Download as pdf or txt
Download as pdf or txt
You are on page 1of 54

Stroke: Pathophysiology, Diagnosis,

And Management 7th Edition James


Grotta
Visit to download the full and correct content document:
https://ebookmass.com/product/stroke-pathophysiology-diagnosis-and-management-
7th-edition-james-grotta/
More products digital (pdf, epub, mobi) instant
download maybe you interests ...

Stroke: Pathophysiology, Diagnosis, And Management 7e


2021 7th Edition James Grotta

https://ebookmass.com/product/stroke-pathophysiology-diagnosis-
and-management-7e-2021-7th-edition-james-grotta/

Sex Differences in Cardiac Diseases: Pathophysiology,


Presentation, Diagnosis and Management Niti R. Aggarwal

https://ebookmass.com/product/sex-differences-in-cardiac-
diseases-pathophysiology-presentation-diagnosis-and-management-
niti-r-aggarwal/

Goodwin and Guze’s Psychiatric Diagnosis 7th Edition

https://ebookmass.com/product/goodwin-and-guzes-psychiatric-
diagnosis-7th-edition/

Critical Care Nursing: Diagnosis and Management 8th


Edition

https://ebookmass.com/product/critical-care-nursing-diagnosis-
and-management-8th-edition/
Differential Diagnosis of Common Complaints 7th Edition

https://ebookmass.com/product/differential-diagnosis-of-common-
complaints-7th-edition/

Goodwin and Guze's Psychiatric Diagnosis 7th Edition


Carol S. North

https://ebookmass.com/product/goodwin-and-guzes-psychiatric-
diagnosis-7th-edition-carol-s-north/

Yen & Jaffe’s Reproductive Endocrinology: Physiology,


Pathophysiology, and Clinical Management, 8e Yen &
Jaffe’s Reproductive Endocrinology: Physiology,
Pathophysiology, and Clinical Management, 8e 2018 8th
Edition 8E Yen & Jaffe’S Reproductive Endocrinology:
https://ebookmass.com/product/yen-jaffes-reproductive-
endocrinology-physiology-pathophysiology-and-clinical-
Physiology
management-8e-yen-jaffes-reproductive-endocrinology-physiology-
pathophysiology-and-clinical-m/

Stroke Care Harwood

https://ebookmass.com/product/stroke-care-harwood/

Urgent Care Dermatology: Symptom-Based Diagnosis James


E. Fitzpatrick

https://ebookmass.com/product/urgent-care-dermatology-symptom-
based-diagnosis-james-e-fitzpatrick/
Stroke
Pathophysiology, Diagnosis, and Management

SEVENTH EDITION

JAMES C. GROTTA, MD ENG H. LO, PhD


Director of Stroke Research and Mobile Stroke Unit Professor of Neurology and Radiology
Clinical Innovation and Research Institute Harvard Medical School
Memorial Hermann Hospital-Texas Medical Center Boston, Massachusetts
Houston, Texas Director, Neuroprotection Research Laboratories
Massachusetts General Hospital
Charlestown, Massachusetts
GREGORY W. ALBERS, MD
Professor
Department of Neurology and Neurological Sciences RALPH L. SACCO, MD
Stanford University Chairman, Department of Neurology
Stanford, California Olemberg Family Chair in Neurological Disorders
Miller Professor of Neurology, Public Health Sciences,
Human Genetics, and Neurosurgery
JOSEPH P. BRODERICK, MD University of Miami Miller School of Medicine
Professor Chief of Service, Neurology
Department of Neurology and Rehabilitation Medicine Jackson Health System
University of Cincinnati Gardner Neuroscience Institute Miami, Florida
Cincinnati, Ohio

LAWRENCE K.S. WONG, MD


ARTHUR L. DAY, MD Professor
Professor and Co-Chairman Department of Medicine and Therapeutics
Director of Cerebrovascular Surgery Chinese University of Hong Kong
Residency Program Director Shatin, Hong Kong, China
Department of Neurosurgery
University of Texas Medical School at Houston
Houston, Texas

SCOTT E. KASNER, MD
Professor, Department of Neurology
University of Pennsylvania
Director, Comprehensive Stroke Center
University of Pennsylvania Health System
Philadelphia, Pennsylvania
Elsevier
1600 John F. Kennedy Blvd.
Ste 1600
Philadelphia, PA 19103-2899

STROKE: PATHOPHYSIOLOGY, DIAGNOSIS, AND MANAGEMENT:  ISBN: 978-0-323-69424-7


SEVENTH EDITION
Copyright © 2022 by Elsevier, Inc. All rights reserved.

No part of this publication may be reproduced or transmitted in any form or by any means, electronic or
mechanical, including photocopying, recording, or any information storage and retrieval system, without
permission in writing from the publisher. Details on how to seek permission, further information about the
Publisher’s permissions policies, and our arrangements with organizations such as the Copyright Clearance
Center and the Copyright Licensing Agency can be found at our website: www.elsevier.com/permissions.

This book and the individual contributions contained in it are protected under copyright by the Publisher
(other than as may be noted herein).

Notice

Practitioners and researchers must always rely on their own experience and knowledge in evaluating and
using any information, methods, compounds, or experiments described herein. Because of rapid advances
in the medical sciences in particular, independent verification of diagnoses and drug dosages should be
made. To the fullest extent of the law, no responsibility is assumed by Elsevier, authors, editors, or con-
tributors for any injury and/or damage to persons or property as a matter of products liability, negligence
or otherwise, or from any use or operation of any methods, products, instructions, or ideas contained in
the material herein.

Previous editions copyrighted © 2016 by Elsevier Inc; 2011 by Saunders, an imprint of Elsevier Inc; 2004, 1998,
1992, 1986 by Churchill Livingstone.

Library of Congress Control Number: 2020952390

Senior Acquisitions Editor: Melanie Tucker


Senior Content Development Specialist: Ann Ruzycka Anderson
Publishing Services Manager: Catherine Albright Jackson
Senior Project Manager: Doug Turner
Designer: Ryan Cook

Printed in the United States of America

Last digit is the print number: 9 8 7 6 5 4 3 2 1


Video Contents

Video 30.1 Important concepts regarding Video 32.9 TTE of large posterior mitral annular
angioarchitecture, classification, and risk calcification (MAC) with central echo lucency,
factors for arteriovenous malformations. suggesting caseous MAC.
Video 30.2 Endovascular embolization of a frontal Video 33.1 Large atherosclerotic plaque in the mid-portion
arteriovenous malformation. of the aortic arch with large superimposed,
Video 30.3 Endovascular and surgical techniques in the mobile thrombus (also Fig. 33.6).
treatment of dural arteriovenous fistulae. Video 33.2 Mobile aortic plaque in the superior aspect of
Video 32.1 Mobile atheroma: An intraoperative video 3D the aortic arch, just proximal to the takeoff of
TEE image of a mobile atheroma in the aortic the innominate artery (also Fig. 33.10A and B).
arch, visualized during a transcatheter aortic Video 35.1 3D reconstruction of carotid angiography
valve replacement (TAVR), consistent with in a 40-year-old woman with right-sided
grade 5 atheromatous disease. numbness. Anterior, posterior, and left lateral
Video 32.2 (A) Positive bubble study: Markedly positive rotational video of the left internal carotid
bubble study consistent with a large atrial artery demonstrates a web.
level shunt. (B) Negative bubble study: Video 60.1 Knee-ankle-foot-orthosis: A knee-ankle-foot
Agitated saline contrast (“bubble”) study orthosis enabled this hemiplegic person with
showing no evidence of an interatrial sensorimotor loss to prevent the knee from
communication. snapping back in the mid- to late stance phase
Video 32.3 TEE bubble study with intrapulmonary of gait, prevent catching the forefoot when
shunt: A bubble study on TEE with delayed initiating swing, and improved control of the
passage of bubbles into the left atrium via the stability of foot placement during the stance
right upper pulmonary vein, suggesting an phase.
intrapulmonary shunt, such as a pulmonary Video 72.1 Minimally invasive endoscopic ICH
atriovenous malformation. evacuation. The procedure begins with a
Video 32.4 Takotsubo cardiomyopathy: TTE revealing 1.5-cm incision, a 1-cm craniectomy, and
akinesis of mid- and apical left ventricular stereotactic placement of a 19F (6.3-mm)
segments with hypercontractile basal sheath into the hematoma. The introducer
segments, consistent with a stress-mediated is then removed from the sheath, and the
cardiomyopathy (Takotsubo cardiomyopathy). endoscope is placed down the sheath with
an adjunctive aspiration device inserted
Video 32.5 Mobile atrial septum with positive bubble in the working channel of the endoscope.
study: Mobile atrial septum, consistent with
Continuous irrigation with lactated Ringers is
an atrial septal aneurysm. Agitated saline
used throughout the procedure.
contrast (“bubble”) study reveals passage
of bubbles into the left heart after the atrial Video 72.2 Minimally invasive endoport-mediated ICH
septum bulges into the left atrium, suggesting evacuation. The procedure begins with a
a transient increase in right atrial pressure. 4-cm incision, a 2.5-cm craniotomy, and
This finding is consistent with the presence of stereotactic placement of a 13.5-cm endoport
a patent foramen ovale (PFO). into the hematoma. The introducer is
then removed from the endoport, and the
Video 32.6 TTE of a large left atrial myxoma, filling the
endoport is fixed in place. A microscope or
entire left atrium and resulting in obstruction
an exoscope is then positioned to provide
to left ventricular filling.
visibility down the endoport and into the
Video 32.7 Papillary fibroelastoma: TEE of the aortic cavity.
valve revealing a linear echo density on the
Video 75.1 Surgical management of a cerebral cavernous
ventricular surface of the valve, consistent
malformation.
with a papillary fibroelastoma.
Video 32.8 2D (A) and 3D (B) images of a large burden of Video 77.1 Superficial temporal artery to middle cerebral
artery bypass.
clot in the left atrium and left atrial appendage
in a patient with a cardiomyopathy and atrial Video 78.1 Decompressive craniectomy in a patient with
flutter, presenting with a stroke. malignant middle cerebral artery infarction.

ix
Foreword to the Seventh Edition
This original editor continues to marvel at the advances in The growing participation in stroke management by
the field of stroke, justifying the seventh edition of this book. those in allied fields has done nothing to displace the role
Among other topics, the first edition had a mere 15 chap- of neurovascular clinicians, whose commitment includes
ters, and in 347 pages covered “Stroke Therapy.” The subjects studying how the brain works. Insights from modern basic
ranged from management of risk factors to rehabilitation. biology, increasingly sophisticated imaging, prospective
This edition has no less than 28 such chapters, clustered in clinically detailed databases, and even access to video Zoom
stand-alone sections for medical and interventional therapy. follow-ups are providing windows into what was formerly
The page length for all of subjects has steadily expanded by called semiology. Decades ago, the neurology literature was
editions: proof, if needed, of progress. Gone—and good rid- dotted with titles beginning with “The Neurology of…” by
dance!—are the days when those interested in stroke were which the author(s) implied how a clinical syndrome allowed
considered clinically irrelevant for lack of definitive therapies. insight into diagnosis or prognosis. Today, a surprising number
Instead, far from an arcane subspecialty, stroke prevention of outcomes for acute focal syndromes formerly considered
and management now has an impact on the clinical practice static, prevented from their full development, or deemed
of many medical and surgical fields whose training not long modified favorably by acute interventions, are yielding insights
ago scarcely touched on the subject. into the mysteries of functional reorganization. The increasing
Stroke clinicians now find their clinical judgment opportunities to understand this effect offers literature-
tested—sometimes to their vexation—by the application of oriented neurovascular clinicians the chance to be links in an
hyperacute management algorithms driven mainly by scor- unbroken chain of inquiry dating back to antiquity.
ing systems, meta-analyses, and outcomes from the wave of
clinical trials. Few can argue with the positive effect of rapid J.P. Mohr, MD
assessment and intervention, especially for acute ischemic Daniel Sciarra Professor of Neurology
strokes. Insights spanning genetics, basic biology, computer- Department of Neurology
driven population studies, web-based meta-analyses, and Director and Neurologist
increasingly common longitudinal outcome reports are wel- Doris and Stanley Tanenbaum Stroke Center
come signs of progress. Only novelists should designate their Columbia University Irving Medical Center
published work free from revision. The current contributors New York, New York
can expect further changes to justify an eighth edition in the
foreseeable future.

x
Preface
The seventh edition of the text has a number of important Other unsolved areas that receive substantial updating
changes. First, this edition has even more on-line features, include intracerebral hemorrhage by new author Dr. Anderson
making it easier to access its content in a digital-friendly for- and arteriovenous malformations by Drs. Samaniego, Roa,
mat. The eBook includes the entire book plus full reference Ortega-Gutierrez, and Derdeyn. In addition, the chapters
lists (as opposed to the Key References that appear in the chap- on the surgical management of different types of brain
ters) and a larger number of videos than the previous edition. hemorrhage have all been updated by new authors.
Access to the Expert Consult eBook version is included with The previous edition appeared just as the trials demonstrating
print purchase. This enhanced eBook experience allows you the benefit of endovascular thrombectomy were published, so
to search all of the text, figures, and references on a variety of the coverage of this revolution in treatment was incomplete.
devices. The content can also be downloaded to tablets and In this edition, Dr. Broderick’s section, Interventional Therapy,
smart phones for offline use. and in particular the chapter by Drs. Saver and Jahn on the
Another important change includes our new Surgical endovascular treatment of acute ischemic stroke, have been
Therapy section editor, Arthur Day, MD. Dr. Day is an substantially updated to include the results of all those pivotal
international authority on the surgical management of clinical trials, as well as the myriad studies that followed.
cerebral aneurysms, intracranial hemorrhage, and extracranial The final unresolved topic receiving increased coverage
vascular disease. He is the recipient of numerous neurosurgical in this edition is how best to deliver these effective new
leadership awards, and from first-hand experience I can attest treatments (e.g., stroke systems of care). We have added a new
to his passion for teaching and the wisdom that has grown chapter on this topic, written by Drs. Czap, Harmel, Audebert,
out of decades of skillfully managing the complexities of the and myself, that explores different models and approaches to
entire array of neurovascular surgical cases. Of particular value reorganizing our stroke centers, resources, and staffing. and
for his role as editor, Dr. Day has been an important leader In addition, first-time contributors to this title, Drs. Kircher
of the neurosurgical field as it has emerged from open to and Adeoye, have expanded the chapter on prehospital and
endovascular approaches and as it has partnered with vascular emergency care.
neurology in the conduct of clinical trials. As a result of his While I have focused my editorial spotlight on a few of the
intimate knowledge of the entire neurovascular landscape and major unresolved topics that are receiving substantial and well-
its leaders, you will see that the authors of almost all of the deserved increased attention, I want to emphasize that each and
chapters in the Surgical Therapy section have changed and the every chapter has been updated with new information. There
chapters have all been updated. I think that the readers will be is a new chapter on posterior reversible encephalopathy, which
impressed by the combined experience, fresh perspective, and replaces the old chapter on hypertensive encephalopathy with
new information in every chapter in the section. new authors (Drs. Balu and Fischer); the imaging chapters on
Other notable changes in this edition justified enlarging CT and MRI have been updated, with expanded discussion of
attention given to several underappreciated and yet unresolved the important role of imaging in patient selection for acute
problems in the field. In line with the increasing evidence of therapy; the chapters on cardiac disease, cryptogenic stroke,
vascular disease as the most important modifiable contributor and secondary prevention provide more information on
to dementia and much-needed attention to the biology atrial fibrillation detection, other possible causes of embolic-
underlying small vessel disease, a new chapter on this topic appearing stroke without known source, and their long
has been added to the Pathophysiology section, which has term management; the antiplatelet therapy chapter includes
been overseen by the senior editor, Dr. Lo. In addition, the updated data from recent trials of dual antiplatelet therapy;
chapters on the clinical aspects of vascular dementia and and the design of stroke clinical trials chapter has been
small vessel disease have been updated by new authors (Drs. rewritten by new authors (Drs. Perez, Elm, and Saver) and
Rundek, Seshadri, and Caunca) in the Epidemiology and Risk includes emerging novel approaches to figuring out if new
Factors section, and important new information is found in the treatments work.
chapters on genetics and CADASIL. Somewhat linked to this All in all, I hope that the exciting relevant new data that fill
topic and also reflecting a maturing interest in non-imaging the pages of our journals and make stroke such a dynamic and
stroke biomarkers in general is an entirely new chapter on interesting field are distilled into these pages in a readable and
“OMICs,” written by Drs. Jickling and Sharp. authoritative format that will help the reader understand their
Disparities in stroke incidence and outcomes has become patients and their underlying disease, which they see every
a hot topic, accentuated recently by the spotlight cast on this day, and also provide the foundation for new knowledge that
issue during the COVID-19 pandemic and the racial unrest in will be the substrate for the next edition.
the United States. The already outstanding chapter on stroke
disparities by Drs. Howard, Howard, and McCullough has James C. Grotta, MD
been updated, and this topic has also been woven through
other chapters where relevant.

xi
Contributors

Harold P. Adams Jr., MD Hugo J. Aparicio, MD, MPH Hakan Ay, MD


Professor Assistant Professor Associate Professor
Department of Neurology Department of Neurology Departments of Neurology and
Carver College of Medicine Boston University School of Medicine Radiology
University of Iowa Investigator, The Framingham Heart Massachusetts General Hospital
Iowa City, Iowa Study Harvard Medical School
Boston, Massachusetts Boston, Massachusetts
Opeolu Adeoye, MD
Takeda Pharmaceutical Company
Vice Chair, Research Ken Arai, PhD
Limited
Co-Director, UC Stroke Team Associate Professor
Cambridge, Massachusetts
Professor Neuroprotection Research Laboratory
Department of Emergency Medicine Departments of Radiology and Selva Baltan, MD, PhD
University of Cincinnati Neurology Professor and Vice Chair for Basic
Cincinnati, Ohio Massachusetts General Hospital Research
Harvard Medical School Anesthesiology and Peri-Operative
Gregory W. Albers, MD
Boston, Massachusetts Medicine
Professor
Oregon Health and Science University
Department of Neurology and Jaroslaw Aronowski, PhD, MD
School of Medicine
Neurological Sciences Professor and Vice Chair
Portland, Oregon
Stanford University Roy M. and Phyllis Gough Huffington
Stanford, California Chair in Neurology Ramani Balu, MD, PhD
McGovern Medical School Assistant Professor
Andrei V. Alexandrov, MD, RVT
University of Texas Health Science Division of Neurocritical Care
Semmes-Murphey Professor and Chairman
Center at Houston Department of Neurology
Department of Neurology
Houston, Texas University of Pennsylvania
The University of Tennessee Health
Philadelphia, Pennsylvania
Science Center Kunakorn Atchaneeyasakul, MD
Memphis, Tennessee Clinical Instructor of Neurology Mandana Behbahani, MD
StrokeNet Fellow Resident
Sepideh Amin-Hanjani, MD
Department of Neurology Department of Neurosurgery
Professor and Program Director
University of Pittsburg School of University of Illinois at Chicago
Department of Neurosurgery
Medicine Chicago, Illinois
Co-Director, Neurovascular Surgery
Pittsburg, Pennsylvania
University of Illinois at Chicago Oscar R. Benavente, MD
Stroke Vascular Fellow
Chicago, Illinois Professor
UCLA Health
Department of Medicine
Hongyu An, PhD Los Angeles, California
Director
Associate Professor
Heinrich Audebert, MD Stroke and Cerebrovascular Health
Department of Radiology
Senior Physician Research
Washington University School of
Assistant Director of the Department CBF Brain Research Center
Medicine
Department of Neurology Division of Neurology
St. Louis, Missouri
Center for Stroke Research University of British Columbia
Craig S. Anderson, MD, PhD Charité University Medicine Berlin Vancouver, British Columbia, Canada
Professor of Neurology Berlin, Germany
Eric M. Bershad, MD
The George Institute for Global Health
Roland N. Auer, MD, PhD Associate Professor
University of New South Wales
Professor and Neuropathologist Neurology, Neurosurgery, and Space
Sydney, Australia
Department of Pathology Medicine
Josef Anrather, VMD Royal University Hospital Baylor College of Medicine
Professor of Neuroscience Saskatoon, Saskatchewan, Canada Houston, Texas
Feil Family Brain and Mind Research
Issam A. Awad, MD Jimmy V. Berthaud, MD, MPH
Institute
The John Harper Seeley Professor of Assistant Professor
Weill Cornell Medicine
Surgery (Neurosurgery) Department of Neurology
New York, New York
Director, Neurovascular Surgery University of Michigan Medical School
Department of Neurosurgery Ann Arbor, Michigan
University of Chicago
Chicago, Illinois

xii
Contributors xiii

Spiros L. Blackburn, MD Louis R. Caplan, MD Greg Christorforids, MD


Associate Professor Professor Professor
Department of Neurosurgery Department of Neurology Department of Radiology
University of Texas Houston Health Harvard University University of Chicago
Science Center Beth Israel Deaconess Medical Center Chicago, Illinois
Houston, Texas Boston, Massachusetts
E. Sander Connolly, Jr., MD
Leo H. Bonati, MD Julián Carrión-Penagos, MD Bennett M. Stein Professor
Professor of Neurology Neurology Resident Chairman
Head Stroke Center Department of Neurology Department of Neurological Surgery
Department of Neurology University of Chicago Columbia University Medical Center
University Hospital Basel Chicago, Illinois New York, New York
Department of Clinical Research
Mar Castellanos, MD Steven C. Cramer, MD
University of Basel
Department of Neurology Professor
Basel, Switzerland
Complexo Hospitalario Universitario A Department of Neurology
Julian Bösel, MD Coruña David Geffen School of Medicine
Professor Biomedical Research Institute of A University of California, Los Angeles
Department of Neurology Coruña Los Angeles, California
Klinikum Kassel A Coruña, Spain
Brett L. Cucchiara, MD
Kassel, Germany
Michelle R. Caunca, PhD Professor
Marie Germaine Bousser, MD Medical Scientist Training Program Department of Neurology
Department of Neurology and CERVCO University of Miami Miller School of University of Pennsylvania
Reference Center for Rare Vascular Medicine Philadelphia, Pennsylvania
Diseases of the Eye and Brain Miami, Florida
Alexandra L. Czap, MD
Hôpital Lariboisiére
Hugues Chabriat, MD, PhD Neuro-oncologist
Assistance Publique Hôpital de Paris;
Department of Neurology and CERVCO Assistant Professor
Université of Paris
Reference Center for Rare Vascular Department of Neurology
Paris, France
Diseases of the Eye and Brain McGovern Medical School
Joseph P. Broderick, MD Hôpital Lariboisiére University of Texas Health Science
Professor Assistance Publique Hôpital de Paris Center at Houston
Department of Neurology and Ambroise Paré Houston, Texas
Rehabilitation Medicine INSERM U 1161
Mark J. Dannenbaum, MD
University of Cincinnati Gardner Genetics and Physiopathology of
Assistant Professor
Neuroscience Institute ­Cerebrovascular Diseases
Department of Neurosurgery
Cincinnati, Ohio Université of Paris
University of Texas Houston Health
Service de Neurologie
Martin M. Brown, MA, MD Science Center
Hôpital Lariboisière
Emeritus Professor of Stroke Medicine Houston, Texas
Paris, France
UCL Queen Square Institute of
Patricia H. Davis, MD
Neurology Angel Chamorro, MD, PhD
Professor Emeritus
University College London Professor of Neurology
Department of Neurology
London, United Kingdom Department of Neurosciences
Carver College of Medicine
Hospital Clinic of Barcelona
Wendy Brown, MD University of Iowa
Barcelona, Spain
Stroke Director Iowa City, Iowa
Sutter Roseville Medical Center Jieli Chen, MD
Ted M. Dawson, MD, PhD
Roseville California Senior Scientist
Director, Institute for Cell Engineering
Department of Neurology
John C.M. Brust, MD Professor of Neurology
Henry Ford Hospital
Professor John Hopkins University School of
Detroit, Michigan
Department of Neurology Medicine
Columbia University College of Jun Chen, MD, PhD Baltimore, Maryland
Physicians and Surgeons Professor
Valina L. Dawson, PhD
New York, New York Department of Neurology
Director, Neuroregeneration and Stem
University of Pittsburgh
Cheryl Bushnell, MD, MHS Cell Programs
Pittsburgh, Pennsylvania
Department of Neurology Institute for Cell Engineering
Wake Forest School of Medicine Michael Chopp, PhD Professor of Neurology, Neuroscience,
Winston-Salem, North Carolina Senior Scientist and Physiology
Department of Neurology Johns Hopkins School of Medicine
Patrícia Canhão, MD, PhD
Henry Ford Hospital Baltimore, Maryland
Department of Neurosciences
Detroit, Michigan
(Neurology) Arthur L. Day, MD
Distinguished Professor
Hospital de Santa Maria-CHULN Professor and Co-Chairman
Department of Physics
Instituto de Medicina Molecular João Director of Cerebrovascular Surgery
Oakland University
Lobo Antunes Residency Program Director
Rochester, Michigan
Faculdade de Medicina Department of Neurosurgery
Universidade de Lisboa University of Texas Medical School at
Lisbon, Portugal Houston
Houston, Texas
xiv Contributors

