighter packing of the tails, enhancing chain cohe-
evance are the multilamellar (L) phase and the cubic (Q) sion within the hydrophobic membrane core.
phase including its disordered bicontinuous version known Below, we briefly outline a few of the numerous pioneer-
as the sponge phase (L3). Tightly packed multilayers are ing experimental and theoretical studies of bilayer phase
present, for instance, in the nervous system, playing a key transitions and lipid order in membranes. Related topics,
role as low capacitance layers in myelin sheath of axons. such as lipid–protein interactions, as well as membrane
A unique feature of the Q-phase is that its constituent lipid defects, dynamics, and curvature elasticity, are discussed in
molecules form an ordered 3D network of interconnected, subsequent sections.
water-filled, bilayer tubes [18]. Upon swelling, the cubic
symmetry may disappear, but the network topology remains
1.2.3 The Lβ → Lα and Other Bilayer Transitions
intact, resulting in the formation of the L3 phase. Interest
in this phase increased recently following the discovery by Common to all lipid bilayers is a first-order chain-melting
Rapoport and coworkers that the endoplasmic reticulum transition (Lβ → L α) from the gel phase to a 2D fluid phase,
(ER) can form tubular networks extending throughout the taking place at a well-defined temperature Tm. Above this
entire cell [19,20]. temperature, in the L α phase, the membrane is a 2D fluid,
The early findings about the polymorphism of lipid– enabling lateral translation of the lipids in the membrane
water assemblies indicated the possible role of lipid-phase plane, as well as limited conformational freedom of their
transition in controlling biological membrane processes. flexible hydrocarbon tails (see Figure 1.3). The extent of chain
For instance, the L β to L α transition discussed below allows conformational freedom depends sensitively on the cross-
cells to adjust the elasticity and fluidity of their membranes sectional area per lipid head group a, whose value depends
in response to changing environmental conditions [21]. on the balance of repulsive and attractive interlipid forces:
These notions stimulated the development and application The effective attraction between lipid tails, which tends to
of various new experimental techniques, as well as a burst minimize the unfavourable surface energy associated with
of theoretical studies. Important insights into the molecular exposing the hydrocarbon region to contact with water, ver-
order and dynamics of fluid phases were gained by mea- sus the repulsive forces due to electrostatic and excluded
surements of lipid orientational order parameters using volume interactions between head groups, as well as the
Deuterium nuclear magnetic resonance (NMR) [22,23], entropic repulsion between the hydrocarbon lipid chains in
infrared [24], and spin label electron spin resonance (ESR) order to increase their conformational freedom. This balance
spectroscopy [25]. Chain segment distributions and dynam- of forces is schematically described later in Figure 1.4.
ics were derived by neutron scattering methods [26–28], and Widely varied in their mathematical approaches, most
correlations between vesicle shape changes and phase tran- theories of the L β → L α transition agree that its primary
sitions were gained by freeze-fracture electron microscopy thermodynamic driving force is the gain in conforma-
(EM) (for review, see reference [29]). In parallel, thermo- tional (trans/gauche) entropy of the hydrocarbon lipid
dynamic properties could be measured with high precision chains above Tm, compensating for the partial loss of
using dilatometry [30–32], light scattering, Fourier trans- cohesive energy of the hydrocarbon chains in the passage
form infrared spectroscopy (FTIR) [21,30], and sensitive to the (~4%) less dense L α phase. This interplay between
calorimetric methods [33]. ­interchain attraction energy and conformational entropy is
reflected in the chain length dependence of Tm . Thus, for
example, in the case of bilayers composed of PC lipids with
1.2.2 Thermotropic Polymorphism of Lipid Bilayers
two saturated hydrocarbon tails, Tm increases from 23°C to
In addition to the rich polymorphism of lyotropic 3D lipid 41°C to 55°C as the lipid chain length increases from n = 14
phases, lipid bilayers exhibit a number of thermotropic to 16 to 18 methylene groups, respectively [29,36]. While
phase transitions taking place within the 2D membrane both the enthalpy and entropy changes in the transition
plane. Their number and character are even richer and more increase with n, the monotonic (roughly linear) increase
intricate in multicomponent lipid bilayer and lipid–protein of Tm = ∆H m /∆Sm with n indicates a stronger chain length
membranes [18,29,34–37]. At low temperatures, the lipid dependence of the transition enthalpy. The presence of a
molecules are immobile, forming a “gel” phase, in which double bond along the hydrocarbon chains has a dramatic
the lipid head groups are generally densely packed, orga- effect on the chain-melting transition. Depending on the
nizing typically in orthorhombic crystalline order. The position of the C=C bond, Tm may be 1°C–60°C lower upon
hydrocarbon lipid tails in the gel phase are stretched, with shifting the position of this bond from the vicinity of the
their main axes parallel to each other and directed either head group to the centre of the chain and then up again by
parallel or tilted with respect to the membrane normal, with about 50°C when the double bond is close to the chain end,
A Short History of Membrane Physics 5

as observed for dioctadecanoyl-phosphatidylethanolamine


(PE) (C18:1-PC) bilayers [38]. A possible explanation to this
behaviour is that when the double bond is located near the
beginning or end of the chain, most of the chain can still
behave as a saturated one enabling relatively efficient pack-
ing in the gel phase, while its presence in the middle of the
chain introduces chain tilting resulting in a perturbation to
lipid packing and thus a lower transition temperature.
Among the early theories of the chain-melting transition
are exact mathematical solutions to approximate bilayer
models, e.g., Nagle’s dimer model of hydrocarbon chains FIGURE 1.3 (a) The heat capacity of a suspension of vesi-
on a 2D lattice [39], Scott’s [40] and Pink’s [41] “few- cles composed of dipalmitoylphosphatidyl-glycerol (DPPG,
states” chain models, as well as Landau theories using the C:16,0 PG), showing the chain-melting transition (L α→ L β) and the
chain segment density (ρ), the average orientational order pretransition (L β → Pβ′). (b) Schematic illustration of the molecu-
parameter (η), or the area per head group (a) as the relevant lar architecture of the fluid (L α) and the three solid phases (Pβ, L β
thermodynamic order parameter [42–44]. Realistic, albeit and L c). The nontilted crystalline L c -phase forms by annealing the
approximate, molecular-level theories have also been pro- L β -phase at temperatures below the pretransition temperature TP.
(c) The solid phases are frustrated because the head groups favour
posed. First among these is Marcelja’s pioneering theory
the orthorhombic symmetry, while the chains prefer the triangu-
[45,46], where all rotational isomeric states of the hydro- lar lattice (of lattice constant a = 0.42 nm).
carbon chains are taken into account, and their interactions
and bond orientational order parameters are treated in anal-
ogy to the Maier–Saupe theory of the isotropic–nematic orientation of the crystal axes) is maintained. This new
transition. In addition to the chain-melting transition, his type of a solid state is known as the hexatic phase. Among
theory and subsequent molecular-level models, followed by others, it was suggested to characterize the s­ tructure of the
a growing number of increasingly sophisticated and exten- spectrin–actin network of erythrocytes (see, e.g., [29]).
sive MD simulations, have provided detailed information In many cases, predominantly in membranes composed
about chain conformational properties in the fluid phase, of lipids with large head groups, the chain-melting transi-
as discussed in little more detail in Section 1.2.5. Many of tion is preceded by a weak pretransition, taking place at
these approaches were later extended and applied to anal- a temperature Tp<Tm, whereby the bilayer transforms from
yse the phase behaviour of multicomponent lipid bilayers the crystalline Lβ phase to a corrugated phase, commonly
and lipid–protein membranes (Section 1.4) and to explain known as the ripple phase and denoted as Pβ [36,47].
and predict the molecular aspects of membrane elasticity Unravelling the origin of the ripple phase has challenged
(Section 1.8). many experimentalists and theorists. One rather consistent
Another topic of physicists’ interest has been the phase conclusion from the various studies is that the lipids in the
behaviour of the lipid bilayer below Tm [47]. While crys- long edge of the ripples are densely packed – resembling the
talline, the gel phases prevailing at these temperatures are Lβ order, while the shorter edge is thinner and lipid packing
remarkably flexible, resembling the glassy state of polyeth- there resembles the L α phase. The theories suggested range
ylenes. The hydrocarbon chain dynamics in the bilayers from continuum models, through molecular-level calcula-
involves kink formation and rapid diffusion along the hydro- tions, to MD simulations, yet a full picture is still missing
carbon chains, as discussed in Section 1.6 (see Figure 1.11). and the nature of the pretransition is still a matter of con-
Among the challenging issues pertaining to the crystalline troversy. Early models of the ripple phase can be found in
phases is their rich defect structure, which is largely due to Ref. [29] (for a recent study and survey of the ripple phase,
the frustration of lipid packing owing to two reasons: first, see, e.g., [49]).
because the fatty acid chains cannot rotate freely about the
carboxyl groups linking them to the head group phosphates 1.2.4 Lipid Monolayers
(in contrast to ether lipids) and, second, because the hydro-
carbon chains prefer forming a triangular lattice while the In the early 1970s, lipid monolayers were intensively stud-
head groups prefer orthorhombic packing (see Figure 1.3c). ied as potential model systems of biomembranes, offering
The defect structures associated with these frustrations unique possibilities to establish phase diagrams of lipid
[29] became popular paradigms for studying the physics of mixtures as functions of temperature and lateral pressure.
flexible two-dimensional solids such as polymerized mem- Monolayers are also useful to study the nucleation and
branes. These solids exhibit a novel type of a phase tran- growth (Ostwald ripening) of microdomains of lipid mix-
sition with diverging specific heat, termed the crumpling tures [50–52]. Furthermore, monolayer experiments pro-
transition, which is a consequence of the dissociation of vide direct determinations of the thermal expansivity and
dislocations [48]. The spatial long-range order of the crys- compressibility of lipid layers, enabling parallel character-
talline lattice of the low-temperature phase is lost in this ization of their molecular architecture by high-resolution
transition, while the long-range orientational order (i.e., the X-ray and neutron surface scattering [53].
6 Handbook of Lipid Membranes

Lipid monolayers have been extensively used to study the core is spatially uniform and equal to that in liquid alkanes
adsorption and enzymatic activity of proteins as a function [64,65]. An explicit expression for P(α ) – the probability of
of the lipid packing density. Striking examples are the cleav- finding the lipid chain in conformations α – can be derived
age of phospholipids by phospholipases and the exchange of (for any given area per lipid head group a) by minimizing
proteins between vesicles of different composition mediated the chain free energy subject to the constraint of constant
by lipid exchange proteins, showing that enzymatic activity segment density [62,63]. The result is
in the fluid–solid coexistence region is far more efficient
than in either the fluid or the gel phase [53]. Underlying
this behaviour is the diverging membrane compressibility
in the phase transition, enabling the enzyme to efficiently
P(α ) =
  
1
q {
exp −[ε (α ) +
∫ π(z)ϕ (α ; z) dz]/k T }(1.1)
B

grab lipids in the domain boundaries and membrane defects where ε (α ) is the chain conformational (e.g., trans/gauche)
accompanying the transition (see Section 1.7). Monolayer energy in conformation α ; ϕ (α ; z )dz is the number of chain
studies are also expected to provide important insights into segments whose centres in this conformation fall within
the mechanisms associated with the recruitment of oppo- the layer z , z + dz parallel and at distance z from the hydro-
sitely charged lipids to the vicinity of peripherally bound carbon–water interface; k B is Boltzmann’s constant; and
macromolecules such as proteins or DNA. T is the temperature. The π( z ) are Lagrangian parameters
representing the lateral pressure profile (see Figure 1.4),
1.2.5 Lipid Chain Order in the Lα Phase evaluated by solving the self-consistency ­equations:
Information concerning chain ordering and conforma- ϕ (z) = ∑ α
P(α )ϕ (α ; z ) = ρ ( z ), with ρ ( z ) = ρ denoting
tional freedom in the (biologically relevant) L α phase has the local chain density in the bilayer core. Given P(α ), one
been the goal of numerous experimental, theoretical, and can calculate any desired single chain property, e.g., the order
computer simulation studies. Of particular interest are the parameter profile, as illustrated in Figure 1.4. Furthermore,
orientational bond order parameter profiles of the lipid thermodynamic quantities, in the mean field approxima-
chains, which are usually provided by deuterium NMR tion, can be derived using statistical ­thermodynamic rela-
measurements [22,54] and the spatial distributions of lipid tionships involving the partition function, q, and the lateral
chain segments obtained by X-ray and neutron scattering pressure profile, π( z ). Such properties include lipid–­protein
[26,55,56]. The period from the late 1970s to the 1990s wit- interaction free energies and membrane e­ lastic moduli, as
nessed the appearance of numerous theories and simula- briefly mentioned in subsequent sections.
tion studies of lipid chain order, including elegant lattice
models [57,58], pioneering MD simulations [59], as well as
a number of mean field theories [45,60–62] showing very 1.3 THE LIPIDOME: MEMBRANES AS
good agreement with experiment (for a review of the early MULTICOMPONENT LIPID MIXTURES
theories, see, e.g., [63]).
The orientational bond order profile, {Sk }, represents the In the 1970s, biochemists began to analyse the complex
average relative orientations of the Ck –Ck-1 bonds along the lipid composition of biomembranes and to explore the
hydrocarbon lipid tails relative to the membrane normal. mechanism of lipid synthesis (see Refs. [29,69] for reviews).
They are directly related to the C–H bond orientations, Their ­studies showed that each cell organelle exhibits a
which can be determined by selective deutration of the Ck –H ­characteristic lipid composition, which is maintained during
the cell’s lifetime, despite the rapid exchange of ­material via
bond, yielding Sk = (1/2) 3cos2 θ k − 1 , where θ k is the angle
endocytosis, exocytosis, and intracellular vesicle ­trafficking.
between bond Ck –D bond along the hydrocarbon chain and In another series of elegant biochemical experiments, the
the membrane normal. The angular brackets denote aver- van Deenen group showed that the lipids are asymmetri-
aging over all possible chain conformations. In Marcelja’s cally distributed between the two leaflets of the erythrocyte
pioneering theory [45], {Sk } appears explicitly in the self- PM: the sphingomyelins (SPHMs) and oligosugar-carrying
consistency equations for the nematic-like molecular field, ­gangliosides residing only in the outer leaflet, along with
Φ = V0 (ntr /n)∑S k
k , with ntr /n denoting the fraction of
PCs. The inner leaflet was found to ­contain most of the PEs
and all the anionic lipids, specifically phosphatidylserines
C–H bonds in trans conformation. V0 ≈ 700 cal/mole was (PSs) that comprise about 7% of the acidic membrane lipids
used as the average van der Waals energy per chain in the and phosphatidylinositol PI-4,5-P2 comprising about 2%.
corresponding polyethylene liquid. His solution, for any Significant differences were also found with respect to the
given chain length, n, yields both {Sk } and the average area lipid composition of different organelles. Sphingomyelins
per head group, a, as a function of temperature. and oligosugar-carrying gangliosides reside mainly in
Other molecular field theories [61–63] evaluated the the PM and the late endosomes, a­ mounting to about 20%
probability distribution of chain conformations based on of the total phospholipid content of these organelles, as
the “hydrocarbon droplet” assumption, according to which ­compared with only 3%–10% of these ­lipids in other
the hydrocarbon segment density within the hydrophobic organelles. Deveaux and coworkers provided evidence
A Short History of Membrane Physics 7

(a) (b)

(c)

FIGURE 1.4 (a) The lateral pressure profile in a lipid bilayer. Head group repulsion resulting from electrostatic and excluded v­ olume
interactions and interchain repulsion due to conformational restrictions are balanced by the tendency of the bilayer to minimize
water–hydrocarbon contact. (b) Characterization of dynamics of chain conformations in terms of mean square displacement of chain
segments in horizontal and normal directions. The local diffusion coefficients in the two directions increase linearly with the distance
from the head group. This gradient in segment motion is equivalent to the gradient of static order parameter [27]. (c) Orientational bond
order parameters of the Ck–H bonds along the palmitoyl chain (left), and the oleoyl chain (right) of POPC. The palmitoyl chain is fully
saturated, while the oleoyl chain contains one double bond (between carbons 10 and 11), which perturbs chain order, resulting in the
sharp dip in the order parameter profile. The inset illustrates the angles θ k appearing in the definition of the order parameter. (For an
all-trans chain along the membrane normal, θ k = π/2, implying Sk = −1/2.) The triangles describe the experimental results of Seelig
and Waespe-Šarčević [66], the squares are from the molecular dynamics simulations by Heller et al. [67], and the circles are from mean
field calculations based on Eq. (1.1) [68].

that the asymmetric lipid distribution is maintained by t­ emperatures by increasing the fraction of unsaturated
specific ATP-dependent translocases called flippases [70]. ­lipids [72].
Differences in cholesterol concentrations are also strik- Taken together, these observations showed that the lipid
ing; the molar ­concentration of cholesterol in the nuclear composition is an intrinsic property of each cell type, that
membrane, the ER, and Golgi is less than 10%, as com- their synthesis is genetically controlled, and that lipid and
pared with 15% in lysosomes and 42% in the PM which protein biosyntheses are closely regulated. In analogy to the
is close to the ­solubility limit (see Figure 1.5b). Steck terms genome and proteome, we may therefore regard the
and ­coworkers showed that this gradient of cholesterol sets of lipids in the various organelles as the lipidome.
­distribution is actively maintained [71]. The fine-tuning of Unravelling the homeostatic control of lipid composi-
lipid ­homeostasis is mediated by specific exchange proteins, tion of cell organelles triggered numerous physicochemi-
transferring lipids between the outer leaflets of intracellular cal studies of the phase behaviour of multicomponent lipid
organelles (see Figure 1.12). and lipid–cholesterol layers. A wealth of experimental
Correlations between the thermodynamic phase state techniques were developed for this purpose, including ESR
and the biological function of the membrane became spectroscopy [21,34], NMR techniques [34,73], FTIR and
­evident following the observation that cells can adapt to the Raman spectroscopy [24], differential calorimetry [74],
ambient temperature by proper choice of lipid chain length, densitometry [75], and freeze-fracture EM (see [29] for ref-
the fraction of unsaturated lipids, and the ­number of dou- erences). All of these techniques determine phase boundar-
ble bonds of these lipids. In particular, specific enzymes ies by scanning the temperature, which means that the lipid
were found to help plant cells adjust to low environmental composition of the segregated phases changes continuously
8 Handbook of Lipid Membranes

