Discover millions of ebooks, audiobooks, and so much more with a free trial

Only $11.99/month after trial. Cancel anytime.

Kidney Development, Disease, Repair and Regeneration
Kidney Development, Disease, Repair and Regeneration
Kidney Development, Disease, Repair and Regeneration
Ebook2,190 pages96 hours

Kidney Development, Disease, Repair and Regeneration

Rating: 3.5 out of 5 stars

3.5/5

()

Read preview

About this ebook

Kidney Development, Disease, Repair and Regeneration focuses on the molecular and cellular basis of kidney development, exploring the origins of kidney lineages, the development of kidney tissue subcompartments, as well as the genetic and environmental regulation of kidney development. Special coverage is given to kidney stem cells and possible steps towards kidney repair and regeneration. Emphasis is placed on the fetal origins of postnatal renal disease and our current understanding of the molecular basis of damage and repair. Biomedical researchers across experimental nephrology and developmental biology will find this a key reference for learning how the underlying developmental mechanisms of the kidney will lead to greater advances in regenerative medicine within nephrology.

  • Offers researchers a single comprehensive resource written by leaders from both the developmental biology and the experimental nephrology communities
  • Focuses on understanding the molecular basis of organogenesis in the kidney as well as how this can be affected both genetically and environmentally
  • Explains the underlying developmental mechanisms which influence the kidney’s inherent repair capacity
  • Demonstrates how a deeper understanding of mechanisms will lead to greater advances in regenerative medicine
LanguageEnglish
Release dateAug 6, 2015
ISBN9780128004388
Kidney Development, Disease, Repair and Regeneration

Related to Kidney Development, Disease, Repair and Regeneration

Related ebooks

Biology For You

View More

Related articles

Reviews for Kidney Development, Disease, Repair and Regeneration

Rating: 3.5 out of 5 stars
3.5/5

2 ratings0 reviews

What did you think?

Tap to rate

Review must be at least 10 words

    Book preview

    Kidney Development, Disease, Repair and Regeneration - Melissa Helen Little

    Kidney Development, Disease, Repair and Regeneration

    Editor

    Melissa H. Little

    Table of Contents

    Cover image

    Title page

    Copyright

    Contributors

    Foreword

    Acknowledgments

    Section I. Development

    Introduction

    Chapter 1. Zebrafish Renal Development and Regeneration

    Overview

    The Zebrafish Pronephros as a Model of Nephron Development and Injury

    Development and Repair of the Zebrafish Mesonephros

    Conclusions

    Chapter 2. Early Specification and Patterning of the Intermediate Mesoderm: Genetics and Epigenetics

    Introduction

    Early Development of the Kidney

    Epigenetic Regulation during Kidney Development

    Role of Epigenetics in the Adult Kidney

    Conclusions

    Chapter 3. The Human Kidney: Parallels in Structure, Spatial Development, and Timing of Nephrogenesis

    Introduction

    Anatomic Structure of the Mature Human Kidney

    Anatomic Development of the Kidney

    Other Mammalian Species: How They Differ

    Nephron Endowment

    Conclusion

    Chapter 4. RET Signaling in Ureteric Bud Formation and Branching

    Short Introduction to Ureteric Bud Formation and Branching

    The Role of GDNF/GFRα1/RET Signaling in Mouse Kidney Development

    Mutations in RET, GFRA1, and GDNF in Human Congenital Kidney Defects

    Regulation of Ret and Gdnf Gene Expression

    Signaling Pathways Downstream of RET

    Effects of Point Mutations That Remove RET Tyrosine Residues Involved in Signal Transduction

    Genes That Act Downstream of GDNF/GFRα1/RET Signaling

    Cooperation Between GDNF and FGF10 Signaling in UB Outgrowth and Branching

    Does GDNF/RET Signaling Specify the Normal Pattern of UB Branching?

    Cellular Mechanisms of Branching Morphogenesis and the Role of RET Signaling

    Chapter 5. Quantification of Developmental Branching Morphogenesis

    General Introduction

    Stereotypy in Renal Branching

    Challenges in Translating Renal Branching Knowledge

    Chapter 6. Transcriptional Regulation of the Nephrogenic Mesenchyme and Its Progeny

    Introduction

    Counteraction and Cooperation Between Six2 and Wnt/β-Catenin Signaling in Regulation of Nephron Progenitors

    Identification of Direct Transcriptional Targets of Six2 and β-Catenin in Nephron Progenitors

    Cooperation Between Six2 and Osr1 in the Repression of Wnt4

    Sall1 and Wt1 in Nephron Progenitor Programs

    Maintenance of Nephron Progenitors

    Gene Regulatory Networks Established by Transient Activation of Wnt/β-Catenin Signaling

    Closing Remarks

    Chapter 7. The Role of Growth Factors in Balancing Cap Mesenchyme Survival and Differentiation

    The Cap Mesenchyme

    Growth Factor Signaling in Cap Mesenchyme Differentiation

    Fibroblast Growth Factor Signaling

    Wnt Signaling

    Bone Morphogenetic Protein Signaling

    Intersections between FGF, Wnt, and BMP Pathways

    Future Perspectives

    Chapter 8. Notch Signaling in Nephron Segmentation

    Introduction

    Signal Transduction of Notch Signaling

    Notch Signaling Acts after Wnt/β-Catenin Signaling during Nephrogenesis

    Nephron Segmentation

    Rbpj-Dependent Notch Signaling Is Essential for Nephron Segmentation

    Notch Gain-of-Function Studies

    Notch2 Is Activated More Effectively Than Notch1

    Alagille Syndrome and Notch

    Conclusion

    Chapter 9. Genetic and Epigenetic Regulation of Nephron Number in the Human

    Overview

    Monogenic Disorders Causing Suboptimal Ureteric Bud Branching

    Monogenic Disorders Affecting the Renal Progenitor Cell Pool

    Subtle Renal Hypoplasia and Common Polymorphic Variants of Genes That Affect Ureteric Bud Branching

    Subtle Renal Hypoplasia and Common Polymorphic Gene Variants That Regulate the Renal Progenitor Cell Pool

    Epigenetic Regulation of Renal Progenitor Cells

    Conclusion

    Chapter 10. Formation and Maintenance of a Functional Glomerulus

    Introduction

    Patterning of Nephrons and Specification of Glomeruli

    Development of Podocytes and SD

    Formation of Glomerular Vasculature

    Structure and Development of the GBM

    Development of the Mesangium

    Development of PECs and Bowman’s Capsule

    Summary and Perspectives

    Chapter 11. Maturation and Roles of Collecting Ducts and Loops of Henle in Renal Medulla Development

    Introduction

    Composition and Organization of the Renal Medulla

    Overview of Renal Medulla Development

    Medullary Collecting Ducts

    The Loop of Henle

    Medullary Microvasculature

    Concluding Remarks

    Chapter 12. Developmental Roles of the Stroma

    What Is the Renal Stroma?

    Developmental Origins of Renal Stromal Cells

    Role of Stroma in Kidney Development

    Future Directions

    Chapter 13. The Origin and Regulation of the Renal Vasculature

    Introduction

    Anatomy of the Renal Vasculature

    Mechanisms Underlying Renal Vascular Morphogenesis

    Embryonic Origin of the Renal Vasculature

    Lineage of Vascular Cells

    Signaling Pathways and Regulatory Factors

    Remaining Issues

    Section II. Disease

    Introduction

    Chapter 14. Variation in Human Nephron Number and Association with Disease

    Introduction

    Estimating Nephron Number

    Human Nephron Number: Variability Is the Rule

    Human Nephron Number and Blood Pressure

    Relationship between Glomerular Number and Glomerular Volume

    Human Nephron Number and Renal Pathology

    Conclusion

    Chapter 15. The Effect of the In utero Environment on Nephrogenesis and Renal Function

    Introduction

    The Developmental Origins of Health and Disease

    Evidence for Programming in the Human

    Links between Impaired Kidney Development and Hypertension

    Animal Models of Developmental Programming: Consequences for Kidney Development

    Why Is the Kidney Susceptible to Programming?

