1
|
Naatz A, Bohl KS, Jones Lipinski RA, Nord JA, Gehant AL, Hansen PA, Smith BC, Corbett JA. Role of SIRT3 in the regulation of Gadd45α expression and DNA repair in β-cells. J Biol Chem 2025:108451. [PMID: 40147772 DOI: 10.1016/j.jbc.2025.108451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 03/09/2025] [Accepted: 03/22/2025] [Indexed: 03/29/2025] Open
Abstract
In previous studies, we have shown that growth arrest and DNA damage (Gadd) 45α is required for the repair of nitric oxide-mediated DNA damage in β-cells. Gadd45α expression is stimulated by nitric oxide and requires forkhead box protein (Fox) O1 and NAD+-dependent deacetylase activity. Based on inhibitor studies, we attributed this activity to Sirtuin (SIRT)1; however, the inhibitors used in this previous study also attenuate the deacetylase activity of SIRT2, 3, and 6. We now provide experimental evidence that SIRT1 is dispensable for β-cell expression of Gadd45α and that the mitochondrial localized isoform SIRT3, is required for DNA repair in β-cells. We show that siRNA knockdown of Sirt3 attenuates nitric oxide-stimulated Gadd45α mRNA accumulation in both wildtype and Sirt1-/- INS 832/13 cells as well as isolated rat islets, and that SIRT3 inhibition increases FoxO1 acetylation and attenuates DNA repair in response to nitric oxide. While SIRT3 is predominantly localized to mitochondria, a small fraction is localized in the nucleus of insulin-containing cells and functions to participate in the regulation of FoxO1-dependent, nitric oxide-stimulated DNA repair.
Collapse
Affiliation(s)
- Aaron Naatz
- From the Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, 53226
| | - Kelsey S Bohl
- From the Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, 53226
| | - Rachel A Jones Lipinski
- From the Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, 53226
| | - Joshua A Nord
- From the Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, 53226
| | - Alyssa L Gehant
- From the Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, 53226
| | - Polly A Hansen
- From the Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, 53226
| | - Brian C Smith
- From the Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, 53226
| | - John A Corbett
- From the Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, 53226.
| |
Collapse
|
2
|
Varghese SS, Hernandez-De La Peña AG, Dhawan S. Safeguarding genomic integrity in beta-cells: implications for beta-cell differentiation, growth, and dysfunction. Biochem Soc Trans 2024; 52:2133-2144. [PMID: 39364746 PMCID: PMC11555696 DOI: 10.1042/bst20231519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2024]
Abstract
The maintenance of optimal glucose levels in the body requires a healthy reserve of the insulin producing pancreatic beta-cells. Depletion of this reserve due to beta-cell dysfunction and death results in development of diabetes. Recent findings highlight unresolved DNA damage as a key contributor to beta-cell defects in diabetes. Beta-cells face various stressors and metabolic challenges throughout life, rendering them susceptible to DNA breaks. The post-mitotic, long-lived phenotype of mature beta-cells further warrants robust maintenance of genomic integrity. Failure to resolve DNA damage during beta-cell development, therefore, can result in an unhealthy reserve of beta-cells and predispose to diabetes. Yet, the molecular mechanisms safeguarding beta-cell genomic integrity remain poorly understood. Here, we focus on the significance of DNA damage in beta-cell homeostasis and postulate how cellular expansion, epigenetic programming, and metabolic shifts during development may impact beta-cell genomic integrity and health. We discuss recent findings demonstrating a physiological role for DNA breaks in modulating transcriptional control in neurons, which share many developmental programs with beta-cells. Finally, we highlight key gaps in our understanding of beta-cell genomic integrity and discuss emerging areas of interest.
Collapse
Affiliation(s)
- Sneha S. Varghese
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA 91010, U.S.A
| | | | - Sangeeta Dhawan
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA 91010, U.S.A
| |
Collapse
|
3
|
Naatz A, Yeo CT, Hogg N, Corbett JA. β-Cell-selective regulation of gene expression by nitric oxide. Am J Physiol Regul Integr Comp Physiol 2024; 326:R552-R566. [PMID: 38586887 PMCID: PMC11381020 DOI: 10.1152/ajpregu.00240.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 03/07/2024] [Accepted: 04/02/2024] [Indexed: 04/09/2024]
Abstract
Nitric oxide is produced at low micromolar levels following the induction of inducible nitric oxide synthase (iNOS) and is responsible for mediating the inhibitory actions of cytokines on glucose-stimulated insulin secretion by islets of Langerhans. It is through the inhibition of mitochondrial oxidative metabolism, specifically aconitase and complex 4 of the electron transport chain, that nitric oxide inhibits insulin secretion. Nitric oxide also attenuates protein synthesis, induces DNA damage, activates DNA repair pathways, and stimulates stress responses (unfolded protein and heat shock) in β-cells. In this report, the time- and concentration-dependent effects of nitric oxide on the expression of six genes known to participate in the response of β-cells to this free radical were examined. The genes included Gadd45α (DNA repair), Puma (apoptosis), Hmox1 (antioxidant defense), Hsp70 (heat shock), Chop (UPR), and Ppargc1α (mitochondrial biogenesis). We show that nitric oxide stimulates β-cell gene expression in a narrow concentration range of ∼0.5-1 µM or levels corresponding to iNOS-derived nitric oxide. At concentrations greater than 1 µM, nitric oxide fails to stimulate gene expression in β-cells, and this is associated with the inhibition of mitochondrial oxidative metabolism. This narrow concentration range of responses is β-cell selective, as the actions of nitric oxide in non-β-cells (α-cells, mouse embryonic fibroblasts, and macrophages) are concentration dependent. Our findings suggest that β-cells respond to a narrow concentration range of nitric oxide that is consistent with the levels produced following iNOS induction, and that these concentration-dependent actions are selective for insulin-containing cells.