(L β) phases (fluid–solid demixing). Examples of the sec-


ond situation are fluid phospholipid–cholesterol mixtures
(as shown in Figure 1.5b) and fluid mixtures composed
of nonsaturated PC and PE lipids, as studied by Wu and
McConnell [73]. Examples of the third case are mixtures
of saturated lipids differing in chain length by more than
two CH2 groups, such as DMPC (C14:0-PC) and DSPC
(P18:0-PC). This mixture exhibits a peritectic phase dia-
gram with the solid–solid miscibility gap penetrating into
the fluid–solid region, resulting in precritical phenomena
above the liquidus temperature [77,78].
The phase behaviour of binary and ternary mixtures of
FIGURE 1.5 (a) Demonstration of a phase transition of vesicles
phospholipids with cholesterol has been the focus of numer-
of brain sphingomyelin (SPHM), reproduced from [85]. (b) Phase ous studies. Mixtures of zwitterionic saturated lipids (such
diagram of a binary mixture of cholesterol and PC lipid, in this as PCs or SPHMs) with cholesterol exhibit phase diagram
example DMPC with Tm = 23°C. Below Tm, a solid–solid miscibil- similar to the one shown in Figure 1.5 for the special case of
ity gap forms, which separates two homogeneous low-­temperature DMPC/cholesterol. SANS experiments showed that, below
phases described in the text. The vertical bar marks a stoichio- the chain-melting transition (T<Tm), this mixture exhibits
metric mixture containing 75% PC. Above Tm, two fluid phases a miscibility gap between xchol ≈ 0.08 and xchol ≈ 0.25. At
­coexist. They are separated by a miscibility gap that is expected xchol < 0.08, a solid solution is formed, while at xchol > 0.25, a
to end in a critical point at Tc. It separates the cholesterol-poor
homogenous liquid solution of cholesterol in DMPC forms,
L α phase and the cholesterol-rich condensed L αc phase. Image
adapted from Ref. [86].
which saturates at xchol ≈ 0.42. To fulfil the phase rule, the
phase line at xchol ≥ 0.25 is assumed to mark a stoichiometric
mixture. Several experiments providing evidence for this
interpretation are summarized in Refs. [77,78].
while scanning through the coexistence region. Therefore, The packing density in the saturated solution (xchol > 0.2)
these methods do not allow to determine ­equilibrium-phase is 20% higher than that of the solid solution. It is thus often
diagrams or to directly observe the segregation of phases referred to as a “condensed ordered phase”, despite being
of equal symmetry, such as solid–solid or ­fluid–fluid a 2D fluid. Above Tm, there is a fluid–fluid miscibility gap,
immiscibility. At present, small-angle neutron s­ cattering separating a cholesterol poor expanded phase, L α, from a
(SANS) in combination with contrast matching is the only cholesterol-rich and condensed, yet liquid, phase L αc (where
method enabling the establishment of phase diagrams the index c stands for “condensed”). Such fluid–fluid coex-
of mixed bilayers of mixed vesicles in thermodynamic istence of phospholipid–cholesterol mixtures was also
equilibrium [76]. suggested by theoretical studies [79,80]. The molecular
An ongoing issue since the early days of membrane structure and phase changes in lipid–cholesterol systems
physics has been whether nature makes use of lipid-phase have been studied in great detail, both experimentally [81],
separation in order to maintain the characteristic lipid and and by MD simulations [82].
protein composition of cellular organelles. Two major ques- The biological relevance of the phase diagram in Figure
tions that arise in this context are (1) why is cholesterol 1.5b becomes evident when we consider the fatty acid chain
mainly accumulated in the PM? and (2) how are proteins in distribution of membrane lipids (see Table 1.1 in Ref. [29]).
the PM recycled during membrane turnover? It shows that SPHMs expose mainly long-chain lipids with
Numerous studies on binary lipid–lipid mixtures and few double bonds (in particular C16:0, C18:0, C24:0, and
ternary lipid–lipid–cholesterol mixtures, in parallel to the C24:1), Therefor the natural SPHM fraction exhibits a
search for specific lipid–protein interaction mechanism, chain-melting transition near physiological temperature, as
provided partial answers to these questions. Research along shown in Figure 1.5a [75,76]. In contrast, the glycerol-based
these lines was guided by the expectation that phase sepa- lipids, such as PCs, contain mainly C16:0, C18:0, C18:1,
ration processes in phospholipid mixtures, liquid crystals, C18:2, and C20:4 and melt well below 10°C. Other nota-
and metal alloys are determined by the same thermody- ble facts are that cholesterol associates preferentially with
namic concepts, as outlined in the regular solution theory saturated lipids [83,84] and acts as fluidizer of crystalline
of Hildebrand or the Bragg–Williams lattice models (see membranes [29,76].
Ref. [29]). These concepts predicted that at constant tem- Taken together, these findings suggest that the lipid moi-
perature, phase separation is expected to occur if the com- ety of mammalian cells can be regarded as quasi-binary
ponents exhibit different symmetries (called symmetry mixture composed of glycerol lipid (which melts at T ≤ 0°C)
rule in liquid crystal physics) or if the molecular shapes of and SPHM (with Tm≈40°C). Since the glycerol lipids exhibit
two components (forming the same type of phase) differ a high content of unsaturated fatty acids, the cholesterol
remarkably. An example of the first case is the decomposi- content (xc ≤ 10 mole %) and the lipid packing density are
tion of DMPC and DPPC mixtures into fluid (L α) and solid small [83]. In contrast, the high-melting temperature SPHM
A Short History of Membrane Physics 9

fraction is saturated with cholesterol and thus forms a con- of interfacial hydrophobicity scales provide important
densed (glass-like) state called Lα c . information pertaining to the folding free energies of the
A possible role of the pseudo-binary lipid mixtures for peptides and their propensity to insert into the lipid hydro-
the generation of functional domains in cell membranes phobic core [87]. While preferring the hydrocarbon lipid
and lipid–protein sorting became only evident after the chains over the aqueous environment, the presence of a
establishment of elastic and electrostatic mechanisms of hydrophobic protein inclusion in the membrane core incurs
selective lipid–protein interaction, which will be discussed a nonnegligible free energy penalty associated with the
in the following. perturbation of the local lipid order of their surrounding
lipid environment. The perturbation energy is proportional
1.4 LIPID–PROTEIN SORTING, to the number of lipid molecules affected by the presence
of the integral inclusion and hence to the circumference of
INTERACTIONS, AND
the protein. Indeed, experiments with spin-labelled lipids
DOMAIN FORMATION reveal that the fraction of lipids associated with intramem-
Biological functions depend critically on the interaction of branous proteins is proportional to their perimeter [88]. It
the lipid membrane with peripheral and integral proteins also depends on the hydrophobic mismatch, δh = hP −hL ,
and most often with proteins composed of both hydropho- which measures the difference between the hydrophobic
bic and polar domains. The interaction between peripheral height of the protein and the thickness of the unperturbed
proteins and lipid membranes is primarily electrostatic, lipid bilayer [89], as illustrated in Figure 1.6.
involving generally attractive forces between polybasic In multicomponent lipid membranes, the presence
domains and anionic lipids. These interactions will be of integral inclusions may lead to lipid sorting, attract-
addressed in the next section, while here we shall comment ing lipids of matching height and chemical affinity to
on the interactions between hydrophobic integral proteins their immediate vicinity. Furthermore, in analogy to the
and their surrounding environment of lipid chains. hydrophobic interaction – whereby two nonpolar solutes in
water are attracted to each other because in contact they
cause a lesser perturbation of the water structure around
1.4.1 Lipid –Protein Interaction
them – the interaction between two hydrophobic inclu-
The free energy changes associated with the partitioning sions in a lipid membrane is attractive because, together,
of polypeptide chains onto the hydrocarbon–water inter- they perturb a smaller number of neighbouring lipids as
face of lipid membranes depend sensitively on their length compared with their being far apart of each other. If the
and amino acid composition. Detailed measurements perturbation of lipid order is large enough, as in the case

FIGURE 1.6 (a) Elastic distortion of a lipid bilayer induced by the mismatch of the hydrophobic thicknesses of the bilayer (hL) and
the membrane spanning domain of the protein (hP). (b) Model of the plasma membrane suggested by the pseudo-binary phase diagram
of Figure 1.5 and the Bretcher–Munroe analysis of hydrophobic length distribution of proteins [107]. The membrane spanning parts of
band III comprise 25 amino acid residues ( hP ≈ 3.8 nm ) and is expected to be surrounded in the outer monolayer primarily by SPHM
with C24:0 chains and PE or PC with C18: 0 chains. The cholesterol is expected to accumulate in the outer leaflet. (c) Above a threshold
concentration of short peptides (hP < hL ) in a bilayer containing long-chain lipids and/or lipids with negative spontaneous curvature, a
transition takes place to the inverted hexagonal phase, allowing more conformational freedom to the long lipid chains.
10 Handbook of Lipid Membranes

of large δh = hP −hL , the lipid-mediated attraction between width of the perturbation annulus surrounding an isolated
proteins can overcome their translational (mixing) entropy, inclusion involves about three lipid diameters. The range of
resulting in their 2D condensation to form protein-rich 2D perturbation and its magnitude are larger for nonzero hydro-
domains. phobic mismatches because the lipid chains adjacent to the
Treating the lipid–protein membrane as a 2D liquid mix- inclusion must stretch (δh = hP −hL > 0) or compress (δh < 0)
ture of N lipids of cross-sectional area a, and N P proteins in order to avoid exposing hydrophobic protein regions to
of area ga, its free energy in the mean-field approximation water [92]. The lipid-mediated interaction between hydro-
is given by phobic inclusions has also been studied by quite a few con-
tinuum elastic theories. Thus, for example, Schröder [44]
∆GLP = Nk BT {ϕ L ln ϕ L + (1/g)ϕ P ln ϕ P + χϕ Lϕ P } predicted that the attraction between inclusions decreases
exponentially as a function of their distance R, while
(1.2) Goulian et al. [93] found a 1/R4 dependence. Safran and
with ϕ L and ϕ P denoting the area fractions of lipid and pro- coworkers emphasized the importance of the lipid sponta-
teins, respectively. Proteins whose hydrophobic domain neous curvature, revealing the possible existence of barriers
consists of a single α-helix occupy about the same hydro- to protein attraction resulting in nonmonotonic interaction
carbon–water surface area as that of one lipid molecule potentials [94].
(typically around 70 Å2) implying g ≈ 1. Larger proteins, In addition to in-plane protein aggregation, the ­elastic
such as the band III ion channel, occupy larger membrane deformation of the lipid bilayer by hydrophobic i­ nclusions
areas amounting to as many as g ≈ 20 lipids. As usual, the can sometimes lead to rather dramatic structural trans-
nonideality interaction parameter χ is proportional to the formations of the lipid–protein matrix. For example, it
difference between the lipid–protein interaction poten- was found that beyond a certain critical concentration of
tial and the average of the lipid–lipid and protein–protein short hydrophobic peptides (δh <0) in a lipid bilayer, a
potentials. It should be noted, however, that unlike the case morphological transition of the lipid–peptide mixture can
of flexible polymers in solutions, integral proteins in mem- take place from the lamellar (Lα ) to the inverse-hexagonal
brane are rigid, so that χ depends on their circumference (H II) phase, with the peptides serving as bridges between
length rather than on their length. The critical point implied neighbouring water tubes (see, e.g., Refs [95,96]). The
by the mixing free energy Eq. (1.2) is specified by a lower critical peptide concentration depends on the spontane-
critical protein concentration than that of ordinary ( g = 1) ous curvature of the lipids (preferred by lipids with nega-
mixtures, namely ϕ Pc = 1 / (1 + g ) → 1/ g , and a smaller tive spontaneous curvature) and the bending rigidity of the
embedding lipid matrix [97]. Another well-known peptide-
( )
2
critical interaction strength, χ = 1 + g 2 g → 1/2. induced morphological transition that has been extensively
Resembling polymer solutions, the binodal curve for large g studied, both e­ xperimentally (see, e.g., Refs [98,99]) and
is strongly skewed, corresponding to very low protein solu- theoretically (see, e.g., Refs [100–103]), is the formation of
bility. For example, the solubility limit of Band III at room membrane pores by amphipathic, antimicrobial, peptides.
temperature is ϕ P = 10 −4. Similarly, many studies have been concerned with the
The strength of lipid–protein interactions can be deter- kinetics and thermodynamics of gramicidin channel forma-
mined by measuring the shift in the lipid chain-melting tion [100,101,104,105].
transition temperature caused by small amounts of mem- Like many other membrane properties, lipid–protein
brane proteins, or by the shift of the solidus and liquidus interactions have also been studied using computer simu-
lines [29]. Using light scattering and EM imaging, Riegler lations, including both Monte Carlo (MC) simulations and
and Möhwald [90] studied the gel–liquid crystal transition especially and in rapidly growing numbers, MD simula-
of vesicles composed of PC lipids of different chain lengths tions. In recent years, with the advent of fast computers
containing integral (photosynthesis reaction centre) pro- and the development of sophisticated computational algo-
teins. Consistent with the “mattress model” of Mouritsen rithms, most of the theoretical–computational studies of
and Bloom [89], they found that the lipid-mediated attrac- lipid–­protein membranes involve MD simulations. The
tion between the proteins increases with the magnitude of qualitative insights that have been provided by earlier (both
the hydrophobic mismatch, δh =hP −hL . In parallel, several molecular-level and continuum) theory approaches are
molecular-level models and quite a few continuum elastic ­nevertheless invaluable, especially when large-scale coop-
theories have been proposed to account for the interactions erative phenomena such as protein-mediated phase transi-
between membrane inclusions. In 1975, using liquid crystal tions or biomembrane elastic properties are concerned.
elasticity theory, Gruler pointed out the changes imposed The adsorption of proteins exposing hydrophobic mem-
on membrane curvature due to conical inclusions [91]. A brane anchors can mediate the formation of local buds by
year later, Marcelja applied his molecular field theory to the lateral expansion of one monolayer. It has been postulated
case δh = 0, where the only perturbation to lipid order is the that such mechanisms of induced budding can trigger the
presence of the rigid protein wall [46]. He found that the formation of coated pits [86] and caveoli [106].
A Short History of Membrane Physics 11

1.4.2 Lipid and Protein Sorting and the Formation EM (reviewed in Ref. [29] and later by microfluorescence
of Functional Microdomains (R afts) techniques [111]). Theoretical studies were based on the
combination of the bending elasticity model and the Cahn–
A detailed survey of the length distribution of the hydro- Hilliard theory of spinodal decomposition [112].
phobic thickness of integral proteins residing in the PM and Unfortunately, the interest in the homeostasis of mem-
the Golgi apparatus due to Bretcher and Munro provided brane lipid composition and the physics of structure for-
new insights into the physical mechanisms of lipid and pro- mation by specific lipid–protein interactions has faded
tein sorting during membrane turnover by endocytosis and considerably in recent years, perhaps because these are very
exocytosis [107]. They found that the hydrophobic domains complex fields of research. We must remember, however,
of integral proteins (consisting of 15 amino acid residues; that proper physiological function of cells and hence our
hP ≈ 2.5nm ) in the Golgi apparatus are about 0.5 nm health depend critically on the control of lipid and mem-
shorter than those present in the PM (hL ≈ 3nm). Together brane protein patterns. Diseases are often associated with
with the selective lipid–protein interaction by hydrophobic dysfunctions of the lipidome. Thus, for example, eryth-
matching shown in Figure 1.6a, the Munroe Bretcher analy- rocytes of patients suffering from diabetes exhibit a 1–2
sis suggests that protein sorting within the PM as well as mole% higher cholesterol content than those of healthy peo-
between the PM and intracellular organelles could be con- ple, indicating a correlated metabolic disorder. The higher
trolled by lateral phase separation. As illustrated in Figure cholesterol content reduces the erythrocytes deformability
1.6b, SPHM and the gangliosides reside in the outer leaflet and could possibly result in blood flow dysfunction.
of the PM. Comprising only 12%–30% of the lipid popula-
tion in the PM, SPHM can only cover part of the cell enve-
lope. This suggests that the cholesterol-rich SPHM fraction 1.5 MEMBRANES AS ELECTRIFIED
forms lateral domains within the PM which can accommo- INTERFACES: ELECTROSTATIC
date proteins with long hydrophobic domains, as illustrated SWITCHING OF FUNCTIONAL PROTEINS
in Figure 1.6b. One biologically important example is the
The electrical surface potential of biomembranes is dic-
transferrin receptor [29,108].
tated by two contributions: first, the presence of about 10%
This type of domain should not be confused with “rafts”
of charged lipids exposing their acidic head groups to the
[109]. Rafts are generally observed as Triton-insoluble
cytoplasmic surface and, second, the electric dipole layer
membrane fragments, and it cannot be excluded yet that
formed by the carboxyl groups of the glycerol backbone,
they are artefacts generated following the modification of
which is located about 0.5 nm below the lipid–water inter-
the lipid structure by Triton (108B).
face (reviewed in Ref. [29]). The 2D density and spatial
Another case where lipid–protein sorting plays a key role
distribution of surface charges affect the structure and ther-
is the recycling of receptors after endocytosis by coated pits
modynamics of isolated membranes and play a crucial role
and caveoli. A well-studied example is the coated vesicle–
in the recruitment, binding, and activation of functional
mediated transfer of iron ions into cells via the Fe carrier
proteins with polybasic domains, as discussed below.
transferrin bound to transferrin receptors (reviewed in Ref.
[86]).
Evidence has been accumulated indicating that some 1.5.1 Role of Lipid Charge on the
cell surface receptors are directly recycled from the early Chain-Melting Transition
endosomes to the PM by the Rab GTPases Rab 4 and Rab
5, thus avoiding the detour about the multibody particles One of the first questions that fascinated physicists has
[110]. For these special (but vital) cases, the lipid–protein been concerned with the influence of the lateral interac-
sorting could be controlled by the phase separation of the tion between charged lipid head groups on the gel-to-liquid
sphingomyelines and the glycerol lipid. The transferrin crystal transition temperature, Tm. As first shown by Träuble
receptor exhibits a particularly long hydrophobic domain and Eibl [113] and further elaborated by Jähnig [114]), the
(hP ≈ 4.5 nm [107]) and could be recycled through the surface lateral pressure associated with the electrical dou-
above mechanism. A major step of the recycling is the for- ble layer, π el, can be regulated by varying the pH or the
mation of microdomains of different curvatures by the two ionic strength of the embedding solution. The correspond-
segregated phases, which grow via Ostwald ripening until ing shift in transition temperature is given by
the endosomal vesicle becomes unstable and eventually
decays into two vesicles [85,111]. Model membrane studies ∆Fel ∆A
δ Tm = ≈− π el (1.3)
suggest that fission of vesicles from large endosomes may ∆S ∆S
also be triggered by cholesterol at molar fractions close to
the solubility limit of xc ~ 0.42 [85]. where π el = − ∂ Fel / ∂ A with ∆Fel denoting the excess electro-
The phase separation–induced shape changes, and vesi- static free energy of the charged membrane relative to the
cle fission is an ongoing theme of membrane physics. It was neutral one, with ∆S and ∆A denoting the changes in molar
extensively studied experimentally, first by freeze-fracture entropy and molar area in the chain-melting transition,
12 Handbook of Lipid Membranes

respectively, both of which are positive. The transition discovery that acidic lipids, in particular phosphoinositides
temperature of the charged membrane is lower because (PI), such as PI-4,5-P2 and PI-3,4,5-P3, play a key role in
the electrostatic pressure, which tends to increase the area the activation of many of the proteins involved in signal
per molecule, favours the more expanded fluid state, thus transmission across membranes and the coupling of the
enhancing the transition. actin cortex to the PM. These electrostatically switchable
Application of the classical Gouy–Chapman–Overbeek proteins contain polybasic sequences comprising up to
theory of electrical double layers led to important quanti- about 10 basic residues (generally lysines) and posttransla-
tative relationships between the change in transition tem- tionally coupled fatty acid chains (see Figures 1.7 and 1.8).
perature and the degree of dissociation of the lipid head In the resting state of cells, these proteins reside in a
groups. Theories developed by Jähnig [114] and others (as sleeping conformation within the cytoplasm, hiding their
discussed in detail by Cevc [115] and Andelman [116,117]) charged residues and fatty acids. They are activated by
enabled the calculation of the surface pH as a function of exposing their membrane binding sequences (for instance,
the electrostatic surface potential and the ionic strength of through phosphorylation) resulting in their electrostatic–
the aqueous phase. An important consequence of the nega- hydrophobic binding to the membrane surface, as ­illustrated
tive membrane surface charge revealed by these early stud- in Figure 1.7. Important examples are the molecular switches
ies is the lowering of the surface pH, which plays a key of the GTPase family and their associated helper proteins,
role in controlling the degree of dissociation of lipids and such as GTP exchange proteins (GEF), which activate the
adsorbed proteins [118]. At low ionic strengths (I ≤ 100 mM), GTPases by replacing GDP by GTP. The GTPases can be
the effect can be dramatic, inducing significant pH shifts; switched on either directly by the electrostatic hydrophobic
ΔpH ≥ 2 [29]. The effects are, however, small at physiologi- binding (as in the case of the Ras proteins in Figure 1.7a)
cal ionic strengths of I ≈ 400 mM, explaining the fading or indirectly by coupling to the membrane-anchored GEF.
interest in electrically induced phase transitions. The active GTPase attracts and triggers the function of one
The electrostatic interactions between lipid membranes or several enzymes, e.g., kinases.
containing charged lipids and oppositely charged macro- The delicate balance of forces underlying the physics
molecules (such as proteins exposing polybasic sequences of the electrostatic–hydrophobic membrane recruitment
or DNA) are of great biological relevance. Many of the early of natural proteins has been first systematically explored
studies of these interactions have focused on the calcium- by McLaughlin and coworkers [122–124] who studied the
induced membrane binding of annexin to PS-containing binding of the myristoylated alanine-rich C-kinase sub-
membranes [119]. Other early studies demonstrated the strate (MARCKS) protein to partially charged membranes.
charge-induced switching of protein function. One intrigu- MARCKS – serving as a paradigm of proteins recruited
ing example is the electrostatic switching of the orientation to membranes by electrostatic–hydrophobic forces –
of amphiphatic helices exposing charged head groups in ­consists of a polybasic “effector” domain (ED) contain-
pH-dependent manner from a horizontally adsorbed state ing 13 lysine residues and 4 intercalated serine groups.
into a membrane spanning state, whereby peptide oligo- The ED is flanked by two flexible chains, both comprising
mers organize into ion channels, enabling their function as ~150 amino acid ­residues. The N-terminus flexible domain
antimicrobial peptides [120]. ends with ­myristoyl anchor (see Figure 1.8b). Similar
The interest in electrostatic membrane–protein interac- to the case of K-Ras binding described in Figure 1.7, the
tion increased dramatically in recent years following the membrane-­binding free energy of MARCKS involves a