    Programming of Disease Is More than Just Nephron Number

    Mechanism Contributing to the Formation of Low Nephron Endowment

    Conclusion

    Chapter 16. Wilms’ Tumor: A Case of Persistence of the Nephrogenic Mesenchyme

    The History of Wilms’ Tumors

    Identification of WT1 Gene

    Structure of WT1

    Molecular Studies on the Function of WT1 in the Nephrogenic Mesenchyme

    Embryonic Expression of WT1 and Phenotypes of WT1 Mutant Mice

    Other Tumor Suppressor Genes for Wilms’ Tumor

    The Cell of Origin for Wilms’ Tumor

    Other Influences on Kidney Progenitor Cell Expansion

    Relationship of Wilms’ Tumors to Nephron Progenitor Cells

    Conclusion

    Chapter 17. Wnt, Notch, and Tubular Pathology

    Wnt and Notch

    Wnt and Notch in Acute Kidney Injury Repair

    Wnt and Notch in Kidney Fibrosis

    Wnt and Notch in Polycystic Kidney Disease

    Wnt and Notch in Renal Cell Cancer

    Conclusion

    Chapter 18. Regulation of Ureteric Bud Outgrowth and the Consequences of Disrupted Development

    Introduction

    Nephric Duct Development and UB Outgrowth

    Ureteric Budding Site Determination and GDNF-Dependent UB Outgrowth

    GDNF–Independent Induction of UB Outgrowth

    Common Human Disease Caused by Abnormal UB Outgrowth

    Conclusions

    Chapter 19. Vesicoureteral Obstruction and Vesicoureteral Reflux: Different Congenital Defects With a Common Cause

    Introduction

    Development of the Urinary Tract

    Conclusion

    Chapter 20. Polycystic Kidney Diseases and Other Hepatorenal Fibrocystic Diseases: Clinical Phenotypes, Molecular Pathobiology, and Variation between Mouse and Man

    Introduction

    Hepatorenal Fibrocystic Diseases: Clinical Phenotypes

    Hepatorenal Fibrocystic Diseases: Genetic Defects and Molecular Pathobiology

    Genetic Testing

    Mouse Models

    Conclusion

    Chapter 21. Genetic Aspects of Human Congenital Anomalies of the Kidney and Urinary Tract

    Introduction

    Budding Hypothesis

    HNF1β Nephropathy

    RET, Hirschsprung Disease, and CAKUT

    ITGA8 Mutations in Autosomal Recessive Bilateral Kidney Agenesis

    PAX2 in Renal Coloboma Syndrome

    EYA1, SIX1, and SIX5 in Branchiootorenal Syndrome

    Renal Tubular Dysgenesis

    BMP4

    Other Genes Involved in CAKUT

    Ureteral Anomalies

    Bladder Anomalies with Functional Bladder Outflow Obstruction

    Detection of Chromosomal Microimbalances

    Concepts of Oligogenic Inheritance

    MiRNAs as Potential Regulators of Renal Developmental Genes

    Conclusions

    Chapter 22. Inherited Kidney Disorders in the Age of Genomics

    Introduction

    From DNA to Genomics

    Interpretation of NGS Data

    Impact of NGS in the Nephrology Field

    NGS Challenges

    Personalized Therapeutics

    Conclusions and Future Directions

    Chapter 23. Fibrosis: A Failure of Normal Repair and a Common Pathway to Organ Failure

    Fibrosis is a Characteristic Feature of Chronic Kidney Diseases

    Injury–Repair Processes Gone Awry

    Cellular Mechanisms of Fibrosis

    Source of Fibrogenic Cells

    Molecular Pathways in Fibrogenesis

    Matrix Turnover

    Resolution of Fibrosis

    Section III. Repair

    Introduction

    Chapter 24. Postnatal Cell Turnover in the Nephron Epithelium: What Can This Tell Us?

    Postnatal Glomerular Epithelial Cell Turnover

    Glomerular Cell Turnover in Disease

    Postnatal Tubular Epithelial Cell Turnover

    Conclusion

    Chapter 25. Plasticity within the Collecting Ducts: What Role Does This Play in Response to Injury?

    Development of the Collecting Duct

    Acute Kidney Injury and Response of the CD

    CD-Specific Mechanisms of Epithelial Injury and Repair

    Summary

    Chapter 26. The Onset and Resolution of Renal Fibrosis: A Human Perspective

    Introduction

    Renin-Angiotensin System

    Transforming Growth Factor-β

    Connective Tissue Growth Factor

    Epigenetic Control of Renal Fibrosis

    Conclusions

    Chapter 27. The Molecular Response to Renal Injury: How Does Chronic Renal Damage Suppress Normal Repair Processes?

    The Renal Response to Chronic Kidney Disease

    Adaptive Repair in the Kidney

    Risk Factors and Features of Maladaptive Repair

    Maladaptive Repair and Recurrent AKI

    Mechanisms of Cell Cycle Control

    Cell Cycle Control in Healthy and Diseased Kidneys

    Senescent Cells, G2/M Arrest, and Kidney Injury

    Epigenetic Changes after AKI

    Pericytes and Kidney Scarring

    Microvascular Loss and Renal Hypoxia

    Conclusions

    Chapter 28. Investigating the Process of Renal Epithelial Repair to Develop New Therapies

    Introduction

    Cellular Origins of Epithelial Cells during Repair

    Targeting Epithelial Repair Pathways

    Preventing Maladaptive Responses after AKI to Promote Repair

    Conclusions

    Chapter 29. Evidence for Renal Progenitors in the Human Kidney

    Introduction

    Renal Progenitors in the Human Nephron

    Tubular-Committed Progenitors

    Tubular Progenitors in the Pathogenesis of Kidney Disorders

    Renal Progenitors in the Glomerulus of Adult Human Kidney

    Glomerular Progenitors in the Pathogenesis of Kidney Disorders

    Human Progenitor Cultures and Their Possible Application for Kidney Disease Modeling

    Conclusion

    Chapter 30. Label-Retaining Cells and Progenitor Cells in Renal Epithelial Homeostasis and Regeneration

    Stem Cells in Adult Organs

    Markers and Labels of Stem Cells

    Label-Retaining Cells in the Kidney Papilla

    A Genetic Label for Renal LRCs

    Label-Retaining Cells Proliferate in a Restricted Compartment at the Top of the Papilla

    In Vivo Demonstration of Migration of LRCs from the Papilla in Response to Ischemic Injury

    SDF1/CXCR4 Cause Proliferation and Migration of LRCs in Response to Ischemic Injury

    Microarray Analysis of LRCs: The Wnt Pathway

    Controversies in Renal Stem Cells

    Multiple Pools of Stem Cells in the Kidney?

    Conclusion

    Chapter 31. A Reparative Role for Macrophages in Kidney Disease

    Introduction

    Macrophage Phenotype

    Reparative Macrophages in Kidney Disease

    The Potential for Macrophage Therapy in Kidney Disease

    Frontiers of Kidney Macrophage Research

    Conclusion

    Chapter 32. The Use of Mesenchymal Stromal Cells for Treating Renal Injury and Promoting Allograft Survival after Renal Transplantation

    Introduction

    Mesenchymal Stromal Cells

    Immunomodulatory Properties of MSCs

    Reparative Properties of MSCs

    Experimental Models of MSCs in Acute Kidney Injury and after Transplantation

    Mesenchymal Stromal Cells for Clinical Therapeutic Use

    Clinical Data of MSCs in Kidney Injury and after Renal Transplantation

    Potential Risks

    Conclusions

    Section IV. Regeneration

    Introduction

    Chapter 33. Reprogramming to Kidney

    Introduction

    Reprogramming to Pluripotency

    How Does Reprogramming to Pluripotency Occur?