T. Michael De Silva, PhD Bruce H. Dobkin, MD Myriam Fornage, PhD


Lecturer in Physiology Professor Professor, Center for Human Genetics
Department of Physiology, Anatomy, Department of Neurology Laurence and Johanna Favrot
and Microbiology David Geffen School of Medicine Distinguished Professor in Cardiology
School of Life Sciences University of California, Los Angeles Brown Foundation Institute of
La Trobe University Los Angeles, California Molecular Medicine
Melbourne, Victoria, Australia McGovern Medical School
Imanuel Dzialowski, MD
University of Texas Health Science
Diana Aguiar de Sousa, MD, PhD Head of ELBLAND Center for
Center at Houston
Department of Neurosciences Neuro-Rehabilitation
Houston, Texas
(Neurology) Teaching Faculty
Hospital de Santa Maria-CHULN Technical University Dresden Karen L. Furie, MD, MPH
Instituto de Medicina Molecular João Dresden, Germany Neurologist-in-Chief
Lobo Antunes Rhode Island Hospital
Mitchell S.V. Elkind, MD
Faculdade de Medicina The Miriam Hospital and Bradley
Professor
Universidade de Lisboa Hospital
Department of Neurology
Lisbon, Portugal Samuel I. Kennison, MD, and Bertha
Columbia University College of
S. Kennison Professor of Clinical
Victor J. Del Brutto, MD Physicians and Surgeons
Neuroscience
Assistant Professor Professor
Chair of Neurology
Department of Neurology Department of Epidemiology
The Warren Alpert Medical School of
Stroke Division Columbia University Mailman School
Brown University
University of Miami Miller School of of Public Health
Providence, Rhode Island
Medicine New York, New York
Miami, Florida Lidia Garcia-Bonilla, PhD
Jordan Elm, PhD
Assistant Professor of Research in
Gregory J. del Zoppo, MD Associate Professor
Neuroscience
Professor Department of Public Health Sciences
Feil Family Brain and Mind Research
Division of Hematology Medical University of South Carolina
Institute
Department of Neurology Charleston, South Carolina
Weill Cornell Medicine
University of Washington
Valery L. Feigin, MD, PhD New York, New York
Seattle, Washington
Professor of Epidemiology and
Steven L. Giannotta, MD
Colin P. Derdeyn, MD Neurology
Professor
Professor and Chair National Institute for Stroke and
Chair, Neurological Surgery
Radiology and Neurology Applied Neurosciences
Department of Neurosurgery
Department of Neurology Faculty of Health and Environmental
Keck School of Medicine
Carver College of Medicine Studies
University of Southern California
University of Iowa Auckland University of Technology
Los Angeles, California
Iowa City, Iowa Auckland, New Zealand
Y. Pierre Gobin, MD
Marco R. Di Tullio, MD José Manuel Ferro, MD, PhD
Professor of Radiology in Neurology
Professor of Medicine Department of Neurosciences
and Neurosurgery
Division of Cardiology (Neurology)
Director, Interventional Neurology
Columbia University College of Hospital de Santa Maria-CHULN
Weill Cornell Medical Center
Physicians and Surgeons Principle Investigator
New York Presbyterian Hospital
Associate Director Instituto de Medicina Molecular João
New York, New York
Adult Cardiovascular Ultrasound Lobo Antunes
Laboratories Faculdade de Medicina Mark P. Goldberg, MD
Cardiologist Professor Professor of Neurology
Cardiology Division Universidade de Lisboa Associate Vice Chair of Institutional
Columbia University Irving Medical Lisbon, Portugal Advancement
Center University of Texas Southwestern
Thalia S. Field, MD
New York, New York Medical Center
Associate Professor
Dallas, Texas
Hans Christoph Diener, MD, PhD Department of Medicine
Professor of Neurology Emeritus University of British Columbia Larry B. Goldstein, MD
Medical Faculty of the University Vancouver, British Columbia, Canada Ruth L Works Professor and Chairman
Duisburg-Essen Department of Neurology
Marlene Fischer, MD, PhD
Institute for Medical Informatics, University of Kentucky
University Medical Center Hamburg-
Biometry and Epidemiology Co-Director, Kentucky Neuroscience
Eppendorf
Essen, Germany Institute
Center for Anesthesiology and Intensive
Lexington, Kentucky
Michael N. Diringer, MD Care Medicine
Professor Department of Intensive Care Medicine Nicole R. Gonzales, MD
Department of Neurology Hamburg, Germany Professor
Washington University School of Department of Neurology
Medicine McGovern Medical School
St. Louis, Missouri University of Texas Health Science
Center at Houston
Houston, Texas
Contributors xv

David M. Greer, MD Glen C. Jickling, MD Charles E. Kircher, MD


Richard B. Slifka Chief of Neurology Associate Professor Assistant Professor
Boston Medical Center Department of Medicine Department of Emergency Medicine
Professor and Chair Division of Neurology University of Cincinnati
Department of Neurology University of Alberta Neurointensivist
Boston University School of Medicine Edmonton, Alberta, Canada Gardner Neuroscience Institute
Boston, Massachusetts University of Cincinnati Medical Center
Anne Joutel, MD, PhD
Cincinnati, Ohio
James C. Grotta, MD INSERM U 1266
Director of Stroke Research and Mobile Pathogenesis of Small Vessel Diseases of Timo Krings, MD, PhD
Stroke Unit the Brain; The David Braley and Nancy
Clinical Innovation and Research Université of Paris Gordon Chair in Interventional
Institute Paris, France Neuroradiology
Memorial Hermann Hospital-Texas Chief, Diagnostic and Interventional
Scott E. Kasner, MD
Medical Center Neuroradiology
Professor
Houston, Texas Toronto Western Hospital and
Department of Neurology
University Health Network
Ruiming Guo University of Pennsylvania
Program Director, Interventional
Department of Neurology Director, Comprehensive Stroke Center
Neuroradiology
Pittsburgh Institute of Brain Disorders University of Pennsylvania Health
Toronto Western Hospital
and Recovery System
Professor
University of Pittsburgh Philadelphia, Pennsylvania
Departments of Radiology and Surgery
Pittsburgh, Pennsylvania
Mira Katan, MD, MS University of Toronto
Jose Gutierrez, MD, MPH Assistant Professor Toronto, Ontario, Canada
Assistant Professor of Neurology Department of Neurology
Rita V. Krishnamurthi, BSc, MApplSc,
Department of Neurology University Hospital of Zurich
PhD
Columbia University College of Zurich, Switzerland
Associate Professor
Physicians and Surgeons
Christopher P. Kellner, MD National Institute for Stroke and
New York, New York
Assistant Professor Applied Neurosciences
Peter Harmel, MD Department of Neurosurgery Faculty of Health and Environmental
Department of Neurology Icahn School of Medicine at Mount Studies
Charité University Medicine Berlin Sinai Auckland University of Technology
Berlin, Germany New York, New York Auckland, New Zealand
George Howard, DrPH Muhib Khan, MD Tobias Kurth, MD, ScD
Professor Clinical Assistant Professor Professor of Public Health and
Department of Biostatistics Department of Clinical Neuroscience Epidemiology
School of Public Health Michigan State University College of Institute of Public Health
University of Alabama at Birmingham Human Medicine Charité-Universitätsmedizin Berlin
Birmingham, Alabama Division Chief, Inpatient Neurology Berlin, Germany
Director, Comprehensive Stroke Center
Virginia J. Howard, PhD Maarten G. Lansberg, MD, PhD
Spectrum Health
Professor Professor
Grand Rapids, Michigan
Department of Epidemiology Neurology and Neurological Sciences
School of Public Health Chelsea S. Kidwell, MD Stanford University
University of Alabama at Birmingham Professor Stanford, California
Birmingham, Alabama Vice Chair of Research
Elad I. Levy, MD, MBA
Department of Neurology
Jee-Yeon Hwang, PhD Professor and L. Nelson Hopkins, MD,
University of Arizona College of
Assistant Professor Chairman
Medicine
Department of Pharmacology and Department of Neurological Surgery
Tuscon, Arizona
Neuroscience Jacobs School of Medicine and
Creighton University Helen Kim, MPH, PhD Biomedical Sciences
Omaha, Nebraska Professor University at Buffalo
Department of Anesthesia and Director, Interventional Stroke Services
Costantino Iadecola, MD
Perioperative Care Endovascular Neurosurgery Fellowship
Director and Chair
Director, Center for Cerebrovascular Kaleida Health
Feil Family Brain and Mind Research
Research Buffalo, New York
Institute
University of California, San Francisco
Weill Cornell Medicine David S. Liebeskind, MD
San Francisco, California
New York, New York Professor of Neurology
Jong S. Kim, MD, PhD Director, Neurovascular Imaging
Reza Jahan, MD
Professor Research Core
Professor
Department of Neurology Director, Outpatient Stroke and
Division of Interventional
University of Ulsan Neurovascular Programs
Neuroradiology
Asan Medical Center Director, UCLA Cerebral Blood Flow
Department of Radiology
Seoul, South Korea Laboratory
David Geffen School of Medicine
Director, UCLA Vascular Neurology
University of California, Los Angeles
Residency Program
Los Angeles, California
UCLA Department of Neurology
Los Angeles, California
xvi Contributors

Sook-Lei Liew, PhD, OTR/L Jason M. Meckler, MD Maiken Nedergaard, MD, PhD
Assistant Professor Neurologist Professor and Director
Chan Division of Occupational Science Norton Neurology Services Center for Translational Neuromedicine
and Occupational Health Louisville, Kentucky University of Rochester Medical Center
Division of Biokinesiology and Physical Rochester, New York
James Frederick Meschia, MD
Therapy Professor and Director
Professor
Department of Neurology Center for Basic and Translational
Department of Neurology
Keck School of Medicine Neuroscience
Mayo Clinic
University of Southern California University of Copenhagen
Jacksonville, Florida
Los Angeles, California Copenhagen, Denmark
Steven R. Messé, MD
David J. Lin, MD Justin A. Neira, MD
Professor
Clinical Fellow Resident
Department of Neurology
Center for Neurotechnology and Department of Neurological Surgery
Perelman School of Medicine at the
Neurorecovery Columbia University Medical Center
University Hospital of Pennsylvania
Department of Neurology NY-Presbyterian Hospital
Philadelphia, Pennsylvania
Massachusetts General Hospital New York, New York
Boston, Massachusetts J Mocco, MD
Sarah Newman, NP
Professor
Benjamin Lisle, PM, PhD Beth Israel Lahey Health
Department of Neurosurgery
Department of Neurology Lahey Hospital and Medical Center
Icahn School of Medicine at Mount
University of Missouri Medical School Burlington, Massachusetts
Sinai
and Cox Health
New York, New York Patrick J. Nicholson, MB, BCh, BAO
Springfield, Missouri
Diagnostic and Interventional
Maxim Mokin, MD, PhD
Eng H. Lo, PhD Neuroradiologist
Associate Professor
Professor of Neurology and Radiology Toronto Western Hospital and
Department of Neurosurgery
Harvard Medical School University Health Network
University of South Florida College of
Boston, Massachusetts University of Toronto
Medicine
Director, Neuroprotection Research Toronto, Ontario, Canada
Vascular Neurologist
Laboratories
Neurosciences Center Bo Norrving, MD, PhD
Massachusetts General Hospital
Tampa General Hospital Professor
Charlestown, Massachusetts
Tampa, Florida Department of Clinical Sciences
Patrick D. Lyden, MD Neurology Division
Michael A. Mooney, MD
Professor Lund University
Instructor
Department of Neurology Lund, Sweden
Department of Neurosurgery
Cedars-Sinai Medical Center
Brigham and Women’s Hospital Martin O’Donnell, MB, PhD
Los Angeles, California
Harvard Medical School Department of Medicine
Takakuni Maki, MD Boston, Massachusetts NUI Galway and Saolta University
Neuroprotection Research Laboratory Healthcare Group
Lewis B. Morgenstern, MD
Departments of Radiology and Galway, Ireland
Professor
Neurology
Department of Neurology Dimitry Ofengeim, PhD
Massachusetts General Hospital
Director, Stroke Program Department of Cell Biology
Harvard Medical School
University of Michigan Medical School Harvard Medical School
Boston, Massachusetts
Ann Arbor, Michigan Boston, Massachusetts
Department of Neurology
Kyoto University Graduate School of Michael A. Moskowitz, MD Jun Ogata, MD, PhD
Medicine Professor of Neurology Internal Medicine
Kyoto, Japan Harvard Medical School Hirakata General Hospital for
Senior Neuroscientist Developmental Disorders
Georgios A. Maragkos, MD
Departments of Radiology and Hirakata-shi, Osaka, Japan
Post-Doctoral Research Fellow
Neurology
Department of Neurosurgery Christopher S. Ogilvy, MD
Massachusetts General Hospital
Beth Israel Deaconess Medical Center Professor
Boston, Massachusetts
Harvard Medical School Department of Neurosurgery
Boston, Massachusetts Michael T. Mullen, MD Harvard Medical School
Assistant Professor Director, Brain Aneurysm Institute
Miklos Marosfoi, MD
Department of Neurology Director, Endovascular and Operative
Assistant Professor of Radiology
Perelman School of Medicine at the Neurovascular Surgery
Tufts University School of Medicine
University Hospital of Pennsylvania Beth Israel Deaconess Medical Center
Beth Israel Lahey Health
Philadelphia, Pennsylvania Boston, Massachusetts
Lahey Hospital and Medical Center
Burlington, Massachusetts Steffen Nägel, MD Emanuele Orrù, MD
Departement of Neurology Assistant Professor of Radiology
Louise D. McCullough, MD, PhD
Martin-Luther-University Tufts University School of Medicine
Professor and Chair
Halle-Wittenberg Beth Israel Lahey Health
Department of Neurology
University Hospital Halle Lahey Hospital and Medical Center
McGovern Medical School
Halle, Germany Burlington, Massachusetts
University of Texas Health Science
Center at Houston
Houston, Texas
Contributors xvii

Santiago Ortega-Gutiérrez, MD Miguel A. Perez-Pinzon, PhD Christina P. Rossitto, BS


Associate Clinical Professor Professor and Vice-Chair for Basic Medical Student
Neurology, Neurosurgery, Radiology, Science of Neurology Icahn School of Medicine at Mount
and Anesthesia Peritz Scheinberg Endowed Professor in Sinai
Department of Neurology Neurology New York, New York
Carver College of Medicine Director, Peritz Scheinberg Cerebral
Tatjana Rundek, MD, PhD
University of Iowa Vascular Disease Research
Professor of Neurology
Iowa City, Iowa Laboratories
Department of Neurology
Department of Neurology
Matthew Maximillian Padrick, MD Evelyn F. McKnight Endowed Chair for
University of Miami Miller School of
Resident Physician Learning and Memory in Aging
Medicine
Department of Neurology Scientific Director, Evelyn F. McKnight
Miami, Florida
Cedars-Sinai Medical Center Brain Institute
Los Angeles, California John M. Picard, MD University of Miami Miller School of
Fellow Medicine
Kaushik Parsha, MD
Division of Neurocritical Care and Miami, Florida
Institute for Stroke and Cerebrovascular
Emergency Neurology
Diseases Jonathan J. Russin, MD
Yale New Haven Hospital
Department of Neurology Assistant Professor
New Haven, Connecticut
McGovern Medical School Director, Cerebrovascular Surgery
University of Texas Health Science Sean P. Polster, MD Department of Neurosurgery
Center at Houston Neurosurgery Resident Keck School of Medicine
Houston, Texas Department of Neurosurgery University of Southern California
University of Chicago Los Angeles, California
Mark Parsons
Chicago, Illinois
Professor of Medicine and Neurology Ralph L. Sacco, MD
Department of Neurology William J. Powers, MD Chairman, Department of Neurology
South Western Sydney Clinical School Professor of Neurology Olemberg Family Chair in Neurological
University of New South Wales Department of Neurology Disorders
Sydney, Australia University of North Carolina School of Miller Professor of Neurology, Public
Liverpool Hospital Medicine Health Sciences, Human Genetics,
Ingham Institute for Applied Medical Chapel Hill, North Carolina and Neurosurgery
Research University of Miami Miller School of
Volker Puetz, MD
Liverpool, Australia Medicine
Department of Neurology
Chief, Department of Neurology
Neil V. Patel, MD University Clinics Carl Gustav Carus
Jackson Memorial Hospital
Assistant Professor of Radiology Technische Universität Dresden
Miami, Florida
Tufts University School of Medicine Dresden, Germany
Beth Israel Lahey Health Apostolos Safouris, MD, PhD
Jukka Putaala, MD, PhD
Lahey Hospital and Medical Center Acute Stroke Unit
Associate Professor
Burlington, Massachusetts Metropolitan Hospital
Department of Neurology
Piraeus, Greece
Virendra I. Patel, MD, MPH Helsinki University Hospital and
Chief of Vascular Surgery University of Helsinki Edgar A. Samaniego, MD
Co-Director, Aortic Center Helsinki, Finland Associate Professor
Columbia University Irving Medical Neurology, Neurosurgery, and Radiology
Margarita Rabinovich, NP
Center Department of Neurology
Beth Israel Lahey Health
New York, New York Carver College of Medicine
Lahey Hospital and Medical Center
University of Iowa
Ludmila Pawlikowska, PhD Burlington, Massachusetts
Iowa City, Iowa
Associate Professor
Bruce R. Ransom, MD, PhD
Department of Anesthesia and Lauren H. Sansing, MD, MSTR
Professor and Chair
Perioperative Care Associate Professor
Department of Neuroscience
Center for Cerebrovascular Research Academic Chief
City University of Hong Kong
University of California, San Francisco Division of Stroke and Vascular
Hong Kong, China
San Francisco, California Neurology
Jorge A. Roa, MD Department of Neurology
Adriana Pérez, PhD
Post-doctoral Research Fellow Yale School of Medicine
Professor
Neurology and Neurosurgery New Haven, Connecticut
Department of Biostatistics and Data
University of Iowa Hospitals and Clinics
Science Nikunj Satani, MD, MPH
Iowa City, Iowa
The University of Texas Health Science Institute for Stroke and Cerebrovascular
Center at Houston Gary A. Rosenberg, MD Diseases
Austin, Texas Professor Department of Neurology
Department of Neurology McGovern Medical School
University of New Mexico Health University of Texas Health Science
Sciences Center Center at Houston
Director, Center for Memory and Aging Houston, Texas
University of New Mexico
Albuquerque, New Mexico
xviii Contributors

Ronald J. Sattenberg, MD Omar K. Siddiqi, MD Hiroo Takayama, MD, PhD


Radiologist Assistant Professor Director of Cardiovascular Institute
Louisville, Kentucky Section of Cardiovascular Medicine Co-Director, HCM Program, Division of
Department of Medicine Cardiac, Vascular & Thoracic Surgery
Jeffrey L. Saver, MD
Boston University School of Medicine Co-Director, Aortic Program
Professor of Neurology
Boston, Massachusetts Co-Director, Marfan Clinic
David Geffen School of Medicine
Columbia University Irving Medical Center
Director, UCLA Stroke Center Aneesh B. Singhal, MD
New York, New York
University of California, Los Angeles Vice Chair of Neurology
Los Angeles, California Massachusetts General Hospital Joseph Tarsia, MD
Associate Professor of Neurology Neurologist
Sean I. Savitz, MD
Harvard Medical School Ochsner Health
Professor of Neurology
Boston, Massachusetts New Orleans, Louisiana
Stroke Program Director
Institute for Stroke and Cerebrovascular Christopher G. Sobey, PhD Turgut Tatlisumak, MD, PhD
Diseases NHMRC Senior Research Fellow and Professor of Neurology and Stroke
Department of Neurology Professor in Physiology Medicine
McGovern Medical School Department of Physiology, Anatomy, Department of Clinical Neuroscience
University of Texas Health Science and Microbiology Institute of Neuroscience and
Center at Houston School of Life Sciences Physiology
Houston, Texas La Trobe University Sahlgrenska Academy at University of
Melbourne, Victoria, Australia Gothenburg
Christian Schmidt, MD
Chief Physician
Neurologist Clemens J. Sommer, MD
Department of Neurology
Department of Neurology Director, Institute of Neuropathology
Sahlgrenska University Hospital
Klinikum Kassel University Medical Center of the
Gothenburg, Sweden
Kassel, Germany Johannes Gutenberg-University Mainz
Department of Neurology Mainz, Germany Ajith J. Thomas, MD
Faculty of Human Medicine Assistant Professor
Robert F. Spetzler, MD
Georg August University Department of Neurosurgery
Emeritus President and Chief Executive
Göttingen, Germany Harvard Medical School
Officer
Co-Director
Sudha Seshadri, MD Emeritus Chair, Department of
Brain Aneurysm Institute
Director, Glenn Biggs Institute for Neurosurgery
Beth Israel Deaconess Medical Center
Alzheimer’s and Neurodegenerative Barrow Neurological Institute
Boston, Massachusetts
Diseases Phoenix, Arizona
University of Texas Health Sciences Center John W. Thompson, PhD
Christopher J. Stapleton, MD
San Antonio, Texas Director of Basic Science Research
Instructor
Senior Investigator Department of Neurosurgery
Department of Neurosurgery
Framingham Heart Study University of Miami Miller School of
Massachusetts General Hospital
Framingham, Massachusetts Medicine.
Boston, Massachusetts
Adjunct Professor Miami, Florida
Department of Neurology Ben A. Strickland, MD
Georgios Tsivgoulis, MD, PhD, RVT
Boston University School of Medicine Department of Neurosurgery
Professor of Neurology
Boston, Massachusetts Keck School of Medicine
Second Department of Neurology
University of Southern California
Vijay K. Sharma, MD National and Kapodistrian University of
Los Angeles, California
Associate Professor Athens
Yong Loo Lin School of Medicine Hua Su, MD School of Medicine
National University of Singapore Professor Attikon University Hospital
Senior Consultant Department of Anesthesia and Athens, Greece
Department of Neurology Perioperative Care
Elizabeth Tournier-Lasserve, MD, PhD
National University Hospital Center for Cerebrovascular Research
Molecular Genetics Department and
Singapore University of California, San Francisco
CERVCO Reference Center for Rare
San Francisco, California
Frank R. Sharp, MD Vascular Diseases of the Eye and Brain
Professor José I. Suarez, MD Hopital Lariboisiére
Department of Neurology Director, Division of Neurosciences Assistance Publique Hôpital de Paris;
School of Medicine Critical Care Université of Paris
University of California, Davis Professor Paris, France
Sacramento, California Departments of Anesthesia and Critical
Gabriel Vidal, MD
Care Medicine, Neurology, and
Kevin N. Sheth, MD Neurologist
Neurosurgery
Professor of Neurology and System Stroke Medical Director
The Johns Hopkins University School of
Neurosurgery Ochsner Health
Medicine
Chief, Division of Neurocritical Care New Orleans, Louisiana
Baltimore, Maryland
and Emergency Neurology
Ajay K. Wakhloo, MD PhD FAHA
Associate Chair, Clinical Research
Professor of Radiology
Department of Neurology
Tufts University School of Medicine
Yale School of Medicine and Yale New
Beth Israel Lahey Health
Haven Hospital
Lahey Hospital and Medical Center
New Haven, Connecticut
Burlington, Massachusetts
Contributors xix