Collapse
Affiliation(s)
- Aaron Naatz
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Chay Teng Yeo
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Neil Hogg
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - John A Corbett
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| |
Collapse
|
4
|
Yousefzadeh MJ, Huerta Guevara AP, Postmus AC, Flores RR, Sano T, Jurdzinski A, Angelini L, McGowan SJ, O’Kelly RD, Wade EA, Gonzalez-Espada LV, Henessy-Wack D, Howard S, Rozgaja TA, Trussoni CE, LaRusso NF, Eggen BJ, Jonker JW, Robbins PD, Niedernhofer LJ, Kruit JK. Failure to repair endogenous DNA damage in β-cells causes adult-onset diabetes in mice. AGING BIOLOGY 2023; 1:20230015. [PMID: 38124711 PMCID: PMC10732477 DOI: 10.59368/agingbio.20230015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
Age is the greatest risk factor for the development of type 2 diabetes mellitus (T2DM). Age-related decline in organ function is attributed to the accumulation of stochastic damage, including damage to the nuclear genome. Islets of T2DM patients display increased levels of DNA damage. However, whether this is a cause or consequence of the disease has not been elucidated. Here, we asked if spontaneous, endogenous DNA damage in β-cells can drive β-cell dysfunction and diabetes, via deletion of Ercc1, a key DNA repair gene, in β-cells. Mice harboring Ercc1-deficient β-cells developed adult-onset diabetes as demonstrated by increased random and fasted blood glucose levels, impaired glucose tolerance, and reduced insulin secretion. The inability to repair endogenous DNA damage led to an increase in oxidative DNA damage and apoptosis in β-cells and a significant loss of β-cell mass. Using electron microscopy, we identified β-cells in clear distress that showed an increased cell size, enlarged nuclear size, reduced number of mature insulin granules, and decreased number of mitochondria. Some β-cells were more affected than others consistent with the stochastic nature of spontaneous DNA damage. Ercc1-deficiency in β-cells also resulted in loss of β-cell function as glucose-stimulated insulin secretion and mitochondrial function were impaired in islets isolated from mice harboring Ercc1-deficient β-cells. These data reveal that unrepaired endogenous DNA damage is sufficient to drive β-cell dysfunction and provide a mechanism by which age increases the risk of T2DM.
Collapse
Affiliation(s)
- Matthew J. Yousefzadeh
- Department of Molecular Medicine and the Center on Aging, The Scripps Research Institute, 130 Scripps Way #3B3, Jupiter FL, 33458, USA
- Department of Biochemistry, Molecular Biology and Biophysics and Institute on the Biology of Aging and Metabolism, University of Minnesota, 6-155 Jackson Hall, 321 Church St., Minneapolis, MN 55455, USA
| | - Ana P. Huerta Guevara
- Department of Pediatrics, Section Molecular Metabolism and Nutrition, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Andrea C. Postmus
- Department of Pediatrics, Section Molecular Metabolism and Nutrition, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Rafael R. Flores
- Department of Molecular Medicine and the Center on Aging, The Scripps Research Institute, 130 Scripps Way #3B3, Jupiter FL, 33458, USA
- Department of Biochemistry, Molecular Biology and Biophysics and Institute on the Biology of Aging and Metabolism, University of Minnesota, 6-155 Jackson Hall, 321 Church St., Minneapolis, MN 55455, USA
| | - Tokio Sano
- Department of Molecular Medicine and the Center on Aging, The Scripps Research Institute, 130 Scripps Way #3B3, Jupiter FL, 33458, USA
| | - Angelika Jurdzinski
- Department of Pediatrics, Section Molecular Metabolism and Nutrition, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Luise Angelini
- Department of Molecular Medicine and the Center on Aging, The Scripps Research Institute, 130 Scripps Way #3B3, Jupiter FL, 33458, USA
- Department of Biochemistry, Molecular Biology and Biophysics and Institute on the Biology of Aging and Metabolism, University of Minnesota, 6-155 Jackson Hall, 321 Church St., Minneapolis, MN 55455, USA
| | - Sara J. McGowan
- Department of Molecular Medicine and the Center on Aging, The Scripps Research Institute, 130 Scripps Way #3B3, Jupiter FL, 33458, USA
- Department of Biochemistry, Molecular Biology and Biophysics and Institute on the Biology of Aging and Metabolism, University of Minnesota, 6-155 Jackson Hall, 321 Church St., Minneapolis, MN 55455, USA
| | - Ryan D. O’Kelly
- Department of Molecular Medicine and the Center on Aging, The Scripps Research Institute, 130 Scripps Way #3B3, Jupiter FL, 33458, USA
- Department of Biochemistry, Molecular Biology and Biophysics and Institute on the Biology of Aging and Metabolism, University of Minnesota, 6-155 Jackson Hall, 321 Church St., Minneapolis, MN 55455, USA
| | - Erin A. Wade
- Department of Molecular Medicine and the Center on Aging, The Scripps Research Institute, 130 Scripps Way #3B3, Jupiter FL, 33458, USA
| | - Lisa V. Gonzalez-Espada
- Department of Molecular Medicine and the Center on Aging, The Scripps Research Institute, 130 Scripps Way #3B3, Jupiter FL, 33458, USA
| | - Danielle Henessy-Wack
- Department of Molecular Medicine and the Center on Aging, The Scripps Research Institute, 130 Scripps Way #3B3, Jupiter FL, 33458, USA
| | - Shannon Howard
- Department of Molecular Medicine and the Center on Aging, The Scripps Research Institute, 130 Scripps Way #3B3, Jupiter FL, 33458, USA
| | - Tania A. Rozgaja
- Department of Molecular Medicine and the Center on Aging, The Scripps Research Institute, 130 Scripps Way #3B3, Jupiter FL, 33458, USA
| | - Christy E. Trussoni
- Division of Gastroenterology and Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, MN 55905, USA
| | - Nicholas F. LaRusso
- Division of Gastroenterology and Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, MN 55905, USA
| | - Bart J.L. Eggen
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Johan W. Jonker