    Directed Differentiation of Pluripotent Cells

    Direct Reprogramming: Fate Conversion from One Somatic Cell Type to Another

    Pioneer versus Lineage-Specifying Factors

    Evidence that Reprogramming to Nephron Progenitor is Feasible

    Overlapping Actions for the Nephron Progenitor Reprogramming Genes Suggests Redundancy

    Alternative Options for the Specification of CM

    Transcription Factors Required for Reprogramming to Mature Renal Cell Types

    Challenges to Direct Reprogramming and Cellular Therapies Using Reprogrammed Cells

    Conclusion

    Chapter 34. From Development to Regeneration: Kidney Reconstitution In vitro and In vivo

    Strategies for Regenerative Medicine of the Kidney

    Lessons from Kidney Development in Frogs and Mice

    Nephron Progenitors in the Mouse Embryonic Kidney

    The Newly Identified Origin of the Kidney

    Generation of Nephron Progenitors In vitro

    Reconstitution of 3D Kidney Structures from Nephron Progenitors

    Generation of the Ureteric Bud In vitro

    Challenges for Reconstitution of a Functional Kidney In vitro

    Kidney Reconstitution In vivo

    Conclusions

    Chapter 35. Directing the Differentiation of Pluripotent Stem Cells to Renal End Points

    Introduction

    Pluripotent Stem Cells

    Kidney Development

    Differentiation Formats, Inducers, and Cell Lines

    Animal Cap in Fertilized Eggs of Amphibians

    Differentiation of Mouse ESCs and iPSCs into Kidney Lineages

    Human ESCs and iPSCs

    Cell Therapy, Disease Modeling, and Toxicology

    Hurdles to Overcome

    Conclusions

    Chapter 36. Patient-Derived Induced Pluripotent Stem Cells to Target Kidney Disease

    Induced Pluripotent Stem Cells

    Induced Pluripotent Stem Cells Targeting the Kidney

    Cell Memory and iPS Differentiation

    Generating Specific Mature Renal Cell Types From iPS Cells

    Potential Application of iPS Cell-Derived Kidney Podocytes

    Applications of iPS Cells in Disease Modeling and Toxicity Screening

    Understanding an Inherited Basis of Kidney Disease Using iPS Cells

    Developing Novel Treatments Based on iPS Disease Modeling of Inherited Kidney Disease

    Gene Targeting and Editing of iPS Cells

    Cell Replacement Strategies

    Conclusion

    Chapter 37. Xenotransplantation in the Kidney: A Historical Perspective

    Introduction

    Xenotransplantation of Developed Kidneys

    Transplantation of Developing Kidneys

    Allotransplantation of Renal Primordia

    Xenotransplantation of Renal Primordia

    Xenotransplantation of Pig Renal Primordia

    Use of Non-renal Precursor Cells Integrated into Renal Primordia or Xenobiotic Nephrogenesis

    Challenges in the Application of Embryonic Kidney Transplantation

    Recapitulation of Filtration, Reabsorption, and Secretion

    Conclusions

    Chapter 38. Use of the Nephrogenic Niche in Xeno-Embryos for Kidney Regeneration

    Introduction

    Niche in the Blastocyst

    Niche in the Growing Embryo

    Niche in the Adult Organ

    Conclusions

    Chapter 39. Human Fetal Kidney for Regenerative Medicine: From Embryonic Rudiments to Renal Stem/Progenitor Cells

    Introduction

    Pre- and Post-Mesenchymal-to-Epithelial Transition Stages of Nephrogenesis

    Human Embryonic Kidney Rudiments

    Biomarking the hFK and Wilms’ Tumor as a Step for Progenitor Cell Identification

    Isolation of Expandable Human Nephric Progenitor Cells From Fetal Kidney

    Identification of WT Stem Cells

    Clinical Relevance and Concluding Remarks

    Chapter 40. Renal Replacement Approaches Using Deceased Donor Cell Sources

    Introduction

    Section I

    Section II

    Section III

    Conclusions

    Chapter 41. Tissue Engineering through Additive Manufacturing: Hope for a Bioengineered Kidney?

    Additive Manufacturing Meets Tissue Engineering

    Strategies for Building Kidney Tissue through Additive Manufacturing

    Renal Tissue Design Considerations for Regenerative Applications

    Considerations for Tissue Maturation

    Summary

    Chapter 42. Decellularized Whole Organ Scaffolds for the Regeneration of Kidneys

    Introduction

    Tissue Engineering Strategy

    Extracellular Matrix

    Whole Organ Decellularization

    Re-endothelialization

    Renal Recellularization

    In vivo Functionality

    Conclusions

    Index

    Copyright

    Academic Press is an imprint of Elsevier

    125 London Wall, London EC2Y 5AS, UK

    525 B Street, Suite 1800, San Diego, CA 92101-4495, USA

    225 Wyman Street, Waltham, MA 02451, USA

    The Boulevard, Langford Lane, Kidlington, Oxford OX5 1GB, UK

    Copyright © 2016 Elsevier Inc. All rights reserved.

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

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

    Notices

    Knowledge and best practice in this field are constantly changing. As new research and experience broaden our understanding, changes in research methods, professional practices, or medical treatment may become necessary.

    Practitioners and researchers must always rely on their own experience and knowledge in evaluating and using any information, methods, compounds, or experiments described herein. In using such information or methods they should be mindful of their own safety and the safety of others, including parties for whom they have a professional responsibility.

    To the fullest extent of the law, neither the Publisher nor the authors, contributors, or editors, assume any liability for any injury and/or damage to persons or property as a matter of products liability, negligence or otherwise, or from any use or operation of any methods, products, instructions, or ideas contained in the material herein.

    Medical Disclaimer

    Medicine is an ever-changing field. Standard safety precautions must be followed, but as new research and clinical experience broaden our knowledge, changes in treatment and drug therapy may become necessary or appropriate. Readers are advised to check the most current product information provided by the manufacturer of each drug to be administered to verify the recommended dose, the method and duration of administrations, and contraindications. It is the responsibility of the treating physician, relying on experience and knowledge of the patient, to determine dosages and the best treatment for each individual patient. Neither the publisher nor the authors assume any liability for any injury and/or damage to persons or property arising from this publication.

    ISBN: 978-0-12-800102-8

    British Library Cataloguing-in-Publication Data

    A catalogue record for this book is available from the British Library

    Library of Congress Cataloging-in-Publication Data

    A catalog record for this book is available from the Library of Congress

    For information on all Academic Press publications visit our website at http://store.elsevier.com/

    Typeset by TNQ Books and Journals

    www.tnq.co.in

    Printed and bound in the United States of America

    Contributors

    Qais Al-Awqati,     Department of Medicine, Columbia University College of Physicians & Surgeons, New York, NY, USA

    H.H. Arts,     Department of Human Genetics, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands

    Anthony Atala,     Wake Forest School of Medicine, Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA

    Felicity J. Barnes,     Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia

    Ariela Benigni,     IRCCS — Istituto di Ricerche Farmacologiche Mario Negri, Centro Anna Maria Astori, Science & Technology Park Kilometro Rosso Bergamo, Italy

    John F. Bertram,     Department of Anatomy and Developmental Biology, School of Biomedical Sciences, Monash University, Melbourne, VIC, Australia

    M. Jane Black,     Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, Australia

    Joseph V. Bonventre

    Renal Division and Biomedical Engineering Division, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Boston, MA, USA

    Division of Health Sciences and Technology, Harvard-Massachusetts Institute of Technology, Cambridge, MA, USA

    Harvard Stem Cell Institute, Cambridge, MA, USA

    Deborah A. Buffington,     Innovative BioTherapies, Incorporated, Ann Arbor, MI, USA

    Kevin T. Bush,     Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA

    Qi Cao,     Centre for Transplant and Renal Research, Westmead Millennium Institute, University of Sydney at Westmead Hospital, Westmead, NSW, Australia

    Thomas Carroll,     Departments of Molecular Biology and Internal Medicine (Nephrology), University of Texas Southwestern Medical Center, Dallas, TX, USA

    Melanie Cosgrove,     Department of Experimental Medicine, McGill University, Montreal, QC, Canada

    Frank Costantini,     Department of Genetics and Development, Columbia University, New York, NY, USA

    Luise Cullen-McEwen,     Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, Australia

    Alan J. Davidson,     Department of Molecular Medicine & Pathology, School of Medical Sciences, Faculty of Medical Health Sciences, The University of Auckland, Auckland, New Zealand

    Benjamin Dekel

    Pediatric Stem Cell Research Institute, Edmond & Lili Safra Children’s Hospital, Sheba Medical Center, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel

    Division of Pediatric Nephrology, Edmond & Lili Safra Children’s Hospital, Sheba Medical Center, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel

    Rachel C. Dodd,     Department of Molecular Medicine & Pathology, School of Medical Sciences, Faculty of Medical Health Sciences, The University of Auckland, Auckland, New Zealand

    Gregory R. Dressler,     Department of Pathology, University of Michigan, Ann Arbor, MI, USA