Babette B. Weksler, MD Shadi Yaghi, MD John H. Zhang, MD, PhD


Professor Emerita Associate Professor Professor of Neurosurgery,
Department of Medicine Department of Neurology Anesthesiology, Neurology, and
Weill Cornell Medicine NYU Grossman School of Medicine Physiology and Pharmacology
New York, New York Director, Vascular Neurology Director of Neuroscience Research
NYU Langone Hospital-Brooklyn Associate Chair and Physiology
Joshua Z. Willey, MD
Director, Clinical Vascular Neurology Graduate Program Coordinator
Associate Professor
Research Loma Linda University School of
Department of Neurology
NYU Langone Health Medicine
Columbia University Irving Medical
New York, New York Loma Linda, California
Center
New York, New York Takenori Yamaguchi, MD, PhD Zhitong Zheng, MD
President Emeritus Visiting Fellow
Max Wintermark, MD
National Cerebral and Cardiovascular Department of Neurology
Professor
Center Henry Ford Hospital
Department of Radiology/Neuroimaging
Suita, Osaka, Japan Detroit, Michigan
and Neurointervention
Stanford University Tuo Yang, MD R. Suzanne Zukin, PhD
Stanford, California Research Instructor Professor
Department of Neurology Department of Neuroscience
Lawrence K.S. Wong, MD
Pittsburgh Institute of Brain Disorders F.M. Kirby Chair in Neural Repair and
Professor
and Recovery Protection
Department of Medicine and
University of Pittsburgh Director, Neuropsychopharmacology
Therapeutics
Pittsburgh, Pennsylvania Center
Chinese University of Hong Kong
Albert Einstein College of Medicine
Shatin, Hong Kong, China Masahiro Yasaka, MD, PhD
Bronx, New York
Director, Cerebrovaascular Center
Guohua Xi, MD
National Hospital Organization Kyushu Richard M. Zweifler, MD
Professor of Neurosurgery
Medical Center Associate Medical Director
Associate Director, Crosby Neurosurgical
Fukuoka, Japan Medical Services
Laboratories
Co-Medical Director of Neurosciences
Richard C. Schneider Research Professor Darin B. Zahuranec, MD
System Chair of Neurology
University of Michigan Associate Professor
Ochsner Health
Ann Arbor, Michigan Department of Neurology
New Orleans, Louisiana
University of Michigan Medical School
Jinchong Xu, PhD
Ann Arbor, Michigan
Assistant Professor
Neuroregeneration and Stem Cell Feng Zhang, MD, PhD
Programs Assistant Professor
Institute for Cell Engineering Department of Neurology
Johns Hopkins University School of Pittsburgh Institute of Brain Disorders
Medicine and Recovery
Baltimore, Maryland University of Pittsburgh
Pittsburgh, Pennsylvania
AHA Evidence-Based Classifications
TABLE 1 Applying Classification of Recommendations and Level of Evidence

SIZE OF TREATMENT EFFECT


CLASS III No Benefit
CLASS I CLASS IIa CLASS IIb or CLASS III Harm
Benefit > > > Risk Benefit > > Risk Benefit ≥ Risk Procedure/ Treatment
Additional studies with Additional studies with Test
Procedure/
focused objectives broad objectives
Treatment COR Not No Proves
needed needed; additional
SHOULD be III: No Helpful Benefit
registry data would be
performed/ IT IS REASONABLE to benefit
helpful
administered perform procedure/
administer treatment Procedure/Treatment COR Excess Harmful to
MAY BE CONSIDERED III: Cost w/o Patients
Harm Benefit or
Harmful
LEVEL A • Recommendation • Recommendation in • Recommendation’s • Recommendation that procedure or
ESTIMATE OF CERTAINTY (PRECISION) OF TREATMENT EFFECT

Multiple that procedure or favor of treatment usefulness/efficacy treatment is not useful/effective


populations treatment is or procedure being less well established and may be harmful
evaluated* useful/effective useful/effective • Greater conflicting • Sufficient evidence from multiple
Data derived from • Sufficient • Some conflicting evidence from randomized trials or meta-analyses
multiple evidence from evidence from multiple randomized
randomized multiple multiple randomized trials or meta-
clinical trials or randomized trails trials or meta- analyses
meta-analyses or meta-analyses analyses
LEVEL B • Recommendation • Recommendation in • Recommendation’s • Recommendation that procedure or
Limited that procedure or favor of treatment usefulness/efficacy treatment is not useful/effective
populations treatment is or procedure being less well established and may be harmful
evaluated* useful/effective useful/effective • Greater conflicting • Evidence from single randomized
Data derived from • Evidence from • Some conflicting evidence from single trial or nonrandomized studies
a single single evidence from randomized trial or
randomized trial randomized trial single randomized nonrandomized
or nonrandomized or nonrandomized trial or studies
studies studies nonrandomized
studies
LEVEL C • Recommendation • Recommendation in • Recommendation’s • Recommendation that procedure or
Very limited that procedure or favor of treatment usefulness/efficacy treatment is not useful/effective
populations treatment is or procedure being less well established and may be harmful
evaluated* useful/effective useful/effective • Only diverging expert • Only expert opinion, case studies,
Only consensus • Only expert • Only diverging opinion, case or standard of care
opinion of experts, opinion, case expert opinion, case studies, or standard
case studies, or studies, or studies, or standard of care
standard of care standard of care of care
Suggested phrases should is reasonable may/might be considered COR III COR III
for writing is recommended can be useful/effective/ may/might be reasonable No Benefit Harm
recommendations† is indicated beneficial usefulness/effectiveness is
is not potentially
is useful/effective/ is probably unknown/unclear/
recommended harmful
beneficial recommended or uncertain or not well
is not indicated causes harm
indicated established
should not be associated with
Comparative treatment/strategy A treatment/strategy A is done excess morbidity/
effectiveness is recommended/ probably recommended/ is not useful/ mortality
phrases† indicated in indicated in preference beneficial/effective should not be done
preference to to treatment B
treatment B treatment it is reasonable to
A should be chosen choose treatment A over
over treatment B treatment B

Reprinted with permission Circulation. 2010;121:1544–1579 ©2010, American Heart Association, Inc.

xx
AHA Evidence-Based Classifications xxi

BOX 1   Evidence Classifications 


1. Size of treatment effect 2. Certainty of treatment effect
• Class I: Benefit >>> Risk. Procedure/treatment SHOULD be • Level A: Data derived from multiple randomized clinical trials or
performed/administered. meta-analyses.
• Class IIa: Benefit >> Risk. IT IS REASONABLE to perform • Level B: Data derived from a single randomized trial or
procedure/administer treatment. nonrandomized studies.
• Class IIb: Benefit ≥ Risk. Procedure/treatment MAY BE • Level C: Only consensus opinion of experts, case studies, or
CONSIDERED. standard of care.
• Class III: No Benefit/Harm. Procedure/treatment is not useful/
effective and may be harmful.

Adapted from Sacco RL, Adams R, Albers G, et al. Guidelines for prevention of stroke in patients with ischemic stroke or transient ischemic attack: a statement
for healthcare professionals from the American Heart Association/American Stroke Association Council on Stroke. Stroke. 2006;37:577–617.
SECTION
I Pathophysiology

Eng H. Lo

The first section in this new edition of Stroke provides an immune cells. New sections describe emerging opportunities
updated and comprehensive survey of the molecular, cellular, in tolerance and preconditioning, as well as interactions
and pathophysiologic mechanisms that underlie the brain’s between the immune system and the microbiome. The
reaction to ischemia and hemorrhage. At the cellular level, chapter on stroke recovery reviews a complex spectrum of
stroke affects pathways of hemostasis and perturbs interac­ compensatory response in resident precursor and circulating
tions between circulating blood elements, the blood vessel progenitor cells. New insights have been added to explore
itself, and brain parenchyma. At the functional level, the regu­ the role of exosomes and micro-RNA that may transfer and
lation and dysregulation of hemodynamics and metabolism coordinate signals between all cell types in the remodeling
mediates an integrated neurologic response. At the organ level, neurovascular unit. The chapter on white matter has also
stroke induces histopathologic reactions in all neural, glial, been expanded, with added material that links exercise to
and vascular cells. Hence this section begins with three chap­ oligodendrocyte homeostasis and resilience. The chapter
ters that define basic principles of vascular biology, cerebral on cerebral hemorrhage surveys advances in molecular and
blood flow and metabolism, and brain tissue injury. Updates cellular phenomena with new ideas that may link ferroptosis
include new information on hemodynamic responses to to translational opportunities and clinical trials. The chapter
thrombectomy and reperfusion, as well as new sections that on vascular malformations has been updated to link signaling
discuss correlations between experimental animal models and cascades in advanced zebrafish and mouse models with genes
clinical pathology. that are implicated in clinical disease. Finally, this section ends
Building on these fundamental principles, the next few with the addition of a new chapter that defines novel pathways
chapters then explore the molecular mechanisms of cell death in the neurovascular unit that mediate vascular contributions
and survival. Genes and pathways underlying necrosis and to cognitive impairment and dementia.
programmed cell death are balanced against an expanding Optimal translation for cerebrovascular disease cannot
family of endogenous neuroprotection mediators. The neuro­ occur without a rigorous dissection of the molecular and
vascular unit chapter remains a centerpiece for the overall cellular fundamentals in neurovascular and gliovascular
concept of cell-cell signaling. However, beyond the brain biology. The basic principles established in this section should
itself, interactions with other organ systems are also discussed provide not only mechanistic foundations but also a rational
in terms of crosstalk with neuroinflammatory cascades and basis for pursuing therapeutics and diagnostics in stroke.

1
SECTION
I Pathophysiology

1 Cerebral Vascular Biology in Health and Disease


T. Michael De Silva, Christopher G. Sobey

KEY POINTS collateral flow is thought to be important when blood flow in


one region is compromised.2 The pial arterioles then dive into
• C erebral artery tone is substantially modulated under the brain to give rise to parenchymal arterioles. Parenchymal
physiologic conditions by endothelium-derived arterioles are long, relatively unbranched arterioles that per-
nitric oxide, by reactive oxygen species, and through fuse a distinct area of brain tissue.3 The capillary network arises
hyperpolarization mediated by several types of K+ from the parenchymal arterioles, which is where the majority
channels. of nutrient and gas exchange occurs. Although much less is
known about their function during health or disease, cerebral
• Cerebral vascular function is very sensitive to venules and veins are also important components of the cere-
endothelial dysfunction that occurs during chronic bral circulation. For example, major disruption to blood-brain
disease, resulting in impairment of vasodilator barrier function during acute hypertension occurs in the pial
mechanisms. venules.4
• Oxidative stress and inflammation occur in the
cerebral circulation in response to cardiovascular risk PHYSIOLOGIC REGULATION OF CEREBRAL
factors present during atherosclerosis and chronic
hypertension, such as elevated plasma levels of
VASCULAR TONE
cholesterol and angiotensin II, respectively. Numerous mechanisms regulate cerebral artery function. Most
of the recent experimental evidence regarding such mecha-
nisms has come from pharmacologic studies and the use of
genetically modified mice. Major mechanisms include the
release of nitric oxide (NO) from the endothelium to underly-
INTRODUCTION ing smooth muscle cells (discussed in the Nitric Oxide and
The brain has a limited supply of nutrients; thus normal brain Cyclic Guanosine Monophosphate section); potassium ion
function relies on adequate perfusion by the cerebral circula- (K+) channels (see K+ Channels), which includes a discussion
tion for the delivery of oxygen and nutrients, as well as the of the newly described two-pore domain (K2P) channels, Rho/
removal of waste products. It is for this reason that cerebral Rho-kinase activity (see RhoA/Rho-Kinase); reactive oxygen
vascular tone is tightly regulated, and why any alterations in species (ROS), which are discussed in the Reactive Oxygen
mechanisms that modulate cerebral vessel function can pre- Species section; and the recently described transient receptor
dispose to cerebrovascular disease and stroke. Atherosclerosis potential (TRP) channels (discussed in the Transient Receptor
is the underlying pathologic process for both coronary and Potential Channels section).
cerebral artery disease, which are the two most common forms
of cardiovascular disease.1
The purpose of this chapter is thus to provide insight into
Nitric Oxide and Cyclic Guanosine Monophosphate
major mechanisms that regulate cerebral artery function, A major mechanism for maintenance of vascular tone by
and alterations in these mechanisms in two major clinical the endothelium involves the production of endothelium-
conditions that have a significant negative impact on health derived NO. In endothelium, NO is synthesized from endo-
worldwide—hypertension and atherosclerosis. The scope thelial nitric oxide synthase (eNOS); it then diffuses to the
is mostly limited to discussion of cerebral blood vessels underlying smooth muscle, where it activates soluble gua-
and mechanisms that regulate their tone, either under nylate cyclase, which in turn leads to increased intracellu-
basal conditions or in response to physiologically relevant lar cyclic guanosine monophosphate levels and subsequent
agonists. relaxation of the smooth muscle.5 Experimental evidence
for modulation of cerebral vascular tone by endothelium-
derived NO has been obtained by applying inhibitors of
ORGANIZATION OF THE CEREBRAL CIRCULATION NOS to cerebral blood vessels from several different species,
The brain is predominantly perfused by three pairs of intracra- both in vivo and in vitro, and has involved such inhibitors
nial arteries: the anterior, middle, and posterior cerebral arter- causing vasoconstriction (reviewed extensively in Faraci and
ies (ACA, MCA, and PCA, respectively). These arise from the Heistad6).
circle of Willis, a ring of arteries formed by the anterior and pos- NO release from the endothelium can also be stimulated
terior communicating arteries that connect the terminal ends in response to receptor- (e.g., acetylcholine, bradykinin)
of the basilar and internal carotid arteries. The ACA, MCA, and or non-receptor-mediated agonists, or in response to shear
PCA travel along the pial surface of the brain, branching into stress. Endothelium-dependent, NO-mediated cerebral
smaller arterioles. Importantly, anastomoses exist between vascular relaxation in response to such agonists is often used
the smaller arterioles of these three major arterial trees, and to determine the functional integrity of the endothelium.

3
4 SECTION I Pathophysiology

Endothelial dysfunction, manifested as diminished NO Recent evidence of the importance of Ca2+ spark activity
bioavailability experimentally by impaired endothelium- and BKCa channels as mediators of vasodilators has emerged,
dependent vasodilation, or reduced vasoconstriction in as TEA and iberiotoxin inhibit vasodilator responses in
response to a NOS inhibitor, is a common feature of many response to vasodilators that activate adenylate cyclase and
cerebrovascular-related diseases (discussed in the Alterations guanylate cyclase.12 Acidosis markedly increased Ca2+ spark
in Cerebral Vascular Function During Hypertension and activity and caused dilatation of brain parenchymal arterioles.
Atherosclerosis section). Such exogenously applied agonists Dilatation was inhibited by inhibitors of ryanodine receptors
are often useful in this way experimentally, and they may (ryanodine) and BKCa channels (paxilline), as well as in mice
also be important endogenously. For example, neurovascular lacking the BKCa channel.13 Hydrogen sulfide (an important
coupling in some brain regions is mediated by neuronally signaling molecule in the regulation of vascular tone and
released acetylcholine acting on the endothelium to blood pressure) also increased Ca2+ spark and BKCa current
stimulate eNOS.7 frequency, as well as causing dilatation in cerebral arterioles—
the vasodilatation was inhibited by ryanodine and iberiotoxin,
suggesting Ca2+ spark activity is important in the response.14
K+ Channels Intermittent hypoxia increased myogenic tone through loss of
The activity of K+ channels is a major regulator of smooth hydrogen sulfide activation of KCa channels.15 Hypoxia had
muscle cell membrane potential and, as such, is an important no effect on Ca2+ spark frequency but reduced KCa channel
regulator of vascular tone. This is because vessel diameter is in activity.16 Protein expression of KCa2.2, 2.3, and 3.1,16 as well
large part dependent on cytosolic Ca2+ concentration, which as α- and β1-subunits of BKCa channels17 in cerebral arteries,
in turn is dependent on membrane potential. There are five have been reported.
major types of K+ channels known to be expressed in cerebral
blood vessels: calcium (Ca2+)-activated (KCa) K+ channels, ATP
sensitive K+ (KATP) channels, voltage-sensitive K+ (KV) chan-
KATP Channels
nels, inwardly rectifying K+ (KIR) channels, and tandem-pore KATP channels are defined by their sensitivity to intracellular
(TREK-1) channels, and all are regulators of vascular tone. ATP, with their activity being inhibited by intracellular ATP.18
This is supported by the wealth of information using both Generally, the intracellular concentration of ATP is normally
pharmacologic inhibitors and gene-targeted mice to study sufficient that these channels have a low open probability
the regulation of membrane potential and vascular function. in most vascular smooth muscle cells under normal condi-
Potassium channels are also important mediators of vasodi- tions,19 and this appears to also be the case in the cerebral
lator responses to several vasodilators that regulate vascular circulation, where glibenclamide, a selective inhibitor of KATP
tone, and this will be also be discussed. channels, has no effect on cerebral vascular tone.20 However,
KATP channels appear to be present and functional in cerebral
vessels based on direct evidence for their expression (discussed
KCa-Activated K+ Channels as follows) and a wealth of evidence reporting glibenclamide-
There are three subtypes of KCa channels present in the vascu- sensitive relaxation of cerebral arteries in response to KATP
lature: large-conductance KCa (BKCa) channels, intermediate- channel activators.18
conductance (IKCa) channels, and small-conductance (SKCa) Several more recent studies have investigated the expression
channels. Most research regarding the functional importance of KATP in cerebral vessels. KATP channels are thought to be a
of this channel, especially in cerebral arteries, has centered hetero-multimeric complex of two subunits: one is a pore-
around the BKCa channel. forming inward-rectifying K+ channel type 6 (i.e., 6.1 or 6.2),
As the name suggests, these channels are activated in and the other is a sulfonylurea receptor (SUR), either SUR1 and
response to increases in intracellular Ca2+. Membrane SUR2, with the SUR2 gene generating the two splice variants
depolarization, myogenic responses (i.e., pressure-induced SUR2A and SUR2B.21 Messenger RNA (mRNA) expression for
vasoconstriction, important in development and maintenance both the pore-forming subunits (KIR6.1 and 6.2) and SUR1,
of basal vascular tone), and elevations in arterial pressure are 2A, and 2B has been demonstrated in cerebral arteries,21,22
associated with elevations in intracellular Ca2+ concentration although another study investigating SUR expression found
in cells of the vasculature.8 Thus an important function of no expression of SUR1 and reported only SUR2B expression.23
these channels appears to be to act as a negative feedback Protein expression of KIR6.1 and 6.2, as well as SUR1 and 2B,
mechanism during increases in Ca2+ to limit vasoconstriction. was also reported.22 Cerebral arterioles were found to express
A major mechanism of elevations in intracellular Ca2+ appears KIR6.1 and SUR2B,24 with human cerebral arteries found to
to be via Ca2+ sparks, which are localized elevations in express SUR2B.23
cytosolic Ca2+, due to the opening of ryanodine-sensitive Ca2+ Acidosis and reductions in intracellular pO2 are known
release channels in the sarcoplasmic reticulum to KCa channels to produce cerebral vasodilatation. KATP channels have been
located on the plasma membrane. shown to be involved in cerebral vasodilatation in response
These channels are important in modulating the basal to acidosis,25,26 as well as in vasodilatation to NMDA, which
tone of cerebral arteries, as selective inhibition of BKCa may be important in the coupling of cerebral metabolism
channels with tetraethylammonium ion (TEA) produces and blood flow.27 More direct evidence for a role of KATP
vasoconstriction.8–10 In mice deficient in the β1 subunit of channels in mediating vasodilatation in response to oxygen/
BKCa channels, increased intracellular Ca2+ concentration glucose deprivation was reported in that vasodilatation was
in response to ryanodine (which at low concentrations impaired in SUR-deficient compared with wild-type mice.23
depletes Ca2+ stores from the sarcoplasmic reticulum so Myogenic tone, and vasodilatation in response to hypoxia, are
that intracellular Ca2+ concentration increases) and cerebral not dependent on SUR2 expression,23 although relaxation to
vascular constriction to iberiotoxin (selective inhibitor of BKCa hypoxia is inhibited by glibenclamide,18,28 suggesting a role
channels) was reduced, suggesting that Ca2+ spark activity for KATP channels in hypoxia-induced vasodilatation where the
modulates myogenic tone through BKCa channel activation.11 KATP subunit composition does not involve SUR2. Hydrogen
These channels may be more important in the modulation of sulfide also dilates cerebral arteries, an effect that is inhibited
basal tone in larger cerebral arteries.8 by glibenclamide and in SUR2-deficient mice.24
Cerebral Vascular Biology in Health and Disease 5

