Download as pdf or txt
Download as pdf or txt
You are on page 1of 60

Hypoxic Respiratory Failure in the

Newborn: From Origins to Clinical


Management 1st Edition Shyamala
Dakshinamurti (Editor)
Visit to download the full and correct content document:
https://ebookmeta.com/product/hypoxic-respiratory-failure-in-the-newborn-from-origin
s-to-clinical-management-1st-edition-shyamala-dakshinamurti-editor/
More products digital (pdf, epub, mobi) instant
download maybe you interests ...

Tendinopathy From Basic Science to Clinical Management


Kentaro Onishi

https://ebookmeta.com/product/tendinopathy-from-basic-science-to-
clinical-management-kentaro-onishi/

Goldsmith’s Assisted Ventilation of the Neonate: An


Evidence-Based Approach to Newborn Respiratory Care 7th
Edition Martin Keszler

https://ebookmeta.com/product/goldsmiths-assisted-ventilation-of-
the-neonate-an-evidence-based-approach-to-newborn-respiratory-
care-7th-edition-martin-keszler/

A Relational Approach to Governing Wicked Problems From


Governance Failure to Failure Governance Peeter Selg

https://ebookmeta.com/product/a-relational-approach-to-governing-
wicked-problems-from-governance-failure-to-failure-governance-
peeter-selg/

Clinical Methods in Respiratory Medicine 1st Edition


Jyotirmoy Pal Supriya Sarkar Sekhar Chakraborty

https://ebookmeta.com/product/clinical-methods-in-respiratory-
medicine-1st-edition-jyotirmoy-pal-supriya-sarkar-sekhar-
chakraborty/
Primary Mathematics 3A Hoerst

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

Respiratory Care Assessment and Management 2nd Edition


Deborah Duncan

https://ebookmeta.com/product/respiratory-care-assessment-and-
management-2nd-edition-deborah-duncan/

Learning from Failure in the Design Process


Experimenting with Materials 1st Edition Lisa Huang

https://ebookmeta.com/product/learning-from-failure-in-the-
design-process-experimenting-with-materials-1st-edition-lisa-
huang/

Heading Home with Your Newborn From Birth to Reality


4th Edition Laura A. Jana

https://ebookmeta.com/product/heading-home-with-your-newborn-
from-birth-to-reality-4th-edition-laura-a-jana/

Failure to Thrive and Malnutrition A Practical Evidence


Based Clinical Guide Joyee Goswami Vachani (Editor)

https://ebookmeta.com/product/failure-to-thrive-and-malnutrition-
a-practical-evidence-based-clinical-guide-joyee-goswami-vachani-
editor/
HYPOXIC RESPIRATORY
FAILURE IN THE
NEWBORN
HYPOXIC RESPIRATORY
FAILURE IN THE
NEWBORN

From Origins to Clinical Management

Edited by
Shyamala Dakshinamurti

Section Editors
Steven H. Abman
Po-Yin Cheung
Satyan Lakshminrusimha
Patrick J. McNamara
William K. Milsom
CRC Press
Boca Raton and London
First edition published 2022

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

© 2022 Taylor & Francis Group, LLC

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

This book contains information obtained from authentic and highly regarded sources. While all reasonable efforts
have been made to publish reliable data and information, neither the author[s] nor the publisher can accept any legal
responsibility or liability for any errors or omissions that may be made. The publishers wish to make clear that any
views or opinions expressed in this book by individual editors, authors or contributors are personal to them and do
not necessarily reflect the views/opinions of the publishers. The information or guidance contained in this book is
intended for use by medical, scientific or health-care professionals and is provided strictly as a supplement to the
medical or other professional’s own judgement, their knowledge of the patient’s medical history, relevant manufac-
turer’s instructions and the appropriate best practice guidelines. Because of the rapid advances in medical science, any
information or advice on dosages, procedures or diagnoses should be independently verified. The reader is strongly
urged to consult the relevant national drug formulary and the drug companies’ and device or material manufactur-
ers’ printed instructions, and their websites, before administering or utilizing any of the drugs, devices or materials
mentioned in this book. This book does not indicate whether a particular treatment is appropriate or suitable for a
particular individual. Ultimately it is the sole responsibility of the medical professional to make his or her own profes-
sional judgements, so as to advise and treat patients appropriately. The authors and publishers have also attempted to
trace the copyright holders of all material reproduced in this publication and apologize to copyright holders if permis-
sion 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 pho-
tocopying, microfilming, and recording, or in any information 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 mpkbookspermissions@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.

“Copyright Satyan Lakshminrusimha, used with permission”, for figures provided by Dr. Satyan Lakshminrusimha.

ISBN: 9780367493998 (hbk)


ISBN: 9781032078182 (pbk)
ISBN: 9780367494018 (ebk)

DOI: 10.1201/9780367494018

Typeset in Warnock Pro


by KnowledgeWorks Global Ltd.
CONTENTS

Preface..................................................................................................................................................................................................................................vii
Editor.................................................................................................................................................................................................................................. viii
Section Editors....................................................................................................................................................................................................................ix
Contributors......................................................................................................................................................................................................................... x

Introduction.............................................................................................................................................................................................................. 1
Shyamala Dakshinamurti, William K. Milsom, Satyan Lakshminrusimha, Steven H. Abman, Po-Yin Cheung and
Patrick J. McNamara

Part 1: THE ORIGINS OF HYPOXIA TOLERANCE


Edited by William K. Milsom
The Human Fetus and Metabolic Adaptations to Hypoxia............................................................................................................................ 6
Dominique Singer
Hypoxia as a Neuroinflammatory Stimulus during Development.............................................................................................................13
Peter M. MacFarlane and Allison S. Osborne
Human Adaptations to High Altitude..............................................................................................................................................................19
Tatum Simonson
Fetal Llama Adaptation to Altitude in the Andean Altiplano.....................................................................................................................24
Aníbal J. Llanos, Germán Ebensperger, Emilio A. Herrera, Roberto V. Reyes and María Serón-Ferré
Neonates of Burrowing and Hibernating Mammals: Metabolic and Respiratory Adaptations to Hypoxia.....................................28
Yvonne A. Dzal and William K. Milsom
Diving Response and Hypoxia in Deep Sea Mammals.................................................................................................................................34
Allyson Hindle
The Phylogeny and Ontogeny of Cardiorespiratory Coupling in Vertebrates and Its
Relevance to Noninvasive Monitoring of the Human Fetus........................................................................................................................39
Edwin W. Taylor and Harry Gee

Part 2: FETAL HYPOXIA AND NEONATAL TRANSITION


Edited by Satyan Lakshminrusimha
Fetal and Neonatal Oxygen Environment........................................................................................................................................................46
Payam Vali, Robin H. Steinhorn and Satyan Lakshminrusimha
Fetal Oxygenation during Maternal Hypoxic Illness....................................................................................................................................51
Fabiana Postolow and Shyamala Dakshinamurti
Hemodynamics of the Circulatory Transition................................................................................................................................................57
Graeme R. Polglase, Martin Kluckow, Andrew W. Gill and Stuart B. Hooper
Oxygen during Postnatal Stabilization.............................................................................................................................................................62
Maximo Vento and Ola D. Saugstad

Part 3: BIOLOGY OF HYPOXIC RESPIRATORY FAILURE IN THE NEONATE


Edited by Steven H. Abman
Hypoxia and Pulmonary Artery Structure......................................................................................................................................................68
Csaba Galambos and Steven H. Abman
Animal Models of PPHN and Vasoconstrictor Signaling in Hypoxia....................................................................................................... 74
Candice D. Fike and Judy L. Aschner
Hypoxia and Endothelial Dysfunction in the Lung.......................................................................................................................................80
Emily Callan and Girija Ganesh Konduri
Effects of Hypoxia on Pulmonary Vascular Smooth Muscle Contraction and Relaxation...................................................................87
Shyamala Dakshinamurti, Anjali Y. Bhagirath and Robert P. Jankov
Cellular Oxygen Sensing, Mitochondrial Oxygen Sensing and Reactive Oxygen Species...................................................................96
Jason Boehme and Emin Maltepe

v
vi Contents

Reactive Oxygen Species Signaling in Animal Models of Pulmonary Hypertension..........................................................................101


Stephen Wedgwood, Satyan Lakshminrusimha and Robin H. Steinhorn
Hypoxia, Myocardial Metabolic Adaptation, and Right Ventricular Performance..............................................................................106
Mark K. Friedberg
Hypoxic Remodeling of Neonatal Pulmonary Artery and Myocardium................................................................................................111
Hui Zhang, R. Dale Brown, Jason Williams, Cassidy Delaney and Kurt R. Stenmark
Hypoxia-Induced Epigenetic Mechanisms of Pulmonary Hypertension...............................................................................................119
Kurt H. Albertine

Part 4: HYPOXIA AND COLLATERAL DAMAGE


Edited by Po-Yin Cheung
The Effects of Hypoxia Ischemia on the Term Brain, and a Strategic Approach..................................................................................126
Aravanan Anbu Chakkarapani and Nicola J. Robertson
Effects of Hypoxia on Cerebral Perfusion and the Blood-Brain Barrier.................................................................................................135
Divyen K. Shah, W. L. Amelia Lee, David F. Wertheim and Stephen T. Kempley
Effects of Hypoxia on Airway, Alveolar Function, and Respiration.........................................................................................................142
Y.S. Prakash, Christina M. Pabelick and Peter M. MacFarlane
Hypoxic Respiratory Failure and the Neonatal Kidney..............................................................................................................................148
Catherine Morgan and Po-Yin Cheung
The Effect of Hypoxia on Intestinal Function...............................................................................................................................................152
Eric B. Ortigoza and Josef Neu
Effects of Hypoxia on Perinatal Drug Disposition.......................................................................................................................................155
Karel Allegaert, Anne Smits and Pieter Annaert
Anesthetic Considerations for the Neonate with Hypoxic Respiratory Failure....................................................................................160
M. Ruth Graham

Part 5: DIAGNOSIS AND MANAGEMENT OF NEONATAL HYPOXIC RESPIRATORY FAILURE


Edited by Patrick J. McNamara
Epidemiology and Outcomes of Infants with Hypoxemic Respiratory Failure.....................................................................................168
Souvik Mitra, Roberta L. Keller and Prakesh S. Shah
Clinical Evaluation of Hypoxic Respiratory Failure....................................................................................................................................177
Yasser Elsayed and Shyamala Dakshinamurti
Diagnosis of Pulmonary Hypertension by Echocardiography..................................................................................................................184
Danielle R. Rios, Patrick J. McNamara and Regan E. Giesinger
Right Ventricular Performance and Ventricular Interdependence in Hypoxia.....................................................................................193
Afif El Khuffash and Philip T. Levy
Ventilation Strategies in Neonatal Hypoxemic Respiratory Failure........................................................................................................199
Bradley A. Yoder, Michelle J. Yang, Nicholas R. Carr and Martin Keszler
Pulmonary Hypertension Phenotypes in the Newborn............................................................................................................................. 208
Regan E. Giesinger, John P. Kinsella, Steven H. Abman and Patrick J. McNamara
Special Consideration: HRF in Congenital Diaphragmatic Hernia.........................................................................................................216
Gabriel Altit, Jason Gien, Nolan DeLeon and Richard Keijzer
Special Consideration: Hypoxemic Respiratory Failure among Patients with Hypoxic Ischemic Encephalopathy.....................224
Arvind Sehgal, Satyan Lakshminrusimha and Regan E. Giesinger
Special Consideration: HRF in the Preterm..................................................................................................................................................233
Michelle Baczynski, Amish Jain and Dany E. Weisz
Hypoxemic Respiratory Failure: Neonatal Cases.........................................................................................................................................238
Adrianne Rahde Bischoff, Faith Zhu, Amish Jain and Patrick J. McNamara

Index.................................................................................................................................................................................................................................. 246
PREFACE

Babies with hypoxic respiratory failure were always the sickest in and drawing together international expertise in diagnosis and
the newborn ICU. As a neonatology trainee, I would sit at their treatment. I am grateful to my august co-editors for their tre-
bedsides on call nights, watching their seesawing oxygen satura- mendous insights and enthusiasm in bringing this collection of
tions, all our whirring, thumping machines a poor substitute for knowledge together. And as always, delighted and illuminated by
the safety and sanity of the uterus; willing their fist-sized hearts Satyan Lakshminrusimha’s illustrations.
to pump a little longer, while we figured out a better way to push It must be acknowledged that this book was conceived before,
oxygen into them. but written during the COVID-19 pandemic. There has been no
Yet, at these same oxygen levels, a fetus would have thrived. scientific connection established between COVID and neonatal
What failed these infants was the elemental transition process pulmonary hypertension. But there is an affective connection, in
from fetus to newborn; the key to be born and to live in this world. writing about hypoxic respiratory failure at a crystalline moment
The genesis of this book can be traced back to those roller- when the world is struggling with the toll of hypoxic respiratory
coaster call nights. Also to the writings of evo-devo stalwarts failure. I thank all the contributing authors for their commitment
Stephen Jay Gould and Sean Carroll; to serendipitous lectures to this project despite this pressure.
by the inimitable John West on how the elephant lost its pleu- Every book requires a village; in particular, the organizational
ral space, and Paul Ponganis on the origins of the diving reflex, skills of project managers Nathalie Buissé and Sophia Mbabaali
which felt like bathing one’s brain in pure logic; and a chance from the George & Fay Yee Centre for Healthcare Innovation, and
meeting with Bill Milsom on a mountain in Mongolia. Clear con- the Medical Sciences editorial desk at Taylor & Francis/CRC Press.
nections can be made from human evolution to the process of I thank my family for their support during the compilation
our emergence as creatures capable of independent life. The poet of this volume, including eagle-eyed proofreading, and for their
Mary Oliver urged, Pay attention / Be astonished / Tell about it. gifts of science, mountains and words.
This applies to scientists and clinicians too. There is a compelling Half a century ago, my cousin Sanjay died of hypoxic respira-
poetry to physiology that lurks beneath the myriad details. tory failure soon after birth. How much we have learned since
Gathered here is a collection of reviews by masters in their then. To him and every blue baby since, this effort is in memory.
fields, highlighting the pathways of hypoxia and pulmonary
hypertension from the population to the organism and the cell Shyamala Dakshinamurti

vii
EDITOR

Dr Shyamala Dakshinamurti is a neonatologist and biomedi- organizer of the annual Canadian National Perinatal Research
cal researcher, Professor of Pediatrics and Physiology at the Meeting. Dr Dakshinamurti’s research is on the hemodynamics
University of Manitoba, Canada, and member of the Biology of the neonatal pulmonary circuit and the physiology of hypoxia
of Breathing theme, Children’s Hospital Research Institute during circulatory transition. She is also a writer interested in sci-
of Manitoba. She is research director for the University of ence communication.
Manitoba’s Neonatology fellowship program, and a scientific

viii
SECTION EDITORS

Dr Steven H. Abman is Professor of Pediatrics and Director of and has developed mobile apps of neonatal illustrations and
the Pediatric Heart Lung Center at the University of Colorado infographics.
Anschutz School of Medicine and the Children’s Hospital
Colorado, USA. Having trained in Pediatric Pulmonary and Dr Patrick J. McNamara is a staff neonatologist and Director
Critical Care Medicine, Dr. Abman has had a long-standing of the Division of Neonatology at the University of Iowa Stead
interest in the developing lung circulation and pulmonary Family Children’s Hospital and Professor of Pediatrics and
hypertension in diverse settings, as reflected by his clinical and Internal Medicine, University of Iowa, USA. He is the cur-
laboratory-oriented research. He is currently Director of the rent chair of the PanAmerican Hemodynamic Collaborative,
Pediatric Pulmonary Hypertension Network (PPHNet), Executive Pediatric Academic Society Neonatal Hemodynamics Advisory
Vice-Chairman for the BPD Collaborative and President of the and Neonatal Hemodynamics (TnECHO) Special Interest Group
American Pediatric Society. at the American Society of Echocardiography. His clinical and
research interests include myocardial performance in the set-
Dr Po-Yin Cheung is a neonatologist of the Northern Alberta tings of a hemodynamically significant ductus arteriosus, pulmo-
Neonatal Program of Alberta Health Services and a full Professor nary hypertension, and targeted neonatal echocardiography.
in the Departments of Pediatrics, Pharmacology and Surgery at
the University of Alberta, Canada. He is also an Honorary Clinical Dr William K. Milsom is a comparative physiologist and
Professor of the Department of Paediatrics and Adolescent Emeritus Professor of Zoology at the University of British
Medicine at the University of Hong Kong and Honorary Consultant Columbia, Canada, best known as an authority on the evolu-
at the NICU of the HKU-Shenzhen Hospital. Dr. Cheung is a tion of respiratory processes in vertebrates. He has received the
clinician-scientist and conducts both clinical and basic science highest awards presented by the Canadian Society of Zoologists
research in neonatal hypoxia-reoxygenation injury and neonatal (the Fry Medal), the Comparative and Evolutionary Physiology
transition. Section of the American Physiological Society (the August Krogh
Distinguished Lecture), and the Society for Experimental Biology
Dr Satyan Lakshminrusimha is a neonatologist and the Dennis (UK) (the Bidder Lecture), the three major societies in his disci-
and Nancy Marks Chair and Professor of Pediatrics at the pline. He has also served as the President of the Canadian Society
University of California at Davis, Sacramento, USA. His areas of Zoologists and as President of the International Congress of
of research interest include optimal oxygenation in neonatal Comparative Physiology and Biochemistry. In 2012, Dr Milsom
lung injury, neonatal resuscitation, and pulmonary hyperten- received the Distinguished Service Award from the Canadian
sion. Dr Lakshminrusimha enjoys drawing medical illustra- Society of Zoologists in recognition of contributions to the well-
tions, for which he has developed an international reputation, being of zoology in Canada beyond the call of duty.

ix
CONTRIBUTORS

Steven H. Abman Adrianne Rahde Bischoff


Division of Pulmonary Medicine Department of Pediatrics
Department of Pediatrics University of Iowa Stead Family Children’s Hospital
University of Colorado and Children’s Hospital Colorado Iowa City, IA
Aurora, CO
Jason Boehme
Kurt H. Albertine Department of Pediatrics
Division of Neonatology and Pulmonary University of California, San Francisco
Department of Internal Medicine San Francisco, CA
University of Utah
Salt Lake City, UT R. Dale Brown
Departments of Pediatrics and Medicine
Karel Allegaert Cardiovascular Pulmonary Research Laboratories
Department of Development and Regeneration, Department of University of Colorado
Pharmaceutical and Pharmacological Sciences Anschutz Medical Campus
KU Leuven Aurora, CO
Leuven, Belgium
and Emily Callan
Department of Clinical Pharmacy, Erasmus MC Department of Pediatrics
Rotterdam, The Netherlands Children’s Hospital of Wisconsin
Milwaukee, WI
Gabriel Altit
Division of Neonatology Nicholas R. Carr
Department of Pediatrics Division of Neonatology
McGill University and Montreal Children’s Hospital University of Utah School of Medicine
Montreal, QC, Canada Salt Lake City, UT

Pieter Annaert Aravanan Anbu Chakkarapani


Department of Pharmaceutical and Pharmacological Sciences Neonatology and Clinical Pediatrics
KU Leuven Sidra Medicine and Weill Cornell Medicine
Leuven, Belgium Doha, Qatar
and
Department of Clinical Pharmacy Po-Yin Cheung
Erasmus MC Division of Neonatal-Perinatal Care
Rotterdam, The Netherlands Department of Pediatrics
University of Alberta
Judy L. Aschner Edmonton, AB, Canada
Department of Pediatrics
Hackensack Meridian School of Medicine Shyamala Dakshinamurti
Nutley, NJ Section of Neonatology, Department of Pediatrics
and Department of Physiology
Department of Pediatrics University of Manitoba
Albert Einstein College of Medicine Children’s Hospital Research Institute of Manitoba
Bronx, NY Winnipeg, MB, Canada
and
Joseph M. Sanzari Children’s Hospital Cassidy Delaney
Hackensack University Medical Center Department of Pediatrics
Hackensack, NJ School of Medicine
University of Colorado
Michelle Baczynski Anschutz Medical Campus
Department of Respiratory Therapy Aurora, CO
Mount Sinai Hospital
Toronto, ON, Canada Nolan DeLeon
Children’s Hospital Research Institute of Manitoba
Anjali Y. Bhagirath Winnipeg, MB, Canada
Children’s Hospital Research Institute of Manitoba
Winnipeg, MB, Canada

x
Contributors xi

Yvonne A. Dzal Emilio A. Herrera


Department of Biology and Centre for Forest Interdisciplinary Programa de Fisiopatología
Research Instituto de Ciencias Biomédicas (ICBM)
University of Winnipeg Facultad de Medicina
Winnipeg, MB, Canada International Center for Andean Studies (INCAS)
Universidad de Chile
Germán Ebensperger Putre, Chile
Programa de Fisiopatología
Instituto de Ciencias Biomédicas (ICBM) Allyson Hindle
Facultad de Medicina Integrative Physiology
International Center for Andean Studies (INCAS) University of Nevada
Universidad de Chile Las Vegas, NV
Putre, Chile and
Anesthesia Center for Critical Care Research
Massachusetts General Hospital
Yasser Elsayed
Boston, MA
Section of Neonatology
Department of Pediatrics
Stuart B. Hooper
University of Manitoba
Obstetrics and Gynaecology Monash Health
Winnipeg, MB, Canada
Monash University
Melbourne, Australia
Candice D. Fike
Department of Pediatrics Amish Jain
University of Utah Health Department of Pediatrics
Salt Lake City, UT University of Toronto
Toronto, ON, Canada
Mark K. Friedberg Mount Sinai Hospital
Cardiology Toronto, ON, Canada
The Hospital for Sick Children
Toronto, ON, Canada Robert P. Jankov
Departments of Pediatrics and Cellular and Molecular Medicine
Csaba Galambos University of Ottawa and Children’s Hospital of Eastern Ontario
Pathology Research Institute
Anschutz Medical Campus Ottawa, ON, Canada
Aurora and Children’s Hospital Colorado
Aurora, CO Richard Keijzer
Division of Pediatric Surgery
Harry Gee Department of Surgery
Obstetrics and Gynaecology University of Manitoba and Children’s Hospital Research
Birmingham Women’s Hospital Institute of Manitoba
Birmingham, United Kingdom Winnipeg, MB, Canada

Jason Gien Roberta L. Keller


Neonatal/Perinatal Medicine Department of Pediatrics
University of Colorado University of California
Anschutz Medical Campus San Francisco, CA
Aurora, CO
Stephen T. Kempley
Regan E. Giesinger Royal London Hospital and Barts and the London School of
Department of Pediatrics Medicine
University of Iowa London, United Kingdom
Iowa City, IA
Martin Keszler
Andrew W. Gill Department of Pediatrics
Department of Paediatrics Women and Infants Hospital of Rhode Island
University of Western Australia Providence, RI
Perth, Australia
Afif El Khuffash
M. Ruth Graham Department of Neonatology
Department of Anesthesiology, Perioperative and Pain Medicine The Rotunda Hospital and School of Medicine
University of Manitoba Royal College of Surgeons in Ireland
Winnipeg, MB, Canada Dublin, Ireland
xii Contributors