(a) (b) Sleeping Active


N Enzyme Anchor
+
++
KKKKKSKKCVIM P Kinase
+ +
+ +
Basic
Adaptor domain
O ++
Activators

FIGURE 1.7 (a) Domain of structure of the Ras-proteins p21Ras (K-RAS) as a paradigm of protein activation by recruitment to mem-
branes through electrostatic hydrophobic forces. The Ras-protein family consists of a variable domain at the C-terminus containing
the polybasic domain and the functional N-terminus. Note that the sequence of lysine groups (K) is interrupted by a polar serine group
(S) which can be negatively charged by phosphorylation (see the example of MARCKS). The cysteine group (C) is attached to a fatty
acid serving as a hydrophobic anchor. (b) Enzyme activation by electrostatic–hydrophobic switching. The protein (shown at the top) is
activated by phosphorylation, resulting in the exposure of fatty acid anchors and polybasic peptide sequences followed by binding to
the membrane by electrostatic and hydrophobic forces. It acts as switch that recruits and activates specific adaptor proteins which in
turn attract and activate proteins involved in signal transduction (see also Ref. [121]).
A Short History of Membrane Physics 13

(a) Hydrophilic loop Hydrophilic (b)


tail
MARCKS
Polybasic
segment N Loop Tail
+ + + +
= =

Extracellular KKKKKRFSFKKSFKLSG FSFKKNK


side

(c) IP3 Ca++


Serine
Activated
phosphorylation
PLC- PKC
MARCKS
DAG
++++++ ++ +++ + ++++++ ++++++
-
- -

5-

PI-4,5-P2

+++ Hyperactive
P +++
P binding site
MARCKS
++++++ ++ PI-3K ++ ++++++
-
-7 -7 -

PI-3, 4,5-P2

FIGURE 1.8 (a) Schematic illustration of protein binding to membranes by hydrophobic–electrostatic forces. (b) Snapshot from a
Monte Carlo simulation of MARCKS adsorption, showing the enrichment of the binding region by polyvalent acidic lipids (purple),
and the hydrophobic binding of the myristoyl anchor at the N terminus [127]. (c) Possible control of the access of the MARCKS-covered
phosphoinositides by functional proteins through protein kinase C (PKC)–mediated unbinding of the protective layer of MARCKS
(shown on the right side). PKC is activated by local bursts of Ca++ and DAG anchors, which are generated by phospholipase Cγ
(PLC-Cγ). The activated PKC phosphorylates the serine groups of MARCKS resulting in the explosive displacement of the protective
MARCKS layer and the exposure of PI-4,5 anchors. The right side shows the membrane recruitment and activation of the PI-3,4,5-P3
generating protein PI-3K. It generates a burst of the highly selective lipid anchor PI-3,4,5-P2 which enhances the membrane recruitment
of many functional proteins, including PLC-Cγ and PI-3K. Note that in the absence of MARCKS, the lipid anchor generators PLC-Cγ
and PI-3K would be constantly active, which could result in overexcitation of cells.

subtle interplay between (1) the electrostatic attraction of of −3 or −5) which comprises only ~2% of the ­membrane
the basic domains to acidic lipids, ∆Fel , which provides the lipids (as compared with 10%–30% of the m ­ onovalent PS)
major contribution to the binding free energy, (2) the hydro- [123,127–129]. MARCKS–­membrane interaction is just one
phobic force due to the fatty acid anchor, ∆Ftail, and (3) the of many examples of the very strong and selective ­binding
entropic free energy loss associated with the ­flexible – of functional proteins to p­ hosphoinositides (PI-4,5-P2 and
intrinsically unfolded – amino acid chains extending into PI-3,4,5-P3). Such binding is generally mediated by specific
the aqueous phase, ∆Floops . ∆Ftail can be estimated by the protein homology domains. Important examples, besides
empirical law ∆Ftail = (11 − 3n) kJ M−1 [29] or from mea- pleckstrin homology domains, are the C1 and C2 domains.
surements of unbinding forces by single molecule force The former harbours specific binding pockets for phos-
spectroscopy [125,126]. The electrostatic binding free phoinositides, while the C-domains penetrate the lipid layer
energy of the ­effector domain and the entropy loss of the with two hydrophobic loops [130]. The binding of the C2
flexible chain extending into the cytoplasm were estimated domain is calcium dependent.
by MC studies [127]. A possible role of MARCKS is to prevent the unwanted
Consistent with experiment and theory, efficient binding membrane recruitment and activation of enzymes (such
of the basic domain requires the colocalization of a matching as phospholipases) by inducing the segregation of the
number (namely, 13) of acidic lipid charges to the small ED– ­phosphoinositides and their shielding from enzymatic
membrane contact zone. Importantly, most of the protein attack. Indeed, about 10 µM of MARCKS suffices to inhibit
charge neutralization is achieved through the recruitment of the phospholipase Cγ-mediated generation of the second
the polyvalent PI-4,5-P2 lipid (generally carrying a charge messengers diacylglycerol (DAG) and inositol triphosphate
14 Handbook of Lipid Membranes

(IP3). The electrostatically induced segregation of charged entropy loss in comparison with that of monovalent acidic
lipids by basic polypeptide sequences such a polylysine has lipids [127,128].
indeed been demonstrated earlier [131]. In parallel to the thermodynamic analyses of the
The screening of the PI-4,5-P2 lipids is abolished by ­electrostatic–hydrophobic protein binding, the dynamics
phosphorylation of MARCKS through protein kinase C of this process has also been studied, both experimentally
(PKC), resulting in their unbinding from the membrane. and theoretically. Fluorescence correlation spectroscopy
The exposure of PI-4,5-P2 then enables the activation (FCS) measurements revealed that peptide diffusion on
of molecular switches (such as GTPases) following the the surface of membranes containing PI-4,5-P2 is signifi-
schemes illustrated in Figures 1.7 and 1.8. One important cantly slower than on those containing only monovalent
example is the generation of the specific anchor PI-3,4,5-P3 lipids. It was also found that PI-4.5-P2 diffusion is substan-
by phosphoinositol triphosphate-kinase (PI3K) shown in tially hampered [129], indicating correlated diffusion of
Figure 1.8c. PI-3,4,5-P3 acts as a second messenger which the peptide and the multivalent counterlipids. Theoretical
strongly enforces the specific binding of molecular switches modelling combining the Cahn–Hilliard theory to describe
of the GTPase family or of the phospholipase-Cγ, which the diffusion of lipid components in the inhomogeneous
generates the lipid anchor DAG. Since DAG mediates the peptide-dressed membrane, and MC simulations to model
membrane binding and activation of protein kinase C, the peptide diffusion provide further insights into the coupled
protective shield of MARCKS can be rapidly displaced to peptide–lipid diffusion dynamics. Specifically, strongly
expose PI-4,5-P2. attracted by the electrochemical potential gradient due to
A vital role of MARCKS has been established recently. the peripheral peptide, the PI-4.5-P2 lipids rapidly diffuse
Together with protein kinase C, MARCKS plays a key role toward the interaction zone, while monovalent lipids are too
for the plasticity of synapses, by enabling the rapid remod- slow to follow the motion of the peptide. Furthermore, the
elling of dendritic spines at the tip of axons. Knock-out of relatively strong, albeit reversible, binding of the multiva-
its gene is lethal [132]. lent lipids to the adsorbing peptides introduces an effective
In summary, numerous vital membrane-based ­processes, drag that slows down the diffusion of this “complex” [134].
associated with the restructuring of the actin cortex, such
as cell locomotion and cell adhesion, are controlled by the
1.5.3 Macroion Aggregation on
recruitment of actin-binding proteins to the ­cytoplasmic
the Membrane Surface
leaflet through electrostatic–hydrophobic forces. These
proteins swim in the cytoplasmic space in a sleeping In analogy to the lipid-mediated aggregation of integral
­conformation and are recruited to specific sites at the cell proteins, the perturbation of lipid organization by periph-
envelope in a logistic way by external cues, such as cyto- erally bound macroions can lead to their 2D aggregation
kines or hormones. Most important is the very strong selec- on the membrane surface [131,135]. The underlying driv-
tive binding to the PI-3,4,5-P3 lipids by the specific binding ing forces are quite different, however. One interesting
pockets other than pleckstrin homology domains [130,133], case is that of proteins adsorbing onto a membrane com-
such as the C2 domain mentioned above. In this way, the posed of a ­nonideal, yet subcritical, binary lipid mixture of
functional protein may selectively bind to PI-3,4,5-P3, charged and neutral lipids. That is, the nonideality interac-
although this lipid anchor is by a factor 100 less abundant tion parameter of the lipid mixture (corresponding to χ in
than PI-4,5-P2. Eq. (1.2), for the case g = 1) is not large enough to induce
phase separation of the mixture. However, when macroions
1.5.2 Underlying Thermodynamics and Kinetics adsorb onto the membrane surface, they attract oppositely
charged lipids to their binding zone, resulting in the forma-
Two kinds of entropy play principal roles in the electrostatic tion of small membrane patches rich in charged lipids, as
binding of peripheral “macroions” such as proteins, DNA, illustrated in Figure 1.9. Each of these patches involves a
and other biopolymers to lipid membranes. The first, com- line energy proportional to nχ , where n is the number of
mon to the attraction between any two oppositely charged interlipid contacts at the perimeter of the segregated lipid
macroions, is the gain in 3D translational entropy of the patch [136]. To avoid the unfavourable line energy of the
mobile counterions originally surrounding the isolated mac- macroions–lipid patches, they attract each other with an
roions upon their release to the bulk solution (see Section effective attraction energy of order nχ , which can lead to
1.5.4). The second is loss of 2D translational (“demixing”) 2D phase separation of the dressed membrane into mac-
entropy of the oppositely charged mobile “counterlipids” roion-rich and macroion-poor phases, as illustrated in the
upon their migration and segregation near the macroion- bottom panel of Figure 1.9.
binding site in order to balance its charge, thus enhanc- Interestingly, under certain conditions, the adsorption of
ing its membrane binding. In the abovementioned case of macroions onto oppositely charged membranes can result in
MARCKS adsorption, the neutralization of its basic ED is charge inversion of the membrane. Namely the net surface
primarily due to preferential sequestration of the polyvalent charge of the dressed membrane is of opposite sign to that
PI-4.5-P2, whose clustering involves a significantly smaller of the bare membrane. This charge inversion phenomenon
A Short History of Membrane Physics 15

(a) counterlipids; especially so in the case of fluid membranes


where the counterlipids can diffuse and segregate in the
protein adsorption zone to provide efficient charge match-
ing, as in Figure 1.9. The mobile counterions can now be
released into the bulk solution, providing a major, in some
cases the principal, contribution to the electrostatic free
energy of macroion–membrane binding. The magnitude of
(b)
the entropy associated with the release of the mobile coun-
terions diminishes upon increasing the salt concentration
in solution, consistent with the well-known fact that pro-
tein–membrane binding weakens at elevated levels of salt
in the aqueous solution. The counterion release mechanism
has been demonstrated in protein–DNA binding [139] and
various other systems [140]. A rather dramatic example of
FIGURE 1.9 Adsorbed macroions attract counterlipids to the
this mechanism is the formation of “lipoplexes” – those
interaction zone (a). On nonideal lipid mixtures, this can induce spontaneously formed complexes of cationic lipid mem-
lateral phase separation of the macroion dressed membrane (b). branes and DNA [141–143] – as illustrated schematically
in Figure 1.10a. Conductivity measurements and Poisson–
is commonplace in polyelectrolyte adsorption onto sur- Boltzmann theory calculations suggest that in cases where
faces, as well as in several cases involving biological mole- the cationic membrane charge density matches the charge
cules, e.g., the charge inversion of DNA saturated by bound density of DNA, the entropic contribution to the binding
polyvalent cations [137]. Analogously, charge inversion is free energy may be as high as ~95%, corresponding to the
expected when globular basic proteins bind to acidic lipid release of practically all the initially immobile counterions
layers [138] or when DNA adsorbs onto a lipid membrane (Figure 1.10b [144]).
containing cationic lipids. Charge inversion may also take
place upon the adsorption of intrinsically disordered pro- 1.6 MEMBRANE DYNAMICS: LIPID LATERAL
teins onto lipid membranes.
DIFFUSION AND ITS IMPACT
In a pioneering study on membrane dynamics published in
1.5.4 Counterion Release
1970, Hermann Träuble [146] showed that water permeabil-
Charged proteins, charged membranes, DNA, and other ity of membrane is driven by the diffusion of gauche-trans-
macroions are surrounded by clouds of mobile, gener- gauche (gtg) kink chain defects along the hydrocarbon
ally monovalent, counterions whose confinement to these fatty acid chains (see Figure 1.11a). In the same year, Frye
clouds involves an unfavourable loss of translational and Edidin reported the first experimental evidence of
entropy. When a charged protein approaches a membrane lateral mobility of proteins in cell envelopes [147]. Soon
containing oppositely charge lipids, the mobile counter- afterwards, by analysing the ESR line narrowing due to
ions originally surrounding the macroions are no longer spin–spin exchange of spin-labelled lipids, Träuble and
needed because charge neutralization can be provided by Sackmann provided the first direct data about the lateral

(a) (b)

FIGURE 1.10 (a) Schematic illustration of the counterion release mechanism in the formation of DNA–cationic–lipid complexes.
The entropy gained by the released mobile counterions provides a major contribution to the complexation free energy. The topology of
the lipoplexes formed depends on the spontaneous curvature of the lipids. They are lamellar when the spontaneous curvature is small,
and inverse-hexagonal when the lipid bilayers used contain negative-curvature loving (“helper”) lipids like PE [145]. (b) The fraction
of counterions released to solution in the formation of lamellar lipoplexes, reaching a maximum when the charge density on the DNA
and the lipid membranes are equal in magnitude [144].
16 Handbook of Lipid Membranes

diffusion of lipids in membranes, finding Dlat ≈1–10 −12 m2 s−1 the friction coefficient between juxtaposed monolayers
for the lateral diffusion coefficient [148]. Around the same of supported membranes or between integral proteins in
time, evidence for the high lateral lipid mobility has also supported membrane and the solid surface. In the case of
been demonstrated by NMR spectroscopy [149]. Later large frictional coupling of an integral protein (of radius
on, short-range diffusion coefficients (over ~100 nm) were R) with the surface, the diffusion coefficient becomes
derived by analysing the kinetics of excimer formation Dlat = k B T πbc R 2 , where bc = ηw H ; ηw denotes the fric-
and subsequent fluorescence following collisions between tional coefficient of the fluid layer, and H is the distance
pyrene-labelled lipids [150]. The application of this method between the protein and the surface [157,158]. Recent prog-
to lipid cholesterol mixtures led to the development of the ress was made by Komura et al. who studied the diffusion
free area model of molecular diffusion in membranes. This of proteins in membranes coupled to viscoelastic media
model accounts for the fact that (unlike in solids) molecular [159]. Their model opens new possibilities to explore the
diffusion in liquids is not an activated process but rather impediment of membrane protein diffusion by frictional
driven by density fluctuations in the liquid. Nevertheless, coupling of the extrinsic domains to the actin cytoskeleton
the diffusivity in the membrane obeys an Arrhenius-like or the glycocalyx.
law, where the temperature dependence of Dlat is dictated The most versatile technique for measuring molecular
by the thermal expansivity of the fluid [150]. The quantita- motions in model membranes is quasi-elastic neutron scat-
tive analysis of measurements of diffusion coefficients in tering (QENS). Owing to spin–spin exchange between spins
phospholipid cholesterol mixtures by this model provided of equal quantum number (S = ±1/2), the neutron scattering
one of the first experimental proofs of the condensing effect function of protonated material is incoherent (the scatter-
of cholesterol. The model was extended by Almeida and ing length density becomes negative). This unique feature
Vaz [151] and Saxton [152] and applied to explore the mem- allows us to analyse the motion of single lipid molecules
brane heterogeneity by Monte Carlo studies. (or portions thereof) by embedding fully (or partially) pro-
The next technical advancement arrived with the devel- tonated lipid molecules in a bilayer of fully deuterated lip-
opment of single-molecule tracking techniques, which were ids. The incoherent scattering provides information on the
developed following the invention of high-sensitivity CCD single proton self-correlation function:
cameras, and enabled measuring the time dependence of
   

the mean square displacement of single lipids or proteins,  
2
G inc ( r − r ′ , t ) = δ (r ′ − Ri (0) δ (r − Ri (t ) − n 2
x (t ) − x (t + τ ) = A τ ß. Deviations from the random walk (1.4)
i

model (β = 1) yielded new insights into the heterogeneous


organization of cell membranes [153]. Additional new and By studying oriented lipid multilayers, one can determine
attractive perspectives opened up following the develop- the direction of lipid motion with respect to the membrane
 