KV Channels RhoA/Rho-Kinase 1
KV channels are activated in response to increases in pressure Smooth muscle cell contractility is ultimately governed by the
in cerebral arteries and modulate cerebral vascular tone, in that phosphorylation state of myosin light chain (MLC), vascular
pharmacologic inhibition of KV channels with 4-aminopyri- smooth muscle tone occurring in association with increasing
dine causes cerebral artery depolarization and constriction.29,30 levels of MLC phosphorylation. MLC is phosphorylated by
KV channels are also known to mediate cerebral artery dila- MLC-kinase—a Ca2+-calmodulin-dependent enzyme—and is
tions, including in response to NO.29,31 KV channel subunits dephosphorylated by MLC phosphatase (MLCP). MLC phos-
are expressed in cerebral vessels (e.g., KV1.2 and 1.5,32–34 and phorylation and smooth muscle contractility are not always
KV2.1 and 2.235,36)—including in humans.37 KV2-mediated directly proportional to intracellular Ca2+ concentration.
current is proposed to underlie KV-dependent modulation of Other mechanisms can regulate smooth muscle contractility
cerebral artery tone in that inhibition of the KV2 channel with independent of changes in intracellular Ca2+ concentration, a
stromatoxin-caused cerebral artery constriction.36 phenomenon known as Ca2+-sensitization. Ca2+-sensitization
can occur through several pathways and ultimately results
in inhibition of MLCP. One such pathway is the RhoA/Rho-
KIR Channels kinase (ROCK) pathway. When ROCK is activated, it phos-
This channel is so named since it conducts K+ current more phorylates the myosin-binding (i.e., regulatory) subunit of
readily into than out of the cell over a wide range of mem- MLCP, and thus inhibits MLCP activity, which ultimately leads
brane potentials. However, at membrane potentials within to smooth muscle (and thus vascular) contractility.67,68
the physiologic range, these channels actually conduct a small In vascular muscle, RhoA can be activated by stretch.
outward current. Consequently, when this channel is inhibited This is important since myogenic tone is characterized by
with the pharmacologic blocker, barium ion (Ba2+), depolar- pressure-induced vasoconstriction, making it important for
ization and constriction of cerebral arteries are observed.38–44 the development of basal vascular tone. The contribution of
Furthermore, in mice lacking the KIR2.1 subunit—the subunit ROCK activity to the cerebral artery myogenic response has
thought to be important in mediating vascular KIR current— been studied through the use of Y-27632 and fasudil (HA-
cerebral artery KIR channel currents are absent.45 1077), pharmacologic inhibitors of Rho-kinase.69 For example,
In the cerebral circulation, K+ is released during neuronal Y-27632 relaxes cerebral artery segments following pressure-
activity and may be siphoned to cerebral vessels directly by induced constriction,70 and pressure-induced cerebral artery
astrocytes after neuronal activation.46 Basal concentration of K+ constriction is inhibited by Y-27632 and fasudil.71–73 In vivo,
in cerebrospinal fluid is ∼3 mM and may increase to between where myogenic tone is present, several studies have reported
4 and 7 mM during neuronal activity. In this concentration that Y-27632 and fasudil cause the dilatation of cerebral
range (i.e., from 3 to 10 mM), K+ causes dilatation of cerebral arteries74–78 and arterioles.79 Recent work has begun to define
arteries38,40–42,47,48 and arterioles.39,43,44,49–56 Moreover, the role of ROCK isoforms in the cerebral vasculature. The use
K+-induced hyperpolarization and vasodilatation in this of the selective ROCK2 inhibitor SLX-2119 (also known as
concentration range are inhibited by Ba2+,38–42,48,53–55,57–59 KD025) has revealed that myogenic tone in brain parenchymal
suggesting KIR-mediated K+-induced vasodilation may be an arterioles is ROCK2-dependent.80 In addition, SLX-2119 dilates
important mechanism in the coupling of cerebral metabolism pial arterioles in vivo.80
and blood flow (neurovascular coupling). Furthermore, cerebral ROCK is also important in the regulation of endothelial
vascular relaxation responses to K+ are absent in mice lacking cell function via effects on NO signaling. ROCK has been
the KIR2.1 subunit.45 There have been reports of KIR2.1 channel shown to reduce NO bioavailability, which occurs via
expression in cerebral arteries.38,58 Regarding the role for KIR2.1 reducing NO production via reducing phosphorylation of the
channels in neurovascular coupling, recent work identified stimulatory Ser,11, 77 direct phosphorylation of the inhibitory
KIR2.1 channel on capillaries as critical for sensing neuronal Thr495 residue on endothelial NOS, and/or reducing eNOS
activity (via K+ release) and initiating a retrograde signal to mRNA stability. These findings, in combination with the role
dilate upstream arterioles, thereby increasing local blood flow.60 of ROCK in vascular muscle, provide good evidence that the
RhoA/Rho-kinase pathway is a major mechanism contributing
to cerebral vascular tone.
K2P Channels
A new family of channels—two pore domain K+ (K2P) chan-
nels—have recently been characterized.61 These channels
Reactive Oxygen Species
require two protein subunits, each contributing two pore ROS are known to influence cerebral vascular tone, and this
domains, to form a functional channel. There are several is reviewed extensively elsewhere.81 These ROS include the
members of the K2P family expressed in the vasculature, with parent molecule superoxide (O2−), as well as hydroxyl radical
some reported to be functionally important in the cerebral (OH) and hydrogen peroxide (H2O2). The closely related reac-
vasculature. Expression of TREK-1, TREK-2, TASK-1, TWIK-2, tive nitrogen species (RNS)—peroxynitrite—is also commonly
TRAAK, and THIK-1 has been reported in cerebral arteries, involved in such effects.
with TREK-1 being the most abundant.62,63 Protein and mRNA Superoxide, a negatively charged anion, can elicit either
expression of TREK-1 in the basilar artery was associated with dilatation82–85 or constriction82,86 of cerebral arteries.
vasodilatation induced by polyunsaturated fatty acids (which Superoxide reacts extremely efficiently with NO. As has
are important, as they improve brain resistance against cere- been discussed, NO is a major regulator of cerebral vascular
bral ischemia), such as α-linolenic acid in wild-type mice; tone; thus reduced NO bioavailability following increased
vasodilatation in response to linolenic acid was absent in mice superoxide levels will likely result in vasoconstriction,
deficient in TREK-1.64 Nevertheless, another study reported with vasoconstriction being reported in response to higher
similar vasodilator responses of the basilar artery to α-linolenic concentrations of superoxide82,83 and vasorelaxation at low
acid in wild-type and TREK-1-deficient mice.65 Cerebral artery concentrations.82
expression of TRAAK was associated with an important role in H2O2 is a chemically more stable species than superoxide,
mediating endothelium-independent vasodilatation.66 and it diffuses much more readily across cell membranes, thus
6 SECTION I Pathophysiology

potentially being important as a signaling molecule. Many stress110 and uridine triphosphate.111 TRPV1, TRPV5, and
studies have reported that H2O2 acts as a cerebral vasodilator, TRPV6 channels do not appear to be expressed in cerebral
both in vivo and in vitro,85,87–94 although vasoconstriction has arteries.112 The melastatin TRP channel 4 (TRPM 4) is
also been reported.95 activated by high levels of intracellular Ca2+ and is known
Peroxynitrite, formed from the rapid chemical reaction of to be expressed in cerebral arteries.113 Expression in smooth
superoxide with NO, can also affect cerebral vascular tone, muscle cells is consistent with a role in the myogenic response,
with both dilatation96,97 and constriction97–99 of cerebral in that myogenic vasoconstriction was attenuated in cerebral
arteries reported. Lower concentrations of peroxynitrite appear arteries administered TRPM4 antisense.114 Pharmacologic
to cause cerebral vasoconstriction, with higher concentrations inhibition of the TRPM4 channel with 9-phenanthrol was
typically causing vasodilatation.97,100 able to cause hyperpolarization and prevent the development
and maintenance of myogenic tone, further underlining
its importance in the maintenance of myogenic tone in the
Transient Receptor Potential Channels cerebral circulation.115 Another study also reported cerebral
TRP channels are a superfamily of cation channels compris- vascular expression of TRPM4 protein, which, once inactivated,
ing at least 28 members and are assigned to 6 subfamilies results in reduced myogenic vasoconstriction in response to a
based on their sequence homology.101 These are TRPC (clas- PKC activator.116 TRPA1 channels are known to be expressed
sical), TRPV (vanilloid), TRPM (melastatin), TRPA (ankyrin), in cerebral vessels, specifically in endothelium, and mediate
TRPP (polycystin), and TRPML (mucolipin).102 The structure, endothelium-dependent vasodilatation.117 Finally, TRPP2
expression profile, and function of TRP channels have been channels have been shown to contribute to myogenic tone
reviewed in detail.103 generation in cerebral arteries.118 The role, if any, of other TRP
Depending on the specific TRP channel in question, channels in the cerebral vasculature is presently unknown.
activation can result in constriction or dilation of cerebral
arteries. TRPC1 channels have been shown to mediate
constriction of cerebral arteries via facilitating receptor- ALTERATIONS IN CEREBRAL VASCULAR FUNCTION
operated calcium entry in response to endothelin-1.104 TRPC3 DURING HYPERTENSION AND ATHEROSCLEROSIS
channels also facilitates vasoconstriction to endothelin-1,105
but this does not occur via receptor-operated calcium entry.
Atherosclerosis
TRPC3 has also been shown to mediate constriction to the Atherosclerosis is the underlying pathologic process for both
nucleotide, uridine triphosphate.106 Myogenic tone in cerebral coronary and cerebral artery disease.1 However, atheroscle-
arteries isolated from hypertensive mice was inhibited by rotic lesions progress at a slower rate in intracranial arteries
treatment with SKF93635 (a specific inhibitor of TRPC6 compared with extracranial arteries in both animal models
channels at the concentration used in that study). SKF93635 and humans.119 Atherosclerosis is thought to be initiated by
was without effect in arteries from aged mice, suggesting trapping of lipids in the subendothelial layer, leading to the
TRP channel function is disrupted in cerebral arteries from generation of biologically active oxidized species (i.e., oxi-
aged mice.107 Some TRP channels, such as the vanilloid TRP dized low-density lipoprotein [LDL]), ultimately leading
channel (TRPV3), are chemosensitive. The TRPV3 channel to recruitment of leukocytes to the artery wall.120 Oxidative
is expressed in the endothelium of cerebral arteries, and the modification of LDL present in the intima by ROS may thus
dietary agonist carvacrol, which may be cardioprotective, be a key initiating step in atherosclerosis.121 Endothelial dys-
mediates endothelium-dependent cerebral vasodilatation function is an early step in the development of atherosclerosis,
that is inhibited by a pharmacologic inhibitor of TRPV1-4 and traditional cardiovascular risk factors (e.g., dyslipidemia,
channels.108 TRPV4 channels are expressed in endothelium hypertension) are associated with endothelial dysfunction.122
and vascular muscle cells and appear to mediate vasodilation. Furthermore, atherosclerosis is characterized by chronic
While activation of TRPV4 channels results in calcium entry inflammation of the vasculature; thus these three key pro-
in vascular muscle cells, the resulting calcium sparks activate cesses characteristic of atherosclerosis—oxidative stress, endo-
BK channels and thus hyperpolarization and dilation of the thelial dysfunction, and inflammation—will be discussed here
artery.109 Endothelial TRPV4 channels are activated (resulting (also summarized in Fig. 1.1), with much of the discussion
in calcium influx) and mediate dilation in response to shear referring to data from the apolipoprotein E-deficient (ApoE−/−)

Hypercholesterolemia Hypertension / ↑Ang II

Fig. 1.1. Schematic diagram summarizing cerebrovascular


effects of hypercholesterolemia, and elevated Ang II and ↑ NOX2 oxidase ↑ AT1R
hypertension. Hypercholesterolemia induces oxidative
stress, and ultimately inflammation—comprising leukocyte ↑ NADPH oxidases
and platelet adhesion, and endothelial dysfunction. These ↑ ROS
effects are all attenuated in NOX2 oxidase-deficient mice.
↑ O2–, ONOO –
Ang II increases leukocyte and platelet adhesion, infiltration
of inflammatory/immune cells, and causes endothelial
dysfunction due to reduced nitric oxide (NO) bioavailability. ↑ Leukocyte ↓ NO Inflammation
and
These effects are largely inhibited by AT1 receptor (AT1R) ↑ Leukocyte ↓ NO Infiltration of
platelet
inhibitors and in AT1R-deficient mice; reactive oxygen and inflammatory,
adhesion
species (ROS) scavengers and NOX2 oxidase-deficiency, as platelet immune cells
well as in lymphocyte-deficiency (RAG−/−) mice, implicating Endothelial adhesion
AT1R; and nicotinamide adenine dinucleotide phosphate dysfunction
Endothelial
(NADPH) oxidase-derived ROS and the adaptive immune dysfunction ↑ Pro-inflammatory
system in the detrimental effects of chronic hypertension in cytokines and
the cerebral circulation. chemokines
Cerebral Vascular Biology in Health and Disease 7

mouse. The ApoE−/− mouse, characterized by high levels of in cerebral vessels of hypercholesterolemic mice—leukocyte
plasma cholesterol due to deletion of the APOE gene (impor- and platelet adhesion was prevented by immunoneutraliza- 1
tant in cholesterol metabolism), provides a very useful experi- tion of P-selectin and in NOX2-deficient mice, suggesting that
mental model for understanding the mechanisms of disease P-selectin and NOX2-dependent oxidative stress are important
initiation.1 mechanisms in hypercholesterolemia-induced inflammation
in the brain.123 Arginase type 1 expression was also increased
Cerebral Vascular Oxidative Stress in Models of in cerebral vessels from ApoE−/− mice,134 which is relevant
since oxidized LDL increases arginase activity and decreases
Atherosclerosis endothelial NO levels, ultimately leading to impaired NO
Some evidence suggests the prevalence of oxidative stress in function in the vascular endothelium.135 Vascular cell adhe-
cerebral vessels during hypercholesterolemia or atherosclero- sion molecule-1 (VCAM-1) expression was not altered in brain
sis. For example, in wild-type mice placed on a high choles- microvessels of ApoE−/− mice.136
terol diet for 2 weeks,123 and ApoE−/− mice on high-fat diet for
7 weeks,124 oxidative stress was found to be present in cerebral
arteries. The study by Miller et al.124 went on to suggest that
Hypertension
NOX2 oxidase was the source of the oxidative stress, as the oxi- Hypertension profoundly and negatively impacts the cere-
dative stress present in ApoE−/− was abolished in mice deficient bral circulation and brain, and is a major risk factor for stroke
in both ApoE and NOX2 (i.e., ApoE−/−/NOX2−/y; Fig. 1.2). and a leading cause of cognitive decline and dementia.137
Hypertension may promote the formation of atherosclerotic
Cerebral Vascular Endothelial Dysfunction in Models of plaques in cerebral arteries and arterioles,137 and there is a wealth
of experimental evidence demonstrating detrimental functional
Atherosclerosis consequences of hypertension on the cerebral circulation. Many
Several lines of evidence suggest that atherosclerosis is asso- initial studies focused on the spontaneously hypertensive rat
ciated with reduced NO bioavailability and endothelial dys- (SHR), where augmented NADPH oxidase-derived superoxide
function. In earlier reports, relaxation responses of the basilar production91 and impaired endothelium-dependent responses
artery to acetylcholine were impaired in hypercholesterolemic have been reported.58,75,138–141 What follows is a discussion of
versus normal rabbits,125 although cerebral vascular responses more recent data regarding the influence of hypertension on
to acetylcholine were reportedly preserved126,127 or even aug- the cerebral circulation—specifically, hypertension in response
mented128 during atherosclerosis. In atherosclerotic mon- to elevated angiotensin II (Ang II) levels (also summarized in
keys, contraction of basilar arteries in response to inhibition Fig. 1.1). Ang II is of major importance because it is involved in
of soluble guanylate cyclase was reduced compared with that many of the functional and structural changes occurring in the
in normal monkeys, suggesting the basal influence of soluble cerebral circulation during chronic hypertension.5,119,137
guanylate cyclase on basal tone of cerebral arteries is dimin-
ished during atherosclerosis, perhaps reflecting a reduced pro- Oxidative Stress in Hypertension Involving
duction/activity of NO during atherosclerosis.129 Similarly,
cerebral artery contractions in response to the application
Elevated Ang II
of l-NAME (a NOS inhibitor) were reduced in vessels from Ang II increases ROS production in the cerebral circulation.
ApoE−/− compared to normal mice,124 suggesting reduced NO Work from Iadecola’s group has found that acute intravenous
bioavailability was present during atherosclerosis. Reduced infusion of mice with Ang II increases both blood pressure
cerebral vascular relaxation to acetylcholine in ApoE−/ versus and ROS production by cerebral blood vessels.142–146 Increased
normal mice further suggests that reduced NO bioavailability ROS levels were prevented by treatment with the ROS scavenger
is associated with endothelial dysfunction in the cerebral cir- MnTBAP.146 This treatment also reportedly increases 3-nitro-
culation during atherosclerosis.130,131 Interestingly, magnetic tyrosine immunoreactivity (indicative of nitrosative stress) in
resonance imaging of cerebral arteries in rabbits fed a diet mouse cerebral vascular endothelial cells, an effect that was
high in cholesterol were narrower compared with their control prevented by a peroxynitrite scavenger and a NOS inhibitor,
counterparts,132 which may suggest increased vascular tone or and was also absent in NOX2 oxidase-deficient mice.143 Thus
potentially structural alterations. these findings suggest that Ang II increases peroxynitrite forma-
Further experiments were conducted to provide a link tion in the cerebral vasculature largely via the reaction of NOX2
between oxidative stress and vascular dysfunction. Impaired oxidase-derived superoxide with NO (see Fig. 1.2).
NO-dependent responses of cerebral vessels from ApoE−/−
mice were reversed in vessels from ApoE−/− mice treated Endothelial Dysfunction in Hypertension Involving
with a scavenger of ROS (tempol),124,130 the nicotinamide
adenine dinucleotide phosphate (NADPH) oxidase inhibitor
Elevated Ang II
apocynin,130 or in ApoE−/−/NOX2−/y mice,124 strongly suggesting Acute intravenous administration of Ang II has been reported
that NOX2 oxidase-derived superoxide is a major mediator of to impair NO-dependent increases in cerebral blood
cerebral vascular dysfunction during atherosclerosis (see Fig. 1.2). flow(CBF),145,146 an effect that was reversed by MnTBAP and
Oxidative stress and endothelial dysfunction is present despite the angiotensin type 1 (AT1) receptor antagonist losartan.146
the apparent absence of lesions in cerebral blood vessels.124,130 Topical application of Ang II to cerebral arterioles in vivo
causes impaired NO-dependent responses that can be pre-
Cerebral Vascular Inflammation in Models of vented by the superoxide scavenger tiron.147 Similarly, Ang
II-induced endothelial dysfunction in cerebral arterioles of
Atherosclerosis ECSOD-deficient mice in vivo was reversed by tempol.148 In a
Atherosclerosis is characterized by chronic inflammation of more chronic model of Ang II-dependent hypertension, Ang II
the vasculature. Platelet endothelial cell adhesion molecule-1 increased blood pressure and caused endothelial dysfunction
(PECAM-1) is involved in the inflammatory process and of the basilar artery. This effect of Ang II was absent in NOX2
in leukocyte-endothelial interactions, and its expression is oxidase-deficient mice, and partially attenuated in NOX1
increased in cerebral arterioles of ApoE−/− mice.133 Leukocyte oxidase-deficient mice, suggesting Ang II-induced endothelial
and platelet adhesion, as well as oxidative stress, were elevated dysfunction is dependent on NOX2 oxidase and, perhaps to
8 SECTION I Pathophysiology

some, extent NOX1 oxidase.149 In spite of these findings, Ang dysfunction in Ang II hypertension via a mechanism involving
II increases blood pressure in both NOX2 and NOX1 oxidase TRPV4.153 Thus there is potential cross-talk between AT1R and
deficient mice, suggesting that the cerebral vascular and pressor MR. In a genetic model of hypertension (mice overexpressing
actions of Ang II are independent of one another.149 To further human renin and angiotensinogen), endothelial dysfunction of
confirm this point, previous studies have reported that systemic the basilar artery was completely reversed by the administration
administration of a nonpressor dose of Ang II caused endo- of polyethylene glycol superoxide dismutase (PEG-SOD).154
thelial dysfunction in the cerebral circulation.150 In addition, Taken together, these data suggest that Ang II causes endothe-
endothelial dysfunction precedes the development of hyperten- lial dysfunction in the cerebral circulation by activating AT1R
sion in response to a slow-pressor dose of Ang II.151 Endothelial expressed in the vessel wall, leading to an increase in superox-
dysfunction in response to Ang II was reversed by ROS scav- ide production, and subsequent oxidative inactivation of NO
engers.150,152 It has recently been reported that inhibition of (see Fig. 1.2). Recent evidence suggests that MR may also be
the mineralocorticoid receptor (MR) improves endothelial involved in the dysfunction caused by Ang II.

Hypercholesterolemia/ Hypertension
atherosclerosis

Ang II

Rho kinase AT1

NOX2
eNOS
oxidase Endothelial cell

O2– NO O2–

ONOO–

BKCa KIR KV KATP


K+ K+ K+ K+ Rho kinase P
MLC
sGC
Impaired membrane
hyperpolarization MLCP
Hyperpolarization

MLC

Vasodilation Vasoconstriction

Smooth muscle cell

NOX2
oxidase AT1

Ang II Perivascular macrophage


Hypertension

Fig. 1.2. Atherosclerosis/hypercholesterolemia and hypertension profoundly alter key mechanisms that modulate cerebral artery tone.
Atherosclerosis/hypercholesterolemia and hypertension increase oxidative stress via activation of NOX2 oxidase. The increased superoxide (O2−)
levels scavenges endothelial nitric oxide synthase (eNOS)-derived nitric oxide (NO), resulting in reduced NO bioavailability and hence reduced
NO-mediated vasodilation and peroxynitrite formation (ONOO−). ONOO− can directly influence cerebrovascular tone (see text). K+ channel activity
modulates vascular tone. Most studies have investigated the effect of hypertension on BKCa channel function, with the outcome (i.e., increased
or decreased channel function) depending on the model of hypertension studied (see text). Baseline KV channel function is impaired, as is
KATP-mediated vasodilatation compared with normotensive conditions. Baseline KIR channel function is augmented, whereas KIR-mediated K+-
induced vasodilatation is impaired. In endothelial cells, Rho kinase can reduce eNOS activity (see text for details). In vascular muscle, Rho kinase
phosphorylates (and inactivates) myosin light chain phosphatase (MLCP), leading to enhanced phosphorylation of myosin light chain (MLC) and
increased contractility. During hypertension, Rho kinase activity is increased, impairing normal cerebrovascular regulation.
Cerebral Vascular Biology in Health and Disease 9

