Cell Cycle and Senescence Regulation by Podocyte Histone Deacetylase 1 and 2

Significance Statement The loss of integrity of the glomerular filtration barrier results in proteinuria that is often attributed to podocyte loss. Yet how damaged podocytes are lost remains unknown. Germline loss of murine podocyte-associated Hdac1 and Hdac2 (Hdac1/2) results in proteinuria and collapsing glomerulopathy due to sustained double-stranded DNA damage. Hdac1/2 deletion induces loss of podocyte quiescence, cell cycle entry, arrest in G1, and podocyte senescence, observed both in vivo and in vitro. Through the senescence secretory associated phenotype, podocytes secrete proteins that contribute to their detachment. These results solidify the role of HDACs in cell cycle regulation and senescence, providing important clues in our understanding of how podocytes are lost following injury. Background Intact expression of podocyte histone deacetylases (HDAC) during development is essential for maintaining a normal glomerular filtration barrier because of its role in modulating DNA damage and preventing premature senescence. Methods Germline podocyte-specific Hdac1 and 2 (Hdac1/2) double-knockout mice were generated to examine the importance of these enzymes during development. Results Podocyte-specific loss of Hdac1/2 in mice resulted in severe proteinuria, kidney failure, and collapsing glomerulopathy. Hdac1/2-deprived podocytes exhibited classic characteristics of senescence, such as senescence-associated β-galactosidase activity and lipofuscin aggregates. In addition, DNA damage, likely caused by epigenetic alterations such as open chromatin conformation, not only resulted in podocyte cell-cycle entry as shown in vivo by Ki67 expression and by FUCCI-2aR mice, but also in p21-mediated cell-cycle arrest. Through the senescence secretory associated phenotype, the damaged podocytes secreted proinflammatory cytokines, growth factors, and matrix metalloproteinases, resulting in subsequent podocyte detachment and loss, evidenced by senescent podocytes in urine. Conclusions Hdac1/2 plays an essential role during development. Loss of these genes in double knockout mice leads to sustained DNA damage and podocyte senescence and loss.

[1]  M. Bitzer,et al.  Upregulated PD-1 signaling antagonizes glomerular health in aged kidneys and disease , 2022, The Journal of clinical investigation.

[2]  L. Dworkin,et al.  Age-related GSK3β overexpression drives podocyte senescence and glomerular aging , 2022, The Journal of clinical investigation.

[3]  J. He,et al.  Controversies in Podocyte Loss: Death or Detachment? , 2021, Frontiers in Cell and Developmental Biology.

[4]  A. Jacinto,et al.  The right time for senescence , 2021, eLife.

[5]  Y. Nabeshima,et al.  Alternative Functions of Cell Cycle-Related and DNA Repair Proteins in Post-mitotic Neurons , 2021, Frontiers in Cell and Developmental Biology.

[6]  S. Shankland,et al.  Podocyte Aging: Why and How Getting Old Matters. , 2021, Journal of the American Society of Nephrology : JASN.

[7]  K. Lemley,et al.  Effect of disease progression on the podocyte cell cycle in Alport Syndrome. , 2021, Kidney international.

[8]  L. Dworkin,et al.  The ketone body β-hydroxybutyrate mitigates the senescence response of glomerular podocytes to diabetic insults. , 2021, Kidney international.

[9]  S. Venkatraman,et al.  The cGAS–STING pathway as a therapeutic target in inflammatory diseases , 2021, Nature Reviews Immunology.

[10]  P. Jat,et al.  Mechanisms of Cellular Senescence: Cell Cycle Arrest and Senescence Associated Secretory Phenotype , 2021, Frontiers in Cell and Developmental Biology.

[11]  J. Gil,et al.  Senescence and the SASP: many therapeutic avenues , 2020, Genes & development.

[12]  S. Ishibe,et al.  Murine Epsins Play an Integral Role in Podocyte Function. , 2020, Journal of the American Society of Nephrology : JASN.