ment of the FCS technique, pioneered by the group of Webb normal. If the motion is unlimited, G inc ( r − r ′ , t ) decays
(reviewed in Ref. [154]). By analysing the temporal correla- to zero for t → ∞. In contrast, the self-correlation function
tion function of fluorescence signals, FCS enables to dis- reaches a finite value G∞ if the motion is restricted. G∞ is
tinguish between lateral motional processes and chemical called the elastic incoherent scattering factor (EISF). EISF
reactions in nanometre-size areas. Among the early appli- measurements yield quantitative information on the lateral
cations of this method is the abovementioned measurement extent of the motion of molecules or of molecular segments.
of the on-off kinetics of the binding of the MARCKS pro- Application of different QENS instruments enables study-
tein to charged vesicles by electrohydrophobic forces [155]. ing molecular motions in the time range of 10 −7–10 −9 sec.
Lateral diffusion became most prominent among physi- By combining QENS with NMR, the dynamic range can be
cists following the seminal work of Saffman and Delbrück extended to 1 Hz.
who showed that Dlat depends only logarithmically on the Several important new insights about membrane
radius of integral proteins [156]. This law implies that large ­dynamics that were provided by QENS are depicted in
objects diffuse nearly as fast as lipids, rendering diffusion Figure 1.11. These are as follows:
limited enzyme reaction in membranes very efficient. This (i) The lipid lateral diffusion involves fast local
is another beautiful example demonstrating how smart motions (Dloc ≈ 10 −10 m2 s−1) and long-range lateral motion
nature is in using physical principles to advance biological (Dlat ≈ 10 −12 m2 s−1), in agreement with the free area model of
processes. lateral diffusion. (ii) In the course of the L β →L α ­transition,
The Saffmann–Delbrück model was later extended by the number and mobility of chain defects increase
Evans and Sackmann to more realistic situations where ­continuously, whereas only long-wavelength motions with
integral proteins interact weakly with their environment, Q < 1.9 A−1 (or Δ ≥ 3 A) show a discontinuity at Tm. (iii) The
e.g., through the frictional coupling of membrane proteins dynamic range of QENS (10 −13< ν <10 −8) can be extended to
to the actin cytoskeleton or the diffusion of proteins in the millisecond time range by the combination of QENS and
supported membrane [157]. Using Einstein’s relationship NMR relaxation experiments, providing new insights into
between the diffusion (D) and the frictional (λ ) coeffi- the local motions of the water molecules bound to lipid head
cients, Dlat = k BT λ , the model proved useful in measuring groups [161]. (iv) Measurements of out-of-plane motions
A Short History of Membrane Physics 17

membranes have been reviewed in Ref. [163]. Below, we


briefly mention some pertinent examples.
One example is that of the line defects separating fluid
and gel-like domains. These defects provide pathways for
rapid lateral diffusion of membrane solutes (analogous to
“short-circuit” diffusion in solid-state physics [163–165]).
Another example is that of the chain packing defects
around conically shaped integral proteins such as the volt-
age-dependent Na+ channels [166]. These defects provide
“cavities” – attracting small solutes such as local anaesthet-
ics which can fill the voids and impede the conductivity (see
Figure 1.12). This function of defects as attractive centres is
reminiscent of the Cottrell clouds of solutes formed around
dislocations in solids which play a key role in the condition-
ing of material properties of metal alloys.
Density fluctuations in the lipid matrix (δρ ) may play
a key role in enzyme activity and the conductivity of ion
­channels [166] which are particularly pronounced in liq-
uid–solid domain boundaries, or around SPHM-rich
FIGURE 1.11 Top left: model of gtg-kink diffusion.
domains (Figure 1.6). The lateral density fluctuations may
Simultaneous hindered rotations about two C–C bonds (separated induce correlated changes in the cross-sectional area (∆A)
by one C–C bond) enable the kink performing diffusional jumps of the embedded proteins. In the case of ion channels, for
of length ΔL ≈0. 13 nm. The jump frequency of νj ≈ 10 −11 s−1 cor- instance, the changes in Na+ conductivity were shown to
responds to kink diffusivity of D kink ≈ 10 −5 cm2 s−1. Top right: snap- obey an Arrhenius-type behaviour, k = k0 exp {− K∆A /k BT },
shot of membrane as observed by MD calculations. The clusters where K is the membrane compression modulus and ∆A is
of yellow and green beads indicate molecules of the anaesthetics the area difference of the hydrophobic domain of the ion
halothane, demonstrating the role of the hydrocarbon chain region channel between the nonconducting and the conducting
as an apolar organic solvent of small hydrophobic solutes (image
state. The area compression modulus is inversely propor-
reproduced from Ref. [160] with permission of the authors).
Bottom: Summary of membrane dynamics from the nanosecond
tional to the density ­fluctuations, δρ 2 ρ 2 = K −1 ρ k B T ,
to the ten second time regime as revealed by QENS and NMR or which increases (hence K decreases) drastically around the
ESR relaxation experiments. ­SPHM-rich domains and diverges at the boundaries of
­coexisting liquid–solid domains. In the case of sodium chan-
nels of the heart muscle, ∆A ≈ 0.04 nm 2. For densely packed
of single lipids support the m
­ echanism of ­short-range lipid bilayers, K ≈ 0.8 Nm −1 and hence K∆A /k BT ≈ 8.
protrusion fluctuations suggested by Israelachvili and ­Defect-related local density fluctuations can thus drastically
Wennerström [65,162]. lower K , resulting in significantly higher ion conductivities.

1.7 MEMBRANE DEFECTS: PHYSICS


AND BIOLOGICAL FUNCTION (a) (b)
Ion (c)
The role of chain defects in fluid liquid crystalline bilay- channel
ers has been an ongoing theme since the early 1970s. In
natural membranes, defects can come into play in vari-
_ _ _ _ _

Exchange
_ _ _ _ _

ous ways. The transport of water across membranes and protein


the diffusion of small hydrophobic molecules (such as
local anaesthetics) within membrane by kink diffusion are Solute
among the examples. Chain packing defects are likely to cloud
form in highly curved bilayers and at the rim of SPHM-rich
microdomains (see Figure 1.6). The detailed analysis of the FIGURE 1.12 (a) Defect formation by local splay deformation
topology of defect structures of vesicles by freeze-fracture of the lipid bilayer adjacent to conically shaped integral protein,
such as Na+ channels. The stress caused by the deformation can be
EM showed for the first time that tilted phases, such as the
relaxed when small lipophilic molecules such as local anaesthet-
Pβ ′ and the Lβ ′ phases, or the nontilted L c -phase, form in ics migrate towards the defect, filling up the void, thereby pos-
single bilayer vesicles. These studies provided a direct link sibly impeding the ion conductivity. (b) Defects formed at lipid
between liquid crystal physics and membrane physics and halos around integral proteins (top) or the rims of microdomains
played a key role for the recognition of membrane research (bottom) enhance the exchange of lipids between outer leaflets of
by physicists. These early studies of defect formation in membranes.
18 Handbook of Lipid Membranes

Another physiologically important role of membrane of scale-dependent elastic properties of solid-like (­tethered)
defects is illustrated in Figure 1.12. Studies on model membranes due to thermal excitations of long-wavelength
­membranes indicate that lateral membrane ­inhomogeneities, in-plane phonons and bending deformations [172]. It
such as those associated with asymmetrical p­ rotein inclu- stimulated the development of numerous new techniques,
sions or the boundaries between different lipid domains, can enabling the measurement of the bending, shear, and com-
largely accelerate the exchange of specific ­lipids between pression modulus elastic moduli and the Young modulus
outer leaflets of intracellular organelles [163]. Pronounced (E) of cell envelopes. Great enthusiasm was raised by the
defect-enhanced activities of this kind were reported for discovery of undulation forces [173] and their role as regu-
phospholipases, which appear to be most active at fluid– lators of cell adhesion, as discussed in Section 1.9.
solid phase boundaries. Fluorescence microscopy stud- The Helfrich theory of membrane elasticity is a special
ies showed that the enzyme is most active at the interface case of the general Föppl-von Karman theory of shells. It
between solid and fluid domains (see [29,167]). was stimulated by Frank’s theory of liquid crystal elastic-
Yet another physiologically important effect of chain ity and led to relating membrane curvature deformations
packing defects is the increase in the passive transmem- to the splay deformation of smectic liquid crystals. The
brane permeation of molecules and ions, which appears to membrane bending energy ∆Gel was expressed in terms of
increase with the square of the density fluctuations, δρ 2 . the principal curvatures, 1 / R1 and 1 / R2 , of the bilayer’s
Recently, Heimburg et al. suggested that the propagation of neutral plane, with the inside–out asymmetry of the bilayer
defects along the axon could control action potential propa- accounted for by its mean spontaneous curvature C0 ,
gation [168].
1
Lipid packing defects associated with membrane regions
of high curvature can strongly affect enzyme activity in
∆Gel = κ
   
2 ∫∫ dO(2H − C )
 0
2

∫∫ dOH
 G (1.5)

such regions as compared with planar membranes. The


role of curvature in controlling enzymatic activity was Here, H = (1/R1 + 1/R2 )/2 and HG = 1/R1 R2 are the mean and
first demonstrated by Paul Janmey and his coworkers who Gaussian curvatures, respectively. κ and κ correspond to
studied the generation of PI-3,4,5-P3 lipids by phosphoino- the splay and saddle-splay elastic moduli introduced in liq-
sitol-3-kinase (PI-3K), finding that vesicles with an average uid crystal physics [174]. The integration extends over the
diameter of 100 nm are phosphorylated 100 times faster membrane surface. Based on this equation, vesicle shapes
than vesicles with an average diameter greater than 300 nm and phase diagrams have been derived using two generic
[163]. New interest in curvature-related membrane defects parameters: the spontaneous curvature, C0 , and the ratio
was boosted recently by studies concerning the binding of A /V between the vesicle area to its volume, as discussed in
BAR proteins to the necks connecting coated pits to the detail by Seifert and Lipowsky [175].
cell envelope. The neck formation is known to be con- One major success of the Helfrich homogeneous shell
trolled by BAR proteins (such as endophilins [170]). Recent model was the prediction of some shape changes of eryth-
experimental studies provided evidence that the curvature- rocytes, such as the discocyte to stomatocyte transition (see
induced packing defects resulting from the high curvature Figure 1.13a). Direct experimental evidence for the applica-
of the necks may account for the curvature sensing capacity tion of the model to red blood cells was provided by the
of endophilin. The low lipid packing density in the defects observation that this transition can be mimicked by single
enhances the binding of BAR proteins to the necks by facil- lipid component vesicles through temperature variations of
itating the penetration of their amphipathic alpha helices
[171]. Finally, we note that the role of defects as regulators
of the dynamics of membrane processes is reminiscent of (a) (b) (c)
the role of defects for the plastic deformation of metals, sug-
gesting analogies between the strategy of metallurgists to
control the microstructure of metals and of nature to control
the microanatomy of membranes.

1.8 BIOLOGICAL MEMBRANES
AS ELASTIC SHELLS
1.8.1 Morphology and Shape Transitions
of Soft Elastic Shells FIGURE 1.13 Experimental evidence for the simple Helfrich
model of erythrocyte shape changes. (a) Discocyte-to-stomatocyte
The bending elasticity model of cell shape changes p­ roposed transition of red blood cells. (b) Discocyte-to-stomatocyte shape
by Helfrich [7] stimulated the interest of physicists and transition of vesicles of DMPC triggered by increasing the area-
engineers working at the interface between physics and to-volume ratio A/V, achieved by increasing the temperature from
biology, resulting in the development of new concepts of 41°C to 42°C. (c) Demonstration of the generation of toroidal ves-
soft shell elasticity. One example is the nonclassical theory icles (image reproduced from Ref. [29]).
A Short History of Membrane Physics 19

the area-to-volume ratio (see Ref. [176] and Figure 1.13b). • The prolate and oblate shapes are stable over a
Another success of the Helfrich model was the interpreta- large range of excess areas, which could explain
tion of conformational transitions of toroidal vesicles (origi- the robustness of the discocyte shape of erythro-
nally discovered by Bensimon and coworkers [177], see also cytes against changes in osmolarity.
Ref. [178]) resulting from the interplay between the mean • The phase boundaries between two regimes of
(H) and Gaussian curvatures (HG) [179] (see Figure 1.13c). vesicle shapes may be separated by a spinodal
It was shown that shapes of toroidal topology may involve line (SP). Shape transitions, such as budding, may
one or several holes and that for a given number of holes therefore be drastically delayed, a fact that should
(called the genus of toroidal shapes), transitions between be considered in experimental studies of vesicle
different shapes can occur without changing the bending shape changes.
energy [175,180]. A third success was the interpretation of
the tension-induced pearling instability of membrane tubes
in terms of the Rayleigh instability [180]. 1.8.2 Shape Transitions of Stratified Soft
The homogeneous shell model could not explain shape Shells: The Red Blood Cell
changes of vesicles formed from long-chain lipids, which The compelling beauty of the Canham–Helfrich–Evans–
showed mainly spontaneous budding and formation of Svetina–Zeks model delayed the interest of physicists in
branched tubules but no discocyte to stomatocyte transi- ­biologically important aspects of cell membranes struc-
tion. Svetina and Zeks showed that this shortcoming of turing, such as the control of cell shapes by the intimate
the simple Helfrich model is remedied upon accounting coupling of the lipid–protein bilayer to the membrane-asso-
for the coupling between the two monolayers of the shells ciated cytoskeleton or the calculation of asymmetric cell
[181]. In fact, the important role of the coupling between shapes [184]. To overcome these shortcomings, the homo-
the ­monolayers had already been recognized by Evans in geneous shell model was extended in two directions:
1972 [182] but was ignored by the membrane community Asymmetric cell shapes could be mimicked by consid-
for several years. The theory of Evans accounted for the ering the coupling between membrane curvature elasticity
asymmetry of the membrane by assuming that the shape and in-plane phase separation. This issue has been studied
transitions of vesicles are determined by the initial differ- by Andelman and Leibler [112], who combined the elastic
ence ΔA0 of the areas occupied by the inner (Ai) and outer energy functional ∆Gela of the bending elasticity model with
(Ao) monolayers. the free energy expression of the classical Cahn–Hilliard–
The coupling between the opposing monolayers has Langer theory (see also Ref. [29] for a more extensive dis-
important consequences. If vesicles are prepared by swell- cussion). A few years later, Godzs and Gompper showed by
ing of bilayers in water, the area per lipid is roughly the MC simulations that the disk-like cisternae of the ER could
same in the inner and outer monolayers. Therefore, the be formed by mixtures of lipid components, one preferring
inner monolayer has to be compressed to form a closed positive curvature and the other preferring negative curva-
shell. Since the exchange of lipids between the two mono- ture [185].
layers is very slow (of the order of 12 hours for long-chain Shortcomings of the Helfrich model for the calculation
lipids such as DPPC or DOPC), freshly formed vesicles of nonsymmetric cell shapes were clearly pointed out in the
are initially not in mechanical equilibrium. Consequently, PhD work of Stokke [186]. He argued that nonsymmetric
­vesicles of ­long-chain lipids exhibit only inside budding red cell shapes, such as echinocytes and elliptocytes (in
upon increasing the area-to-volume ratio [183]. Figure 1.14), can be formed by the coupling of bending and
In 1997, a generalized elastic model accounting for the shear deformation. With the shear elastic modulus, a new
prestress generated by the tight coupling of the ­monolayers length scale Λ = κ µ is introduced. For typical values of
and the hindered flip flop was developed. The concept of
spontaneous curvature was extended by introducing an ( )
the bending modulus κ = 5 × 10 −19 J and the shear modu-
effective spontaneous curvature consisting of two contri- ( )
lus µ = 7 × 10 −6 J m −2 , one finds Λ ≈ 300 nm.
butions, namely Co (called local spontaneous curvature) A rigorous elasticity theory of cell envelopes of gen-
and a term accounting for the intrinsic area difference ΔA0/ eral shapes, based on principles of differential geometry,
A0. The spontaneous curvature model and the area differ- has been first developed by Mark Peterson [187,188]. The
ence model were unified into the area difference elasticity theory allowed for the first time to calculate changes of the
(ADE) model, which interpolates between the two models. contour of cells induced by external forces, such as those
The ADE model has been extensively reviewed elsewhere applied by high-frequency electric fields [189] or by shear
[175]. Below we point out some major predictions of the deformation fields generating tank treading motions [190].
models. A comprehensive elastic theory of erythrocyte shapes
was developed 30 years after the pioneering Canham–
• The spontaneous curvature and the ADE models Helfrich by Wortis and collaborators [191]. These authors
predict about the same shape transitions but differ calculated cell shapes by minimizing the total energy func-
concerning the prediction of the order of the tran- tional comprising (i) the bending elasticity expression of
sitions and the existence of metastable phase lines. the ADE model ∆GADE and (ii) the sum of the extensional
20 Handbook of Lipid Membranes

FIGURE 1.14 Variation of surface pressure of the lipid–protein bilayer of red blood cell in the region of the cell surface forming a
spicule. The network is stretched at the tip of the spicule and pulls at the membrane towards the inside. It is compressed at the base and
exerts a pressure directed towards the outside. Note that the deformation of the cytoskeleton is associated with the redistribution of the
membrane proteins, such as band III.

deformation and shear deformation ∆Gext of the cytoskel- Many functions of membrane undulation were discovered
eton, so that ∆Gtot = ∆GADE + ∆Gcyt . after Helfrich’s publication. It became evident that the dis-
The elastic energy of the cytoskeleton is expressed in terms joining pressure plays a key role for the swelling of lipid
of the rubber elasticity model of Mooney (first introduced by multilayers and that it controls the adhesion of vesicles
Skalak and Evans [192]) in the field of membrane physics, and cells, compensating the Van der Waals attraction of
using λ1 and λ2 to denote the principal stretching ratios. lipid vesicles at distances < h >≥ 20 − 30 nm . The tension-
induced freezing-in of the undulation was shown to trigger
K µ  λ1 λ2  cell adhesion [195,196]. This effect was applied to measure


∆Gcyt =
2 ∫∫ do(1 − λ λ )
 1 2
2
+
2 ∫∫ dO  λ
 2
+
λ1
− 2
 the membrane bending modulus κ using the micropipette
(1.6) technique [197].
Further theoretical basis of the entropy-driven interfa-
In conclusion, the Wortis model predicts that the shape cial forces was laid about 8 years later by Lipowsky and
changes of erythrocytes during their passage through blood Leibler [198]. To account for the confinement of membrane
vessels require the ongoing reorganization of the ­spectrin– fluctuations, they extended Helfrich’s model by adding a
actin network. Concomitantly, the membrane proteins harmonic interaction potential V (h) = V0 + V ''(h − h0 )2 / 2
Band III and glycophorin are expected to be constantly that restricts these fluctuations. The total free energy func-
­redistributed during the cell transport through the blood tional can then be expressed as
vessels, which is an ATP consuming process.
κ σ 1 ∂2 V 2
∫∫
 dO  ( ∆u ) + ∇u 2 + 2 {
h − ho } 
2
∆Gadh =
1.8.3 Membrane Bending Excitations and   2 2 2 ∂ x 
Their Physiological Significance (1.8)

