Atp6ap2 deletion causes extensive vacuolation that consumes the insulin content of pancreatic β cells

Significance Cells maintain several mechanisms to ensure their survival, including the removal of old or damaged proteins and organelles. This process must be balanced: too little turnover results in the accumulation of cellular “junk,” while excessive removal can deplete the cell and organism of key components. Here, we show that Atp6ap2 in the pancreatic β cell is essential for insulin granule turnover, as its absence resulted in the accumulation of oversized cytosolic vacuoles which conceivably account for excessive granule degradation, and thereby leads to impaired insulin secretion and diabetes. Pancreatic β cells store insulin within secretory granules which undergo exocytosis upon elevation of blood glucose levels. Crinophagy and autophagy are instead responsible to deliver damaged or old granules to acidic lysosomes for intracellular degradation. However, excessive consumption of insulin granules can impair β cell function and cause diabetes. Atp6ap2 is an essential accessory component of the vacuolar ATPase required for lysosomal degradative functions and autophagy. Here, we show that Cre recombinase-mediated conditional deletion of Atp6ap2 in mouse β cells causes a dramatic accumulation of large, multigranular vacuoles in the cytoplasm, with reduction of insulin content and compromised glucose homeostasis. Loss of insulin stores and gigantic vacuoles were also observed in cultured insulinoma INS-1 cells upon CRISPR/Cas9-mediated removal of Atp6ap2. Remarkably, these phenotypic alterations could not be attributed to a deficiency in autophagy or acidification of lysosomes. Together, these data indicate that Atp6ap2 is critical for regulating the stored insulin pool and that a balanced regulation of granule turnover is key to maintaining β cell function and diabetes prevention.

[1]  J. Chen,et al.  (Pro)renin Receptor RNA Interference Silencing Attenuates Diabetic Cardiomyopathy Pathological Process in Rats. , 2019, Human gene therapy.

[2]  T. Stevens,et al.  ATP6AP2 functions as a V-ATPase assembly factor in the endoplasmic reticulum , 2018, Molecular biology of the cell.

[3]  W. Chiu,et al.  The 3.5-Å CryoEM Structure of Nanodisc-Reconstituted Yeast Vacuolar ATPase Vo Proton Channel. , 2018, Molecular cell.

[4]  J. Debnath,et al.  Beyond self-eating: The control of nonautophagic functions and signaling pathways by autophagy-related proteins , 2018, The Journal of cell biology.

[5]  M. Solimena,et al.  FLIM-based pH measurements reveal incretin-induced rejuvenation of aged insulin secretory granules , 2017, bioRxiv.

[6]  A. Müller,et al.  A 4D view on insulin secretory granule turnover in the β‐cell , 2017, Diabetes, obesity & metabolism.

[7]  M. Hall,et al.  Loss of mTORC1 signalling impairs β-cell homeostasis and insulin processing , 2017, Nature Communications.

[8]  Nicholas J. Stam,et al.  Breaking up and making up: The secret life of the vacuolar H+‐ATPase , 2017, Protein science : a publication of the Protein Society.

[9]  R. Lüllmann-Rauch,et al.  Disruption of the vacuolar-type H+-ATPase complex in liver causes MTORC1-independent accumulation of autophagic vacuoles and lysosomes , 2017, Autophagy.

[10]  C. Robinson,et al.  Atomic model for the membrane-embedded VO motor of a eukaryotic V-ATPase , 2016, Nature.

[11]  F. Trepiccione,et al.  Renal Atp6ap2/(Pro)renin Receptor Is Required for Normal Vacuolar H+-ATPase Function but Not for the Renin-Angiotensin System. , 2016, Journal of the American Society of Nephrology : JASN.

[12]  N. Mohebbi,et al.  Colocalization of the (Pro)renin Receptor/Atp6ap2 with H+-ATPases in Mouse Kidney but Prorenin Does Not Acutely Regulate Intercalated Cell H+-ATPase Activity , 2016, PloS one.

[13]  M. Meima,et al.  Identification of the (Pro)renin Receptor as a Novel Regulator of Low-Density Lipoprotein Metabolism. , 2016, Circulation research.

[14]  M. Wright,et al.  New role for the (pro)renin receptor in T-cell development. , 2015, Blood.

[15]  Beth S. Lee,et al.  Vacuolar ATPase in Phagosome-Lysosome Fusion , 2015, The Journal of Biological Chemistry.

[16]  J. Grobe,et al.  Neuron-Specific (Pro)renin Receptor Knockout Prevents the Development of Salt-Sensitive Hypertension , 2014, Hypertension.

[17]  P. Fossier,et al.  The V-ATPase membrane domain is a sensor of granular pH that controls the exocytotic machinery , 2013, The Journal of cell biology.

[18]  David A. Scott,et al.  Genome engineering using the CRISPR-Cas9 system , 2013, Nature Protocols.

[19]  D. Muller,et al.  Autophagy and the (Pro)renin Receptor , 2013, Front. Endocrinol..

[20]  Ihor V. Yosypiv,et al.  Deletion of the Prorenin Receptor from the Ureteric Bud Causes Renal Hypodysplasia , 2013, PloS one.

[21]  O. Kretz,et al.  Vps34 deficiency reveals the importance of endocytosis for podocyte homeostasis. , 2013, Journal of the American Society of Nephrology : JASN.