- Department of Pediatrics, Section Molecular Metabolism and Nutrition, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Paul D. Robbins
- Department of Molecular Medicine and the Center on Aging, The Scripps Research Institute, 130 Scripps Way #3B3, Jupiter FL, 33458, USA
- Department of Biochemistry, Molecular Biology and Biophysics and Institute on the Biology of Aging and Metabolism, University of Minnesota, 6-155 Jackson Hall, 321 Church St., Minneapolis, MN 55455, USA
| | - Laura J. Niedernhofer
- Department of Molecular Medicine and the Center on Aging, The Scripps Research Institute, 130 Scripps Way #3B3, Jupiter FL, 33458, USA
- Department of Biochemistry, Molecular Biology and Biophysics and Institute on the Biology of Aging and Metabolism, University of Minnesota, 6-155 Jackson Hall, 321 Church St., Minneapolis, MN 55455, USA
| | - Janine K. Kruit
- Department of Pediatrics, Section Molecular Metabolism and Nutrition, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
5
|
Yeo CT, Kropp EM, Hansen PA, Pereckas M, Oleson BJ, Naatz A, Stancill JS, Ross KA, Gundry RL, Corbett JA. β-cell-selective inhibition of DNA damage response signaling by nitric oxide is associated with an attenuation in glucose uptake. J Biol Chem 2023; 299:102994. [PMID: 36773802 PMCID: PMC10023961 DOI: 10.1016/j.jbc.2023.102994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 02/11/2023] Open
Abstract
Nitric oxide (NO) plays a dual role in regulating DNA damage response (DDR) signaling in pancreatic β-cells. As a genotoxic agent, NO activates two types of DDR signaling; however, when produced at micromolar levels by the inducible isoform of NO synthase, NO inhibits DDR signaling and DDR-induced apoptosis in a β-cell-selective manner. DDR signaling inhibition by NO correlates with mitochondrial oxidative metabolism inhibition and decreases in ATP and NAD+. Unlike most cell types, β-cells do not compensate for impaired mitochondrial oxidation by increasing glycolytic flux, and this metabolic inflexibility leads to a decrease in ATP and NAD+. Here, we used multiple analytical approaches to determine changes in intermediary metabolites in β-cells and non-β-cells treated with NO or complex I inhibitor rotenone. In addition to ATP and NAD+, glycolytic and tricarboxylic acid cycle intermediates as well as NADPH are significantly decreased in β-cells treated with NO or rotenone. Consistent with glucose-6-phosphate residing at the metabolic branchpoint for glycolysis and the pentose phosphate pathway (NADPH), we show that mitochondrial oxidation inhibitors limit glucose uptake in a β-cell-selective manner. Our findings indicate that the β-cell-selective inhibition of DDR signaling by NO is associated with a decrease in ATP to levels that fall significantly below the KM for ATP of glucokinase (glucose uptake) and suggest that this action places the β-cell in a state of suspended animation where it is metabolically inert until NO is removed, and metabolic function can be restored.
Collapse
Affiliation(s)
- Chay Teng Yeo
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Erin M Kropp
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Polly A Hansen
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Michael Pereckas
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Bryndon J Oleson
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Aaron Naatz
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Jennifer S Stancill
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Kyle A Ross
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Rebekah L Gundry
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - John A Corbett
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
| |
Collapse
|
6
|
Yang GH, Fontaine DA, Lodh S, Blumer JT, Roopra A, Davis DB. TCF19 Impacts a Network of Inflammatory and DNA Damage Response Genes in the Pancreatic β-Cell. Metabolites 2021; 11:metabo11080513. [PMID: 34436454 PMCID: PMC8400192 DOI: 10.3390/metabo11080513] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/31/2021] [Accepted: 08/02/2021] [Indexed: 12/13/2022] Open
Abstract
Transcription factor 19 (TCF19) is a gene associated with type 1 diabetes (T1DM) and type 2 diabetes (T2DM) in genome-wide association studies. Prior studies have demonstrated that Tcf19 knockdown impairs β-cell proliferation and increases apoptosis. However, little is known about its role in diabetes pathogenesis or the effects of TCF19 gain-of-function. The aim of this study was to examine the impact of TCF19 overexpression in INS-1 β-cells and human islets on proliferation and gene expression. With TCF19 overexpression, there was an increase in nucleotide incorporation without any change in cell cycle gene expression, alluding to an alternate process of nucleotide incorporation. Analysis of RNA-seq of TCF19 overexpressing cells revealed increased expression of several DNA damage response (DDR) genes, as well as a tightly linked set of genes involved in viral responses, immune system processes, and inflammation. This connectivity between DNA damage and inflammatory gene expression has not been well studied in the β-cell and suggests a novel role for TCF19 in regulating these pathways. Future studies determining how TCF19 may modulate these pathways can provide potential targets for improving β-cell survival.
Collapse
Affiliation(s)
- Grace H. Yang
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (G.H.Y.); (D.A.F.); (S.L.); (J.T.B.)
| | - Danielle A. Fontaine
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (G.H.Y.); (D.A.F.); (S.L.); (J.T.B.)
| | - Sukanya Lodh
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (G.H.Y.); (D.A.F.); (S.L.); (J.T.B.)
- Department of Biological Sciences, Marquette University, Milwaukee, WI 53233, USA
| | - Joseph T. Blumer
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (G.H.Y.); (D.A.F.); (S.L.); (J.T.B.)
| | - Avtar Roopra
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI 53705, USA;
| | - Dawn Belt Davis
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (G.H.Y.); (D.A.F.); (S.L.); (J.T.B.)