    Jeremy S. Duffield

    Research & Development, Biogen, Cambridge, MA, USA

    Departments of Medicine & Pathology, University of Washington, Seattle, WA, USA

    Klaudyna Dziedzic,     Pediatric Stem Cell Research Institute, Edmond & Lili Safra Children’s Hospital, Sheba Medical Center, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel

    David A. Ferenbach

    Renal Division and Biomedical Engineering Division, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Boston, MA, USA

    Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK

    Julia B. Finkelstein,     Department of Urology, Columbia University College of Physicians and Surgeons, New York, NY, USA

    Paul Goodyer

    Department of Pediatrics, McGill University, Montreal, QC, Canada

    Department of Human Genetics, McGill University, Montreal, QC, Canada

    L.M. Guay-Woodford,     Center for Translational Science, Children’s National Health System, Washington, DC, USA

    Marc R. Hammerman,     Renal Division, Departments of Medicine, and Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA

    David C.H. Harris,     Sydney Medical School – Western Centre for Transplant and Renal Research, Westmead Millennium Institute, University of Sydney at Westmead Hospital, Westmead, NSW, Australia

    Michael J. Hiatt,     Developmental Biology and Regenerative Medicine Program, The Saban Research Institute, Children’s Hospital Los Angeles, University of Southern California, Los Angeles, CA, USA

    Wendy E. Hoy,     Centre for Chronic Disease, University of Queensland, Brisbane, QLD, Australia

    Michael D. Hughson,     Department of Pathology, University of Mississippi Medical Center, Jackson, MS, USA

    Jennifer C. Huling

    Wake Forest School of Medicine, Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA

    School of Biomedical Engineering and Sciences, Virginia Tech-Wake Forest University, Winston-Salem, NC, USA

    H. David Humes

    Innovative BioTherapies, Incorporated, Ann Arbor, MI, USA

    The University of Michigan, Ann Arbor, MI, USA

    Benjamin D. Humphreys

    Renal Division, Brigham and Women’s Hospital, Boston, MA, USA

    Harvard Medical School, Boston, MA, USA

    Harvard Stem Cell Institute, Cambridge, MA, USA

    Roger Ilagan,     Division of Regenerative Medicine, United Therapeutics Corporation, Research Triangle Park, NC, USA

    Nine V.A.M. Knoers,     Department of Medical Genetics, University Medical Centre Utrecht, Utrecht, The Netherlands

    Raphael Kopan,     Division of Developmental Biology, Cincinnati Children’s Hospital, Cincinnati, OH, USA

    Jordan A. Kreidberg,     Department of Medicine, Boston Children’s Hospital, and Department of Pediatrics, Harvard Medical School, Boston, MA, USA

    Callie S. Kwartler,     Departments of Molecular Biology and Internal Medicine (Nephrology), University of Texas Southwestern Medical Center, Dallas, TX, USA

    Laura Lasagni,     Excellence Centre for Research, Transfer and High Education for the Development of DE NOVO Therapies (DENOTHE), University of Florence, Florence, Italy

    Elena Lazzeri,     Excellence Centre for Research, Transfer and High Education for the Development of DE NOVO Therapies (DENOTHE), University of Florence, Florence, Italy

    Melissa H. Little,     The Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia

    Weining Lu,     Department of Medicine, Boston University School of Medicine, Boston Medical Center, Boston, MA, USA

    Daniela Macconi,     IRCCS — Istituto di Ricerche Farmacologiche Mario Negri, Centro Anna Maria Astori, Science & Technology Park Kilometro Rosso Bergamo, Italy

    Douglas G. Matsell,     Child and Family Research Institute, British Columbia Children’s Hospital, University of British Columbia, Vancouver, BC, Canada

    Andrew P. McMahon,     Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, WM Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA

    Cathy Mendelsohn,     Departments of Urology, Genetics and Development and Pathology, Columbia University, New York, NY, USA

    Marcus J. Moeller,     Department of Nephrology and Immunology, RWTH Aachen University, Aachen, Germany

    Karen M. Moritz,     School of Biomedical Science, The University of Queensland, St Lucia, QLD, Australia

    Sanjay K. Nigam

    Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA

    Department of Medicine, University of California, San Diego, La Jolla, CA, USA

    Department of Cellular & Molecular Medicine, University of California, San Diego, La Jolla, CA, USA

    Ryuichi Nishinakamura,     Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan

    A.K. O’Connor,     Center for Translational Science, Children’s National Health System, Washington, DC, USA

    Juan A. Oliver,     Department of Medicine, Columbia University College of Physicians & Surgeons, New York, NY, USA

    Kenji Osafune,     Center for iPS Cell Research and Application (CiRA), Kyoto University, Shogoin, Sakyo-ku, Kyoto, Japan

    Leif Oxburgh,     Center for Molecular Medicine, Maine Medical Center Research Institute, ME, USA

    Joo-Seop Park

    Division of Pediatric Urology, Cincinnati Children’s Hospital, Cincinnati, OH, USA

    Division of Developmental Biology, Cincinnati Children’s Hospital, Cincinnati, OH, USA

    Anna Peired,     Excellence Centre for Research, Transfer and High Education for the Development of DE NOVO Therapies (DENOTHE), University of Florence, Florence, Italy

    Christopher J. Pino,     Innovative BioTherapies, Incorporated, Ann Arbor, MI, USA

    Oren Pleniceanu,     Pediatric Stem Cell Research Institute, Edmond & Lili Safra Children’s Hospital, Sheba Medical Center, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel

    Sharon Presnell,     Organovo, Inc., San Diego, CA, USA

    Victor G. Puelles,     Department of Anatomy and Developmental Biology, School of Biomedical Sciences, Monash University, Melbourne, VIC, Australia

    Susan E. Quaggin,     Division of Nephrology and Hypertension, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA

    Ton J. Rabelink

    Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands

    Department of Nephrology, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands

    Egon Ranghini,     Department of Pathology, University of Michigan, Ann Arbor, MI, USA

    Scott Rapoport,     Division of Regenerative Medicine, United Therapeutics Corporation, Research Triangle Park, NC, USA

    Marlies E.J. Reinders,     Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands

    Giuseppe Remuzzi

    IRCCS — Istituto di Ricerche Farmacologiche Mario Negri, Centro Anna Maria Astori, Science & Technology Park Kilometro Rosso Bergamo, Italy

    Unit of Nephrology and Dialysis, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy

    Sharon D. Ricardo,     Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia

    Paola Romagnani

    Excellence Centre for Research, Transfer and High Education for the Development of DE NOVO Therapies (DENOTHE), University of Florence, Florence, Italy

    Pediatric Nephrology Unit, Meyer Children’s Hospital, University of Florence, Florence, Italy

    Rizaldy P. Scott,     Division of Nephrology and Hypertension, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA

    Maria Luisa S. Sequeira Lopez,     University of Virginia School of Medicine, Charlottesville, VA, USA

    Benjamin Shepherd,     Division of Regenerative Medicine, United Therapeutics Corporation, Research Triangle Park, NC, USA

    Kieran M. Short,     Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia

    Ian M. Smyth

    Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia

    Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, Australia

    Katalin Susztak,     Renal Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA

    Megan R. Sutherland,     CHU Sainte-Justine Research Center and the University of Montreal, Montreal, QC, Canada

    Atsuhiro Taguchi,     Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan

    Yiping Wang,     Centre for Transplant and Renal Research, Westmead Millennium Institute, University of Sydney at Westmead Hospital, Westmead, NSW, Australia

    Stefanie Weber,     Pediatrics II, University Children’s Hospital Essen, Essen, Germany

    Angela J. Westover,     Innovative BioTherapies, Incorporated, Ann Arbor, MI, USA

    Takashi Yokoo,     Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan

    James J. Yoo,     Wake Forest School of Medicine, Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA

    Jing Yu,     Department of Cell Biology, Child Health Research Center, the Center for Immunity, Inflammation and Regenerative Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA

    Foreword

    According to ancient Greek mythology the Titan Prometheus was punished by Zeus for stealing fire from the gods and handing it over to mortal humans. His punishment consisted of being chained to the Caucasus mountains, where he had to suffer an eagle feasting daily on his liver, which would then regenerate overnight. Out of this mythology arose the great hope that all organs, if damaged, could regenerate. Unfortunately, in humans the enormous regenerative capacity of the liver seems to be the exception rather than the rule. Nonetheless, all organs, including the kidney, have some ability to regenerate or repair injured renal structures. In nephrology this is most obvious in the case of acute tubular injury. The limitations for renal regeneration may be imposed by the very complicated intrinsic anatomical structure of the nephron and the functional interconnection of its different components as the basic building blocks of the kidney. Therefore, and in spite of some regeneration of damaged nephron segments, the overall renal function declines in many forms of renal injury. This leads to progression of injury and chronic kidney disease (CKD), which remains a major problem with unfortunately a dearth of effective therapeutic interventions. It is because of this that possible regenerative therapeutic approaches, whether to prevent or slow down progression of CKD or even for replacement of destroyed nephrons, are increasingly attracting interest. In this context one has to recall that such regenerative therapeutic interventions will draw on the basic understanding of normal embryonic developmental of the kidney, and that understanding the molecular basis of these processes will be the key to the development of regenerative therapies for chronic renal disease.