John P. Kinsella William K. Milsom


Division Pediatrics–Neonatology Department of Zoology
Children’s Hospital Colorado The University of British Columbia
Colorado Fetal Care Center Vancouver, BC, Canada
USC Health–Neonatal Intensive Care Unit
Anschutz and Children’s Hospital Souvik Mitra
Aurora, CO Department of Pediatrics
Dalhousie University and IWK Health Center
Martin Kluckow Halifax, NS, Canada
Neonatology, Department of Pediatrics
University of Sydney and Royal North Shore Hospital Catherine Morgan
Sydney, Australia Division of Pediatric Nephrology
Department of Pediatrics
Girija Ganesh Konduri University of Alberta
Division of Neonatology Edmonton, AB, Canada
Department of Pediatrics
Medical College of Wisconsin and Josef Neu
Children’s Research Institute Division of Neonatology
Milwaukee, WI Department of Pediatrics
University of Florida
Satyan Lakshminrusimha Gainesville, FL
Department of Pediatrics
UC Davis Children’s Hospital Eric B. Ortigoza
Sacramento, CA Division of Neonatal-Perinatal Medicine
Department of Pediatrics
W.L. Amelia Lee UT Southwestern Medical Center
Barts and the London School of Medicine Dallas, TX
London, United Kingdom
Allison S. Osborne
Philip T. Levy Department of Pediatrics
Division of Newborn Medicine Rainbow Babies & Children’s Hospital and Case Western
Boston Children’s Hospital Reserve University
and Cleveland, OH
Department of Pediatrics
Harvard Medical School Christina M. Pabelick
Boston, MA Departments of Anesthesiology and Perioperative Medicine and
Physiology and Biomedical Engineering
Aníbal J. Llanos Mayo Clinic, Rochester
Programa de Fisiopatología Rochester, MN
Instituto de Ciencias Biomédicas (ICBM)
Facultad de Medicina Graeme R. Polglase
International Center for Andean Studies (INCAS) Perinatal Transition Group
Universidad de Chile The Ritchie Centre
Putre, Chile Department of Obstetrics and Gynaecology
Monash University and Hudson Institute of Medical Research
Peter M. MacFarlane Victoria, Australia
Department of Pediatrics
Case Western Reserve University Fabiana Postolow
Cleveland, OH Section of Neonatology
Department of Pediatrics
Emin Maltepe University of Manitoba
Department of Pediatrics Winnipeg, MB, Canada
University of California, San Francisco
San Francisco, CA Y.S. Prakash
Departments of Anesthesiology and Perioperative Medicine and
Patrick J. McNamara Physiology and Biomedical Engineering
Department of Pediatrics and Internal Medicine Mayo Clinic
University of Iowa Stead Family Children’s Hospital Rochester, MN
Iowa City, IA
Contributors xiii

Roberto V. Reyes Dominique Singer


Programa de Fisiopatología Division of Neonatology and Pediatric Critical Care Medicine
Instituto de Ciencias Biomédicas (ICBM) University Medical Center Eppendorf
Facultad de Medicina Hamburg, Germany
International Center for Andean Studies (INCAS)
Universidad de Chile Anne Smits
Putre, Chile Department of Development and Regeneration
KU Leuven
Danielle R. Rios Leuven, Belgium
Clinical Associate Professor Neonatal Intensive Care Unit
University of Iowa University Hospital Leuven
Iowa City, IA Leuven, Belgium

Nicola J. Robertson Robin H. Steinhorn


Perinatal Neuroscience and Neonatology Rady Children’s Hospital
EGA Institute for Women’s Health UC San Diego
University College London San Diego, CA
London, England
Kurt R. Stenmark
Ola D. Saugstad Departments of Pediatrics and Medicine
Department of Pediatrics Cardiovascular Pulmonary Research Laboratories
University of Oslo University of Colorado
Oslo, Norway Anschutz Medical Campus
and Aurora, CO
Ann and Robert H. Lurie Children’s Hospital of Chicago and
Northwestern University Feinberg School of Medicine Edwin W. Taylor
Chicago, IL School of Biosciences
University of Birmingham
Arvind Sehgal Birmingham, United Kingdom
Monash Newborn
Monash Children’s Hospital Payam Vali
Melbourne, Australia Department of Pediatrics
and UC Davis Children’s Hospital
Department of Pediatrics Sacramento, CA
Monash University
Maximo Vento
María Serón-Ferré Department of Pediatrics
Programa de Fisiopatología Health Research Institute La Fe
Instituto de Ciencias Biomédicas (ICBM) Valencia, Spain
Facultad de Medicina
International Center for Andean Studies (INCAS) Stephen Wedgwood
Universidad de Chile Department of Pediatrics
Putre, Chile UC Davis School of Medicine
Sacramento, CA
Divyen K. Shah
Royal London Hospital Dany E. Weisz
Barts and the London School of Medicine Department of Pediatrics
London, United Kingdom University of Toronto
Sunnybrook Health Sciences Centre
Prakesh S. Shah Toronto, ON, Canada
Division of Neonatology, Department of Pediatrics
Mount Sinai Hospital David F. Wertheim
Toronto, ON, Canada Department of Networks and Digital Media
Kingston University
Tatum Simonson London, United Kingdom
Division of Pulmonary, Critical Care, & Sleep Medicine
Department of Medicine Jason Williams
University of California San Diego School of Medicine Department of Biochemistry and Molecular Genetics
San Diego, CA University of Colorado Anschutz Medical Campus, Aurora
Aurora, CO
xiv Contributors

Michelle J. Yang Hui Zhang


Division of Neonatology Departments of Pediatrics and Medicine
University of Utah School of Medicine Cardiovascular Pulmonary Research Laboratories
Salt Lake City, UT University of Colorado Anschutz Medical Campus
Aurora, CO
Bradley A. Yoder
Division of Neonatology – Department Faith Zhu
of Pediatrics Department of Pediatrics
University of Utah University of Toronto
Salt Lake City, UT Toronto, ON, Canada
INTRODUCTION
Shyamala Dakshinamurti, William K. Milsom, Satyan Lakshminrusimha,
Steven H. Abman, Po-Yin Cheung and Patrick J. McNamara

Contents
Part 1: The origins of hypoxia tolerance............................................................................................................................................................................1
Part 2: Fetal hypoxia and neonatal transition...................................................................................................................................................................1
Part 3: Biology of hypoxic respiratory failure in the neonate........................................................................................................................................2
Part 4: Hypoxia and collateral damage..............................................................................................................................................................................2
Part 5: Diagnosis and management of neonatal hypoxic respiratory failure.............................................................................................................3

We have all been hypoxic. run the length of the volume; you may want to read with multiple
We have craved oxygen since the day one of our eukaryotic bookmarks in hand. The diving reflex appears in various forms
ancestors first swallowed an aerobic mitochondrial bacterium in every section, as do cardiorespiratory coupling and reactive
and then, seduced by the prospect of multicellularity, began feed- oxygen species. The introductions to each section of the book are
ing its hungry cytochromes oxygen. gathered below, to highlight their internal connectivity.
Nearly a century ago, Sir Joseph Barcroft postulated “the Everest The past decades have brought substantial advances in the care
in utero,” a fetal environment in the womb comparable to the most of pulmonary hypertension in the newborn. For the sake of our
hostile environmental conditions faced by adventurous mankind. patients and their families, we must learn more, to do better.
Subsequently, expeditions reaching the summit of Everest mapped
out the limits of human tolerance for hypoxemia. Fetal tolerance for Part 1: The origins of hypoxia tolerance
intrauterine hypoxia arises from evolutionarily conserved physiolog-
ical mechanisms, the antecedents of which can be learned from div- William K. Milsom
ing mammals or species at high altitudes. In whimsy we say, the fetus
remembers when it was a turtle or a diving seal; then it is born and The inverse relationship between mass-specific metabolic rate and
forgets. It’s not merely whimsy; several chapters in this book recast body size is well established across all animal taxa. Thus, mammals
Just So Stories in the form of stringently tested comparative physiol- are born with a high demand for oxygen that increases progressively
ogy. The response to hypoxia in animals living in extreme environ- with the development of thermoregulatory capacity. In the human
ments illuminates how the human fetus handles its hypoxemic state, infant, this is accompanied by a loss (or resetting) of tolerance to
and why the human newborn suddenly cannot. The physiological hypoxia. The primary responsibility of the newborn to hypoxia is
changes seen during the transition from intrauterine to extrauter- a reduction in metabolic rate, and this ability also decreases pro-
ine life can be read as retracing the evolution and development of gressively with the development of thermoregulatory capacity.
hypoxia tolerance, critical to biological study and clinical practice Prolonged hypoxia has proinflammatory properties potentially
alike; while postnatal hypoxia creates a multisystem cliffhanger. underlying neurologic vulnerability, particularly with respect to
The conventional terminology “persistent pulmonary hyperten- the respiratory control system. Human populations that reside at
sion of the newborn,” as with the term it once replaced, “persistent altitude in the Himalayas, Andean altiplano, and Ethiopian high-
fetal circulation,” is currently under siege. It is not necessarily persis- lands, however, exhibit a distinct composite of phenotypes, with
tent; it is not only in the newly born; it has bounced from one WHO similarities and differences in various physiological processes lead-
classification category to another while remaining among the most ing to increased hypoxia tolerance. The mechanisms that under-
rapidly progressive vasculopathies. The term “acute pulmonary lie enhanced tolerance can represent instances of acclimatization
hypertension” is more accurate, but it fails to capture the arrest of (phenotypic plasticity), or genetic adaptation (genetic assimilation
pulmonary circuit adaptation as a uniquely neonatal circumstance, of positive traits, or genetic compensation to mitigate maladaptive
triggered by hypoxia, inflammation, or pressure phenomena at a pro- plastic responses to hypoxia). These phenomena are well described
foundly vulnerable moment of development. We, therefore, group in the opening chapters of this section. What follows are chapters
these closely related pathophysiologies under the umbrella defini- describing species in which the fetus, newborn, and/or adult are
tion “hypoxic respiratory failure of the newborn,” examine common extremely hypoxia tolerant. These are species that thrive in hypoxic
pathways of disease, distinguish distinct natural histories and then environments at altitude, in underground burrows, or while breath-
differentiate PPHN (if that’s what we should still call it) from other hold diving. These studies, and our growing knowledge of the phy-
pulmonary hypertensions presenting during infancy. logeny and ontogeny of differences in hypoxia tolerance, inform
This book covers the broad ground: The origins of hypoxia our understanding of the huge physiological shifts of the human
adaptation, the impact of oxygen on the circulatory transition at neonatal transition and the dangers of perinatal hypoxia.
birth, the biochemistry of hypoxia in the pulmonary circulation,
and the diagnosis and clinical management of hypoxic respiratory Part 2: Fetal hypoxia and neonatal transition
failure. In other words: What we once were; why we have been
protected; what happens when that protection is lifted; how to Satyan Lakshminrusimha
diagnose when we have entered the maelstrom; what to do next,
and what we still don’t know. The impact of hypoxia is not limited The current COVID-19 pandemic has highlighted the impor-
to the lung; the “collateral damage” of hypoxia on other systems is tance of maternal oxygenation during pregnancy. The mor-
reviewed, in addition to cardiovascular effects. Common threads bidity and mortality of pregnant women with COVID-19 are

DOI: 10.1201/9780367494018-1 1
2 Hypoxic Respiratory Failure in the Newborn

significantly higher than nonpregnant women. Several pregnant nitric oxide (NO) and prostacyclin (PgI2), and reduced produc-
women with COVID-19 and hypoxemic respiratory failure have tion of potent vasoconstrictors, especially endothelin-1 (ET-1) in
required extracorporeal membrane oxygenation (ECMO). While mediating this dramatic transition of the pulmonary circulation.
the placenta and fetal circulation protect the fetus from deleteri- This past work led to the development of current pharmacologic
ous effects of mild maternal hypoxia, profound hypoxemia results interventions that are commonly used to treat neonatal pulmo-
in hypoxic fetoplacental vasoconstriction and deleterious effects nary hypertension, including inhaled NO, phosphodiesterase
on the fetus. Timely respiratory and cardiovascular support to type 5 inhibitors, diverse forms of synthetic PgI2 analogs and
correct maternal hypoxia is crucial to optimize fetal well-being. ET-1 receptor inhibitors. Despite remarkable success in many
Pulmonary transition at birth is truly a miracle! During fetal settings, PPHN remains associated with substantial morbidity
period, the placenta serves as the organ of gas exchange. All fetal and mortality. In addition, growing recognition of the contribu-
organ systems are functional except the lungs. The fetal lungs are tion of PPHN physiology to hypoxic respiratory failure in preterm
dormant and filled with liquid and has low blood flow, partly due infants has led to further challenges in how to best understand
to hypoxic pulmonary vasoconstriction. The fetus is relatively the underlying pathophysiology and optimal therapies in this
hypoxemic (compared to postnatal standards) but not hypoxic, as fragile population. As a result, more work to better understand
it efficiently delivers oxygen to its tissues with abundant cardiac fundamental mechanisms through which prematurity, antenatal
output and high levels of fetal hemoglobin to compensate for low stress and postnatal injury impair pulmonary vascular, cardiac
PaO2. At birth, with the first cry, air enters the alveoli increas- and respiratory function during the first weeks of life remains an
ing PaO2 resulting in a drop in pulmonary vascular resistance, important target for basic research.
switch in ductal shunt from right-to-left to left-to-right, and an In this section, chapters from outstanding investigators in the
8–10-fold increase in pulmonary blood flow. Expiratory braking field of developmental lung biology, lung vascular disease and
during crying and active absorption of lung liquid enables air to cardiac development, key signaling pathways that are altered in
replace lung liquid in the alveoli. endothelial and smooth muscle cells by hypoxia, molecular biol-
As gas exchange shifts from placenta to the lungs at birth, the ogy of oxygen sensing and injury as related to the generation of
source of left ventricular preload changes from umbilical venous reactive oxygen species, metabolic adaptations from fetal to neo-
return shunting across the oval foramen to pulmonary venous natal life and novel epigenetic mechanisms of hypoxia-induced
return. This switch is gradual (with some much-needed overlap) tissue injury are explored in great detail. Overall, this section
while physiological cord clamping is “delayed” until respira- provides exciting new leads that will yield important translational
tions are established. Abrupt or immediate cord clamping soon insights linking the science of vascular and cardiac function and
after birth, prior to establishing neonatal respiration, can poten- disease with developmental biology, and ultimately, clinical care.
tially lead to hypoxia and bradycardia as the newly born infant
is deprived of both sources of oxygen from placenta and lungs.
Physiological cord clamping is associated with better short-term Part 4: Hypoxia and collateral damage
and long-term outcomes.
Birth asphyxia, defined as failure to establish breathing at Po-Yin Cheung
birth, accounts for 900,000 deaths every year, based on World
Hypoxia is systemic and affects all organs and systems in the neo-
Health Organization estimates, and is one of the leading causes of
nate with hypoxic respiratory failure.
neonatal mortality. Ventilation of the lungs during resuscitation
“Oxygen Radical Disease of Neonatology”, a term originally
with the right concentration of oxygen is the key to minimizing
coined by Professor Ola Saugstad in 1988, remains one of the key
morbidity and mortality from birth asphyxia. While 21% oxygen
concepts in the management of neonates with hypoxic respira-
is adequate for most term infants for initial resuscitation, the
tory failure. Reperfusion or reoxygenation injury remains the
optimal initial oxygen concentration in preterm infants is still an
cornerstone of organ injury and complications. Indeed, oxygen
enigma. Chapters by eminent scientists covering these important
may not be the silver bullet for hypoxia. Coming out of the in
topics are included in Part 2.
utero hypoxic environment, the relative postnatal hyperoxia and
the interplay between oxygen and pathophysiological condi-
Part 3: Biology of hypoxic respiratory tions of the neonate affect the developing respiratory system. As
failure in the neonate a defensive mechanism in the redistribution of blood flow dur-
ing hypoxia, there are differential regional perfusion responses
Steven H. Abman resulting in neuroprotection, intestinal and renal compromise.
Tools for assessing organ perfusion and injury have been increas-
Part 3 of this book addresses biologic mechanisms that contrib- ingly used in the clinical arena. The use of these tools improves
ute to hypoxic respiratory failure in sick newborns. At birth, the the monitoring and management of perfusion deficits. Hypoxic-
lung circulation rapidly responds to birth-related stimuli, includ- ischemic encephalopathy receives the most attention because
ing the sudden rise in alveolar PO2 that leads to the fall in pul- of its significant acute and long-term neurodevelopmental con-
monary vascular resistance and rise in pulmonary blood flow, sequences. While therapeutic hypothermia is the standard care
which is essential for postnatal survival. PPHN, a key component for moderate or severe hypoxic-ischemic encephalopathy of term
of neonatal hypoxic respiratory failure, represents the failure of and near-term neonates, recent reports have fueled the discus-
the lung circulation to undergo sufficient vasodilation, leading sion of its use in other populations. Further, adjunctive therapies
to profound hypoxemia due to marked right-to-left extrapulmo- are emerging to alleviate the cerebral injury. Understanding the
nary shunting across the ductus arteriosus and foramen ovale. pathogenetic course of cerebral injury including biochemical
Past laboratory studies identified the critical role of the enhanced cascades and disruption of the blood-brain barrier helps develop
production of vital endothelium-derived vasodilators, such as therapeutic agents. The kidney is the first important organ that is
Introduction 3

affected by hypoxia. However, similar to the therapeutic agents forms of major congenital heart disease may have a similar presen-
for cerebral injury, clinical treatments cannot reverse, but are tation, which may lead to incorrect treatment choices and adverse
merely limited to the prevention of reno-tubular injury. While patient outcomes. Therefore, immediate access to timely and
the renal injury may heal, the long-term sequelae in neonatal comprehensive echocardiography is imperative to enable accu-
kidneys after hypoxia need further research. Systemic hypoxia rate diagnostic ascertainment and disease/physiology-specific
causes a unique injury to the intestine with ischemia and necro- interventions. Recent advances in echocardiography evaluation,
sis. Hypoxic-ischemic intestinal injury has no clear association and in particular the growth of neonatologist-led hemodynamic
with inflammation or the dysbiosis that is typical for necrotiz- programs, have led to improvements in understanding of the rela-
ing enterocolitis, although the two diseases have similar clinical tionship of pulmonary vascular resistance/pressure to right ven-
and histopathological presentations. An individualized thera- tricular function, the interdependence between both ventricles
peutic approach is the key in precision medicine. It is important and the downstream effects on both pulmonary and end-organ
to better understand clinical pharmacology in hypoxic neonates perfusion. The art of clinical care, and its relevance to manage-
such that we can predict drug-related effects based on drug- and ment, is further emphasized through increased appreciation of
population-specific pharmacokinetics and pharmacodynamics. the importance of mechanical ventilation strategies, optimiza-
Little information is available regarding the interactions between tion of functional residual capacity and the interaction between
drugs and therapeutic hypothermia. Some neonates with hypoxic intrathoracic pressure, ventilation and hemodynamics. It is
respiratory failure will require surgeries and need special anes- imperative that clinicians consider heart-lung interaction as a
thetic considerations. The risk and management of intraoperative biological continuum that is an essential determinant of effective
pulmonary hypertensive crisis require the evaluation of periop- tissue oxygenation and carbon dioxide clearance. Traditionally,
erative risk, evidence-based approaches to respiratory support the terms “persistent fetal circulation” and “persistent pulmonary
and rationalized pathophysiology-driven anesthetic and hemody- hypertension of the newborn” have been used to characterize this
namic management in these neonates. Taken together, collateral clinical syndrome where impaired efficacy of oxygenation is the
effects of hypoxia on the brain, intestine, kidneys and other sys- dominant clinical feature. Several chapters in this section suggest
tems are inter-related in the acute pathophysiology, intercurrent that clinicians should classify the nature of pulmonary hyper-
therapeutic states and long-term outcomes. tension as “acute” or “chronic”, which has both diagnostic and
therapeutic relevance. In addition, both acute and chronic forms
of pulmonary hypertension are distinguished by unique clinical
Part 5: Diagnosis and management of phenotypes that influence the approach to monitoring and treat-
neonatal hypoxic respiratory failure ment choices. The importance of phenotypic characterization
is best demonstrated in patients with congenital diaphragmatic
Patrick J. McNamara hernia, where impaired oxygenation may relate to classic pulmo-
nary arterial hypertension with impaired right heart performance
Part 5 focuses on the approach to diagnosis and management of or may relate to pulmonary venous hypertension secondary to
neonatal hypoxemic respiratory failure, which remains a major a left ventricular phenotype. Timely access to comprehensive
cause of both morbidity and mortality in many parts of the world. echocardiography facilitates enhanced diagnostic precision and
As highlighted in prior sections, the biology of the neonate, dur- the implementation of a disease-specific approach to treatment.
ing the transitional period birth, is at higher risk of impairment Finally, acute pulmonary hypertension is an important consider-
in the normal postnatal fall in pulmonary vascular resistance – ation of hypoxemic respiratory failure in premature infants and
which may lead to impaired efficacy of oxygenation, suboptimal adverse neonatal outcomes; however, timely diagnosis and early
right ventricular function and poor systemic blood flow. One of intervention with targeted pulmonary vasodilator therapy and
the major challenges for clinicians relates to the fact that many heart function support may lead to improved outcomes.
Part 1
The Origins of Hypoxia Tolerance

Edited by William K. Milsom


THE HUMAN FETUS AND METABOLIC ADAPTATIONS TO HYPOXIA
Dominique Singer

Contents
Introduction............................................................................................................................................................................................................................6
Being born as a small endotherm – A delicate challenge..............................................................................................................................................6
Size relationship of metabolic rate................................................................................................................................................................................6
Metabolic cost of temperature regulation...................................................................................................................................................................6
Being satisfied with little – The fetus as a euthermic hibernator.................................................................................................................................7
Hematological adaptations to the low O2 environment...........................................................................................................................................7
Left shift of the O2 dissociation curve....................................................................................................................................................................7
Increase in hemoglobin concentration..................................................................................................................................................................7
Metabolic adaptation to the low O2 environment.....................................................................................................................................................7
Favored gas exchange................................................................................................................................................................................................8
Favored growth rate...................................................................................................................................................................................................8
Favored temperature control...................................................................................................................................................................................8
Staying alive with even less – Oxyconforming responses of the feto-placental unit...............................................................................................8
Metabolic gatekeeper role of the placenta..................................................................................................................................................................8
Metabolic programming of the fetus...........................................................................................................................................................................9
Surviving with a minimum – A deep dive through the birth canal.............................................................................................................................9
Lowering heart rate..........................................................................................................................................................................................................9
Redirecting blood flow....................................................................................................................................................................................................9
Holding breath................................................................................................................................................................................................................10
Summary and conclusions – The mammalian fetus as a paragon of coping with the lack...................................................................................10
References..............................................................................................................................................................................................................................11

Introduction keep their body temperature constant or not. Hence, there must
be a more fundamental explanation that probably involves the
Perinatal hypoxia is one of the greatest, if not the greatest threat to self-adjustment of energy requirements to supply conditions.
human fetuses and newborns. Hypoxic-ischemic encephalopathy Whatever may be the ultimate cause, the overall metabolic rule
(HIE) can result in cerebral palsy and other life-long disabilities. implies that small animals need more food and more oxygen per
Attempts to treat asphyxiated babies by induced hypothermia have unit of body weight and should thus exhibit a worse tolerance to
proven to be fairly effective, yet complementary therapies to fur- starvation and hypoxia than larger species. For mammalian neo-
ther mitigate or even repair HIE are still under investigation (1–3). nates that also fall under this rule (the basal metabolic rate of a
However, HIE would probably occur even more frequently if human term baby amounts to 2.0−2.5 Watts per kg compared to
human fetuses and newborns were not able to protect themselves roughly 1 Watt per kg for adults), this would mean that they are
by a number of physiological mechanisms that show striking inherently maladapted to the risks of undersupply and hypoxia
similarities with natural adaptation strategies to oxygen defi- simply because of their small body size (5–8).
ciency and food scarcity in the animal kingdom. A comparative
physiological analysis of these adaptations may help to better Metabolic cost of temperature regulation
understand both the progression of perinatal hypoxia and the Mammals and birds are endothermic (warm-blooded) animals
diagnostic and therapeutic challenges of HIE. that maintain a higher gradient between body and ambient
temperature than ectothermic (cold-blooded) organisms due to
Being born as a small endotherm an elevated metabolic rate. The specific basal metabolic rate of
mammals is 4(–10) times higher than the resting metabolic rate
– A delicate challenge of reptiles of comparable body size. Endotherms are thus not only
When looking for particularly hypoxia-tolerant creatures, one dependent on a continuous food supply, but they also exhibit a
would actually expect anything but small endotherms. There are lower hypoxia tolerance than ectotherms that can often survive
two main reasons for this. for long periods without any O2 (and food) supply (9–11).
Furthermore, unlike ectotherms that usually tolerate larger
Size relationship of metabolic rate thermal variations (poikilothermy), endothermic animals keep
According to a common biological law, also known as Kleiber’s their body temperature constant (homeothermy). The cold
rule (4), the specific basal metabolic rate (in watts per kilogram) defense reaction includes an increase in metabolic rate with
is higher in small than in large animals (Figure 2.1a). The “allo- decreasing ambient temperatures, which is steeper the smaller
metric” (nonproportional) size relationship of metabolic rate is the body size and the larger the surface-to-volume ratio. In new-
usually explained by the fact that small mammals (or birds) need born mammals, heat is produced by nonshivering thermogenesis
a stronger “internal heater” to compensate for the higher heat (NST) in the brown adipose tissue (BAT), which is based on an
losses caused by their relatively larger body surface area. However, uncoupling of oxidative phosphorylation and thus accompanied
a similar relationship applies to all living beings whether they by a high O2 consumption rate. Thus, newborn babies experience