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
- Correspondence:
| |
Collapse
|
7
|
AboYoussef AM, Khalaf MM, Malak MN, Hamzawy MA. Repurposing of sildenafil as antitumour; induction of cyclic guanosine monophosphate/protein kinase G pathway, caspase-dependent apoptosis and pivotal reduction of Nuclear factor kappa light chain enhancer of activated B cells in lung cancer. J Pharm Pharmacol 2021; 73:1080-1091. [PMID: 33856030 DOI: 10.1093/jpp/rgab049] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 02/23/2021] [Indexed: 12/20/2022]
Abstract
OBJECTIVES Lung cancer is one of the most frequent types of cancers that lead to death. Sildenafil is a potent inhibitor of phosphodiesterase-5 and showed potential anticancer effects, which has not yet been fully evaluated. Thus, this study aims to investigate the potential anticancer effect of sildenafil in urethane-induced lung cancer in BALB/c mice. METHODS Five-week-old male BALB/c mice were treated with either (i) normal saline only, (ii) sildenafil only 50 mg kg-1/ P.O every other day for the last four successive weeks, (iii) urethane 1.5 gm kg-1 i.p (at day 1 and day 60), (iv) carboplatin after urethane induction, or (v) sildenafil after urethane induction. KEY FINDINGS It was shown that sildenafil significantly increased the levels of cGMP and Caspase-3 with a reduction of NF-κB, Bcl-2, Cyclin D1, intercellular adhesion molecule 1, matrix metalloproteinase-2 levels and normalisation of Nrf2 along with pronounced improvement in the histological patterns. CONCLUSIONS These results indicated that sildenafil markedly induces cell cycle arrest, apoptosis and inhibits the metastatic activity through activation of cyclic guanosine monophosphate/protein kinase G pathway and down-regulation of cyclin D1 and nuclear factor kappa light chain enhancer of activated B cells with downstream anti-apoptotic gene Bcl-2, which underscores the critical importance of future using sildenafil in the treatment of lung cancer.
Collapse
Affiliation(s)
- Amira M AboYoussef
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Marwa M Khalaf
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Marina N Malak
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Mohamed A Hamzawy
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Fayoum University, Fayoum, Egypt
| |
Collapse
|
8
|
Yeo CT, Stancill JS, Oleson BJ, Schnuck JK, Stafford JD, Naatz A, Hansen PA, Corbett JA. Regulation of ATR-dependent DNA damage response by nitric oxide. J Biol Chem 2021; 296:100388. [PMID: 33567339 PMCID: PMC7967039 DOI: 10.1016/j.jbc.2021.100388] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 02/02/2021] [Indexed: 02/01/2023] Open
Abstract
We have shown that nitric oxide limits ataxia-telangiectasia mutated signaling by inhibiting mitochondrial oxidative metabolism in a β-cell selective manner. In this study, we examined the actions of nitric oxide on a second DNA damage response transducer kinase, ataxia-telangiectasia and Rad3-related protein (ATR). In β-cells and non-β-cells, nitric oxide activates ATR signaling by inhibiting ribonucleotide reductase; however, when produced at inducible nitric oxide synthase-derived (low micromolar) levels, nitric oxide impairs ATR signaling in a β-cell selective manner. The inhibitory actions of nitric oxide are associated with impaired mitochondrial oxidative metabolism and lack of glycolytic compensation that result in a decrease in β-cell ATP. Like nitric oxide, inhibitors of mitochondrial respiration reduce ATP levels and limit ATR signaling in a β-cell selective manner. When non-β-cells are forced to utilize mitochondrial oxidative metabolism for ATP generation, their response is more like β-cells, as nitric oxide and inhibitors of mitochondrial respiration attenuate ATR signaling. These studies support a dual role for nitric oxide in regulating ATR signaling. Nitric oxide activates ATR in all cell types examined by inhibiting ribonucleotide reductase, and in a β-cell selective manner, inducible nitric oxide synthase-derived levels of nitric oxide limit ATR signaling by attenuating mitochondrial oxidative metabolism and depleting ATP.
Collapse
|
9
|
Sidarala V, Pearson GL, Parekh VS, Thompson B, Christen L, Gingerich MA, Zhu J, Stromer T, Ren J, Reck EC, Chai B, Corbett JA, Mandrup-Poulsen T, Satin LS, Soleimanpour SA. Mitophagy protects β cells from inflammatory damage in diabetes. JCI Insight 2020; 5:141138. [PMID: 33232298 PMCID: PMC7819751 DOI: 10.1172/jci.insight.141138] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 11/11/2020] [Indexed: 12/14/2022] Open
Abstract
Inflammatory damage contributes to β cell failure in type 1 and 2 diabetes (T1D and T2D, respectively). Mitochondria are damaged by inflammatory signaling in β cells, resulting in impaired bioenergetics and initiation of proapoptotic machinery. Hence, the identification of protective responses to inflammation could lead to new therapeutic targets. Here, we report that mitophagy serves as a protective response to inflammatory stress in both human and rodent β cells. Utilizing in vivo mitophagy reporters, we observed that diabetogenic proinflammatory cytokines induced mitophagy in response to nitrosative/oxidative mitochondrial damage. Mitophagy-deficient β cells were sensitized to inflammatory stress, leading to the accumulation of fragmented dysfunctional mitochondria, increased β cell death, and hyperglycemia. Overexpression of CLEC16A, a T1D gene and mitophagy regulator whose expression in islets is protective against T1D, ameliorated cytokine-induced human β cell apoptosis. Thus, mitophagy promotes β cell survival and prevents diabetes by countering inflammatory injury. Targeting this pathway has the potential to prevent β cell failure in diabetes and may be beneficial in other inflammatory conditions.