    In this book Professor Melissa Little has successfully undertaken the very difficult task of bringing together and editing texts encompassing the vast amount of information, ranging from basic embryology of the kidney all the way to exploring potential ways of using this basic knowledge to repair or regenerate parts or even entire nephrons or their function. The list of authors reads like a Who’s who of experts for all aspects of kidney development, injury response, cell differentiation and de-differentiation, repair processes, their signaling processes and the specific cells involved, and the various types of stem cells and their biology, with special emphasis on the potential therapeutic use of renal and non-renal adult and embryonic stem cells.

    In order to connect these various aspects into a logical and understandable framework the initial chapters are devoted to the basics of renal development. They expand to illustrate the similarities and differences between developmental processes and those recapitulated during renal injury and repair using various model systems. This includes aspects of genetics and epigenetics during development as well as in disease processes. Having described this for the entire kidney, subsequent chapters explore these aspects for specific nephron segments, from the glomerulus down to the collecting tubule and upper urinary tract, and also for the renal vasculature and stroma. For each nephron segment, diseases resulting from genetic and environmental developmental abnormalities are discussed in terms of their molecular and cellular pathogenesis. Finally, mechanisms and limitations of repair and/or regeneration of specific cell types along the nephron are critically analyzed.

    In this context, the use of lineage tracing has been critical to differentiate regeneration from intrarenal versus bone-marrow derived progenitor cells. Furthermore, it appears that instead of a single type of kidney stem cell to replace lost or damaged kidney tissue, there are nephron segment-specific progenitor cells which give rise to new cells within each kidney compartment. Equally important is the identification of pathways that govern the proliferation and differentiation of regeneration-competent cells, and the signals that inhibit their activity after injury. In addition to the possibility of intra-renal repair and regeneration of nephrons, the possibilities of growing entirely new structural and functional nephrons or even kidneys are discussed and the creation of bio-engineered kidneys or partial replacement of kidney functions are addressed.

    The authors point out the potential for the application of many of these novel regenerative approaches, but also caution about the many remaining hurdles. These include problems of adequately identifying renal stem cell populations and assessing their therapeutic potential. Also the unique architecture of the kidney complicates the anatomical and functional integration of stem cell–derived nephrons. This may apply even more to the functional capacity of a bioengineered organ to partially or completely replace the endogenous kidney. In spite of these difficulties, the road ahead for regenerative therapy is exciting and many obstacles may be overcome based on the enormous strides forward achieved in genetics, molecular and cell biology, and especially stem cell biology.

    Taken together this book provides a state of the art compendium of our present knowledge and of future perspectives for all aspects of regenerative kidney repair and therapy. As such it represents a pioneering and gigantic step forward in this exciting new field. Just as Prometheus brought fire to mortal humans, the authors of this excellent text bring light and hope to all of us nephrologists dwelling in the present penumbra surrounding the therapy of acute and chronic kidney injury.

    Detlef Schlöndorff

    Detlef Schlöndorff, MD is currently visiting professor of medicine at the Icahn School of Medicine at Mount Sinai in New York, USA, and Editor-in-Chief of Kidney International. He is Professor Emeritus of the Ludwig Maximilians University of Munich, Germany.

    Acknowledgments

    The kidneys, from the perspectives of functional diversity and cellular complexity, are one of the most remarkable organs in our body. This has undoubtedly contributed to the challenge of replacing renal function. Indeed, at a point in history where chronic renal disease is rapidly increasing in prevalence, the need for alternative approaches to ensuring ongoing renal function is acute. Fully understanding the organ itself is surely the basis upon which such solutions will arise. My personal foray into the biology of the kidney commenced as an Honours student in Physiology examining the rate of production and degradation of angiotensinogen in a rat model. Serendipitously, I moved from this to a research position in childhood cancer, focussing on the molecular basis of the childhood kidney cancer, Wilms’ tumour, at a time just before the WT1 gene was identified. As a postdoctoral researcher in the United Kingdom, under the mentorship of Professor Nicholas Hastie (then of the MRC Human Genetics Unit, Edinburgh) we began to unravel the link between WT1 and kidney/gonad development and dysmorphogenesis, identifying mutations in the developmental anomaly syndrome, Denys Drash syndrome. This piqued my interest in how the development of this fascinating organ was controlled. What followed was not only a journey through kidney biology, but one that ranged across developmental molecular biology, through systems biology, experimental nephrology and ultimately stem cell biology and regenerative medicine. Hence, bringing together this collection of stories is a reflection of my own scientific journey. For the first time in one location, the reader can bridge that spectrum from how the kidney forms, what can go wrong, how the organ attempts to cope with injury and finally how we might build on this knowledge to recreate structure and function. The end product is the work of many. As always, my family deserves special mention for their endless patience with my obsession for work. Having their love and support makes me human and the interspersed chaos and joy of normal life brings the light and colour that makes the rest worth it. I particularly thank my son, Nathaniel Rhoades, who helped me with production of graphic plates. The dedicated work of my own team of researchers (past and present), not only in generating some of the science discussed within these pages but continuing to deliver in response to my tireless enthusiasm, definitely needs acknowledgement. I thank Jeff Rossetti and Edward Taylor for calmly overseeing the project as timelines walked into the distance. Most of all, I am indebted to the spectacular scientists who wrote the component chapters. You are also my heroes, colleagues and mentors, and I thank you for your contributions to the book in the face of the many clinical, scientific and personal demands on your own lives. I particularly thank Lisa Guay-Woodford and Weining Lu who accepted either more than was first anticipated or were brought in late with short timelines. In building this book, I have learnt much from the individual chapters and the perspective of each set of authors on their particular areas of expertise. To see it complete reinforces my conviction that it was time to marry together these seemingly disparate areas of research around the commonality of the clinical challenge that is kidney failure. Revealing, but exciting, is what there is yet to understand and what might be achieved in the coming decades.

    Professor Melissa H. Little,     NHMRC Senior Principal Research Fellow, Murdoch Children’s Research Institute & Department of Pediatric University of Melbourne, Australia

    Section I

    Development

    Outline

    Introduction

    Chapter 1. Zebrafish Renal Development and Regeneration

    Chapter 2. Early Specification and Patterning of the Intermediate Mesoderm: Genetics and Epigenetics

    Chapter 3. The Human Kidney: Parallels in Structure, Spatial Development, and Timing of Nephrogenesis

    Chapter 4. RET Signaling in Ureteric Bud Formation and Branching

    Chapter 5. Quantification of Developmental Branching Morphogenesis

    Chapter 6. Transcriptional Regulation of the Nephrogenic Mesenchyme and Its Progeny

    Chapter 7. The Role of Growth Factors in Balancing Cap Mesenchyme Survival and Differentiation

    Chapter 8. Notch Signaling in Nephron Segmentation

    Chapter 9. Genetic and Epigenetic Regulation of Nephron Number in the Human

    Chapter 10. Formation and Maintenance of a Functional Glomerulus

    Chapter 11. Maturation and Roles of Collecting Ducts and Loops of Henle in Renal Medulla Development

    Chapter 12. Developmental Roles of the Stroma

    Chapter 13. The Origin and Regulation of the Renal Vasculature

    Introduction

    A functioning kidney is vital for life. While most are familiar with the excretory functions of the kidney, this complex organ also modulates fluid volume, blood pressure, red cell count and bone density, thereby acting as a central regulator of homeostasis. While a fundamental understanding of normal organ morphogenesis is clearly relevant for the interpretation of congenital anomaly, there is a growing appreciation that normal kidney morphogenesis is crucial for long term renal function. In humans, as in most mammals, nephron formation is a fetal event with final nephron number set before or near birth. Evidence in humans for a 10 fold variation in nephron number between individuals, no capacity to form new nephrons after birth and a clear inverse relationship between nephron number and renal disease, has refocussed our attention on how the kidney forms and what determines an optimal outcome.