Cerebrovascular Inflammation in Hypertension Involving pharmacologic inhibition of these channels (with TEA and
iberiotoxin) elicits greater contraction of cerebral arteries from 1
Elevated Ang II hypertensive vs normotensive rats.10 Consistent with this, ibe-
Hypertension induces inflammation in the cerebral circula- riotoxin elicited enhanced contraction of cerebral arterioles
tion. This includes models that involve Ang II. Ang II results from hypertensive versus normotensive rats, an effect associ-
in elevated leukocyte and platelet adhesion in cerebral ves- ated with enhanced cerebral vascular expression of the KCa
sels, an effect prevented by the AT1 receptor antagonists can- channel α subunit.163 Inhibition of BKCa channels with TEA
desartan and losartan, as well as diphenyleneiodonium, an and charybdotoxin cause cerebral vascular contraction in hy-
inhibitor of flavoproteins such as NADPH oxidase.155 In stud- pertensive but not normotensive rats.164
ies performed to extend these findings, Ang II-induced hyper- By contrast, in a model of Ang II-dependent hypertension,
tension was associated with a marked increase in leukocyte contraction of cerebral arteries in response to iberiotoxin was
and platelet adhesion in cerebral vessels, which was attenu- reduced in hypertensive vs normotensive rats, and this was
ated in RAG−/− mice (i.e., deficient in T and B lymphocytes), associated with reduced coupling efficiency between Ca2+ sparks
AT1R−/− mice, and by treatment with losartan,156 suggesting and BKCa channels, as well as reduced β1 subunit expression,
the involvement of immune cells and AT1 receptors in this although α subunit expression was unaltered during chronic
effect. Leukocyte adhesion in response to Ang II in pial vessels hypertension.165 The mechanism may involve calcineurin/
in vivo was also prevented by tempol,157 further confirming NFATc3 signaling, as Ang II-induced reduction in β1 subunit
that cerebrovascular inflammation involves ROS production expression was absent in calcineurin/NFATc3-deficient mice.
and oxidative stress. Interestingly, although leukocyte and Calcineurin/NFATc3 is also important in the development
platelet adhesion in cerebral vessels was enhanced in models of Ang II-dependent hypertension.166 Furthermore, although
of Ang II and deoxycorticosterone acetate (DOCA)-salt hyper- iberiotoxin caused myogenic constriction in normal mice, in
tension, these effects were prevented in the presence of mild a model of Ang II-dependent hypertension, it had no effect
hypercholesterolemia, possibly due to the involvement of on myogenic constriction in cerebral arteries.107 In a model
high-density lipoprotein (HDL), suggesting that mild eleva- of diet-induced obesity where blood pressure was elevated,
tions in certain types of cholesterol may be beneficial in the cerebral vascular BKCaβ1 subunit expression was increased,
setting of hypertension.158 although myogenic tone was not altered.17 Thus functional
Other models of hypertension have also implicated a role alterations in KCa channels appear to be dependent on the
for Ang II in cerebrovascular inflammation. For example, model of experimental hypertension studied.
in a DOCA-salt model of hypertension, leukocyte and KATP Channels. To our knowledge, there is little information
platelet adhesion was prevented by losartan and in AT1R−/− regarding KATP channel function in hypertension. Vasodilator
mice.159 Furthermore, not only were these effects inhibited responses to the KATP channel activator aprikalim were signifi-
by tempol; they were also inhibited by mito-tempol,159 cantly impaired in cerebral arteries from hypertensive versus
implicating mitochondria-derived ROS in the cerebrovascular normotensive rats, suggesting impaired KATP channel function
inflammatory response. These anti-inflammatory effects during hypertension.138 Although SUR2B expression appears
occurred in the absence of any depressor action, suggesting to be increased in small cerebral arteries from hypertensive
that blood pressure is not necessarily a key mediator of rats compared with their normotensive controls,167 the func-
cerebrovascular inflammation where Ang II is involved. In the tional significance of this finding is unknown.
SHR, increased expression of intracellular adhesion molecule-1
(ICAM-1), as well as an increased number of infiltrating and KV Channels. Experimental hypertension may be associated
adherent macrophages in brain microvessels, were inhibited with cerebral artery depolarization and increased myogenic re-
by candesartan.160 Widespread inflammation in many brain sponse, perhaps indicating impaired KV channel function. Phar-
regions of the SHR was also inhibited by candesartan,161 macologic inhibition of KV channels with correolide and psora-4
further implicating a role for activation of AT1R by Ang II constricted cerebral arteries from normotensive rats, but was
and demonstrating a beneficial use for AT1R inhibitors in without effect in cerebral arteries from two models of hyperten-
preventing inflammation associated with cerebrovascular sion, suggesting a reduced contribution of KV channels to the
disease. modulation of basal tone. This was associated with reduced ex-
Perivascular macrophages appear to play a central pression of the pore-forming α1.2 and α1.5 subunits that compose
role in cerebrovascular dysfunction in response to Ang KV channels in hypertensive versus normotensive rats.168 This is
II.162 Faraco et al. showed that Ang II acts on AT1R on in agreeance with the impaired KV2 channel function of cerebral
perivascular macrophages, resulting in NOX2 oxidase- arteries reported in a model of Ang II-dependent hypertension,
dependent ROS production and endothelial dysfunction.162 in that stromatoxin-induced contraction of cerebral arteries was
Thus inflammation appears to be a key mechanism, decreased in arteries from hypertensive vs normotensive rats.36
leading to both oxidative stress and subsequent endothelial In Dahl salt-sensitive rats, KV channel current density was de-
dysfunction. creased in cerebral artery myocytes from hypertensive vs normo-
tensive rats.169 Lower KV current density was reported in cerebral
vascular smooth muscle cells from SHR compared with WKY.170
K+ Channel Function in Chronic Hypertension
Expression of K+ channels in the cerebral vasculature and the KIR Channels. The first evidence for impaired KIR channel
importance of their role in modulating arterial tone, includ- function during chronic hypertension was the finding that
ing mediation of vasodilator responses, has been described. Ba2+-sensitive cerebral vascular relaxant responses to K+ in hy-
The deleterious actions of chronic hypertension in the cere- pertensive rats were impaired when compared with normo-
bral vasculature are also well known;137 it is thus unsurprising tensive controls.171 A subsequent study reported altered KIR
that K+ channel function is altered in association with chronic channel function during chronic hypertension, whereby KIR
hypertension (see Fig. 1.2). channels were not the predominant mediator of cerebral vaso-
dilator responses to K+, unlike in normal animals. This was
BKCa Channels. Basal activity of BKCa channels may be great- despite responses to K+ being preserved (or even enhanced),
er in cerebral arteries during chronic hypertension, in that KIR2.1 expression being preserved, and an enhanced role for
10 SECTION I Pathophysiology

KIR channels in modulating arterial tone during chronic hy- 59. Marrelli SP, Johnson TD, Khorovets A, Childres WF, Bryan Jr RM.
pertension.58 In the cerebral microvasculature, preserved (or Altered function of inward rectifier potassium channels in cere-
even enhanced) vasorelaxation to K+ during chronic hyper- brovascular smooth muscle after ischemia/reperfusion. Stroke.
tension was mediated by KIR channels.55 KIR channel function 1998;29:1469–1474.
60. Longden TA, Dabertrand F, Koide M, et al. Capillary k(+)-sensing
may thus be preserved in smaller arterioles55 during chronic initiates retrograde hyperpolarization to increase local cerebral
hypertension, as opposed to impaired function in larger arter- blood flow. Nat Neurosci. 2017;20:717–726.
ies,58,171 at least in mediating responses to K+. 75. Chrissobolis S, Sobey CG. Evidence that rho-kinase activity con-
tributes to cerebral vascular tone in vivo and is enhanced during
chronic hypertension: comparison with protein kinase c. Circ
Rho-Kinase in Hypertension Res. 2001;88:774–779.
Enhanced dilator responses of the basilar artery to Y-27632 78. Kitazono T, Ago T, Kamouchi M, et al. Increased activity of cal-
in models of chronic hypertension suggest an increase in cium channels and rho-associated kinase in the basilar artery
Rho-kinase function in hypertension (see Fig. 1.2).75,77,78 during chronic hypertension in vivo. J Hypertens. 2002;20:879–
884.
Furthermore, pressure-dependent development of myogenic 80. De Silva TM, Kinzenbaw DA, Modrick ML, Reinhardt LD, Faraci
tone of cerebral arteries is inhibited by Y-27632 to a greater FM. Heterogeneous impact of rock2 on carotid and cerebrovas-
extent in hypertensive vs normotensive rats,172 thus support- cular function. Hypertension. 2016;68:809–817.
ing an important role for Rho-kinase in increased myogenic 89. Miller AA, Drummond GR, Mast AE, Schmidt HH, Sobey CG.
tone of cerebral arteries. A recent study demonstrated that Effect of gender on nadph-oxidase activity, expression, and
acute systemic elevations in endothelin-1 levels impaired cere- function in the cerebral circulation: role of estrogen. Stroke.
bral vascular endothelial function, an effect that was reversed 2007;38:2142–2149.
by Y-27632.173 Interestingly, the role of Rho-kinase as a key 90. Miller AA, Drummond GR, Schmidt HHW, Sobey CG. Nadph
mediator of cerebral vascular dysfunction during chronic oxidase activity and function are profoundly greater in cerebral
versus systemic arteries. Circ Res. 2005;97:1055–1062.
hypertension may be dependent on the cause of the hyperten- 95. De Silva TM, Broughton BR, Drummond GR, Sobey CG, Miller
sion, as Ang II-induced cerebral endothelial dysfunction was AA. Gender influences cerebral vascular responses to angio-
not reversed by Y-27632.173 tensin ii through nox2-derived reactive oxygen species. Stroke.
2009;40:1091–1097.
97. Maneen MJ, Hannah R, Vitullo L, DeLance N, Cipolla MJ. Per-
CONCLUSION oxynitrite diminishes myogenic activity and is associated with
Experimental evidence for some major mechanisms regulating decreased vascular smooth muscle f-actin in rat posterior cere-
cerebral vascular function has been presented, together with bral arteries. Stroke. 2006;37:894–899.
how many of these mechanisms are altered during hyperten- 103. Earley S, Brayden JE. Transient receptor potential channels in the
vasculature. Physiol Rev. 2015;95:645–690.
sion and atherosclerosis—two disease states that predispose 119. Hu X, De Silva TM, Chen J, Faraci FM. Cerebral vascular dis-
to clinical stroke. Consequently, molecular targets that may be ease and neurovascular injury in ischemic stroke. Circ Res.
of benefit in cerebrovascular disease, and perhaps the preven- 2017;120:449–471.
tion and/or treatment of ischemic stroke, are being identified. 123. Ishikawa M, Stokes KY, Zhang JH, Nanda A, Granger DN. Cere-
However, clearly further work is needed to ultimately iden- bral microvascular responses to hypercholesterolemia: roles of
tify effective therapies, as these diseases are currently poorly nadph oxidase and p-selectin. Circ Res. 2004;94:239–244.
controlled. 124. Miller AA, De Silva TM, Judkins CP, Diep H, Drummond GR,
Sobey CG. Augmented superoxide production by nox2-con-
The complete reference list is available at taining nadph oxidase causes cerebral artery dysfunction during
hypercholesterolemia. Stroke. 2010;41:784–789.
www.expertconsult.inkling.com. 137. Iadecola C, Davisson RL. Hypertension and cerebrovascular dys-
function. Cell Metab. 2008;7:476–484.
KEY REFERENCES 143. Girouard H, Park L, Anrather J, Zhou P, Iadecola C. Cerebrovas-
cular nitrosative stress mediates neurovascular and endothelial
1. Hansson GK, Hermansson A. The immune system in atheroscle- dysfunction induced by angiotensin ii. Arterioscler Thromb Vasc
rosis. Nat Immunol. 2011;12:204–212. Biol. 2007;27:303–309.
2. Chan SL, Sweet JG, Bishop N, Cipolla MJ. Pial collateral reactiv- 146. Girouard H, Park L, Anrather J, Zhou P, Iadecola C. Angiotensin
ity during hypertension and aging: understanding the function ii attenuates endothelium-dependent responses in the cerebral
of collaterals for stroke therapy. Stroke. 2016;47:1618–1625. microcirculation through nox-2-derived radicals. Arterioscler
13. Dabertrand F, Nelson MT, Brayden JE. Acidosis dilates brain Thromb Vasc Biol. 2006;26:826–832.
parenchymal arterioles by conversion of calcium waves to sparks 149. Chrissobolis S, Banfi B, Sobey CG, Faraci FM. Role of nox iso-
to activate bk channels. Circ Res. 2012;110:285–294. forms in angiotensin ii-induced oxidative stress and endothelial
23. Adebiyi A, McNally EM, Jaggar JH. Vasodilation induced by dysfunction in brain. J Appl Physiol. 2012;113:184–191.
oxygen/glucose deprivation is attenuated in cerebral arteries of 151. Capone C, Faraco G, Park L, Cao X, Davisson RL, Iadecola C.
sur2 null mice. Am J Physiol Heart Circ Physiol. 2011;301:H1360– The cerebrovascular dysfunction induced by slow pressor doses
H1368. of angiotensin ii precedes the development of hypertension. Am
38. Chrissobolis S, Ziogas J, Chu Y, Faraci FM, Sobey CG. Role J Physiol Heart Circ Physiol. 2011;300:H397–H407.
of inwardly rectifying k(+) channels in k(+)-induced cere- 157. Zhang M, Mao Y, Ramirez SH, Tuma RF, Chabrashvili T. Angio-
bral vasodilatation in vivo. Am J Physiol Heart Circ Physiol. tensin ii induced cerebral microvascular inflammation and
2000;279:H2704–H2712. increased blood-brain barrier permeability via oxidative stress.
46. Filosa JA, Bonev AD, Straub SV, et al. Local potassium signaling Neuroscience. 2010;171:852–858.
couples neuronal activity to vasodilation in the brain. Nat Neu- 162. Faraco G, Sugiyama Y, Lane D, et al. Perivascular macrophages
rosci. 2006;9:1397–1403. mediate the neurovascular and cognitive dysfunction associated
57. Chrissobolis S, Sobey CG. Inhibitory effects of protein kinase c with hypertension. J Clin Invest. 2016;126:4674–4689.
on inwardly rectifying k+- and atp-sensitive k+ channel-medi- 173. Faraco G, Moraga A, Moore J, Anrather J, Pickel VM, Iadecola
ated responses of the basilar artery. Stroke. 2002;33:1692–1697. C. Circulating endothelin-1 alters critical mechanisms regulating
58. Chrissobolis S, Ziogas J, Anderson CR, Chu Y, Faraci FM, Sobey cerebral microcirculation. Hypertension. 2013;62:759–766.
CG. Neuronal no mediates cerebral vasodilator responses to k+
in hypertensive rats. Hypertension. 2002;39:880–885.
REFERENCES 23. Adebiyi A, McNally EM, Jaggar JH. Vasodilation induced by
oxygen/glucose deprivation is attenuated in cerebral arteries of
1. Hansson GK, Hermansson A. The immune system in atheroscle-
sur2 null mice. Am J Physiol Heart Circ Physiol. 2011;301:H1360–
rosis. Nat Immunol. 2011;12:204–212.
H1368.
2. Chan SL, Sweet JG, Bishop N, Cipolla MJ. Pial collateral reactiv-
24. Liang GH, Adebiyi A, Leo MD, McNally EM, Leffler CW, Jag-
ity during hypertension and aging: understanding the function
gar JH. Hydrogen sulfide dilates cerebral arterioles by activat-
of collaterals for stroke therapy. Stroke. 2016;47:1618–1625.
ing smooth muscle cell plasma membrane katp channels. Am J
3. Nishimura N, Schaffer CB, Friedman B, Lyden PD, Kleinfeld D.
Physiol Heart Circ Physiol. 2011;300:H2088–H2095.
Penetrating arterioles are a bottleneck in the perfusion of neocor-
25. Lindauer U, Vogt J, Schuh-Hofer S, Dreier JP, Dirnagl U. Cerebro-
tex. Proc Natl Acad Sci U S A. 2007;104:365–370.
vascular vasodilation to extraluminal acidosis occurs via com-
4. Mayhan WG, Heistad DD. Role of veins and cerebral venous
bined activation of atp-sensitive and ca2+-activated potassium
pressure in disruption of the blood-brain barrier. Circ Res.
channels. J Cereb Blood Flow Metab. 2003;23:1227–1238.
1986;59:216–220.
26. Rosenblum WI, Wei EP, Kontos HA. Vasodilation of brain surface
5. Chrissobolis S, Miller AA, Drummond GR, Kemp-Harper BK,
arterioles by blockade of na-h+ antiport and its inhibition by
Sobey CG. Oxidative stress and endothelial dysfunction in cere-
inhibitors of katp channel openers. Brain Res. 2004;1005:77–83.
brovascular disease. Front Biosci. 2011;16:1733–1745.
27. Philip S, Armstead WM. Nmda dilates pial arteries by katp and
6. Faraci FM, Heistad DD. Regulation of the cerebral circula-
kca channel activation. Brain Res Bull. 2004;63:127–131.
tion: role of endothelium and potassium channels. Physiol Rev.
28. Taguchi H, Heistad DD, Kitazono T, Faraci FM. Atp-sensitive
1998;78:53–97.
k+ channels mediate dilatation of cerebral arterioles during
7. Zhang F, Xu S, Iadecola C. Role of nitric oxide and acetylcholine
hypoxia. Circ Res. 1994;74:1005–1008.
in neocortical hyperemia elicited by basal forebrain stimulation:
29. Sobey CG, Faraci FM. Inhibitory effect of 4–aminopyridine on
evidence for an involvement of endothelial nitric oxide. Neurosci-
responses of the basilar artery to nitric oxide. Br J Pharmacol.
ence. 1995;69:1195–1204.
1999;126:1437–1443.
8. Faraci FM, Sobey CG. Role of potassium channels in regulation of
30. Quan L, Sobey CG. Selective effects of subarachnoid hemorrhage
cerebral vascular tone. J Cereb Blood Flow Metab. 1998;18:1047–
on cerebral vascular responses to 4-aminopyridine in rats [in
1063.
process citation]. Stroke. 2000;31:246–2465.
9. Sobey CG, Faraci FM. Effect of nitric oxide and potassium chan-
31. Jarajapu YP, Oomen C, Uteshev VV, Knot HJ. Histamine
nel agonists and inhibitors on basilar artery diameter. Am J
decreases myogenic tone in rat cerebral arteries by h2-receptor-
Physiol Heart Circ Physiol. 1997;272:H256–H262.
mediated kv channel activation, independent of endothelium
10. Paterno R, Heistad DD, Faraci FM. Functional activity of
and cyclic amp. Eur J Pharmacol. 2006;547:116–124.
ca2+-dependent k+ channels is increased in basilar artery
32. Albarwani S, Nemetz LT, Madden JA, et al. Voltage-gated k+
during chronic hypertension. Am J Physiol Heart Circ Physiol.
channels in rat small cerebral arteries: molecular identity of the
1997;272:H1287–H1291.
functional channels. J Physiol. 2003;551:751–763.
11. Lohn M, Lauterbach B, Haller H, Pongs O, Luft FC, Gollasch M.
33. Cheong A, Dedman AM, Beech DJ. Expression and function of
Beta(1)-subunit of bk channels regulates arterial wall[ca(2+)] and
native potassium channel [k(v)alpha1] subunits in terminal
diameter in mouse cerebral arteries. J Appl Physiol. 2001;91:1350–
arterioles of rabbit. J Physiol. 2001;534:691–700.
1354.
34. Ishiguro M, Morielli AD, Zvarova K, Tranmer BI, Penar PL,
12. Paterno R, F.M.F., Heistad DD. Role of ca2+-dependent k+ chan-
Wellman GC. Oxyhemoglobin-induced suppression of voltage-
nels in cerebral vasodilatation induced by increases in cyclic gmp
dependent k+ channels in cerebral arteries by enhanced tyrosine
and cyclic amp in the rat. Stroke. 1996;27:1603–1608.
kinase activity. Circ Res. 2006;99:1252–1260.
13. Dabertrand F, Nelson MT, Brayden JE. Acidosis dilates brain
35. Jahromi BS, Aihara Y, Ai J, Zhang ZD, Nikitina E, Macdonald RL.
parenchymal arterioles by conversion of calcium waves to sparks
Voltage-gated k+ channel dysfunction in myocytes from a dog
to activate bk channels. Circ Res. 2012;110:285–294.
model of subarachnoid hemorrhage. J Cereb Blood Flow Metab.
14. Liang GH, Xi Q, Leffler CW, Jaggar JH. Hydrogen sulfide activates
2008;28:797–811.
ca(2)(+) sparks to induce cerebral arteriole dilatation. J Physiol.
36. Amberg GC, Santana LF. Kv2 channels oppose myogenic
2012;590:2709–2720.
constriction of rat cerebral arteries. Am J Physiol Cell Physiol.
15. Jackson-Weaver O, Paredes DA, Gonzalez Bosc LV, Walker BR,
2006;291:C348–C356.
Kanagy NL. Intermittent hypoxia in rats increases myogenic tone
37. Cheong A, Dedman AM, Xu SZ, Beech DJ. K(v)alpha1 channels
through loss of hydrogen sulfide activation of large-conductance
in murine arterioles: differential cellular expression and regula-
ca(2+)-activated potassium channels. Circ Res. 2011;108:1439–
tion of diameter. Am J Physiol Heart Circ Physiol. 2001;281:H1057–
1447.
H1065.
16. Zhao G, Adebiyi A, Xi Q, Jaggar JH. Hypoxia reduces kca chan-
38. Chrissobolis S, Ziogas J, Chu Y, Faraci FM, Sobey CG. Role
nel activity by inducing ca2+ spark uncoupling in cerebral artery
of inwardly rectifying k(+) channels in k(+)-induced cere-
smooth muscle cells. Am J Physiol Cell Physiol. 2007;292:C2122–
bral vasodilatation in vivo. Am J Physiol Heart Circ Physiol.
C2128.
2000;279:H2704–H2712.
17. Howitt L, Sandow SL, Grayson TH, Ellis ZE, Morris MJ, Murphy
39. Horiuchi T, Dietrich HH, Tsugane S, Dacey Jr RG. Role of potas-
TV. Differential effects of diet-induced obesity on bkca {beta}1-
sium channels in regulation of brain arteriolar tone: comparison
subunit expression and function in rat skeletal muscle arteri-
of cerebrum versus brain stem. Stroke. 2001;32:218–224.
oles and small cerebral arteries. Am J Physiol Heart Circ Physiol.
40. Johnson TD, Marrelli SP, Steenberg ML, Childres WF, Bryan RMJ.
2011;301:H29–H40.
Inward rectifier potassium channels in the rat middle cerebral
18. Kitazono T, Faraci FM, Taguchi H, Heistad DD. Role of potas-
artery. Am J Physiol Reg Int Comp Physiol. 1998;274:R541–R547.
sium channels in cerebral blood vessels. Stroke. 1995;26:1713–
41. Knot HJ, Zimmerman PA, Nelson MT. Extracellular k+-induced
1723.
hyperpolarizations and dilatations of rat coronary and cere-
19. Sobey CG. Potassium channel function in vascular disease. Arte-
bral arteries involve inward rectifier k+ channels. J Physiol.
rioscler Thromb Vasc Biol. 2001;21:28–38.
1996;492:419–430.
20. Faraci FM, Heistad DD. Role of atp-sensitive potassium channels
42. Prior HM, Webster N, Quinn K, Beech DJ, Yates MS. K+-induced
in the basilar artery. Am J Physiol Heart Circ Physiol. 1993;264:H8–
dilation of a small renal artery: no role for inward rectifier k+
H13.
channels. Cardiovasc Res. 1998;37:780–790.
21. Jansen-Olesen I, Mortensen CH, El-Bariaki N, Ploug KB. Charac-
43. Sun H, Zhao H, Sharpe GM, Arrick DM, Mayhan WG. Influence
terization of k(atp)-channels in rat basilar and middle cerebral
of chronic alcohol consumption on inward rectifier potassium
arteries: studies of vasomotor responses and mRNA expres-
channels in cerebral arterioles. Microvasc Res. 2008;75:367–372.
sion. Eur J Pharmacol. 2005;523:109–118.
44. Weyer GW, Jahromi BS, Aihara Y, et al. Expression and func-
22. Ploug KB, Edvinsson L, Olesen J, Jansen-Olesen I. Pharmacologi-
tion of inwardly rectifying potassium channels after experi-
cal and molecular comparison of k(atp) channels in rat basilar
mental subarachnoid hemorrhage. J Cereb Blood Flow Metab.
and middle cerebral arteries. Eur J Pharmacol. 2006;553:254–
2006;26:382–391.
262.