Collapse
Affiliation(s)
- Vaibhav Sidarala
- Division of Metabolism, Endocrinology and Diabetes and Department of Internal Medicine, and
| | - Gemma L Pearson
- Division of Metabolism, Endocrinology and Diabetes and Department of Internal Medicine, and
| | - Vishal S Parekh
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Benjamin Thompson
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Lisa Christen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Morgan A Gingerich
- Division of Metabolism, Endocrinology and Diabetes and Department of Internal Medicine, and.,Program in Biological Sciences, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Jie Zhu
- Division of Metabolism, Endocrinology and Diabetes and Department of Internal Medicine, and
| | - Tracy Stromer
- Division of Metabolism, Endocrinology and Diabetes and Department of Internal Medicine, and
| | - Jianhua Ren
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Emma C Reck
- Division of Metabolism, Endocrinology and Diabetes and Department of Internal Medicine, and
| | - Biaoxin Chai
- Division of Metabolism, Endocrinology and Diabetes and Department of Internal Medicine, and
| | - John A Corbett
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | | | - Leslie S Satin
- Division of Metabolism, Endocrinology and Diabetes and Department of Internal Medicine, and.,Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Scott A Soleimanpour
- Division of Metabolism, Endocrinology and Diabetes and Department of Internal Medicine, and.,VA Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
| |
Collapse
|
10
|
Stafford JD, Shaheen ZR, Yeo CT, Corbett JA. Inhibition of mitochondrial oxidative metabolism attenuates EMCV replication and protects β-cells from virally mediated lysis. J Biol Chem 2020; 295:16655-16664. [PMID: 32972972 PMCID: PMC7864063 DOI: 10.1074/jbc.ra120.014851] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 09/06/2020] [Indexed: 12/15/2022] Open
Abstract
Viral infection is one environmental factor that may contribute to the initiation of pancreatic β-cell destruction during the development of autoimmune diabetes. Picornaviruses, such as encephalomyocarditis virus (EMCV), induce a pro-inflammatory response in islets leading to local production of cytokines, such as IL-1, by resident islet leukocytes. Furthermore, IL-1 is known to stimulate β-cell expression of iNOS and production of the free radical nitric oxide. The purpose of this study was to determine whether nitric oxide contributes to the β-cell response to viral infection. We show that nitric oxide protects β-cells against virally mediated lysis by limiting EMCV replication. This protection requires low micromolar, or iNOS-derived, levels of nitric oxide. At these concentrations nitric oxide inhibits the Krebs enzyme aconitase and complex IV of the electron transport chain. Like nitric oxide, pharmacological inhibition of mitochondrial oxidative metabolism attenuates EMCV-mediated β-cell lysis by inhibiting viral replication. These findings provide novel evidence that cytokine signaling in β-cells functions to limit viral replication and subsequent β-cell lysis by attenuating mitochondrial oxidative metabolism in a nitric oxide-dependent manner.
Collapse
Affiliation(s)
- Joshua D Stafford
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Zachary R Shaheen
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Chay Teng Yeo
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - John A Corbett
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
| |
Collapse
|
11
|
Oleson BJ, Corbett JA. Can insulin secreting pancreatic β-cells provide novel insights into the metabolic regulation of the DNA damage response? Biochem Pharmacol 2020; 176:113907. [PMID: 32171728 DOI: 10.1016/j.bcp.2020.113907] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 03/06/2020] [Indexed: 12/18/2022]
Abstract
Insulin, produced by pancreatic β-cells, is responsible for the control of whole-body glucose metabolism. Insulin is secreted by pancreatic β-cells in a tightly regulated process that is controlled by the serum level of glucose, glucose sensing and glucose oxidative metabolism. The regulation of intermediary metabolism in β-cells is unique as these cells oxidize glucose to CO2 on substrate supply while mitochondrial oxidative metabolism occurs on demand, for the production of intermediates or energy production, in most cell types. This review discusses recent findings that the regulation of intermediary metabolism by nitric oxide attenuates the DNA damage response (DDR) and DNA damage-dependent apoptosis in a β-cell selective manner. Specific focus is placed on the mechanisms by which iNOS derived nitric oxide (low micromolar levels) regulates DDR activation via the inhibition of intermediary metabolism. The physiological significance of the association of metabolism, nitric oxide and DDR signaling for cancer biology and diabetes is discussed.
Collapse
Affiliation(s)
- Bryndon J Oleson
- From the Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - John A Corbett
- From the Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
12
|
Sarkar R, Patra U, Lo M, Mukherjee A, Biswas A, Chawla-Sarkar M. Rotavirus activates a noncanonical ATM-Chk2 branch of DNA damage response during infection to positively regulate viroplasm dynamics. Cell Microbiol 2020; 22:e13149. [PMID: 31845505 DOI: 10.1111/cmi.13149] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 11/20/2019] [Accepted: 11/21/2019] [Indexed: 12/14/2022]
Abstract
Surveillance for maintaining genomic pristineness, a protective safeguard of great onco-preventive significance, has been dedicated in eukaryotic cells to a highly conserved and synchronised signalling cascade called DNA damage response (DDR). Not surprisingly, foreign genetic elements like those of viruses are often potential targets of DDR. Viruses have evolved novel ways to subvert this genome vigilance by twisting canonical DDR to a skewed, noncanonical response through selective hijacking of some DDR components while antagonising the others. Though reported for many DNA and a few RNA viruses, potential implications of DDR have not been addressed yet in case of infection with rotavirus (RV), a double-stranded RNA virus. In the present study, we aimed at the modulation of ataxia telangiectasia mutated (ATM)-checkpoint kinase 2 (Chk2) branch of DDR in response to RV infection in vitro. We found activation of the transducer kinase ATM and its downstream effector Chk2 in RV-SA11-infected cells, the activation response being maximal at 6-hr post infection. Moreover, ATM activation was found to be dependent on induction of the upstream sensor Mre11-Rad50-Nbs1 (MRN) complex. Interestingly, RV-SA11-mediated maximal induction of ATM-Chk2 pathway was revealed to be neither preceded by occurrence of nuclear DNA damage nor transduced to formation of damage-induced canonical nuclear foci. Subsequent investigations affirmed sequestration of MRN components as well as ATM-Chk2 proteins away from nucleus into cytosolic RV replication factories (viroplasms). Chemical intervention targeting ATM and Chk2 significantly inhibited fusion and maturation of viroplasms leading to attenuated viral propagation. Cumulatively, the current study describes RV-mediated activation of a noncanonical ATM-Chk2 branch of DDR skewed in favour of facilitated viroplasm fusion and productive viral perpetuation.