The interest of many soft matter physicists in biomaterials This yields two observable length scales: the capillary
was stimulated by the studies of cell membrane fluctuations length λc and the persistence length ζ p given by
by Brochard and Lennon [193] and Helfrich [173]. In the
former study, it was shown that the long-known flickering
of erythrocytes was activated by bending excitations of the λc = κ / σ and ζ p = 4 κ / V '' (1.9)
soft cell envelopes. Helfrich’s work led to the beautiful pic-
ture of a membrane as a dynamically rough surface, which Here, λc is a measure of the maximum wavelength over
can be regarded as being composed of cushions of dimen- which the membrane excitations are dominated by the
sion ζ p × ζ p performing independent Brownian motions in bending excitations, and ζ p accounts for the lateral exten-
the normal direction to the membrane [194]. Each collision sion of the membrane deformation induced by a local point
of random local protrusions with adjacent surfaces was force.
assumed to involve an energy of the order of kBT, in analogy Experimental tests of the theories of membrane bending
to molecules of an ideal gas hitting a solid wall. For ten- excitations became possible following the development of
sion-free membranes at distance h from the wall, the repul- the Fourier spectroscopy of bending excitations of adhering
sive membrane pressure (which is equal to the transmitted giant vesicles and erythrocytes by reflection interference
energy per unit area) was shown to be given by [173,194] contrast microscopy (RICM) combined with fast image
processing [199]. The spatial correlation function of the
pdisj ≈ k B T / ζ p2 h ∝ ( k B T ) / κ h 3 (1.7) excitation amplitude u ( x , t ), i.e.,
2
A Short History of Membrane Physics 21

    u( x )u( x + ξ ) = ζ ⊥ exp −( x − x )2 / ζ p2 (1.10){ } developed by Safran and coworkers [203] allowing to
evaluate the wavelength dependence of the mean square
and hence, ζ p could then be directly measured. By simul- fluctuation amplitude in the membrane wavelength range
taneous measurement of the bending modulus ζ p , the Λ ≈ 0.5 − 2.5µm measured by Zilker’s Fourier spectros-
force constant V '' of the interfacial potential could also be copy [199]. Korenstein and coworkers later provided direct
determined. evidence that the enhanced dynamic surface roughness is
In 1987, Millner and Safran published a rigorous the- associated with ATP turnover [204,205], which provides
ory of shape fluctuations of vesicles by accounting for the some evidence for the model [201] of the enhancement
boundary condition of constant volume in terms of the of bending excitations by tension fluctuations proposed in
membrane tension γ [200]. Their model paved the way for Figure 1.16.
high-precision measurement of membrane bending moduli It has been suggested that the overexcitation of the eryth-
by the Fourier analysis of the contour of giant vesicles [183] rocyte shell by fluctuating chemical forces can be described
with 0.5 µm wavelength resolution. The high-precision in terms of an effective temperature Teff . Teff − T is a mea-
method allowed to determine the number of shells of giant sure of the excess undulation amplitude excited by sto-
vesicles or the modification of κ by small concentrations of chastic chemical forces. It can be generally determined by
solutes and cholesterol. simultaneously measuring the bending modulus by flicker
analysis and other micromechanical techniques. Another
method is based on the measurement of excess membrane
1.8.4 Red Blood Cell Membranes as Actively tension by the micropipette technique [197]. This approach
Driven Semiflexible Statistical Surfaces has been adopted by Bassereau and coworkers to demon-
strate the enhancement of bending membrane excitation
A surprising finding of the high-precision RICM mea-
by active chemical noise generated by ion translocation
surements of bending elasticity of erythrocyte mem-
through membranes. Studying giant vesicles doped with
branes was their astonishingly small bending modulus:
reconstituted bacteriorhodopsin, it was shown that light-
κ ≈ 7 k B T as compared with the value of κ ≈ 50k B T
driven proton pumping activity amplifies the bending undu-
expected for the PM containing 50 mole% cholesterol
lation drastically [206]. The chemical noise generated by
[199,201]. Indeed, a parallel measurement of κ by the
proton influx can give rise to higher excess temperatures.
Fourier analysis of the cell contour derived by phase
The following discussion of the physiological role of
contrast microscopy yielded a value of 50k B T [202], sug-
membrane flickering shows that more detailed studies of
gesting that the thermal excitation of the bending undu-
active excitation of flickering are expected to provide valu-
lations is enforced by random chemical forces. These
able insights into dynamic control of membrane processes.
are expected to be caused by local fluctuations of lateral
tension by the constituent coupling and uncoupling of
the spectrin links from glycophorin, which is associated 1.8.5 The Physiological Role of
with ATP turnover (Figure 1.15). Membrane Flickering
A sophisticated and rigorous two-shell model of
In addition to erythrocytes, membrane bending undulations
the bending elasticity of stratified cell envelopes was
have been observed in various other cells, including endo-
thelial cells and macrophages. These cells show mainly
(a) Spe
short-wavelength excitations of Λ ≤ 1 µm with ~10 nm
ctrin
Ankyrin
Dangling
tetr
am
er
root mean square amplitudes [207]. This is remarkable,
Actin
bond
considering the fact that the bending and shear moduli of
Glycoophorin
Band III these composite shells are much larger (κ ≈ 1000 k B T and
µ ≈ 4 × 10 −4 Jm −2 ) than those of erythrocytes. A challeng-
(b) Kinase
activation ing question is therefore whether membrane bending exci-
Band IV.1 tations may serve some purposes. Several possible answers
Phosphatase
deactivation Actin are outlined below.
First and most importantly, the bending excitations pre-
FIGURE 1.15 (a) Schematic view of composite cell envelope of vent the nonspecific adhesion of erythrocytes and mac-
erythrocytes with the lipid–protein bilayer exhibiting thermally rophages to solid surfaces (see Ref. [207]) or the walls of
and actively driven bending excitations. The undulatory excita- the blood vessels. Second, erythrocytes do not possess
tions are limited by about 10% excess area of the bilayer with intracellular transport mechanisms, and the hydrodynamic
respect to the average area of the spectrin–actin network. They
flow induced by flickering in the cytoplasm (see Ref. [193])
determine the distance between the two subshells and generate an
excess membrane tension [194,203]. (b) The ongoing ATP-driven
could accelerate the exchange of haemoglobin between the
binding and unbinding of band IV.1 from glycophorin associ- rim of the cell and its centre. Third, the disjoining pressure
ated with the formation of dangling bonds enhances the bending between the lipid–protein bilayer and the actin cytoskeleton
amplitudes and could account for the nonthermal contributions to controls the distance between the two subshells of cell enve-
the undulations. lopes [203] and may inhibit the sticking of the actin cortex
22 Handbook of Lipid Membranes

(a) (b)

FIGURE 1.16 Experimental evidence for the homophilic nature of cadherin-mediated intercell adhesion. (a) In vitro self-organization
of a cell mixture into an embryo-like structure. Following the dissociation (by alkali) of a piece of embryonic (amphibian neurola) tis-
sue, the cells were allowed to reassemble. Initially, the cells reassemble to form a uniformly mixed globule. After a while, due to the
homophilic nature of the adhesion, segregation takes place, with neural cells (blue) constituting the inner core, mesodermal (green) in
the middle layer and epithelial cells (red) in the outer shell. (A coloured version of the figure adapted from the original landmark paper
of Towns and Holtfreter [218]). (b) Cadherin segregation during the early stages of embryo morphogenesis. N-cadherins are expressed
in the neural tube developing from the epithelial tissue, where cells are held together by trans binding of E-cadherins [214].

to the membrane. Fourth, the thermal excitations generate


attractive isotropic forces between neighbouring mem-
κ C0 =
∫ zπ(z)dz, ∫
κ = − z 2 π( z )d z (1.11)
brane–cytoskeleton linkers, which is balanced by the ten-
sion of the entropic springs of the spectrin tetramers. This
may serve the isotropic distribution of the spectrin–mem- Using the lateral pressure profile derived from Eq. (1.1),
brane linkers glycophorin and Band III and the dynamic these elastic moduli, as well as an explicit expression for κ ,
softening of the cell envelope, as suggested by the stiffening were derived, enabling to calculate the bending rigidities of
of elliptocytes. single- and multi-component membranes [209,210], show-
ing very good qualitative and quantitative agreement with
experiment. Over the years, additional molecular-level the-
1.8.6 Molecular Aspects of Membrane Elasticity ories and lattice models of membrane elasticity have been
In parallel to the continuum theories of membrane elastic- proposed, yet in recent years – in the spirit of time – most of
ity, cell shape changes, undulations forces, and the various the effort is invested on developing and applying large-scale
elegant experimental methods developed to measure elastic MD computer simulations.
membrane moduli, many molecular-level models and theo-
ries have been proposed to calculate those moduli. These 1.9 CELL ADHESION: A MEMBRANE-
models range from simple pictures to rather elaborate theo-
BASED PROCESS
ries employing realistic chain models and/or sophisticated
electrostatic analyses. A simple yet very useful criterion Among the first questions that motivated the interest of
for assessing the spontaneous curvature corresponding to physicists in cell adhesion was whether intermembrane
a given lipid molecule is provided by the “packing parame- adhesion can be understood in terms of the classical DLVO
ter” p = al /v, where a, v, and l are the optimal area per lipid theory of interfacial forces between charged surfaces.
head group, the volume of the tail, and its length, respec- Studies of this question by Israelachvili and coworkers using
tively [65]; large p favours positive curvature, whereas the surface force apparatus (SFA) [65] and by Parsegian
small p, as in the case with PE for instance, favours negative and coworkers using their osmotic stress technique [211]
curvature, as observed in the inverse hexagonal (HII) phase. resulted in the first measurements of the Hamaker con-
Phenomenological “compression models” have yielded stants that measure the strength of the interaction between
insightful scaling relationships relating the bending rigidity lipid bilayers and between membranes and solid surfaces
to the lipid chain length and area per head group (see, e.g., [65]. A second approach, pioneered by Bell and cowork-
[63,208,209] and references therein). More sophisticated ers [212,213], attempted to explain cell-to-cell adhesion in
theories have also been proposed, taking into account the terms of the competition between specific attractive forces
conformational (e.g., the rotational isomeric) statistics of involving (“lock and key”) adhesion receptors, and generic
the hydrocarbon lipid chains and/or the electrostatic and repulsion forces mediated by the glycoproteins forming the
excluded volume interactions between the head groups. glycocalyx. Analysing the adhesion-induced deformations
Any theory or computer simulation that yields information of adhering cell envelopes, they concluded that cells can
regarding the lateral pressure profile described in Figure 1.4 be regarded as liquid droplets whose adhesion strength is
can be used to calculate the elastic moduli using general determined by the Young–Laplace equation. This pioneer-
relationships, such as ing and fundamental study pointed out analogies between
A Short History of Membrane Physics 23

the adhesion of cells and partial wetting of solids by fluid far stronger than those between unlike cells. Consequently,
droplets. a mixture of cells expressing E- and N-cadherins strongly
Below, we briefly comment on two complementary prefers to “phase segregate”, resembling the behaviour of
points of view of membrane adhesion. (i) In Section 9.2, we a liquid mixture of immiscible molecules. This has been
outline the physicists’ approach, emphasizing the mechani- verified in various in vivo systems and in vitro assays,
cal aspects of cell adhesion and the interplay between mem- [214,216,217].
brane curvature elasticity, surface tension, and the role of Additional physical insights into the liquid droplet con-
“linker” and “repeller” molecules. (ii) In Section 9.1, we cept and more quantitative information on the cohesive
briefly comment on the cell biologists’ point of view where forces between cells were gained by Brochard and cowork-
much emphasis is given to the molecular details and mutual ers [219] by studying spherical cell assemblies by the micro-
interactions of the adhesive receptors. pipette aspiration technique. They showed that the cohesive
force between spherical cell clusters increases monotoni-
cally with the cadherin expression. Studies of the dynamics
1.9.1 Molecular Aspects
of spreading of cells on surfaces showed some interesting
Early biological studies were largely concerned with analogies with the spreading of viscoplastic fluid droplets.
embryogenesis and tissue development, with particu- While confirming the homophilicity of cell adhesion via
lar emphasis on cadherin-mediated cell–cell adhesion. selective (lock and key) forces among cell surface recep-
Takeichi [214], Steinberg [215,216], and others (see, e.g., tors, the adhesive energies associated with these forces
[217] and references therein) have demonstrated that cad- do not explain the magnitude of cell–cell or cell–surface
herin-mediated adhesion is strongly homophilic. Two early energies, as they do not account for the very important role
examples of this important aspect of cell adhesion are played by the coupling of the adhesive receptors to the cyto-
demonstrated in Figure 1.17. The classical experiment of skeleton of the composite membrane, nor for the important
Towns and Holtfreter was actually carried out before the effects due to membrane curvature fluctuations. In other
role of adhesive receptors such as cadherins, for instance, words, these assays provide little insight into the role of
has been realized. The homophilic nature of receptor-medi- the composite cell membrane or into the formation of focal
ated adhesion was cast in simple physicochemical terms by adhesion complexes generally formed at the cell–cell and
Steinberg and coworkers, an approach known as the dif- cell–tissue interface.
ferential adhesion hypothesis (DAH), [215,216]. The DAH
asserts that cell mixtures behave analogously to liquid mix-
1.9.2 Insights Gained by Biomimetic Systems
tures of barely miscible molecules. Thus, for example, cells
expressing epithelial (E) cadherins prefer adhering to cells Important theoretical steps towards understanding the
expressing E-cadherins, rather than to, say, cells express- physical basis underlying the control of cell adhesion by soft
ing neural (N) cadherins. Underlying this behaviour is elastic cell envelopes was made by Lipowsky and Seifert
the fact that although the interaction between E-cadherin [220], Bruinsma [221], and Evans [222]. These authors
and N-cadherin receptors is attractive, its strength showed that the adhesion strength of fluid membranes can
(wNE) is smaller than the average strength of the homophilic be quantified in terms of the spreading pressure W (the free
­attractions (i.e., wNE < ( wNN + wNE ) / 2). The differences energy of adhesion per unit area) given by the following
between the attractive energies of the different kinds of equations:
receptors are small (typically just a few kBT), yet since each
2
cell is covered typically by tens to hundreds of thousands κ 1
W = σ (1 − cosθ c ) , W = (1.12)
of molecules, the adhesive affinities between like cells are 2  Rc 

FIGURE 1.17 (a) Mechanical equilibrium of the interfacial tension and the bending moment at the contact line of the adhesion zone.
The surface tensions σ ij are measures of the interfacial energies. Rc is the radius of membrane curvature at the contact line (see also
[220]). (b) Decay of the cell substrate interface into domains of tight adhesion (formed by integrin clustering) and weakly adhering
zones, which are separated from the substrate by long-range forces mediated by glycoproteins of the glycocalyx and undulation forces.
The binding strength of the initially formed clusters of integrin is enforced first, by the drastic increase of the affinity of the CAMs
for ligands of the tissue (such as fibronectin) and secondly, by the increase of the membrane bending stiffness induced by coupling of
actin gel patches to the cytoplasmic domains of integrin (following Eq. (1.12)). The width of the transition zone between the two states
of adhesion is equal to the persistence length ζ p appearing in Eq. (1.7). (Images adopted from Ref. [178].)
24 Handbook of Lipid Membranes

In this equation, σ is the membrane tension, κ is the bend- key role for the onset of lymphocyte transgres-
ing modulus introduced in Eq. (1.5), θ c is the contact angle, sion through the endothelial cell layers (see Refs.
and R c is the radius of curvature of the shell at the contact [223,224]). In the case of cadherin-mediated cell–
line Lc (defined in Figure 1.17). The first equation on the cell adhesion, clusters of the linkers are stabilized
left side is the classical Young equation of capillary forces. by binding of their cytoplasmic tails to actin by
The second equation accounts for the elastic energy asso- β- and α-catenin. An important consequence
ciated with the bending of the membrane at the transition of this linkage is a drastic increase of the mem-
from the adhering to the free membrane area. The contact brane bending modulus and thus – according to
angle θ c and the contact curvature Rc can be measured with Eq. (1.12) – of the binding strength W.
high precision by using interferometric techniques, such as • A most surprising but important result is the
RICM [196,221]. following. The values of the adhesion ener-
Experimental physics of cell adhesion research was gies measured by contour analysis, namely
boosted by the development of planar solid-supported W = 10 −5 − 10 −6 J m −2 , are several orders of magni-
membranes or patterned polymer films as models of cell tude smaller than the energies estimated from the
and soft tissue surfaces (reviewed until 2005 in Ref. [17]). known density and binding energy of the linker
This design opened up the possibility of reconstructing the pairs, yielding W ≈ 10 −3 J m −2 [224]. It was shown
contour of adhering soft shells, such as vesicles with nano- that this reduction of W is a consequence of the
metre spatial resolution, by the RICM technique, enabling two-dimensional osmotic pressure exerted by the
high-precision measurements of the geometric parameters CAMs and repellers expelled from the adhesion
Rc and θc and hence of the free energies of adhesion (W in domain. A rigorous theory of the initial process of
Eq. (1.5)). adhesion domain formation by interplay of elastic
Simple model systems were designed to gain quanti- and entropic forces was developed by Seifert and
tative insight into the control of the state of adhesion by Smith [225].
interplay of short-range attraction forces mediated by
specific linker pairs, long-range repulsion forces medi- Taken together, the biomimetic studies show that the
ated by glycoproteins and undulation forces, and the repulsive force mediated by the glycoproteins of the cell
adhesion-induced membrane deformation. Giant vesicles surface plays a key role for the softening of the adhesion
doped with ligands of tissue (such as fibronectin or the strength and the rapid formation and dismantling of adhe-
antigenic oligosaccharide Sialyl Lewis X) served as test sion domains. It should be noted, however, that many of
cells. Supported membranes exposing a conjugate cell the glycoproteins (such as Syndcans and CD44) can act as
adhesion molecule (CAM, such as integrin and selectins) CAMs, which mediate the adhesion of stromal cells to mac-
mimicked the tissue or cell surface. To account for the romolecules of the extracellular matrix, such as collagen
repulsion forces mediated by the glycoproteins, lipids networks. Most CAMs expose cytoplasmic domains which
exposing hydrophilic macromolecular head groups were mediate their coupling to the actin cortex through specific
added to one of the membranes. Unbinding forces between linkers, such as talin and ezrin. The global distribution
the linker pairs were measured by magnetic tweezer force of the linkers can be controlled by the actin–microtubule
microscopy. The state of the art of model membrane stud- crosstalk, which plays a key role for the global polariza-
ies was reviewed recently [178]. We therefore summarize tion of cells. Several physiologically relevant examples of
below only few important lessons learned from these the control of cell adhesion by microtubule–actin crosstalk
model membrane studies: have been recently reviewed in Ref. [178].