[13]  Wei Zhang,et al.  Caspase-11/4 and gasdermin D-mediated pyroptosis contributes to podocyte injury in mouse diabetic nephropathy , 2020, Acta Pharmacologica Sinica.

[14]  S. Ishibe,et al.  Identification of Podocyte Cargo Proteins by Proteomic Analysis of Clathrin-Coated Vesicles. , 2020, Kidney360.

[15]  Chi D. Chu,et al.  CKD Awareness Among US Adults by Future Risk of Kidney Failure. , 2020, American journal of kidney diseases : the official journal of the National Kidney Foundation.

[16]  W. Wood,et al.  p16 a biomarker of aging and tolerance for cancer therapy , 2020, Translational cancer research.

[17]  J. Qu,et al.  The ageing epigenome and its rejuvenation , 2020, Nature Reviews Molecular Cell Biology.

[18]  C. Schmitt,et al.  Cellular Senescence: Defining a Path Forward , 2019, Cell.

[19]  P. Greengard,et al.  Loss of SATB1 Induces p21-Dependent Cellular Senescence in Post-mitotic Dopaminergic Neurons. , 2019, Cell stem cell.

[20]  L. Ferrucci,et al.  A proteomic atlas of senescence-associated secretomes for aging biomarker development , 2019, bioRxiv.

[21]  Alireza Hadj Khodabakhshi,et al.  Metascape provides a biologist-oriented resource for the analysis of systems-level datasets , 2019, Nature Communications.

[22]  A. Alimonti,et al.  Cellular Senescence: Aging, Cancer, and Injury. , 2019, Physiological reviews.

[23]  Zhenhai Zhang,et al.  Podocyte histone deacetylase activity regulates murine and human glomerular diseases , 2019, The Journal of clinical investigation.

[24]  Jeffrey N. McKnight,et al.  Condensin-Dependent Chromatin Compaction Represses Transcription Globally during Quiescence. , 2019, Molecular cell.

[25]  T. Ha,et al.  Puromycin aminonucleoside triggers apoptosis in podocytes by inducing endoplasmic reticulum stress , 2018, Kidney research and clinical practice.

[26]  Frédérick A. Mallette,et al.  Cellular Senescence in Postmitotic Cells: Beyond Growth Arrest. , 2018, Trends in cell biology.

[27]  M. Demaria,et al.  Hallmarks of Cellular Senescence. , 2018, Trends in cell biology.

[28]  O. Kovalchuk,et al.  Epigenetic Regulation of Cellular Senescence and Aging , 2017, Front. Genet..

[29]  Kyung Lee,et al.  Reduction in podocyte SIRT1 accelerates kidney injury in aging mice. , 2017, American journal of physiology. Renal physiology.

[30]  V. Gorgoulis,et al.  Sudan Black B, The Specific Histochemical Stain for Lipofuscin: A Novel Method to Detect Senescent Cells. , 2017, Methods in molecular biology.

[31]  J. Tyler,et al.  Epigenetics and aging , 2016, Science Advances.

[32]  M. Nagata,et al.  Podocyte injury and its consequences. , 2016, Kidney international.

[33]  S. Shankland,et al.  Cell cycle re-entry sensitizes podocytes to injury induced death , 2016, Cell cycle.

[34]  S. Somlo,et al.  Essential Role of X-Box Binding Protein-1 during Endoplasmic Reticulum Stress in Podocytes. , 2016, Journal of the American Society of Nephrology : JASN.

[35]  Zhihong Liu,et al.  Toll-like Receptor 9 Can be Activated by Endogenous Mitochondrial DNA to Induce Podocyte Apoptosis , 2016, Scientific Reports.

[36]  T. Uzu,et al.  Impaired Podocyte Autophagy Exacerbates Proteinuria in Diabetic Nephropathy , 2015, Diabetes.

[37]  S. Quaggin,et al.  The cell biology of renal filtration , 2015, The Journal of cell biology.

[38]  W. Huber,et al.  Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2 , 2014, Genome Biology.