Collapse
Affiliation(s)
- Rakesh Sarkar
- Division of Virology, National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India
| | - Upayan Patra
- Division of Virology, National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India
| | - Mahadeb Lo
- Division of Virology, National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India
| | - Arpita Mukherjee
- Division of Virology, National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India
| | - Asim Biswas
- Division of Virology, National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India
| | - Mamta Chawla-Sarkar
- Division of Virology, National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India
| |
Collapse
|
13
|
Oleson BJ, Broniowska KA, Yeo CT, Flancher M, Naatz A, Hogg N, Tarakanova VL, Corbett JA. The Role of Metabolic Flexibility in the Regulation of the DNA Damage Response by Nitric Oxide. Mol Cell Biol 2019; 39:e00153-19. [PMID: 31235477 PMCID: PMC6712938 DOI: 10.1128/mcb.00153-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 05/07/2019] [Accepted: 06/18/2019] [Indexed: 12/24/2022] Open
Abstract
In this report, we show that nitric oxide suppresses DNA damage response (DDR) signaling in the pancreatic β-cell line INS 832/13 and rat islets by inhibiting intermediary metabolism. Nitric oxide is known to inhibit complex IV of the electron transport chain and aconitase of the Krebs cycle. Non-β cells compensate by increasing glycolytic metabolism to maintain ATP levels; however, β cells lack this metabolic flexibility, resulting in a nitric oxide-dependent decrease in ATP and NAD+ Like nitric oxide, mitochondrial toxins inhibit DDR signaling in β cells by a mechanism that is associated with a decrease in ATP. Non-β cells compensate for the effects of mitochondrial toxins with an adaptive shift to glycolytic ATP generation that allows for DDR signaling. Forcing non-β cells to derive ATP via mitochondrial respiration (replacing glucose with galactose in the medium) and glucose deprivation sensitizes these cells to nitric oxide-mediated inhibition of DDR signaling. These findings indicate that metabolic flexibility is necessary to maintain DDR signaling under conditions in which mitochondrial oxidative metabolism is inhibited and support the inhibition of oxidative metabolism (decreased ATP) as one protective mechanism by which nitric oxide attenuates DDR-dependent β-cell apoptosis.
Collapse
Affiliation(s)
- Bryndon J Oleson
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | | | - Chay Teng Yeo
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Michael Flancher
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Aaron Naatz
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Neil Hogg
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Vera L Tarakanova
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - John A Corbett
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
14
|
Somasundaram V, Basudhar D, Bharadwaj G, No JH, Ridnour LA, Cheng RY, Fujita M, Thomas DD, Anderson SK, McVicar DW, Wink DA. Molecular Mechanisms of Nitric Oxide in Cancer Progression, Signal Transduction, and Metabolism. Antioxid Redox Signal 2019; 30:1124-1143. [PMID: 29634348 PMCID: PMC6354612 DOI: 10.1089/ars.2018.7527] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 03/08/2018] [Indexed: 01/03/2023]
Abstract
SIGNIFICANCE Cancer is a complex disease, which not only involves the tumor but its microenvironment comprising different immune cells as well. Nitric oxide (NO) plays specific roles within tumor cells and the microenvironment and determines the rate of cancer progression, therapy efficacy, and patient prognosis. Recent Advances: Key understanding of the processes leading to dysregulated NO flux within the tumor microenvironment over the past decade has provided better understanding of the dichotomous role of NO in cancer and its importance in shaping the immune landscape. It is becoming increasingly evident that nitric oxide synthase 2 (NOS2)-mediated NO/reactive nitrogen oxide species (RNS) are heavily involved in cancer progression and metastasis in different types of tumor. More recent studies have found that NO from NOS2+ macrophages is required for cancer immunotherapy to be effective. CRITICAL ISSUES NO/RNS, unlike other molecules, are unique in their ability to target a plethora of oncogenic pathways during cancer progression. In this review, we subcategorize the different levels of NO produced by cells and shed light on the context-dependent temporal effects on cancer signaling and metabolic shift in the tumor microenvironment. FUTURE DIRECTIONS Understanding the source of NO and its spaciotemporal profile within the tumor microenvironment could help improve efficacy of cancer immunotherapies by improving tumor infiltration of immune cells for better tumor clearance.
Collapse
Affiliation(s)
- Veena Somasundaram
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland
| | - Debashree Basudhar
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland
| | - Gaurav Bharadwaj
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland
| | - Jae Hong No
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seoul, Republic of Korea
| | - Lisa A. Ridnour
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland
| | - Robert Y.S. Cheng
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland
| | - Mayumi Fujita
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland
- Department of Basic Medical Sciences for Radiation Damages, National Institutes of Quantum and Radiological Science and Technology, Chiba, Japan
| | - Douglas D. Thomas
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois
| | - Stephen K. Anderson
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland
| | - Daniel W. McVicar
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland
| | - David A. Wink
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland
| |
Collapse
|
15
|
Oleson BJ, Corbett JA. Dual Role of Nitric Oxide in Regulating the Response of β Cells to DNA Damage. Antioxid Redox Signal 2018; 29:1432-1445. [PMID: 28978225 PMCID: PMC6166691 DOI: 10.1089/ars.2017.7351] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 09/09/2017] [Indexed: 01/09/2023]
Abstract
SIGNIFICANCE Cytokines released in and around pancreatic islets during islet inflammation are believed to contribute to impaired β cell function and β cell death during the development of diabetes. Nitric oxide, produced by β cells in response to cytokine exposure, controls many of the responses of β cells during islet inflammation. Recent Advances: Although nitric oxide has been shown to inhibit insulin secretion and oxidative metabolism and induce DNA damage in β cells, it also activates protective pathways that promote recovery of insulin secretion and oxidative metabolism and repair of damaged DNA. Recent studies have identified a novel role for nitric oxide in selectively regulating the DNA damage response in β cells. CRITICAL ISSUES Does nitric oxide mediate cytokine-induced β cell damage, or is nitric oxide produced by β cells in response to cytokines to protect β cells from damage? FUTURE DIRECTIONS β cells appear to be the only islet endocrine cell type capable of responding to proinflammatory cytokines with the production of nitric oxide, and these terminally differentiated cells have a limited capacity to regenerate. It is likely that there is a physiological purpose for this response, and understanding this could open new areas of study regarding the loss of functional β cell mass during diabetes development.