6 DOI: 10.1201/9780367494018-2
The Human Fetus and Metabolic Adaptations 7

FIGURE 2.1 Metabolic size relationship as a target of perinatal and seasonal metabolic adaptations. (a) Following an overall bio-
logical rule (“mouse-to-elephant curve”), the specific metabolic rate increases with decreasing body mass. (b) The mammalian fetus,
however, behaves more like an organ of its mother. The “disproportionately” low metabolic rate favors tissue oxygenation in spite of
low intrauterine O2 tensions. (c) A similar metabolic adaptation is found in hibernating mammals that exhibit a uniform minimal
metabolic rate that equals the basal metabolic rate achieved by the largest mammals based on body size alone.

a higher thermometabolic stress than adults and are known to be 19 mmHg as opposed to approximately 28 mmHg in human
at higher risk of hypothermia, due to the limits of thermoregula- adults). The markedly increased O2 affinity results in the fact that
tion being reached earlier (12–14). at a pO2 between 25 and 30 mmHg, the mean O2 saturation in the
All in all, small mammals have particularly high energy fetal circulation amounts to 65−70% rather than 50% as would
requirements both due to their small body size and thermoregu- be expected, under comparable ambient conditions, in unaccli-
latory properties, making them particularly susceptible to condi- mated adults.
tions of undersupply. From this point of view, it is not surprising
that over the some 150 million years of mammalian evolution, Increase in hemoglobin concentration
both the intrauterine development and – above all – the process Second, the increase in hemoglobin concentration (up to roughly
of being born have been optimized by a number of self-protective 18−20 g/dl in term neonates as opposed to 13−15 g/dl in adults). This
mechanisms (15–17). is a kind of high-altitude acclimatization responding to the fact that
despite its higher O2 affinity, the O2 saturation of the fetal hemoglo-
bin is still well below the almost 100% in oxygenated adult blood.
Being satisfied with little – The The two adaptive mechanisms ensure that the total O2 con-
fetus as a euthermic hibernator tent of fetal blood is in the order of adult blood which is often
misinterpreted as if the low pO2 in the fetal circulation was fully
The intrauterine environment might be imagined as a Garden of
compensated. However, this is not true, since – independently of
Eden where everything is available in abundance. However, this
the amount of O2 carried by the red cells – the driving force for
is not the case. In particular with respect to O2, the fetus has to
the diffusion of gases is partial pressure. Hence, even the afore-
cope with scarcity even under normal developmental conditions.
mentioned adaptive responses cannot prevent the O2 from being
The mean O2 partial pressure in the fetal circulation amounts
“pressed” into the fetal tissues under a much lower tension than
to 25−30 mmHg and thus corresponds to the arterial blood gas
in adults. This would inevitably affect tissue oxygenation if there
values that have been measured in extreme mountaineers climb-
were not another adaptive mechanism.
ing on the top of the world without additional oxygen (“Everest
in utero”). The exceptionally low O2 tension has previously been Metabolic adaptation to the low O2 environment
thought to reflect a worse gas exchange capacity of the placenta The key to understanding this additional adaptation is Warburg’s
as compared to the lung. Meanwhile, it is assumed that (given law (20), stating that the “critical depth” (of penetration of O2 into
the immaturity of O2 free radical detoxification systems) Mother tissue by diffusion) does not only depend on the partial pressure
Nature deliberately put the mammalian fetus in a hypoxic com- gradient from outside to inside but also on the rate at which O2 is
partment to prevent it from O2 toxicity (18, 19). consumed by the tissue. The lower the O2 consumption rate, the
Hematological adaptations to the low O2 environment higher the penetration depth, or in other words: the adverse effect
Even though these ambient conditions are normal during intra- of a lowered partial pressure can be counteracted by a reduced
uterine life (and should therefore be referred to as “low-oxygen” tissue respiration rate.
rather than as “hypoxic” conditions), the fetus has to compensate This is the background for a widely underestimated self-
for the reduced pO2 to cover its O2 needs. As described in many protective mechanism to be found in mammalian fetuses, namely
physiological textbooks, this compensation primarily consists of the suppression of the usual metabolic size relationship. In fact,
two complementary hematological mechanisms. the general rule that the specific metabolic rate increases with
decreasing body mass (“mouse-to-elephant curve”) seems to be
Left shift of the O2 dissociation curve somehow “switched off” during intrauterine life (Figure 2.1b).
First, the well-known left shift of the O 2 dissociation curve of fetal From a metabolic point of view, the mammalian fetus behaves
hemoglobin (down to a half-saturation pressure of approximately more “like an organ of its mother,” with the metabolic increase
8 Hypoxic Respiratory Failure in the Newborn

up to the level expected from body size occurring only after birth Favored temperature control
(6, 21–23). Third, both marsupial neonates and mammalian fetuses can
The deviation from the usual metabolic size relationship “afford” the suppression of metabolic size relationship because
was first described by Hasselbalch (24) in avian embryos and they are passively thermostated, be it in the maternal abdomi-
explained by the fact that if their energy turnover was as high nal pouch or in the womb, and therefore do not need a stronger
as expected from their small body size, the diffusion capacity “internal heater” to compensate for higher heat losses over their
of the egg shell would not be high enough to cover the resulting relatively larger body surface area. In the case of the mammalian
O2 demand (25). Unlike in avian embryos, metabolic measure- fetus in particular, it could even be that with an appropriately
ments in mammalian fetuses are methodologically difficult and high metabolic rate, the heat transport capacity of the placenta
accordingly rare, starting from the first observations by Bohr (21, might not be sufficient to remove the excess heat from the amni-
26–30). Since then, it has been repeatedly shown that mamma- otic cavity (28).
lian (including human) neonates still have a disproportionately Remarkably, the intrauterine metabolic reduction is the only
low specific O2 consumption rate immediately after birth, before exception to the general metabolic size relationship apart from
rising more or less rapidly to the metabolic level appropriate to hibernating mammals that fall to a uniform minimum specific
their own body size (22, 31–33). As was first observed by Brück metabolic rate that corresponds to the specific basal metabolic
(34) in his pivotal studies on the metabolism of human term and rate of the very largest mammals and might thus reflect a com-
preterm neonates, and later confirmed by our own respiromet- mon limit to metabolic reduction (Figure 2.1c) (40–42). This also
ric measurements (35), the earliest postnatal metabolic rates are applies to black bears that give birth to their offspring while in
remarkably independent of gestational age and birth weight and hibernation and thus provide a “missing link” between intra-
thus reflect the suppression of metabolic size relationship during uterine and seasonal adaptations (43). Similar to fetuses, at least
intrauterine life. some hibernating mammals exhibit a decreased (venous) blood
Another indirect and often overlooked sign of intrauter- pO2 (44, 45), suggesting that a low-oxygen atmosphere could be
ine metabolic reduction in humans is heart rate. After a sharp a common permissive factor of metabolic reduction both in the
increase in the first trimester, the fetal heart rate levels off natural overwintering strategies and in the “euthermic hiberna-
at about 140 beats per minute, where it remains more or less tion” of mammalian fetuses.
unchanged until the expected date of birth (36). This is signifi-
cantly lower than would be expected in adult mammals of com-
parable size, especially in the earlier stages of pregnancy (e.g. a
Staying alive with even less –
100 g mammal corresponding in body weight to a human fetus
at 16 weeks of gestation would have a heart rate of around 400 Oxyconforming responses of
beats per minute) (5, 37). Since the heart rate directly parallels the feto-placental unit
the metabolic rate, the “inappropriately” low heart rate clearly
reflects the metabolic suppression that adapts the human fetus Even though the fetus is already adapted to the low-oxygen
to its low-oxygen habitat. atmosphere through hematological and metabolic adaptations,
The protective effect of the reduced metabolic rate can be there is still a substantial adaptive reserve, in that a number
illustrated in marsupial mammals that are physiologically born of exogenous and endogenous conditions are tolerated with-
in an extremely immature state and spend most of their fetal out jeopardizing the pregnancy as a whole (46). This applies
development in their mother’s abdominal pouch. According to to high-altitude pregnancies, to food scarcity and famines, to
our own studies on Monodelphis neonates, tiny creatures of 100 pregnancy disorders with impaired placental perfusion, and to
mg weight and 1 cm length, these animals show no postnatal cases of severe fetal anemia. In terms of O2 supply, a further
metabolic increase at all and maintain a metabolic level that in reduction by approximately 50% (e.g. in highland pregnancies
their case amounts to only 20% of what would be expected in up to 4000–4500 m of altitude; or in fetal anemias down to 9–8
an adult marsupial of comparable size (35). This has three major g/dl of serum hemoglobin) is usually tolerable without major
implications. adverse effects, except for a mild-to-moderate Intra-Uterine
Growth Restriction (IUGR). This adaptive reserve is due to a
Favored gas exchange coordinated response of the feto-placental unit that consists of
First, it compensates for the scarce O2 supply, which in their case two main factors.
is not due to an “insufficient” placenta, but to a very immature
lung (38). As has been shown by Mortola and coworkers (39) in Metabolic gatekeeper role of the placenta
a slightly different marsupial species, a considerable part of the Recent findings indicate that the placenta plays an important
O2 uptake in these animals during their first days of life occurs gatekeeper role in the allocation of energy flows. The basic
via skin respiration – which would be unimaginable without a premise here is that only 60% of the oxygen supplied to the
unique combination of small size and disproportionately low feto-placental unit is passed on to the fetus, while 40% is con-
metabolic rates (6). sumed by the placental tissue itself. With reduced O2 delivery,
the placenta appears to reduce its own O2 consumption in order
Favored growth rate to maintain fetal O2 supply (Figure 2.2a). The metabolic reduc-
Second, the low maintenance metabolism allows for a high tion that is accompanied by a decrease in mitochondrial density
growth efficiency despite a necessarily limited substrate supply. impairs active transport and synthetic processes (thus resulting
In the aforementioned marsupial species, an increase to 500% of in a more or less pronounced IUGR), yet prevents the fetus from
birth weight within the first 10 days of life was observed, although a critical O2 deficit. It seems that maintaining adequate oxygen-
neither their milk intake nor the caloric content of the milk was ation has priority over unrestricted growth in intrauterine life
exceptionally high. (29, 30, 47, 48).
The Human Fetus and Metabolic Adaptations 9

FIGURE 2.2 Self-protective responses of the feto-placental unit in mammals. (a) Since the placenta itself consumes up to 40% of
the total O2 supplied to the feto-placental unit, it can prevent the fetus from hypoxia by reducing its own metabolic rate. (b) The fetus
is able to compensate for the resulting lack of substrate supply by “refraining” from growth in favor of maintenance metabolism. (c)
A critical O2 deficiency leads to a redistribution of blood flow from the body shell to the central organs (brain-sparing effect, diving
reflex). (d) The gradual adjustment of metabolic demand to energy supply reflects an oxyconforming response that differs from the
usual oxyregulatory behavior of mammalian tissues.

Metabolic programming of the fetus dips or decelerations in the cardiotocographic (CTG) monitoring.
The fact that the metabolic rate in mammalian fetuses is lower They differ from the stress response an adult would show in the
than expected from body size also means that the growth metab- case of suffocation, namely an acceleration in heart rate and an
olism accounts for a relatively high proportion of the total energy increase in cardiac output to maintain an adequate O2 supply to
turnover. This in turn offers the fetus the opportunity to adapt to the tissues. Since this typical oxyregulatory response enables the
an inadequate supply by “refraining” from growth (Figure 2.2b). organism to escape or fight a threatening situation, it has proven
Accordingly, IUGR that from a clinical perspective is usually con- to be an obviously successful adult survival strategy in the course
sidered as a pathological symptom of impaired supply, can be inter- of evolution. Its major disadvantage, however, is that under the
preted as a physiological adaptation that enables the fetus to survive conditions of an already limited O2 supply, the O2 demand will
a further shortage despite an already limited O2, and substrate avail- increase even further. As the fetus completely relies on energy sup-
ability. The earlier the underlying metabolic changes start and the ply via the umbilical cord, any attempt to fight against the O2 lack
longer they last, the more the fetal organism is programmed to “low would be futile and would only lead to a reduced hypoxia tolerance
flame,” meaning that even a normal supply of substrate acts as a sur- due to the increased O2 consumption rate. It is therefore reason-
plus and thus promotes a metabolic syndrome later in life (49–51). able from an evolutionary perspective that the fetus responds to
The placental decrease in O2 consumption in response to an acute (perinatal) hypoxia with a different physiological reaction
decreasing O2 supply, also referred to as hypoxic hypometabo- that is called the “diving response”, by analogy to aquatic mam-
lism (52, 53), is an “oxyconforming” response that differs from the mals (seals) that exhibit a similar pattern during longer periods
usual “oxyregulatory” behavior of mammalian tissues that tend to of submersion (59–61). This comprises three main components.
maintain their metabolic rate until final breakdown (Figure 2.2d).
It has long been assumed that oxyconformism is confined to lower Lowering heart rate
vertebrates (frogs) or even invertebrates (intertidal worms) that A cornerstone of the diving response in both aquatic mammals
are able to adapt to changing supply conditions by adjusting their and mammalian fetuses is the gradual decrease in heart rate.
metabolic rate more flexibly (54, 55). Recent studies suggest that Given that the myocardium, together with the brain, accounts for
not only the placenta but also the fetus itself shows an oxycon- a significant proportion of total O2 consumption in the fetal life,
forming behavior in that its own metabolic rate underlies some bradycardia per se makes a significant contribution to reducing
variations depending on the fluctuating O2 tensions in the fetal energy requirements.
circulation (30). This may reflect, among other things, the varying Redirecting blood flow
contribution of growth metabolism to the overall metabolic rate. Since the slowed heart rate results in a reduced cardiac output,
However, it is also related to a redistribution of blood flow that the an adequate supply of the organism would no longer be possible if
fetus exhibits in response to deteriorating O2 supply conditions there were no redistribution of blood flow. That’s why the diving
(brain-sparing effect), and that may be regarded as a precursor of reflex involves a centralization of the circulation in favor of the
the birth-related diving response (Figure 2.2c) (56, 57). vital organs (heart, brain). The temporary reduction of peripheral
perfusion leads to an accumulation of lactate in peripheral tissues
Surviving with a minimum – A deep and a subsequent washout after re-emergence. As pointed out by
dive through the birth canal Scholander in a landmark paper (62), the post-diving lactate peak
in seals is similar to the transient lactate increase that is observed
Whenever the fetus is at risk of suffering a severe O2 deficiency in human neonates immediately after birth, and that, inciden-
during the birth process (58), a reaction occurs that is very well- tally, makes it difficult to draw a clear line between a beneficial
known to the entire delivery room staff as a red flag: the so-called self-protective response and an actual perinatal asphyxia.
10 Hypoxic Respiratory Failure in the Newborn

Holding breath rate. Most of them are known from adaptations to a predictable
Not unlike at deep rest, all spontaneous fetal motor activity is (e.g. seasonal) undersupply in the animal kingdom (53, 60, 70)
also suppressed under stress, so as to avoid any unnecessary suggesting that, vice versa, the perinatal period might even act
energy expenditure. This includes the intermittent respiratory as a common ontogenetic source of adaptive responses among
movements the fetus is known to exhibit, just to prepare the dia- mammals.
phragm for its life-long activity. Only when a critical degree of With regard to perinatal adaptation, it should be emphasized
hypoxia is reached, the apnea is interrupted by serial gasps that, that the self-protective mechanisms are arranged in a cascade-
if occurring in the birth canal, can lead to a meconium aspira- like manner and exhaust themselves gradually (Figure 2.3).
tion syndrome. Once the baby is born, the gasps provide the This means that the more of them have already been activated
blood with a minimum of O2, just enough to maintain the greatly in utero (e.g., in a growth-restricted fetus), the fewer are left at
slowed heart rate for a while (“autoresuscitation”) (63–65). birth. The cumulative protective benefit is only effective as long as
As a whole, the diving response results in a slower consump- the gap between supply and demand can be narrowed by reduc-
tion of the remaining O2 reserves (that, both in fetuses and div- ing demand to an indispensable minimum. This also means that
ing mammals, are elevated by a high hematocrit). The clinical neonatal hypoxia tolerance is more a “resistance” than a “toler-
appearance of a “depressed” neonate with apnea, bradycardia, ance” in its strict sense, delaying the onset of critical hypoxia
and reduced peripheral perfusion actually corresponds to the rather than attenuating the harmful effects of O2 lack. In fact,
full picture of the diving reflex. Just as in a seal taking a deep there is little, if any, evidence that neonatal tissues really “toler-
breath after re-emergence at the water surface, a rapid increase in ate” hypoxia better than adult ones do. Although lactate acidosis
the baby’s APGAR values from the first to the fifth minute of life plays an important role in the assessment of perinatal asphyxia,
reflects a rapid recovery from a self-protective response (66, 67). an overall (enzymatic) increase in the anaerobic capacity, i.e. the
In case of persisting hypoxia of any cause, the diving reflex will be ability to extract energy from lactic acid fermentation, has never
maintained and completed by a suppression of thermogenesis in been proven in neonatal tissues (71).
the O2- and pH-sensitive BAT which, while preventing an adverse What is unique about placental, fetal, and neonatal tissues
thermoregulatory increase in metabolic rate, increases the risk of is their ability to “deliberately” reduce their metabolic rate
inadvertent hypothermia (28, 52, 68, 69). in response to decreased energy supply – a capability often
misinterpreted as a low metabolic trait. In adult mammals
experiencing a gap between energy demand and supply, counter-
Summary and conclusions – The mammalian regulatory responses (tachycardia) are initiated to compensate
fetus as a paragon of coping with the lack for the reduced supply (oxyregulatory behavior). If these are not
successful, a “passive” breakdown in metabolic rate occurs – as
In summary, mammalian, including human, fetuses and new- if a light bulb goes out as soon as the battery is exhausted. An
borns are equipped with a number of self-protective mechanisms alternative way to respond to shortened supply is to “actively”
that prevent them from the risks of intrauterine and perinatal life. reduce demand (oxyconforming behavior) – as if a light bulb is
Since the most important of these risks is a temporary lack of provisionally dimmed in view of the imminent exhaustion of the
O2 and nutrient supply, being in contrast to the particularly high battery. The phenomenological similarity between active and
metabolic demands of small endotherms, it is not surprising that passive reduction in energy consumption (the light bulb becomes
these mechanisms are mainly based on a reduction in metabolic darker) partly explains the clinical problems in determining the

FIGURE 2.3 Cascade-like arrangement of self-protective mechanisms in mammalian fetuses and neonates. The declining curves
represent the “passive” metabolic breakdown resulting from an imbalance between O2 demand and supply. Whenever the metabolic
rate falls below a critical lower limit, this results in irreversible damage. However, any “active” reduction in metabolic rate leads to a
gradually slowed metabolic breakdown and an accordingly enhanced hypoxia tolerance (schematic view, arbitrary units).
The Human Fetus and Metabolic Adaptations 11

boundaries between protective reactions and signs of damage in 15. Mortola JP. Respir Physiol. 1999; 116:95–103.
16. Singer D. Comp Biochem Physiol A. 1999; 123:221–234.
perinatal asphyxia. 17. Singer D. Anästhesiol Intensivmed Notfallmed Schmerzther. 2002; 37:441–460.
Animal experiments on newborn mammals have revealed 18. Ar A, et al. Israel J Zool. 1994; 40:307–326.
19. Reich B, et al. Dev Neurosci. 2016; 38:311–330.
that their extra hypoxia tolerance is the more pronounced the
20. Warburg O. Biochem Z. 1923; 142:317–333.
smaller and more immature they are at birth (72, 73). In addition 21. Rahn H. A companion to animal physiology. Taylor CR, Johansen K, Bolis L, eds.
to a suppression of thermogenesis, a reduction of the metabolic Cambridge: Cambridge University Press; 1982. pp. 124–137.
22. Wieser W. Respir Physiol. 1984; 55:1–9.
rate at normal body temperature and thus a hypoxic hypome- 23. Singer D, et al. Comp Biochem Physiol A. 2007; 148:780–784.
tabolism was identified as the essential mechanism (15, 52, 24. Hasselbalch AK. Skand Arch Physiol. 1900; 10:353–402.
74). Interestingly, however, this ability (obviously some kind of 25. Paganelli CV, et al. In: Respiration and metabolism of embryonic vertebrates. Seymour
RS, ed. Dordrecht: Dr W Junk Publishers; 1984. pp. 193–204.
transient return to the fetal metabolic level) is lost more or less 26. Bohr C. Skand Arch Physiol. 1900; 10:413–424.
quickly, much in parallel with the postnatal increase in metabolic 27. Dawes GS, et al. J Physiol. 1959; 146:295–315.
28. Schröder HJ, et al. Exp Physiol. 1997; 82:403–414.
rate and its link to body size (23, 75). It may be that some of the
29. Murray AJ. Placenta. 2012; 33:e16–e22.
natural adaptation strategies (e.g., hibernation) imply a tempo- 30. Postigo L, et al. J Physiol. 2009; 587:693–708.
rary disabling of this link in a low-oxygen environment. 31. Hill JR, et al. J Physiol. 1965; 180:239–265.
32. Bauer K, et al. J Pediatr. 2003; 142:390–396.
Finally, the optimization of the birth process over millions 33. Bauer J, et al. Am J Clin Nutr. 2009; 90:1517–1524.
of years of mammalian evolution may explain the difficulties of 34. Brück K. In: Perinatal physiology, 2nd. Stave U, ed. New York: Plenum; 1978.
finding effective treatments for HIE. It must be assumed that pp. 455–498.
35. Singer D. Thermochim Acta. 1998; 309:39–47.
nature has left out nothing that could have contributed to reduc- 36. DuBose TJ. Fertil Steril. 2009; 92:e57–e58.
ing vulnerability. External cooling as an attempt to reduce the 37. Lindstedt SL, et al. Quart Rev Biol. 1981; 56:1–16.
38. Szdzuy K, et al. J Anat. 2008; 212:164–179.
newborn’s metabolic rate differs from the endogenous metabolic
39. Mortola JP, et al. Nature. 1999; 397:660.
suppression occurring in natural adaptations. However, the lat- 40. Geiser F. J Comp Physiol. 1988; 158B:25–37.
ter is only effective as long as the hypoxic state persists. Should 41. Singer D, et al. In: Surviving hypoxia: mechanisms of control and adaptation. Hochachka
PW, Lutz PL, Sick T et al., eds. Boca Raton FL: CRC Press; 1993. pp. 447–458.
it eventually be possible to activate the “fetal hibernation gene” 42. Heldmaier G, et al. Respir Physiol Neurobiol. 2004; 141:317–329.
even after a hypoxic-ischemic event, this might be of huge ben- 43. Tøien, et al. Science. 2011; 331:906–909.
efit not only for asphyxiated neonates and adult stroke or cardiac 44. Musacchia XJ, et al. Am J Physiol. 1971; 221:128–130.
45. Revsbech IG, et al. J Comp Physiol B. 2017; 187:847–856.
arrest patients. 46. Bennet L. J Physiol. 2017; 595:1865–1881.
In the meantime, it is essential to make use of the time margins 47. Carter AM. Placenta. 2000; 21(Suppl A):S31–S37.
48. Schneider H. Placenta. 2000; 21(Suppl A):S38–S44.
built into the birth process through a cascade of self-protective
49. Godfrey KM, et al. Public Health Nutr. 2001; 4:611–624.
mechanisms. This offers the opportunity to identify perinatal 50. Rinaudo P, et al. Annu Rev Physiol. 2012; 74:107–130.
risks in time and to avert them by taking appropriate obstetric 51. Myers DA, et al. Adv Exp Med Biol. 2014; 814:147–157.
52. Mortola JP. Respir Physiol Neurobiol. 2004; 141:345–356.
and/or neonatological measures. 53. Gorr TA. Acta Physiol. 2017; 219:409–440.
54. Boutilier RG, et al. J Exp Biol. 1997; 200:387–400.
55. Buchner T, et al. Comp Biochem Physiol B. 2001; 129:109–120.
References 56. Jensen A, et al. Eur J Obstet Gynecol Reprod Biol. 1999; 84:155–172.
57. Cohen E, et al. Neonatology. 2015; 108:269–276.
1. Shankaran S. Curr Opin Pediatr. 2015; 27:152–157. 58. Turner JM, et al. Am J Obstet Gynecol. 2020; 222:17–26.
2. Davidson JO, et al. Front Biosci. 2018; 23:2204–2226. 59. Elsner R, et al. Monogr Physiol Soc. 1983; 40:1–168.
3. Gunn AJ, et al. Handb Clin Neurol. 2019; 162:217–237. 60. Hill RW, et al. Animal physiology. Sunderland (Mass): Sinauer; 2004.
4. Kleiber M. The fire of life: an introduction to animal energetics. New York: Wiley; 1961. 61. Panneton WM. Physiology. 2013; 28:284–297.
5. Schmidt-Nielsen KC. Cambridge: Cambridge University Press; 1984. 62. Scholander PF. Sci Am. 1963; 209:92–106.
6. Singer D. Respir Physiol Neurobiol. 2004; 141:215–228. 63. Sanocka UM, et al. J Appl Physiol. 1992; 73:749–753.
7. Singer D. Thermochim Acta. 2006; 446:20–28. 64. Fewell JE. Respir Physiol Neurobiol. 2005; 149:243–255.
8. Bejan A, et al. Design in nature. New York: Anchor Books; 2012. 65. Manole MD, et al. Pediatr Res. 2006; 60:174–179.
9. Belkin DA. Science. 1963; 139(3554):492–493. 66. Casey BM, et al. N Engl J Med. 2001; 344:467–471.
10. Suarez RK, et al. Am J Physiol. 1989; 257:R1083–R1088. 67. Iliodromiti S, et al. Lancet. 2014; 384:1749–1755.
11. Else PL, et al. Physiol Biochem Zool. 2004; 77:950–958. 68. Malan A, et al. J Comp Physiol B. 1988; 158:487–493.
12. Okken A, Koch J, eds. Thermoregulation of sick and low birth weight neonates. Berlin: 69. Jayasinghe D. Neonatology. 2015; 107:220–223.
Springer; 1995. 70. Larson J, et al. J Exp Biol. 2014; 217:1024–1039.
13. Agren J. Fanaroff & Martin’s neonatal-perinatal medicine: Diseases of the fetus 71. Jones CT, et al. Physiol Rev. 1985; 65:357–430.
and infant, 10th. ed. Martin RJ, Fanaroff AA, Walsh MC, eds. Philadelphia: Elsevier 72. Himwich HE, et al. Am J Physiol. 1941; 133:327–339.
Saunders; 2015. pp. 502–512. 73. Lutz PL, et al. Comp Biochem Physiol B Biochem Mol Biol. 1996; 113:3–13.
14. Singer D, et al. Innovations and frontiers in neonatology (Pediatr Adolesc Med, vol. 22); 74. Rohlicek CV, et al. J Appl Physiol. 1998; 84:763–768.
Basel: Karger; 2020. pp. 95–111. 75. Mourek J. Physiol Bohemoslov. 1959; 8:106–111.
HYPOXIA AS A NEUROINFLAMMATORY STIMULUS DURING DEVELOPMENT