• An inevitable consequence of the balance of


1.9.3 Synopsis
interfacial forces and elastic stresses summa-
rized above is the decay of the adhesion zone into The design of sophisticated model systems of cell adhesion,
microdomains of tight adhesion (formed by clus- together with the development of new experimental tech-
ters of bound pairs of linkers), which are separated niques and thermomechanical theories have provided new
by nonadhering zones. insights into the control of cell adhesion by intermolecular
• Cell adhesion is a two-step process. The first step forces and elastic stresses [178]. It is certainly a long way to
consists in the formation of clusters of linker pairs design model systems, which can mimic the active control
by the mechanism described above. In a second- of cell adhesion mediated by coupling of CAM–clusters to
ary step, these clusters are stabilized by coupling the actin cortex.
of actin gel patches to the cytoplasmic tails of the At present, a more promising strategy is to apply the
CAMs. In the case of integrin, this coupling is methods and concepts developed over the last 40 years to
mediated by the integrin–actin linkers talin and study the adhesion of living cells on biofunctionalized sur-
ezrin (illustrated in Figure 1.17), which results in faces. The bending moduli, the membrane tensions, and the
a drastic enhancement of the affinity of integrins adhesion strengths can be measured by applying lift forces
for ligands (such as ICAM-I). This step plays a and hydrodynamic shear forces [213,226]. Insights into the
A Short History of Membrane Physics 25

control of the adhesion strength mediated by coupling of capacity to adapt their physical properties to environmental
the actin cortex to the intracellular domains of the CAM– conditions.
clusters can be gained by mutations of the proteins regulat-
ing actin membrane coupling, such as talin (see Ref. [226]).
LIST OF ABBREVIATIONS
An intriguing example of the benefit of combined studies
of biomimetic and natural systems is mentioned in the next ADE model: area difference elasticity model
section. BLMs: black lipid membranes
The combined study of in vitro systems and natural cells DLVO theory: Derjaguin–Landau–Verwey–Overbeek
has shed new light on the long-standing question: how does theory of the stability of colloidal
myelin sheath form tightly packed multilayers of collapsed suspensions
cell lobes around axons? The first insights were provided EISF: elastic incoherent scattering factor
by Israelachvili and coworkers who measured the forces FCS: fluorescence correlation spectroscopy
between the inner leaflets of the PMs mediated by the FTIR: Fourier transform infrared spectroscopy
electrostatic interaction of the myelin basic protein [227]. MARCKS: myristoylated alanine-rich C-kinase
Recent experiments by Janshoff and coworkers on natu- substrate
ral systems solved another essential part of the enigma. PLC-Cγ: phospholipase-Cγ (an enzyme
They showed that membrane lobes protruding from oli- generating inositol 1,4,5-trisphosphate
godentrocytes (cells forming myelin sheets in the brain) and diacylglycerol (DAG) from
adhere strongly to axon surface after downregulation of PI-4,5-P2)
the genetic expression of both the repellent glycoproteins PI-3K: phosphoinositid-3-kinase (generator of
and the glycolipids [226]. The intermembrane distance of PI-3,4,5-P3)
2 nm is determined by the van der Waals attraction, which RICM: reflection interference contrast
is balanced by the repulsion mediated by uncharged inte- microscopy (also called interference
gral proteins. reflection microscopy [IRM]).
SFA: surface force apparatus
1.10 A SHORT OUTLOOK: WHERE WE
STAND AND WHERE TO GO LIPID ABBREVIATIONS AND
In the last 40 years, our understanding of the physical NOMENCLATURE
basis of the self-organization of biological membrane has The structure of each type of lipid is characterized by the
advanced dramatically, owing to the comparative study structure of the head groups and the number of C-atoms (n)
of model systems and biological membranes. This break- and double bonds (m) of the hydrocarbon chains.
through was made possible by the interactive development
of new physical techniques and novel theoretical concepts
and computational tools. There are still many unsolved TYPES OF LIPIDS
problems. Many of these are due to our difficulties to deal PC: phosphatidylcholine
with the thermomechanics of multicomponent systems PE: phosphatidylethanolamine
under nonequilibrium conditions. PS: phosphatidylserine
It appears that there are two directions to go. One strat- PI: phosphoinositides (including PI-4,5-P2 and
egy is to mimic the structure and function of real composite PI-3,4,5-P3, also abbreviated as PtdIns(4,5)P2
membranes by reconstitution of active actin and micro- and PtdIns (3,4,5)P3)
tubule networks into giant vesicles with reconstituted ion DAG: diacylglycerol
channels and CAMs. Such artificial cell organelles are also
of growing interest as smart drug delivery systems.
An easier strategy may be to study natural mem- EXAMPLES
branes and their structural and functional modifications PC: C16:0; C16:0 and PC: C16:0, C18;1 stand for dipalmi-
by specific mutations or signalling molecules. To this end toylphosphatidylcholin and palmitol-strearoyl phosphati-
physicists should realize that biological membranes are dylcholine, respectively.
composite shells and that their molecular architecture is
constantly remodelled by biochemical processes to adapt
ACKNOWLEDGEMENTS
the cell structure and function to the cell’s physiological
needs. E.S. gratefully acknowledges the financial support from the
Studies of natural membrane processes teach us how to Excellence Initiative of the Technical University Munich
regulate the structure and physical properties of complex and the Center for NanoScience (CeNS) at the Ludwig-
materials by local changes of the chemical composition in Maximilian-University (LMU) Munich. A.B.S. thanks the
a logistic way. A future benefit of such painstaking studies support of the Fritz Haber Research Center supported by
may be that we learn how to design smart materials with the the Minerva Foundation.
26 Handbook of Lipid Membranes

BIBLIOGRAPHY 20. Sackmann, E. 2014. Endoplasmatic reticulum shaping by


generic mechanisms and protein-induced spontaneous
1. Gorter, E., and F. Grendel. 1925. On bimolecular layers of ­curvature. Adv. Colloid Interface Sci. 208: 153–160.
lipoids on the chromocytes of the blood. J. Exp. Med. 41: 21. Sackmann, E., H. Träuble, H.J. Galla, and P. Overath.
439–43. 1973. Lateral diffusion, protein mobility, and phase transi-
2. Danielli, J.F., and H. Davson. 1935. A contribution to the tions in Escherichia coli membranes. A spin label study.
theory of permeability of thin films. J. Cell. Comp. Physiol. Biochemistry. 12: 5360–5369.
5: 495–508. 22. Seelig, J. 1977. Deuterium magnetic-resonance theory
3. Singer, S.J., and G.L. Nicolson. 1972. The fluid mosaic and applications to lipid membranes. Q. Rev. Biophys. 10:
model of the structure of cell membranes. Science 175(80): 353–418.
720–731. 23. Mely, B., J. Charvolin, and P. Keller. 1975. Disorder of lipid
4. Sackmann, E. 1983. Physical foundation of the molecular chains as a function of their lateral packing in lyotropic liq-
organization and dynamics of membranes in biophysics. In: uid crystals. Chem. Phys. Lipids. 15: 161–173.
Hoppe, W., Lohmann, W., Markl, H., Ziegler, H., editors. 24. Mantsch, H.H., and R.N. McElhaney. 1991. Phospholipid
Biophysics. Springer Verlag. Berlin, pp. 425–457. phase transitions in model and biological membranes as
5. Luzzati, V. 1968. X-ray diffraction studies of lipid-water studied by infrared spectroscopy. Chem. Phys. Lipids. 57:
systems. In: Chapman D., editor. Biological Membranes, 213–226.
Vol. 1. Academic Press, New York, pp. 71–123. 25. Esmann, M., and D. Marsh. 1985. Spin-label studies on
6. Luzzati, V., and a Tardieu. 1974. Lipid phases: Structure the origin of the specificity of lipid-protein interactions
and structural transitions. Annu. Rev. Phys. Chem. 25: in Na+,K+-ATPase membranes from Squalus acanthias.
79–94. Biochemistry. 24: 3572–3578.
7. Helfrich, W. 1973. Elastic properties of lipid bilayers: 26. König, B.W., S. Krueger, W.J. Orts, C.F. Majkrzak, N.F.
Theory and possible experiments. Z. Naturforsch. 28: Berk, J. V Silverton, and K. Gawrisch. 1996. Neutron
693–703. reflectivity and atomic force microscopy studies of a lipid
8. Bessis, M. 1973. Living Blood Cells and Their bilayer in water adsorbed to the surface of a silicon single
UltrastructueNo Title. Berlin-Heidelberg-New York: crystal. Langmuir. 12: 1343–1350.
Apringer Verlag. 27. König, S., W. Pfeiffer, T. Bayerl, D. Richter, and E.
9. Shen, B.W., R. Josephs, and T.L. Steck. 1986. Ultrastructure Sackmann. 1992. Molecular dynamics of lipid bilayers
of the intact skeleton of the human erythrocyte membrane. studied by incoherent quasi-elastic neutron scattering. J.
J. Cell Biol. 102: 997–1006. Phys. II. 2: 1589–1615.
10. Legendre, K., S. Safieddine, P. Küssel-Andermann, C. Petit, 28. Buldt, G., H.U. Gally, A. Seelig, and J. Seelig. 1978. Neutron
and A. El-Amraoui. 2008. alphaII-betaV spectrin bridges diffraction studies on selectively deuterated phospholipid
the plasma membrane and cortical lattice in the lateral wall bilayers. Nature. 271: 182–184.
of the auditory outer hair cells. J. Cell Sci. 121: 3347–3356. 29. Sackmann, E. 1995. Physical basis of self-organization
11. Hladky, S.B., and D. a Haydon. 1972. Ion transfer across and function of membranes: Physics of vesicles. In: R.
lipid membranes in the presence of gramicidin A. I. Studies Lipowsky, E. Sackmann, editors. Structure and Dynamics
of the unit conductance channel. Biochim. Biophys. Acta. of Membranes. Amsterdam: Elsevier. pp. 213–304.
274: 294–312. 30. Overath, P., and H. Trauble. 1970. Phase transitions in cells,
12. Bamberg, E., and P. Läuger. 1973. Channel formation kinet- membranes, and lipids of Escherichia coli by fluorescent
ics of gramicidin A in lipid bilayer membranes. J. Membr probes, light scattering and dilatometry. Biochemistry. 12:
Biol. 11: 177–194. 2625–2634.
13. Kolb, H.-A., and G. Boheim. 1978. Analysis of the multi- 31. McIntosh, T.J., and S.A. Simon. 1986. Area per mol-
pore system of alamethicin in a lipid membrane. J. Membr. ecule and distribution of water in fully hydrated dilauro-
Biol. 38: 151–191. ylphosphatidylethanolamine bilayers. Biochemistry. 25:
14. Conti, F., and Wanke, E. 1975. Channel noise in nerve 4948–4952.
membranes and lipid bilayers. Q. Rev. Biophys. 8: 451–506. 32. Wilkinson, D.a, and J.F. Nagle. 1981. Dilatometry and calo-
15. Chernomordik, L., A. Chanturiya, J. Green, and J. rimetry of saturated phosphatidylethanolamine dispersions.
Zimmerberg. 1995. The hemifusion intermediate and its Biochemistry. 20: 187–92.
conversion to complete fusion: Regualtion by membrane 33. Privalov, G., V. Kavina, E. Freire, and P.L. Privalov. 1995.
composition. Biophys. J. 69: 922–929. Precise scanning calorimeter for studying thermal proper-
16. Li, F., F. Pincet, E. Perez, C.G. Giraudo, D. Tareste, and ties of biological macromolecules in dilute solution. Anal.
J.E. Rothman. 2011. Complexin activates and clamps Biochem. 232: 79–85.
SNAREpins by a common mechanism involving an 34. Shimshick, E.J., and H.M. McConnell. 1973. Lateral phase
intermediate energetic state. Nat. Struct. Mol. Biol. 18: separation in phospholipid membranes. Biochemistry. 12:
941–946. 2351–2360.
17. Tanaka, M., and E. Sackmann. 2005. Polymer-supported 35. Mouritsen, O.G. 1991. Theoretical models of phospholipid
membranes as models of the cell surface. Nature. 437: phase transitions. Chem. Phys. Lipids. 57: 179–194.
656–663. 36. Mouritsen, O.G. 2005. Life–as a Matter of Fat: The
18. Seddon, J.M., and R.H. Templer. 1995. Polymorphism of Emerging Science of Lipidomics. Heidelberg: Springer.
lipid-water systems. In: Lipowsky R, E Sackmann, edi- 37. Silvius, D.R. Thermotropic Phase Transitions of Pure
tors. Structure and Dynamics of Membranes. Amsterdam: Lipids in Model Membranes and Their Modifications by
Elsevier. pp. 98–160. Membrane Proteins. New York: John Wiley & Sons, Inc.
19. Voeltz, G.K., W.A. Prinz, Y. Shibata, J.M. Rist, and T.A. 1982.
Rapoport. 2006. A class of membrane proteins shaping the 38. Coolbear, K.P., C.B. Berde, and K.M. Keough. 1983. Gel to
tubular endoplasmic reticulum. Cell. 124: 573–586. liquid-crystalline phase transitions of aqueous dispersions
A Short History of Membrane Physics 27

of polyunsaturated mixed-acid phosphatidylcholines. 59. van der Ploeg, P. Berendsen, H.J.C., and P. van der Ploeg.
Biochemistry. 22: 1466–1473. 1982. Molecular dynamics simulation of a bilayer mem-
39. Nagle, J.F. 1973. Theory of biomembrane phase transitions. brane. J. Chem. Phys. 76: 3271.
J. Chem. Phys. 58: 252. 60. Jähnig, F. 1979. Molecular theory of lipid membrane order.
40. Scott Jr., H.L. 1975. Some models for lipid bilayer and J. Chem. Phys. 70: 3279.
biomembrane phase transitions. J. Chem. Phys. 62: 61. Gruen, D.W.R. 1985. A model for the chains in amphiphi-
1347–1353. lic aggregates. 1. Comparison with a molecular dynamics
41. A. Caillé, D. Pink, F. De Verteuil, and M.J.Z. 1980. simulation of a bilayer. J. Phys. Chem. 89: 146–153.
Theoretical models for quasi-two-dimensional mesomor- 62. Ben-Shaul, A., I. Szleifer, and W.M. Gelbart. 1985. Chain
phic monolayers and membrane bilayers. Can. J. Physics, organization and thermodynamics in micelles and bilayers.
58(5): 581–611. Doi: 10.1139/p80-083. I. Theory. J. Chem. Phys. 83: 3597–3611.
42. Jähnig, F. 1981. Critical effects from lipid-protein interac- 63. Ben-Shaul, A. 1995. Molecular theory of chain packing,
tion in membranes. I. Theoretical description. Biophys. J. elasticity and lipid protein interaction in lipid bilayers. In:
36: 329–345. Lipowsky R, E Sackmann, editors. Structure and Dynamics
43. de Gennes, P.G. 1974. General features of lipid organiza- of Membranes. Amsterdam: Elsevier. pp. 359–402.
tion. Phys. Lett. A. 47: 123–124. 64. Israelachvili, J.N., J. Mitchell, and B.W. Ninham. 1976.
44. Schröder, H. 1977. Aggregation of proteins in membranes. Theory of self-assembly of hydrocarbon amphiphiles
An example of fluctuation-induced interactions in liquid into micelles and bilayers. J. Chem. Soc. Farad. 2. 72:
crystals. J. Chem. Phys. 67: 1617–1619. 1525–1568.
45. Marčelja, S. 1974. Chain ordering in liquid crystals II. 65. Israelachvili, J.N. 2011. Intermolecular and surface forces.
Structure of bilayer membranes. Biochim. Biophys. Acta. 3rd Edition. London: Academic Press.
367: 165–176. 66. Seelig, J., and N. Waespe-Sarcevic. 1978. Molecular
46. Marčelja, S. 1976. Lipid-mediated protein interaction in order in cis and trans unsaturated phospholipid bilayers.
membranes. Biochim. Biophys. Acta. 455: 1–7. Biochemistry. 17: 3310–3315.
47. Cevc, G. 1991. Polymorphism of the bilayer membranes 67. Heller, H., M. Schaefer, and K. Schulten. 1993. Molecular
in the ordered phase and the molecular origin of the lipid dynamics simulation of a bilayer of 200 lipids in the
pretransition and rippled lamellae. Biochim. Biophys. Acta gel and in the liquid crystal phase. J. Phys. Chem. 97:
- Biomembr. 1062: 59–69. 8343–8360.
48. Nelson, D. 1987. Theory of the crumpling transition. In: 68. Fattal, D.R., and A. Ben-shaul. 1994. Mean-field calcula-
Nelsonj, D, Piran, T and Weinberg S, editor. Statistical tions of chain packing and conformational statistics in
Mechanics of Surfaces and Membranes. Singapore: World lipid bilayers: Comparison with experiments and molecular
Scientific, pp. 137–155. dynamics studies. Biophys. J. 67: 983–995.
49. Akabori, K., and J.F. Nagle. 2015. Structure of the DMPC 69. Op den Kamp, J. a, M.T. Kauerz, and L.L. van Deenen.
lipid bilayer ripple phase. Soft Matter. 11: 918–926. 1975. Action of pancreatic phospholipase A2 on phospha-
50. Albrecht, O, Gruler, H, and Sackmann, E. 1978. tidylcholine bilayers in different physical states. Biochim.
Polymorphism of phospholipid monolayers. J. Phys. 39: Biophys. Acta. 406: 169–177.
301–313. 70. Seigneuret, M., and P.F. Devaux. 1984. ATP-dependent
51. McConnell, H.M., and V.T. Moy. 1988. Shapes of finite­two- asymmetric distribution of spin-labeled phospholipids
dimensional lipid domains. J. Phys. Chem. 92: 4520–4525. in the erythrocyte membrane: Relation to shape changes.
52. McConnell, H.M. 1989. Theory of hexagonal and Proc. Natl. Acad. Sci. U. S. A. 81: 3751–3755.
stripe phases of monolayers. Proc. Natl. Acad. Sci. 86: 71. Steck, T.L., and Y. Lange. 2010. Cell cholesterol homeosta-
3452–3455 sis: Mediation by active cholesterol. Trends Cell Biol. 20:
53. Möhwald, H. 1995. Lipid Monolayers. In: Lipowsky, R and 680–687.
Sackmann E, editor. Handbook of Biological Physics Vol 72. Zheng, G., B. Tian, F. Zhang, F. Tao, and W. Li. 2011. Plant
1A. Amsterdam: North Holland. pp. 161–211. adaptation to frequent alterations between high and low
54. Seelig, J., and W. Niederberger. 1974. Two pictures of a temperatures: Remodelling of membrane lipids and main-
lipid bilayer. A comparison between deuterium label and tenance of unsaturation levels. Plant. Cell Environ. 34:
spin-label experiments. Biochemistry. 13: 1585–1588. 1431–42.
55. König, S., T.M. Bayerl, G. Coddens, D. Richter, 73. Wu, S., and H. McConnell. 1975. Phase separations in
and E. Sackmann. 1995. Hydration dependence ­phospholipid membranes. Biochemistry. 14: 847–854.
of chain dynamics and local diffusion in L-alpha- 74. Vist, M.R., and J.H. Davis. 1990. Phase equilibria of choles-
dipalmitoylphosphtidylcholine multilayers studied by terol/dipalmitoylphosphatidylcholine mixtures: 2H nuclear
incoherent quasi-elastic neutron scattering. Biophys. J. 68: magnetic resonance and differential scanning calorimetry.
1871–1880. Biochemistry. 29: 451–464.
56. Wiener, M.C., and S.H. White. 1992. Structure of a fluid 75. Mabrey, S., P.L. Mateo, and J.M. Sturtevant. 1978. High-
dioleoylphosphatidylcholine bilayer determined by joint sensitivity scanning calorimetric study of mixtures of
refinement of x-ray and neutron diffraction data. III. cholesterol with dimyristoyl- and dipalmitoylphosphatidyl-
Complete structure. Biophys. J. 61: 434–447. cholines. Biochemistry. 17: 2464–2468.
57. Dill, K.A., and P.J. Flory. 1980. Interphases of chain mole- 76. Evans, E., and D. Needham. 1986. Giant vesicle bilayers
cules: Monolayers and lipid bilayer membranes. Proc. Natl. composed of mixtures of lipids, cholesterol and polypep-
Acad. Sci. 77: 3115–3119. tides. Thermomechanical and (mutual) adherence proper-
58. Leermakers, F.A.M., and J.M.H.M. Scheutjens. 1988. ties. Faraday Discuss. Chem. Soc. 81: 267–280.
Statistical thermodynamics of association colloids. III. The 77. Knoll, W., G. Schmidt, and E. Sackmann. 1983. Critical
gel to liquid phase transition of lipid bilayer membranes. J. demixing in fluid bilayers of phospholipid mixtures. A neu-
Chem. Phys. 89: 6912. tron diffraction study. J. Chem. Phys. 79: 3439–3442.
28 Handbook of Lipid Membranes

