Download as pdf or txt
Download as pdf or txt
You are on page 1of 58

Pediatric Hypertension 5th Joseph T

Flynn Julie R Ingelfinger Tammy M


Brady Eds
Visit to download the full and correct content document:
https://ebookmeta.com/product/pediatric-hypertension-5th-joseph-t-flynn-julie-r-ingelfi
nger-tammy-m-brady-eds/
More products digital (pdf, epub, mobi) instant
download maybe you interests ...

Lovell and Winter s Pediatric Orthopaedics 8th Edition


John M. Flynn

https://ebookmeta.com/product/lovell-and-winter-s-pediatric-
orthopaedics-8th-edition-john-m-flynn/

Robotics by R R Ghorpade First Edition Dr R R Ghorpade


M T Puranik Dr M M Bhoomkar A J Patil

https://ebookmeta.com/product/robotics-by-r-r-ghorpade-first-
edition-dr-r-r-ghorpade-m-t-puranik-dr-m-m-bhoomkar-a-j-patil/

Essentials of Pediatric Urology 3rd Edition Duncan T


Wilcox David F M Thomas

https://ebookmeta.com/product/essentials-of-pediatric-
urology-3rd-edition-duncan-t-wilcox-david-f-m-thomas/

Hypertension A Case Based Approach 1st Edition


Elizabeth W Edwards Donald J Dipette Eds

https://ebookmeta.com/product/hypertension-a-case-based-
approach-1st-edition-elizabeth-w-edwards-donald-j-dipette-eds/
Examination pediatric 5th Edition Wayne Harres

https://ebookmeta.com/product/examination-pediatric-5th-edition-
wayne-harres/

Avery 2 1st Edition Erin R Flynn

https://ebookmeta.com/product/avery-2-1st-edition-erin-r-flynn/

Avery 1 1st Edition Erin R Flynn

https://ebookmeta.com/product/avery-1-1st-edition-erin-r-flynn/

Absorbing Inevitability 1st Edition Erin R Flynn

https://ebookmeta.com/product/absorbing-inevitability-1st-
edition-erin-r-flynn/

A Companion to Free Will 1st Edition Joseph Keim


Campbell Kristin M Mickelson V Alan White Eds

https://ebookmeta.com/product/a-companion-to-free-will-1st-
edition-joseph-keim-campbell-kristin-m-mickelson-v-alan-white-
eds/
Joseph T. Flynn
Julie R. Ingelfinger
Tammy M. Brady
Editors

Pediatric
Hypertension
Fifth Edition
Pediatric Hypertension
Joseph T. Flynn • Julie R. Ingelfinger •
Tammy M. Brady
Editors

Pediatric Hypertension
Fifth Edition

With 104 Figures and 111 Tables


Editors
Joseph T. Flynn Julie R. Ingelfinger
Department of Pediatrics Pediatric Nephrology Unit
University of Washington Mass General for Children at MGB
School of Medicine Harvard Medical School
Seattle, WA, USA Boston, MA, USA
Division of Nephrology
Seattle Children’s Hospital
Seattle, WA, USA

Tammy M. Brady
Department of Pediatrics
Division of Pediatric Nephrology
Johns Hopkins University School of Medicine
Baltimore, MD, USA

ISBN 978-3-031-06230-8 ISBN 978-3-031-06231-5 (eBook)


https://doi.org/10.1007/978-3-031-06231-5
1st edition: © Humana Press 2004
2nd edition: © Springer Science+Business Media, LLC 2011
3rd edition: © Springer Science+Business Media New York 2013
4th edition: © Springer International Publishing AG 2018
© Springer Nature Switzerland AG 2023
This work is subject to copyright. All rights are reserved by the Publisher, whether the whole or
part of the material is concerned, specifically the rights of translation, reprinting, reuse of
illustrations, recitation, broadcasting, reproduction on microfilms or in any other physical way,
and transmission or information storage and retrieval, electronic adaptation, computer software, or
by similar or dissimilar methodology now known or hereafter developed.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this
publication does not imply, even in the absence of a specific statement, that such names are exempt
from the relevant protective laws and regulations and therefore free for general use.
The publisher, the authors, and the editors are safe to assume that the advice and information in this
book are believed to be true and accurate at the date of publication. Neither the publisher nor the
authors or the editors give a warranty, expressed or implied, with respect to the material contained
herein or for any errors or omissions that may have been made. The publisher remains neutral with
regard to jurisdictional claims in published maps and institutional affiliations.

This Springer imprint is published by the registered company Springer Nature Switzerland AG.
The registered company address is: Gewerbestrasse 11, 6330 Cham, Switzerland.
Preface to the Fifth Edition

The importance of good cardiovascular health in adulthood is obvious but


cannot be overemphasized, especially for children with health problems such
as obesity and elevated blood pressure. This new edition of Pediatric Hyper-
tension emphasizes data that have appeared over the last several years that are
relevant to children with elevated blood pressure, as well as to children with
obesity and other health challenges. Cross-sectional and longitudinal studies in
large cohorts have led to notable advances in the field of pediatric hyperten-
sion. While there is still much work remaining, important knowledge gaps
have been narrowed by new studies. Advances include improved understand-
ing of which blood pressure thresholds lead to intermediate outcomes in
children such as left ventricular hypertrophy, changes in carotid intima
media thickness, and other markers as well as long-term cardiovascular out-
comes in adults; enhanced assessment and interpretation of noninvasive mea-
sures of cardiovascular disease risk and early vascular aging; recognition of
nontraditional cardiovascular disease risk factors in youth such as the role of
early life stress; and the evolving description of the neurocognitive sequelae of
hypertension. Further, the results of work over the last several years have
demonstrated the importance of emergency preparedness and adaptability in
the face of natural disasters and pandemics.
These substantial advancements, some of which challenge prevailing
thought and have already prompted changes in professional society and expert
clinical recommendations, are summarized in this Fifth Edition. As with the
Fourth Edition, this current text is part of the Springer Major Reference Work
program, which is available both in print and online, a format that allows for
real-time updating. In addition to new chapters that address the topics noted
above, this Edition provides updates to previously published chapters, includ-
ing those exploring the epidemiology of hypertension; the genetic, perinatal,
and lifestyle contributions to pediatric blood pressure and hypertension; sec-
ondary causes of hypertension and comorbidities; and the practical and prag-
matic approaches to auscultatory, home, and ambulatory blood pressure
measurement.
We are proud of the breadth of topics covered in this Edition of Pediatric
Hypertension. It truly does take a village, and we are most grateful to our
expert contributors, who shared their expertise and time to make this Edition as
compelling and current as we believe it to be. We hope that this text will not

v
vi Preface to the Fifth Edition

only inform and educate existing clinicians and experts immersed in the field,
but that it will also inspire students and trainees to choose a career that focuses
on cardiovascular health promotion across the lifespan.

Seattle, USA Joseph T. Flynn


Boston, USA Julie R. Ingelfinger
Baltimore, USA Tammy M. Brady
December 2022
Preface to the Fourth Edition

We are delighted to present this expanded fourth edition of Pediatric Hyper-


tension, which is intended to capture and update the ongoing progress in
childhood hypertension. There is a growing recognition that adult cardiovas-
cular disease has its origins in childhood, supported by many recent studies.
Additionally, the assessment of the short-term sequelae of childhood hyper-
tension is providing new and important data, reviewed herein. Further, there
are increasing numbers of studies that are delineating mechanisms of blood
pressure elevation in the young. While the obesity epidemic appears to be
leveling off (at least in the United States), it remains an important contributor
to the higher prevalence of childhood hypertension reported in recent years;
numerous epidemiologic studies have become available since publication of
the third edition of this text and are detailed here. With publication of this new
fourth edition, we hope to bring further focus on the importance of under-
standing and addressing the role of the obesity epidemic in pediatric
hypertension.
As our publisher, Springer, has transitioned this text to its Major Reference
Work program, which is available not only in print but also online, which
allows for continual updating, we have been able not only to retain the topics
covered in previous editions of Pediatric Hypertension but also to add new
chapters that address additional and important aspects of childhood hyperten-
sion. One new chapter addresses the controversy over routine childhood blood
pressure screening raised by the 2014 US Preventive Services Task Force
Report. Obesity hypertension is now covered in two chapters, one focusing on
mechanisms and the other on clinical aspects. Another important mechanism
of cardiovascular disease, vascular dysfunction, is covered in a new chapter in
the first section of the text. We also now address the important topic of home
blood pressure measurement, while continuing to cover casual and ambulatory
blood pressure measurement in detail. Expanded chapters on ESRD-related
hypertension, substance-induced hypertension, hypertension in oncology
patients, and hypertension in young adults should be of substantial interest
to clinicians who care for such patients. We have also expanded the section on
hypertension research with a new chapter on cohort studies and meta-analyses
and their role in studying childhood hypertension. Finally, we have added a
short Appendix summarizing the major changes of the 2017 American Acad-
emy of Pediatrics clinical practice guideline on childhood hypertension, which
was completed as this new edition was in progress.

vii
viii Preface to the Fourth Edition

It is impossible to put together a comprehensive text such as Pediatric


Hypertension without more than “a little help from our friends.” We are greatly
indebted to our returning authors as well as to our new authors, all of whom
were asked to contribute to the text because of their acknowledged expertise in
childhood hypertension. We also thank Daniela Graf and Rebecca Urban from
Springer for helping keep everyone on task. We are certain that you will agree
that the tremendous amount of work that has been devoted to this edition of
Pediatric Hypertension has led to a comprehensive and useful text, which we
hope you will consult often in your clinics and research laboratories.

Seattle, WA, USA Joseph T. Flynn


Boston, MA, USA Julie R. Ingelfinger
Boise, ID, USA Karen M. Redwine
Preface to the Third Edition

We are excited to offer you this third edition of Pediatric Hypertension.


Interest in childhood hypertension has increased markedly since the publica-
tion of the prior editions of this text, fueled in part by the increase in the
prevalence of hypertension in children and adolescents, owing to the obesity
epidemic. Investigators have continued to explore many aspects of hyperten-
sion in the young, resulting in better understanding of the mechanisms,
manifestations and management of this important clinical problem. Cardio-
vascular disease remains the leading medical cause of death in the world. Only
by understanding important risk factors such as hypertension at the earliest
stages of disease, during childhood, can substantial progress at eradicating this
disease be made.
In this edition, we have retained most of the topics from the prior two
editions, but have made some important additions and replacements that we
feel will increase the usefulness of the text to clinicians and researchers alike.
New clinically oriented chapters on obesity-related hypertension, endocrine
hypertension and renovascular hypertension should help guide the evaluation
and management of these major causes of hypertension in the young. A new
chapter on models of hypertension should help both researchers and clinicians
to better understand the investigative approaches that have been employed to
study childhood hypertension. There are also new chapters on hypertension in
pregnancy and ethnic influences on hypertension in the young, which should
be of particular interest to those who care for large numbers of teens and
minority patients, respectively.
A text such as this would not have been possible without contributions from
many busy people, all of whom are acknowledged experts in the field. We are
profoundly grateful to our colleagues who agreed to contribute chapters to this
text, especially those who willingly took on new topics only 2–3 years after
writing their chapters for the second edition! It has been a privilege to work
with such a talented and generous group of collaborators, and we are sure that
you will agree that their efforts have resulted in an enhanced third edition.

Seattle, WA, USA Joseph T. Flynn


Boston, MA, USA Julie R. Ingelfinger
Princeton, NJ, USA Ronald J. Portman

ix
Preface to the Second Edition

Interest in pediatric hypertension dates back nearly half a century, when it was
first recognized that a small percentage of children and adolescents had elevated
blood pressures – and in those days, the same normal values for adult blood
pressure were utilized in children! The many advances since that time have led to
a much clearer understanding of how to identify, evaluate, and treat hypertensive
children and adolescents. At the same time, many questions remain: What causes
hypertension in children without underlying systemic conditions? What are the
long-term consequences of high blood pressure in the young? What is the
optimal therapy of childhood hypertension? and Does such treatment benefit
the affected child or adolescent? Can we identify children at risk of developing
hypertension and intervene to prevent its occurrence? Readers conversant with
the history of hypertension in the young will recognize that these questions were
being asked decades ago and may still be unanswered for many years to come.
The first text focusing on pediatric hypertension was published in 1982.
The book you are about to read is a direct descendant of that first effort to
summarize what is known about hypertension in the young. We are fortunate
to have been given the first opportunity to produce a second edition of such a
text, which reflects the increased interest in hypertension in the young that has
developed since the publication of the first edition of Pediatric Hypertension.
Many chapters from the first edition have been revised and updated by their
original authors; others have been written by new authors. New chapters on
topics of recent interest in pediatric hypertension such as the metabolic
syndrome and sleep disorders have been added. We hope that the reader will
find this new edition of Pediatric Hypertension to be an up-to-date, clinically
useful reference as well as a stimulus to further research in the field.
It is also our hope that the advances summarized in this text will ultimately
lead to increased efforts toward the prevention of hypertension in the young,
which, in turn, should ameliorate the burden of cardiovascular disease in
adults. We thank our many colleagues who have taken time from their busy
schedules to contribute to this text – and we are sure that you will agree with us
that their combined efforts have resulted in a valuable reference to those
interested in hypertension in the young.