Peter M. MacFarlane and Allison S. Osborne

Contents
Fetal “hypoxia” as a stimulus for development...............................................................................................................................................................13
Inflammation........................................................................................................................................................................................................................13
Hypoxia as an inflammatory stimulus.............................................................................................................................................................................14
Effects of pro-inflammatory mediators on respiratory control..................................................................................................................................14
Hypoxia, inflammation, and breathing......................................................................................................................................................................14
Sensitivity of carotid body chemoreceptors to inflammatory mediators...........................................................................................................15
Sensitivity of brainstem respiratory neurons to inflammatory mediators.........................................................................................................15
Peripheral-to-central inflammatory transmission.........................................................................................................................................................15
Sepsis and other pro-inflammatory insults.....................................................................................................................................................................16
Critical windows of vulnerability to inflammatory insults..........................................................................................................................................16
Vulnerability in the fetal-neonatal transition – Postnatal loss of hypoxia tolerance.............................................................................................16
Conclusions...........................................................................................................................................................................................................................17
Acknowledgments...............................................................................................................................................................................................................17
References..............................................................................................................................................................................................................................17

Fetal “hypoxia” as a stimulus Inflammation


for development Inflammation is a physiological response of the immune system
The fetal environment is relatively hypoxemic compared to the triggered by a wide variety of insults including injury, pathogens,
extra-uterine air-breathing one, and the “Oxygen Cascade” and toxins, and even excessive or deficient O2 exposure (6). Both
describes the O2 gradients and associated transport mechanisms infectious and noninfectious insults activate immune cells that
from the atmosphere to mitochondria (1). The atmospheric par- initiate inflammatory signaling pathways; these responses are
tial pressure of O2 (PO2) at sea level, which is perceived post- intended to be beneficial through the removal of harmful material
natally as “normoxia,” is ∼160 mmHg. The PO2 of the umbilical and activation of the healing process (7). On the other hand, uncon-
artery that returns fetal blood to the placenta is ∼40 mmHg and trolled or chronic inflammatory responses can have inadvertent
the fetal arterial PO2 (PaO2) reaches ∼30 mmHg (2). The placen- effects and, therefore, there are both benefits and consequences
tal environment, therefore, is classically categorized as relatively associated with immune system activation (8). Abnormalities
hypoxic from an atmospheric air-breathing standpoint. However, in inflammatory responses have been proposed in autism spec-
it is a necessary “normoxic” state for the fetus especially since trum disorders (9), fragile X syndrome (10), sudden infant death
O2 supply to the tissue is adequate and does not necessitate syndrome (SIDS), and fetal inflammatory syndrome (11, 12).
anaerobic metabolic pathways to support energy demands. The Responses are also vastly diverse and can vary by location (e.g.
O2 demand of the fetus is low compared to the newborn infant cell/tissue-specific, peripheral [systemic], central [CNS]), dura-
primarily because two significant energy-demanding processes tion, age, and nature of the insult or pathogen. Although immune
are minimal – thermogenesis is low because it is provided and inflammatory activation mechanisms are complex, a simpli-
maternally, and the infrequent fetal breathing movements also fied characterization could generally be categorized as a response
minimize energy costs. Growth and development comprise the that involves activation of immune cells (peripherally – leukocyte
predominant energy demands of the fetus, and the intrinsic recruitment; centrally – microglia, astrocytes) and/or release of
O2 levels comprise a necessary stimulus to guide appropriate pro-inflammatory cytokines/­chemokines (6). While the profile of
development. The “hypoxic” embryonic environment stimulates cytokine/chemokine responses are also complex, increased expres-
trophoblast cell differentiation and placentation during early sion, synthesis, and release of pro-inflammatory cytokines IL-1β,
pregnancy, while in later stages of pregnancy, the low-O2 ten- IL-6, and TNFα are commonly recognized.
sions promote organ development including airway branching, Inflammatory responses can also involve increased (transcrip-
lung morphogenesis, and angiogenesis (3, 4). Cellular (adap- tional) cellular signaling of MAP kinases, NF-kB, JAK, and STAT
tive) responses to hypoxia are largely modulated by transcrip- pathways, but discussion of the complexities of these responses
tional activity involving hypoxia-inducible factor signaling (5). are beyond the scope of this chapter. Changes in endogenous O2
However, there are of course limits below which abnormally levels (particularly hypoxia) can also activate immune responses
­low-O2 availability (e.g. severity of hypoxia) can have unfortunate leading to inflammation. In this context, the severity/intensity,
consequences for the developing organism and the vulnerability duration (acute, chronic), and pattern (continuous/sustained,
to hypoxia is age- and tissue-specific. Given the susceptibility of intermittent/recurrent) of O2 exposure can be expected to deter-
exposure to hypoxic insults during development, in this chapter, mine the characteristics of the immune/inflammatory response.
we highlight the pro-inflammatory properties of hypoxia and the The effect of hypoxia in the context of an inflammatory stimulus
ways in which it can affect various elements of the respiratory can impact individuals of all ages particularly the fetus and devel-
neural control system during development. oping neonate.

DOI: 10.1201/9780367494018-3 13
14 Hypoxic Respiratory Failure in the Newborn

Hypoxia as an inflammatory stimulus and disproportional head/body size (26–28). Of interest is that
the carotid body chemoreceptors play an important role in this
A variety of clinical scenarios are associated with fetal hypoxia fetal response to hypoxia, since it is largely blunted following
including umbilical cord compression, pre-eclampsia, maternal carotid body denervation (29). The effects of hypoxia and immu-
anemia, substance abuse, multiple pregnancies, chorioamnio- nosensing capabilities of the carotid body are discussed later.
nitis, gestational diabetes, and sleep-disordered breathing (13). Physiologically, hypoxia during development manifests in many
The fetus is fairly well-equipped with various physiological adap- forms and the subsequent inflammatory response can vary with
tations to defend against excessive or inadvertent hypoxia. The age and as a function of hypoxia (e.g. acute, chronic sustained
higher fetal hemoglobin affinity, ability to shunt blood prefer- [CSH], and intermittent hypoxia [CIH]).
entially to vulnerable organs, bradycardia, and implementation
of hypoxic-hypometabolism are important defense mechanisms
to avoid ischemic injury, most of which are largely lost postna- Effects of pro-inflammatory
tally. However, there are limits to the functional benefits of these mediators on respiratory control
hypoxia defense strategies especially with prolonged/chronic
exposure. The concept of fetal origins of adult disease has been Following birth, the increase in arterial PO2 has a profound effect
postulated for several decades (14) and a large body of evidence on the respiratory control system. The rapid oxygenation leads
has arisen from studies focused on the effects of fetal nutritional to resetting of the O2-sensing properties of carotid bodies, the
compromise and stress. More recent studies have shed light on primary peripheral chemoreceptors responsible for initiating the
developmental reprogramming following fetal hypoxia, which can ventilatory defense response to hypoxemia, or HVR (an index of
lead to cardiovascular and metabolic susceptibility later in post- respiratory control [dys]function, pertaining to the capacity to
natal life (15). Delineating the basic mechanisms has been chal- mount an increase in minute ventilation to defend against hypox-
lenging, but emerging evidence has revealed pro-inflammatory emia). The respiratory system rapidly adapts to the postnatal envi-
properties of hypoxia at multiple ages. In adults suffering from ronment, and over several days (animal studies, see Bavis (30) for
mountain sickness, increased serum levels of IL-6, IL-6 receptor, a review), or weeks (human infants), the HVR then becomes more
and C-reactive protein are some markers of inflammation (16) in robust and begins to mature (31), although the HVR is blunted
association with physiological effects of altitude hypoxia includ- in preterm infants (32). A similar phenomenon probably explains
ing vascular leakage, pulmonary, and cerebral edema (17, 18). the blunted HVR in infants born at high altitude (33). In rodents,
Hypoxia can also amplify the innate immune response to invad- chronic postnatal hypoxia blunts the HVR and delays its matura-
ing pathogens by recruiting adaptive immune cells; inflamma- tion largely through a disruption in the resetting of carotid body
tory pathologies in which tissue hypoxia has been documented O2 sensitivity (34–36) and impairment of carotid body growth
include inflammatory bowel disease, ischemia, cancer, obesity, and development. Since hypoxia has pro-inflammatory proper-
and arthritis (19). ties resulting in increased expression of various cytokines such as
However, rather than hypoxia manifesting as a consequence IL-1β, IL-6, and TNFα in multiple brain regions, it is pertinent to
of inflamed tissue, it can also activate inflammatory responses discuss the effects of inflammation on the peripheral and central
through O2-sensitive signaling pathways such as hypoxia- neural circuitry controlling breathing.
inducible factor (HIF) in a variety of immune cell types (20).
Microglia and astrocytes are the main CNS inflammatory media- Hypoxia, inflammation, and breathing
tors and are responsive to hypoxia (21). They are the resident Two of the most prominent examples in which IH stimulates an
immune cells of the CNS; their inflammatory role is complex inflammatory response is sleep apnea (adolescents and adults) and
and has adverse effects on the respiratory neural control system, apnea of prematurity (preterm infants) (20, 37). IH is a distinctly
particularly during development (discussed in more detail later). different stimulus than chronic-sustained hypoxia because of the
Microglial activation increases microglia motility by releasing repetitive reoxygenation and, therefore, IH likely involves a com-
cytokines and chemokines, which also play a role in the removal bination of both an inflammatory and oxidative stress response.
of cellular debris and phagocytosis of synapses as well as modula- In preterm infants, the incidence of IH events commonly associ-
tion of neurotransmitters critically involved in respiratory neuron ated with apnea of prematurity increases over the first 2 post-
excitability (22). Previous studies have demonstrated increased natal weeks (38), which has also been associated with increased
microglia expression in specific brainstem cardiorespiratory serum oxidative stress markers (39). In the in vitro brainstem slice
control regions following neonatal hypoxia, resulting in altered preparation of neonatal rats, IH-induced increases in respiratory
neurotransmitter expression and respiratory control dysfunc- rhythmic activity were attenuated by the microglia inhibitor
tion (23). Hypoxia has similar effects on various brain autonomic minocycline, which implies an inflammatory-mediated stimu-
regions in adult rats leading to increased inflammatory mediators lation of breathing (40). In a rat model of CIH exposure, blood
IL-1β, IL-6, MMP9, and TNFα (24). In neonatal mice, a transient cytokine levels were elevated in both serum and brain (specifi-
anoxic event was sufficient to induce microsomal prostaglan- cally brainstem) (41). In pregnant women, IH occurs more fre-
din E synthase-1 (mPGES-1) activity (25). In the same study, the quently, which can lead to developmental disturbances in fetal
C-reactive protein was correlated with elevated PGE2 in cere- somatic growth leading to reduced birthweight (42) as well as pre-
bral spinal fluid, which was associated with increased apnea term delivery, NICU admissions, and intrauterine growth restric-
frequency. Finally, in utero, fetal brain sparing in which cardio- tions (43). Pregnant rats exposed to CIH caused impairments in
vascular responses to hypoxia include increased carotid (i.e. ventilatory defense responses to acute hypoxic challenge in their
brain) blood flow relative to peripheral regions (e.g. hindlimbs) offspring (44). In contrast, early postnatal CIH enhances the HVR
is a mechanism intended to protect the CNS from hypoxic injury (45, 46). Also of interest is that gestational IH increases suscep-
(13). The long-term impact of this response, however, manifests tibility of the offspring to subsequent inflammatory insults (47)
as asymmetrical growth restriction, small for gestational age, suggesting reprogramming of fetal immune responses.
Hypoxia as a Neuroinflammatory Stimulus 15

The carotid body chemoreceptors contain specialized cells to Sensitivity of brainstem respiratory
detect changes in arterial oxygenation (both hypoxia and hyper- neurons to inflammatory mediators
oxia) (48). Type I glomus cells contain the O2-sensing machinery Hypoxia and various pro-inflammatory cytokines/chemokines
that results in the release of various neurotransmitters as part of also affect brainstem respiratory neural control regions. IL-1β
the afferent hypoxemic stimulus that gets transmitted into brain- injections into the fourth ventricle increased c-Fos activity in
stem respiratory control regions, specifically the nucleus tractus tyrosine hydroxylase-expressing neurons of the nTS, the brain-
solitarius (nTS). Peripheral hypoxic signals are received in nTS stem integrative site of vagal (and carotid body) afferent inputs
via the vagus nerve where the inputs are integrated and transmit- (61). Similarly, in a neonatal rat model of sepsis, nTS neuron
ted to other brainstem regions to stimulate an increase in minute excitability was increased (62). CSH increased the excitability
ventilation (i.e. the HVR) (48). Type II cells of the carotid body of nTS neurons, which was suspected to involve a disturbance
are sustentacular supportive cells and resemble glial cells, which in astrocyte-neuron interactions (63). Neonatal rats exposed to
could imply the carotid body itself has inflammatory responsive CSH followed by CIH had increased spontaneous bursting of nTS
capabilities. Many of these components involved in respiratory neurons (64), although neonatal CIH alone depressed nTS neu-
control have demonstrated sensitivity to inflammatory cyto- ron activity through a mechanism related to reduced numbers
kines/chemokines and, therefore, result in a functional effect on of active synapse (65). While the reduced activity of nTS neurons
respiratory output. IL-1β administration decreased respiratory could translate into a depressant effect on breathing, increased
frequency, hypoxic gasping, and autoresuscitation, a mechanism excitability could be a compensatory response to reduced carotid
related to activation of prostaglandin E2 (PGE2) pathways (25). body inputs, or a form of over-compensation (64). Increased
PGE2 administration to neonatal rats resulted in unstable breath- excitation of brainstem neurons could also reflect loss of effi-
ing and increased sigh-related apneas (49). In the latter case, these cient neurotransmission and, therefore, an inability to respond
effects could be prevented by pre-treatment with caffeine and an to hypoxia resulting from excessive glutamate release and cyto-
adenosine receptor antagonist. Caffeine has anti-inflammatory toxicity (66). PGE2 administration, which binds to EP3 receptors,
properties and is widely used clinically in preterm infants to decreased respiratory rhythm generation and increased apnea in
mitigate apnea (50). Intraperitoneal injection of IL-1β decreased brainstem spinal cord preparation of neonatal mice (25). Acute
respiration in neonatal mice and also worsened anoxic survival hypoxia has direct effects on the activity of preBotzinger (pre-
(25). IL-1β also attenuated the ventilatory defense response to BotC) neurons, a pre-motor respiratory nucleus critical for gen-
acute hypercapnic challenge in neonatal mice (51), whereas PGE2 erating inspiratory rhythm. The initial responses of a single cell
depressed breathing in movements in several species including PreBotC neuron to acute hypoxia reflects the biphasic ventilatory
humans (25, 52, 53). The effects of individual inflammatory ele- response of newborns, comprising an initial increase in inspira-
ments on breathing could be mediated at multiple sites of the tory bursting, followed by a rapid decline over several minutes
respiratory control system. (67). However, in vivo neonatal CIH increases irregularity of
respiratory rhythm of these same neurons (68) and enhances
Sensitivity of carotid body chemoreceptors respiratory frequency following in vitro IH, which was attenuated
to inflammatory mediators by microglial inhibitors (40). Overall, there is evidence demon-
The presence of the glial-like sustentacular cells of the carotid strating pro-inflammatory effects on excitability of neurons in
body makes it a promising candidate for having immunosens- both central and peripheral respiratory control regions.
ing properties (54). Chronic hypoxia upregulates expression of
proinflammatory cytokines in the adult carotid body. As little as
3 days of hypoxia (10% FIO2) increased IL-1β, IL-6, TNFαR1, and Peripheral-to-central
their cytokines in the carotid body and some were localized to inflammatory transmission
glomus cells (55). Further, exogenous application of bath-applied
IL-1β, IL-6, and TNFα to dissociated glomus cells enhanced the Immunoresponsive cells including macrophages, dendritic
hypoxic-induced intracellular calcium response (55, 56), whereas cells, and B cells located in the spleen and lymphatic system
IL-1β administration caused glomus cell depolarization (57). In are some of the “first responders” to peripheral detection of
contrast, TNFα application decreased carotid body sensitivity to foreign or pro-inflammatory challenges (69). A large portion of
acute hypoxia (54), suggesting different cytokines/chemokines the immune response includes cell proliferation and secretion
could have differential effects on carotid body excitation, which of pro-inflammatory cytokines often leading to a large-scale,
could depend on interactions with other stimuli. CIH-induced system-wide inflammatory cascade. There are several proposed
increases in carotid body sensory activity were associated with mechanisms by which the peripheral pro-inflammatory response
increased TNFα and IL-1β (58, 59). However, although ibupro- can be transmitted to the CNS including activation of endothe-
fen treatment had anti-inflammatory effects, it failed to decrease lial cells, leading to direct release of other cytokines into the
the augmented sensory responses, suggesting mechanisms CNS. Cytokines acting indirectly across the blood-brain barrier
underlying carotid body potentiation might not necessarily be include circulating IL-1β activation of IL-1 receptors located on
linked to increased inflammatory expression. In vivo studies on vascular endothelial cells of the BBB, leading to increased COX-2
anesthetized adult rats also showed exogenous/topical applica- and microsomal PGE1 activity (25, 70).
tion of IL-1β to the carotid body increased carotid sinus nerve More recently, the novel concept of localized peripheral CNS
activity, which was blocked with an IL-1 receptor antagonist inflammatory responses in regions that receive afferent/peripheral
(57). Similarly, topical application of LPS to the carotid body also nerve inputs has gained traction. Effects on peripheral nerves and
caused tachypnea in anesthetized rats (54). Collectively, these associated tissue can transmit afferent signals to corresponding
data support the immunosensing capabilities of the carotid body brain regions that initiate localized CNS inflammatory responses.
both under generalized inflammatory environments and follow- How this takes place is unknown, but could involve direct trans-
ing hypoxia (60). port of peripheral signals (e.g. cellular proteins or content), using
16 Hypoxic Respiratory Failure in the Newborn

the nerve as a physical conduit into the CNS, or through modi- of vulnerability, the risk/outcome associated with an environ-
fication of electrical activity (sensory denervation or increased mental challenge may be loss of homeostasis, injury, or death
excitation). From a respiratory control standpoint, modulation rather than the induction of phenotypic or adaptive plasticity,
of brainstem inflammation via the afferent origins of the vagus which would normally be intended to be beneficial (80). There
nerve is a prominent example. Intravenous administration of LPS are windows of development that are revealed by natural changes
increased c-FOS expression in the nTS, which was blunted follow- in respiratory control. The HVR doubles in magnitude between
ing ipsilateral cervical vagotomy (71). Loss or diminished periph- P10 and P15 (81), despite a transient loss of an effective HVR on
eral inputs can also cause localized CNS effects. Unilateral nodose day P13 (82). Such large swings in respiratory control capabilities
ganglionectomy increased OX-42 expression in the ipsilateral, but coincide with a time of rapid and in some cases transient changes
not contralateral, side of the NTS, DMNV, and nucleus ambiguous in brainstem neurochemistry (83–86). For example, glutamate,
(NA), which was prevented with the microglia inhibitor minocy- GABA, glycine, and 5-HT receptor expression change dramati-
cline (72). The localized response on the ipsilateral side adds fur- cally in brainstem cardio-respiratory control regions during the
ther support for a CNS method of inflammatory responsiveness via second week of life in the rat (∼P12–P13) (83, 87). Similar unique
alterations in peripheral inputs. This concept offers novel avenues and transient changes in various components of the ECM also
to explore therapeutic targets using compounds (or technology, e.g. take place during postnatal development (88). An extensive dis-
nerve stimulation) that aren’t required to cross the BBB, which can cussion on the role of the ECM in brain development is beyond
often have otherwise unintended nonspecific effects. the scope of this chapter; however, various ECM components
interact with microglia and in some cases also comprise impor-
Sepsis and other pro-inflammatory insults tant inflammatory responses leading to altered neurotransmit-
ter expression (e.g. 5-HT). Constitutive expression of TNFα and
Common causes of neonatal inflammation include infection iNOS mRNA increases transiently at P10 whereas TLR4 increases
from sepsis, pneumonia, and meningitis. Sepsis and associated at ∼3 weeks of age in brainstem (89). Similarly, there is a transient
irreversible organ dysfunction are the major contributing factor reduction in brain metabolites during acute hypoxia that occur
in infant morbidity and mortality, particularly in preterm infant in 9- to 13-day-old rats (90), suggesting key neurodevelopmen-
populations. tal events related to brain energy metabolism. However, these
Neonatal infections account for ∼1/3 of infant deaths (73) and changes appear to represent necessary events required for archi-
increase apnea in preterm infants. In utero, chorioamnionitis is a tectural organization for appropriate brainstem development.
common cause of prematurity, and neonatal infections are asso- Such critical periods also define a time when developmental plas-
ciated with meningitis, intraventricular hemorrhage, and white ticity can be elicited, although they can also comprise transient
matter damage including periventricular leukomalacia (74). periods during which the organism is particularly vulnerable to
These conditions, in turn, are associated with long-term neuro- environmental insults (80).
developmental impairments including cerebral palsy (75, 76). LPS This is especially the case with respect to pro-inflammatory
is commonly used experimentally as a potent pro-inflammatory/ challenges including prolonged (days) hypoxia and acute LPS
endotoxic insult and has also been used to mimic sepsis (54). It exposure. CSH during these key neurochemical adjustments
is a glycolipid portion of gram-negative bacterial surface mol- increased microglia expression and decreased 5-HT immuno-
ecule that initiates a potent inflammatory response largely via reactivity in two distinct brainstem cardio-respiratory control
activation of toll-like receptors. In adult cats, LPS infusion caused regions, the nTS, and DMNV (23). These effects of CSH were asso-
tachypnea, tachycardia, and hypotension, although the effects on ciated with an unexplained high mortality that was not observed
ventilation were absent following carotid body denervation (54). in younger or older rats exposed to CSH (23, 91) and could be
LPS also increased basal frequency of carotid body discharge sug- prevented with the microglia inhibitor minocycline. These data
gesting an excitatory effect even in the absence of hypoxia (54). indicate that the heightened vulnerability during the critical win-
LPS application to the in vitro brainstem spinal cord prepara- dow of development may involve a microglia-mediated disruption
tion of ∼2-day-old rats decreased respiratory burst frequency, to brainstem neurochemical development. Similarly, LPS had a
but the magnitude of the depressant effect of LPS was reduced in similar lethal effect during the same critical window of develop-
rats preconditioned with gestational IH, a model aimed to mimic ment (89). LPS administration to neonatal rats close to the time
maternal IH associated with sleep apnea in pregnancy (47). point of these neurochemical adjustments also caused a uniquely
The possibility that in utero insults can re-program CNS elevated degree of mortality. In this case, the mortality could be
inflammatory processes in a way that modifies subsequent (e.g. predicted in rats by prior assessment of their HVR, which offers
postnatal) challenges is largely under-investigated, yet likely some insight into the association between the lethality of inflam-
has fundamentally important implications for infant mortal- matory insults and disturbances in respiratory control. How
ity and morbidity. More recent studies have begun to shed light these windows of development compare to humans is not clear,
on inflammatory responses to substance of abuse. Animal stud- although it should be noted that there are multiple windows of
ies have demonstrated increased CNS cytokine expression and postnatal vulnerability to pro-inflammatory insults, especially
glial activation/neuroinflammation following prenatal exposure surrounding birth.
to opioids (77) and alcohol (78) and postnatally to nicotine (79).
Vulnerability in the fetal-neonatal transition
Critical windows of vulnerability – Postnatal loss of hypoxia tolerance
to inflammatory insults
With normal transition to extrauterine life, lung fluid clears and
The terms critical window of plasticity and critical period of vul- the newborn’s first breaths begin to establish the lung’s func-
nerability refer to time periods of heightened sensitivity to envi- tional residual capacity, concurrent with a rapid drop in pulmo-
ronment insults or challenges. However, during a critical period nary vascular resistance (PVR) and increase in pulmonary blood
Hypoxia as a Neuroinflammatory Stimulus 17