    In all amniotes, three excretory organs formed during embryogenesis. The permanent kidney in humans (and all mammals) is the third of these structures to form; the metanephros. Despite this, there are lessons to be learnt even from the non-mammalian kidney. The molecular paradigms described in pronephric development in the fish show remarkable parallels with those used for the elaboration of the much more complex mammalian metanephros. Chapter 1 will describe the differences in approach to nephron endowment, repair and response to growth that exist in fish. The developmental origin of these excretory structures is addressed in Chapter 2. Studies on metanephric development have focussed on mammalian models such as mouse, rat, sheep, pig and baboon. Of these, mouse is the most amenable to genetic manipulation, including lineage tracing to investigate cellular ontogeny as well as compartment specific gene deletion or activation. The accessibily of such model organisms, together with the simplicity of genetic manipulation and visualisation, has allowed us to acquire much information about morphogenesis in simpler excretory organs. While studies of kidney morphogenesis in mouse have significantly improved our knowledge of the pathways involved in kidney morphogenesis, a mouse is not a human. Indeed, substantial differences exist in the histological structure or kidneys between different mammals. Chapter 3 will address human kidney morphogenesis and how this differs from morphogenesis in other model organisms. Understanding these differences may also inform us about approaches to modify development or repair in the human.

    The metanephros is generated from four major progenitor populations that give rise to the branching collecting duct tree (ureteric bud), the nephrons (cap mesenchyme), interstitial elements, including the mesangium and pericytes (stroma) and the vasculature (vascular progenitors). Chapter 4 discusses the regulation of patterning of the ureteric epithelium. The challenge in studying morphogenesis in all mammalian species remains the size and cellular complexity. Chapter 5 begins to address this in the mouse, illustrating both what level of quantitative morphogenetic analyses might be possible as well as highlighting what we have missed to date. The nephrons arise from the cap mesenchyme. Our understanding of what specifies this population and how the balance between self-renewal and commitment to nephron formation is regulated is discussed in Chapters 6 and 7. The nascent nephron has to undergo substantial elongation, patterning and segmentation to generate the required functional compartmentalisation essential for nephron function (Chapter 8). Regulation of nephron number in the human is discussed in Chapter 9. Most critical for nephron function is the differentiation of a vascularised glomerulus with podocytes and endothelial cells forming a functional glomerular basement membrane. The molecular regulation of this process will be discussed in Chapter 10. Chapter 11 then discusses how different segments of these elongating and maturing nephrons are directed to the appropriate location to form the final organ. The influence of the surrounding stroma on this process is covered by Chapter 12. Finally, our current understanding of how the vasculature of the kidney arises will be presented in Chapter 13.

    Chapter 1

    Zebrafish Renal Development and Regeneration

    Rachel C. Dodd,  and Alan J. Davidson     Department of Molecular Medicine & Pathology, School of Medical Sciences, Faculty of Medical Health Sciences, The University of Auckland, Auckland, New Zealand

    Abstract

    The zebrafish is a useful genetic and embryological model system for investigating renal development, regeneration, and disease. In Zebrafish, kidney development progresses through two types of kidney: the pronephros, a fully functional, two-nephron embryonic kidney and the mesonephros, the adult kidney comprising hundreds of nephrons. Zebrafish nephrons show considerable conservation in structure and function compared with mammalian nephrons, with both exhibiting a common glomerular architecture and a similar tubule segmentation pattern. Unlike mammals, however, the zebrafish mesonephric kidney maintains a nephron progenitor population into adulthood and continues to generate new nephrons throughout life in response to growth or injury. The zebrafish model is used to study nephron formation and to understand the neonephrogenic growth/regenerative response, which may lead to the development of novel therapies for the treatment of human renal disease.

    Keywords

    Embryo; Kidney; Mesonephros; Nephron; Pronephros; Regeneration; Zebrafish

    Overview

    The vertebrate kidney plays critical roles in body fluid homeostasis and metabolic waste removal via blood filtration and the excretion/absorption of solutes and water. The workhorse of the kidney is the nephron, which is structurally and functionally conserved among vertebrates and comprises two major parts: the glomerular blood filter and the renal tubules. The ultrafiltrate produced by the glomerulus enters the tubules and is sequentially modified via selective solute reabsorption and secretion [1]. In general, the tubule can be broadly divided into proximal tubule segments, where the bulk of filtrate reabsorption takes place; intermediate segments for the concentration of urine (found only in reptiles, birds, and mammals); and distal tubule segments, where more specialized fine-tuning of salt and acid–base balance takes place. This process is vital for the regulation of electrolyte levels, osmolarity, blood pressure, and pH balance, and disruption of nephron function manifests as a variety of clinical conditions.

    How the nephron forms during kidney organogenesis remains poorly understood, in part because the complex architecture of the mammalian kidney renders close investigation of nephrogenesis a challenge. In contrast, lower vertebrate models such as the zebrafish develop comparatively simple embryonic kidneys. The transparency of zebrafish embryos coupled with their external development, rapid growth, and a range of genetic tools means that gene expression and function can be readily assessed at a high cellular resolution. As a result, the zebrafish is providing new insights into how the nephron forms during embryogenesis.

    Nephron regeneration also has recently become the focus of intense study. The mammalian nephron has a limited capacity for regeneration but is able to repair localized lesions in the proximal tubule. However, the ability of mammals to generate nephrons is restricted to stages of embryonic or early postnatal life, when the kidney is undergoing organogenesis. In contrast, zebrafish and other teleosts retain the ability to generate new nephrons during adulthood in response to growth or injury. This pathway of de novo nephron formation (called neonephrogenesis) is poorly understood but might provide key insights into developing novel regeneration-based therapies for humans. This chapter focuses on the use of zebrafish to understand nephrogenesis in both developmental and regenerative contexts. How zebrafish have advanced our understanding of the developmental pathways controlling nephron formation, as well as an examination of the neonephrogenic response that occurs in the adult zebrafish kidney, are discussed.

    The Zebrafish Pronephros as a Model of Nephron Development and Injury

    Overview of the Zebrafish Pronephros

    Whereas the mammalian pronephros is vestigial and restricted to the nephric duct lineage, the zebrafish pronephros is a fully functional kidney that is essential for osmoregulation in the free-swimming embryo. The zebrafish pronephros consists of two bilaterally paired nephrons that are fused rostrally at the midline, via a shared glomerulus, and caudally at the cloaca [2] (Figure 1.1). The zebrafish glomerulus shows a high degree of conservation with mammalian glomeruli, with centrally located fenestrated endothelial cells supported by mesangial cells [3], a thick glomerular basement membrane, and an outer layer of podocytes with long, interdigitating foot processes. Zebrafish podocytes are linked together via slit diaphragm protein bridges and express a number of genes identified from mammalian studies as being important for podocyte function, including slit diaphragm components (nephrin, neph1, podocin), glomerular basement membrane adhesion (integrin α3), and electronegative surface charge (podocalyxin) [4–7].

    Linking the glomerulus to the tubules is a short neck region made up of low cuboidal epithelial cells and scattered multiciliated cells [6]. The neck region is not found in mammals but has been observed in other fish species; it is likely to play a role in propelling the filtrate from the glomerulus into the proximal tubule [8,9]. In support of this, disruptions in cilia function lead to a characteristic accumulation of fluid in the neck region, resulting in bilateral dilatations or cysts that compress the adjacent glomerulus [5,10,11]. The neck region is characterized by the expression of the pax2a transcription factor and a salt-and-pepper pattern of genes implicated in cilia function, including rfx2, foxj1a, and dnah9 [6,12]. Although the neck region was initially thought to function as a tubule segment, it lacks expression of conserved proximal tubule genes, does not exhibit a brush border, and fails to take up fluorescent tracers that pass through the glomerular filter [6]. Instead, knockdown studies of pathways required for podocyte formation suggest that neck cells may be more closely related to the podocyte lineage, perhaps sharing a common progenitor [6].