Seattle, WA, USA Joseph T. Flynn


Boston, MA, USA Julie R. Ingelfinger
Princeton, NJ, USA Ronald J. Portman

xi
Preface to the First Edition

More than a quarter of a century has elapsed since the first Task Force on Blood
Pressure Control in Children was published in 1977. Since that seminal
publication, normative data have been obtained for both casual and ambulatory
children’s blood pressure. Blood pressure measurement in infants, children,
and adolescents, once an afterthought, has become routine. Pediatric Hyper-
tension discusses the many aspects of pediatric hypertension – a multi-
disciplinary subspecialty that is comprised of pediatric nephrologists,
cardiologists, endocrinologists, pharmacologists, and epidemiologists.
Although some areas of our discipline have become well established, others,
such as routine use of ambulatory blood pressure recording and well-designed
trials in pediatric hypertension, are still emerging. Accordingly, we have
included chapters that focus on aspects of blood pressure control and hyper-
tension in the very young that are particularly relevant to those caring for
infants, children, and adolescents.
Pediatric Hypertension opens with chapters concerning blood pressure
regulation in the very young: the transition from fetal life to infant circulation,
the factors that regulate blood pressure in early childhood, and the chronobi-
ology of pediatric blood pressure. We then move on to the assessment of blood
pressure in children. The book addresses both casual and ambulatory blood
pressure measurement methodologies and norms, as well as the epidemiology
of hypertension in children.
Definitions of hypertension in children, predictors of future hypertension,
risk factors, and special populations are discussed at length. Comprehensive
chapters on both primary and secondary hypertension in children point out
differences in presentation of hypertension in the pediatric, in comparison to
the adult, population. The contributions of genetics to the understanding of
hypertension are presented, as well as those events during gestation and
perinatal life that may influence the development of later hypertension. Risk
factors that are discussed include the influences of race and ethnicity, diet,
obesity, and society. Special populations, including the neonate with hyper-
tension and the child with chronic renal failure or end-stage renal disease, are
each discussed in a separate chapter. In those chapters, the pathophysiology
insofar as it is known is also considered.
This text concludes with a section that focuses on the evaluation and
management of pediatric hypertension. Suggestions for evaluation are pre-
sented, and both nonpharmacologic and pharmacologic therapy are discussed

xiii
xiv Preface to the First Edition

at length. The 1997 Food and Drug Administration Modernization Act, which
offers extension of market exclusivity in return for approved clinical trials of
medications with pediatric indication, has had a major impact on the conduct
of pediatric antihypertensive medication trials. The current status of such
pediatric antihypertensive trials is presented. In the appendix, the reader will
find the latest tables for the definition of hypertension in children from the
Fourth Report on the Diagnosis, Evaluation, and Treatment of High Blood
Pressure in Children and Adolescents, to be published in Pediatrics in the
summer of 2004.
We hope that Pediatric Hypertension provides a catalyst for more interest in
pediatric hypertension as well as a guide for the interested clinician or clinical
researcher already active in this discipline. Very shortly, the results of addi-
tional trials concerning new antihypertensive agents in children will be avail-
able with the mandate that new antihypertensive medications be evaluated in
children. An update by the Task Force on Blood Pressure Control in Children
will also be completed in 2004. A number of groups that have a special interest
in blood pressure and its control in the very young will continue to contribute
to the field, among them, most notably, the International Pediatric Hyperten-
sion Association; the National Heart, Lung, and Blood Institute; the American
Society of Hypertension; and the American Society of Pediatric Nephrology.
These initiatives will lead to a better understanding of the definition, causes,
consequences, prevention, and treatment of pediatric hypertension. In addition
to advances in molecular and genetics laboratories, new technologies in
assessment of human cardiac and vascular anatomy and physiology will help
to elucidate the pathophysiology of hypertension and its response to manage-
ment. In so doing, our hope is that the trend towards reduction in cardiovas-
cular morbidity and mortality will continue for the current generation of
children.
Finally, we wish to acknowledge the pioneering work of so many in the
field of pediatric hypertension that has given us the foundation and tools to
advance our field.

International Pediatric Hypertension Association Ronald J. Portman, M.D.


Jonathan M. Sorof, M.D.
Julie R. Ingelfinger, M.D.
Contents

Part I Regulation of Blood Pressure and Pathophysiological


Mechanisms of Hypertension . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1

1 Neurohumoral and Autonomic Regulation of Blood


Pressure . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
Empar Lurbe and Josep Redon
2 Cardiovascular Influences on Blood Pressure . . . . . . . . . . . . 21
Manish D. Sinha and Phil Chowienczyk
3 Vasoactive Factors and Blood Pressure in Children . . . . . . . 41
Ihor V. Yosypiv
4 Ions and Fluid Dynamics in Hypertension . . . . . . . . . . . . . . . 59
Avram Z. Traum
5 Uric Acid in the Pathogenesis of Hypertension . . . . . . . . . . . 71
Daniel I. Feig
6 Insulin Resistance and Other Mechanisms of Obesity
Hypertension . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 91
Vidhu Thaker and Bonita Falkner
7 Monogenic and Polygenic Contributions to
Hypertension . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 113
Julie R. Ingelfinger
8 Antenatal Programming of Blood Pressure . . . . . . . . . . . . . . 133
Andrew M. South
9 Familial Aggregation of Blood Pressure and the
Heritability of Hypertension . . . . . . . . . . . . . . . . . . . . . . . . . . 159
Sujane Kandasamy and Rahul Chanchlani
10 The Role of Dietary Electrolytes and Childhood Blood
Pressure Regulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 169
Dawn K. Wilson, Tyler C. McDaniel, and Sandra M. Coulon
11 Endothelial Dysfunction and Vascular Remodeling in
Hypertension . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 195
Julie Goodwin

xv
xvi Contents

12 Adverse Childhood Experiences and Their Relevance to


Hypertension in Children and Youth . . . . . . . . . . . . . . . . . . . 217
Julie R. Ingelfinger
13 Salt Sensitivity in Childhood Hypertension . . . . . . . . . . . . . . 229
Coral D. Hanevold
14 Early Vascular Aging in Pediatric Hypertension
Patients . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 249
Mieczysław Litwin

Part II Assessment of Blood Pressure in Children:


Measurement, Normative Data, and Epidemiology . . . . . . . . . . . . 271

15 Methodology of Office Blood Pressure Measurement . . . . . . 273


Tammy M. Brady
16 Value of Routine Screening for Hypertension in
Childhood . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 285
Joseph T. Flynn
17 Development of Blood Pressure Norms and Definition of
Hypertension in Children . . . . . . . . . . . . . . . . . . . . . . . . . . . . 297
Bonita Falkner
18 Ambulatory Blood Pressure Monitoring Methodology and
Norms in Children . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 311
Elke Wühl
19 Methodology and Applicability of Home Blood Pressure
Monitoring in Children and Adolescents . . . . . . . . . . . . . . . . 345
George S. Stergiou and Angeliki Ntineri
20 Epidemiology of Hypertension and Cardiovascular
Disease in Children and Adolescents . . . . . . . . . . . . . . . . . . . 367
Elyse O. Kharbanda

Part III Hypertension in Children: Etiologies and Special


Populations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 387

21 Ethnic Differences in Childhood Blood Pressure . . . . . . . . . . 389


Joshua Samuels and Xamayta Negroni-Balasquide
22 Obesity Hypertension: Clinical Aspects . . . . . . . . . . . . . . . . . 405
Ian Macumber and Joseph T. Flynn
23 Hypertension in Children with Type 2 Diabetes or the
Metabolic Syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 421
Grace Kim and Joseph T. Flynn
24 Primary Hypertension in Children . . . . . . . . . . . . . . . . . . . . . 439
Manpreet K. Grewal, Tej K. Mattoo, and Gaurav Kapur
Contents xvii

25 White Coat and Masked Hypertension . . . . . . . . . . . . . . . . . 461


Yosuke Miyashita and Coral D. Hanevold
26 Hypertension in Chronic Kidney Disease . . . . . . . . . . . . . . . . 477
Susan M. Halbach
27 Hypertension in End-Stage Kidney Disease: Dialysis . . . . . . 499
Franz Schaefer
28 Hypertension in End-Stage Kidney Disease:
Transplantation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 515
Tomáš Seeman
29 Renovascular Hypertension, Vasculitis, and Aortic
Coarctation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 533
Kjell Tullus and Jelena Stojanovic
30 Endocrine Hypertension . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 549
Perrin C. White
31 Neonatal and Infant Hypertension . . . . . . . . . . . . . . . . . . . . . 573
Janis M. Dionne
32 Obstructive Sleep Apnea and Hypertension in Children . . . 601
Amee Revana and Alisa A. Acosta
33 Hypertension in the Pregnant Teenager . . . . . . . . . . . . . . . . . 615
Tracy E. Hunley and Deborah P. Jones
34 Neurocognition in Childhood Hypertension . . . . . . . . . . . . . 645
Marc B. Lande and Juan C. Kupferman
35 Stroke and Childhood Hypertension . . . . . . . . . . . . . . . . . . . 659
Juan C. Kupferman, Marc B. Lande, and Stella Stabouli
36 Medication and Substance-Induced Hypertension:
Mechanisms and Management . . . . . . . . . . . . . . . . . . . . . . . . 683
Sandeep K. Riar and Douglas L. Blowey
37 Hypertension in Oncology and Stem Cell Transplant
Patients . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 701
Benjamin L. Laskin and Sangeeta R. Hingorani
38 Hypertension in Older Adolescents and Young Adults . . . . . 723
Matthew B. Rivara
39 Hypertension in the Developing World . . . . . . . . . . . . . . . . . 739
Vera H. Koch

Part IV Evaluation and Management of Pediatric


Hypertension . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 753

40 Diagnostic Evaluation of Pediatric Hypertension . . . . . . . . . 755


Nicholas Larkins and Derek Roebuck
xviii Contents

41 Sequelae of Hypertension in Children and Adolescents . . . . 771


Donald J. Weaver Jr. and Mark M. Mitsnefes
42 Cardiovascular Assessment of Childhood Hypertension . . . 785
Edem Binka and Elaine M. Urbina
43 The Role of ABPM in Evaluation of Hypertensive
Target-Organ Damage . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 805
Stella Stabouli and Vasilios Kotsis
44 Exercise Testing in Hypertension and Hypertension
in Athletes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 827
Carissa M. Baker-Smith and Takeshi Tsuda
45 Nonpharmacologic Treatment of Pediatric Hypertension . . . 843
Stephen R. Daniels and Sarah C. Couch
46 Pharmacologic Treatment of Pediatric Hypertension . . . . . . 857
Michael A. Ferguson and Deborah R. Stein
47 Management of Hypertensive Emergencies . . . . . . . . . . . . . . 883
Craig W. Belsha
48 Hypertension Care During Emergencies and Pandemics . . . 899
Joyce P. Samuel and Bradley K. Hyman

Part V Hypertension Research in Pediatrics . . . . . . . . . . . . . . . . . . 907

49 Hypertensive Models and Their Relevance to Pediatric


Hypertension . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 909
Julie R. Ingelfinger
50 Cohort Studies, Meta-analyses, and Clinical Trials in
Childhood Hypertension . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 921
Nicholas Larkins and Jonathan Craig
51 Changes in Drug Development Regulations and Their
Impact on Clinical Trials . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 945
Elizabeth J. Thompson, Kevin D. Hill, Rachel D. Torok, and
Jennifer S. Li
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 959
About the Editors

Dr. Joseph T. Flynn MD, MS, is the Dr. Robert


O. Hickman Endowed Chair in Pediatric Nephrol-
ogy; Professor of Pediatrics, University of
Washington; and Chief, Division of Nephrology,
Seattle Children’s Hospital. He is an internation-
ally recognized expert in the treatment of hyper-
tension in children and recently chaired the
American Academy of Pediatrics Subcommittee
that developed the updated clinical practice guide-
line on high blood pressure in children and ado-
lescents. Dr. Flynn served on the Council of the
International Pediatric Nephrology Association
from 2010 to 2019, and also on the Council of
the American Society of Pediatric Nephrology,
including 2 years as ASPN President
(2012–2014). He currently is a board member of
the Renal Physicians Association and a member of
the Nephrology sub-board of the American Board
of Pediatrics. He currently serves as one of the
Editors-in-Chief of the journal Pediatric Nephrol-
ogy and is a member of the editorial boards of
Hypertension, Blood Pressure Monitoring, and
The Journal of Pediatrics. He has contributed
chapters to all five editions of the Pediatric Hyper-
tension and edited the three previous editions.

Julie R. Ingelfinger MD, is Professor of Pediat-


rics at Harvard Medical School, Senior Consultant
in pediatric nephrology at Mass General for Chil-
dren at MGB, and Deputy Editor of the New
England Journal of Medicine. She is an interna-
tionally recognized hypertension specialist and
consultative pediatric nephrologist. Her commit-
ment to teaching has been reflected by receiving
the Henry L. Barnett Award from the AAP in
2009, the Founders Award from the American
xix
xx About the Editors

Society of Pediatric Nephrology in 2012, the


National Kidney Foundation’s Honors Award in
2018, the Alumni Lifetime Achievement Award
from Albert Einstein College of Medicine (2018),
and the Barbara T. Murphy Award from the Amer-
ican Society of Nephrology (2022).
Dr. Ingelfinger has been involved in studies of
the intrarenal renin angiotensin aldosterone sys-
tem (RAAS) for many years. Her current projects
focus on the role of the intrarenal renin angioten-
sin system in disease states and the role of mater-
nal diabetes in kidney development and perinatal
programming.
She is interested in innovative ways to teach
writing and communication and has been an editor
of Pediatric Hypertension in each of its five
editions.