78. Knoll, W., G. Schmidt, K. Ibel, and E. Sackmann. 1985. 98. Ludtke, S.J., K. He, W.T. Heller, T.A. Harroun, L. Yang, and
Small-angle neutron scattering study of lateral phase sepa- H.W. Huang. 1996. Membrane pores induced by magainin.
ration in dimyristoylphosphatidylcholine-cholesterol mixed Biochemistry. 35: 13723–13728.
membranes. Biochemistry. 24: 5240–5246. 99. Epand, R.M., Y. Shai, J.P. Segrest, and G.M.
79. Komura, S., H. Shirotori, P.D. Olmsted, and D. Andelman. Anantharamaiah. 1995. Mechanisms for the modulation of
2004. Lateral phase separation in mixtures of lipids and membrane bilayer properties by amphiphatic helical pep-
cholesterol systems. EPL (Europhysics Letters) 321: 7. tides. Biopolym. (Peptide Sci. 37: 319–338.
80. Elliott, R., I. Szleifer, and M. Schick. 2006. Phase diagram 100. Huang, H.W. 1986. Deformation free energy of bilayer
of a ternary mixture of cholesterol and saturated and unsat- membrane and its effect on gramicidin channel lifetime.
urated lipids calculated from a microscopic model. Phys. Biophys. J. 50: 1061–1070.
Rev. Lett. 96: 098101. 101. Zemel, A., D.R. Fattal, and A. Ben-Shaul. 2003. Energetics
81. Reinl, H., T. Brumm, and T.M. Bayerl. 1992. Changes of and self-assembly of amphipathic peptide pores in lipid
the physical properties of the liquid-ordered phase with membranes. Biophys. J. 84: 2242–2255.
temperature in binary mixtures of DPPC with cholesterol. 102. Zuckermann, M.J., and T. Heimburg. 2001. Insertion
Biophys. J. 61: 1025–1035. and pore formation driven by adsorption of proteins onto
82. Tu, K., M.L. Klein, and D.J. Tobias. 1998. Constant- lipid bilayer membrane-water interfaces. Biophys. J. 81:
pressure molecular dynamics investigation of cholesterol 2458–2472.
effects in dipalmitoylphosphatidylcholine bilayer. Biophys. 103. Lin, J.-H., and A. Baumgaertner. 2000. Stability of a melit-
J. 75: 2147–2156. tin pore in a lipid bilayer: A molecular dynamics study.
83. Brzustowicz, M.R., V. Cherezov, M. Caffrey, W. Stillwell, Biophys. J. 78: 1714–1724.
and S.R. Wassall. 2002. Molecular organization of choles- 104. Helfrich, P., and E. Jakobsson. 1990. Calculation of defor-
terol in polyunsaturated membranes: Microdomain forma- mation energies and conformations in lipid membranes
tion. Biophys. J. 82: 285–298. containing gramicidin channels. Biophys. J. 57: 1075–1084.
84. Maulik, P.R., and G.G. Shipley. 1996. N-palmitoyl sphingo- 105. Goulian, M., O.N. Mesquita, D.K. Fygenson, C. Nielsen,
myelin bilayers: Structure and interactions with cholesterol O.S. Andersen, and a Libchaber. 1998. Gramicidin channel
and dipalmitoylphosphatidylcholine. Biochemistry. 35: kinetics under tension. Biophys. J. 74: 328–337.
8025–8034. 106. Sens, P., and M.S. Turner. 2004. Theoretical model for the
85. Döbereiner, H.G., J. Käs, D. Noppl, I. Sprenger, and E. formation of caveolae and similar membrane invaginations.
Sackmann. 1993. Budding and fission of vesicles. Biophys. Biophys. J. 86: 2049–2057.
J. 65: 1396–403. 107. Bretscher, M.S., and S. Munro. 1993. Cholesterol and the
86. Sackmann, E. 2006. Thermo-elasticity and adhesion as golgi apparatus phe. Science 261(80): 1280–1281.
regulators of cell membrane architecture and function. J. 108. Kurrle, A., P. Rieber, and E. Sackmann. 1990. Reconstitution
Phys. Condens. Matter. 18: R785–R825. of transferrin receptor in mixed lipid vesicles. An example
87. Wimley, W.C., and S.H. White. 1996. Experimentally of the role of elastic and electrostatic forces for protein/lipid
determined hydrophobicity scale for proteins at membrane assembly. Biochemistry. 29: 8274–8282.
interfaces. Nat. Struct. Biol. 3: 842–848. 109. Simons, K., and E. Ikonon. 1997. Functional rafts in cell
88. Marsh, D. 1990. Lipid-protein interactions in membranes. membranes. Nature. 387: 569–572.
FEBS Lett. 268: 371–375. 110. Schwartz, S.L., C. Cao, O. Pylypenko, A. Rak, and A.
89. Mouritsen, O.G., and M. Bloom. 1984. Mattress model of Wandinger-Ness. 2008. Rab GTPases at a glance. J. Cell
lipid-protein interactions in membranes. Biophys. J. 46: Sci. 121: 246–246.
141–153. 111. Baumgart, T., T.S. Hess, and W.W. Webb. 2003. Imaging
90. Riegler, J., and H. Möhwald. 1986. Elastic interactions of coexisting fluid domains in biomembrane models coupling
photosynthetic reaction center proteins affecting phase tran- curvature and line tension. Nature. 425: 821–824.
sitions and protein distributions. Biophys. J. 49: 1111–1118. 112. Leibler, S., and D. Andelman. 2013. Ordered and curved
91. Gruler, H. 2015. Chemoelastic effect of membranes. Z. meso-structures in membranes and amphiphilic films.
Naturforsch. C. 30: 608–614. Journal de physique. 48: 2013–2018.
92. Fattal, D.R., and A. Ben-Shaul. 1993. A molecular model 113. Träuble, H., and H. Eibl. 1974. Electrostatic effects on lipid
for lipid protein interaction in membranes: The role of phase transitions: Membrane structure and ionic environ-
hydrophobic mismatch. Biophys. J. 65: 1795–1809. ment. PNAS. 71: 214–219.
93. Goulian, M., R. Bruinsma, and P. Pincus. 1993. Long-range 114. Jähnig, F. 1976. Electrostatic free energy and shift of the
forces in heterogeneous fluid membranes. Eur. Lett. 22: phase transition for charged lipid membranes. Biophys.
145–150. Chem. 4: 309–318.
94. Aranda-Espinoza, H., A. Berman, N. Dan, P. Pincus, and 115. Cevc, G. 1990. Membrane electrostatics. Biochim. Biophys.
S.A. Safran. 1996. Interaction between inclusions embed- Acta. 1031: 311–382.
ded in membranes. Biophys. J.J. 71: 648–656. 116. Andelman, D. 1995. Electrostatic properties of mem-
95. Killian, J.A. 1998. Hydrophobic mismatch between pro- branes: The Poisson-Boltzmann theory. In: Lipowsky R., E.
teins and lipids in membranes. Biophys. Biochim. 1376: Sackmann, editors. Structure and Dynamics of Membranes.
401–416. Amsterdam: Elsevier. pp. 603–642.
96. Siegel, D.P., and R.M. Epand. 1997. The mechanism of 117. Markovich, T., D. Andelman, and R. Podgornik. 2016.
lamellar-to-inverted hexagonal phase transitions in phos- Charged membranes: Poisson-Boltzmann theory, DLVO
phatidylethanolamine: Implications for membrane fusion paradigm and beyond. In: Safinya C, J Rädler, editors.
mechanisms. Biophys. J. 73: 3089–3111. Handbook of Lipid Membranes.
97. May, S., and A. Ben-Shaul. 1999. Molecular theory of lipid- 118. Parsegian, V.A., and D. Gingell. 1972. On the electrostatic
protein interaction and the Lalpha-HII transition. Biophys. interaction across a salt solution between two bodies bear-
J. 76: 751–767. ing unequal charges. Biophys. J. 12: 1192–1204.
Another random document with
no related content on Scribd:
CHAPTER X.
MAJOR BYNG’S SUGGESTION.

Major Byng, a wiry, dried-up little officer, with remarkably thin legs
and sporting proclivities, was reclining in a long chair, in the
verandah of the Napier Hotel, Poonah, smoking his after-breakfast
“Trichy,” and running his eye over the “Asian” pocket-book.
“Hullo, Byng, old man!” cried a loud cheerful voice, and looking up,
his amaze was depicted in the countenance he turned upon
Clarence Waring.
“Waring! Why—I thought,” putting down his book and sitting erect.
“Thought I had gone home—sold out and was stone broke. But
here I am, you see, on my legs again.”
“Delighted to hear it,” with a swift glance at Waring’s well-to-do air
and expensive-looking clothes. “Sit down, my dear boy,” he cried
cordially, “sit down and have a cheroot, and tell me all about yourself
and what has brought you back again to the land of regrets? Is it tea,
coffee, or gold?”
“Gold, in one sense. I am companion to a young millionaire, or
rather to the nephew of a man who has so much money—and no
children—that he does not know what to do.”
“And who is the young man? Does he know what to do?”
“His name is Jervis—his rich uncle is married to my sister; we are
connections, you see, and when he expressed a desire to explore
the gorgeous East, my sister naturally suggested me for the post of
guide, philosopher, and friend.”
Here Major Byng gave a short sharp laugh, like a bark.
“We landed in Bombay ten days ago, and are going to tour about
and see the world.”
“What is the programme?”
“My programme is as follows: Poonah races, Secunderabad races,
Madras races, a big game shoot in Travancore, expense no object,
elephants, beaters, club-cook, coolies with letters, and ice for the
champagne. Then I shall run him about in the train a bit, and show
him Delhi, Agra, Jeypore; after that we will put in the end of the cold
weather in Calcutta. I have lots of pals there, and from Calcutta we
will go to the hills, to Shirani. I shall be glad to see the old club again
—many a fleeting hour have I spent there!”
“That same club had a shocking bad name for gambling and bear
fighting,” said Major Byng significantly.
“I believe it had, now you mention it; but you may be sure that it
has reformed—like myself.”
“And this young fellow—what is he like?”
“Quiet, gentlemanly, easy-going, easily pleased, thinks every one
a good sort,” and Waring laughed derisively; “abhors all fuss or
show, never bets, never gets up in the morning with a head, no
expensive tastes.”
“In fact, his tastes are miserably beneath his opportunities! What a
pity it is that the millionaire is not your uncle!”
“Yes, instead of merely brother-in-law, and brothers-in-law are
notoriously unfeeling. However, I have adopted mine as my own
blood relation, for the present. I boss the show. Come and dine with
me to-night, and tell me all the ‘gup,’ and give me the straight tip for
the Arab purse.”
“All right. Is this young Jervis a sportsman?”
“He is a first-class man on a horse, and he plays polo, but he does
not go in for racing—more’s the pity!”
“Plays polo, does he? By Jove!” and an eager light shone in the
major’s little greenish eyes. “I’ve a couple of ponies for sale——”
“He does not want them now, whatever he may do later in Calcutta
or in the hills. I shall be looking out for three or four for myself, good
sound ones, mind you, Byng, up to weight. I’ve put on flesh, you see,
but I dare say my anxious responsibilities will wear me down a bit.
Jervis does not weigh more than ten stone, and, talk of the devil,
here he comes.”
Major Byng turned his head quickly, as at this moment Waring’s
travelling companion, a slight, active-looking young man, entered the
compound, closely pursued by a swarm of hawkers, and their
accompanying train of coolies, bearing on their heads the inevitable
Poonah figures, hand-screens, pottery, beetle-work, silks, silver, and
jewellery.
“I say, Waring,” he called out as he approached, “just look at me!
One would think I was a queen bee. If this goes on, you will have to
consign me to a lunatic asylum, if there is such a place out here.”
“Mark, let me introduce you to my old friend, Major Byng.”
Major Byng bent forward in his chair—to stand up was too great
an exertion even to greet a possible purchaser of polo ponies—
smiled affably, and said—
“You are only just out, I understand. How do you like India?”
“So far, I loathe it,” sitting down as he spoke, removing his topee,
and wiping his forehead. “Ever since I landed, I have lived in a state
of torment.”
“Ah, the mosquitoes!” exclaimed Major Byng, sympathetically; “you
will get used to them. They always make for new arrivals and fresh
blood.”
“No, no; but human mosquitoes! Touts, hawkers, beggars,
jewellers, horse-dealers. They all set upon me from the moment I
arrived. Ever since then, my life is a burthen to me. It was pretty bad
on board ship. Some of our fellow-travellers seemed to think I was a
great celebrity, instead of the common or ordinary passenger; they
loaded me with civil speeches, and the day we got into Bombay I
was nearly buried alive in invitations, people were so sorry to part
with me!”
“Here is a nice young cynic for you!” exclaimed Captain Waring,
complacently. “He is not yet accustomed to the fierce light that beats
upon a good-looking young bachelor, heir to thirty thousand a year
——”
“Why not make it a hundred thousand at once, while you are about
it?” interrupted the other impatiently. “How could they tell I was heir
to any one? I’m sure I am a most everyday-looking individual. My
uncle’s income is not ticketed on my back!”
“It was in one sense,” exclaimed Waring, with a chuckle.
“It was only with the common, vulgar class that I was so
immensely popular.”
“My dear fellow, you are much too humble minded. You were
popular with every one.”
“No, by no means; I could have hugged the supercilious old dame
who asked me with a drawl if I was in any way related to Pollitt’s
patent fowl food? I was delighted to answer with effusion, ‘Nephew,
ma’am.’ She despised me from the very bottom of her soul, and
made no foolish effort to conceal her feelings.”
“Ah! She had no daughters,” rejoined Waring, with a scornful
laugh. “The valet told all about you. He had nothing on earth to do,
but magnify his master and consequently exalt himself. Your value is
reflected in your gentleman’s gentleman, and he had no mock
modesty, and priced you at a cool million! By the way, I saw him
driving off just now in the best hotel landau, with his feet on the
opposite cushions, and a cigarette in his mouth. He is a magnificent
advertisement.”
They were now the centre of a vast mob of hawkers, who formed a
squatting circle, and the verandah was fully stocked. The jewellers
had already untied their nice little tin boxes from their white calico
wrappers, and their contents were displayed on the usual enticing
squares of red saloo.
“Waring Sahib!” screamed an ancient vendor with but one eye.
“Last time, three four years ago, I see you at Charleville Hotel,
Mussouri, I sell your honour one very nice diamond bangle for one
pretty lady——”
“Well, Crackett, I’m not such a fool now. I want a neat pearl pin for
myself.” He proceeded to deliberately select one from a case, and
then added with a grin, “That time, I paying for lady; this time,
gentleman,” pointing to Jervis, “paying for me.”
“I can’t stand it,” cried Jervis, jumping to his feet. “Here is the man
with the chestnut Arab and the spotted cob with pink legs, that has
been persecuting me for two days; and here comes the boy with the
stuffed peacock who has stalked me all morning; and—I see the girl
in the thunder and lightning waistcoat. I know she is going to ask me
to ride with her,” and he snatched up his topee and fled.
Major Byng noticed Jervis at the table d’hôte that evening. He had
been cleverly “cut off” from Waring, and was the prey of two over-
dressed, noisy young women. Mrs. Pollitt was mistaken, second-rate
people did come to India.
“I’ll tell you what, Waring!” he said to that gentleman, who was in
his most jovial, genial humour, “that young fellow is most shamefully
mobbed. His valet has given him away. If you don’t look out, he will
slip his heel ropes and bolt home. Pray observe his expression! Just
look at those two women, especially at the one who is measuring the
size of her waist with her serviette, for his information. He will go
back by the next steamer; it is written on his forehead!”
“No, he won’t do that,” rejoined Clarence, with lazy confidence.
“He has a most particular reason for staying out here for a while; but
I grant you that he is not enjoying himself, and does not appear to
appreciate seeing the world—and it is not a bad old world if you
know the right way to take it. Now, if I were in his shoes,” glancing
expressively across the table, “I’d fool that young woman to the very
top of her bent!”
In the billiard-room, when Mark joined them, Major Byng said—
“I saw your dismal plight at dinner, and pitied you. If you want to
lead a quiet life, and will take an old soldier’s advice, I would say, get
rid of the valet, send him home with half your luggage. Then start
from a fresh place, where no one knows you, with a good
Mussulman bearer, who is a complete stranger to your affairs. Let
Clarence here be paymaster—he can talk the language, and looks
wealthy and important—he won’t mind bearing the brunt, or being
taken for a rich man if the trouble breaks out again, and you can live
in peace and gang your ain gait.”
The Major’s advice was subsequently acted upon,—with most
excellent results. The cousins meanwhile attended the Poonah
races, where Clarence met some old acquaintances.
One of them privately remarked to Major Byng—
“Waring seems to have nine lives, like a cat, and looks most
festive and prosperous. I saw him doing a capital ready-money
business with the ‘Bookies’ just now—and he is a good customer to
the Para Mutual. It is a little startling to see him in the character of
mentor. I only hope he won’t get into many scrapes!”
“Oh, Telemachus has his head screwed on pretty tight, and he will
look after Waring—the pupil will take care of the teacher. He is a real
good sort, that boy. I wonder if his people know how old Clarence
used to race, and carry on and gamble at the lotteries, and generally
play the devil when he was out here?”
“Not they!” emphatically.
“He owes me one hundred rupees this three years, but he is such
a tremendous Bahadur now, that I am ashamed to remind him of
such a trifling sum. I sincerely hope that he has turned over a new
leaf and is a reformed character. What do you say, Crompton?”
“I say ‘Amen,’ with all my heart,” was the prompt response.
Mark Jervis had gone straight to the agents, Bostock & Bell’s, the
day he had landed in Bombay, and asked for his father’s address.
He only obtained it with difficulty and after considerable delay. The
head of the firm, in a private interview, earnestly entreated him to
keep the secret, otherwise they would get into trouble, as Major
Jervis was a peculiar man and most mysterious about his affairs,
which were now entirely managed by a Mr. Cardozo. Major Jervis
had not corresponded with them personally, for years. He then
scribbled something on a card, which he handed to the new arrival,
who eagerly read, “Mr. Jones, Hawal-Ghât, via Shirani, N.W.P.” The
major’s son despatched a letter with this superscription by the very
next post.
CHAPTER XI.
A RESERVED LADY.