[22]  B. Xiang,et al.  Pancreatic islet cell autophagy during aging in rats. , 2013, Clinical and investigative medicine. Medecine clinique et experimentale.

[23]  S. Beck,et al.  Drosophila ATP6AP2/VhaPRR functions both as a novel planar cell polarity core protein and a regulator of endosomal trafficking , 2013, The EMBO journal.

[24]  M. Foti,et al.  Improved characterization of the insulin secretory granule proteomes. , 2012, Journal of proteomics.

[25]  A. Danser,et al.  (Pro)renin and its receptors: pathophysiological implications. , 2012, Clinical science.

[26]  A. Müller,et al.  Novel standards in the measurement of rat insulin granules combining electron microscopy, high-content image analysis and in silico modelling , 2012, Diabetologia.

[27]  H. Eguchi,et al.  Rapamycin Causes Upregulation of Autophagy and Impairs Islets Function Both In Vitro and In Vivo , 2012, American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons.

[28]  H. Itoh,et al.  Prorenin receptor is essential for normal podocyte structure and function. , 2011, Journal of the American Society of Nephrology : JASN.

[29]  F. Luft,et al.  Prorenin receptor is essential for podocyte autophagy and survival. , 2011, Journal of the American Society of Nephrology : JASN.

[30]  R. de Cabo,et al.  Deletion of the mammalian INDY homolog mimics aspects of dietary restriction and protects against adiposity and insulin resistance in mice. , 2011, Cell metabolism.

[31]  Alexis A Gonzalez,et al.  Soluble Form of the (Pro)Renin Receptor Is Augmented in the Collecting Duct and Urine of Chronic Angiotensin II–Dependent Hypertensive Rats , 2011, Hypertension.

[32]  G. Walz,et al.  Regulation of Frizzled-Dependent Planar Polarity Signaling by a V-ATPase Subunit , 2010, Current Biology.

[33]  K. Fukuda,et al.  The (Pro)renin Receptor/ATP6AP2 is Essential for Vacuolar H+-ATPase Assembly in Murine Cardiomyocytes , 2010, Circulation research.

[34]  M. Cooper,et al.  RILLKKMPSV Influences the Vasculature, Neurons and Glia, and (Pro)Renin Receptor Expression in the Retina , 2010, Hypertension.

[35]  C. Cruciat,et al.  Requirement of Prorenin Receptor and Vacuolar H+-ATPase–Mediated Acidification for Wnt Signaling , 2010, Science.

[36]  K. Connelly,et al.  The (Pro)Renin Receptor: Site-Specific and Functional Linkage to the Vacuolar H+-ATPase in the Kidney , 2009, Hypertension.

[37]  U. Boggi,et al.  Autophagy in human type 2 diabetes pancreatic beta cells , 2009, Diabetologia.

[38]  Eeva-Liisa Eskelinen,et al.  Autophagy: a lysosomal degradation pathway with a central role in health and disease. , 2009, Biochimica et biophysica acta.

[39]  Kun Wook Chung,et al.  Loss of autophagy diminishes pancreatic beta cell mass and function with resultant hyperglycemia. , 2008, Cell metabolism.

[40]  Masaaki Komatsu,et al.  Autophagy is important in islet homeostasis and compensatory increase of beta cell mass in response to high-fat diet. , 2008, Cell metabolism.

[41]  A. Roebroek,et al.  Role of furin in granular acidification in the endocrine pancreas: Identification of the V-ATPase subunit Ac45 as a candidate substrate , 2008, Proceedings of the National Academy of Sciences.

[42]  D. Campbell,et al.  Critical review of prorenin and (pro)renin receptor research. , 2008, Hypertension.

[43]  Brad J Marsh,et al.  Regulated autophagy controls hormone content in secretory-deficient pancreatic endocrine beta-cells. , 2007, Molecular endocrinology.

[44]  P. Pandolfi,et al.  A Novel Signal Transduction Cascade Involving Direct Physical Interaction of the Renin/Prorenin Receptor With the Transcription Factor Promyelocytic Zinc Finger Protein , 2006, Circulation research.

[45]  A. Yamamoto,et al.  The a3 isoform of V-ATPase regulates insulin secretion from pancreatic β-cells , 2006, Journal of Cell Science.

[46]  M. Bader,et al.  Prorenin and its ancient receptor. , 2006, Hypertension.

[47]  S. Bonner-Weir,et al.  Five stages of evolving beta-cell dysfunction during progression to diabetes. , 2004, Diabetes.

[48]  B. Wolf,et al.  Rapamycin has a deleterious effect on MIN-6 cells and rat and human islets. , 2003, Diabetes.

[49]  G. Nguyen,et al.  Pivotal role of the renin/prorenin receptor in angiotensin II production and cellular responses to renin. , 2002, The Journal of clinical investigation.

[50]  K. Pfeiffer,et al.  Identification and Characterization of a Novel 9.2-kDa Membrane Sector-associated Protein of Vacuolar Proton-ATPase from Chromaffin Granules* , 1998, The Journal of Biological Chemistry.

[51]  L. Orci,et al.  Conversion of proinsulin to insulin occurs coordinately with acidification of maturing secretory vesicles , 1986, The Journal of cell biology.

[52]  L. Orci,et al.  Insulin, not C-peptide (proinsulin), is present in crinophagic bodies of the pancreatic B-cell , 1984, The Journal of cell biology.