Dr. Tammy M. Brady MD, PhD, is Associate


Professor of Pediatrics at Johns Hopkins Univer-
sity School of Medicine where she serves as Vice
Chair for Clinical Research and as the Associate
Director of the Welch Center for Prevention, Epi-
demiology and Clinical Research. She has
focused her career on improving the care of chil-
dren at increased cardiovascular disease risk
through research and clinical innovation. She
serves as the Medical Director of the Harriet
Lane Kidney Center Pediatric Hypertension Pro-
gram, and she directs a multidisciplinary obesity
hypertension clinic (ReNEW clinic; Reversing the
Negative cardiovascular Effects of Weight).
Dr. Brady is an expert in blood pressure measure-
ment and blood pressure device validation. She is
the co-chair of the Association for the Advance-
ment of Medical Instrumentation (AAMI) Sphyg-
momanometer committee, the American
professional society that develops American
National Standards and technical reports regard-
ing sphygmomanometer devices. She was Chair
of a two-day meeting at the WHO where
published guidance and specifications on blood
pressure measuring devices were updated. She is
also Co-chair of the American Medical Associa-
tion’s Validated Device Listing Committee.
Contributors

Alisa A. Acosta Department of Pediatrics Renal Section, Baylor College of


Medicine/Texas Children’s Hospital, Houston, TX, USA

Carissa M. Baker-Smith Nemours Cardiac Center, Nemours Children’s


Hospital, Wilmington, DE, USA

Craig W. Belsha SSM Health Cardinal Glennon Children’s Medical Center,


Saint Louis University, St. Louis, MO, USA

Edem Binka Division of Pediatric Cardiology, University of Utah, Salt Lake


City, UT, USA

Douglas L. Blowey Pediatric Nephrology, Children’s Mercy Hospital, Uni-


versity of Missouri, Kansas City, MO, USA

Tammy M. Brady Department of Pediatrics, Division of Pediatric Nephrol-


ogy, Johns Hopkins University School of Medicine, Baltimore, MD, USA

Rahul Chanchlani Division of Nephrology, Department of Pediatrics,


McMaster University, Hamilton, ON, Canada

Phil Chowienczyk Kings College London British Heart Foundation Centre,


London, UK

Sarah C. Couch Department of Rehabilitation, Exercise and Nutrition Sci-


ences, University of Cincinnati Medical Center, Cincinnati, OH, USA

Sandra M. Coulon West Texas Veterans Healthcare System, Big Spring, TX,
USA

Jonathan Craig College of Medicine and Public Health, Flinders University,


Adelaide, SA, Australia

Stephen R. Daniels Department of Pediatrics, University of Colorado School


of Medicine, Children’s Hospital Colorado, Aurora, CO, USA

Janis M. Dionne Division of Nephrology, Department of Pediatrics, Univer-


sity of British Columbia, BC Children’s Hospital, Vancouver, BC, Canada

Bonita Falkner Departments of Medicine and Pediatrics, Thomas Jefferson


University, Philadelphia, PA, USA

xxi
xxii Contributors

Daniel I. Feig Division of Nephrology, Department of Pediatrics, University


of Alabama, Birmingham, AL, USA
Michael A. Ferguson Division of Nephrology, Boston Children’s Hospital,
Harvard Medical School, Boston, MA, USA
Joseph T. Flynn Department of Pediatrics, University of Washington School
of Medicine, Seattle, WA, USA
Division of Nephrology, Seattle Children’s Hospital, Seattle, WA, USA
Julie Goodwin Department of Pediatrics, Yale University School of Medi-
cine, New Haven, CT, USA
Manpreet K. Grewal Division of Nephrology and Hypertension, Depart-
ment of Pediatrics, Children’s Hospital of Michigan, Detroit, MI, USA
Susan M. Halbach Division of Nephrology, Seattle Children’s Hospital,
University of Washington School of Medicine, Seattle, WA, USA
Coral D. Hanevold Division of Nephrology, Department of Pediatrics, Uni-
versity of Washington School of Medicine, Seattle, WA, USA
Kevin D. Hill Department of Pediatrics, Duke Clinical Research Institute,
Durham, NC, USA
Sangeeta R. Hingorani Division of Nephrology, Seattle Children’s Hospital,
Seattle, WA, USA
Tracy E. Hunley Pediatric Nephrology, Monroe Carell Jr. Children’s Hospi-
tal at Vanderbilt, Vanderbilt University Medical Center, Nashville, TN, USA
Bradley K. Hyman Department of Pediatrics- Division of Pediatric Nephrol-
ogy & Hypertension, McGovern Medical School at University of Texas Health
Science Center, Houston, TX, USA
Julie R. Ingelfinger Pediatric Nephrology Unit, Mass General for Children at
MGB, Harvard Medical School, Boston, MA, USA
Deborah P. Jones Pediatric Nephrology, Monroe Carell Jr. Children’s Hos-
pital at Vanderbilt, Vanderbilt University Medical Center, Nashville, TN, USA
Sujane Kandasamy Department of Health Research Methods, Evidence &
Impact, McMaster University, Hamilton, ON, Canada
Gaurav Kapur Division of Nephrology and Hypertension, Department of
Pediatrics, Children’s Hospital of Michigan, Detroit, MI, USA
Pediatric Nephrology, Central Michigan University, Detroit, MI, USA
Elyse O. Kharbanda Research, HealthPartners Institute, Minneapolis, MN,
USA
Grace Kim Division of Endocrinology and Diabetes, Seattle Children’s
Hospital, Seattle, WA, USA
Department of Pediatrics, University of Washington School of Medicine,
Seattle, WA, USA
Contributors xxiii

Vera H. Koch Pediatric Nephrology, University of Sao Paulo Medical


School, São Paulo, Brazil
Department of Pediatrics, Pediatric Nephrology Unit, Instituto da Criança
Hospital das Clinicas, University of São Paulo Medical School, Sao Paulo,
Brazil
Medical Residency, University of Sao Paulo Medical School, Sao Paulo,
Brazil
Vasilios Kotsis Department of Internal Medicine, Aristotle University of
Thessaloniki, Thessaloniki, Greece
Juan C. Kupferman Department of Pediatrics, Division of Pediatric
Nephrology and Hypertension, Maimonides Medical Center, Brooklyn, NY,
USA
Marc B. Lande Department of Pediatrics, Division of Pediatric Nephrology,
University of Rochester Medical Center, Rochester, NY, USA
Nicholas Larkins Department of Nephrology and Hypertension, Perth Chil-
dren’s Hospital, Nedlands, WA, Australia
Paediatrics Division, Medical School, University of Western Australia, Perth,
WA, Australia
Benjamin L. Laskin Division of Nephrology, The Children’s Hospital of
Philadelphia, Philadelphia, PA, USA
Jennifer S. Li Department of Pediatrics, Duke Clinical Research Institute,
Durham, NC, USA
Mieczysław Litwin Department of Nephrology and Arterial Hypertension,
The Children’s Memorial Health Institute, Warsaw, Poland
Empar Lurbe Pediatric Department, Consorcio Hospital General, Univer-
sity of Valencia, Valencia, Spain
CIBER Fisiopatología Obesidad y Nutrición (CB06/03), Instituto de Salud
Carlos III, Madrid, Spain
Ian Macumber Keck School of Medicine, University of Southern California,
Los Angeles, CA, USA
Division of Nephrology, Children’s Hospital Los Angeles, Los Angeles, CA,
USA
Tej K. Mattoo Departments of Pediatrics (Nephrology) and Urology, Wayne
State University School of Medicine, Detroit, MI, USA
Tyler C. McDaniel Department of Psychology, Barnwell College, University
of South Carolina, Columbia, SC, USA
Mark M. Mitsnefes Division of Nephrology and Hypertension, Cincinnati
Children’s Hospital Medical Center, Cincinnati, OH, USA
Yosuke Miyashita Department of Pediatrics, University of Pittsburgh Med-
ical Center Children’s Hospital of Pittsburgh, University of Pittsburgh School
of Medicine, Pittsburgh, PA, USA
xxiv Contributors

Xamayta Negroni-Balasquide Department of Pediatrics, University of


Puerto Rico Medical Science Campus, San Juan, PR, USA

Angeliki Ntineri Third Department of Medicine, Sotiria Hospital, School of


Medicine, Hypertension Center STRIDE-7, National and Kapodistrian Uni-
versity of Athens, Athens, Greece

Josep Redon CIBER Fisiopatología Obesidad y Nutrición (CB06/03),


Instituto de Salud Carlos III, Madrid, Spain
Cardiovascular and Renal Research Group, INCLIVA Research Institute,
University of Valencia, Valencia, Spain

Amee Revana Department of Pediatrics, Pulmonary and Sleep Medicine


Section, Baylor College of Medicine/Texas Children’s Hospital, Houston,
TX, USA

Sandeep K. Riar Pediatric Nephrology, Children’s Mercy Hospital, Univer-


sity of Missouri, Kansas City, MO, USA

Matthew B. Rivara Division of Nephrology, Department of Medicine, Kid-


ney Research Institute, University of Washington, Seattle, WA, USA

Derek Roebuck Medical School, University of Western Australia, Perth,


WA, Australia
Department of Medical Imaging, Perth Children’s Hospital, Nedlands, WA,
Australia

Joyce P. Samuel Department of Pediatrics- Division of Pediatric Nephrology


& Hypertension, McGovern Medical School at University of Texas Health
Science Center, Houston, TX, USA

Joshua Samuels Department of Pediatrics, University of Puerto Rico Med-


ical Science Campus, San Juan, PR, USA

Franz Schaefer Division of Pediatric Nephrology, Center for Pediatrics and


Adolescent Medicine, Heidelberg University Hospital, Heidelberg, Germany

Tomáš Seeman Department of Pediatrics, Charles University Prague, 2nd


Medical Faculty, Prague, Czech Republic
Department of Pediatrics, Dr. von Hauner Children’s Hospital, LMU Munich,
Munich, Germany

Manish D. Sinha Department of Paediatric Nephrology, Evelina London


Children’s Hospital, Guys & St Thomas’ NHS Foundation Trust, London, UK
Kings College London British Heart Foundation Centre, London, UK

Andrew M. South Department of Pediatrics, Section of Nephrology, Brenner


Children’s, Wake Forest University School of Medicine, Winston Salem, NC,
USA

Stella Stabouli Department of Pediatrics, Aristotle University of


Thessaloniki, Hippocratio Hospital, Thessaloniki, Greece
Contributors xxv

Deborah R. Stein Division of Nephrology, Boston Children’s Hospital,


Harvard Medical School, Boston, MA, USA
George S. Stergiou Third Department of Medicine, Sotiria Hospital, School
of Medicine, Hypertension Center STRIDE-7, National and Kapodistrian
University of Athens, Athens, Greece
Jelena Stojanovic Great Ormond Street Hospital for Children, London, UK
Vidhu Thaker Divisions of Molecular Genetics, and Pediatric Endocrinol-
ogy, Department of Pediatrics, Columbia University Medical Center, New
York, NY, USA
Division of Endocrinology, Boston Childrens Hospital, Harvard Medical
School, Boston, MA, USA
Elizabeth J. Thompson Department of Pediatrics, Duke Clinical Research
Institute, Durham, NC, USA
Rachel D. Torok Department of Pediatrics, University of Pittsburgh Medical
Center, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
Avram Z. Traum Division of Nephrology, Boston Children’s Hospital,
Boston, MA, USA
Department of Pediatrics, Harvard Medical School, Boston, MA, USA
Takeshi Tsuda Nemours Cardiac Center, Nemours Children’s Hospital,
Wilmington, DE, USA
Kjell Tullus Great Ormond Street Hospital for Children, London, UK
Elaine M. Urbina Preventive Cardiology, Cincinnati Children’s Hospital
Medical Center, University of Cincinnati, Cincinnati, OH, USA
Donald J. Weaver Jr. Department of Pediatrics, Division of Nephrology and
Hypertension, Atrium Health Levine Children’s, Charlotte, NC, USA
Perrin C. White Division of Pediatric Endocrinology, Department of Pedi-
atrics, UT Southwestern Medical Center and Children’s Medical Center,
Dallas, TX, USA
Dawn K. Wilson Department of Psychology, Barnwell College, University
of South Carolina, Columbia, SC, USA
Elke Wühl Center for Pediatrics and Adolescent Medicine, University Hos-
pital Heidelberg, Heidelberg, Germany
Ihor V. Yosypiv Department of Pediatrics, Tulane University, New Orleans,
LA, USA
Part I
Regulation of Blood Pressure and
Pathophysiological Mechanisms of
Hypertension
Neurohumoral and Autonomic Regulation
of Blood Pressure 1
Empar Lurbe and Josep Redon

Contents
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
Neural Components of BP Regulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
Central Nervous System . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
Autonomic Nervous System . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
Sympathetic Activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
Parasympathetic Activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
Baro- and Chemoreflexes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
Humoral Components of BP Regulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
Renin-Angiotensin-Aldosterone System . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
Atrial Natriuretic Peptides . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
The Apelin System . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
The Kidney, Fluid Volume, and Salt Intake . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
The Vasculature and Nitric Oxide . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15

E. Lurbe (*)
Pediatric Department, Consorcio Hospital General,
University of Valencia, Valencia, Spain
CIBER Fisiopatología Obesidad y Nutrición (CB06/03),
Instituto de Salud Carlos III, Madrid, Spain
e-mail: empar.lurbe@uv.es
J. Redon
CIBER Fisiopatología Obesidad y Nutrición (CB06/03),
Instituto de Salud Carlos III, Madrid, Spain
Cardiovascular and Renal Research Group, INCLIVA
Research Institute, University of Valencia, Valencia, Spain
e-mail: josep.redon@uv.es

© Springer Nature Switzerland AG 2023 3


J. T. Flynn et al. (eds.), Pediatric Hypertension,
https://doi.org/10.1007/978-3-031-06231-5_1
4 E. Lurbe and J. Redon

Circadian Variation of Blood Pressure . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16


Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
Cross-References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17

Abstract Keywords

Maintenance of BP requires the coordinated Angiotensin II · Atrial natriuretic peptides ·


regulation of many components that interact Baroreflex · Blood pressure · Circadian
in an interdependent network that ultimately rhythm · Endothelin · Nitric oxide ·
controls the main determinants of BP, cardiac Sympathetic activity · Parasympathetic ·
output, peripheral resistance, and blood vol- Vasoconstriction · Vasopressin
ume. Neurohumoral and autonomic mecha-
nisms are key components of a complex Abbreviations
network. The central nervous system, orga-
nized into multiple levels of integrative cen- ACE1 Angiotensin-converting enzyme 1
ters, regulates sympathetic activity and ACE2 Angiotensin-converting enzyme 2
vasopressin release in response to nervous ACh Acetylcholine
and humoral input. The autonomic system, by ADMA Asymmetric dimethylarginine
means of the efferent sympathetic and para- ALD Aldosterone
sympathetic pathways, regulates heart rate, Ang I Angiotensin I
vasoconstriction, and activation of the renin- Ang I–7 Angiotensin 1–7
angiotensin-aldosterone system. Humoral Ang II Angiotensin II
components, angiotensin II, aldosterone, and Ang III Angiotensin III
atrial natriuretic peptides regulate vasocon- Ang IV Angiotensin IV
striction and sodium excretion. Short-term reg- AngIVr Angiotensin IV receptor
ulation of cardiac output and peripheral ANGN Angiotensinogen
resistance is interconnected by autonomic ner- ANP Atrial natriuretic peptide
vous system reflexes with the intervention of ANS Autonomic nervous system
baroreceptors. Mid-term control is achieved by Apr Apelin receptor
the interplay between sympathetic nervous AS Apelinergic system
traffic, humoral systems, and the kidney, con- AT1r AT1-receptor
trolling vascular peripheral resistance and AT2r AT2-receptor
blood volume. The kidney, through both BNP Brain natriuretic peptide
pressure-natriuresis and arteriolar mecha- BP Blood pressure
nisms, as well as by flow-mediated vasodilata- CNP C-type natriuretic peptide
tion and myogenic reactivity, participates CNS Central nervous system
broadly in the process. Finally, a circadian CV Circadian variability
rhythm of BP levels is regulated by a clock CVLM Caudal ventrolateral medulla
mechanism driven by the suprachiasmatic DMV Dorsal motor nucleus of the vagus
nucleus, which is stimulated directly by light. eNOS Nitric oxide synthase
Advances in molecular biology techniques and EP Epinephrine
genetic experimental models continue to pro- ErB Endothelin receptor B
vide more precise information about these GABA Gamma-aminobutyric acid
complex networks. ILM Intermediate lateral medulla
1 Neurohumoral and Autonomic Regulation of Blood Pressure 5