10.e1
10.e2 References

45. Zaritsky JJ, Eckman DM, Wellman GC, Nelson MT, Schwarz TL. 68. Somlyo AP, Somlyo AV. Ca2+ sensitivity of smooth muscle and
Targeted disruption of kir2.1 and kir2.2 genes reveals the essen- nonmuscle myosin ii: modulated by g proteins, kinases, and
tial role of the inwardly rectifying k(+) current in k(+)-mediated myosin phosphatase. Physiol Rev. 2003;83:1325–1358.
vasodilation. Circ Res. 2000;87:160–166. 69. Chrissobolis S, Sobey CG. Recent evidence for an involvement
46. Filosa JA, Bonev AD, Straub SV, et al. Local potassium signaling of rho-kinase in cerebral vascular disease. Stroke. 2006;37:2174–
couples neuronal activity to vasodilation in the brain. Nat Neu- 2180.
rosci. 2006;9:1397–1403. 70. Gokina NI, Park KM, McElroy-Yaggy K, Osol G. Effects of rho
47. Fujii K, Heistad DD, Faraci FM. Ionic mechanisms in spon- kinase inhibition on cerebral artery myogenic tone and reactiv-
taneous vasomotion of the rat basilar artery in vivo. J Physiol. ity. J Appl Physiol. 2005;98:1940–1948.
1990;430:389–398. 71. Lagaud G, Gaudreault N, Moore ED, Van Breemen C, Laher I.
48. McCarron JG, Halpern W. Potassium dilates rat cerebral arteries Pressure-dependent myogenic constriction of cerebral arteries
by two independent mechanisms. Am J Physiol Heart Circ Physiol. occurs independently of voltage-dependent activation. Am J
1990;259:H902–H908. Physiol Heart Circ Physiol. 2002;283:H2187–H2195.
49. Busija DW, Chen J. Reversal by increased csf [h+] and [k+] of 72. Lim M, Choi SK, Cho YE, et al. The role of sphingosine kinase 1/
phorbol ester-induced arteriolar constriction in piglets. Am J sphingosine-1-phosphate pathway in the myogenic tone of pos-
Physiol Heart Circ Physiol. 1992;263:H1455–H1459. terior cerebral arteries. PloS One. 2012;7:e35177.
50. Iadecola C, Kraig RP. Focal elevations in neocortical interstitial 73. Kold-Petersen H, Brondum E, Nilsson H, Flyvbjerg A, Aalkjaer C.
k+ produced by stimulation of the fastigial nucleus in rat. Brain Impaired myogenic tone in isolated cerebral and coronary resis-
Res. 1991;563:273–277. tance arteries from the goto-kakizaki rat model of type 2 diabe-
51. Kuschinsky W, Wahl M, Bosse O, Thurau K. Perivascular potas- tes. J Vasc Res. 2012;49:267–278.
sium and pH as determinants of local pial arterial diameter in 74. Chrissobolis S, Budzyn K, Marley PD, Sobey CG. Evidence that
cats. Circ Res. 1972;31:240–247. estrogen suppresses rho-kinase function in the cerebral circula-
52. McCulloch J, Edvinsson L, Watt P. Comparison of the effects of tion in vivo. Stroke. 2004;35:2200–2205.
potassium and pH on the calibre of cerebral veins and arteries. 75. Chrissobolis S, Sobey CG. Evidence that rho-kinase activity con-
Pflugers Arch. 1982;393:95–98. tributes to cerebral vascular tone in vivo and is enhanced during
53. Nguyen TS, Winn HR, Janigro D. Atp-sensitive potassium chronic hypertension: comparison with protein kinase c. Circ
channels may participate in the coupling of neuronal activ- Res. 2001;88:774–779.
ity and cerebrovascular tone. Am J Physiol Heart Circ Physiol. 76. Erdos B, Snipes JA, Kis B, Miller AW, Busija DW. Vasocon-
2000;278:H878–H885. strictor mechanisms in the cerebral circulation are unaffected
54. Paisansathan C, Xu H, Vetri F, Hernandez M, Pelligrino DA. Inter- by insulin resistance. Am J Physiol Regul Integr Comp Physiol.
actions between adenosine and k+ channel-related pathways in 2004;287:R1456–R1461.
the coupling of somatosensory activation and pial arteriolar dila- 77. Kitayama J, Kitazono T, Ooboshi H, Takada J, Fujishima M,
tion. Am J Physiol Heart Circ Physiol. 2010;299:H2009–H2017. Ibayashi S. Long-term effects of benidipine on cerebral vasoreac-
55. Nakahata K, Kinoshita H, Tokinaga Y, et al. Vasodilation medi- tivity in hypertensive rats. Eur J Pharmacol. 2002;438:153–158.
ated by inward rectifier k+ channels in cerebral microvessels of 78. Kitazono T, Ago T, Kamouchi M, et al. Increased activity of cal-
hypertensive and normotensive rats. Anesth Analg. 2006;102:571– cium channels and rho-associated kinase in the basilar artery
576. during chronic hypertension in vivo. J Hypertens. 2002;20:879–
56. Jin CZ, Kim HS, Seo EY, et al. Exercise training increases inwardly 884.
rectifying k(+) current and augments k(+)-mediated vasodilata- 79. Didion SP, Lynch CM, Baumbach GL, Faraci FM. Impaired
tion in deep femoral artery of rats. Cardiovasc Res. 2011;91:142– endothelium-dependent responses and enhanced influence
150. of rho-kinase in cerebral arterioles in type ii diabetes. Stroke.
57. Chrissobolis S, Sobey CG. Inhibitory effects of protein kinase c 2005;36:342–347.
on inwardly rectifying k+- and ATP-sensitive k+ channel-medi- 80. De Silva TM, Kinzenbaw DA, Modrick ML, Reinhardt LD, Faraci
ated responses of the basilar artery. Stroke. 2002;33:1692–1697. FM. Heterogeneous impact of rock2 on carotid and cerebrovas-
58. Chrissobolis S, Ziogas J, Anderson CR, Chu Y, Faraci FM, Sobey cular function. Hypertension. 2016;68:809–817.
CG. Neuronal no mediates cerebral vasodilator responses to k+ 81. Miller AA, Drummond GR, Sobey CG. Reactive oxygen species in
in hypertensive rats. Hypertension. 2002;39:880–885. the cerebral circulation: are they all bad? Antioxid Redox Signal.
59. Marrelli SP, Johnson TD, Khorovets A, Childres WF, Bryan Jr RM. 2006;8:1113–1120.
Altered function of inward rectifier potassium channels in cere- 82. Didion SP, Faraci FM. Effects of nadh and nadph on superoxide
brovascular smooth muscle after ischemia/reperfusion. Stroke. levels and cerebral vascular tone. Am J Physiol Heart Circ Physiol.
1998;29:1469–1474. 2002;282:H688–H695.
60. Longden TA, Dabertrand F, Koide M, et al. Capillary k(+)-sensing 83. Park L, Anrather J, Zhou P, Frys K, Wang G, Iadecola C. Exog-
initiates retrograde hyperpolarization to increase local cerebral enous nadph increases cerebral blood flow through nadph
blood flow. Nat Neurosci. 2017;20:717–726. oxidase-dependent and -independent mechanisms. Arterioscler
61. Ketchum KA, Joiner WJ, Sellers AJ, Kaczmarek LK, Goldstein SA. Thromb Vasc Biol. 2004;24:1860–1865.
A new family of outwardly rectifying potassium channel proteins 84. Wei EP, Christman CW, Kontos HA, Povlishock JT. Effects of oxy-
with two pore domains in tandem. Nature. 1995;376:690–695. gen radicals on cerebral arterioles. Am J Physiol. 1985;248:H157–
62. Lloyd EE, Marrelli SP, Bryan Jr RM. Cgmp does not activate two- H162.
pore domain k+ channels in cerebrovascular smooth muscle. Am 85. Wei EP, Kontos HA, Beckman JS. Mechanisms of cerebral vasodi-
J Physiol Heart Circ Physiol. 2009;296:H1774–H1780. lation by superoxide, hydrogen peroxide, and peroxynitrite. Am J
63. Bryan Jr RM, You J, Phillips SC, et al. Evidence for two-pore Physiol. 1996;271:H1262–H1266.
domain potassium channels in rat cerebral arteries. Am J Physiol 86. Cosentino F, Sill JC, Katusic ZS. Role of superoxide anions in the
Heart Circ Physiol. 2006;291:H770–H780. mediation of endothelium-dependent contractions. Hyperten-
64. Blondeau N, Petrault O, Manta S, et al. Polyunsaturated fatty sion. 1994;23:229–235.
acids are cerebral vasodilators via the trek-1 potassium channel. 87. Iida Y, Katusic ZS. Mechanisms of cerebral arterial relaxations to
Circ Res. 2007;101:176–184. hydrogen peroxide. Stroke. 2000;31:2224–2230.
65. Namiranian K, Lloyd EE, Crossland RF, et al. Cerebrovascular 88. Leffler CW, Busija DW, Armstead WM, Mirro R. H2o2 effects on
responses in mice deficient in the potassium channel, trek-1. Am cerebral prostanoids and pial arteriolar diameter in piglets. Am J
J Physiol Regul Integr Comp Physiol. 2010;299:R461–R469. Physiol. 1990;258:H1382–H1387.
66. Parelkar NK, Silswal N, Jansen K, Vaughn J, Bryan Jr RM. Andre- 89. Miller AA, Drummond GR, Mast AE, Schmidt HH, Sobey CG.
sen J. 2,2,2-trichloroethanol activates a nonclassical potassium Effect of gender on nadph-oxidase activity, expression, and
channel in cerebrovascular smooth muscle and dilates the mid- function in the cerebral circulation: role of estrogen. Stroke.
dle cerebral artery. J Pharmacol Exp Ther. 2010;332:803–810. 2007;38:2142–2149.
67. Somlyo AP, Somlyo AV. Signal transduction by g-proteins, rho- 90. Miller AA, Drummond GR, Schmidt HHW, Sobey CG. Nadph
kinase and protein phosphatase to smooth muscle and non- oxidase activity and function are profoundly greater in cerebral
muscle myosin ii. J Physiol (Lond). 2000;522(Pt 2):177–185. versus systemic arteries. Circ Res. 2005;97:1055–1062.
References 10.e3

91. Paravicini TM, Chrissobolis S, Drummond GR, Sobey CG. 115. Gonzales AL, Garcia ZI, Amberg GC, Earley S. Pharmacological
Increased nadph-oxidase activity and nox4 expression during inhibition of trpm4 hyperpolarizes vascular smooth muscle. Am 1
chronic hypertension is associated with enhanced cerebral vaso- J Physiol Cell Physiol. 2010;299:C1195–C1202.
dilatation to nadph in vivo. Stroke. 2004;35:584–589. 116. Crnich R, Amberg GC, Leo MD, et al. Vasoconstriction result-
92. Sobey CG, Heistad DD, Faraci FM. Mechanisms of bradykinin- ing from dynamic membrane trafficking of trpm4 in vascular
induced cerebral vasodilatation in rats. Evidence that reactive smooth muscle cells. Am J Physiol Cell Physiol. 2010;299:C682–
oxygen species activate k+ channels. Stroke. 1997;28:2290–2294. C694.
discussion 2295. 117. Earley S, Gonzales AL, Crnich R. Endothelium-dependent cere-
93. Sobey CG, Heistad DD, Faraci FM. Potassium channels mediate bral artery dilation mediated by trpa1 and ca2+-activated k+
dilatation of cerebral arterioles in response to arachidonate. Am channels. Circ Res. 2009;104:987–994.
J Physiol. 1998;275:H1606–H1612. 118. Narayanan D, Bulley S, Leo MD, et al. Smooth muscle cell
94. You J, Golding EM, Bryan Jr RM. Arachidonic acid metabolites, transient receptor potential polycystin-2 (trpp2) channels con-
hydrogen peroxide, and edhf in cerebral arteries. Am J Physiol tribute to the myogenic response in cerebral arteries. J Physiol.
Heart Circ Physiol. 2005;289:H1077–H1083. 2013;591:5031–5046.
95. De Silva TM, Broughton BR, Drummond GR, Sobey CG, Miller 119. Hu X, De Silva TM, Chen J, Faraci FM. Cerebral vascular dis-
AA. Gender influences cerebral vascular responses to angio- ease and neurovascular injury in ischemic stroke. Circ Res.
tensin ii through nox2-derived reactive oxygen species. Stroke. 2017;120:449–471.
2009;40:1091–1097. 120. Charo IF, Taub R. Anti-inflammatory therapeutics for the treat-
96. Li J, Li W, Altura BT, Altura BM. Peroxynitrite-induced relaxation ment of atherosclerosis. Nat Rev Drug Discov. 2011;10:365–376.
in isolated canine cerebral arteries and mechanisms of action. 121. Weber C, Noels H. Atherosclerosis: current pathogenesis and
Toxicol Appl Pharmacol. 2004;196:176–182. therapeutic options. Nat Med.17:1410–1422
97. Maneen MJ, Hannah R, Vitullo L, DeLance N, Cipolla MJ. Per- 122. Sitia S, Tomasoni L, Atzeni F, et al. From endothelial dysfunction
oxynitrite diminishes myogenic activity and is associated with to atherosclerosis. Autoimmun Rev. 2011;9:830–834.
decreased vascular smooth muscle f-actin in rat posterior cere- 123. Ishikawa M, Stokes KY, Zhang JH, Nanda A, Granger DN. Cere-
bral arteries. Stroke. 2006;37:894–899. bral microvascular responses to hypercholesterolemia: roles of
98. Brzezinska AK, Gebremedhin D, Chilian WM, Kalyanaraman B, nadph oxidase and p-selectin. Circ Res. 2004;94:239–244.
Elliott SJ. Peroxynitrite reversibly inhibits ca(2+)-activated k(+) 124. Miller AA, De Silva TM, Judkins CP, Diep H, Drummond GR,
channels in rat cerebral artery smooth muscle cells. Am J Physiol Sobey CG. Augmented superoxide production by nox2-con-
Heart Circ Physiol. 2000;278:H1883–H1890. taining nadph oxidase causes cerebral artery dysfunction during
99. Elliott SJ, Lacey DJ, Chilian WM, Brzezinska AK. Peroxynitrite is hypercholesterolemia. Stroke. 2010;41:784–789.
a contractile agonist of cerebral artery smooth muscle cells. Am J 125. Rossitch Jr E, Alexander 3rd E, Black PM, Cooke JP. L-argi-
Physiol. 1998;275:H1585–H1591. nine normalizes endothelial function in cerebral vessels from
100. Maneen MJ, Cipolla MJ. Peroxynitrite diminishes myogenic tone hypercholesterolemic rabbits. J Clin Invest. 1991;87:1295–
in cerebral arteries: role of nitrotyrosine and f-actin. Am J Physiol 1299.
Heart Circ Physiol. 2007;292:H1042–H1050. 126. Simonsen U, Ehrnrooth E, Gerdes LU, et al. Functional proper-
101. Earley S. Trpa1 channels in the vasculature. Br J Pharmacol. ties in vitro of systemic small arteries from rabbits fed a choles-
2012;167:13–22. terol-rich diet for 12 weeks. Clin Sci (Lond). 1991;80:119–129.
102. Earley S. Endothelium-dependent cerebral artery dilation medi- 127. Kanamaru K, Waga S, Tochio H, Nagatani K. The effect of athero-
ated by transient receptor potential and ca2+-activated k+ chan- sclerosis on endothelium-dependent relaxation in the aorta and
nels. J Cardiovasc Pharmacol. 2011;57:148–153. intracranial arteries of rabbits. J Neurosurg. 1989;70:793–798.
103. Earley S, Brayden JE. Transient receptor potential channels in the 128. Stewart-Lee AL, Burnstock G. Changes in vasoconstrictor and
vasculature. Physiol Rev. 2015;95:645–690. vasodilator responses of the basilar artery during maturation
104. Guibert C, Beech DJ. Positive and negative coupling of the endo- in the Watanabe heritable hyperlipidemic rabbit differ from
thelin eta receptor to ca2+-permeable channels in rabbit cerebral those in the New Zealand white rabbit. Arterioscler Thromb.
cortex arterioles. J Physiol. 1999;514(Pt 3):843–856. 1991;11:1147–1155.
105. Xi Q, Adebiyi A, Zhao G, et al. Ip3 constricts cerebral arteries 129. Didion SP, Heistad DD, Faraci FM. Mechanisms that produce
via ip3 receptor-mediated trpc3 channel activation and inde- nitric oxide-mediated relaxation of cerebral arteries during ath-
pendently of sarcoplasmic reticulum ca2+ release. Circ Res. erosclerosis. Stroke. 2001;32:761–766.
2008;102:1118–1126. 130. Kitayama J, Faraci FM, Lentz SR, Heistad DD. Cerebral vascular
106. Reading SA, Earley S, Waldron BJ, Welsh DG, Brayden JE. Trpc3 dysfunction during hypercholesterolemia. Stroke. 2007;38:2136–
mediates pyrimidine receptor-induced depolarization of cerebral 2141.
arteries. Am J Physiol Heart Circ Physiol. 2005;288:H2055–H2061. 131. Yamashiro K, Milsom AB, Duchene J, et al. Alterations in nitric
107. Toth P, Csiszar A, Tucsek Z, et al. Role of 20–hete,trp channels oxide and endothelin-1 bioactivity underlie cerebrovascular
& bkca in dysregulation of pressure-induced ca2+ signaling and dysfunction in apoe-deficient mice. J Cereb Blood Flow Metab.
myogenic constriction of cerebral arteries in aged hypertensive 2010;30:1494–1503.
mice. Am J Physiol Heart Circ Physiol. 2013;305:H1698–H1708. 132. Schreurs BG, Smith-Bell CA, Lemieux SK. Dietary cholesterol
108. Earley S, Gonzales AL, Garcia ZI. A dietary agonist of transient increases ventricular volume and narrows cerebrovascular diam-
receptor potential cation channel v3 elicits endothelium-depen- eter in a rabbit model of Alzheimer’s disease. Neuroscience.
dent vasodilation. Mol Pharmacol. 2010;77:612–620. 2013;254C:61–69.
109. Earley S, Heppner TJ, Nelson MT, Brayden JE. Trpv4 forms a 133. Bengrine A, Da Silveira C, Massy ZA, Boullier A, Bugnicourt JM,
novel ca2+ signaling complex with ryanodine receptors and bkca Chillon JM. Cerebral arterioles preparation and pecam-1 expres-
channels. Circ Res. 2005;97:1270–1279. sion in c57bl/6j and apoe-/- mice. Front Biosci (Landmark Ed).
110. Hartmannsgruber V, Heyken WT, Kacik M, et al. Arterial response 2011;16:2367–2371.
to shear stress critically depends on endothelial trpv4 expression. 134. Badaut J, Copin JC, Fukuda AM, Gasche Y, Schaller K, da Silva
PloS One. 2007;2:e827. RF. Increase of arginase activity in old apolipoprotein-e deficient
111. Marrelli SP, O’Neil RG, Brown RC, Bryan Jr RM. Pla2 and trpv4 mice under western diet associated with changes in neurovascu-
channels regulate endothelial calcium in cerebral arteries. Am J lar unit. J Neuroinflammation. 2012;9:132.
Physiol Heart Circ Physiol. 2007;292:H1390–H1397. 135. Ryoo S, Lemmon CA, Soucy KG, et al. Oxidized low-den-
112. Inoue R, Jensen LJ, Shi J, et al. Transient receptor potential sity lipoprotein-dependent endothelial arginase ii activa-
channels in cardiovascular function and disease. Circ Res. tion contributes to impaired nitric oxide signaling. Circ Res.
2006;99:119–131. 2006;99:951–960.
113. Earley S. Trpm4 channels in smooth muscle function. Pflugers 136. Bugnicourt JM, Da Silveira C, Bengrine A, et al. Chronic renal
Arch. 2013;465:1223–1231. failure alters endothelial function in cerebral circulation in mice.
114. Earley S, Waldron BJ, Brayden JE. Critical role for transient recep- Am J Physiol Heart Circ Physiol. 2011;301:H1143–H1152.
tor potential channel trpm4 in myogenic constriction of cerebral 137. Iadecola C, Davisson RL. Hypertension and cerebrovascular dys-
arteries. Circ Res. 2004;95:922–929. function. Cell Metab. 2008;7:476–484.
10.e4 References

138. Kitazono T, Heistad DD, Faraci FM. Atp-sensitive potassium 156. Vital SA, Terao S, Nagai M, Granger DN. Mechanisms underlying
channels in the basilar artery during chronic hypertension. the cerebral microvascular responses to angiotensin ii-induced
Hypertension. 1993;22:677–681. hypertension. Microcirculation. 2010;17:641–649.
139. Kitazono T, Heistad DD, Faraci FM. Enhanced responses of 157. Zhang M, Mao Y, Ramirez SH, Tuma RF, Chabrashvili T. Angio-
the basilar artery to activation of endothelin-b receptors in tensin ii induced cerebral microvascular inflammation and
stroke-prone spontaneously hypertensive rats. Hypertension. increased blood-brain barrier permeability via oxidative stress.
1995;25:490–494. Neuroscience. 2010;171:852–858.
140. Mayhan WG. Impairment of endothelium-dependent dilatation 158. Rodrigues SF, Vital SA, Granger DN. Mild hypercholesterol-
of basilar artery during chronic hypertension. Am J Physiol Heart emia blunts the proinflammatory and prothrombotic effects of
Circ Physiol. 1990;259:H1455–H1462. hypertension on the cerebral microcirculation. J Cereb Blood Flow
141. Sobey CG, Moffatt JD, Cocks TM. Evidence for selective Metab. 2013;33:483–489.
effects of chronic hypertension on cerebral artery vasodi- 159. Rodrigues SF, Granger DN. Cerebral microvascular inflam-
latation to protease-activated receptor-2 activation. Stroke. mation in doca salt-induced hypertension: role of angioten-
1999;30:1933–1941. sin ii and mitochondrial superoxide. J Cereb Blood Flow Metab.
142. Kazama K, Anrather J, Zhou P, et al. Angiotensin ii impairs neu- 2012;32:368–375.
rovascular coupling in neocortex through nadph oxidase-derived 160. Ando H, Zhou J, Macova M, Imboden H, Saavedra JM. Angio-
radicals. Circ Res. 2004;95:1019–1026. tensin ii at1 receptor blockade reverses pathological hypertrophy
143. Girouard H, Park L, Anrather J, Zhou P, Iadecola C. Cerebrovas- and inflammation in brain microvessels of spontaneously hyper-
cular nitrosative stress mediates neurovascular and endothelial tensive rats. Stroke. 2004;35:1726–1731.
dysfunction induced by angiotensin ii. Arterioscler Thromb Vasc 161. Benicky J, Sanchez-Lemus E, Honda M, et al. Angiotensin ii at1
Biol. 2007;27:303–309. receptor blockade ameliorates brain inflammation. Neuropsycho-
144. Capone C, Faraco G, Anrather J, Zhou P, Iadecola C. Cyclooxy- pharmacology. 2011;36:857–870.
genase 1-derived prostaglandin e2 and ep1 receptors are required 162. Faraco G, Sugiyama Y, Lane D, et al. Perivascular macrophages
for the cerebrovascular dysfunction induced by angiotensin ii. mediate the neurovascular and cognitive dysfunction associated
Hypertension. 2010;55:911–917. with hypertension. J Clin Invest. 2016;126:4674–4689.
145. Capone C, Anrather J, Milner TA, Iadecola C. Estrous cycle 163. Liu Y, Hudetz AG, Knaus HG, Rusch NJ. Increased expression
dependent neurovascular dysfunction induced by angiotensin ii of ca2+-sensitive k+ channels in the cerebral microcirculation
in the mouse neocortex. Hypertension. 2009;54:302–307. of genetically hypertensive rats: evidence for their protection
146. Girouard H, Park L, Anrather J, Zhou P, Iadecola C. Angiotensin against cerebral vasospasm. Circ Res. 1998;82:729–737.
ii attenuates endothelium-dependent responses in the cerebral 164. Kamouchi M, Kitazono T, Nagao T, Fujishima M, Ibayashi S.
microcirculation through nox-2-derived radicals. Arterioscler Role of ca(2+)-activated k+ channels in the regulation of basilar
Thromb Vasc Biol. 2006;26:826–832. arterial tone in spontaneously hypertensive rats. Clin Exp Phar-
147. Didion SP, Faraci FM. Angiotensin ii produces superoxide-medi- macol Physiol. 2002;29:575–581.
ated impairment of endothelial function in cerebral arterioles. 165. Amberg GC, Bonev AD, Rossow CF, Nelson MT, Santana LF.
Stroke. 2003;34:2038–2042. Modulation of the molecular composition of large conductance,
148. Kitayama J, Chu Y, Faraci FM, Heistad DD. Modulation of dilator ca(2+) activated k(+) channels in vascular smooth muscle during
responses of cerebral arterioles by extracellular superoxide dis- hypertension. J Clin Invest. 2003;112:717–724.
mutase. Stroke. 2006;37:2802–2806. 166. Nieves-Cintron M, Amberg GC, Nichols CB, Molkentin JD, San-
149. Chrissobolis S, Banfi B, Sobey CG, Faraci FM. Role of nox iso- tana LF. Activation of nfatc3 down-regulates the beta1 subunit
forms in angiotensin ii-induced oxidative stress and endothelial of large conductance, calcium-activated k+ channels in arterial
dysfunction in brain. J Appl Physiol. 2012;113:184–191. smooth muscle and contributes to hypertension. J Biol Chem.
150. Chrissobolis S, Faraci FM. Sex differences in protection against 2007;282:3231–3240.
angiotensin ii-induced endothelial dysfunction by manganese 167. Kirsch T, Wellner M, Luft FC, Haller H, Lippoldt A. Altered gene
superoxide dismutase in the cerebral circulation. Hypertension. expression in cerebral capillaries of stroke-prone spontaneously
2010;55:905–910. hypertensive rats. Brain Res. 2001;910:106–115.
151. Capone C, Faraco G, Park L, Cao X, Davisson RL, Iadecola C. 168. Tobin AA, Joseph BK, Al-Kindi HN, et al. Loss of cerebrovascular
The cerebrovascular dysfunction induced by slow pressor doses shaker-type k(+) channels: a shared vasodilator defect of genetic
of angiotensin ii precedes the development of hypertension. Am and renal hypertensive rats. Am J Physiol Heart Circ Physiol.
J Physiol Heart Circ Physiol. 2011;300:H397–H407. 2009;297:H293–H303.
152. Capone C, Faraco G, Park L, Cao X, Davisson RL, Iadecola C. 169. Wellman GC, Cartin L, Eckman DM, et al. Membrane depo-
The cerebrovascular dysfunction induced by slow pressor doses larization, elevated ca2+ entry, and gene expression in cerebral
of angiotensin ii precedes the development of hypertension. Am arteries of hypertensive rats. Am J Physiol Heart Circ Physiol.
J Physiol Heart Circ Physiol. 2011;300:H397–H407. 2001;281:H2559–H2567.
153. Diaz-Otero JM, Yen TC, Fisher C, Bota D, Jackson WF, Dorrance 170. Xie MJ, Zhang LF, Ma J, Cheng HW. Functional alterations in
AM. Mineralocorticoid receptor antagonism improves parenchy- cerebrovascular k(+) and ca(2+) channels are comparable
mal arteriole dilation via a trpv4-dependent mechanism and pre- between simulated microgravity rat and shr. Am J Physiol Heart
vents cognitive dysfunction in hypertension. Am J Physiol Heart Circ Physiol. 2005;289:H1265–H1276.
Circ Physiol. 2018;315:H1304–H1315. 171. McCarron JG, Halpern W. Impaired potassium-induced dilata-
154. Faraci FM, Lamping KG, Modrick ML, Ryan MJ, Sigmund tion in hypertensive rat cerebral arteries does not reflect altered
CD, Didion SP. Cerebral vascular effects of angiotensin ii: na+, k+-atpase dilation. Circ Res. 1990;67:1035–1039.
new insights from genetic models. J Cereb Blood Flow Metab. 172. Jarajapu YP, Knot HJ. Relative contribution of rho-kinase and
2006;26:449–455. pkc to myogenic tone in rat cerebral arteries in hypertension. Am
155. Ishikawa M, Sekizuka E, Yamaguchi N, et al. Angiotensin ii type 1 J Physiol Heart Circ Physiol. 2005;289:H1917–H1922.
receptor signaling contributes to platelet-leukocyte-endothelial 173. Faraco G, Moraga A, Moore J, Anrather J, Pickel VM, Iadecola C.
cell interactions in the cerebral microvasculature. Am J Physiol Circulating endothelin-1 alters critical mechanisms regulating
Heart Circ Physiol. 2007;292:H2306–H2315. cerebral microcirculation. Hypertension. 2013;62:759–766.
2 Mechanisms of Thrombosis and Thrombolysis
Gregory J. del Zoppo