flow. Clamping of the umbilical cord removes the low-resistance instrumental in advancing the clinical care of (particularly) pre-
placenta from the circuit of systemic blood flow and increases term infants.
systemic blood pressure. Together with the drop in PVR, this
decreases the right-to-left shunt across the ductus arteriosus,
which further increases the PaO2 and stimulates closure of the Acknowledgments
ductus. Finally, the other physiologic right-to-left shunt (the
Supported by grants from US National Institutes of Health R01
foramen ovale) closes as pulmonary blood flow increases and
HL056470 and R01 HL138402.
left atrial pressure exceeds the right side. PaO2 rises quickly
to ∼35–40 mmHg within a few minutes of birth, increasing to
∼60–65 mmHg within an hour. These physiological adjustments References
associated with birth promote postnatal viability in an O2-rich
1. Weibel ER, et al. Respir Physiol. 1981; 44(1):151–164.
air-breathing environment. 2. Meschia G In: Maternal-fetal medicine: principles and practice. Creasy R, ed;
While the low fetal O2 levels provide a necessary stimulus Philadelphia: Saunders; 1989.
3. Soares M, et al. Birth Defects Res. 2017; 109(17):1309–1329.
to encourage normal growth, there is also an apparent and 4. Gebb SA. Adv Exp Med Biol. 2003; 543:117–125.
unexplained loss of tolerance to hypoxia immediately follow- 5. Semenza GL. Wiley Interdiscip Rev Syst Biol Med. 2010; 2(3):336–361.
ing birth. Rats born into hypoxia, which would in part mimic a 6. Chen L, et al. Oncotarget. 2017; 9(6):7204–7218.
7. Nathan C, et al. Cell. 2010 Mar 19; 140(6):871–882.
postnatal continuation of fetal hypoxia, resulted in unexplained 8. Chertov O, et al. Immunol Rev. 2000; 177:68–78.
mortality (92). The mortality occurred precipitously at ∼2 weeks 9. Mitchell RH, et al. J Am Acad Child Adolesc Psychiatry. 2014; 53(3):274–296.
10. Higashimori H, et al. Hum Mol Genet. 2013; 22(10):2041–2054.
of age, but not when initiation of hypoxia was delayed for a few
11. Vennemann MM, et al. Int J Legal Med. 2012; 126(2):279–284.
days after birth. This could imply that the postnatal hypoxia 12. Ferrante L, et al. Hum Immunol. 2008; 69(6):368–373.
intolerance is initiated by events immediately surrounding, or 13. Giussani DA. J Physiol. 2016; 594(5):1215–1230.
14. Barker DJ. J Int Med. 2007; 261(5):412–417.
directly associated with birth (93). While the underlying mech- 15. Giussani DA, et al. J Dev Orig Health Dis. 2013; 4(5):328–337.
anisms of loss of hypoxia tolerance are speculative, the mecha- 16. Hartmann G, et al. Cytokine. 2000; 12(3):246–252.
nisms may be O2-sensitive, since being born into hypoxia would 17. Semenza GL. Science. 2007; 318(5847):62–64.
18. Hackett PH, et al. N Engl J Med. 2001; 345(2):107–114.
have eliminated the sudden hyperoxic stimulus. One conse- 19. Eltzschig HK, et al. N Engl J Med. 2011; 364(7):656–665.
quence of birthing into hypoxia is the resultant delay in normal 20. Taylor CT, et al. J Clin Invest. 2016; 126(10):3716–3724.
21. Bilbo S, et al. Front Behav Neurosci. 2009; 3:14.
postnatal processes including resetting of carotid body hypoxia
22. Bilbo SD, et al. Front Neuroendocrinol. 2012; 33(3):267–286.
sensing (36). Carotid body denervation can be lethal, especially 23. MacFarlane PM, et al. J Physiol. 2016; 594(11):3079–3094.
in neonates (94), suggesting that carotid body activity may pro- 24. Silva TM, et al. Exp Physiol. 2018; 103(6):884–895.
25. Hofstetter AO, et al. Proc Natl Acad Sci USA. 2007; 104(23):9894–9899.
vide necessary inputs into brainstem cardiorespiratory control 26. Soria R, et al. Pediatr Res. 2013; 74(6):633–638.
regions (95). Whether postnatal hypoxia-induced inflammation 27. Halliday HL. Best Pract Res Clin Obstet Gynaecol. 2009; 23(6):871–880.
of the carotid bodies plays a role in the vulnerability could be 28. McMillen IC, et al. Reproduction. 2001; 122(2):195–204.
29. Giussani DA, et al. J Physiol. 1993; 461:431–449.
speculated; although at a minimum, these data suggest early 30. Bavis RW. Respir Physiol Neurobiol. 2005; 149(1–3):287–299.
postnatal survival is critically dependent on peripheral carotid 31. Williams BA, et al. J Physiol. 1991; 442:81–90.
32. Gerhardt T, et al. Pediatrics. 1984; 74(1):58–62.
bodies (96).
33. Gamboa A, et al. J Appl Physiol (1985). 2003; 94(3):1255–1256.
The postnatal hypoxia intolerance is also true for preterm 34. Eden GJ, et al. J Physiol. 1987; 392:11–19.
infants, which are prematurely expelled from their hypoxic in- 35. Eden GJ, et al. J Physiol. 1987; 392:1–9.
36. Sterni LM, et al. Am J Physiol. 1999; 277(3 Pt 1):L645–L652.
utero environment. A meta-analysis combined data from five 37. Ryan S, et al. Circulation. 2005; 112(17):2660–2667.
clinical trials that randomized preterm infants into either a low 38. Di Fiore JM, et al. J Pediatr. 2012; 161(6):1047–1052.
target range of baseline arterial O2 saturation (85–89%) (i.e. mim- 39. Shah VP, et al. Neonatology. 2020:1–7.
40. Camacho-Hernández NP, et al. Respir Physiol Neurobiol. 2019; 265:9–18.
icking very mild CSH) or a higher range (91–95%) starting within 41. Snyder B, et al. Physiological Reports. 2017; 5(9):e13258.
24 hours of birth (97). One unexpected and concerning finding 42. Loube DI, et al. Chest. 1996; 109(4):885–889.
43. Ding XX, et al. Sleep Breath. 2014; 18(4):703–713.
was a higher mortality rate in the low target (ie. relatively hypoxic)
44. Gozal D, et al. Am J Respir Crit Care Med. 2003; 167(11):1540–1547.
group compared to the high target group of infants. However, 45. Reeves SR, et al. Pediatr Res. 2006; 60(6):680–686.
within the low target group of infants, there was an increased risk 46. Julien CA, et al. Respir Physiol Neurobiol. 2011; 177(3):301–312.
47. Johnson SM, et al. Respir Physiol Neurobiol. 2018; 256:128–142.
of death in infants who received the low oxygen <6 hours after 48. Feldman JL, et al. Annu Rev Neurosci. 2003; 26:239–266.
birth, which was not seen in infants who received low oxygen >6 49. Mitchell LJ, et al. Am J Physiol Regul Integr Comp Physiol. 2020; 319(2):R233–R242.
hours after birth (97). These data add further support to the con- 50. Abu-Shaweesh JM, et al. Semin Fetal Neonatal Med. 2017; 22(5):342–347.
51. Siljehav V, et al. J Appl Physiol (1985). 2014; 117(9):1027–1036.
cept of a postnatal loss of hypoxia tolerance, particularly when it 52. Kitterman JA, et al. J Appl Physiol Respir Environ Exerc Physiol. 1983; 54(3):687–692.
occurs closer to the time of birth. 53. Tai TC, et al. Am J Physiol Regul Integr Comp Physiol. 2000; 278(6):R1460–R1473.
54. Zapata P, et al. Respir Physiol Neurobiol. 2011; 178(3):370–374.
55. Lam SY, et al. Histochem Cell Biol. 2008; 130(3):549–559.

Conclusions 56. Fan J, et al. J Neurosci Res. 2009; 87(12):2757–2762.


57. Shu HF, et al. Eur J Neurosci. 2007; 25(12):3638–3647.
58. Rio D, et al. Adv Exp Med Biol. 2012; 758:199–205.
Overall, antenatal hypoxia exposure manifests in many forms, 59. Del Rio R, et al. Eur Respir J. 2012; 39(6):1492–1500.
60. Gauda EB, et al. Respir Physiol Neurobiol. 2013; 185(1):120–131.
patterns, and severities, which can depend on associated clinical
61. DeBoer MD, et al. Peptides. 2009; 30(2):210–218.
or pathological conditions. Hypoxia clearly has pro-inflammatory 62. Eftekhari G, et al. Shock. 2020; 54(2):265–271.
properties affecting both peripheral and central mechanisms of 63. Accorsi-Mendonça D, et al. J Neurosci. 2015; 35(17):6903–6917.
64. Mayer CA, et al. Respiratory physiology & neurobiology. 2015; 205:28–36.
respiratory neural control. An understanding of the mechanis- 65. Almado CE, et al. J Neurosci. 2012; 32(47):16736–46.
tic basis for the vulnerabilities associated with hypoxia exposure, 66. Zhang W, et al. Am J Physiol Regul Integr Comp Physiol. 2008; 295(5):R1555–R1562.
particularly during critical windows of development, may help 67. Garcia AJ 3rd, et al. Respir Physiol Neurobiol. 2019; 270:103259.
68. Garcia AJ 3rd, et al. Front Neuroscience. 2016; 10:4.
to tease out underlying features associated with postnatal loss 69. Chaplin DD. J Allergy Clin Immunol. 2010; 125(2 Suppl 2):S3–S23.
of hypoxia tolerance. Key breakthroughs in these areas will be 70. Ek M, et al. Nature. 2001; 410(6827):430–431.
18 Hypoxic Respiratory Failure in the Newborn

71. Hermann GE, et al. Am J Physiol Regul Integr Comp Physiol. 2001; 280(1):R289–R299. 85. Liu Q, et al. Neuroscience. 2010; 165(1):61–78.
72. Roulston CL, et al. Neuroscience. 2005; 135(4):1241–1253. 86. Wong-Riley MT, et al. Respir Physiol Neurobiol. 2005; 149(1-3):83–98.
73. Black RE, et al. Lancet. 2010; 375(9730):1969–1987. 87. Liu Q, et al. J Comp Neurol. 2010; 518(7):1082–1097.
74. Dammann O, et al. Ment Retard Dev Disabil Res Rev. 2002; 8(1):46–50. 88. Stryker C, et al. Am J Physiol Regul Integr Comp Physiol. 2018; 314(2):R216–R227.
75. Heep A, et al. Arch Dis Child Fetal Neonatal Ed. 2003; 88(6):F501–F504. 89. Rourke KS, et al. Respir Physiol Neurobiol. 2016; 232:26–34.
76. Moscuzza F, et al. Gynecol Endocrinol. 2011; 27(5):319–323. 90. Jensen F, et al. Brain Res Dev Brain Res. 1993; 73(1):99–105.
77. Jantzie LL, et al. Brain Behav Immun. 2020; 84:45–58. 91. Mayer CA, et al. J Appl Physiol (1985). 2014; 116(5):514–521.
78. Noor S, et al. Front Immunol. 2018; 9:1107. 92. Mortola JP Respiratory physiology of newborn mammals. Baltimore, MD: Johns Hopkins
79. Peixoto TC, et al. Neuroscience. 2019; 418:69–81. University Press; 2001. 334 p.
80. Bavis RW, et al. Exp Neurol. 2017 Jan; 287(Pt 2):176–191. 93. Mitchell L, et al. Respir Physiol Neurobiol. 2020; 273:103318.
81. Ohtake PJ, et al. Am J Respir Crit Care Med. 2000; 162(3 Pt 1):1140–1147. 94. Serra A, et al. J Appl Physiol (1985). 2001; 91(3):1298–1306.
82. Liu Q, et al. J Appl Physiol. 2009; 106(4):1212–1222. 95. MacFarlane PM, et al. Respir Physiol Neurobiol. 2013; 185(1):170–176.
83. Liu Q, et al. J Appl Physiol. 2002; 92(3):923–934. 96. Donnelly DF, et al. J Appl Physiol (1985). 1990 Mar; 68(3):1048–1052.
84. Liu Q, et al. J Appl Physiol. 2005; 98(4):1442–1457. 97. Askie LM, et al. JAMA. 2018; 319(21):2190–2201.
HUMAN ADAPTATIONS TO HIGH ALTITUDE

Tatum Simonson

Contents
Introduction..........................................................................................................................................................................................................................19
Phenotypic differences among highland populations...................................................................................................................................................19
Evidence for genetic adaptations to high altitude.........................................................................................................................................................20
Genetic adaptation in Tibetan highlanders..............................................................................................................................................................21
Genetic adaptation in Andean highlanders..............................................................................................................................................................21
Genetic adaptation in Ethiopian highland populations.........................................................................................................................................21
Cross-population and cross-species insights into adaptive genetic factors.............................................................................................................22
Functional insights into genetic adaptation....................................................................................................................................................................22
References..............................................................................................................................................................................................................................22

Introduction Phenotypic differences among


Populations have resided in the Himalayas, Andean Altiplano,
highland populations
and Ethiopian highlands for hundreds of generations. Present- One of the most extensively examined traits among highlanders
day highlanders on each continent exhibit a distinct composite is hemoglobin concentration, which varies markedly between
of phenotypes, with similarities and differences noted in various Tibetan and Andean populations (10). Tibetans’ relatively lower
physiological assessments, including but not limited to hema- hemoglobin concentration at high altitude (comparable to values
tological parameters, ventilatory control, and birth weight. In observed at sea level) is associated with greater exercise capacity
addition to these physiological assessments, recent large-sale (11) and reproductive success (12). Some Ethiopian populations
genomic efforts aim to understand how genetic factors contribute also exhibit relatively lower hemoglobin concentrations at high
to such different responses despite comparable environmental altitude (13–15), although additional associations remain to be
stresses. Investigation into -omic-level signatures (e.g. epigenom- determined. Lower hemoglobin concentration in Tibetans has
ics, transcriptomics, proteomics, and metabolomics) provides an been attributed to a reduced erythropoietic response to hypox-
additional layer of information regarding the genetic pathways emia. However, hemoglobin mass is elevated in Sherpa compared
involved in responses to hypoxia and promising insights into to lowlanders, and Sherpa has a larger plasma volume and com-
generational as well as short-term pathway responses to hypoxia. parable total blood volume at a lower hemoglobin concentration
The term adaptation, specifically in reference to highland relative to Andeans, which may minimize high-viscosity-related
populations, is distinct from more immediate changes that may damage of microcirculatory blood flow (16).
occur in response to an environmental challenge over a relatively Increased hemoglobin-oxygen-binding affinity has been noted
brief period of time (i.e. acclimatization). In highland popula- among various high-altitude species, and may provide benefit at
tions, adaptations have occurred by means of increased survival altitude through an improved rate of oxygen equilibration across
and/or ability to reproduce (1, 2). Genome-wide scans of selec- the alveolar-capillary barrier. While many studies provide vary-
tion, which search for outlier adaptive patterns in the genome, are ing conclusions among highland populations examined, recent
especially powerful given the strong selective pressure imposed studies in Andeans (17) and Tibetans (18) indicate that these
by hypoxia over thousands of years and provide important clues populations exhibit a reduced P50 at high altitude.
to human adaptations to high altitude (1). Another hallmark difference between populations is in rest-
The thousands of years that populations have lived at alti- ing ventilation, which is higher in many Tibetans compared
tude provide sufficient time for selection to occur and for the to Aymara Andeans as is the hypoxic ventilatory responses
frequency of adaptive alleles to increase in the population. The (HVR), which varies much more in Tibetans relative to mini-
Tibetan highlands have been occupied for 30,000 and possibly mal responses observed among Andeans (19). However, most
40,000 years (3), more than double the amount of time popula- studies report that arterial oxygen saturation is lowest among
tions have resided in the Andes (∼12,000 to 14,000 years ago) (4) Himalayan highlanders, slightly higher in Andeans, and highest
with notable European contribution to the gene pool nearly 500 in Amhara Ethiopians (19, 20). Tibetan infants, relative to Han
years ago (5). While the history of various Ethiopian populations Chinese or Andean infants, have higher oxygen saturation, which
at high altitude involves migrations into and out of the highland is hypothesized to underlie susceptibility to high altitude pulmo-
regions throughout the past 70,000 years, more recent migrations nary hypertension and chronic mountain sickness in Andeans
are noted as recent as 500 years ago (6–9). The number of genera- (21). The potential for hypoxia’s long-term effects are also noted
tions a population has spent at altitude is important to consider in among individuals with excessive erythrocytosis, who were more
studies of adaptation as distinct evolutionary histories, and ensu- likely to have been born to mothers with hypertension and peri-
ing phenotypes, have been uniquely shaped in each population. natal hypoxia (22).

DOI: 10.1201/9780367494018-4 19
20 Hypoxic Respiratory Failure in the Newborn

Recent studies suggest that the effects of hypoxia during preg- Evidence for genetic adaptations
nancy are proportional to the hypoxia severity (23), and at alti-
tudes greater than 2500 meters, birth weights generally decrease
to high altitude
(24–28). Reproductive success and infant survival are crucial Recent advances in genomics provide opportunities to exam-
components of transgenerational adaptation in highlanders. ine thousands to millions of genetic markers at once, provid-
Compared to individuals of lowland ancestry at high altitude, ing important information about adaptive genetic signatures in
Tibetans and Andeans exhibit increased common iliac blood highland populations. Microarrays, which provide information
flow into uterine arteries and less intrauterine growth restric- about variable sites scattered throughout the entire genome,
tion during pregnancy with Tibetans tending to have less pre- as well as exome and whole genome sequence analyses, which
and postnatal mortality (29–33). Birth weights are also higher provide information about protein coding and both coding and
among Tibetan and Andean babies (34). It appears that adverse noncoding sites, respectively, have been utilized over the past
reproductive effects are lessened among highlander groups who decade to pinpoint key genetic targets for high-altitude adapta-
have resided at high altitude for many generations, suggesting a tion. In addition to these genomic tools, other -omics techniques
genetic basis for these differences (31). (epigenomics, transcriptomics, metabolomics, proteomics) have
The prevalence of chronic mountain sickness (CMS), char- become increasingly accessible and provide important comple-
acterized by excessive erythrocytosis, hypoxemia, and in some mentary information to genomic and physiological data from
cases pulmonary hypertension and possible development of cor these populations.
pulmonale and congestive heart failure, varies across popula- Genetic signatures of adaptation may be detected by examin-
tions resident more than 2500 m above sea level. While Tibetans ing patterns of variation within a collection of genomes from a
appear to be at lower risk of developing CMS, it is more common population exposed to strong selective pressures. Many tests of
in Andeans and noted in nearly 30% of men more than 60 years of selection search for outlier signals exhibiting reduced genetic
age (34–35). Research in Ethiopian highlanders is limited. CMS diversity. This reduction in variation results from beneficial
is also noted among Han Chinese who have moved to high alti- genetic variant(s), and surrounding linked genetic markers,
tude (36) and residents of Leadville, CO, in the United States (37). increasing rapidly in the population over hundreds of generations
Population differences in susceptibility to CMS may be attributed as they provide an advantage for survival (20). Extreme or even
to genetic, epigenetic, and environmental factors. subtle allele frequency differences in highland genomes relative
Various reviews regarding physiological traits exhibited by high- to lowland populations may also be used to identify potentially
land populations are available and provide insight into remaining adaptive genetic markers. These approaches, based on patterns in
questions in this field (2, 14, 20, 38, 39). Integration of key phe- genomic regions or specific sites, may also be examined in com-
notype information, longitudinal outcomes, and the genetic and bination; such a technique was recently applied to whole genome
molecular basis of adaptation within and across Tibetan, Andean, sequence data from Tibetans (40).
and Ethiopian populations will be necessary to address the com- One of the key genetic pathways identified within regions
plexities of human adaptation to altitude (Figure 4.1). exhibiting a signal of positive selection in multigenerational
highland populations is the hypoxia-inducible factor (HIF) path-
way. HIFs are transcription factors that bind to specific DNA
sequences and regulate the expression of messenger RNA that
encodes for hypoxia-response proteins. There are three isoforms
of HIF with HIF-1α, HIF-2α, and HIF-3α subunits that form a
heterodimer with a constitutively expressed HIF-β subunit.
HIF-1α and HIF-2α have distinct patterns of expression with
tissue- and temporal-specific effects (41, 42), although HIF-3α
activity is not as well characterized. During conditions of nor-
moxia, the α subunits are targeted for degradation via oxygen-
dependent prolyl hydroxylase domain (PHD) proteins (PHD-1,
PHD-2, PHD-3) and ubiquitination that occurs through interac-
tion with the von Hippel-Lindau (VHL). HIFs play various roles in
response to cellular hypoxia (43) that involve thousands of down-
stream genes involved in erythropoiesis, angiogenesis, metabolic
pathways, vasomotor tone, cell proliferation, and survival (42).
Most oxygen-dependent species utilize this important pathway.
Genomic studies of high-altitude populations provide impor-
tant insight into key hypoxia-response genes as well as non-
HIF-associated pathways important for long-term adaptation.
Cross-population comparisons further highlight potential
mechanisms of adaptation that underlie distinct physiological
aspects of adaptation. While various genetic pathways and, in
FIGURE 4.1 Genomic signatures of adaptation identified in some cases, specific genes have been identified in more than one
more than one continental highland population. Gene names continental highland population, the precise putatively adap-
provided for findings reported in genomic studies of Tibetan, tive variants remain to be identified with the exception of only a
Andean, and Ethiopian populations. *HIF pathway genes few cases. Additional studies will be necessary to determine the
include EPAS1 in all three populations, EGLN1 in Tibetans, and functional relevance of adaptive signals reported thus far and
BHLHE41 in Ethiopians. determine whether the specific variants are associated with key
Human Adaptations to High Altitude 21

phenotypes, as reported in many studies of Tibetan adaptation understanding distinct population histories, and unique genetic
thus far (2, 20, 44). backgrounds, in studies of genetic adaptation to high altitude.