MCA4R Melanocortin neurological (central nervous and autonomic


MR Mineralocorticoid receptor systems), humoral (circulating peptides), barore-
NA Nucleus ambiguus ceptors and chemoreceptors, arteries, heart, and
NEP Norepinephrine kidneys, are crucial to maintain the equilibrium
NO Nitric oxide of the system. In the short-term, cardiac output
NOX NADPH oxidase and peripheral resistance are interconnected by
NPRA Natriuretic peptide receptor A autonomic nervous system reflexes. In the
NPRB Natriuretic peptide receptor B mid-term, vascular peripheral resistance and
NTS Nucleus of tractus solitarius blood volume control are achieved through the
OVLT Organum vasculosum of the lamina interplay between sympathetic nervous system
terminalis activity (SNA), humoral systems, and the kidney.
PRR Prorenin receptor The mission of each of these components men-
PSN Parasympathetic nucleus tioned above, as a sensor or as an effector, is
PVN Paraventricular nucleus regulated and/or modulated by complex organ-
RAAS Renin-angiotensin-aldosterone specific mechanisms (Fig. 2).
system Dysregulation in one or more BP determi-
RGS Regulator of G protein signaling nants produces a response by the system to
RVLM Rostral ventrolateral medulla restore BP values, returning to equilibrium.
SNA Sympathetic nervous activity However, if the dysregulation persists, the feed-
back set point will need to be reset; if it is ele-
vated, a hypertensive state develops. The
Introduction complexity of the interactions among the differ-
ent systems in hypertensive patients or animal
Intravascular pressure, known as blood pressure models is well represented in Page’s mosaic
(BP), is necessary in order to create blood flow concept, which reflects the many interactions of
that transports oxygen and nutrients to body the mechanisms involved in hypertension (Page
organs. Required to achieve this crucial function, 1949). The mosaic theory has been continuously
many components are orchestrated in an updated since Page created it with the addition of
interdependent network. The determinants of new elements due to advances in the knowledge
BP are cardiac output, peripheral resistance, of the physiopathology of BP regulation (Page
and blood volume (Fig. 1). Diverse elements, 1974; Harrison et al. 2021).

Fig. 1 Basic scheme of


main factors controlling BP
and their interactions
6 E. Lurbe and J. Redon

Fig. 2 Determinants,
effectors – sensors and
modulators of BP
regulation

In the 1970s, Guyton and Coleman published


a model of BP regulation that attempted to pro- Neural Components of BP Regulation
vide concepts to explain the mechanism for long-
term BP control, based on the link between blood Both the central nervous system (CNS) and auto-
pressure and sodium balance (Guyton et al. nomic nervous system (ANS) play key roles in BP
1972), and subsequent updates have refined the regulation (Johnson et al. 2015). Generally, while
model. In it, nevertheless, the role of the nervous they integrate multiple afferent and efferent ele-
system was specifically lacking (Guyton 1990). ments, the CNS predominantly commands the
Currently, it is recognized that integrated neuro- level of activation and the ANS predominantly
humoral and autonomic mechanisms are the cen- executes the orders.
tral components that coordinate the control
process even when their activity is modulated
by other relevant physiological and pathological Central Nervous System
processes (Pluznick 2017; Raizada et al. 2017;
Carnagarin et al. 2019; Yang et al. 2020; Madhur Several structures of the CNS are involved in BP
et al. 2021). control, organized as multiple levels of integrative
This chapter reviews the most relevant mecha- centers (Ghali 2017a). Usually, these centers
nisms involved in determining BP levels – effec- respond to signals received through neurogenic
tors and sensors. The anatomy and function of afferent connexons or circulating humoral pep-
each of the elements requires an individual tides from the blood. The integration of all these
description, and the present chapter emphasizes elements results in SNA and vasopressin release
their interplay in driving BP regulation. (Fig. 3).
1 Neurohumoral and Autonomic Regulation of Blood Pressure 7

Fig. 3 Main central and autonomic neural pathways in BP by the interaction of neurons from the lamina terminalis,
regulation. Elements controlling the role of the CNS PVN, RVLM, CVLM, and NTS transmitting signals to the
receiving signals from forebrain structures, baroreceptors, preganglionic fibers of the ILM and then to the ganglionic
chemoreceptors, homeostatic parameters, and organs, ones that innervate vessels and organs. Parasympathetic
through autonomic fibers, cranial nerves, as well as from activity is exerted by the NTS which recruits neurons in the
humoral factors in areas with an absent blood-brain barrier. NA and DNV innervating sinus node and the heart-lungs,
In response, sympathetic and parasympathetic systems respectively. Sympathetic activity and humoral peptides
are activated or blunted. Sympathetic activity is exerted interact both in CNS and in systemic actions (see text).
8 E. Lurbe and J. Redon

Sympathetic Activity (Fry and Ferguson 2021), and with a practically


Activation of the sympathetic nervous system nonexistent blood-brain barrier. Consequently,
produces not only an increase in the heart rate this lack of a blood-brain barrier exposes the neu-
and vasoconstriction of the arterioles, but it also rons to humoral factors – angiotensin II among
increases renin production in the juxtaglomerular others (Leenen 2014) – facilitating their detection
apparatus of the kidney. The most relevant nodes of signals from the circulating blood. Beginning at
and pathways of the SNS include the nucleus of the lamina terminalis, the downstream SNA path-
tractus solitarius (NTS), the rostral (RVLM) and ways involve the PNV, upper cervical spinal cord,
caudal ventrolateral medulla (CVLM), and the and the RVLM. In addition, the SNA pathways
intermedium lateral medulla (ILM). Furthermore, also impact other processes that, in part, contrib-
elements present in the lamina terminalis of the ute to BP regulation, such as thirst, secretion of
third ventricle (circumventricular organ) and antidiuretic hormone, and vascular reactivity in
dorsomedial hypothalamus are also involved in the skin vessels.
SNA (Briant et al. 2016). SNA is generated through different neurotrans-
Although the above nodes and pathways have mitters. Fast SNA is transmitted by mechanisms
direct functions in establishing SNA, the NTS and in which glutamate or gamma-aminobutyric acid
the RVLM play the key roles. The NTS, located in (GABA) binds to ligand-gated ionotropic recep-
the dorsomedial medulla, contains different neu- tors, opening the ion channels of the neurons to
ronal clusters that receive the diverse afferent initiate membrane depolarization and generate the
sensory signals (Martinez and Kline 2021). The potential action in the relevant axon. The response
NTS is the first relay station for general visceral depends on the quantity of presynaptic neuro-
afferents, and it has a critical position in the initi- transmitter, postsynaptic receptors, and the level
ation and integration of a wide variety of reflexes of activity in the intraneuronal signaling path-
controlling cardiovascular and respiratory func- ways. In the mid-term SNA, angiotensinogen
tions, matching the process of tissue perfusion (ANGN) neurons play an important role. In
and pulmonary ventilation. From the NTS, effer- them, angiotensin II (Ang II) is released in the
ent sympathetic fibers go to the RVLM and effer- synaptic cleft and binds to the AT1-receptor
ent parasympathetic (PSN) fibers projected in the (AT1r). This receptor stimulates a G protein sig-
dorsal motor nucleus of the vagus (DMV) and naling pathway with activation of NADPH-
nucleus ambiguus (NA) from which fibers emerge oxidases (NOX) increasing intracellular reactive
to innervate the heart and vessels (Cutsforth- oxygen species (ROS). The final result is the
Gregory and Benarroch 2017; Zanutto et al. activation of several mechanisms and gene
2010). The RVLM integrate information from expressions in the neurons transmitting the signals
PVN, NTS, and baroreflexes (Ghali 2017b). (Leenen et al. 2017). Evidence of the relevance of
The lamina terminalis of the third ventricle, a more slowly acting pathway which maintains
formed by the subfornical organ and the organum the stimulation elicited by Ang II is available
vasculosum of the lamina terminalis (OVLT), nowadays. This slow-acting pathway involves
is a small structure juxtaposed to the third aldosterone (ALD) and the mineralocorticoid
ventricle that has connections to the hypothala- receptor (MR), elements co-localized in the
mus, limbic system, thalamus, and cerebral cortex ANGN neurons.

Fig. 3 (continued) Green arrows show afferent pathways, nucleus ambiguus, NTS, PVN paraventricular nucleus,
and black arrows show efferent pathways. CVLM caudal PSNA parasympathetic nervous activity, RVLM rostral
ventrolateral medulla, DNV dorsal nucleus of the vagus, ventrolateral medulla, SNA sympathetic nervous system
GN ganglionic, ILM intermediolateral medulla, NA activity
1 Neurohumoral and Autonomic Regulation of Blood Pressure 9

Vasopressin ganglions. While preganglionic neurons use ace-


Vasopressin, a potent antidiuretic and vasoconstric- tylcholine (ACh) as their neurotransmitter, the
tor hormone, intervenes in the control of plasma autonomic ganglionic fiber uses ACh or norepi-
volume. It is produced in the PVN and the supra- nephrine (NEP) (Benarroch 2020).
optic nucleus that contain magnocells and
parvocells (Pittman 2021). The magnocells project
to the neurohypophysis which secretes vasopressin Sympathetic Activity
when a reduction in blood volume or changes in
natremia are detected. The parvocells of the PVN The sympathetic preganglionic neurons are
project to the lower brainstem and spinal cord in located in the ILM in the spinal column from
two separate ways. One is the RVLM excitatory thoracic 1 to lumbar 3 segments. With the poten-
pathway, and the other is the CVLM modulatory tial of being activated by the segmental visceral or
pathway of SNA (Koba et al. 2018; Brown et al. somatic afferents, they work in different ways to
2020). In addition to the above mechanisms, stim- mediate sympathetic responses driven by inputs
ulation of neuronal vasopressin-receptors by coming from the hypothalamus and the upper part
hyperosmolarity and stress is implicated in SNA of the medulla. The preganglionic sympathetic
(Carmichael and Wainford 2015). axons arrive at paravertebral, prevertebral, termi-
nal ganglia, and adrenal medulla. Paravertebral
Melanocortin and prevertebral ganglia innervate tissues and
The melanocortin system, MC4R, also appears to organs releasing NEP. The adrenal medulla
modulate SNS activity in response to various releases epinephrine (EP) into the blood (Coote
stimuli that are accompanied by sympathetic over- and Spyer 2018).
activity. The regions with the greatest abundance Norepinephrine and EP stimulate specific recep-
of MC4R are all important sites for the regulation tors (Farzam et al. 2021), the adrenoreceptors α1,2
of autonomic function: PVN, lateral hypothala- and β1,2,3 in the heart, vessels, bronchus, bladder,
mus, amygdala, NTS, DMV, and preganglionic and uterus. Activation of the postsynaptic α1 recep-
sympathetic neurons of the intermediolateral tor by NEP and EP produces vasoconstriction and
medulla (ILM) (do Carmo et al. 2017). The changes in renal hemodynamics, while the α2 pre-
MC4R, induced by leptin in the presence of obe- sents pre- and postsynaptically, reducing the liber-
sity, has been implicated in SNA (da Silva et al. ation of NEP and decreasing SNA. Activation of β1
2019). by EP, which binds with greater affinity than does
NE, increases heart rate, cardiac contractibility, and
drives renin release. Therefore, the effects of SNA
Autonomic Nervous System are the increment of peripheral resistance, cardiac
output, and renin-angiotensin-aldosterone system
The autonomic nervous system, previously (RAAS) activity. In contrast, the β2 receptor, acti-
defined functions, integrates visceral afferent vated by epinephrine, produces mild smooth mus-
inputs with descending influences from forebrain cle relaxation. Although the β3 receptor plays a role
areas. The ANS is involved in homeostasis, emo- in metabolic components, it has none in BP control.
tions, and responses to survival, and its relevance The SNA is of special relevance to BP control
on BP regulation has been acknowledged based in the innervation of the kidney. The sympathetic
on multiple experimental, pharmacologic, and fibers in the kidney, both efferent and afferent with
clinical studies (Dampney 2016). a proportion of approximately 90% and 10%,
The autonomic system is composed of the respectively, penetrate along the renal artery
efferent sympathetic and parasympathetic, the vis- wall. In the kidney ilium, they branch out and
ceral afferents, and the enteric nervous system. penetrate the cortex and the juxtaglomerular area
Both sympathetic and parasympathetic go (Ana Lusch et al. 2014). Renal SNA efferent
through preganglionic neurons and autonomic increases tubular reabsorption of water and
10 E. Lurbe and J. Redon

sodium, renin release, renal vascular resistance, vasodilatation. In parallel, cardiovagal neurons
and it reduces glomerular filtration. The afferent of the NA produce bradycardia. In response to a
signal travels to the CNS boosting SNA. The decrease in pulsatile blood pressure, the afferent
relevance of renal SNA in BP regulation is input of the baroreceptor decreases with an incre-
emphasized by the use of renal denervation to ment in SNA, which results in vasoconstriction
reduce BP values in patients with hypertension and tachycardia due to a reduction in vagal tone.
that is difficult to control (Azizi 2021), even in In addition, as a result of the relationship between
the absence of an antihypertensive drug regimen cardiovagal output and respiration, heart rate
(Mahfoud et al. 2021). decreases during inspiration and increases during
expiration.
A second task integrated with breathing is the
Parasympathetic Activity regulation of tissue perfusion. Cardiac output, BP,
and SNA can be modified according to necessity
In contrast to the widely spread SNS, the PSN has in response to signals coming from the arterial
the preganglionic neurons in the visceral efferent chemoreceptors (Iturriaga et al. 2021). These che-
part of the brainstem and in the sacral spinal moreceptors, located in the carotid body and in the
region (Garamendi-Ruiz and Gómez-Esteban aortic wall, respond mainly to PO2 in the blood
2019). The ganglia are outside or within the wall and less to PCO2 and pH. The generated output
of the target organs in which ACh exerts the signals travel through the glossopharyngeal and
function through the muscarinic receptors, μ1,2,3. vagus nerves to the NTS and the dorsal respira-
The structure of the PSN results in organ-specific tory group, composed of inspiratory neurons
reflexes. Relevant for BP regulation are those located in the medulla. The response, controlled
driven by the vagus nerve which provides pregan- by the RVLM, differentially regulates cardiac out-
glionic innervation to autonomous ganglia in the put, BP, and the perfusion of organs (Guyenet
thorax. Fibers emerging from the DMV innervate et al. 2010). Furthermore, the signals of the carotid
ganglia of the heart and lungs, whereas neurons of body and aortic chemoreceptors interact with sig-
the NA innervate ganglia of the sinus node in the nals from the pCO2 neuron-sensitive in the CNS,
heart with a reduction in heart rate (Standish et al. contributing to the modification of the SNA.
1994).