[39]  Emmette R. Hutchison,et al.  Are there roles for brain cell senescence in aging and neurodegenerative disorders? , 2014, Biogerontology.

[40]  Atsushi Miyawaki,et al.  Fucci2a: A bicistronic cell cycle reporter that allows Cre mediated tissue specific expression in mice , 2014, Cell cycle.

[41]  Paul A. Wiggins,et al.  Estimating podocyte number and density using a single histologic section. , 2014, Journal of the American Society of Nephrology : JASN.

[42]  S. Ishibe,et al.  Podocyte-associated talin1 is critical for glomerular filtration barrier maintenance. , 2014, The Journal of clinical investigation.

[43]  Sharon Y. R. Dent,et al.  Chromatin modifiers and remodellers: regulators of cellular differentiation , 2013, Nature Reviews Genetics.

[44]  Wei Shi,et al.  featureCounts: an efficient general purpose program for assigning sequence reads to genomic features , 2013, Bioinform..

[45]  H. Anders,et al.  New insights into the pathology of podocyte loss: mitotic catastrophe. , 2013, The American journal of pathology.

[46]  Kai-Uwe Eckardt,et al.  Evolving importance of kidney disease: from subspecialty to global health burden , 2013, The Lancet.

[47]  H. Anders,et al.  Podocyte loss involves MDM2‐driven mitotic catastrophe , 2013, The Journal of pathology.

[48]  Kelly J. Morris,et al.  A complex secretory program orchestrated by the inflammasome controls paracrine senescence , 2013, Nature Cell Biology.

[49]  L. Cantley,et al.  The Terminator mouse is a diphtheria toxin receptor knockin mouse strain for rapid and efficient enrichment of desired cell lineages , 2013, Kidney international.

[50]  M. Moeller,et al.  Renal albumin filtration: alternative models to the standard physical barriers , 2013, Nature Reviews Nephrology.

[51]  A. D’Andrea,et al.  Chromatin Remodeling at DNA Double-Strand Breaks , 2013, Cell.

[52]  Jian-Kang Chen,et al.  mVps34 deletion in podocytes causes glomerulosclerosis by disrupting intracellular vesicle trafficking. , 2013, Journal of the American Society of Nephrology : JASN.

[53]  H. Anders,et al.  Podocyte Mitosis – A Catastrophe , 2012, Current molecular medicine.

[54]  J. Bartek,et al.  Specific lipofuscin staining as a novel biomarker to detect replicative and stress-induced senescence. A method applicable in cryo-preserved and archival tissues , 2012, Aging.

[55]  Thomas R. Gingeras,et al.  STAR: ultrafast universal RNA-seq aligner , 2013, Bioinform..

[56]  P. De Camilli,et al.  Role of dynamin, synaptojanin, and endophilin in podocyte foot processes. , 2012, The Journal of clinical investigation.

[57]  R. Fridman,et al.  Assessment of gelatinases (MMP-2 and MMP-9) by gelatin zymography. , 2012, Methods in molecular biology.

[58]  S. Jackson,et al.  Human HDAC1 and HDAC2 function in the DNA-damage response to promote DNA nonhomologous end-joining , 2010, Nature Structural &Molecular Biology.

[59]  Steven S. Foster,et al.  Histone deacetylase inhibitor induces DNA damage, which normal but not transformed cells can repair , 2010, Proceedings of the National Academy of Sciences.

[60]  S. Hatakeyama,et al.  Inhibition of podocyte FAK protects against proteinuria and foot process effacement. , 2010, Journal of the American Society of Nephrology : JASN.

[61]  C. Cohen,et al.  Autophagy influences glomerular disease susceptibility and maintains podocyte homeostasis in aging mice. , 2010, The Journal of clinical investigation.

[62]  J. Campisi,et al.  The senescence-associated secretory phenotype: the dark side of tumor suppression. , 2010, Annual review of pathology.