Genetic adaptation in Tibetan highlanders Genetic adaptation in Andean highlanders


Some of the strongest evidence for adaptation in Tibetans is based While fewer genomic studies have been completed in Andeans
on assessments of genotype-phenotype relationships between relatives Tibetans, various high-altitude candidate genes in this
putatively adaptive markers and unique physiological traits exhib- population have been identified and, in few cases, replicated in
ited by Tibetans. Using microarray analyses of nearly one million independent studies. The first genome-wide analysis was based
genetic markers, in 2010 Simonson et al. conducted within and on nearly a half million SNPs examined in Aymara and Quechua
across-population (comparing Han Chinese and Tibetan popula- Andeans (53), which was followed by subsequent analysis com-
tions) tests of selection (45). The list of adaptive candidate genes paring Andean and Tibetan populations that concluded genomic
identified in this analysis was compared to an a priori list of candi- adaptation was dissimilar, although an exception was noted at
date genes based on known function in hypoxia-related pathways the EGLN1 gene that appeared to have different haplotype struc-
(45). Two HIF pathway genes, EPAS1 which encodes HIF-2α, and ture in Andeans and Tibetans (54). Sequence analysis of this
EGLN1 which encodes prolyl hydroxylase PHD2, were identified region revealed that while both Andeans and Tibetans exhibit
among the top ten genes reported, and adaptive copies of EGLN1 an adaptive signal at this locus, the putatively adaptive variants
and PPARA gene candidates were associated with relatively lower identified in Tibetans were absent or found at low frequency in
hemoglobin concentration. These genes were also associated with Andeans (55). Among the genes originally identified by Bigham
metabolic parameters in Tibetans (46), and PPARA was associ- et al., EDNRA (endothelin receptor type A) and PRKAA1 (pro-
ated with metabolic alterations in skeletal muscle of Sherpa (47, tein kinase AMP-activated catalytic subunit alpha 1) were found
48). The top candidate gene HMOX2 reported by Simonson et al. to be associated with birthweight, and PRKAA1 with metabolic
(45) was also associated with hemoglobin concentration in an homeostasis (56).
analysis of data from 1250 Tibetan men (49). Additional analyses suggest genes related to cardiac function
Based on analysis of exome sequence data from 50 Tibetans, have been important for adaptive processes in Andeans. Genes
Yi et al. in 2010 identified variants in the EPAS1 gene that were identified in larger genome-wide SNP analyses identified VEGFB,
highly differentiated between Tibetans and Han Chinese (50). which encodes vascular endothelial growth factor B and ELTD1, the
A single variant, which differed by 78% frequency between the adhesion G protein-coupled receptor L4 (57). Low-coverage whole
two populations, was associated with hemoglobin concentration genome sequencing revealed signals of positive selection at BRINP3
in Tibetans. Many of the same genes (e.g. EPAS1, EGLN1, the (BMP/retinoic acid inducible neural specific 3), NOS2 (nitric oxide
hemoglobin gene cluster, PKLR, and HFE) were reported by both synthase 2), and TBX5 (T-box transcription factor 5) loci (58).
Simonson et al. (45) and Yi et al. (50), despite the use of different Another whole genome sequence analysis, based on 10 indi-
techniques (20). viduals with and 10 without CMS, identified SENP1 (SUMO spe-
In an effort to identify major differences in allele frequencies cific peptidase 1) and ANP32D (acidic nuclear phosphoprotein 32
between Han Chinese and Tibetans, Beall et al. analyzed data family member D) as top targets for adaptation (59). Decreased
from approximately 500,000 SNPs and found eight variants close levels of these genes were detected in Andeans without CMS and
to the EPAS1 gene that were highly differentiated and further decreased expression in a Drosophila genetic model was asso-
associated with hemoglobin concentration in Tibetans (51). The ciated with greater survival under conditions of hypoxia (59).
findings of these three studies, which were the first to provide Additional studies based on SNP microarray analysis indicate
phenotypic associations with adaptive genetic factors in Tibetans, FAM213A (peroxiredoxin like 2A) and SFTPD (surfactant pro-
were replicated in numerous subsequent analyses (2, 20, 39). Given tein D) are also strong targets of selection in Andeans (60). Whole
that HIFs regulate hundreds to thousands of genes and pathways, genome sequence analysis based on highland Argentinians and
these findings provide a glimpse into the complexity of genetic lowland Native Americans also revealed an adaptive signature
adaptation to high altitude. In addition, hundreds of distinct puta- at the EPAS1 locus previously identified as a top candidate gene
tively adaptive genes have been identified across studies, including in Tibetans, and GPR126 (adhesion G protein-coupled receptor
HIF and non-HIF pathway genes. Efforts to systematically inves- G6). Several additional genes reported in these studies have since
tigate the overlap of these signatures across studies will provide been reported as adaptive in other Andean populations or identi-
important insight for prioritization in future studies. fied in Tibetans (e.g., the B hemoglobin locus and EDNRA) (20),
Each highland population’s evolutionary history, which as well as IL6 (interleukin 6), ALDH2 (aldehyde dehydrogenase 2
impacts present-day physiology, is distinct, and recent findings family member), and alcohol dehydrogenase genes, among others,
based on comparisons of Tibetan and archaic genomes illustrate including EPAS1 (61).
this important point. Increased technological advancements
have provided opportunity to sequence ancient DNA samples Genetic adaptation in Ethiopian
from populations no longer living, including Neanderthals and highland populations
Denisovans who were contemporaries of modern humans until Fewer physiological and genomic studies have been completed
tens of thousands of years ago. Genetic material from these popu- in highland Ethiopian populations. Populations have resided in
lations is, however, still present in the genomes of extant modern the Ethiopian highlands for more than 70,000 years (6), although
humans. Genetic variants in the EPAS1 gene region in Tibetans, people likely moved in and out of high-altitude areas, with a per-
noted as one of the strongest adaptive signatures in Tibetans, manent settlement (at 4200 m) recently dated to 31,000–47,000
is most similar to Denisovan DNA compared to DNA of other years ago (62). While the genomic findings thus far suggest
human populations (40, 52). Therefore, archaic genetic admix- involvement of the HIF pathway in Ethiopian adaptation (63),
ture provided variation that helped Tibetans to adapt to the high- which is similar to various reports in Tibetans and Andeans,
altitude environment. This finding highlights the importance of other distinct adaptive pathways have been reported (2, 8, 20).
22 Hypoxic Respiratory Failure in the Newborn

Some of the putatively adapted gene regions identified in (Asp4Glu, Cys127Ser) exhibits a gain of function (a lower Km
Ethiopians are associated with hemoglobin concentration. For for oxygen) and is associated with increased HIF degradation in
example, THRB (thyroid hormone receptor beta) and the gene hypoxia and potential erythroid progenitor proliferation disrup-
regions of PPARA and EPAS1 are reported as adaptive targets of tion (66). Additional reports suggest a loss-of-function role via
selection and/or associated with hemoglobin concentration (45, defective binding of co-chaperone p23 and increased HIF activ-
50, 51) in Tibetans, are also associated with this phenotype in ity (67, 68). In addition to this protein-coding change, variation
Amhara Ethiopians (8). Another HIF pathway gene BHLHE41 within the first intron of EGLN1 was associated with increased
(basic helix-loop-helix family member E41) is also a top selection expression and high-altitude pulmonary edema (HAPE) in a pop-
candidate identified across three Ethiopian populations (Amhara, ulation from India (69).
Oromo, and Tigray Ethiopians) (9). Increased availability of whole genome sequence data indi-
Other genes not associated with phenotypes but previously iden- cate that regions of the genome with heterochromatic marks,
tified in other highland population include EDNRB (endothelial DNA methylations sites, and non-coding variants are crucial for
receptor B) identified in Amhara Ethiopians as well as in Andeans adaptation [e.g. in Andeans (59) and Tibetans (40)]. Genetic fac-
(54), while EDNRA, within the same gene family, is also a selec- tors that influence patterns of gene expression provide mecha-
tion candidate gene in Tibetans (45). In a mouse model, decreased nisms for variation across tissues and/or stages of development
expression of EDNRB is associated with hypoxia tolerance (64). in contrast to variants that impact protein function, resulting in
Others non-hypoxia-associated genes identified thus far uniform alterations across all cells. These fine-tuned, context-
in Amhara Ethiopians include VAV3 (vav guanine nucleotide specific changes could underlie important molecular mecha-
exchange factor 3) and RORA (RAR-related orphan receptor A) nisms of adaptation including key changes in hypoxia-related or
(8). Adaptive targets of a regions in chromosome 19 in Oromo and hypoxia-independent pathways.
Simen Ethiopians include CIC (capicua transcriptional repressor), In addition to progress regarding whole genome sequence analy-
LIPE (lipase E hormone-sensitive type), and PAFAH1B3 (plate- sis, insights have been more recently obtained using complementary
let-activating factor acetylhydrolase 1b catalytic subunit 2), and -omics strategies, such as transcriptomics, epigenomics, metabolo-
appear to afford tolerance to hypoxia in a Drosophila model (64). mics, and proteomics. Given various -omics factors may be impacted
by environmental cues, this is an important and active area of
Cross-population and cross-species study. For example, variation in DNA methylation may impact
gene expression, although limited data are currently available for
insights into adaptive genetic factors populations resident at high altitude. A study of saliva samples
Many studies of genetic adaptation in humans have converged on from Oromo Ethiopians at high and low altitudes showed distinct
HIF pathway genes as top candidates of selection. In many cases, patterns of CpG methylation at four sites (7). Based on a limited
it appears that key distinct pathway genes, and in some cases, study of Andeans with and without excessive erythrocytosis,
the same genes may underlie adaptation. Various studies have differential methylation was identified within the EGLN1gene
searched the genomes of other high-altitude species and identi- (70, 71). A more recent study showed that the number of years
fied overlapping targets of selection related to various conditions spent living at high altitude was associated with decreased DNA
of environmental hypoxia (65). Although there is major overlap in methylation at the promoter region of EPAS1, and higher levels at
genetic pathways highlighted thus far (i.e. the hemoglobin gene LINE-1 that also correlated with altitude at birth, based on epi-
cluster, associated with blood-oxygen affinity, HIF pathway genes genetic analyses of Quechua individuals living at high versus low
such as EPAS1 and EGLN1), different variants with putatively dis- altitude in Peru (72).
tinct functions are most commonly reported as targets of selec- Additional genomic and -omics studies that integrate infor-
tion (65). mation with physiological findings and functional analyses of
The genes mentioned in this chapter by no means provide a precise variants will provide greater insight into the adaptive
comprehensive list of candidates to compare, as many studies landscape of highland populations. Candidate variants, priori-
based on human genomic data thus far highlight only prioritized tized based on identification as targets of positive selection and,
sets within regions of the genome exhibiting an adaptative signal in some cases, phenotype associations, may be evaluated through
and/or report hundreds of genes or subsets of the genes that meet gene-editing techniques in various cell lines and under varying
criteria established by the authors of the study. In addition, vari- physiological conditions (73). Such studies may yield important
ous analytical approaches may be employed across studies, sam- insight into causality and further understanding of ancestry-
pling strategies may vary, and adaptive signals may be detected specific responses to hypoxia relevant in the contexts of health
or not based on individual or population histories within a and disease.
region. Efforts to more fully capture genomic information across
geographical regions and to perform standardize analyses will References
provide greater insight into individual variation needed to fully 1. Simonson TS. High Alt Med Biol. 2015; 16(2):125–137.
appreciate unique and shared signatures of adaptation in humans. 2. Moore LG. J Appl Physiol. 2017; 123:1371–1385.
3. Zhang XL, et.al. Science. 2018; 362(6418):1049–1051.
4. Rademaker K, et.al. Science. 2014; 346(6208):466–469.
5. Cook ND. Born to die: disease and New World conquest, 1492-1650. Cambridge, NY:
Functional insights into genetic adaptation Cambridge University Press; 1998. p. 268.
6. Hassen M. The Oromo of Ethiopia: a history, 1570–1860. Trenton, NJ: Red Sea Press;
1990.
While thousands of high-altitude selection candidate genes and 7. Alkorta-Aranburu G, et al. PLoS Genet. 2012; 8:e1003110.
gene regions have been reported, the functional variant(s) under- 8. Scheinfeldt LB, et.al. Genome Biol. 2012; 13:R1.
lying adaptation largely remain to be determined. Some prog- 9. Huerta-Sánchez E, et.al. Mol Biol Evol. 2013; 30:1877–1888.
10. Beall CM. Proc Natl Acad Sci USA. 2007; 104:8655–8660.
ress has been made for the EGLN1 gene, which encodes PHD2, a 11. Simonson TS, et al. J Physiol. 2015; 593:3207–3218.
top target of selection in Tibetans. One variant in the first exon 12. Jeong C, et al. Nat Commun. 2014; 5:3281.
Human Adaptations to High Altitude 23

13. Beall CM, et al. Proc Natl Acad Sci USA. 2002; 99:17215–17218. 44. Azad P, et al. J Mol Med (Berl). 2017; 95:1269–1282.
14. Beall CM. Integr Comp Biol. 2006; 46:18–24. 45. Simonson TS, et al. Science. 2010; 329:72–75.
15. Cheong HI, et al. Am J Physiol Lung Cell Mol Physiol. 2017; 312:L172–L177. 46. Ge R-L, et al. Mol Genet Metab. 2012; 106:244–247.
16. Stembridge M, et al. Proc Natl Acad Sci USA. 2019; 116:16177–16179. 47. Horscroft JA, et al. Proc Natl Acad Sci USA. 2017; 114(24):6382–6387.
17. Balaban DY, et al. Respir Physiol Neurobiol. 2013; 186:45–52. 48. O’Brien KA, et al. Curr Opin Endocr Metab Res. 2019; 11:33–41.
18. Simonson TS, et al. Exp Physiol. 2014; 99:1624–1635. 49. Yang D, et al. Hum Mutat. 2016; 37:216–223.
19. Beall CM. Hum Biol. 2000; 72:201–228. 50. Yi X, et al. Science. 2010; 329:75–78.
20. Simonson TS. High Alt Med Biol. 2015b; 16:125–137. 51. Beall CM, et al. Proc Natl Acad Sci USA. 2010; 107:11459–11464.
21. Niermeyer S, et al. Pulm Circ. 2015; 5:48–62. 52. Huerta-Sánchez E, et al. Nature. 2014; 512:194–197.
22. Julian CG, et al. Am J Physiol Heart Circ Physiol. 2015; 309:H565–H573. 53. Bigham AW, et al. Hum Genomics. 2009; 4:79–90.
23. Jang EA, et al. Front Physiol. 2015; 6:176. 54. Bigham A, et al. PLoS Genet. 2010; 6:e1001116.
24. Jensen GM, et al. Am J Public Health. 1997; 87:1003–1007. 55. Heinrich EC, et al. Ann Hum Genet. 2019; 83:171–176.
25. Krampl E, et al. Ultrasound Obstet Gynecol. 2000; 16:9–18. 56. Bigham AW, et al. Physiol Genomics. 2014; 46:687–697.
26. Giussani DA, et al. J Dev Orig Health Dis. 2013; 4:328–337. 57. Eichstaedt CA, et al. PloS one. 2014; 9(3):e93314
27. Giussani DA, et al. Pediatr Res. 2001; 49:490–494. 58. Crawford JE, et al. Am J Hum Genet. 2017; 101:752–767.
28. Levine LD, et al. Am J Obstet Gynecol. 2015; 212:210 e211–218. 59. Zhou D, et al. Am J Hum Genet. 2013; 93:452–462.
29. Zamudio S, et al. Am J Phys Anthropol. 1993; 91:215–224. 60. Valverde G, et al. PLoS One. 2015; 10(5):e0125444
30. Moore LG, et al. Am J Phys Anthropol. 1998; Suppl 27:25–64. 61. Foll M, et al. Am J Hum Genet. 2014; 95:394–407.
31. Moore LG. High Alt Med Biol. 2001; 2:257.–279. 62. Ossendorf G, et al. Science. 2019; 365:583–587.
32. Moore LG, et al. Am J Hum Biol. 2001; 13:635–644. 63. Bigham AW, et al. Genes Dev. 2014; 28:2189–2204.
33. Tripathy V, et al. Am J Hum Biol. 2005; 17:442–450. 64. Udpa N, et al. Genome Biol. 2014; 15:R36.
34. Monge C, et al. N Engl J Med. 1989; 321(18):1271. 65. Pamenter ME, et al. Front Genet. 2020; 11:743.
35. Villafuerte FC, et al. High Alt Med Biol. 2016; 17:61–69. 66. Lorenzo FR, et al. Nat Genet. 2014; 46:951–956.
36. Jiang C, et al. BMC Public Health. 2014; 14:701. 67. Song D, et al. J Biol Chem. 2014; 289:14656–14665.
37. Asmus IV. In: Health and behavioral sciences. Denver, CO: University of Colorado; 2002. 68. Song D, et al. Proc Natl Acad Sci USA. 2020; 117:12230–12238.
38. Gilbert-Kawai ET, et al. Physiology. 2014; 29:388–402. 69. Aggarwal S, et al. Proc Natl Acad Sci. 2010; 107:18961–18966.
39. Bhandari S, et al. Front Physiol. 2019; 10:1116. 70. Julian CG. J Appl Physiol. 2017; 123:1362–1370.
40. Hu H, et al. PLoS Genet. 2017; 13:e1006675. 71. Julian CG. Front Physiol. 2019; 10:1397.
41. Hu CJ, et al. Mol Cell Biol. 2003; 23:9361–9374. 72. Childebayeva A, et al. Epigenetics. 2019; 14:1–15.
42. Semenza GL. Ann Rev Genomics Hum Genet. 2020; 21:183–204. 73. Hall JE, et al. Front Genet. 2020; 11:471.
43. Samanta D, et al. Wiley Interdiscip Rev Syst Biol Med. 2017; 9(4):10.1002/wsbm.1382.
FETAL LLAMA ADAPTATION TO ALTITUDE IN THE ANDEAN ALTIPLANO

Aníbal J. Llanos, Germán Ebensperger, Emilio A. Herrera,


Roberto V. Reyes and María Serón-Ferré

Contents
Introduction..........................................................................................................................................................................................................................24
The adult llama.....................................................................................................................................................................................................................24
The fetal llama.......................................................................................................................................................................................................................24
The fetal llama cardiovascular system and adrenergic mechanisms...................................................................................................................24
Hypoxia tolerance and the llama brain......................................................................................................................................................................26
The newborn llama pulmonary circulation....................................................................................................................................................................26
Acknowledgments...............................................................................................................................................................................................................27
References..............................................................................................................................................................................................................................27

Introduction disappear at lowland, indicating that they were acclimatization


instead of adaptation (14).
Aquí la hebra dorada salió de la vicuña
a vestir los amores, los túmulos, las madres,
el rey, las oraciones, los guerreros The fetal llama
(The fleece of the vicuña was carded here Species survival requires the ability to produce kin. To describe
to clothe men’s love in gold, their tombs and mothers, fetal oxygenation in the womb, Professor Joseph Barcroft coined
the king, the prayers, the warriors) his famous phrase summarized as Everest in utero (15, 16). At
lowland, the umbilical vein has the highest fetal PO2 (30 mmHg),
“The Heights of Macchu Picchu,” Pablo Neruda two-thirds lower than the maternal aortic PO2. Barcroft’s meta-
(translated by Nathaniel Tarn) phor is apt, as years later, at the summit of Mount Everest, at 8848
m of altitude, Christopher Pizzo’s alveolar gas PO2 was 28 mmHg
High on the Andes, the whole Altiplano is immersed in thin atmo- (17). Nonetheless, the natural habitat of the maternal and fetal
spheric air, but despite of the latter, it sustains a good number llama is 4000 m. We measured PO2 in fetal and maternal llamas
of animals, including the llama (Lama glama), a member of the at 4400 m above sea level, and the values dropped from 21 mmHg
Camelidae family, presently residing over 4000 m above sea level. at sea level to 15 mmHg at 4400 m (18), while in the maternal
The llama’s journey began about 2 million years ago, as the llama llama, PO2 went from 98 to 44 mmHg at 4400 m (Llanos, unpub-
North American ancestor climbed and dived onto the Andes’ lished data). Consequently, in Andean pregnancies, we were wit-
atmospheric ocean of the Alto Andino (1). Between 7000 and nessing hypoxia inside hypoxia, exceeding Professor Barcroft’s
6000 years ago, the llama and the alpaca became domesticated description.
by the Amerindians in today’s Peruvian Andes (2), the guanaco In this chapter, we describe three rather unknown features,
being the ancestor of the llama and the vicuña of the alpaca (3). paramount in determining the ability of the fetal and neona-
tal llamas to thrive in the high-altitude plateau. First, the fetal
llama has an augmented function of the α-adrenergic receptors
The adult llama to sustain cardiovascular function. Second, the fetal llama has
mechanisms that allow the brain to withstand hypoxia. Third,
The adult llama displays important physiological adaptations per- the neonatal llama lung has increased production of carbon mon-
mitting it to thrive at altitudes over 4000 m. Some traits may be oxide (CO), a mechanism preventing neonatal pulmonary high-
the expression of genetic changes since they persist after many altitude arterial hypertension. A brief description of these notable
generations of residing at lowlands. Among them are a low P50 (4, changes gives a glimpse of how the fetal and neonatal llama cope
5), small elliptical red cells with a slight increase in blood hemo- with the high-altitude hypoxia of the Alto Andino.
globin content (6, 7), and decreased expression of the carbonic
anhydrase II isozyme (CA II) (8), permitting better O2 extraction
by the tissues (9). More recently, it has been found that alpacas The fetal llama cardiovascular system
hold an archaic helix-loop-helix deletion in the HIF-1 alpha pro- and adrenergic mechanisms
tein, which could be reducing the hypoxic responses at high alti- We assessed fetal cardiovascular function in our model of a
tudes (10), since HIFs could produce maladaptation to chronic chronically catheterized fetal llama at Santiago (580 m) (19), by
hypoxia, inducing polycythemia and pulmonary arterial hyper- comparing values with fetal sheep at similar gestational ages. The
tension (11). In this regard, a notable physiological attribute of the fetal and neonatal sheep are the “gold standard,” since most of
highland llama is an arterial pulmonary pressure like lowland lla- the perinatal research with results translated to humans took
mas, without remodeling of pulmonary vessels (12), and a lesser place in this species. Our comparative studies detected differ-
pulmonary pressure increase to acute hypoxia than sheep at low ences between fetal llama and fetal sheep, already in a situation
altitude (13). Conversely, the high muscle myoglobin concentra- of normoxia, at which PO2 levels were rather similar in both
tion and lactic dehydrogenase activity present in highland llamas species. Nevertheless, the fetal llama systemic arterial pressure,