Collapse
Affiliation(s)
- Bryndon J. Oleson
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - John A. Corbett
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
16
|
Oleson BJ, Naatz A, Proudfoot SC, Yeo CT, Corbett JA. Role of Protein Phosphatase 1 and Inhibitor of Protein Phosphatase 1 in Nitric Oxide-Dependent Inhibition of the DNA Damage Response in Pancreatic β-Cells. Diabetes 2018; 67:898-910. [PMID: 29444892 PMCID: PMC5909998 DOI: 10.2337/db17-1062] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 02/02/2018] [Indexed: 11/13/2022]
Abstract
Nitric oxide is produced at micromolar levels by pancreatic β-cells during exposure to proinflammatory cytokines. While classically viewed as damaging, nitric oxide also activates pathways that promote β-cell survival. We have shown that nitric oxide, in a cell type-selective manner, inhibits the DNA damage response (DDR) and, in doing so, protects β-cells from DNA damage-induced apoptosis. This study explores potential mechanisms by which nitric oxide inhibits DDR signaling. We show that inhibition of DDR signaling (measured by γH2AX formation and the phosphorylation of KAP1) is selective for nitric oxide, as other forms of reactive oxygen/nitrogen species do not impair DDR signaling. The kinetics and broad range of DDR substrates that are inhibited suggest that protein phosphatase activation may be one mechanism by which nitric oxide attenuates DDR signaling in β-cells. While protein phosphatase 1 (PP1) is a primary regulator of DDR signaling and an inhibitor of PP1 (IPP1) is selectively expressed only in β-cells, disruption of either IPP1 or PP1 does not modify the inhibitory actions of nitric oxide on DDR signaling in β-cells. These findings support a PP1-independent mechanism by which nitric oxide selectively impairs DDR signaling and protects β-cells from DNA damage-induced apoptosis.
Collapse
Affiliation(s)
- Bryndon J Oleson
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI
| | - Aaron Naatz
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI
| | - Sarah C Proudfoot
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI
| | - Chay Teng Yeo
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI
| | - John A Corbett
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
17
|
Gorrepati KDD, Lupse B, Annamalai K, Yuan T, Maedler K, Ardestani A. Loss of Deubiquitinase USP1 Blocks Pancreatic β-Cell Apoptosis by Inhibiting DNA Damage Response. iScience 2018; 1:72-86. [PMID: 30227958 PMCID: PMC6135944 DOI: 10.1016/j.isci.2018.02.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 01/31/2018] [Accepted: 02/01/2018] [Indexed: 01/09/2023] Open
Abstract
Impaired pancreatic β-cell survival contributes to the reduced β-cell mass in diabetes, but underlying regulatory mechanisms and key players in this process remain incompletely understood. Here, we identified the deubiquitinase ubiquitin-specific protease 1 (USP1) as an important player in the regulation of β-cell apoptosis under diabetic conditions. Genetic silencing and pharmacological suppression of USP1 blocked β-cell death in several experimental models of diabetes in vitro and ex vivo without compromising insulin content and secretion and without impairing β-cell maturation/identity genes in human islets. Our further analyses showed that USP1 inhibition attenuated DNA damage response (DDR) signals, which were highly elevated in diabetic β-cells, suggesting a USP1-dependent regulation of DDR in stressed β-cells. Our findings highlight a novel function of USP1 in the control of β-cell survival, and its inhibition may have a potential therapeutic relevance for the suppression of β-cell death in diabetes. Genetic and chemical inhibition of USP1 promoted β-cell survival USP1 inhibitors blocked β-cell death in human islets without affecting β-cell function USP1 inhibition reduced DDR signals in stressed β-cells
Collapse
Affiliation(s)
- Kanaka Durga Devi Gorrepati
- Islet Biology Laboratory, University of Bremen, Centre for Biomolecular Interactions Bremen, Leobener Straße NW2, Room B2080, 28359 Bremen, Germany
| | - Blaz Lupse
- Islet Biology Laboratory, University of Bremen, Centre for Biomolecular Interactions Bremen, Leobener Straße NW2, Room B2080, 28359 Bremen, Germany
| | - Karthika Annamalai
- Islet Biology Laboratory, University of Bremen, Centre for Biomolecular Interactions Bremen, Leobener Straße NW2, Room B2080, 28359 Bremen, Germany
| | - Ting Yuan
- Islet Biology Laboratory, University of Bremen, Centre for Biomolecular Interactions Bremen, Leobener Straße NW2, Room B2080, 28359 Bremen, Germany
| | - Kathrin Maedler
- Islet Biology Laboratory, University of Bremen, Centre for Biomolecular Interactions Bremen, Leobener Straße NW2, Room B2080, 28359 Bremen, Germany.
| | - Amin Ardestani
- Islet Biology Laboratory, University of Bremen, Centre for Biomolecular Interactions Bremen, Leobener Straße NW2, Room B2080, 28359 Bremen, Germany.
| |
Collapse
|
18
|
Sidarala V, Kowluru A. Exposure to chronic hyperglycemic conditions results in Ras-related C3 botulinum toxin substrate 1 (Rac1)-mediated activation of p53 and ATM kinase in pancreatic β-cells. Apoptosis 2018; 22:597-607. [PMID: 28220272 DOI: 10.1007/s10495-017-1354-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Chronic hyperglycemia (HG) promotes pancreatic islet dysfunction which leads to the onset of T2DM. This study is aimed at defining regulatory roles of Rac1, a small G-protein, in the activation of p53 and ATM kinase in pancreatic β-cells, under the duress of HG conditions. We report significant stimulatory effects of HG (20 mM; 24 h) on p53 activation in INS-1 832/13 cells, normal rodent and human islets. Pharmacological inhibition of Rac1 (EHT1864 or NSC23766) significantly suppressed HG-induced p53 activation in INS-1 832/13 cells and rat islets, suggesting novel roles for this small G-protein in the activation of p53. Inhibition of Rac1 geranylgeranylation with simvastatin or GGTI-2147, significantly attenuated HG-induced p53 activation, suggesting requisite roles for this signaling step in HG-mediated effects on β-cells. HG-induced p53 activation was also suppressed by SB203580, a known inhibitor of p38MAPK. Additionally, we observed increased activation of ATM kinase under HG conditions, which was blocked in presence of EHT1864. Furthermore, pharmacological inhibition of ATM kinase (KU55933) reduced activation of ATM kinase, but not p53, suggesting that HG-mediated activation of p53 and ATM could represent independent pro-apoptotic events. In conclusion, these data indicate that sustained activation of Rac1-p38MAPK signaling axis leads to activation of p53 leading to β-cell dysfunction under the duress of chronic hyperglycemic conditions.