Humoral Components of BP
Baro- and Chemoreflexes Regulation

The principal mechanism for the short-term con- Besides the activity of the nervous system,
trol of BP is the baroreflex, providing continuous humoral factors also play a key role in BP regula-
buffering of acute changes in BP by sympathetic tion. Neural and humoral mechanisms continu-
vasomotor and cardiovagal outputs (Lauder et al. ously interact; both become either activated or
2020). The arterial baroreceptors are located in the downregulated (Fig. 4). This interaction results
wall of the carotid sinus and the aortic arch, inner- in mid-term control of peripheral resistance and
vated by the glossopharyngeal and the vagus blood volume.
nerves, respectively. Under normal resting condi-
tions, heart rate is regulated by the NA, providing
a tonic vagal activity controlling beat-to-beat in Renin-Angiotensin-Aldosterone
the sinus node of the heart, and the SNA influ- System
ences the peripheral resistance. An increment in
stretch by pulse pressure stimulates the afferent The renin-angiotensin-aldosterone system (RAAS)
synapse on neurons of the NTS initiating a regulates many vital body functions, making it one
response which inhibits the SNA, producing of the key mechanisms in BP regulation. Although
1 Neurohumoral and Autonomic Regulation of Blood Pressure 11

Fig. 4 Main humoral system in BP regulation. Peptides peptides. Shaded blue promotes BP reduction, and shaded
stimulate or decrease diverse mechanisms in the vessels white promotes BP increment. GN ganglionic, ILM
(vasoconstriction/vasodilatation) and in the kidney (diure- intermediolateral medulla, NTS, PVN paraventricular
sis and sodium reabsorption). Angiotensin II activates the nucleus, RVLM rostral ventrolateral medulla, SNA sympa-
SNA which in turn contributes to the activity of systemic thetic nervous activity

the relevance to BP regulation was the first to be The peptides of the system not only produce sys-
recognized, its role in many other physiological temic action as an endocrine achievement but also
processes extends its importance. Continuous work locally in a paracrine/intracrine manner
research has identified both the components of the (Leenen et al. 2017).
system, peptides and receptors, as well as the intra- The initial RAAS components are prorenin and
cellular mechanisms its activation triggers (see renin (RN), produced mainly in the juxtaglomerular
▶ Chap. 3, “Vasoactive Factors and Blood Pressure apparatus in response to a low sodium concentration
in Children”). in the distal tubule or reduction in the renal perfusion
Precursors and enzymes for the formation pressure or increased activity of the SNS. There is a
and degradation of biologically active peptides circulating RAAS and local tissue RAASs. In the
form a complex network ubiquitous in the body. circulating RAAS, once secreted in its inactive
12 E. Lurbe and J. Redon

form, prorenin, binds to the prorenin receptor angiotensin II type 2 receptor (AT2r), and the
(PRR), activating renin, which then cleaves the MAS receptor. Also present is one Ang IV bind-
peptide angiotensin I (Ang I) from angiotensinogen, ing site, a type II transmembrane zinc protein
which is synthesized in the liver. Subsequently, (Karnik et al. 2015). The distribution of these
angiotensin-converting enzyme (ACE1), present in receptors in the CNS is variable. The AT1-recep-
the endothelial pulmonary cells and within multiple tors are present not only in areas poorly protected
tissues, cleaves two amino acids from the by the blood-brain barrier, the circumventricular
C-terminus of Ang I resulting in angiotensin II organs, but also in other areas, such as the supra-
(Ang II), the most active peptide of the RAAS. Of optic, preoptic, and ventral medial nuclei, indicat-
note, ACE1 also reduces bradykinin concentration. ing the relevance of the neuronal synthesis of the
Ang II is further cleaved into other peptides – RAAS elements. The AT2r are located in areas
(i) angiotensin 1–7 (Ang I–7) by the action of the distinct from those of the AT1r (Guimond and
angiotensin-converting enzyme 2 (ACE2), which Gallo-Payet 2012). Moreover, the AngIVr,
cleaves one amino acid from the C-terminus of although similarly distributed as the AT2r, are
Ang II; (ii) angiotensin III (Ang III) by the enzyme also present in the hippocampus, a location at
aminopeptidase A in the CNS; and (iii) angiotensin which no other AT-receptors exist.
IV (Ang IV) a peptide converted from angiotensin The principal effects of RAAS peptides in BP
III by the angiotensin IV receptor (AngIVr) (Fig. 5) regulation are the following:
(Carey 2013; Shu et al. 2021).
The actions elicited by the peptides described • Angiotensin II exerts its action mainly by stim-
above originate in three G protein-coupled recep- ulating the AT1r. This increases intracellular
tors: the angiotensin II type 1 receptor (AT1r), the MAP kinase inducing gene expression,

Fig. 5 Renin-angiotensin-aldosterone system: active pep- angiotensin IV. These four peptides then are able to bind to
tides, enzymes, receptors, and non-active peptides. Pro- three G protein-coupled receptors, AT1r, AT2r, and MASr,
renin binds to the prorenin receptor (PRR), activating and a type II transmembrane zinc protein ATIVr. Angio-
renin, which then cleaves angiotensinogen to produce tensin II binding to AT1r stimulates aldosterone synthesis
angiotensin I, which is then cleaved to angiotensin II by and secretion. Active peptides (black square), enzymes
ACE1. Subsequently, ACE2 cleaves angiotensin II to pro- (light blue square), receptors (dark blue square), and non-
duce angiotensin 1–7, aminopeptidase A produces angio- active peptides (red)
tensin III, which through the receptor ATIVr is converted in
1 Neurohumoral and Autonomic Regulation of Blood Pressure 13

producing vasoconstriction and aldosterone synthesized de novo. Astrocytes are the main
secretion in the cortical layer of the adrenal angiotensinogen source in the brain, and neurons
glands. It regulates water and electrolyte bal- have an intracellular RAAS with membrane-
ance, peripheral resistance, and BP levels. bound receptors, as well as receptors in neuronal
Besides vasoconstriction, Ang II induces mitochondria and nuclei (Sumners et al. 2020;
cellular growth and cell migration, inflamma- Cosarderelioglu et al. 2020).
tion, and oxidative stress by activating other Angiotensin II and Ang III in the adrenal
intracellular mechanisms. Activation of the gland stimulate the synthesis and secretion of
AT2r partly reduces the contraction induced aldosterone, which promotes transepithelial
by the AT1r by inhibiting cell proliferation sodium transport, chloride reabsorption, and
and releasing nitric oxide (NO). Besides its potassium/magnesium secretion in the kidney.
systemic action, Ang II is secreted in the CNS Aldosterone activity is mediated by the activa-
at synaptic clefts, contributing to stimulation of tion of the mineralocorticoid receptor (MR),
the postsynaptic AT1r and consequently boosts ubiquitous in many cellular systems (Bollag
SNA (Hussain and Awan 2018). 2014). Due to its low selectivity, MRs bind not
• Angiotensin 1–7, in contrast to Ang II, through only aldosterone but also cortisol in humans. The
coupling with the MAS-receptor and AT2r, can distribution of this receptor in many organs
restore endothelial function by increasing involved in cardiovascular homeostasis, such as
endothelial nitric oxide synthase (eNOS) and the brain, heart, kidney, and vessels, explains
inhibiting NOX. Additionally, as a partial why, in addition to modifying sodium re-
antagonist of AT1r, Ang 1–7 also contributes absorption, the MR mediates other relevant
to the final effect of vasodilation. Furthermore, physiological actions triggered by cortisol bind-
Ang 1–7 may be a regenerative agent in the ing (Sztechman et al. 2018; please also see
cardiovascular system, since it stimulates ▶ Chap. 2, “Cardiovascular Influences on
endothelial and endothelial-progenitor cells Blood Pressure”). Among these is a prolonged
(Roks et al. 2011; Diz et al. 2011). response in presynaptic neurons of the ILM trig-
• Angiotensin III, through AT1r and AT2r, gered by binding of aldosterone to the MR
increases SNA and vasopressin release, and is (Nakagaki et al. 2012).
more potent than Ang II in the brain (Huber et al.
2017).
• Angiotensin IV is critical for dopamine and Atrial Natriuretic Peptides
acetylcholine release, and appears relevant for
enhancing memory, but not BP regulation Atrial natriuretic peptides (ANPs) are a group of
(Gard 2008). hormones that contribute to BP regulation
throughout the vascular tree, kidney, and neural
The RAAS, as the key humoral component of tissues. After the description of the first ANP, two
BP regulation, exerts its impact by increasing other peptides, the brain natriuretic peptide (BNP)
peripheral resistance through vasoconstriction and C-type natriuretic peptide (CNP), were iden-
and by controlling blood volume through actions tified. While ANP is mainly synthesized in the
in the kidney and in stimulating aldosterone secre- cardiac atria, BNP is synthesized in cardiac ven-
tion (Ames et al. 2019). In the CNS, components tricles, and CNP is synthesized in the
of the RAAS exert both systemic and local endothelial vascular cells and the CNS. Once syn-
actions. Systemic actions of the RAAS are driven thesized, these peptides bind to plasma membrane
by the concentration of Ang II in the blood and receptors, receptor A (NPRA) and B (NPRB).
within the CNS has greatest effect in those areas While NPRA is activated by both ANP and
of the brain in which there is a minimal or nonex- BNP, NPRB is only activated by CNP.
istent blood-brain barrier. Further, a local tissue ANP increases the glomerular filtration rate
RAAS exists in the CNS in which components are and inhibits sodium and water reabsorption in
14 E. Lurbe and J. Redon

the kidney, resulting in natriuresis and diuresis The pressure-natriuresis curve is a mechanism
(Goetze et al. 2020). Moreover, these natriuretic whereby BP elevation increases diuresis and natri-
peptides decrease renin secretion and aldosterone uresis to restore equilibrium (O’Connor and Cow-
synthesis in the adrenal gland, produce relaxation ley 2010). Changes in renal interstitial pressure
of vascular smooth muscle cells, and increase and in proximal tubular sodium transporters
vascular permeability. Thus, they counteract the largely control sodium reabsorption (Palmer and
actions of Ang II and vasopressin (Hodes and Schnermann 2015). Renal medullary blood flow
Lichtstein 2014). Natriuretic peptide receptors is relatively poorly autoregulated, and when kid-
are broadly expressed in the CNS, and their pres- ney perfusion increases, glomerular efferent arte-
ence in the area of the lamina terminalis of the rioles of the juxtamedullary nephrons augment the
third ventricle suggests that the ANPs can be flow to the vasa recta. Consequently, capillary
synthetized by neurons or bound from the circu- hydrostatic pressure increases and is transmitted
lation. Although no central relevant mission in BP to the renal interstitium (Sadowski and Bądzyńska
control has been identified, they seem to modulate 2020). This pressure increment produces a rapid
the activation of Ang II and vasopressin in the reduction in sodium reabsorption in the proximal
CNS (dos Santos Moreira et al. 2017). tubule by internalization of apical Na+/H+
exchanger isoform 3 and the sodium-phosphate
cotransporter along the microvilli of the proximal
The Apelin System tubule. Under normal conditions, the proximal
tubule reabsorbs two-thirds of the filtered sodium
Recently, a novel apelinergic system (AS) with a (McDonough 2010). Concurrently, the RAAS and
putative role in water homeostasis and blood pres- SNA contribute to the dynamic intracellular trans-
sure regulation has been described (Janssens et al. position of the sodium transporters. Further,
2021; Chapman et al. 2021). This system, com- washout of the medullary solute gradient contrib-
posed of the two short-lived peptide ligands, utes to the volume regulation in the distal neph-
apelin and elabela, and a G protein-coupled apelin rons (Franco and Oparil 2006).
receptor, appears to possess activity reciprocal to Excess salt intake is rapidly eliminated by the
that of the RAAS. Apelin receptors (Apr), identi- kidney, although when the capacity is reduced or
fied in endothelial cells, vascular smooth muscle, overload occurs, intravascular volume will
and cardiomyocytes, once stimulated, produce increase. Relatively reduced sodium excretion
arterial and venous vasodilatation (Gupta et al. capacity is present in about 25% of people, cate-
2016). In the kidney, they increase renal blood gorized as salt-sensitivity (Päivä et al. 2006). In
flow and diuresis with mechanisms linked to NO such persons, reduced flow-mediated vasodilation
generation. Vasopressin, apelin, and the receptors is observed as a consequence of several elements –
are co-localized in the hypothalamus; they also enhanced vasoconstriction, decreased capacity to
interact in the CNS. modulate SNA, and increased asymmetric
dimethylarginine (ADMA), an endogenous nitric
oxide synthase inhibitor that reduces NO activity
The Kidney, Fluid Volume, and Salt (Päivä et al. 2006). Sodium accumulation in the
Intake dermal and lymphatic interstitium and other tis-
sues, stored in hypertonic concentrations with pro-
The kidney is the main organ that modulates vol- teoglycans, may activate immune reactions (Wiig
ume homeostasis, in close relation with neurohu- et al. 2013). The putative role of sodium stored in
moral factors. As discussed above, renin secretion this manner is not yet well understood (Chachaj
by the juxtaglomerular apparatus and sympathetic and Szuba 2020).
activation by the efferent and afferent nerves con- Changes in body water/sodium balance are
tribute to BP regulation. The other main contrib- also controlled by ANPs and the CNS. Atrial
utor is the pressure-natriuresis curve. natriuretic peptide induces natriuresis and diuresis
1 Neurohumoral and Autonomic Regulation of Blood Pressure 15