24 DOI: 10.1201/9780367494018-5
Fetal Llama Adaptation 25

cardiac output, and organ blood flows were lower than in fetal reabsorbing a great deal of water, explaining the high osmolality
sheep, together with a higher total vascular resistance pointing to found in the fetal llama urine (36).
important differences between both species (19–21). Administration of phentolamine (alpha-1- and alpha-2-adren-
These differences are further highlighted when fetal llamas are ergic receptor antagonist) during normoxia had major effects,
exposed to an episode of acute hypoxia, as described here. We eliciting systemic hypotension and tachycardia, with a reduction
studied the fetal llama cardiovascular responses to acute hypoxia in vascular resistance, and rising blood flow in carotid and femo-
extensively (18, 20, 22–26). Our results showed that in fetal llama ral territories. Furthermore, it elicited dramatic effects when the
at 0.6- to 0.7-gestation, at lowland, acute hypoxia elicited a rise in fetal llamas were submitted to acute hypoxia, inducing a rapid
mean arterial pressure, a short-lived bradycardia (23), and a redis- and pronounced systemic hypotension, paralleled with marked
tribution of blood flow towards to heart and adrenals glands, but bradycardia plus deterioration of all the measured cardiovascular
oddly not to the brain (22, 25). The latter is a feature not observed variables, that led all six fetuses to cardiovascular collapse and
in the fetal sheep or in any other mammalian species exposed death. The continent (the vessels’ territory) was greater than the
to acute hypoxia. Another notable finding was the considerable content (the blood), producing the collapse. To identify which of
peripheral vasoconstriction produced by hypoxia, exemplified the two alpha-adrenergic receptors brought this dramatic result,
by a femoral resistance almost fivefold greater than that of fetal we repeated the experiments using the more specific alpha-1-
sheep at the same gestational age (23, 27, 28). adrenergic receptor antagonist, prazosin, finding a similar outcome.
The mechanisms mediating the fetal cardiovascular responses In contrast, administration of yohimbine, an alpha-2-adrenergic
to acute hypoxia involve neural (chemoreflex), endocrine, and receptor antagonist, showed no cardiovascular effects either in
vasoactive local molecules, such as NO, CO, endothelin, and normoxia or in hypoxia (26). Therefore, the alpha-1-adrenergic
others (28). A potent carotid chemoreflex mediates cardiovas- receptor function appears to be the fundamental mediator in
cular responses to hypoxia in the fetal sheep. Surprisingly, and the vasoconstrictor response to hypoxia in the fetal llama. This
in marked contrast to the fetal sheep, we found that the che- outcome was in marked contrast to what is observed in the fetal
moreflex plays a minor role in the fetal llama. Bilateral carotid sheep with hypoxia, in which alpha-adrenergic receptor blockade
body denervation did not change the substantial femoral vas- also abolished femoral vasoconstriction; and only when hypoxia
cular vasoconstriction induced by hypoxia (23). However, the was accompanied with carotid sinus nerve section the fetal sheep
chemoreflex may trigger other mechanisms, such as fetal llama did not survive (27). Thus, in lowland species such as sheep,
adrenocortical responses to hypoxia, since carotid body dener- two mechanisms are important for fetal survival during acute
vation abolished cortisol increase, despite maintaining plasma hypoxia: alpha-adrenergic function and carotid chemoreflexes.
ACTH (29). In contrast, the rise in plasma cortisol ensued, but For fetal llamas, only alpha-1-adrenergic mechanisms are funda-
more slowly than controls in fetal sheep (23). Therefore, in the mental to cope with acute hypoxia.
fetal llama, the neural input is a principal regulator for the adre- The importance of the fetal alpha-1-adrenergic mechanisms
nal cortex. during chronic hypoxia is further highlighted in fetal sheep made
Given the limited cardiovascular effect of the chemoreflex, chronically hypoxic by placental embolization (37), and in chron-
we next addressed the role of endocrine and local mechanisms ically hypoxic fetal sheep at 3600 m (Llanos, unpublished data). In
in regulating peripheral vascular response to hypoxia in the fetal both situations, administration of phentolamine during a super-
llama (23, 30). imposed episode of acute hypoxia produced cardiovascular col-
The fetal llama has a greater basal concentration of plasma cat- lapse and death, as in the fetal llama.
echolamines than the fetal sheep, revealing an important basal Thus, fetal chronic hypoxia in lowland species, or evolutionary
sympathetic tone (31, 32). Hypoxia induced a norepinephrine chronic hypoxia in highland species at near sea level, amplifies
(NE) surge that was more important than that of epinephrine (E) the contribution of the alpha-adrenergic system in the defense
(32), associated with increases of neuropeptide Y (NPY) and vaso- against episodes of acute hypoxia.
pressin, but no changes in angiotensin II (18). Neuropeptide Y is These findings are critical on clinical grounds, since adminis-
colocalized and cosecreted with catecholamines (33). Further, tration of alpha-adrenergic blockers to pregnant mothers, either
fetal sheep treated with exogenous NPY mimic the vasoconstric- at high altitudes where more than 140 million people live (38) or
tion produced by hypoxia. Due to this notable vasoconstriction in pregnancies complicated with chronic fetal hypoxia at low-
effect and the high NPY levels attained in fetal llama hypoxia, lands, would block in the fetus, the meaningful alpha-adrenergic
NPY could be participating in the potent vasoconstriction compensation during chronic hypoxia leads to a poor outcome.
observed in the fetal llama (34). The intensity of the peripheral vasoconstriction in the hypoxic
To dissect the role of some hormones and local factors in the fetal llama has a robust counterpart of vasodilators at the vascu-
intense vasoconstriction experienced in hypoxia by the fetal lar level. Indeed, local endothelial NO production has a vital role
llama, we administered the respective endothelin (ET), arginine in maintaining basal blood flow literally in all local circulations
vasopressin (AVP), and alpha-adrenergic receptors antagonists. (cerebral, carotid, heart, adrenal and femoral vascular beds) in
Endothelin-1 acts through its receptors ETA and ETB located normoxia, and it lessens the robust vasoconstrictor influence on
in the vascular smooth muscle. ET-1 contributed to the augment these beds during acute hypoxia as demonstrated by L-NAME
of peripheral resistance in the fetal llama under acute hypoxia, blockade of NO synthesis (26, 39).
since the infusion of an ETA receptor blocker, BQ123, precluded These results show an essential role of NO signaling in the
the rise of femoral vascular resistance without modifying the preservation of a dilator tone in the cerebral and femoral circula-
basal femoral tone (26). In contrast, when vasopressin V1 recep- tions, lessening the strong vasoconstrictor influences upon these
tor antagonist was administered in normoxia, it decreased fetal territories during acute hypoxia in the fetal llama. Additionally,
femoral vascular resistance, increasing the blood flow, but did these experiments show that NO plays a central role in augment-
not alter cardiovascular variables during acute hypoxia (24, 35). ing the heart blood flow in acute hypoxia in this species (26, 39).
Additionally, vasopressin could also act on the renal V2 receptor, Furthermore in hypoxia, L-NAME did not affect the rise in fetal
26 Hypoxic Respiratory Failure in the Newborn

plasma ACTH but precluded the increments in adrenal blood lactate by the fetal llama brain and absence of PARP protein deg-
flow and cortisol and adrenaline concentrations in the fetal radation. All these findings support the hypothesis that the fetal
llama. In contrast, L-NAME further augmented the fetal plasma llama responded with brain hypometabolism to cope with an epi-
noradrenaline. All these data support a regulatory role of NO on sode of prolonged hypoxia (45). The hypometabolic responses and
adrenal blood flow and function in the fetal llama (32). hypoxia tolerance have been described in a wide range of species
The pronounced intensity of the peripheral vasoconstriction that, for limitation of space, are not possible to describe.
response to cope with acute hypoxia in the fetal llama opens an The strategy of hypoxic hypometabolism found in the fetal
important question about its adaptative function(s). Certainly, we llama brain is an effective adaptation to dwell amidst the thin air
do not have the answers, but we can speculate. First, reducing milieu of the Alto Andino. Whether this is present in other fetal
the blood flow and O2 supply to several organs and reducing the tissues needs to be investigated.
O2 consumption and temperature could induce a hypometabolic
adaptation of these organs. Second, it permits redirection of the The newborn llama pulmonary circulation
blood flow and O2 supply toward the vital organs, in this case,
heart and adrenal glands. The brain blood flow did not increase To emerge from the womb “into this breathing world” (Shakespeare,
with hypoxia but showed certain degree of hypometabolism, as Richard the Third) is one of the most dramatic events in the life of
we will discuss later. Third, reducing the blood flow and O2 sup- the individual, since the neonate in barely 2–3 minutes establishes
ply to several organs may trigger the release of molecules that its complete pulmonary function, including the pulmonary circula-
may produce ischemic preconditioning in the brain, heart, adre- tion. The latter strikingly changes from constricted fetal pulmonary
nals, in the tributary organ of the constricted artery, or in the to neonatal dilated vessels accommodating ten times more blood
whole body (40). In this regard, alpha-1-adrenergic receptors are flow than before (46). A necessary condition is a rapid fall of pulmo-
involved in adult heart preconditioning and may do so in fetal nary vascular resistance, and failure to do so results in pulmonary
llama heart as well in other organs (41). hypertension in the newborn. In the Altiplano, the llama neonate
rapidly establishes an efficient pulmonary circulation without pul-
Hypoxia tolerance and the llama brain monary hypertension, in contrast, the neonatal lambs end with dif-
An outstanding difference between fetal llama and sheep ferent degrees of hypoxic pulmonary hypertension (47).
responses to hypoxia resides in the brain. During an episode of Neonatal llama systemic circulation maintains some features
acute hypoxia in the fetal sheep at 0.85 gestation, cerebral vas- already present in utero, like a femoral vasoconstriction 3.6 times
cular resistance decreases, enhancing blood flow threefold to greater than in newborn sheep, in response to acute hypoxia at
all regions of the brain. This preserves brain oxygen supply and low altitude. This is mediated by an enhanced peripheral vasocon-
maintains oxygen consumption until ascending aortic blood oxy- strictor sensitivity to norepinephrine and femoral vascular bed
gen content fall to approximately 2.2 ml/dl. Below this threshold, expression of alpha-1B-adrenergic instead of alpha-1A- adrenergic
the brain oxygen consumption begins to fall (21, 42–44). receptor. The latter, less sensitive to adrenergic agonists, is more
In the fetal llama, the response was utterly different. Acute abundant in the femoral circulation of the newborn sheep (48).
hypoxia in the fetal llama neither changes cerebral vascular resis- Thus, the fetal and newborn llama alpha-adrenergic tone may be
tance nor cerebral blood flow (20, 22, 23, 25). As carotid vascu- paramount to withstand the harsh life at the Andean plateau.
lar resistance and carotid blood flow remain unaltered (23, 24), Regarding the pulmonary circulation, the increase of mean arte-
the cerebral oxygen delivery was reduced (23), whereas cerebral rial pulmonary pressure (mPAP) in newborn llamas has a biphasic
oxygen extraction was maintained, reducing cerebral oxygen shape during acute hypoxia, suggesting changes in vasoconstric-
consumption in the acutely hypoxic fetal llama. In marked con- tors and vasodilators functions along the course of hypoxia. The
trast with the sheep, the electrocorticogram flattened under this newborn llama at high altitude have a lower rise in mPAP than
episode of acute hypoxia, but seizure activity did not take place newborn llama at lowlands, suggesting increased vasodilator
(25). Analyzing the data more in detail, the total brain, cerebel- mechanisms in the former. In contrast, the high-altitude newborn
lum, and pons did not modify the vascular resistance or blood sheep, already with a higher basal mPAP, augmented mPAP further
flow, while the medulla had a reduction in vascular resistance than low-altitude newborn sheep and did not have the biphasic
and a rise in blood flow during acute hypoxia in the fetal llama response observed in the newborn llama (49, 50).
(25). The preferential redistribution of blood flow within the fetal At least two mechanisms determine the lack of pulmonary
llama brain to the medulla suggests that this area is important in artery hypertension and pulmonary vascular remodeling in high-
cardiovascular control during hypoxia in this species. land newborn llama. One of them is the hemoxygenase-carbon
A fundamental question is which are the mechanisms used monoxide pathway (HO–CO). Hemoxygenases 1 and 2 (HO1 and
by the fetal llama to prevent the hypoxic neural injury? As men- HO2), found in vessels, degrade the heme group producing equi-
tioned, the electrocorticogram flattened under the episode of molar amounts of CO, Fe2+, and biliverdin. Fe2+ rapidly combines
acute hypoxia, but seizure activity did not take place (25). This with ferritin, while biliverdin generates bilirubin. Importantly,
result suggested the absence of hypoxic damage by an adaptive CO has vasodilator, anti-inflammatory and antiproliferative
brain hypometabolism. We look for further evidence by submit- properties, whereas biliverdin, bilirubin, and ferritin have anti-
ting the fetal llama to an extended episode of hypoxia, lasting 24 oxidant properties (30). Chronically administered inhaled CO
hours, determining brain temperature, Na+ channel density, and reduces pulmonary hypertension in rodents submitted to chronic
Na-K-ATPase activity. Furthermore, we looked at the poly ADP- hypoxia, while CO administered acutely diminishes pulmonary
ribose polymerase (PARP) protein degradation, as an index of cell resistance in hypoxic adult sheep (13, 51). CO induces vasodila-
death in the brain cortex. We found a fall of 0.56°C in the brain tation through activation of sGC and BKCa potassium channels,
cortex temperature, along with a 51% decrease in Na-K-ATPase increasing the smooth muscle cGMP and hyperpolarizing the cell
activity and a 44% drop in protein level of NaV1.1, a voltage-gated membrane, respectively. Additionally, it decreases smooth muscle
sodium channel. Additionally, there was a little production of proliferation (52). High-altitude newborn llamas have augmented
Fetal Llama Adaptation 27

HO1 and HO2 protein expression and a pulmonary CO produc- Acknowledgments


tion almost sevenfold higher than high-altitude newborn sheep
(30, 52, 53). Altogether, these data indicate that HO–CO signal- Supported by grant 1040647, FONDECYT, Chile.
ing is a crucial player in preventing the occurrence of pulmonary
hypertension in the newborn llama at the Alto Andino.
The second mechanism regulating pulmonary vasodilation is
References
nitric oxide (NO). We studied the role of NO in mPAP regulation in 1. Stanley H, et al. Proc Biol Sci. 1994; 256(1345):1–6.
2. Wheeler JC. Biol J Linn Soc. 1995; 54(3):271–295.
lowland and highland newborn llama and newborn sheep, basally 3. Kadwell M, et al. Proc Biol Sci. 2001; 268(1485):2575–2584.
and under an episode of acute hypoxia, by blocking NO synthesis 4. Meschia G, et al. Exp Physiol. 1960; 45(3):284–291.
with L-NAME. These experiments showed that basally the NO role 5. Moraga F, et al. Comp Biochem Physiol A Physiol. 1996; 115(2):111–115.
6. Sanhueza E, et al. Pediatr Res. 1992; 32(6):737.
is limited in lowland and highland newborn llamas. However, it is 7. Reynafarje C, et al. J Appl Physiol. 1968; 24(1):97–98.
important in both during acute hypoxia. In contrast, in newborn 8. Yang H, et al. Biochem Genet. 2000; 38(7–8):241–252.
9. Banchero N, et al. Respir Physiol. 1971; 13(1):102–115.
sheep, NO plays an important basal role in maintaining mPAP 10. Moraga FA, et al. J Health Med Sci. 2020; 6(2):97–106.
in pulmonary normotensive lowland and pulmonary hyperten- 11. Prabhakar NR, et al. Physiol Rev. 2012; 92(3):967–1003.
sive highland sheep. In the latter, NO is maximally stimulated, as 12. Harris P, et al. Thorax. 1982; 37(1):38–45.
13. Nachar R, et al. High Alt Med Biol. 2001; 2(3):377–385.
mPAP does not increase further with an acute hypoxia challenge. 14. Reynafarje C, et al. J Appl Physiol. 1975; 38(5):806–810.
Thus, NO does not suffice for compensating the pulmonary hyper- 15. Barcroft J, et al. J Physiol. 1933; 77(2):194–206.
tension suffered by newborn sheep in the Andean plateau (49, 50). 16. Allison BJ, et al. J Physiol. 2016; 594(5):1247–1264.
17. West JB. Science. 1984; 223(4638):784–788.
Basal NOS activity, eNOS, and soluble guanylyl cyclase (sGC) pro- 18. Llanos AJ, et al. High Alt Med Biol. 2003; 4(2):193–202.
tein expression in lung tissue were consistent with L-NAME find- 19. Benavides CE, et al. Respir Physiol. 1989; 75:327–334.
20. Llanos A, et al. Comp Biochem Physiol. 1998; 119(3):705–709.
ings. As expected, eNOS activity and protein were much higher in 21. Perez R, et al. Am J Physiol Regul Integr Comp Physiol. 1989; 256:R1011–R1018.
lowland and highland newborn sheep than in newborn llama (53). 22. Llanos AJ, et al. Reprod Fertil Dev. 1995; 7(3):549–552.
However, differences between newborn sheep and newborn 23. Giussani DA, et al. Am J Physiol. 1996; 271:R73–R83.
24. Giussani DA, et al. J Physiol. 1999; 515(Pt 1):233–241.
llama were found for other proteins. The highland newborn 25. Llanos AJ, et al. J Physiol. 2002; 538(3):975–983.
llama lung has a lower expression of the vasoconstrictor Rho- 26. Llanos AJ, et al. Respir Physiol Neurobiol. 2007; 158(2–3):298–306.
associated kinase 2 (ROCK 2) and phospho-serine19 myosin light 27. Giussani DA, et al. J Physiol. 1993; 461:431–449.
28. Giussani DA, et al. J Physiol. 1994; 477:81–87.
chain than lowland newborn llama, thus favoring vasodilation in 29. Riquelme RA, et al. Endocrinology. 1998; 139:2564–2570.
the pulmonary circulation at high altitude (49). Finally, highland 30. Reyes RV, et al. J Appl Physiol. 2020; 129:152–161.
31. Jones CT, et al. J Physiol. 1975; 285(1):381–393.
and lowland newborn llama had an exceptionally low asymmet- 32. Riquelme RA, et al. J Physiol. 2002; 544(1):267–276.
ric dimethylarginine (ADMA) and homocysteine plasma concen- 33. Fletcher AJW, et al. Endocrinology. 2000; 141(11):3976–3982.
trations, and a low arginase type II activity in lung parenchyma. 34. Sanhueza EM, et al. J Physiol. 2003; 546(3):891–901.
35. Herrera EA, et al. High Alt Med Biol. 2000; 1(3):175–184.
ADMA and homocysteine are inhibitors of NOS while arginase 36. Wintour EM, et al. Reprod Fertil Dev. 1995; 7:1311–1319.
hydrolyzes arginine is the main substrate for NO production. In 37. Block BS, et al. Am J Obstet Gynecol. 1984; 148(7):878–885.
contrast, both groups of newborn sheep had significantly higher 38. Moore LG, et al. Am J Phys Anthropol. 1998; 107(Suppl 27):25–64.
39. Sanhueza EM, et al. Am J Physiol Regul Integr Comp Physiol. 2005; 289(3):R776–R783.
plasma concentrations of ADMA and homocysteine, and ten 40. Perez-Pinzon MA. Comp Biochem Physiol Part A. 2007; 147:291–299.
times more lung arginase II activity than newborn llamas (54). 41. Jensen BC, et al. J Cardiovasc Pharmacol. 2014; 63(4):291–301.
42. Field DR, et al. J Dev Physiol. 1990; 14:131–137.
Plasma ADMA and homocysteine increase in adult pulmonary 43. Jones MD, et al. J Appl Physiol. 1977; 43:1080–1084.
hypertension (55). Thus, modest arginase activity and low con- 44. Richardson BS, et al. J Dev Physiol. 1989; 11(1):37–43.
centrations of ADMA and homocysteine in the newborn llama 45. Ebensperger G, et al. J Physiol. 2005; 567:963–975.
46. Rudolph AM. Ann Rev Physiol.1979; 41:383–395
constitute new and interesting mechanisms of pulmonary adap- 47. Herrera EA, et al. Am J Physiol Regul Integr Comp Physiol. 2007; 292:R2234–R2240.
tation to deal with chronic hypoxia. 48. Moraga FA, et al. Am J Physiol Regul Integr Comp Physiol. 2011; 301(4):R1153–R1160.
After finishing to describe and discuss these notable traits 49. Reyes RV, et al. J Physiol. 2018; 596:5907–5923.
50. Herrera EA, et al. Nitric Oxide. 2019; 89:71–80.
found in the fetal and neonatal llamas, we just adapted a phrase 51. Zuckerbraun BS, et al. J Exp Med. 2006; 203:2109–2119.
coined by 19th century French physiologist Claude Bernard: “No 52. Llanos AJ, et al. Respir Physiol Neurobiol. 2012; 184:186–191.
53. Herrera EA, et al. Cardiovasc Res. 2008; 77:197–201.
matter how far the Lama glama wanders at sea level it will always 54. López V, et al. Front Physiol. 2018; 9:606.
carry the Altiplano in its milieu intérieur.” 55. Arrigoni FI, et al. Circulation. 2003; 107:1195–1201.
NEONATES OF BURROWING AND HIBERNATING MAMMALS
Metabolic and Respiratory Adaptations to Hypoxia
Yvonne A. Dzal and William K. Milsom

Contents
Introduction..........................................................................................................................................................................................................................28
Physiological responses to hypoxia in adult mammals................................................................................................................................................29
Metabolic and respiratory responses to hypoxia in neonatal rodents......................................................................................................................30
Metabolic and respiratory responses to hypoxia in adult rodents.......................................................................................................................30
Effects of progressive hypoxia within the thermoneutral zone......................................................................................................................30
Effects of progressive hypoxia in the cold...........................................................................................................................................................32
Metabolic and respiratory responses to hypoxia in adult naked mole rats and big brown bats..........................................................................32
Conclusion: Are hibernators just big babies?.................................................................................................................................................................32
Acknowledgments...............................................................................................................................................................................................................33
Literature cited.....................................................................................................................................................................................................................33

Introduction different mammals use to combat low environmental O2 and


how these physiological strategies change with postnatal devel-
In this review, we explore the different hypoxic ventilatory and opment. The similarities and differences between species high-
metabolic responses employed by neonates and adults of species light the unity and the diversity of evolutionary strategies for
of small mammals, which may underlie differences in their resis- coping with O2 deprivation and allow us to examine the extent
tance to hypoxic respiratory failure. In the process, we address to which species that hibernate retain strategies common to
the hibernator as neonate hypothesis: are hibernators just big mammalian neonates.
babies? Neonatal mammals exhibit several physiological dif-
ferences from their adult counterparts. Interestingly, there are
striking parallels in physiological traits between neonatal mam-
mals and adults of species capable of hibernation (1–3) (Table TABLE 6.1 Diversity of Physiological Traits Common to
6.1). These similarities are numerous and not likely confined to Hibernating Mammals That May Have Originated
just the traits listed in Table 6.1. Nevertheless, this extensive from the Retention of Neonatal Traits
list of similarities has given rise to the hypothesis that the abil-
ity of heterothermic mammals to hibernate has evolved from
the retention of neonatal traits and that the genetic potential
for heterothermy is expressed, to some extent, in all neonatal
mammals (2).
Among the striking parallels between neonatal mammals
and adult hibernators is their remarkable tolerance to low
environmental O2 (i.e. hypoxia). While most adult mammals
are not hypoxia tolerant, hibernating mammals are excep-
tional in this regard (4–7) (Figure 6.1A). This was beautifully
demonstrated in the pioneering study of Hiestand et al. (4),
in which survival times were measured in several species of
adult mammals exposed to extremely low levels of environ-
mental O2 (2.8% O2 ; Figure 6.1A). Nonhibernators did quite
poorly, with no species surviving more than 3 minutes (Figure
6.1A). Hibernators, however, survived significantly longer
than nonhibernators, with some species surviving 2.8% O2 for
over 3 hours (4) (Figure 6.1A).
While adult hibernators are far more hypoxia tolerant than
adult nonhibernators, as neonates, both groups are more toler-
ant of hypoxia than their adult counterparts (8–19) (Figure 6.1B).
Thus, postnatal changes occur in hypoxia tolerance of both hiber-
nators and nonhibernators, but the changes are greater in nonhi-
bernating species (Figure 6.1B).
The basis of neonatal hypoxia tolerance and the postnatal
changes that occur in different species have, thus far, attracted
little attention. In this chapter, we focus on recent research
on hibernating species to identify the physiological strategies Source: Data from (4, 7, 12–14, 22, 23, 34, 50, 56, 72–91).

28 DOI: 10.1201/9780367494018-6
Metabolic and Respiratory Adaptations 29

FIGURE 6.1 Hibernators are more hypoxia tolerant than nonhibernators. (A) Survival time in hypoxia of adult nonhibernators and
hibernators exposed to extremely low levels of hypoxia (2.8% O2); modified from (4). (B) Postnatal changes in the survival of nonhi-
bernators (circles) [data from (36)] and predicted trend for hibernators (dotted line) exposed to mild hypoxia (11.8% O2) from birth.