KEY POINTS Notably, all substances that promote plasmin formation have
the potential to increase the risk of hemorrhage.
• T he fundamental processes involved in thrombus The acute use of PAs has been associated with detectable
formation, thrombus dissolution, and thrombus clinical improvement in selected patients with symptoms
stability and their relevance to the central nervous of focal cerebral ischemia.1-9 Acute thrombolysis has thus
system (CNS) are described. attained pride of place in the treatment of ischemic stroke
so far. Currently, recombinant tissue plasminogen activator
• The role(s) of endogenous plasminogen activators (rt-PA) is licensed in the United States, Japan, Europe, and
(PAs, including tissue-type plasminogen activator, many other countries for the treatment of ischemic stroke
urokinase-type plasminogen activator) in within 3 hours of symptom onset, and up to 4.5 hours in
thrombus dissolution are presented, together with some jurisdictions.6,9 Early studies, a phase III prospective
considerations of their regulation in vivo. Their trial, and more recent experience suggest that extension of the
relevance to derived therapeutics is emphasized. treatment window is possible with strict limitations to patient
• Fibrinolytic agents tested or used as pharmaceuticals selection.3-5,9 Early on, few studies of acute rt-PA delivery
including recombinant and purified endogenous correlated improvement in patient outcome with imaging
PAs and exogenous PAs (including streptokinase, evidence of recanalization of an occluded brain-supplying
staphylokinase, PAs derived from Desmodus species, artery, however.4
and novel plasminogen activators) are presented. The development of agents that promote fibrin degradation
• The molecular basis for PA inhibition and modulation in the clinical setting stems from observations in the 19th
of vascular fibrinolysis is made. century of the spontaneous liquefaction of clotted blood and
the dissolution of fibrin thrombi. A growing understanding of
• These considerations form a basis for exploration of plasma proteolytic digestion of fibrin paralleled enquiry into
current information about the impact of PAs and of the mechanisms of streptococcal fibrinolysis. Streptokinase
plasmin generation on CNS vessel and microvessel (SK) was the first PA employed to dissolve closed space
integrity. (intrapleural) fibrin clots, but purified preparations were
• Exploration of the role(s) of endogenous PAs in required for lysis of intravascular thrombi. The development
CNS development, CNS integrity, and on neuronal of PAs for therapeutic lysis of vascular thrombi has progressed
function in the CNS is presented, and the potential along with insights into the mechanisms of thrombus
effects of therapeutic PAs on the CNS. formation and degradation. It should be remembered that
• The pioneering use of therapeutic plasminogen the concentrations of PAs used to degrade fibrin thrombi
activation in the acute setting in ischemic/thrombotic clinically far exceed those required to perform the same task
stroke, acute cerebral arterial recanalization, and its endogenously.
consequences are described.
• The use of PAs in experimental cerebral ischemia, THROMBUS FORMATION
recanalization and tissue injury reduction, and their
The relative platelet-fibrin composition of a specific thrombus
limitations and relevance to the clinical setting are
depends on the vascular bed, the local development of fibrin,
discussed.
platelet activation, and regional blood flow or shear stress.
• The risks of PAs in the acute intervention in ischemic Even in the same arterial territory there may be considerable
stroke and the quantitative effects on intracerebral variability and local heterogeneity in thrombus composition
hemorrhage are presented. Limitations to the clinical use as evidenced by thrombi removed in situ.10-13 Pharmacologic
of fibrinolytic agents in ischemic stroke are considered. inhibition of the platelet activation/aggregation and coagula-
tion processes can also alter thrombus composition and vol-
ume. At arterial flow rates thrombi are predominantly platelet
rich, whereas at lower shear rates characteristic of venous flow,
Thrombosis, and thrombus growth, dissolution, and migra- activation of coagulation seems to predominate. It has been
tion are inextricably connected. Thrombus formation involves suggested that the efficacy of pharmacologic thrombus lysis
activation of platelets, activation of the coagulation system, depends on (i) the relative fibrin content and (ii) the extent of
and the processes of fibrin dissolution. The central feature of fibrin cross-linking of the thrombus that may reflect thrombus
each of these processes is the generation of thrombin from age and thrombus remodeling. The latter may vary with loca-
prothrombin. Thrombin, in turn, generates the thrombus tion within a vascular bed (e.g., arterial, capillary, or venular).
fibrin network by the cleavage of circulating fibrinogen with Thrombin (factor IIa) is the central player in clot formation
formation of the fibrin network. Excess local vascular fibrin (Fig. 2.1). Thrombin, a serine protease, cleaves fibrinogen to
deposition can contribute to thrombus growth, while vascular generate fibrin, which forms the scaffolding for the growing
injury and excess degradation of fibrin in “hemostatic plugs” thrombus. Inter-fibrin strand cross-linking requires active
at sites of vascular injury can lead to hemorrhage. Plasmin can factor XIII, a transglutaminase bound to fibrinogen that
degrade fibrin and fibrinogen. Plasminogen activators (PAs), is itself activated by thrombin. Factor XIIIa stabilizes the
which convert plasminogen to plasmin, have been exploited fibrin network (Fig. 2.2).14,15 Thrombin-mediated fibrin
to dissolve clinically significant vascular thrombi acutely. polymerization leads to the generation of fibrin I and fibrin

11
EXTRINSIC PATHWAY INTRINSIC PATHWAY

HMWK
TF
VII TF: VIIa XI XII Pre-kallikrein
Ca2
X IX
Kallikrein

Ca2 XIIa
Fibrin
Fibrinogen
XIa
IIa

IXa VIII
Va Xa  VIIIa
II 
PF 1.2
Ca2 Ca 2

EXTRINSIC PATHWAY INTRINSIC PATHWAY

HMWK
TF
VII TF: VIIa XI XII Pre-kallikrein
Ca2
X IX
Kallikrein

Ca2 XIIa
Fibrin Fibrinogen
XIa
IIa

Xa IXa  VIIIa VIII


Va
II 
PF 1.2
Ca2 Ca 2

EXTRINSIC PATHWAY INTRINSIC PATHWAY

HMWK
TF
XI XII Pre-kallikrein
VII TF: VIIa
Ca2
X IX
Kallikrein

Ca2 XIIa Protein C


Fibrin Fibrinogen Thrombomodulin
XIa
IIa
AT APC
HC-III IXa VIII Protein S
Va Xa  VIIIa
II 
PF 1.2
Ca2 Ca 2

C
Fig. 2.1. Intrinsic and extrinsic coagulation pathways (see text). Phospholipid-containing membranes (e.g., platelets) provide the scaffold for
accelerating coagulation pathway activation. Both intrinsic and extrinsic pathways lead to prothrombin (factor II) activation, with fibrin generation
from circulating fibrinogen. The extrinsic pathway initiates coagulation through the interaction of factor VII with tissue factor (TF) in the vascular
adventitia, brain perivascular parenchyma, and activated monocytes. The TF:VIIa complex catalyzes activation of factor X and acceleration
of thrombin generation. The intrinsic system involves activation of components within the vascular lumen. Initiation of coagulation through
this pathway involves pre-kallikrein, kallikrein, high-molecular-weight kininogen (HMWK), and factors XI and XII. (A) Thrombin generation. The
intrinsic system activates factor X through the “tenase” complex (factors VIIIa and IXa, and Ca2+ on phospholipid). Both intrinsic and extrinsic
pathways activate prothrombin through the common “prothrombinase” complex (factors Xa and Va, and Ca2+). The platelet surface has
receptors for factors Va and VIIIa. Cleavage of prothrombin generates the prothrombin fragment 1.2 (PF 1.2) and thrombin (factor IIa). (B)
Thrombin has multiple stimulatory positive feedback effects. It catalyzes activation of factors XI and VIII as well as the activities of the tenase
and prothrombinase complexes. Thrombin also stimulates activation of platelets and granule secretion via specific thrombin receptors on their
surface. (C) Coagulation activation is regulated by interleaving inhibitor pathways. The effects of factors Va, Xa, and VIIIa are modulated by the
protein C pathway. Activated protein C (APC), generated by the action of the endothelial cell receptor thrombomodulin on protein C, with its
cofactor protein S, inhibits the action of factor V. AT, Antithrombin; HC-III, heparin cofactor-III.
Another random document with
no related content on Scribd:
this is done he waits until the [667]loaa mua iaia ka uala i ka wa
stormy months are over, and i ka lala, hoi aku oia me ka
long after that, in the month of wahie a ho-a i ka imu a kalua a
Kaaona, he goes to throw up the moa. Kahea aku oia penei:
earth on the hills of the potatoes; “Keaonui i ka maka o ka opua
when he first obtains potato, ilalo iho, e hoi e ai i ka ai.” A hala
whether in between hills or from ka malama o Welehu, o Makalii,
the running vine, he returns with a o Hinaiaeleele, a komo i ka
wood, lights the fire in the imu malama o Nana i ka la o Mauli
and cooks the potato until it is hoonana kane, hoonana ka
done. He prays in this manner: wahine, kau na pualei i ke poo,
“Keaonui, hanging below the eye hele e eli i ka uala a ike na kane
of the narrow pointed clouds, ame na wahine. Penei ka olelo a
come to partake of the food.” ka mea palaualelo: “E! eia ka’u
When the months of Welehu, 247 pue uala,” a “eia no hoi kau,” a
Makalii 248 and Hinaiaeleele are papa aku kekahi i kekahi; “mai
past, and during the month of kii mai oe i ka’u” o ka eli no ia a
Nana on the day called Mauli, he loaa, ka uala. Aohe nae he nunui
shows off before men and he makalii wale no; hookahi mea
women, and placing a wreath of e ola ai o ka ohana o ka malama
flowers on his head, he would i ka palula, oia ka ai e ola ai ke
sally forth to dig potatoes in the ole ka hua. A olelo ae kekahi
presence of these men and penei: “Aole no keia o na
women. Then the improvidents malama e hua nui ai ka uala; he
would call out, “Say, this is my malama ulu palula wale no keia,
potato hill;” [and another] “And he aa noi ke kumu. Eia ka
this is mine;” they would forbid malama e kanu ai i hua.” I ka la
each other by saying, “Don’t you o Hilo kii aku ka mahiai i na lau;
come to get mine.” They would eia ka inoa: kola, nika,
then dig and obtain potatoes. But pukeleawe, hiiaka, lapa,
they are not of large size; they huamoa; waiho aku e like me ka
are small. There is one way of mea i hoike ia. Aia hiki mai ka la
keeping the family in food, [that o Hoaka oia ka wa e kanu ai, e
is] to care for the leaves [of the kanu e like me ka mea i hoike ia
potato]; that is the food with maluna, a hala o Ikiiki pau ke
which to feed the family if there kaumaha o ke kanaka mahiai,
be no tubers. One would say manao ae oia o ke ola o ka
thus: “Anyway, these are not the ohana; a i ka la o Olekukahi,
months when potatoes bear Olekulua, Olekupau, oia na la e
plentifully; these are months pue ai i ka uala; noho aku oia a
when leaves grow rank and the hala napoo Mohalu, o Hua, o
stalks swell large. This month is Akua, o Hoku, o Mahealani, o
the time to plant in order to Kulu, a i ka la hope o Akua hele
bear.” On the day called Hilo the oia ma kona mala i ka wa e puka
planter obtains some stalks, the mai ai na kao, oia hoi ka lalani, a
ones called kola, nika, ku oia ma ke kihi o ka mala,
pukeleawe, hiiaka, lapa, and penei oia e kahea ai: “E
huamoa. 249 Prepare them in the Kanepuaa, eku i uka, eku i kai,
manner previously stated. When eku i nae, eku i lalo, eku iwaena
the day called Hoaka arrives o ka kaua mala uala nei la, e
then is the time to plant; set Kanepuaa, eku oe mai kela kihi
them out in the manner a i keia kihi, mai kela kaika a i
explained above; and when Ikiiki keia kaika, mai kela iwi a i keia
is past the planter would be iwi, i hua i ka mole, i hua i ke
relieved, for he realizes that his kano, i hua i ke aakolo i ka wa.”
family is saved; the days called A pau kana pule ana, noho aku
Olekukahi, Olekulua and oia a hala Kaloakukahi,
Olekupau are the days during Kaloakulua, Kaloapau, a i ka la o
which to hill up potatoes; then he Kane, hoomakaukau ke kane
waits until the days called ame ka wahine i mau koko no ka
Mohalu, Hua, Akua, Hoku, uala, a i ka la o Lono, hoomaka
Mahealani and Kulu, and the last ka eli ana o ka uala. Ekolu no
day, Akua, he visits the potato uala o ka pue, nunui nohoi ka
field at the time that the uala. Ua hai mai kekahi
constellation called Taurus rises; elemakule i ke ana, a ua ana
arriving at the corner of his field, wau ekolu kapuai ke anapuni,
he prays in this manner: “O olioli wale ae e mahiai uala i ka
Kanepuaa, 250 root towards the nunui o ka hua: Hoi mai ke kane
mountain, root towards the sea; ame ka wahine, kalua ka puaa
root towards the wind, root me ka uala.
towards the calm, root in the
middle of this our potato field! O
Kanepuaa! do thou root from that
corner to this corner, from that
border to this border, from that
side to this side, so fruit would
appear at the end of the stalk,
along the stalk, and the roots
which creep between hills.”

After he has finished this prayer [aole i pau.] [669]


he waits until Kaloakukahi,
Kaloakulua, and Kaloapau are
past, and on the day called
Kane, the husband and the wife
prepare ko-ko (nets) for the
potatoes; on the day called
Lono, prepare for digging the
potatoes. There are only three
potatoes in a hill; large indeed.
(An old man showed me the
measure, and when I measured
it, it was three feet in
circumference; it made me feel
like cultivating potatoes on
account of those big tubers.) The
husband and wife then return
and cook a pig with the potatoes.
[unfinished.] [668]

CONCERNING THE TI- NO KA LA-I.


LEAF.

The ti-leaf is one of the plants O ka la-i oia no kekahi o na laau


growing in the mountains. It e ulu ana ma kuahiwi. Ua ulu oia
grows in valleys, on hills, ma na awawa, na puu, na
mountain ridges and side-hills. kualapa, na kipapali. O keia laau
This ti-leaf plant grows tall, but it o ka la-i he loloa no, aohe nae
is not large round like other ona nunui e like me kekahi mau
trees. I think the ti-leaf grows on laau e ae. Ua ulu no paha ka la-i
all inhabited islands, but I am not ma na mokupuni i noho ia e
sure. Where it originated I do not kanaka, aole i maopopo loa ia’u.
know; it may have been brought
from some place, or it may have O kahi i loaa mai ai ka la-i aole i
been indigenous. Let us consider loaa ia’u; he loaa mai paha mai
the uses of the ti-leaf. iloko mai o kahi mea, a i ole he
mea ulu wale mai no. E nana
kakou i na hana a ka la-i.

the uses of its leaf. na hana a kona lau.

The leaf of the ti is something O ka lau la-i, he mea no ia e


that alleviates the trouble of hoopau ai i ka pilikia o ke
man, in that it is used in the kanaka, oia hoi ma ka hana ia
construction of houses to be ana i hale i wahi e noho ai ke
occupied by man; such a house kanaka, a kapaia ia hale, “hale
is called hale la-i 251 (ti-leaf la-i.” E like me ke ’lii i kukulu ai i
house). Just like a chief who built hale la-i nona ma Puulaina, a
a ti-leaf house for himself at kapaia ia puu ia manawa o
Puulaina and because of the fact Puulai, e like me ka mea a
that la-i was used, the hill was kakou i lohe ai mamua iho nei.
called Puula-i, 252 as we heard He kapa no hoi ia no kekahi poe
heretofore. It is also used by ma ka haku ia ana a paa, alaila
some people for wearing aahu.
[apparel] by braiding it, and,
when done, wearing it.

It is used for lau 253 fishing; when He mea lau lawaia ke hili ia a
braided long it is used to frighten loihi, nana e hoa mai ka ia iloko
and drive the fish into the net. It o ka upena. He mea kauwewe
is used for covering the imu to imu i mea e paa ai ka mahu a
retain the steam and thus cook moa ka ai. He mea pulehu i’a
the food. It is used for covering nohoi. He mea pai ai. He moku
for fish to be roasted on coals. It nohoi ia na kekahi poe i ka wa
is used for paiai 254 covers. It was kahiko. He pau hula ke haku ia a
also used as a boat by some paa, na ka poe hula. He mea
people in the olden time. It is ahaaina nohoi. He pulumi
braided into hula skirts and used moena i pau ai ka lepo. A o ka
by the dancers. It is used at iwi owaena o ka lau, he mea
feasts. It is used as a broom to hana papale ia. O ka lau nohoi
clean the dirt from the mat. The he mea hana ia i pu-la-i i mea
midrib is used for braiding into hookani ma ke puhi ana aku
hats. The leaf is made into pula-i nohoi.
(lai whistle) which gives forth
sound when blown upon.

By the use of the pula-i were the Ma ka pu la-i nohoi i lilo ai na


sisters of Aiwohikupua kaikuahine o Aiwohikupua i poe
befriended by Laieikawai, and aikane na Laieikawai, a noho
they lived together happily. It like lakou me ka oluolu. Pela
was the case when Malio, the nohoi o Malio ke kaikuahine o
sister of Halaaniani, went to get Halaaniani i kii ai ia Laielohelohe
Laielohelohe the younger sister me ka pu la-i, ke kaikaina o
of Laieikawai. She used the Laieikawai, i ka wa i makemake
pula-i when Halaaniani wished to ai o Halaaniani e moe ia
get Laielohelohe for wife; they Laielohelohe, aole nae i loaa ia
did not succeed in their desire, laua.
however.

The leaf of the ti is also used to O ka lau no hoi o ka la-i, he mea


drive away illness from a sick kuehu i ka mai, i pau ka mai i ka
person. If the sickness be [in the wa kahiko. Ina he puupuu ka
nature of] sores, then ti-leaf is mai, alaila, kii i ka la-i, i mea
obtained and placed on the hoomoe pu, i ole e pipili ka
body, so the sores would not puupuu i ka moena a eha, ina he
touch the mat and cause pain; if lau la-i, aole e pipili, a he mau
ti-leaf [is used] the sores do not nohoi ka ma-u, aole e wela ka ili.
adhere; they are cooling; the A he mea lei ia no e kekahi poe
skin is not heated. It is worn on ma ka a-i, i ole e loaa i ka mai,
the neck by some people to na ka la-i e pale aku i ka mai a
prevent illness; the ti-leaf wards pakele.
off and safeguards from
sickness.

the stalk of the ti-leaf o ke kumu o ka la-i.


plant.