by increasing the glomerular filtration rate and vasoconstrictors (Jackson 2017) and SNA (Martí-
inhibiting sodium reabsorption, thereby nez et al. 2009).
counteracting the activity of the RAAS (Bie and Endothelin-1, produced and released by the
Evans 2017). Through fibers from the lamina endothelial cells, in low levels stimulates the
terminalis and the VIIth, IXth, and Xth cranial endothelin receptor B (ErB), which in turn
nerves, the CNS receives information about extra- increases the synthesis of vasodilatory NO and
cellular fluid osmolarity, sodium concentration, prostacyclin PGI2, relevant to limiting the vaso-
volume receptors, arterial/cardiopulmonary baro- constriction produced for the stimulation of endo-
receptors, sense of taste, and BP. Once integrated thelin receptor A (Kostov 2021).
in the NTS, this information triggers appropriate The vasodilatory capacity of arterial vessels
sympathetic, endocrine, and behavioral responses. differs, depending on their structure: large arter-
ies, branched or arteriolar. While the large vessels
utilize preferent NO, the arteriolar ones use the
The Vasculature and Nitric Oxide endothelium-dependent hyperpolarization factor,
which through the myoendothelial junctions is
The vascular structure and function of arteries and transmitted to the smooth muscle cells (Lemmey
arteriolar vessels are key elements of BP regula- et al. 2020). Flow-mediated vasodilatation is an
tion. Peripheral resistance in the vasculature is endothelial mechanism which involves generation
determined by the caliber, reactivity, and elasticity of NO by eNOS, prostacyclin, and the opening of
of vessels. Autonomic activation, circulating calcium-sensitive potassium channels (Tomiyama
humoral factors, and mechanical forces generate et al. 2017). Laminar flow elongates fibers in the
an environment in which endothelial and smooth direction of flow and activates mechano-sensors,
muscle cells establish the caliber of the vessels. G protein-coupled receptors, in the endothelial
Additionally, changes in the muscular layer and surface (Hu et al. 2021). In contrast, turbulent
extracellular matrix of vessels form the basis of flow, which is generated in bifurcations, activates
vascular remodeling, which increases peripheral inflammatory pathways (Amaya et al. 2015).
resistance. The vasodilatation induced by NO can be
Beyond responses to various extravascular blunted by the production of reactive oxygen
stimuli, the endothelium modulates vascular tone species (ROS) and by the presence of
by synthesizing and releasing vasoactive factors asymmetric-dimethylarginine (ADMA) (Tsikas
with vasodilatory capacity (NO, prostacyclin, 2020). Activation of NOX by Ang II, in the vessels,
hyperpolarizing factor, low level endothelin-1) or produces superoxide and other ROS, inactivating
through vasoconstriction (thromboxane A2, high NO and the cofactor for NO synthase, tetra-
level endothelin-1). Peripheral resistance increases hydrobiopterin, producing vasoconstriction (Pautz
due to the action of circulating Ang II, catechol- et al. 2021). The impact of ROS from other sources
amines, vasopressin, high levels of endothelin-1, is not relevant for BP regulation. An endogenous
and thromboxane A2. This is modulated by the competitive inhibitor of NO synthase, the post-
intracellular machinery present in the endothelial translational modification of arginine, ADMA,
and small muscle cells. In endothelial cells, the reduces the vasodilatory capacity (Jankovic et al.
small GTPase RhoA and the downstream target 2017; Poeggeler et al. 2021).
Rho kinase reduces the bioavailability of NO, Beside the flow-induced vasodilatation,
favoring vasoconstriction. On the other hand, in smooth muscle cells are able to change the status
the smooth muscle cells, the G signaling proteins of vasoconstriction or vasodilatation in response
(RGS) 1 and 2, and the RhoA increase vasocon- to intravascular pressure. In this myogenic reac-
striction. Furthermore, the activation of potassium tivity, several mechanisms have been implicated
channels (Shvetsova et al. 2021) produces cellular (Kim and Hong 2021), among them the role of a
hyperpolarization, increasing vasorelaxation and variety of transient receptor potential channels
counteracting contraction induced by different (Liu et al. 2021) which are in charge of
16 E. Lurbe and J. Redon

maintaining the resting membrane potential and induces a circadian secretion of neurotransmitters,
the intracellular calcium dynamics in smooth stimulates SNA, and reduces PSNA activity. The
muscle cells. gears of the clock are composed of activators,
CLOCK and BMAL1, that induce the expression
of their own repressors, PER and CRY, forming a
Circadian Variation of Blood Pressure negative feedback loop (Allada and Bass 2021). The
duration of the stimulus is regulated post-
Recent evidence supports the concept that cardio- translationally, including phosphorylation by
vascular and kidney function have variations kinases. Once astrocytes become activated, they
through the circadian clock. Under normal condi- synchronize the peripheral clock genes located
tions, BP rises during the morning, starting to mainly in the kidney, brain, heart, and vessels
increase about 1 h prior to awakening, reaching resulting in rhythms of clock gene expression
its highest level later, which is maintained until (Patke et al. 2020). This system regulates the diurnal
late afternoon. Thereafter, BP decreases progres- rhythm of many biochemical pathways with the
sively until its nadir, around 3:00 AM (Costello rhythmic production/secretion of peptides and hor-
and Gumz 2021). Circadian variation (CV) in the mones with cardiovascular and renal actions
brain, heart, kidney, and vessels prepares the tran- (Lecarpentier et al. 2020).
sition from sleep to activity. This pattern seems
independent of an individual’s sleep/wake or
fasting/feeding patterns, although recent studies Conclusion
have challenged the total independence of
CV. Physical activity impacts CV, increasing BP Blood pressure control involves the interaction of
during activity and decreasing it with night’s rest multiple systems with a close link between neu-
(Bass and Lazar 2016). The presence of rohumoral and autonomic mechanisms to main-
hypertension-mediated organ damage, mainly in tain in equilibrium the main BP determinants,
the kidney, blunts the physiological BP nocturnal cardiac output, peripheral resistance, and blood
fall (Redon and Lurbe 2008). volume. Overall, the CNS paces the activity of
Circadian rhythm in the levels of peptides the different mechanisms implicated, processing
(melatonin, plasma renin activity, ACE activity, and responding to stimuli coming from both cor-
Ang II, aldosterone, ANP, NO, endothelin-1) with tical, subcortical, and visceral signals, as well as
activity on BP regulation around the clock has from circulating humoral factors. Autonomic
been described (Zhang et al. 2021). Melatonin is functions, coupled with humoral peptides, exe-
inhibited by light; plasma renin and ACE activity, cute the orders mainly in the vessels, kidney, and
Ang II and aldosterone peak just before the usual heart, which themselves possess complex mecha-
time of awakening; ANP is antiphase to BP; and nisms of autoregulation that modulate the
NO increases during wakefulness, the early stages received inputs. Advances in molecular biology
of sleep deprivation and the diurnal increment of techniques and genetic experimental models con-
endothelin-1. If these changes are in response to tinue to provide more precise information about
BP oscillation or play a relevant role in the circa- the intracellular signals that regulate cellular com-
dian variability it is not well established for some munication among these many systems.
of them.
In mammals, CV control is located in the supra-
chiasmatic nucleus (SCN) which is stimulated Cross-References
directly by light through the retinal hypothalamic
tract (Dibner et al. 2010). Astrocytes of the SCN ▶ Cardiovascular Influences on Blood Pressure
generate a complex light-induced mechanism ▶ Vasoactive Factors and Blood Pressure in
involving glutamate as the neurotransmitter, which Children
1 Neurohumoral and Autonomic Regulation of Blood Pressure 17

References of apelin in kidney disease. Nat Rev Nephrol 13:1–14.


https://doi.org/10.1038/s41581-021-00461-z
Allada R, Bass J (2021) Circadian mechanisms in medi- Coote JH, Spyer KM (2018) Central control of autonomic
cine. N Engl J Med 384:550–561 function. Brain Neurosci Adv 13:2398212818812012.
Amaya R, Pierides A, Tarbell JM (2015) The interaction https://doi.org/10.1177/2398212818812012
between fluid wall shear stress and solid circumferential Cosarderelioglu C, Nidadavolu LS, George CJ, Oh ES,
strain affects endothelial gene expression. PLoS One 10: Bennett DA, Walston JD, Abadir PM (2020) Brain
e0129952. https://doi.org/10.1371/journal.pone.0129952 renin-angiotensin system at the intersect of physical
Ames MK, Atkins CE, Pitt B (2019) The renin- and cognitive frailty. Front Neurosci 14:586314.
angiotensin-aldosterone system and its suppression. https://doi.org/10.3389/fnins.2020.586314
J Vet Intern Med 33:363–382 Costello HM, Gumz ML (2021) Circadian rhythm, clock
Ana Lusch A, Leary R, Heidari E, Liss MA, Okhunov Z, genes, and hypertension: recent advances in hyperten-
Perez-Lanzac De Lorca A, Huang J, Wikenheiser J, sion. Hypertension 78:1185–1196
Landman J (2014) Intrarenal and extrarenal autonomic Cutsforth-Gregory JK, Benarroch EE (2017) Nucleus of
nervous system redefined. J Urol 191:1060–1065 the solitary tract, medullary reflexes, and clinical impli-
Azizi M, Sanghvi K, Saxena M, Gosse P, Reilly JP, Levy T, cations. Neurology 88:1187–1196
Rump LC, Persu A, Basile J, Bloch MJ, Daemen J, Lobo da Silva AA, do Carmo JM, Wang Z, Hall JE (2019)
MD, Mahfoud F, Schmieder RE, Sharp ASP, Weber Melanocortin-4 receptors and sympathetic nervous sys-
MA, Sapoval M, Fong P, Pathak A, Lantelme P, Hsi D, tem activation in hypertension. Curr Hypertens Rep 21:
Bangalore S, Witkowski A, Weil J, Kably B, Barman 46–53
NC, Reeve-Stoffer H, Coleman L, McClure CK, Kirtane Dampney RA (2016) Central neural control of the cardio-
AJ, RADIANCE-HTN Investigators (2021) Ultrasound vascular system: current perspectives. Adv Physiol
renal denervation for hypertension resistant to a triple Educ 40:283–296
medication pill (RADIANCE-HTN TRIO): a Dibner C, Schibler U, Albrecht U (2010) The mammalian
randomised, multicentre, single-blind, sham-controlled circadian timing system: organization and coordination of
trial. Lancet 397:2476–2486 central and peripheral clocks. Annu Rev Physiol 72:
Bass J, Lazar MA (2016) Circadian time signatures of 517–549
fitness and disease. Science 354:994–999 Diz DI, Arnold AC, Nautiyal M, Isa K, Shaltout HA, Tallant
Benarroch EE (2020) Physiology and pathophysiology of EA (2011) Angiotensin peptides and central autonomic
the autonomic nervous system. Continuum 26:12–24 regulation. Curr Opin Pharmacol 11:131–137
Bie P, Evans RG (2017) Normotension, hypertension and do Carmo JM, da Silva AA, Wang Z, Fang T, Aberdein N,
body fluid regulation: brain and kidney. Acta Physiol Perez de Lara CE, Hall JE (2017) Role of the
219:288–304 brain melanocortins in blood pressure regulation.
Bollag WB (2014) Regulation of aldosterone synthesis and Biochim Biophys Acta Mol Basis Dis 1863(10
secretion. Compr Physiol 4:1017–1055 Pt A):2508–2514
Briant LJ, Charkoudian N, Hart EC (2016) Sympathetic dos Santos Moreira MC, Naves LM, Marques SM, Silva EF,
regulation of blood pressure in normotension and hyper- Rebelo AC, Colombari E, Pedrino GR (2017) Neuronal
tension: when sex matters. Exp Physiol 101:219–229 circuits involved in osmotic challenges. Physiol Res 66:
Brown CH, Ludwig M, Tasker JG, Stern JE (2020) Somato- 411–423
dendritic vasopressin and oxytocin secretion in endo- Farzam K, Kidron A, Lakhkar AD (2021) Adrenergic
crine and autonomic regulation. J Neuroendocrinol drugs. In: StatPearls [Internet]. StatPearls Publishing,
32(6):e12856. https://doi.org/10.1111/jne.12856 Treasure Island
Carey RM (2013) Newly discovered components and Franco V, Oparil S (2006) Salt sensitivity, a determinant of
actions of the renin–angiotensin system. Hypertension blood pressure, cardiovascular disease and survival.
62:818–822 J Am Coll Nutr 25(3 Suppl):247S–255S
Carmichael CY, Wainford RD (2015) Hypothalamic sig- Fry WM, Ferguson AV (2021) The subfornical organ and
naling mechanisms in hypertension. Curr Hypertens organum vasculosum of the lamina terminalis: critical
Rep 17:39–48 roles in cardiovascular regulation and the control of
Carnagarin R, Matthews V, Zaldivia MTK, Peter K, fluid balance. Handb Clin Neurol 180:203–215
Schlaich MP (2019) The bidirectional interaction Garamendi-Ruiz I, Gómez-Esteban JC (2019) Cardiovas-
between the sympathetic nervous system and immune cular autonomic effects of vagus nerve stimulation.
mechanisms in the pathogenesis of hypertension. Br Clin Auton Res 29:183–194
J Pharmacol 176:1839–1852 Gard PR (2008) Cognitive-enhancing effects of angioten-
Chachaj A, Szuba A (2020) Skin lymphatic system in the sin IV. BMC Neurosci 9(Suppl 2):S15–S21
pathogenesis of arterial hypertension – review and cri- Ghali MGZ (2017a) The brainstem network controlling
tique. Lymphology 53:99–108 blood pressure: an important role for pressor sites in
Chapman FA, Nyimanu D, Maguire JJ, Davenport AP, the caudal medulla and cervical spinal cord.
Newby DE, Dhaun N (2021) The therapeutic potential J Hypertens 35:1938–1947
18 E. Lurbe and J. Redon