    The pronephric tubules exhibit a segmentation pattern that is characterized by two proximal tubule segments (proximal convoluted tubule [PCT] and proximal straight tubule [PST]) and two distal segments (distal early [DE] and distal late [DL]), as revealed by the expression of segment-specific genes and cellular morphology [12–15] (Figure 1.1). As in mammals, the zebrafish proximal tubule segments display all the hallmarks of highly absorptive epithelia, including a prominent brush border, uptake of fluorescent tracers, expression of megalin and cubilin (encoding endocytic scavenging receptors), and specific solute transporters [12,16]. The DE and DL segments express NaCl transporters (slc12a1 and slc12a3, respectively), as well as the chloride channel clcnkb, and are likely to play a key role in NaCl reabsorption similar to that of the distal nephron in mammalians. Notable differences in tubule structure between zebrafish and mammals include a lack of the loop of Henle in zebrafish, which is an adaptation for concentrating the urine in metanephric kidneys, and the presence of motile multiciliated cells in the zebrafish PCT, PST, and DE segments.

    Induction of the Zebrafish Pronephros from the Intermediate Mesoderm

    As in all vertebrates, the zebrafish pronephros develops from the intermediate mesoderm during embryogenesis. However, zebrafish differ from other model vertebrates in that the pronephros arises in situ from most of the intermediate mesoderm, and there is not extensive migration of nephric duct precursors, such as that seen in mammalian embryos (Figure 1.2). As a result, the different cell types along the proximodistal axis of the nephron arise from progenitors laid out along the anteroposterior axis of the intermediate mesoderm. The zebrafish intermediate mesoderm can first be discerned shortly after the end of gastrulation by the expression of the transcription factor genes pax2a, pax8, osr1, and lhx1a [17–19]. Of these genes, pax2a and pax8 play a key role as inducers of pronephros development in zebrafish. Embryos deficient in pax2a and pax8 fail to form pronephric tubules and lack the expression of a number of early acting renal genes, including the transcription factor hnf1b [13]. This result is consistent with mouse studies in which embryos deficient in both Pax2 and Pax8 do not form the nephric duct (the earliest renal lineage to be specified in mammals), and there is a corresponding failure in mesonephric and metanephric kidney induction [20]. How Pax2/8 specify renal fates remains poorly understood and in mammals is complicated by their roles at multiple stages and in different cell types during kidney development. A number of candidate targets of Pax2 and Pax8 have recently been identified in the mouse using microarray profiling and include several other renal transcriptional regulators, such as Lhx1, Gata3, Mecom, and Plac8 [21]. This suggests that Pax2/8 act near the top of a much larger transcriptional network that controls the specification of kidney fate (Figure 1.3). During pronephros development in zebrafish, the expression of Lhx1, Mecom, and Plac8 orthologs also is dependent on Pax2/8, suggesting that the Pax2/8 transcriptional cascade is conserved across vertebrates (unpublished observations and Refs [19,22]).

    Figure 1.1  Schematic showing the development and structure of the zebrafish pronephros.

    The pronephros derives from bilateral stripes of intermediate mesoderm (blue) that are discernible after gastrulation by the expression of transcription factor genes such as pax2a and pax8. As development proceeds, the intermediate mesoderm can be subdivided into podocyte/neck segment progenitors, rostral progenitors, and caudal progenitors. Podocyte/neck cells migrate toward the midline and fuse (indicated by arrows). By 48  h after fertilization, the pronephros comprises a two-nephron structure with a shared glomerulus (green ovals), two proximal convoluted tubule segments (red), two proximal straight tubule segments (yellow), two distal early segments (blue), and two distal late segments (purple).

    Figure 1.2  Comparison of zebrafish (left) and mouse (right) kidney development from the intermediate mesoderm.

    Both zebrafish and mouse kidneys derive from the intermediate mesoderm along the trunk (blue). Unlike other vertebrate models, the zebrafish pronephros differentiates in situ from most of the anteroposterior axis of the intermediate mesoderm, such that progenitors along this axis sequentially give rise to the different cell types of the pronephric nephron (see Figure 1.1 for details). By contrast, during mouse kidney development, the rostral intermediate mesoderm gives rise to nephric duct precursors that migrate caudally and induce the formation of the mesonephros and metanephros.

    Formation of the Pronephric Glomerulus

    Laser ablation studies have demonstrated that podocytes arise from the rostral-most domain of the pax2/8/lhx1a+ intermediate mesoderm, adjacent to somite 3 [6]. In addition to expressing pax2/8/lhx1a, this region also expresses the zinc finger transcription factors Wilms’ tumor suppressor 1a (wt1a) and odd-skipped related 1 (osr1) [10,23]. During later stages of pronephros development, these podocyte progenitors initiate expression of mafba and wt1b, migrate to the midline [6], and fuse (Figures 1.1 and 1.4). During this time, there is transient expression of the Notch target gene hey1 in these cells (discussed in more detail below). Expression of podocyte differentiation markers such as nephrin, podocin, integrin α3, and podocalyxin become expressed just before fusion, concomitant with a downregulation of pax2/8, lhx1a, osr1, and hey1 and stronger expression of wt1a [6]. These molecular and morphological patterns suggest a sequential process whereby the intermediate mesoderm is converted into podocyte progenitors, followed by their terminal differentiation (Figure 1.4). Vascular sprouts from the overlying dorsal aorta invade the mass of fused podocytes and establish the glomerular capillary tuft, followed by mesangial cell recruitment [3]. Glomerular filtration initiates around 48  h after fertilization, with mature size selectivity being achieved 4  days after fertilization [5].

    Considerable evidence in mammals has implicated WT1 as a key regulator of podocyte fate [24–28]. Consistent with this, morpholino-mediated knockdown of wt1a in zebrafish causes a reduction, but not a complete loss, of podocyte progenitors and a failure of these cells to express differentiation markers such as nephrin and podocalyxin [6,29,30]. Knockdown of the wt1b paralog alone does affect early podocyte development, but at later stages it leads to the formation of neck cysts in some animals [30]. Embryos deficient in wt1a and wt1b seem to lack all podocytes, neck cells, and a portion of the proximal tubule [30,31]. While these data suggest that wt1a and wt1b may act redundantly during podocyte, neck, and proximal tubule development, a more detailed analysis of the doubly deficient phenotype is needed to understand the cause of these defects, given that endogenous transcripts for wt1a and wt1b have not been reported in proximal tubule progenitors. Furthermore, wt1b expression persists in podocytes in wt1a-deficient embryos, yet the podocytes still fail to undergo terminal differentiation [6]. This result suggests that wt1a and wt1b have independent functions, at least with regard to podocyte terminal differentiation.

    Figure 1.3  Overview of gene expression changes during podocyte formation in zebrafish.

    Podocyte formation during zebrafish pronephros development is characterized by the sequential differentiation of the intermediate mesoderm, first into podocyte progenitors and then mature podocytes. Selected genes expressed by cells at each stage are listed. The expression patterns of Notch ligands (jagged2 and jagged3), the Notch target hey1, and functional in vivo studies (see text for details) suggest that Notch signaling operates during the early, but not late phases, of podocyte formation.

    Figure 1.4  Proposed gene regulatory network controlling proximodistal patterning of the intermediate mesoderm in zebrafish.

    Diffusion of retinoic acid (blue) across the rostral domain of the intermediate mesoderm likely establishes the proximodistal pattern of the pronephros through a morphogenetic gradient. Pax2/8 and Hnf1b are key regulators of tubule segmentation and maturation, acting downstream or in parallel with RA signaling, although the targets of these transcription factors are still being identified. Mecom, a potential Pax2/8 target, may repress RA signaling. Irx3b, a potential Hnf1b target, is needed to maintain the identity of the distal early segment. Hnf1b factors also play a role in restricting podocyte fate, acting upstream or in parallel to the Wt1a/Notch pathway. Putative positive and negative interactions are indicated with green and red lines, respectively.

    An antagonistic relationship may exist between wt1a and pax2a in zebrafish because pax2a mutants show ectopic expression of wt1a and vegf (normally restricted to podocytes) in the neck region [32]. This suggests that in the absence of pax2a, the neck region may adopt a podocyte fate. Mammalian studies have provided good evidence that WT1 can directly repress Pax2 expression based on in vitro promoter assays, and observations in vivo have revealed an inverse relationship between WT1 and Pax2 expression in podocytes [33,34]. Although wt1a and pax2a are coexpressed during early stages of podocyte development, the finding that pax2a transcripts are downregulated around the time that wt1a expression increases suggests that the antagonistic relationship between Wt1 and Pax2 homologs is conserved in zebrafish.