Physiological responses to reduced, then the ventilatory equivalent may remain constant or
even increase, while pulmonary O2 extraction remains the same
hypoxia in adult mammals or even decreases with decreasing levels of inspired O2. Thus,
When mammals are exposed to low O2 environments, their one of the primary characteristics of hypoxia tolerance is rebal-
immediate compensatory response is to hyperventilate (20, 21). ancing O2 demand with O2 supply (6, 38–40). This remarkable
Exposure to hypoxia, particularly in small mammals, produces resilience to O2 deprivation is achieved through controlled, yet
two opposing effects on the drive to breathe, reflecting a balance fully reversible, reductions in O2-demanding processes such as
between: (1) an increase in chemosensitive drive, due to the stim- thermoregulation, excitatory neurotransmitter release, and ion
ulation of the peripheral chemoreceptors and (2) a reduced meta- permeability (channel arrest and action potential spike arrest)
bolic drive, linked to the depression of body temperature and O2 (7, 8, 41). Although it is possible that the hypoxia-induced reduc-
consumption rate (20–22). Thus, hyperventilation in hypoxia may tion in O2 demand is a passive response due to a lack of O2 sup-
occur either through an increase in ventilation (i.e. the hypoxic ply, a large body of evidence has accumulated in support of the
ventilatory response) (20), a decrease in metabolic demand (i.e. hypothesis that the hypoxic metabolic response is a regulated and
the hypoxic metabolic response) (23), or both. reversible oxy-conforming mode of O2 uptake; i.e. it is controlled
The immediate hypoxic response is typically not sustained. metabolic suppression and NOT hypometabolism (an oft misused
Most mammals studied to date elicit a well-documented biphasic term) (42–47).
ventilatory response in hypoxia (20–21). This is particularly true Although the biphasic ventilatory response to hypoxia is typical
of small mammals (24). The first phase of the hypoxic ventila- of mammals, the relative contributions of different components of
tory response is an immediate increase in ventilation within one the response (the components of the Fick equation for pulmonary
breath of a change in arterial O2 levels, mediated by the stimula- exchange; changes in metabolic rate, ventilation, and pulmonary O2
tion of the peripheral chemoreceptors (20–22, 25, 26). The second extraction) can vary depending on many factors, both within and
phase of the ventilatory response begins with a slow decline in between species. These factors include the level of hypoxia, che-
ventilation to a steady lower level. This is mediated by: (1) cen- moreceptor sensitivity, levels of CO2, pH, time course of hypoxic
tral mechanisms, such as the reduction in the thermoregulatory exposure, pattern of repeated exposure, O2 consumption rate, body
set-point — the temperature at which body temperature is regu- temperature, and developmental stage (20–22).
lated, accompanied by a fall in metabolism (26–30); as well as (2) We now take a look at the strategies employed by neonates of
peripheral mechanisms — due to the time dependent decline in hibernating and nonhibernating species. Unlike adults of non-
the sensitivity of the carotid bodies to hypoxic stimuli (26, 31–33). hibernating species, adults of species capable of hibernating are
If metabolic rate is to be maintained in hypoxia, then any fall capable of extreme reductions in metabolism, body temperature,
in ventilation must be accompanied by an increase in pulmonary heart rate, and ventilation. We examine the extent to which the
O2 extraction and would result in a decrease in the ventilatory neonates of rodents ranging in their degree of heterothermic
equivalent or air convection requirement (the ratio of ventila- expression (homeotherm: rats, facultative heterotherms: mice
tion/metabolism) (23, 34–37). Given that the O2 content of each and hamsters, and obligate heterotherms: ground squirrels) differ
breath is progressively reduced in hypoxia extracting more O2 in the balance of metabolic suppression and increased ventilation
per breath is not sustainable. On the other hand, if metabolism is employed under hypoxic conditions.
30 Hypoxic Respiratory Failure in the Newborn

Metabolic and respiratory responses fall in hypoxia (24) (Figure 6.2A). In adult mammals, it appears
that the fall in O2 consumption rates in hypoxia are the result
to hypoxia in neonatal rodents of resetting the thermoregulatory set-point for body temperature
The mass-specific metabolic rates of rodent neonates are similar regulation to a lower level, leading to an inhibition of both shiver-
to those of their mothers, which are extremely low for a mam- ing, and nonshivering thermogenesis (42, 48, 53, 54), as well as
mal of their size. It is only as rodents grow and develop the adult an increase in heat dissipation until body temperature falls to the
capacity for thermogenesis that their mass-specific metabolic new set point (47). The reduction in O2 consumption rate in neo-
rates rise to allometrically predicted levels (3). For neonates of nates, however, occurs despite little or no fall in body tempera-
altricial species, this can take several weeks. At birth, their pri- ture (Figure 6.2A). Whether this body temperature independent
mary response to reduced O2 availability is to reduce O2 demand fall in O2 consumption rate is due to O2 limitation or active sup-
(14, 16, 19, 36, 48). With exposure to progressive hypoxia, neo- pression by some other means is not yet known. Because of the
nates of species with varying degrees of heterothermy all respond lack of thermogenic capacity, cooling cannot be used to deter-
in a similar fashion, with decreases in O2 consumption rates. For mine whether metabolism can be elevated further, and whether
the species shown in Figure 6.2A, decreases range from 35 to this hypoxia-induced reduction in O2 consumption rate is in fact
50% when exposed to a 9% O2 gas mixture. Accompanying these controlled.
large falls in O2 demand are modest increases in ventilation (40 to In summary, neonates of all rodent species, whether hibernat-
100% for the species in Figure 6.2A). In general, the increases in ing or nonhibernating primarily respond to reduced O2 avail-
ventilation have been reported to vary in an inverse manner with ability in hypoxia with modest increases in ventilation and in
the falls in metabolic rate (24, 49–51). These hypoxia-induced pulmonary O2 extraction, and large decreases in their need for
increases in ventilation are primarily the result of increases in O2 by decreasing metabolism. It is not clear whether their ven-
breathing frequency, as tidal volume either increases slightly, tilatory responses are inadequate and the fall in metabolism is
decreases, or remains constant (24). due to limited O2 availability, or whether this is an orchestrated
The balance between changes in ventilation and changes in O2 reduction in metabolism that is not quite sufficient to accommo-
consumption is reflected in the ventilatory equivalent (the ratio date the reduced O2 supply resulting in modest increases in ven-
of ventilation/O2 consumption rate). Under normoxic conditions, tilation and in pulmonary O2 extraction. Metabolic suppression
changes in O2 consumption rate (such as during exercise) are in response to low O2 conditions becomes disadvantageous when
matched by changes in ventilation and the ratio remains relatively hypoxic conditions persist. In the developing rodent, prolonged
constant. This is not the case in hypoxia, as the amount of O2 in O2 consumption rate suppression inhibits tissue and organ
each breath decreases. Whether the response to hypoxia is an growth, tissue differentiation, and cell repair (55).
increase in ventilation or a fall in O2 consumption rates, the ratio
between the two will increase and the % change in this ratio will Metabolic and respiratory responses
reflect the % change in the O2 composition of the inspired gas. to hypoxia in adult rodents
If the amount of O2 extracted from each breath were to remain Effects of progressive hypoxia within
constant, for animals breathing 9% O2 the ventilatory equiva- the thermoneutral zone
lent should increase by 133% (as indicated by the dotted line Similar to neonatal rodents, adult rodents also respond to hypoxia
in Figure 6.2). As described by the Fick equation [O2 consumption by reducing O2 consumption rates, to some extent. The magni-
rate = total ventilation × (fraction of O2 in inspired air – fraction tude of this reduction has been shown to be size dependent, being
of O2 in expired air)], any difference in these ratios from 133% must greater in smaller mammals (24). The magnitude of the hypoxic
be due to changes in the amount of O2 extracted from the lung metabolic response has also been shown to be greater in adult
with each breath. The neonates of the species shown in Figure 6.2 hibernators than in adult nonhibernators (23, 56), although the
when breathing 9% O2 exhibit increases ranging from 108 to extent of this difference is temperature dependent (see next sec-
307% spanning the ratio of the change in O2 content of inspired tion). The species of rodents shown in Figure 6.2B are all relatively
gas (133%) required to match O2 supply and demand based on small (i.e. less than a kilogram); and exhibit significant falls in
changes in O2 consumption rate and ventilation alone. The high- O2 consumption rate when inspiring 9% O2. The decreases in O2
est value is for the rat and reflects a significant decrease in pulmo- consumption rates of hibernating species within their thermo-
nary O2 extraction. The lowest value is in the mouse and reflects neutral zones are marginally (but not significantly) greater than
an increase in pulmonary O2 extraction. Pulmonary O2 extrac- in the non-hibernating rat. The magnitude of these reductions
tion remains relatively constant in hamsters and ground squirrels in O2 consumption rates, however, are also significantly reduced
(Figure 6.2A). The differences seen in pulmonary O2 extraction compared to the reductions seen in the neonates of the same spe-
in the different species could reflect species differences in lung cies (Figure 6.2A, B).
morphology, lung perfusion, or the carrying capacity of the blood. As mentioned above, the hypoxic metabolic response in
The greater hemoglobin concentration and hemoglobin-O2 affin- adult mammals appears to initially be mediated by a reduc-
ity of neonatal rodents undoubtedly contribute to their reported tion in the thermoregulatory set-point, accompanied by a fall in
blunted hypoxic ventilatory responses (52). On the one hand, this metabolism (26-30). If exposure to hypoxia is prolonged, there
indicates that hibernators maintain a greater diffusion gradient is a time-dependent decline in the sensitivity of the carotid
for O2 uptake at the lung. On the other hand, it indicates that the bodies to hypoxic stimuli (26, 31–33). Thus, in adult rats, it has
differences in the magnitude of the ventilatory responses seen in been shown that there are time domains to the hypoxic meta-
the different species are largely due to the differences in pulmo- bolic response, that the immediate suppression of metabolism
nary O2 extraction and not to differences in O2 chemosensitivity. in hypoxia is not sustained, and that the magnitude of the
Given the poor thermogenic capacity of neonatal rodents, their hypoxic metabolic response decreases over time in an ambient
body temperatures approximate ambient temperature when held temperature-dependent fashion (57, 58). To our knowledge, this
in isolation. Thus, there is little scope for body temperature to has not been examined in other species.
Another random document with
no related content on Scribd:
PLEASE READ THIS BEFORE YOU DISTRIBUTE OR USE THIS WORK

To protect the Project Gutenberg™ mission of promoting the


free distribution of electronic works, by using or distributing this
work (or any other work associated in any way with the phrase
“Project Gutenberg”), you agree to comply with all the terms of
the Full Project Gutenberg™ License available with this file or
online at www.gutenberg.org/license.

Section 1. General Terms of Use and


Redistributing Project Gutenberg™
electronic works
1.A. By reading or using any part of this Project Gutenberg™
electronic work, you indicate that you have read, understand,
agree to and accept all the terms of this license and intellectual
property (trademark/copyright) agreement. If you do not agree to
abide by all the terms of this agreement, you must cease using
and return or destroy all copies of Project Gutenberg™
electronic works in your possession. If you paid a fee for
obtaining a copy of or access to a Project Gutenberg™
electronic work and you do not agree to be bound by the terms
of this agreement, you may obtain a refund from the person or
entity to whom you paid the fee as set forth in paragraph 1.E.8.

1.B. “Project Gutenberg” is a registered trademark. It may only


be used on or associated in any way with an electronic work by
people who agree to be bound by the terms of this agreement.
There are a few things that you can do with most Project
Gutenberg™ electronic works even without complying with the
full terms of this agreement. See paragraph 1.C below. There
are a lot of things you can do with Project Gutenberg™
electronic works if you follow the terms of this agreement and
help preserve free future access to Project Gutenberg™
electronic works. See paragraph 1.E below.
1.C. The Project Gutenberg Literary Archive Foundation (“the
Foundation” or PGLAF), owns a compilation copyright in the
collection of Project Gutenberg™ electronic works. Nearly all the
individual works in the collection are in the public domain in the
United States. If an individual work is unprotected by copyright
law in the United States and you are located in the United
States, we do not claim a right to prevent you from copying,
distributing, performing, displaying or creating derivative works
based on the work as long as all references to Project
Gutenberg are removed. Of course, we hope that you will
support the Project Gutenberg™ mission of promoting free
access to electronic works by freely sharing Project
Gutenberg™ works in compliance with the terms of this
agreement for keeping the Project Gutenberg™ name
associated with the work. You can easily comply with the terms
of this agreement by keeping this work in the same format with
its attached full Project Gutenberg™ License when you share it
without charge with others.

1.D. The copyright laws of the place where you are located also
govern what you can do with this work. Copyright laws in most
countries are in a constant state of change. If you are outside
the United States, check the laws of your country in addition to
the terms of this agreement before downloading, copying,
displaying, performing, distributing or creating derivative works
based on this work or any other Project Gutenberg™ work. The
Foundation makes no representations concerning the copyright
status of any work in any country other than the United States.

1.E. Unless you have removed all references to Project


Gutenberg:

1.E.1. The following sentence, with active links to, or other


immediate access to, the full Project Gutenberg™ License must
appear prominently whenever any copy of a Project
Gutenberg™ work (any work on which the phrase “Project
Gutenberg” appears, or with which the phrase “Project
Gutenberg” is associated) is accessed, displayed, performed,
viewed, copied or distributed:

This eBook is for the use of anyone anywhere in the United


States and most other parts of the world at no cost and with
almost no restrictions whatsoever. You may copy it, give it
away or re-use it under the terms of the Project Gutenberg
License included with this eBook or online at
www.gutenberg.org. If you are not located in the United
States, you will have to check the laws of the country where
you are located before using this eBook.

1.E.2. If an individual Project Gutenberg™ electronic work is


derived from texts not protected by U.S. copyright law (does not
contain a notice indicating that it is posted with permission of the
copyright holder), the work can be copied and distributed to
anyone in the United States without paying any fees or charges.
If you are redistributing or providing access to a work with the
phrase “Project Gutenberg” associated with or appearing on the
work, you must comply either with the requirements of
paragraphs 1.E.1 through 1.E.7 or obtain permission for the use
of the work and the Project Gutenberg™ trademark as set forth
in paragraphs 1.E.8 or 1.E.9.

1.E.3. If an individual Project Gutenberg™ electronic work is


posted with the permission of the copyright holder, your use and
distribution must comply with both paragraphs 1.E.1 through
1.E.7 and any additional terms imposed by the copyright holder.
Additional terms will be linked to the Project Gutenberg™
License for all works posted with the permission of the copyright
holder found at the beginning of this work.

1.E.4. Do not unlink or detach or remove the full Project


Gutenberg™ License terms from this work, or any files
containing a part of this work or any other work associated with
Project Gutenberg™.
1.E.5. Do not copy, display, perform, distribute or redistribute
this electronic work, or any part of this electronic work, without
prominently displaying the sentence set forth in paragraph 1.E.1
with active links or immediate access to the full terms of the
Project Gutenberg™ License.

1.E.6. You may convert to and distribute this work in any binary,
compressed, marked up, nonproprietary or proprietary form,
including any word processing or hypertext form. However, if
you provide access to or distribute copies of a Project
Gutenberg™ work in a format other than “Plain Vanilla ASCII” or
other format used in the official version posted on the official
Project Gutenberg™ website (www.gutenberg.org), you must, at
no additional cost, fee or expense to the user, provide a copy, a
means of exporting a copy, or a means of obtaining a copy upon
request, of the work in its original “Plain Vanilla ASCII” or other
form. Any alternate format must include the full Project
Gutenberg™ License as specified in paragraph 1.E.1.

1.E.7. Do not charge a fee for access to, viewing, displaying,


performing, copying or distributing any Project Gutenberg™
works unless you comply with paragraph 1.E.8 or 1.E.9.

1.E.8. You may charge a reasonable fee for copies of or


providing access to or distributing Project Gutenberg™
electronic works provided that:

• You pay a royalty fee of 20% of the gross profits you derive from
the use of Project Gutenberg™ works calculated using the
method you already use to calculate your applicable taxes. The
fee is owed to the owner of the Project Gutenberg™ trademark,
but he has agreed to donate royalties under this paragraph to
the Project Gutenberg Literary Archive Foundation. Royalty
payments must be paid within 60 days following each date on
which you prepare (or are legally required to prepare) your
periodic tax returns. Royalty payments should be clearly marked
as such and sent to the Project Gutenberg Literary Archive
Foundation at the address specified in Section 4, “Information
about donations to the Project Gutenberg Literary Archive
Foundation.”

• You provide a full refund of any money paid by a user who


notifies you in writing (or by e-mail) within 30 days of receipt that
s/he does not agree to the terms of the full Project Gutenberg™
License. You must require such a user to return or destroy all
copies of the works possessed in a physical medium and
discontinue all use of and all access to other copies of Project
Gutenberg™ works.

• You provide, in accordance with paragraph 1.F.3, a full refund of


any money paid for a work or a replacement copy, if a defect in
the electronic work is discovered and reported to you within 90
days of receipt of the work.

• You comply with all other terms of this agreement for free
distribution of Project Gutenberg™ works.

1.E.9. If you wish to charge a fee or distribute a Project


Gutenberg™ electronic work or group of works on different
terms than are set forth in this agreement, you must obtain
permission in writing from the Project Gutenberg Literary
Archive Foundation, the manager of the Project Gutenberg™
trademark. Contact the Foundation as set forth in Section 3
below.

1.F.

1.F.1. Project Gutenberg volunteers and employees expend


considerable effort to identify, do copyright research on,
transcribe and proofread works not protected by U.S. copyright
law in creating the Project Gutenberg™ collection. Despite
these efforts, Project Gutenberg™ electronic works, and the
medium on which they may be stored, may contain “Defects,”
such as, but not limited to, incomplete, inaccurate or corrupt
data, transcription errors, a copyright or other intellectual
property infringement, a defective or damaged disk or other
medium, a computer virus, or computer codes that damage or
cannot be read by your equipment.

1.F.2. LIMITED WARRANTY, DISCLAIMER OF DAMAGES -


Except for the “Right of Replacement or Refund” described in
paragraph 1.F.3, the Project Gutenberg Literary Archive
Foundation, the owner of the Project Gutenberg™ trademark,
and any other party distributing a Project Gutenberg™ electronic
work under this agreement, disclaim all liability to you for
damages, costs and expenses, including legal fees. YOU
AGREE THAT YOU HAVE NO REMEDIES FOR NEGLIGENCE,
STRICT LIABILITY, BREACH OF WARRANTY OR BREACH
OF CONTRACT EXCEPT THOSE PROVIDED IN PARAGRAPH
1.F.3. YOU AGREE THAT THE FOUNDATION, THE
TRADEMARK OWNER, AND ANY DISTRIBUTOR UNDER
THIS AGREEMENT WILL NOT BE LIABLE TO YOU FOR
ACTUAL, DIRECT, INDIRECT, CONSEQUENTIAL, PUNITIVE
OR INCIDENTAL DAMAGES EVEN IF YOU GIVE NOTICE OF
THE POSSIBILITY OF SUCH DAMAGE.

1.F.3. LIMITED RIGHT OF REPLACEMENT OR REFUND - If


you discover a defect in this electronic work within 90 days of
receiving it, you can receive a refund of the money (if any) you
paid for it by sending a written explanation to the person you
received the work from. If you received the work on a physical
medium, you must return the medium with your written
explanation. The person or entity that provided you with the
defective work may elect to provide a replacement copy in lieu
of a refund. If you received the work electronically, the person or
entity providing it to you may choose to give you a second
opportunity to receive the work electronically in lieu of a refund.
If the second copy is also defective, you may demand a refund
in writing without further opportunities to fix the problem.

1.F.4. Except for the limited right of replacement or refund set


forth in paragraph 1.F.3, this work is provided to you ‘AS-IS’,
WITH NO OTHER WARRANTIES OF ANY KIND, EXPRESS
OR IMPLIED, INCLUDING BUT NOT LIMITED TO
WARRANTIES OF MERCHANTABILITY OR FITNESS FOR
ANY PURPOSE.

1.F.5. Some states do not allow disclaimers of certain implied


warranties or the exclusion or limitation of certain types of
damages. If any disclaimer or limitation set forth in this
agreement violates the law of the state applicable to this
agreement, the agreement shall be interpreted to make the
maximum disclaimer or limitation permitted by the applicable
state law. The invalidity or unenforceability of any provision of
this agreement shall not void the remaining provisions.

1.F.6. INDEMNITY - You agree to indemnify and hold the


Foundation, the trademark owner, any agent or employee of the
Foundation, anyone providing copies of Project Gutenberg™
electronic works in accordance with this agreement, and any
volunteers associated with the production, promotion and
distribution of Project Gutenberg™ electronic works, harmless
from all liability, costs and expenses, including legal fees, that
arise directly or indirectly from any of the following which you do
or cause to occur: (a) distribution of this or any Project
Gutenberg™ work, (b) alteration, modification, or additions or
deletions to any Project Gutenberg™ work, and (c) any Defect
you cause.

Section 2. Information about the Mission of


Project Gutenberg™
Project Gutenberg™ is synonymous with the free distribution of
electronic works in formats readable by the widest variety of
computers including obsolete, old, middle-aged and new
computers. It exists because of the efforts of hundreds of
volunteers and donations from people in all walks of life.

Volunteers and financial support to provide volunteers with the


assistance they need are critical to reaching Project
Gutenberg™’s goals and ensuring that the Project Gutenberg™
collection will remain freely available for generations to come. In
2001, the Project Gutenberg Literary Archive Foundation was
created to provide a secure and permanent future for Project
Gutenberg™ and future generations. To learn more about the
Project Gutenberg Literary Archive Foundation and how your
efforts and donations can help, see Sections 3 and 4 and the
Foundation information page at www.gutenberg.org.

Section 3. Information about the Project


Gutenberg Literary Archive Foundation
The Project Gutenberg Literary Archive Foundation is a non-
profit 501(c)(3) educational corporation organized under the
laws of the state of Mississippi and granted tax exempt status by
the Internal Revenue Service. The Foundation’s EIN or federal
tax identification number is 64-6221541. Contributions to the
Project Gutenberg Literary Archive Foundation are tax
deductible to the full extent permitted by U.S. federal laws and
your state’s laws.

The Foundation’s business office is located at 809 North 1500


West, Salt Lake City, UT 84116, (801) 596-1887. Email contact
links and up to date contact information can be found at the
Foundation’s website and official page at
www.gutenberg.org/contact

Section 4. Information about Donations to


the Project Gutenberg Literary Archive
Foundation
Project Gutenberg™ depends upon and cannot survive without
widespread public support and donations to carry out its mission
of increasing the number of public domain and licensed works
that can be freely distributed in machine-readable form
accessible by the widest array of equipment including outdated
equipment. Many small donations ($1 to $5,000) are particularly
important to maintaining tax exempt status with the IRS.

The Foundation is committed to complying with the laws


regulating charities and charitable donations in all 50 states of
the United States. Compliance requirements are not uniform
and it takes a considerable effort, much paperwork and many
fees to meet and keep up with these requirements. We do not
solicit donations in locations where we have not received written
confirmation of compliance. To SEND DONATIONS or
determine the status of compliance for any particular state visit
www.gutenberg.org/donate.

While we cannot and do not solicit contributions from states


where we have not met the solicitation requirements, we know
of no prohibition against accepting unsolicited donations from
donors in such states who approach us with offers to donate.

International donations are gratefully accepted, but we cannot


make any statements concerning tax treatment of donations
received from outside the United States. U.S. laws alone swamp
our small staff.

Please check the Project Gutenberg web pages for current


donation methods and addresses. Donations are accepted in a
number of other ways including checks, online payments and
credit card donations. To donate, please visit:
www.gutenberg.org/donate.

Section 5. General Information About Project


Gutenberg™ electronic works
Professor Michael S. Hart was the originator of the Project
Gutenberg™ concept of a library of electronic works that could
be freely shared with anyone. For forty years, he produced and
distributed Project Gutenberg™ eBooks with only a loose
network of volunteer support.

Project Gutenberg™ eBooks are often created from several


printed editions, all of which are confirmed as not protected by
copyright in the U.S. unless a copyright notice is included. Thus,
we do not necessarily keep eBooks in compliance with any
particular paper edition.

Most people start at our website which has the main PG search
facility: www.gutenberg.org.

This website includes information about Project Gutenberg™,


including how to make donations to the Project Gutenberg
Literary Archive Foundation, how to help produce our new
eBooks, and how to subscribe to our email newsletter to hear
about new eBooks.

You might also like