Ghali MGZ (2017b) Role of the medullary lateral tegmen- blood pressure and hypertension. Am J Physiol Regul
tal field in sympathetic control. J Integr Neurosci 16: Integr Comp Physiol 309:R1309–R1325
189–208 Karnik SS, Unal H, Kemp JR, Tirupula KC, Eguchi S,
Goetze JP, Bruneau BG, Ramos HR, Ogawa T, de Bold Vanderheyden PM, Thomas WG (2015) International
MK, de Bold AJ (2020) Cardiac natriuretic peptides. Union of Basic and Clinical Pharmacology. XCIX.
Nat Rev Cardiol 17:698–717 Angiotensin receptors: interpreters of pathophysiolog-
Guimond MO, Gallo-Payet N (2012) The angiotensin II ical angiotensinergic stimuli [corrected]. Pharmacol
type 2 receptor in brain functions: an update. Int Rev 67:754–819
J Hypertens 2012:351758. https://doi.org/10.1155/ Kim K, Hong KS (2021) Transient receptor potential
2012/351758 channel-dependent myogenic responsiveness in small-
Gupta MD, Girish MP, Shah D, Rain M, Mehta V, Tyagi S, sized resistance arteries. J Exerc Rehabil 17:4–10
Trehan V, Pasha Q (2016) Biochemical and genetic role Koba S, Hanai E, Kumada N, Kataoka N, Nakamura K,
of apelin in essential hypertension and acute coronary Watanabe T (2018) Sympathoexcitation by hypotha-
syndrome. Int J Cardiol 15(223):374–378 lamic paraventricular nucleus neurons projecting to
Guyenet PG, Stornetta RL, Bayliss DA (2010) Central the rostral ventrolateral medulla. J Physiol 596:
respiratory chemoreception. J Comp Neurol 518: 4581–4595
3883–3906 Kostov K (2021) The causal relationship between
Guyton AC (1990) Long-term arterial pressure control: an endothelin-1 and hypertension: focusing on endothelial
analysis from animal experiments and computer and dysfunction, arterial stiffness, vascular remodeling, and
graphic models. Am J Physiol Regul Integr Comp blood pressure regulation. Life 11:986–932
Physiol 28:R865–R877 Lauder L, Azizi M, Kirtane AJ, Böhm M, Mahfoud F
Guyton AC, Coleman TG, Granger HJ (1972) Circulation: (2020) Device-based therapies for arterial hyperten-
overall regulation. Annu Rev Physiol 34:13–46 sion. Nat Rev Cardiol 17:614–628
Harrison DG, Coffman TM, Wilcox CS (2021) Pathophys- Lecarpentier Y, Schussler O, Hébert JL, Vallée A (2020)
iology of hypertension: the mosaic theory and beyond. Molecular mechanisms underlying the circadian
Circ Res 128:847–863 rhythm of blood pressure in normotensive subjects.
Hodes A, Lichtstein D (2014) Natriuretic hormones in Curr Hypertens Rep 22:50–63
brain function. Front Endocrinol 5:201. https://doi. Leenen FH (2014) Actions of circulating angiotensin II and
org/10.3389/fendo.2014.00201 aldosterone in the brain contributing to hypertension.
Hu Y, Chen M, Wang M, Li X (2021) Flow-mediated Am J Hypertens 27:1024–1032
vasodilation through mechanosensitive G protein- Leenen FHH, Blaustein MP, Hamlyn JM (2017) Update on
coupled receptors in endothelial cells. Trends angiotensin II: new endocrine connections between the
Cardiovasc Med S1050-1738(21):1–3. https://doi.org/ brain, adrenal glands and the cardiovascular system.
10.1016/j.tcm.2020.12.010 Endocr Connect 6:R131–R145
Huber G, Schuster F, Raasch W (2017) Brain renin- Lemmey HAL, Garland CJ, Dora KA (2020) Intrinsic
angiotensin system in the pathophysiology of cardio- regulation of microvascular tone by myoendothelial
vascular diseases. Pharmacol Res 125(Pt A):72–90 feedback circuits. Curr Top Membr 85:327–355
Hussain M, Awan FR (2018) Hypertension regulating Liu L, Guo M, Lv X, Wang Z, Yang J, Li Y, Yu F, Wen X,
angiotensin peptides in the pathobiology of cardiovas- Feng L, Zhou T (2021) Role of transient receptor
cular disease. Clin Exp Hypertens 40:344–352 potential vanilloid 4 in vascular function. Front Mol
Iturriaga R, Alcayaga J, Chapleau MW, Somers VK (2021) Biosci 8:677661. https://doi.org/10.3389/fmolb.2021.
Carotid body chemoreceptors: physiology, pathology, 677661
and implications for health and disease. Physiol Rev Madhur MS, Elijovich F, Alexander MR, Pitzer A,
101:1177–1235 Ishimwe J, Van Beusecum JP, Patrick DM, Smart CD,
Jackson WF (2017) Potassium channels in regulation of Kleyman TR, Kingery J, Peck RN, Laffer CL, Kirabo A
vascular smooth muscle contraction and growth. Adv (2021) Hypertension: do inflammation and immunity
Pharmacol 78:89–144 hold the key to solving this epidemic? Circ Res 128:
Jankovic A, Korac A, Buzadzic B, Stancic A, Otasevic V, 908–933
Ferdinandy P et al (2017) Targeting the NO/superoxide Mahfoud F, Bloch MJ, Azizi M, Wang Y, Schmieder RE,
ratio in adipose tissue: relevance to obesity and diabetes Lobo MD, Sharp ASP, Daemen J, Basile J, Weber MA,
management. Br J Pharmacol 174:1570–1590 Scicli AP, McClure CK, Kirtane AJ (2021) Changes in
Janssens P, Decuypere JP, Bammens B, Llorens-Cortes C, blood pressure after crossover to ultrasound renal
Vennekens R, Mekahli D (2021) The emerging role of denervation in patients initially treated with sham in
the apelinergic system in kidney physiology and dis- the RADIANCE-HTN SOLO trial. EuroIntervention.
ease. Nephrol Dial Transplant 2021:gfab070. https:// https://doi.org/10.4244/EIJ-D-21-00295
doi.org/10.1093/ndt/gfab070 Martinez D, Kline DD (2021) The role of astrocytes in the
Johnson AK, Zhang Z, Clayton SC, Beltz TG, Hurley SW, nucleus tractus solitarii in maintaining central control
Thunhorst RL, Xue BL (2015) The roles of sensitiza- of autonomic function. Am J Physiol Regul Integr
tion and neuroplasticity in the long-term regulation of Comp Physiol 320:R418–R424
1 Neurohumoral and Autonomic Regulation of Blood Pressure 19

Martínez AC, Pagán RM, Prieto D, Recio P, García- Roks AJ, Rodgers K, Walther T (2011) Effects of the renin
Sacristán A, Hernández M, Benedito S (2009) Modu- angiotensin system on vasculogenesis-related progeni-
lation of noradrenergic neurotransmission in isolated tor cells. Curr Opin Pharmacol 11:162–174
rat radial artery. J Pharmacol Sci 111:299–311 Sadowski J, Bądzyńska B (2020) Altered renal medullary
McDonough AA (2010) Mechanisms of proximal tubule blood flow: a key factor or a parallel event in control of
sodium transport regulation that link extracellular fluid sodium excretion and blood pressure? Clin Exp
volume and blood pressure. Am J Physiol Regul Integr Pharmacol Physiol 47:1323–1332
Comp Physiol 298:R851–R861 Shu Z, Wan J, Read RJ, Carrell RW, Zhou A (2021)
Nakagaki T, Hirooka Y, Matsukawa R, Nishihara M, Angiotensinogen and the modulation of blood pressure.
Nakano M, Ito K, Hoka S, Sunagawa K (2012) Acti- Front Cardiovasc Med 8:645123. https://doi.org/10.
vation of mineralocorticoid receptors in the rostral ven- 3389/fcvm.2021.645123
trolateral medulla is involved in hypertensive Shvetsova AA, Gaynullina DK, Tarasova OS, Schubert R
mechanisms in stroke-prone spontaneously hyperten- (2021) Remodeling of arterial tone regulation in post-
sive rats. Hypertens Res 35:470–476 natal development: focus on smooth muscle cell potas-
O’Connor PM, Cowley AW Jr (2010) Modulation of sium channels. Int J Mol Sci 22:5413–5425
pressure-natriuresis by renal medullary reactive oxygen Standish A, Enquist LW, Schwaber JS (1994) Innervation
species and nitric oxide. Curr Hypertens Rep 12:86–92 of the heart and its central medullary origin defined by
Page IH (1949) Pathogenesis of arterial hypertension. J Am viral tracing. Science 263:232–234
Med Assoc 140:451–458 Sumners C, Alleyne A, Rodriguez V, Pioquinto DJ, Ludin
Page IH (1974) Arterial hypertension in retrospect. Circ JA, Kar S, Winder Z, Ortiz Y, Liu M, Krause EG, Kloet
Res 34:133–142 AD (2020) Brain angiotensin type-1 and type-2 recep-
Päivä H, Laakso J, Kähönen M, Turjanmaa V, Kööbi T, tors: cellular locations under normal and hypertensive
Majahalme S, Lehtimäki T, Ruokonen I, Laaksonen R conditions. Hypertens Res 43:281–295
(2006) Asymmetric dimethylarginine and hemodynamic Sztechman D, Czarzasta K, Cudnoch-Jedrzejewska A,
regulation in middle-aged men. Metabolism 55:771–777 Szczepanska-Sadowska E, Zera T (2018) Aldosterone
Palmer LG, Schnermann J (2015) Integrated control of Na and mineralocorticoid receptors in regulation of the
transport along the nephron. Clin J Am Soc Nephrol 10: cardiovascular system and pathological remodelling
e676–e687 of the heart and arteries. J Physiol Pharmacol 69(6).
Patke A, Young MW, Axelrod S (2020) Molecular mech- https://doi.org/10.26402/jpp.2018.6.01
anisms and physiological importance of circadian Tomiyama H, Saisu T, Yamashina A (2017) Measurement
rhythms. Nat Rev Mol Cell Biol 21:67–84 of flow-mediated vasodilatation. Int Heart J 58:
Pautz A, Li H, Kleinert H (2021) Regulation of NOS 158–162
expression in vascular diseases. Front Biosci 26:85–101 Tsikas D (2020) Urinary dimethylamine (DMA) and its
Pittman QJ (2021) Vasopressin and central control of the precursor asymmetric dimethylarginine (ADMA) in
cardiovascular system: a 40-year retrospective. clinical medicine, in the context of nitric oxide
J Neuroendocrinol 33:e13011. https://doi.org/10.1111/ (NO) and beyond. J Clin Med 9:1843–1863
jne.13011 Wiig H, Schröder A, Neuhofer W, Jantsch J, Kopp C,
Pluznick JL (2017) Microbial short-chain fatty acids and Karlsen TV, Boschmann M, Goss J, Bry M,
blood pressure regulation. Curr Hypertens Rep 19: Rakova N, Dahlmann A, Brenner S, Tenstad O,
25–30 Nurmi H, Mervaala E, Wagner H, Beck FX, Müller
Poeggeler B, Robenek H, Pappolla MA (2021) Editorial. DN, Kerjaschki D, Luft FC, Harrison DG, Alitalo K,
Pharmacology of L-arginine and L-arginine-rich food. Titze J (2013) Immune cells control skin lymphatic
Front Pharmacol 12:743788. https://doi.org/10.3389/ electrolyte homeostasis and blood pressure. J Clin
fphar.2021.743788 Invest 123:2803–2815
Raizada MK, Joe B, Bryan NS, Chang EB, Dewhirst FE, Yang F, Chen H, Gao Y, An N, Li X, Pan X, Yang X,
Borisy GG, Galis ZS, Henderson W, Jose PA, Ketchum Tian L, Sun J, Xiong X, Xing Y (2020) Gut microbiota-
CJ, Lampe JW, Pepine CJ, Pluznick JL, Raj D, Seals derived short-chain fatty acids and hypertension: mech-
DR, Gioscia-Ryan RA, Tang WHW, Oh YS (2017) anism and treatment. Biomed Pharmacother 130:
Report of the National Heart, Lung, and Blood Institute 110503–110518
Working Group on the role of microbiota in blood Zanutto BS, Valentinuzzi ME, Segura ET (2010) Neural set
pressure regulation: current status and future directions. point for the control of arterial pressure: role of the
Hypertension 2017(70):479–485 nucleus tractus solitarius. Biomed Eng Online 9:4–17
Redon J, Lurbe E (2008) Nocturnal blood pressure versus Zhang J, Sun R, Jiang T, Yang G, Chen L (2021) Circadian
nondipping pattern: what do they mean? Hypertension blood pressure rhythm in cardiovascular and renal
51:41–42 health and disease. Biomolecules 11:868–882
Cardiovascular Influences on Blood
Pressure 2
Manish D. Sinha and Phil Chowienczyk

Contents
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
Relevance of BP Level During Childhood . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
Hemodynamic Phenotypes of Primary Hypertension and Their Clinical
Relevance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
Components of Blood Pressure: Static and Pulsatile . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 23
Pulsatile Components of Blood Pressure . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25
Physiological Changes in CO, SVR, and HR During Childhood . . . . . . . . . . . . . . . . . . . . 26
Evidence for a “Hyperdynamic State” in Hypertension . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27
Evidence of a Vascular Change in the Early Phase of Hypertension . . . . . . . . . . . . . . . 28
Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 35
Cross-References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 36
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 36

Abstract

The study of hemodynamic mechanisms in


children and young people with primary hyper-
tension has been neglected historically. An
understanding of the abnormalities in the func-
M. D. Sinha (*) tion of the large arteries, the left ventricle, and
Department of Paediatric Nephrology, Evelina London
their interaction may be important to elucidate
Children’s Hospital, Guys & St Thomas’ NHS Foundation
Trust, London, UK the development and observed effects of
hypertension. In this chapter, we discuss car-
Kings College London British Heart Foundation Centre,
London, UK diovascular influences that determine blood
e-mail: manish.sinha@nhs.net pressure; highlighting the static and pulsatile
P. Chowienczyk components of blood pressure and review the
Kings College London British Heart Foundation Centre, published literature evaluating hemodynamics
London, UK in children with hypertension. We compare
e-mail: phil.chowienczyk@kcl.ac.uk