[63]  L. Magnaghi-Jaulin,et al.  Histone deacetylase inhibitors and genomic instability. , 2009, Cancer letters.

[64]  L. Gnudi,et al.  Mechanical forces and TGF β 1 reduce podocyte adhesion through α 3 β 1 integrin downregulation , 2009 .

[65]  L. Barisoni,et al.  Current views on collapsing glomerulopathy. , 2008, Journal of the American Society of Nephrology : JASN.

[66]  K. Suszták,et al.  The Notch pathway in podocytes plays a role in the development of glomerular disease , 2008, Nature Medicine.

[67]  J. Alao,et al.  Molecular Cancer BioMed Central Review The regulation of cyclin D1 degradation: roles in cancer development and the potential for therapeutic invention , 2007 .

[68]  Alain Verreault,et al.  Chromatin Challenges during DNA Replication and Repair , 2007, Cell.

[69]  M. Narita,et al.  Cellular senescence and chromatin organisation , 2007, British Journal of Cancer.

[70]  R. A. Baker,et al.  Cell Cycle Activation in Postmitotic Neurons is Essential for DNA Repair , 2007, Cell cycle.

[71]  W. Carter,et al.  Abnormal development of glomerular endothelial and mesangial cells in mice with targeted disruption of the lama3 gene. , 2006, Kidney international.

[72]  Steven F. Dowdy,et al.  Regulation of Late G1/S Phase Transition and APCCdh1 by Reactive Oxygen Species , 2006, Molecular and Cellular Biology.

[73]  D. DiMaio,et al.  Senescence-associated beta-galactosidase is lysosomal beta-galactosidase. , 2006, Aging cell.

[74]  G. Camussi,et al.  Statins prevent oxidized LDL-induced injury of glomerular podocytes by activating the phosphatidylinositol 3-kinase/AKT-signaling pathway. , 2005, Journal of the American Society of Nephrology : JASN.

[75]  Jian Kuang,et al.  Induction of p21 by p53 following DNA damage inhibits both Cdk4 and Cdk2 activities , 2005, Oncogene.

[76]  L. Holzman,et al.  Podocyte‐specific expression of cre recombinase in transgenic mice , 2003, Genesis.

[77]  M. Bitzer,et al.  Apoptosis in podocytes induced by TGF-beta and Smad7. , 2001, The Journal of clinical investigation.

[78]  V. D’Agati,et al.  Differential expression of cyclin-dependent kinase inhibitors in human glomerular disease: role in podocyte proliferation and maturation. , 2000, Kidney international.

[79]  S. Jackson,et al.  Regulation of p53 in response to DNA damage , 1999, Oncogene.

[80]  M. Davies,et al.  Myocyte loss in chronic heart failure , 1999, The Journal of pathology.

[81]  H. Nagase Cell surface activation of progelatinase A (proMMP-2) and cell migration , 1998, Cell Research.

[82]  C. Norbury,et al.  Cellular responses to DNA damage: cell-cycle checkpoints, apoptosis and the roles of p53 and ATM. , 1995, Trends in biochemical sciences.

[83]  C Roskelley,et al.  A biomarker that identifies senescent human cells in culture and in aging skin in vivo. , 1995, Proceedings of the National Academy of Sciences of the United States of America.

[84]  Y. Komatsu,et al.  Mitosis and the presence of binucleate cells among glomerular podocytes in diseased human kidneys. , 1995, Nephron.

[85]  G. Hannon,et al.  The p21 inhibitor of cyclin-dependent kinases controls DNA replication by interaction with PCNA , 1994, Nature.

[86]  Johannes Gerdes,et al.  Production of a mouse monoclonal antibody reactive with a human nuclear antigen associated with cell proliferation , 1983, International journal of cancer.

[87]  P. Vinay,et al.  Isolation of a pure suspension of rat proximal tubules. , 1981, The American journal of physiology.

[88]  A. Pardee,et al.  A restriction point for control of normal animal cell proliferation. , 1974, Proceedings of the National Academy of Sciences of the United States of America.