    In mammals the Notch pathway is required for podocyte formation during metanephric kidney formation, and a similar role is conserved in zebrafish [6,35–37]. Knockdown of rbpj (a transcriptional mediator of Notch) or the Notch ligands jagged2 and jagged3 reduces the number of podocytes, similar to what occurs in wt1a-deficient embryos, but does not block their differentiation [6]. Embryos deficient in both wt1a and rbpj lack all podocytes, suggesting that these two factors act together, perhaps in a partially redundant fashion, to induce podocyte fate. One possibility is that Notch signaling and Wt1a converge on common downstream targets, with both pathways contributing fractionally to the final transcription level of the target. In support of this, Rbpj and Wt1 physically associate, and the Notch pathway and Wt1 show additive stimulatory effects on the mouse Hey1 promoter in vitro [6]. The role of hey1 in zebrafish podocytes is not clear, but its transient expression suggests that Notch signaling is restricted to the progenitor phase and is downregulated before terminal differentiation (Figure 1.4). Notch signaling does not seem to be compatible with the fully differentiated state of podocytes; ectopic Notch activation in podocytes induces a de-differentiated-like phenotype characterized by reactivation of Pax2 and Jagged expression and a downregulation of WT1 and mature markers such as Nephrin and Podocin [36,38].

    Osr1, encoding a zinc finger transcription factor belonging to the odd-skipped family of genes, has recently been shown to play a role in zebrafish podocyte formation [31]. Knockdown of osr1 leads to a glomerular phenotype similar to that of wt1a-deficient embryos, including fewer podocytes that remain in a progenitor state and fail to express nephrin and podocin [17,31]. Expression of wt1a is unaffected in osr1-deficient embryos, ruling out that osr1 acts as an upstream inducer of wt1a [17], but it may act downstream or parallel to wt1a and the Notch pathway [31]. Support for the latter pathway comes from Drosophila work, where the odd-skipped gene family, downstream of Notch, has been implicated in promoting morphological changes associated with joint formation during leg development [39]. One potential target of osr1 is the lhx1a gene, which is lost in osr1 knockdown embryos. Morpholino-mediated knockdown of lhx1a does not cause an overt podocyte defect (our unpublished observations). However, overexpression of a constitutively active version of Lhx1a has been reported to partially restore nephrin expression in osr1-deficient animals, raising the possibility that an osr1  →  lhx1a  →  nephrin pathway may exist in zebrafish [31]. This is a surprising result because lhx1a downregulates around the time that nephrin becomes expressed [6], and nephrin is considered a WT1 target [40]. Thus, further work is needed to clarify the requirement of lhx1a in the podocyte lineage.

    Proximodistal Patterning of the Pronephros

    Dynamic Expression Patterns of Renal Transcription Factor Genes

    Development of the segmental pattern of the tubule is vital for establishing the regulated reabsorption and secretion of solutes. Currently, there is only a poor understanding of the developmental pathways that establish nephron segmentation and maintain segment identity. This is an area where the zebrafish model excells because gene function can be easily assessed using morpholinos or reverse genetic approaches, and the relatively linear tubules of the pronephros facilitate fine mapping of gene expression patterns. Proximodistal subdivisions of the pax2/pax8/lhx1a+ intermediate mesoderm are evident shortly after gastrulation has completed, with rostrally restricted expression of wt1a and jagged2 (including both podocyte/neck progenitors and some proximal tubule progenitors) and caudally restricted expression of the zinc finger transcription factor gene mecom [12,15,41,42]. Following this early rostral–caudal patterning, additional transcription factor genes are expressed in nested domains, including the iroquois homeobox protein 3b (irx3b) that demarcates progenitors of the PST and DE segments [15]. The expression domains of many of these transcription factors fluctuate over the course of kidney development, but further studies are needed to elucidate how many of these changes reflect alterations in gene expression and how many result from changes in cell migration or cell shape [15]. By 24  h after fertilization, the four nephron segments (PCT, PST, DE, and DL) can be distinguished based on the expression of distinct cohorts of solute transporter genes and transcription factors (Figure 1.1). For instance, the PCT is demarcated by expression of slc20a1a, the PST by trpm7, the DE segment by slc12a1, and the DL segment by slc12a3 [12,14].

    Role of the Retinoic Acid Pathway

    Retinoic acid (RA), the active derivative of vitamin A, is a diffusible morphogen that exerts its effects through concentration-dependent activity and plays a major role in establishing the proximodistal pattern of the pronephros (Wingert et al. [12]). Upon entering the nucleus, RA binds to the RA receptor (RAR) family of transcription factors, resulting in transcriptional activation of RA-responsive target genes [43]. Studies have examined the effects of blocking RA signaling during zebrafish development, either in mutants deficient in aldh1a2 (encoding an enzyme responsible for RA synthesis) or via the application of pathway inhibitors such as diethylaminobenzaldehyde that blocks Aldh1a2. When the RA pathway is compromised during gastrulation, the resulting RA-deficient embryos lack podocytes, show a reduction in the length of the proximal segments, and have an expansion of the distal segments [12,44]. Exogenous RA treatment induces the opposite phenotype and proximalizes the pronephros, resulting in tubules made up of PCT and PST segments only. Adding RA and diethylaminobenzaldehyde for different lengths of time varies the severity of the proximalization and distalization phenotypes, respectively, leading to the suggestion that the pronephric segmentation pattern is set by a gradient of RA, similar to that proposed for the hindbrain [12,45,46]. Consistent with such a model, expression of aldh1a2 is restricted to the anterior paraxial mesoderm, adjacent to proximal nephron progenitors, during early stages of pronephros development [12]. Direct visualization of RA gradients in the zebrafish embryo was recently achieved using a fluorescent reporter capable of detecting free RA [47]. This reporter showed that in the early postgastrula embryo a gradient of RA extends out from the upper trunk and is likely to encompass rostral portions of the intermediate mesoderm, consistent with the notion that a morphogenic gradient specifies different nephron fates. Whether a nephron-patterning role for RA is conserved across vertebrates remains uncertain. While RA is needed for the branching of the mammalian metanephric-collecting duct system, a requirement for nephrogenesis has not been shown [48–52]. Despite this, a number of RA biosynthetic and degradative pathway genes are expressed during mammalian nephron formation, and it is possible that redundancy between these genes masks a patterning role for RA in mammals [53].

    Establishment of Proximodistal Segment Identities Downstream of RA

    How RA modulates patterning of the pronephros remains largely unknown. It is clear that RA exerts its actions on the intermediate mesoderm at very early stages of pronephros development. In the case of glomerular fates, the effect of RA may be direct; the wt1a promoter is bound by members of the RAR family and is responsive to RA concentrations in vivo [54]. In addition to a loss of wt1a, RA-deficient embryos also fail to express jagged2 (consistent with a lack of podocyte specification) and show an expansion of the caudal domain marker mecom [12,41].

    In the case of tubule progenitors, there is growing evidence that several transcription factors act downstream of RA, either directly or indirectly, to establish pronephric segment boundaries. Mecom was initially identified as an oncogene for myeloid tumors in mice, and null mutants die around embryonic day 10.5, with widespread abnormalities including hypocellular mesonephroi [55,56]. During pronephros development in zebrafish, RA signaling seems to restrict mecom expression to the caudal domain of the intermediate mesoderm, where RA concentrations are low [15,31] (Figure 1.3). Knockdown of mecom results in a proximalized pronephros phenotype, similar to that of embryos exposed to exogenous RA, and suggests that Mecom antagonizes RA signaling [41]. Consistent with this, treating mecom-deficient embryos with low concentrations of exogenous RA enhances the proximalized phenotype, suggesting that mecom deficiency sensitizes the intermediate mesoderm to the proximalizing effects of RA. How exactly Mecom interacts with the RA pathway is not yet known. Mecom can interfere directly with the stimulatory action of RARs on its own promoter, raising the possibility that it acts during pronephros development as a direct repressor of RA target genes [57]. How

    Enjoying the preview?
    Page 1 of 1