© Springer Nature Switzerland AG 2023 21


J. T. Flynn et al. (eds.), Pediatric Hypertension,
https://doi.org/10.1007/978-3-031-06231-5_3
22 M. D. Sinha and P. Chowienczyk

findings in hypertensive children with those hypertension in adulthood (Carrico et al. 2013;
with normotension. Increasing data support a Chen and Wang 2008; Tirosh et al. 2010). Recent
cardiac and large artery component in early population-based data suggest that hypertension
hypertension in children and young people. during adulthood and its associated adverse car-
Increase in cardiac output appears to be the diovascular (CV) outcomes has its origin in ele-
earliest identifiable abnormality in children vated BP during adolescence (Lurbe et al. 2016).
and young people (CYP) with increased sys- Similarly, a recent systematic review confirmed
temic vascular resistance (SVR) in hyperten- that elevated BP in childhood or adolescence is
sive young adults. Given the tracking of associated with several intermediate markers and
hypertension from children to adults, the find- hard outcomes of cardiovascular disease in adult-
ing of a cardiac/aortic rather than peripheral hood (Yang et al. 2020). Convincing evidence
vascular changes associated with primary regarding the consequences of childhood blood
hypertension has implications for the etiology pressure trajectories on adult outcomes was
of hypertension both in children and adults. It recently demonstrated by the Bogalusa Heart
also has implications for the best treatment in Study investigators, who observed that persis-
children. tently high BP particularly through adolescence
associated with higher risk for adult left ventricu-
lar hypertrophy (LVH) when compared with nor-
Introduction mal blood pressure (Zhang et al. 2018). The
authors reported that these associations remained
Primary hypertension (PH) is a major health bur- consistent across race and sex groups, with stron-
den globally and one of the main contributors to ger relationship of concentric LVH with adoles-
excess morbidity and mortality, accounting for a cent BP trajectories when compared with
1/3rd of ischemic heart disease and 2/3rds of eccentric LVH (Zhang et al. 2018). Overall, evi-
strokes among adults (Egan et al. 2019). In chil- dence suggests an increasing prevalence of PH in
dren and young people (CYP), there is an increas- CYP, tracking of BP from childhood to adulthood,
ing prevalence of PH, mirroring the childhood and increased risk of adverse cardiovascular out-
obesity epidemic, with a steady increase over the comes as a result. These observations highlight
last two decades (Litwin and Kułaga 2021; Lurbe the need to improve our understanding of the
et al. 2016; Song et al. 2019). Thus, although the potential mechanisms underlying early increase
prevalence of PH is reported between 3% and 5% in blood pressure in children and adolescents.
in those aged <18 years, during adolescence prev-
alence rates are reported to increase to 10–11%,
similar to reports in young in young adults (Flynn Hemodynamic Phenotypes of Primary
et al. 2017; Litwin and Kułaga 2021; McNiece Hypertension and Their Clinical
et al. 2007; Noubiap et al. 2017; Sharma et al. Relevance
2018; Symonides et al. 2010; Urbina et al. 2019a).
Nearly two decades ago, Sorof and colleagues
reported that isolated systolic hypertension (ISH)
Relevance of BP Level During was the predominant hypertension phenotype in
Childhood 12–16-year-old adolescents, affecting 88% of
youth with hypertension (Sorof et al. 2002). In
In childhood PH, despite only modestly elevated those with ISH, most were obese and had increased
blood pressure, hypertension-mediated organ heart rates and BP variability (Sorof et al. 2002;
damage, including left ventricular hypertrophy Sorof 2002). Isolated diastolic hypertension (IDH),
and arteriopathy, can be detected (Azukaitis in contrast, was less common (Sorof 2002).
et al. 2021; Urbina et al. 2019b). Sustained hyper- Recent data from the United States evaluating the
tension in childhood is also a strong predictor of National Health and Nutrition Examination Survey
2 Cardiovascular Influences on Blood Pressure 23

(NHANES, 1999–2016) including over 17,000 chil- It is likely, therefore, that there are different
dren aged 8–18 reported the prevalence of ISH at hemodynamic mechanisms contributing to the
11.1%, but IDH nearly tenfold lower (1.9%) development of hypertension across the age spec-
(Alsaeed et al. 2021). Children with IDH were trum with important differences in clinical out-
more likely to be younger, female, white, and comes. An improved understanding of the
leaner with lower rates of excess weight than underlying mechanisms will help plan appropriate
those with ISH (Alsaeed et al. 2021). In the intervention and follow-up of CYP with elevated
ENIGMA study of ~900 University students in BP and prevent future adverse outcomes.
the United Kingdom, aged 20–27 years, ISH was
described in 8% and the combination of systolic
and diastolic hypertension (SDH) in 4% Components of Blood Pressure: Static
(McEniery et al. 2005). In contrast, the phenotype and Pulsatile
of hypertension, in adults, is strikingly different
with IDH predominating in young adults aged Typically, we measure blood pressure and express
<40 years and ISH predominating among it as “systolic BP,” which relates to the pressure in
middle-aged and elderly hypertensive persons the artery during ventricular contraction, and
(Franklin et al. 2001). The clinical relevance of “diastolic BP,” which relates to the pressure in
the hypertension phenotype was shown clearly by the artery during ventricular relaxation. A more
the Chicago Heart Study Investigators (Yano et al. physiological description would be to express the
2015). In their study, which included >35,000 changes in the measured arterial pressure as
persons, over a follow-up period of 31 years, “static” and “pulsatile” components (Laosiripisan
ISH was reported in 25% of men and 13% of et al. 2017; Roman and Devereux 2014; Tanaka
women, with a prevalence of ISH in younger et al. 2016).
adults up to 50% (Yano et al. 2015). The relative Thus, mean arterial pressure (MAP) represents
risk of mortality from cardiovascular and coro- the steady-state or static BP. It can be regarded as
nary heart disease in adults with ISH was higher the perfusion pressure driving blood flow to the
than in those with normal BP (BP <130/85 mm organs and is dependent on cardiac output (CO)
Hg) (Yano et al. 2015). and the total systemic peripheral vascular resis-
ISH in older adults is widely thought to result tance (SVR), a relationship, represented by the
from “stiffening” of the aorta and large arteries familiar equation of MAP ¼ CO  SVR [31].
secondary to an age-related degenerative process Accordingly, elevated MAP may be a result of
(Franklin et al. 2001). This vascular change leads to increase in CO, in SVR, or in both (Fig. 1).
a widening of pulse pressure (PP, the difference Cardiac output, which represents pump func-
between systolic and diastolic BP) as discussed tion of the heart, is, in turn, the product of heart
below. Aging is associated with gradual physiolog- rate (HR) and stroke volume (SV):
ical changes in arterial structure and function with
arterial stiffening observed even in non-hyperten- CO ¼ HR  SV:
sive elderly persons (Najjar et al. 2005). Hyperten-
sion may also contribute to arterial stiffening as a Heart rate is determined by the relative influ-
consequence of damage to the elastic component ences of sympathetic vs. parasympathetic nerve
of the artery following sustained elevated BP fibers at the sinoatrial node in the heart. Stroke
leading to positive feedback in the relationship volume is determined by the contractile function
between hypertension and stiffening (Cecelja and of the heart and by the “afterload” presented to the
Chowienczyk 2009). In CYP with ISH, an heart by the arterial tree. The heart must generate a
age-related degenerative process leading to arterial force of contraction which exceeds the pressure in
stiffness seems implausible, given the relatively the aorta to open the aortic valve and eject blood
short duration of hypertension (Cecelja and into the systemic arterial circulation. Subsequent
Chowienczyk 2010; Mitchell 2014). ejection of blood depends on the afterload, and it
24 M. D. Sinha and P. Chowienczyk

P1 − P2: difference between MAP and CVP


CVP = 0 mmHg, so P1 − P2 is equivalent to MAP
Q = flow, equivalent to
CO

( 1 − 2) R = resistance,
Q= equivalent to SVR

= =

MAP, mean arterial pressure; CVP, central venous pressure; CO, cardiac output; SVR, total systemic peripheral
vascular resistance

Fig. 1 Schematic showing the equation of mean arterial pressure (MAP). MAP, mean arterial pressure; CVP, central
venous pressure; CO, cardiac output; SVR, total systemic peripheral vascular resistance

is important to appreciate that in a dynamic situ- SVR (also known as total peripheral resistance,
ation this is dependent on the stiffness of the aorta TPR) is determined by the terminal arteries and
and large arteries as well as the SVR. arterioles. Resistance to flow arises from internal
Contractile energy is affected by the following: friction between the fluid and the walls of the
vessel and internally within the fluid. When the
(i) Preload, defined as the pressure in the atria at flow in a vessel segment approximates fully
the end of diastole, when the ventricles are at developed laminar flow, the resistance to flow is
their fullest, i.e., ventricular filling pressure. given by the Hagen–Poiseuille relationship:
Cardiac myocytes are unique in exhibiting
increased force of contraction with increased Resistance ðRÞ ¼ 8ηL=πr4
stretch, which on a whole organ level trans-
lates to increased stroke volume as diastolic where η is blood viscosity, L the length of the
volume increases. This phenomenon first vessel, and r, the radius of the vessel. Flow
noted independently by Otto Frank and Ernest through the vessel is given by the pressure drop
Henry Starling in the early twentieth century ΔP across the tube (the vessel):
is known as the Frank-Starling mechanism.
(ii) Contractility, the strength of a contraction for Q ¼ ΔP πr4 =8ηL:
any given degree of stretch, which is regu-
lated by sympathetic activity. Arrival of an Thus, flow through vessels is highly sensitive to
action potential at the sympathetic postgan- the radius of the resistance vessels, and halving the
glionic terminal leads to release of epineph- lumen size will decrease flow by 93% for any given
rine, which binds (predominantly) to β1 perfusion pressure. When several vessels are joined
receptors on the cardiac myocyte. Sympa- together in series (e.g., terminal artery and arteri-
thetic actions are also by circulating adrena- ole), their total resistance is the sum of the individ-
line released from the adrenal cortex. The ual resistances. When vessels are joined together in
effects of sympathetic activity on the cardiac parallel (e.g., a capillary bed), the total resistance is
action potential, contractility, and resultant the reciprocal of all their summed conductances
ventricular function are summarized in Fig. 2. (conductance being the reciprocal of resistance).
Given this phenomenon, the terminal arteries and
Thus, SV and CO can be modified by preload, arterioles contribute most towards SVR, whereas
the contractile state of the heart, and afterload. resistance in the capillary beds is surprisingly low,
Another random document with
no related content on Scribd:
PLEASE READ THIS BEFORE YOU DISTRIBUTE OR USE THIS WORK

To protect the Project Gutenberg™ mission of promoting the free


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

Section 1. General Terms of Use and


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

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


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

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

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

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


immediate access to, the full Project Gutenberg™ License must
appear prominently whenever any copy of a Project Gutenberg™
work (any work on which the phrase “Project Gutenberg” appears, or
with which the phrase “Project Gutenberg” is associated) is
accessed, displayed, performed, viewed, copied or distributed:
This eBook is for the use of anyone anywhere in the United
States and most other parts of the world at no cost and with
almost no restrictions whatsoever. You may copy it, give it away
or re-use it under the terms of the Project Gutenberg License
included with this eBook or online at www.gutenberg.org. If you
are not located in the United States, you will have to check the
laws of the country where you are located before using this
eBook.

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


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

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


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

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


Gutenberg™ License terms from this work, or any files containing a
part of this work or any other work associated with Project
Gutenberg™.

1.E.5. Do not copy, display, perform, distribute or redistribute this


electronic work, or any part of this electronic work, without
prominently displaying the sentence set forth in paragraph 1.E.1 with
active links or immediate access to the full terms of the Project
Gutenberg™ License.
1.E.6. You may convert to and distribute this work in any binary,
compressed, marked up, nonproprietary or proprietary form,
including any word processing or hypertext form. However, if you
provide access to or distribute copies of a Project Gutenberg™ work
in a format other than “Plain Vanilla ASCII” or other format used in
the official version posted on the official Project Gutenberg™ website
(www.gutenberg.org), you must, at no additional cost, fee or expense
to the user, provide a copy, a means of exporting a copy, or a means
of obtaining a copy upon request, of the work in its original “Plain
Vanilla ASCII” or other form. Any alternate format must include the
full Project Gutenberg™ License as specified in paragraph 1.E.1.

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


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

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


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

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

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


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

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


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

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

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


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

1.F.

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


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

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


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

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


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

1.F.4. Except for the limited right of replacement or refund set forth in
paragraph 1.F.3, this work is provided to you ‘AS-IS’, WITH NO
OTHER WARRANTIES OF ANY KIND, EXPRESS OR IMPLIED,
INCLUDING BUT NOT LIMITED TO WARRANTIES OF
MERCHANTABILITY OR FITNESS FOR ANY PURPOSE.

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


warranties or the exclusion or limitation of certain types of damages.
If any disclaimer or limitation set forth in this agreement violates the
law of the state applicable to this agreement, the agreement shall be
interpreted to make the maximum disclaimer or limitation permitted
by the applicable state law. The invalidity or unenforceability of any
provision of this agreement shall not void the remaining provisions.
1.F.6. INDEMNITY - You agree to indemnify and hold the
Foundation, the trademark owner, any agent or employee of the
Foundation, anyone providing copies of Project Gutenberg™
electronic works in accordance with this agreement, and any
volunteers associated with the production, promotion and distribution
of Project Gutenberg™ electronic works, harmless from all liability,
costs and expenses, including legal fees, that arise directly or
indirectly from any of the following which you do or cause to occur:
(a) distribution of this or any Project Gutenberg™ work, (b)
alteration, modification, or additions or deletions to any Project
Gutenberg™ work, and (c) any Defect you cause.

Section 2. Information about the Mission of


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

Volunteers and financial support to provide volunteers with the


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

Section 3. Information about the Project


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

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


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

Section 4. Information about Donations to


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

The Foundation is committed to complying with the laws regulating


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

While we cannot and do not solicit contributions from states where


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

International donations are gratefully accepted, but we cannot make


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

Please check the Project Gutenberg web pages for current donation
methods and addresses. Donations are accepted in a number of
other ways including checks, online payments and credit card
donations. To donate, please visit: www.gutenberg.org/donate.

Section 5. General Information About Project


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

Project Gutenberg™ eBooks are often created from several printed


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

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

This website includes information about Project Gutenberg™,


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

You might also like