A hot moonless night towards the end of March, and the up-mail
from Bombay to Calcutta has come to a standstill. The glare from the
furnace and the carriage lamps lights up the ghostly looking
telegraph-posts, the dusty cactus hedge, and illuminates a small
portion of the surrounding jungle. Anxiously gazing eyes see no sign
of a station, or even of a signalman’s hut, within the immediate glare
—and beyond it there looms a rocky, barren tract, chiefly swallowed
up in inscrutable darkness.
There is a babel of men’s voices, shrill and emotional, and not
emanating from European throats, a running of many feet, and
above all is heard the snorting of the engine and the dismal shrieks
of the steam whistle.
“What does it all mean?” inquired a silvery treble, and a fluffy head
leant out of a first-class ladies’ compartment.
“Nothing to be alarmed about,” responded a pleasant tenor voice
from the permanent way. “There has been a collision between two
goods trains about a mile ahead, and the line is blocked.”
“Any one killed?” she drawled.
“Only a couple of niggers,” rejoined the pleasant voice, in a
cheerful key.
“Dear me!” exclaimed the lady with sudden animation; “why,
Captain Waring, surely it cannot be you!”
“Pray why not?” now climbing up on the foot-board. “And do I
behold Mrs. Bellett?” as the head and shoulders of a good-looking
man appeared at the window, and looked into the carriage, which
contained a mountain of luggage, two ladies, a monkey, and a small
green parrot.
“Where have you dropped from?” she inquired. “I thought you had
left India for ever and ever. What has brought you back?”
“The remembrance of happier days,” he answered, with a
sentimental air, “and a P. and O. steamer.”
“But you have left the service, surely?”
“Yes, three years ago; it was too much of a grind at home.
Formerly I was in India on duty, now I am out here for pleasure. No
bother about over-staying my leave—no fear of brass hats.”
“Meanwhile, is there any fear of our being run into by another
train?” inquired the second lady nervously, a lady who sat at the
opposite side of the compartment with her head muffled up in a pink
shawl.
“Not the smallest; we are perfectly safe.”
“Captain Waring, this is my sister, Mrs. Coote,” explained Mrs.
Bellett. “And now perhaps you can tell us where we are, and what is
to become of us?”
“As to where you are, you are about three miles from Okara
Junction; as to what will happen to you, I am afraid that you will have
to walk there under my escort—if I may be permitted that honour.”
“Walk three miles!” she repeated shrilly. “Why, I have not done
such a thing for years, and I have on thin shoes. Could we not go on
the engine?”
“Yes, if the engine could fly over nearly a hundred luggage
waggons. It is a fine starlight night; we will get a lamp, and can keep
along the line. They have sent for a break-down gang, and we shall
catch another train at Okara. We will only have about an hour or two
to wait.”
“Well, I suppose we must make the best of it!” said Mrs. Coote,
“like others,” as numbers of natives flocked past, chattering volubly,
and carrying their bedding and bundles.
“I wish we could get supper at Okara,” said her sister. “I am sure
we shall want it after our tramp; but I know we need not build on
anything better than a goat chop, and the day before yesterday’s
curry. However, I have a tea-basket.”
“I can go one better,” said Captain Waring. “I have a tiffin-basket,
well supplied with ice, champagne, cold tongue, potted grouse—
cake—fruit——”
“You are making me quite ravenous,” cried Mrs. Bellett. “But how
are you to get all these delicacies to Okara?”
“By a coolie, I hope. If the worst comes to the worst, I will carry
them on my head, sooner than leave them behind. However, rupees
work wonders, and I expect I shall get hold of as many as will carry
the basket, and also your baggage; I suppose fifty will do?” and with
a grin, he climbed down out of sight.
“What a stroke of luck, Nettie!” exclaimed Mrs. Bellett. “He used to
be such a friend of mine at Mussouri, and imagine coming across
him in this way! He seems to be rolling in money; he must have
come in for a fortune, for he used to be frightfully hard up. I’m so
glad to meet him.”
“Yes, it’s all very fine for you, who are dressed,” rejoined the other
in a peevish voice; “but just look at me in an old tea-jacket, with my
hair in curling-pins!”
“Oh, you were all right! I’m certain he never noticed you!” was the
sisterly reply. “Let us be quick and put up our things. I wish to
goodness the ayah was here,” and she began to bustle about, and
strap up wraps and pillows, and collect books and fans.
Every one in the train seemed to be in a state of activity, preparing
for departure, and presently many parties on foot, with lanterns,
might be seen streaming along the line. Captain Waring promptly
returned with a dozen coolies, and soon Mrs. Bellett’s carriage was
empty. She and her sister were assisted by Captain Waring and a
young man—presumably his companion. Ere descending, Mrs.
Bellett, who had a pretty foot, paused on the step to exhibit the
thinness of her shoes, and demanded, as she put out her Louis-
Quatorze sole, “how she was to walk three miles in that, along a
rough road?”
The two ladies were nevertheless in the highest spirits, and
appeared to enjoy the novelty of the adventure. Ere the quartette
had gone twenty yards, the guard came shouting after them—
“Beg pardon, sir,” to Captain Waring, “but there is a lady quite
alone in my charge. I can’t take her on; I must stay and see to the
baggage, and remain here. And would you look after her?”
“Where is she?” demanded Waring, irritably.
“Last carriage but one—reserved ladies, first-class.”
“I say, Mark,” turning to his friend, “if she is a reserved lady, you
are all right. He is awfully shy, this young fellow,” he explained to his
other companions, with a loud laugh. “I don’t mind betting that she is
old—and you know you are fond of old women—so just run back like
a good chap. You see, I have Mrs. Bellett and her sister—you won’t
be five minutes behind us, bring on the reserved lady as fast as you
can.”
The other made no audible reply, but obediently turned about, and
went slowly past the rows of empty carriages until he came nearly to
the end of the train. Here he discovered a solitary white figure
standing above him in the open door of a compartment, and a girlish
voice called down into the dark—
“Is that you, guard?”
“No,” was the answer; “but the guard has sent me to ask if I can
help you in any way.”
A momentary pause, and then there came a rather doubtful
“Thank you.”
“Your lamp has gone out, I see, but I can easily strike a match and
get your things together. There is a block on the line, and you will
have to get down and walk on to the next station.”
“Really? Has there been an accident? I could not make out what
the people were saying.”
“It is not of much consequence—two goods trains disputing the
right of way; but we shall have to walk to Okara to catch the
Cawnpore mail.”
“Is it far?”
“About three miles, I believe.”
“Oh, that is not much! I have not many things—only a dressing-
bag, a rug, and a parasol.”
“All right; if you will pass them down, I will carry them.”
“But surely there is a porter,” expostulated the lady, “and I need
not trouble you.”
“I don’t suppose there is what you call a porter nearer than
Brindisi, and all the coolies are taking out the luggage. Allow me to
help you.”
In another second the young lady, who was both light and active,
stood beside him on the line. She was English; she was tall; and she
wore a hideously shaped country-made topee—that was all that he
could make out in the dim light.
“Now, shall we start?” he asked briskly, taking her bag, rug, and
parasol.
“Please let me have the bag,” she entreated. “I—I—that is to say, I
would rather keep it myself. All my money is in it.”
“And I may be a highwayman for what you know,” he returned,
with a laugh. “I give you my word of honour that, if you will allow me
to carry it, I will not rob you.”
“I did not mean that,” she stammered.
“Then what did you mean? At any rate I mean to keep it. The other
passengers are on ahead—I suppose you are quite alone?”
“Almost. There is a servant in the train who is supposed to look
after me, but I am looking after him, and seeing that he is not left
behind at the different junctions. We cannot understand one word we
exchange, so he grins and gesticulates, and I nod and point; but it all
comes to nothing, or worse than nothing. I wanted some tea this
morning, and he brought me whisky and soda.”
“And have you no one to rely on but this intelligent attendant?”
“No. The people I came out with changed at Khandala, and left me
in charge of the guard, and in a through carriage to Allahabad; and of
course we never expected this.”
“So you have just come out from home?” he observed, as they
walked along at a good pace.
“Yes; arrived yesterday morning in the Arcadia.”
“Then this is the first time you have actually set foot on Indian soil,
for trains and gharries do not count?”
“It is. Are there”—looking nervously at the wild expanse on either
hand—“any tigers about, do you think?”
“No, I sincerely hope not, as I have no weapon but your parasol.
Joking apart, you are perfectly safe. This”—with a wave of the
aforesaid parasol—“is not their style of hunting-ground.”
“And what is their style, as you call it?”
“Oh, lots of high grass and jungle, in a cattle country.”
“Have you shot many tigers?”
“Two last month. My friend and I had rather good sport down in
Travancore.”
“I suppose you live out here?”
“No, I have only been about six months in the country.”
“I wish I had been six months in India.”
“May I ask why?”
“Certainly you may. Because I would be going home in six months
more.”
“And you only landed forty-eight hours ago! Surely you are not
tired of it already. I thought all young ladies liked India. Mind where
you are going! It is very dark here. Will you take hold of my arm?”
“No, thank you,” rather stiffly.
“Then my hand? You really had better, or you will come a most
awful cropper, and trip over the sleepers.”
“Here is an extraordinary adventure!” said Honor to herself. “What
would Jessie and Fairy say, if they could see me now, walking along
in the dark through a wild desolate country, hand-in-hand with an
absolutely strange young man, whose face I have never even seen?”
A short distance ahead were groups of chattering natives—women
with red dresses and brass lotahs, which caught the light of their
hand-lanterns (a lantern is to a native what an umbrella is to a
Briton); turbaned, long-legged men, who carried bundles, lamps, and
sticks. The line was bordered on either hand by thick hedges of
greyish cactus; here and there glimmered a white flower; here and
there an ancient bush showed bare distorted roots, like the ribs of
some defunct animal. Beyond stretched a dim mysterious landscape,
which looked weird and ghostly by the light of a few pale stars. The
night was still and oppressively warm.
“You will be met at Allahabad, I suppose?” observed Honor’s
unknown escort, after a considerable silence.
“Yes—by my aunt.”
“You must be looking forward to seeing her again?”
“Again! I have never seen her as yet.” She paused, and then
continued, “We are three girls at home, and my aunt and uncle
wished to have one of us on a visit, and I came.”
“Not very willingly, it would seem,” with a short laugh.
“No; I held out as long as I could. I am—or rather was—the useful
one at home.”
“And did your aunt and uncle stipulate for the most useful niece?”
“By no means—they—they, to tell you the truth, they asked for the
pretty one, and I am not the beauty of the family.”
“No? Am I to take your word for that, or are you merely fishing?”
“I assure you that I am not. I am afraid my aunt will be
disappointed; but it was unavoidable. My eldest sister writes, and
could not well give up what she calls her literary customers. My next
sister is—is—not strong, and so they sent me—a dernier ressort.”
She was speaking quite frankly to this stranger, and felt rather
ashamed of her garrulity; but he had a pleasant voice, he was the
first friendly soul she had come across since she had left home, and
she was desperately home-sick. A long solitary railway journey had
only increased her complaint, and she was ready to talk of home to
any one—would probably have talked of it to the chuprassi,—if he
could have understood her!
Her escort had been an unscrupulous, selfish little woman, whose
nurse, having proved a bad sailor, literally saddled her good-natured,
inexperienced charge with the care of two unruly children, and this in
a manner that excited considerable indignation among her fellow
passengers.
“Why should you call yourself a dernier ressort?” inquired her
companion, after a pause, during which they continued to stumble
along, she holding timidly by the young man’s arm.
“Because I am; and I told them at home with my very last breath
that I was not a bit suited for coming out here, and mixing with
strangers—nothing but strangers—and going perpetually into what is
called ‘smart’ society, and beginning a perfectly novel kind of life. I
shall get into no end of scrapes.”
“May I ask your reason for this dismal prophecy?”
“Surely you can guess! Because I cannot hold my tongue. I blurt
out the first thing that comes into my head. If I think a thing wrong, or
odd, I must say so; I cannot help it, I am incurable. People at home
are used to me, and don’t mind. Also, I have a frightful and wholly
unconscious habit of selecting the most uncomfortable topics, and
an extremely bad memory for the names and faces of people with
whom I have but a slight acquaintance; so you see that I am not
likely to be a social success!”
“Let us hope that you take a gloomy view of yourself. For instance,
what is your idea of an uncomfortable topic?”
“If I am talking to a person with a cast in the eye, I am positively
certain ere long to find myself conversing volubly about squints; or, if
my partner wears a wig, I am bound to bring wigs on the tapis. I
believe I am possessed by some mischievous imp, who enjoys my
subsequent torture.”
“Pray how do you know that I have not a squint, or a wig, or both?
A wig would not be half a bad thing in this hot climate; to take off
your hair as you do your hat would often be a great relief! Ah, here
we are coming to the scene of the collision at last,” and presently
they passed by a long row of waggons, and then two huge engines,
one across the line, the other reared up against it; an immense
bonfire burnt on the bank, and threw the great black monsters into
strong outline. Further on they came to a gate and level crossing.
The gate of the keeper’s hut stood wide open, and on the threshold
a grey-haired old woman sat with her head between her knees,
sobbing; within were moans, as if wrung from a sufferer in acute
anguish. Honor’s unknown companion suddenly halted, and
exclaimed impulsively—
“I’m afraid some one has been badly hurt; if you don’t mind, I’ll just
go and see.”
Almost ere she had nodded a quick affirmative, he had vaulted
over the gate, and left her.
CHAPTER XII.
TWO GOOD SAMARITANS.

In all her life, the youngest Miss Gordon had never felt so utterly
solitary or forsaken as now, when she stood alone on the line of the
Great Indian Peninsular Railway. Before her the party of natives, with
their twinkling lanterns, were gradually reaching vanishing point;
behind her was a long, still procession of trucks and waggons, that
looked like some dreadful black monster waiting for its prey; on
either hand stretched the greyish unknown mysterious landscape,
from which strange unfamiliar sounds, in the shape of croakings and
cries, were audible. Oh! when would her nameless companion
return? She glanced anxiously towards the hut, it was beyond the
gate, and down a steep bank, away from the road; animated figures
seemed to pass to and fro against the lighted open door. Ah! here
came one of them, her escort, who had in point of fact been only
absent five minutes, and not, as she imagined, half an hour.
“It is a stoker who has been cut about the head and badly
scalded,” he explained breathlessly. “They are waiting for an
apothecary from Okara, and meanwhile they are trying a native herb
and a charm. They don’t seem to do the poor chap much good. I
think I might be able to do something better for him, though I have no
experience, beyond seeing accidents at football and out hunting; but
I cannot leave you here like this, and yet I cannot well ask you inside
the hut, the heat is like a furnace—and—altogether—it—it would be
too much for you, but if you would not mind waiting outside just for a
few minutes, I’d get you something to sit on.”
“Thank you, but I would rather go in—I have attended an
ambulance class—‘first aid,’ you know, and perhaps I may be of
some little use; there is sticking-plaster, eau-de-Cologne, and a pair
of scissors in my bag.”
“Well, mind; you must brace up your nerves,” he answered, as he
pushed open the gate, and led her down the crumbling sandy incline.
The heat within the hut was almost suffocating; as the girl,
following her guide, entered, every eye was instantly fixed upon her
in wide surprise.
By the light of a small earthen lamp, which smoked horribly, she
distinguished the figure of a man crouching on the edge of a
charpoy; he was breathing in hard hoarse gasps, and bleeding from
a great gash above his eye.
A Eurasian, in a checked cotton suit, stood by, talking incessantly
—but doing nothing else. There were also present, besides the old
woman—a veritable shrivelled-up hag—two native men, possibly the
“bhai-bands,” or chums of the sufferer; in a corner, a large black
pariah sat watching everything, with a pair of unwinking yellow eyes;
and on another charpoy lay a still figure, covered with a sheet. A few
earthen chatties, a mat, a huka, and some gaudy English prints—for
the most part nailed upside down—completed the picture. Hitherto
the travelling companions had been to each other merely the
embodiment of an undefined figure and a voice; the light of the little
mud lamp, whose curling smoke threw outlines of dancing black
devils on the walls, now introduced them for the first time face to
face. To Honor Gordon stood revealed an unexpectedly good-looking
young man, slight and well built, with severely cut features, and a
pair of handsome hazel eyes, which were surveying her gravely. A
gentleman, not merely in his speech and actions, but in his bearing.
He, on his part, was not in the least surprised to behold a pale but
decidedly pretty girl; by means of some mysterious instinct he had
long made up his mind that the owner of such a delicate hand and
sweet clear voice could not be otherwise than fair to see.
“The apothecary cannot be here for one hour!” exclaimed the
Eurasian, glibly. “He,” pointing to the patient, “is very bad. We have
put some herbs to his arm, and the back of his head; but I, myself,
think that he will die!” he concluded with an air of melancholy
importance.
Some kind of a bandage was the first thing Honor asked for, and
asked for in vain; she then quickly unwound the puggaree from her
topee, and tore it into three parts.
Then she bathed and bandaged the man’s head, with quick and
sympathetic fingers, whilst Jervis held the lamp, offered suggestions,
and looked on, no less impressed than amazed; he had hitherto had
an idea that girls always screamed and shrank away from the sight
of blood and horrors.
This girl, though undeniably white, was as cool and self-
possessed, as firm, yet gentle, as any capable professional nurse.
The scalded arm and hand—a shocking spectacle—were attended
to by both. The great thing was to exclude the air, and give the
sufferer at least temporary relief. With some native flour, a bandage
was deftly applied, the arm placed in a sling, and the patient’s head
was bathed with water and eau-de-Cologne. Fanned assiduously by
the girl’s fan, he began to feel restored, he had been given heart, he
had been assured that his hurts were not mortal, and presently he
languidly declared himself better.
The natives who stood round, whilst the sahib and Miss Sahib
ministered so quickly and effectually to their friend, now changed
their lamentations to loud ejaculations of wonder and praise. Miss
Gordon was amazed to hear her companion giving directions to
these spectators in fluent and sonorous Hindustani, and still more
astounded when, as she took up her topee, preparatory to departure,
the Eurasian turned to him, and said in an impressive squeak—
“Sir, your wife is a saint—an angel of goodness”—and then, as an
hasty afterthought, he added, “and beauty!”
Before Jervis could collect his wits and speak, she had replied—
“I am not this gentleman’s wife; we are only fellow-passengers.
Why should you think so?” she demanded sharply.
“Because—oh, please do not be angry—you looked so suitable,”
he answered with disarming candour. “Truly, I hope you may be
married yet, and I wish you both riches, long life, and great
happiness,” he added, bowing very low, lamp in hand.
Honor passed out of the hut, with her head held extraordinarily
high, scrambled up the bank, and proceeded along the line at a
headlong pace in indignant silence.
She now maintained a considerable distance between herself and
her escort; no doubt her eyes were becoming accustomed to the dim
light, and at any rate there was that in her air which prevented him
offering either arm or hand. In spite of the recent scene in which they
had both been actors, where he had clipped hair and cut plaster, and
she had applied bandages and scanty remedies to the same “case,”
they were not drawn closer together; on the contrary, they were
much further apart than during the first portion of their walk, and the
young lady’s confidences had now entirely ceased. She confined
herself exclusively to a few bald remarks about the patient, and the
climate, remarks issued at intervals of ten minutes, and her answers
to his observations were confined to “Yes” and “No.” At last Okara
station was reached; and, to tell the truth, neither of them were sorry
to bring their tête-à-tête to a conclusion. The dazzling lights on the
platform made their eyes blink, as they threaded their way to the
general refreshment room, discovering it readily enough by sounds
of many and merry voices, who were evidently availing themselves
of its somewhat limited resources.
It was not a very large apartment, but it was full. The table was
covered with a thin native tablecloth, two large lamps with punkah
tops, and two cruet-stands and an American ice-pitcher were placed
at formal intervals down the middle. It was surrounded with people,
who were eating, drinking, and talking. At the further end sat Captain
Waring, supported on either hand by his two fair companions, three
men—young and noisy, whom they evidently knew—and a prim,
elderly woman, who looked inexpressibly shocked at the company,
and had pointedly fenced herself off from Mrs. Bellett with a teapot
and a wine-card. Captain Waring’s friends had not partaken of tea
(as the champagne-bottle testified). The tongue, cake, and fruit had
also evidently received distinguished marks of their esteem. Mrs.
Bellett put up her long eyeglass, and surveyed exhaustively the pair
who now entered.

You might also like