The stalk is used as a spear for He auhau nohoi ia, he mea kao-
fire-brand 255 to be thrown from a ahi ke maloo. A o kona wahi
height when dry. The plant is nohoi ia e ulu ai o ka auki, ina
propagated from this part; if a la-i makemake i pa la-i, alaila, kii, a
fence is desired, secure some, kanu a puni, a mahope ulu. Ka
plant them around and after a [671]mea hoi i olelo ia no ka auki.
while they will grow. [670] Aia ma Waipio he muliwai, a he
mano aikanaka ko laila, penei: I
[Here is] a story concerning its ka wa e hele aku ai a hiki ilaila,
wood: At Waipio is a stream aohe uapo ia wa, ku iho ma kae
wherein lived a man-eating o ka muliwai, a kiloi aku i ka
shark; when one came to the auki; a i nalowale koke ka auki,
place (there were no bridges alaila hoi, he mano o loko, aohe i
then) he stood on the bank of the hele, ina e lele pau loa, aka, i
stream and threw in a stalk of ti- kiloi a aohe nalowale iki o ka
plant. If it disappeared quickly, auki, alaila, aohe mano, ua hele i
go back, the shark was there, kahi i hele ai, lele ino a au
and was not gone; to jump in wikiwiki, nokamea, aole akea loa
was to be eaten. But, if when ka; ina e au lohi loaa koke mai
thrown in, the stalk did not no i ka mano ke ahikanana o ka
disappear, then there was no moana, a loaa kaiala mea ai,
shark; it had gone to another kani kaiala aka, ua loaa iaiala.
place; then jump in and swim
across quickly, because the
stream is not wide; but if you
tarried or were slow in swimming
you would be caught by the
shark, the champion of the
ocean, and he would secure a
morsel of food, and he would
laugh for he had obtained
something.

the root: that is, the o ka mole, oia hoi kahi


part under the soil. malalo o ka lepo.
The root was used a great deal O ka mole, he mea hana nui ia
by the Hawaiians for cooking in ia e na kanaka, a kalua i ka imu.
the imu (underground oven). Elua, a ekolu la, alaila moa, lawe
After two or three days it would ia aela no hoi i mea ai, i ka wa
be cooked, then it was used as wi, a i ka wa wi ole, aka, i ka wa
food during times of plenty and wi ka hana nui ia o keia mea. A i
during famine; but it was used ole he mea ai wale no, alaila,
mostly during times of famine. hana ia i mea ona, oia hoi ka
Again it was used for simply okolehao, i mea inu, na na kane,
eating; and then again it was na wahine, na keiki; a oia kekahi
made into a liquor called mea e wi ai o ka lilo ma ia mau
okolehao, 256 a drink for men, hana lealea. A o ua mole nei no,
women and children; that was he aila hoopaa lauoho ia, i ole e
one cause of famine when time puehu i ka makani, a ua kapa ia
was taken up with those kinds of hoi ua mole la-i nei he ki.
pleasure. This same root was
used for oil to keep the hair
together so that it would not be
blown by the wind. It is called ki.

This is all I have obtained by O ia wale iho la no kahi mea i


asking questions. The leaf is the loaa ia’u me ko’u ninaninau ana
most useful part of this plant. aku no hoi; ma ka lau nae kahi
ka hana nui loa ia o keia laau.
John Mana.
John Mana.

THE KUKUI TREE. NO KE KUKUI.


This tree, the kukui, 257 we are O keia laau o ke kukui, ua ike
familiar with; we know its kind kakou a ua hoomaopopo i kona
and what it looks like, and that is, ano, a me kona helehelena a
it has a stocky, stout trunk with pau, oia hoi, he puipui kona kino,
many branches, and green a he nui kona mau lala, a he
leaves. This tree commonly uliuli kona mau lau. O keia laau
grows on the mountains, in the no hoi ua ulu mau ia ma na
forests and hill tops. It grows mauna, ma kuahiwi, ame na
also on all of the islands; but kualono; o keia laau nohoi, ua
there is no place where it came ulu ia ma na mokupuni apau,
from; it is indigenous to Hawaii aka, o kahi nae i loaa mai ai o
nei. Here are the uses of the keia laau aohe wahi i loaa mai
kukui: ai, he laau kahiko no ia no
Hawaii nei. Eia na hana a ke
kukui.

the uses of the na hana a ka lau.


leaves. 258

They are used for hastening the He mea hoopala maia ia i kekahi
ripening of bananas, and are manawa; he mea lauwalu ia ia i
also used in the roasting of fish; kekahi manawa, oia iho la na
those are the uses of the leaves hana a ka lau i loaa ia’u.
which I know about.

concerning the nut. no ka hua.

The nut is edible, after being He mea ai ia ka hua, ma ka


roasted and the kernel pounded pulehu ana a moa, lomi pu me
with salt. 259 It is used in the place ka paakai. A he ia ia no ka noho
of fish when sojourning in the ana i kuahiwi, a me ka la maka
mountain, and the day when the pehu loa no hoi i ka ia. O kekahi,
eye [672]bulges out for [want of] [673]he laau keia i hamo ai i ke
fish. This is also used as kino, i na he puupuu ka mai,
medicine for rubbing on the ame ka piele. He kukui no hoi
body, if the disease be sores on keia no ka manawa kahiko.
the body or on the head. This Mamuli o ke kike ia ana a pau ka
was also used for lamp light in iwi, koe iho ka io, kui ia alawa ke
the olden times. It is prepared by koi me ka niau niu nae e kui ia
cracking the shell, leaving the ai. Ina he hale ahaaina, i ka po,
kernel, which is strung together ua ike ia o keia kukui ka mea
on the midrib of a coconut leaf hana nui ia. Mamuli o ke kui ana
(segment). If a feast were held at a lawa na koi eha a elima paha,
night in a house this kind of lamp huihui ia a kahi hookahi owili iho
was the light mostly used. It is i ka lau o ka la-i mawaho, i ole e
prepared by stringing four or five pau koke. o kekahi, he hana aila
sticks, when they are bunched ia, mamuli nae o ke kalua ia ana
together and wrapped in ti- a moa, kike ia nohoi ka iwi apau,
leaves so that they would not lawe ia a luna o kekahi papa
burn out quickly. It is also made pohaku a i ole he papa laau
into oil by cooking it. When done paha, lu ia iluna olaila, kau iho i
the shells are cracked and the laau nui maluna; me ia laau e
kernel taken and thrown on a olokaa ai io ia nei a o ka wai, oia
stone or wooden board and a no ka aila.
large block of wood is placed on
top. This block is rolled all over
[the kernels] and the juice
[obtained therefrom] is the oil. 260

This is also used to obtain the O kekahi he mea keia e loaa ai o


hamauleo 261 fish of Ewa; the ka ia hamauleo o Ewa; oia hoi,
shell is broken off, leaving only ke kike ia ana a pau ka iwi, koe
the kernel; this is placed in the iho ka io, hookomo iloko o ke
clothes, or else in the container eke o ka lole, a i ole iloko no hoi
where said fish would be placed o ka ipu kahi e hookomo ia iho ai
when caught, or in any other ua ia ala, a i kekahi mea e ae
thing. Proceed to where that fish paha. Hele no a hiki i kahi o ua
is generally caught, chew some ia ala, naunau iho i ua kukui ala,
of this kukui kernel, and blow it a puhi aku iloko o ke kai, a
on the sea; the sea would be malino aela ke kai, a ike ia i hola
smooth and oily, and that fish ua ia ala. Pela no ka lawaia ana i
could be seen. So it is done ka hee, elike me ka lawaia ana o
when fishing for squid; like ka ia hamauleo. O kekahi, he
catching clams. It is also used mea lamalama ia no hoi i ka po,
for torches at night, when fishing ke hele nae i ka lawaia puhi. O
for eels. These are the uses of ia iho la na hana a ka hua i loaa
the nut which I know about. ia’u.

concerning the smoke no ka uahi o keia hua.


from this nut.

The soot from the smoke was O ka uahi he mea hana ia i mea
used in tatuing on the arms. kakau i ka lima, oia hoi ka pa’u.
Here is the method of Eia ke kumu i loaa ai; kui ia
preparation: It is strung on sticks nohoi alawa ke koi, hoa ia no hoi
until each is full; one is lighted, a a, lawe ia a malalo o kekahi
and is taken and placed in a pohaku i eli ia a poopoo,
hollowed stone in such a way hookupono ia ka uahi a kupono
that the smoke would fill the iloko o ua pohaku ala, a mahope
hollow in said stone; after a while manoanoa ae kela uahi i pili mau
the soot would adhere to the i ka pohaku, a kii aku ohikihiki a
stone, when it is dug out and loko o kekahi wahi mea kupono
placed in a container prepared no ia mea; hana pu me ka wai
for it; it is mixed with sugar-cane ko, a lilo ae i mea kakau i ka
juice, and then used for tatuing lima. O ia iho la na hana o ka
hua i loaa ia’u.
the arms. Those are the uses of
the nut which I have obtained.

concerning the shell. no ka iwi.

That is, the part immediately Oia hoi ka mea mawaho ae o ka


outside of the kernel. This thing io. O keia mea he mea
was greatly valued by the old makemake nui ia keia e ka poe
men and the old women in the elemakule ame ka poe luahine,
olden times. When matches had aia i ka manawa kahiko. I ka
not come into use here in manawa aole i laha mai keia ahi,
Hawaii, that is, the sulphur oia hoi ke kukaepele, hana nui
match, the old men and women na elemakule ame na luahine
used to crack the nuts open, me ke kike ia nae a pau ka io
take out the kernel, leaving only koe iho ka iwi, houluulu a ma
the shells, gather them by the kapuahi, hoa i ke ahi a he loihi
fireside and burn them; it burns ka a ana oia mea, aole e pau
for a long time; it does not koke, o ia iho la ka hana a ka iwi
disappear quickly. That is all the i loaa ia’u.
use of the shell 262 which I have
obtained.

concerning the bark. no ka ili.

That is, the part enveloping the Oia ka mea mawaho ae o ke


trunk. This thing was used a kumu. O keia mea he mea hana
great deal by the canoe builders. nui ia keia e ka poe kalai waa,
It was gathered, prepared and mamuli o ka lawe ana, a hana, a
pounded, and its juice was kui a o ka wai, oia no ka mea
mixed with the ashes of the hana pu ia me ka lehu o ke
bulrush or cane-tops. 263 It was akaakai a ha-ko paha, he mea
also spread on the blackboard hana ia no hoi i ka papa eleele i
so that the writing would show. It kohu ke kakau aku. He mea
was also used for printing kapa. kakau ia kekahi i ke kapa pa-
[674] upa-u. [675]

concerning the trunk. no ke kino.

This was often used for fences to He mea hana pinepine ia eia e
confine animals and to protect kanaka i mea pa holoholona
plants; it was also used in the ame ka pa mea kanu, a he mea
construction of houses, and as kukulu hale ia nohoi, a he wahie
firewood for imus. hoa imu nohoi.

concerning the gum. no ka pilali.

When we were young a great Aia i ko makou manawa liilii, he


deal of this thing was eaten; mea ai nui loa ia keia, mamuli o
when the parents went up to the ka pii ana o na makua i ka imi
mountains to look for kukui pepeiao, no ka mea, ua kupu
fungus 264 (mushroom) they mai no ia mea no loko mai oia
would also come across gum laau, a loaa aku la ka pilali e hu
oozing from the tree, and all of it ana a o ka hamu ia aku la ia
would be eaten. It was also used apau. O kekahi, he mea hamo
for daubing on the hair; it was lauoho ia, me ka hookomo ia
placed in a bowl or a cup, with nae iloko o ka bola a kiaha paha,
some water added, and left to ukuhi ia i wai, waiho aku a liuliu
stand for a while, and when kii aku, ua lilo ae la ka wai a
taken up the water has become uuluhaku, a he mea maikai loa ia
lumpy; it was very good when i ka hamo lauoho ana, he pahee
applied to the hair; it made it a me ka pakika. Oia iho la na
smooth, shiny and slippery. hana a ke kukui i loaa iau.
Those are the uses of the kukui
which I have been able to obtain.
the famous kukui no na ulu kukui
groves.—grove of kaulana.—ulu kukui o
kaukaweli. 265 kaukaweli.

This grove was so named during O ke kapa ia ana o keia inoa


the time when Mr. Pogue was mamuli no ia o ka manawa e
teacher [at Lahainaluna] and noho kumu ana o Pokue, no ka
because of his sternness with nui o kona huhu i na haumana.
the pupils. The pupils were Ua kau ka weli o na haumana ia
possessed with fear at his ia no kona huhu, a o kekahi no
sternness, and also because ka makau o na haumana i na
they were afraid of human skulls poo kanaka ame kekahi mau
and other things, so they named mea e ae. A ua kapa lakou o ka
it the Kukui Grove of Kaukaweli. ulukukui o Kaukaweli. O keia
This grove of kukui is where wahi ulu kukui, oia kahi e luana
visitors while away the time mau ai o na malihini ke hiki aku i
during commencement days, na la hoike, a he mea maikai ia.
and it was a good thing.

grove of lanikaula. ulu kukui o lanikaula.

This kukui grove is on Molokai; it O keia ulu kukui aia no ia ma


was named in that way on Molokai, o ka mea i kapa ia ai o
account of a prophet of Molokai keia ulu kukui mamuli oia inoa;
named Lanikaula. When he died he kaula no Molokai, oia hoi o
he was taken to and buried at Lanikaula, i ka make ana o ua
this kukui grove, and that is why kaula nei, lawe ia no a ma ua ulu
its name is Ulukukui a kukui aia kanu ia, a oia ka mea i
Lanikaula. 266 kapa ia ai kona inoa, ulu kukui o
Lanikaula.
concerning the grove no ka ulu kukui o
of lilikoi. lilikoi.

This kukui grove grows on this O keia ulu kukui aia no ia ma


island of Maui, at Makawao. It Maui nei kahi i ulu ai, aia nae ma
was famous because the chiefs Makawao. O ka mea i kaulana ai
went there in the olden times, o keia ulu kukui, no ka hele mau
and perhaps even to this day; it o na ’lii ilaila i ka manawa kahiko
was noted as a place often a hiki paha i keia manawa, he
visited by strangers. It was also wahi makaikai nui ia e na
the place where were procured malihini. O kekahi, kahi no ia e
the kukui nuts for the chiefs, for kii ia ai o na hua kukui ai na na
the kernel of its nuts was alii, nokamea, o ko laila kukui he
fragrant and good to the taste. ala ame ka ono. O ia iho la na
This is all that I have learned. mea i loaa ia’u.

John Moo. John Moo.

where the kukui is kahi i loaa mai ai ke


obtained and its uses. kukui ame na hana.

Where it is obtained: The kukui Kahi i loaa mai ai: O ke kukui, he


is a large tree and is good to laau nui no keia a maikai no hoi
look upon; it is obtained from its ke nana aku, ua loaa mai keia
fruit. When the fruit of the kukui laau mai loko mai no o kona
dries and falls off, then some of hua, aia i ka wa e maloo ai o ka
[676]them will grow into a distinct [677]hua o ke kukui, a helelei mai
kukui tree; that is where the luna aku ona, alaila, ulu mai ana
kukui tree which we know now no kekahi hua ona a lilo i kukui
comes from. But the bark of this okoa; a oia kahi i loaa mai ai ke
tree is used for dyeing nets, kukui a kakou e ike nei, aka, aia
printing kapa, and blackening ma ka ili o keia laau ua hana ia i
canoes; [but in the latter case] it mea hooluu upena, a i mea
is mixed with the ashes of sugar- hooluu kapa no kekahi, a i mea
cane leaves; the leaves were paele waa kekahi, he awili ia nae
burnt to produce the ashes. me ka lau ko, i puhi ia i ke ahi, a
loaa ka nanahu.

Its flower is used for medicine for Pela no hoi kona pua, ua hana ia
certain ailments such as i laau no kekahi mau mai, oia hoi
stomachache, weakness or ke nahu, paaoao, a me ka e’a, a
ulcers of the mouth. Those kinds ua ola no hoi ia ano mai i keia
of diseases can be cured by that laau.
medicine.

The use of its nut: Its nut was Na hana a kona hua: Aia ma
strung into candles; that is, when kona hua ua hana ia i mea
the kukui nut is dried a person ihoiho kukui, oia hoi, i ka wa e
goes for it and gathers plenty, maloo ai ka hua o ke kukui, kii
then he returns to the house, aku ke kanaka, ohi a nui, alaila,
cooks them and when done hoi mai a hiki i ka hale, kalua i ka
cracks them; then string them on imu a moa, kike aku a pau,
a coconut stem, and when that is alaila, kui aku me ka niau a paa,
done it becomes the kukui a o ia iho la ka ihoiho kui i imihia,
candle with which to look at each he inoa hou nae ia, he kali kukui
other, but that is a new name; no ka inoa kahiko.
the old name was kali kukui. 267
Here is another thing: The nut of Eia kekahi; o ka hua no o ua
the kukui is also used in place of kukui nei, oia iho la no ka i-a, aia
fish; it is cooked and when it is nae i ka wa e pulehu ia ai a moa,
done that is the time to eat it. It is a oia iho la ka wa e ai ia ai, a ua
then called inamona, because it kapa ia kona inoa he inamona,
was sweet to the taste when
eaten. Still another thing: The no kona momona a ono ka ke ai
nut is used in fishing for uhu or in aku.
spying for squid; if it were not for
the kukui, these kinds of fishing Eia no kekahi; he mea lawaia
could not be carried on; for the kaka uhu, a akilo hee ia no hoi
oil of the kukui is the thing which ka hua o ke kukui, i na aole ka
enables one to see the dark hua o ke kukui, aole paha e
places of the ocean. That is one pono keia mau lawaia ana, ke
of its uses. ole ka hua kukui, nana e
hoomoakaka aku i na wahi
pouliuli o ka moana, oia iho la
kekahi hana.

Here is still another use of the Eia hou no kekahi hana a ka hua
kukui fruit: It is made into oil. It is kukui, ua hana ia no o ua hua
first gathered, and when there is nei i aila kui; ma ka hoiliili ana a
plenty it is cooked in the imu; nui, alaila, kalua i ka imu a moa,
when cooked it is cracked and kike aku a nahaha, ka iwi
the shell separated from the owaho, o ka io oloko oia ka mea
kernel. The kernel is made into e hana ai i aila ma ke ku’i ana
oil by pounding it on a board until iluna o ka papa a wali, olokaa
it is pulverized; then a large aku oe me kekahi pohaku nui aia
smooth ala 268 stone is rolled on nemonemo, maluna o ke kukui i
this pulverized kukui meat. The ku’i ia a wali; alaila, o ke kahe
juice is then run into a container aku la no ia o ka wai o ka kukui
through a strainer that would iloko o kekahi po’i a’u i hana ai
keep out the dregs. That is one me ke kanana, i mea e komo ole
use of the fruit of the kukui—for ai ke oka iloko, o ia iho la kekahi
oil; but perhaps there are more hana i ka hua kukui i aila, aka,
uses of the fruit of the kukui, but he nui aku no paha na hana o ka
these are what I have seen hua o ke kukui, o ka’u mau mea
being done with my own eyes, nae keia i ike maka i ka hana ia,
and that is why I have brought a oia ka’u mea i lawe mai ai i
these things for your keia mau mea, a hoikeike aku
enlightenment, my friends. iwaena o oukou e o’u mau hoa.

Timothy Lililea. Timothy Lililea.

AN ACCOUNT OF THE MOOLELO NO KA ULU.


BREADFRUIT.

There are two places where Elua wahi i loaa ai ka Ulu. 1. Ma


breadfruit is to be found. 1. At Kaawaloa, Kona, Hawaii. He
Kaawaloa, Kona, Hawaii. Man kanaka ke kumu o ko laila ulu.
was the origin of the breadfruit of Ma ka make ana ona, ua olelo
that place. Before his death he mua nae kela i kana mau keiki:
had said to his children: “If I die, “Ina wau e make, e nana olua i
both of you watch the tree that ka laau e ulu ae ana ma ka puka
may grow at the door of our o ka hale o kakou; o ka hua ona,
house; its fruit shall be your food; oia ka olua ai; o na lima a me na
the hands and hairs are the roots lauoho, oia ke a-a o lalo o ua ulu
of that breadfruit tree, the legs nei, o na wawae oia na lala, o ka
are its branches, the testicle is opea, oia ka hua o luna o ua ulu
the fruit thereon.” At that time nei.” la wa, oia ka ai i ola ai keia
that was the food that saved this ohana a pau, a hiki i ka wa a kini
whole family. At the time the forty o ke akua, a me ka lehu o ke
thousand gods and the four akua ike ana no ka hua o keia
hundred thousand gods saw the ulu, kii lakou e hoao e ai maka,
fruit of this breadfruit tree, they aole ono ke ai, hoao lakou e
went and plucked it and tried to pulehu ike i ka ono, olelo aku
eat it green, but it was not lakou ia Kane ma laua o
palatable; they then tried cooking Kanaloa i ka ono o keia hua,
it on hot coals and found it olelo mai laua: “Aole na he hua
relishable. They spoke to Kane e, he opea na no kekahi kanaka
and Kanaloa 269 of the sweetness i make.” Ia wa luai lakou [679]a
of this fruit; to which they replied pau ke aho, mai Kona ka luai
that the fruit was no other than hele ana ma kuahiwi a hiki i
the testes of a certain man who Waipio. Oia ke kumu i laha ai ko
is now dead. At this they vomited laila ulu i keia wa, no ka luai hele
until they were exhausted; ana o na akua ma ke kuahiwi a
[678]they started to vomit in Kona laha loa.
and continued through the
mountains till they reached
Waipio. That is why the
breadfruit trees became so
plentiful, at that time, because
the gods vomited on their way
through the mountains, hence its
wide spread.

2. At Puuloa, Oahu. Its breadfruit 2. Ma Puuloa i Oahu. Ko laila ulu


plant came from no Kanehunamoku mai, na
Kanehunamoku, 270 brought by kekahi mau kanaka o Puuloa i
two men of Puuloa who were out hele i ka lawaia a puhia e ka ino
fishing and were blown off by a nui, makani a me ka ua, a pae i
heavy wind and rain storm and keia aina kanaka ole, he akua
landed at the uninhabited land, wale no; nolaila mai ka laua lawe
save gods only. Therefore by ana mai i keia ulu a hiki i Puuloa,
them it was introduced at Puuloa kanu a ulu i kekahi lua nui a hua,
and planted in a large excavation ai keia mau kanaka; mahope ike
where it grew and bore fruit, mai o Haumea ma i keia mea o
which they ate. Haumea and ka ulu i lawe malu ia mai e keia
others afterwards knew of this mau kanaka, kii mai oia a ike,
breadfruit tree having been hele oia e nana i keia mau pae
brought away secretly by these moku, aole i ike ia keia ano laau.
men, so she came to see it Nolaila, o ka mea ia ona i hoolei
herself and made a visit to these hele ai i na aina a pau i ka ulu a
islands, but this variety of tree laha loa. O keia aina o
was not found. That is the Kanehunamoku aia i Kahiki, he
reason she scattered the aina i loaa ole i ka poe imi aina e
breadfruit in all lands; hence its hele mai nei e imi, no ka mea,
wide distribution. he aina akua keia. Ina ike ia e
kanaka emi i ka moana aole
This land of Kanehunamoku is in loaa, a pela wale aku. He nui na
Kahiki, a land not found by mea maikai o kela aina, aia ilaila
explorers who are endeavoring ka wai ola a Kane ma laua me
to discover, because this is a Kanaloa. O ka poe make a lehu
mythical land; if this land is seen ke kino ola i kela waiola.
by man it is then submerged in
the ocean, it can never be found,
and so continues on. The good
things of that land are many; the
living-water of Kane and Kanaloa
is found there; those who have
died and their bodies turned into
ashes can be brought back to life
by that water.

Its sap: The sap was very No Ke Kepau. He mea waiwai


valuable in the olden time for the loa ia i ka wa kahiko no ke kapili
snaring of birds; it was manu ana. O ka oi aku ia o ka
considered superior for such use mea hana nui ia e ka poe kia
by those persons who snared manu o na aina a pau loa. O
birds in all the islands. The Hawaii nae ka oi o ka aina kapili
island of Hawaii was the most manu mau. O ka manu e kapili ai
prominent in this method of bird o ka oo hulu pala o ka uka, o

You might also like