Collapse
Affiliation(s)
- Vaibhav Sidarala
- Beta-Cell Biochemistry Laboratory, John D. Dingell VA Medical Center, B-4237 Research Service, 4646 John R, Detroit, MI, 48201, USA.,Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI, 48201, USA.,Division of Metabolism, Endocrinology and Diabetes (MEND), Department of Internal Medicine, Brehm Diabetes Research Center, University of Michigan Medical School, Ann Arbor, MI, 48105-1912, USA
| | - Anjaneyulu Kowluru
- Beta-Cell Biochemistry Laboratory, John D. Dingell VA Medical Center, B-4237 Research Service, 4646 John R, Detroit, MI, 48201, USA. .,Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI, 48201, USA.
| |
Collapse
|
19
|
Xia L, Qin K, Wang XR, Wang XL, Zhou AW, Chen GQ, Lu Y. Pyruvate kinase M2 phosphorylates H2AX and promotes genomic instability in human tumor cells. Oncotarget 2017; 8:109120-109134. [PMID: 29312595 PMCID: PMC5752508 DOI: 10.18632/oncotarget.22621] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 10/28/2017] [Indexed: 01/02/2023] Open
Abstract
Pyruvate kinase (PK) catalyzes the conversion of phosphoenolpyruvate and ADP to pyruvate and ATP, a rate-limiting reaction in glycolysis. M2 isoform of PK (PKM2) is the predominant form of PK expressed in tumors. In addition to its well established cytosolic functions as a glycolytic enzyme, PKM2 displays nuclear localization and important nonmetabolic functions in tumorigenesis. Herein, we report that nuclear PKM2 interacts with histone H2AX under DNA damage conditions. Depletion of PKM2 decreased the level of serine 139-phosphorylated H2AX (γ-H2AX) in response to DNA damage. The in vitro kinase assay reveals that PKM2 directly phosphorylates H2AX at serine 139, which is abolished by the deletion of FBP-binding pocket of PKM2 (PKM2-Del515-520). Replacement of wild type PKM2 with the kinase dead mutant PKM2-Del515-520 leads to decreased cell proliferation and chromosomal aberrations under DNA damage conditions. Together, we propose that PKM2 promotes genomic instability in tumor cells which involves direct phosphorylation of H2AX. These findings reveal PKM2 as a novel modulator for genomic instability in tumor cells.
Collapse
Affiliation(s)
- Li Xia
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China
| | - Kang Qin
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China
| | - Xin-Ran Wang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China
| | - Xiao-Ling Wang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China
| | - Ai-Wu Zhou
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China
| | - Guo-Qiang Chen
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China
| | - Ying Lu
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China
| |
Collapse
|
20
|
Mujoo K, Pandita RK, Tiwari A, Charaka V, Chakraborty S, Singh DK, Hambarde S, Hittelman WN, Horikoshi N, Hunt CR, Khanna KK, Kots AY, Butler EB, Murad F, Pandita TK. Differentiation of Human Induced Pluripotent or Embryonic Stem Cells Decreases the DNA Damage Repair by Homologous Recombination. Stem Cell Reports 2017; 9:1660-1674. [PMID: 29103969 PMCID: PMC5831054 DOI: 10.1016/j.stemcr.2017.10.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 10/04/2017] [Accepted: 10/05/2017] [Indexed: 12/16/2022] Open
Abstract
The nitric oxide (NO)-cyclic GMP pathway contributes to human stem cell differentiation, but NO free radical production can also damage DNA, necessitating a robust DNA damage response (DDR) to ensure cell survival. How the DDR is affected by differentiation is unclear. Differentiation of stem cells, either inducible pluripotent or embryonic derived, increased residual DNA damage as determined by γ-H2AX and 53BP1 foci, with increased S-phase-specific chromosomal aberration after exposure to DNA-damaging agents, suggesting reduced homologous recombination (HR) repair as supported by the observation of decreased HR-related repair factor foci formation (RAD51 and BRCA1). Differentiated cells also had relatively increased fork stalling and R-loop formation after DNA replication stress. Treatment with NO donor (NOC-18), which causes stem cell differentiation has no effect on double-strand break (DSB) repair by non-homologous end-joining but reduced DSB repair by HR. Present studies suggest that DNA repair by HR is impaired in differentiated cells. Spontaneous and S-phase-specific chromosome aberrations in differentiated cells Higher frequency of residual γ-H2AX foci after exposure to DNA-damaging agents Higher frequency of cells with 53BP1 and RIF1 co-localization in differentiated cells Higher frequency of cells with a reduced number of RAD51 or BRCA1 foci
Collapse
Affiliation(s)
- Kalpana Mujoo
- Department of Radiation Oncology, Weill Cornell Medical College, The Houston Methodist Hospital Research Institute, Houston, TX 77030, USA; Institute of Molecular Medicine, University of Texas Health at Houston, Houston, TX 77030, USA.
| | - Raj K Pandita
- Department of Radiation Oncology, Weill Cornell Medical College, The Houston Methodist Hospital Research Institute, Houston, TX 77030, USA
| | - Anjana Tiwari
- Department of Radiation Oncology, Weill Cornell Medical College, The Houston Methodist Hospital Research Institute, Houston, TX 77030, USA
| | - Vijay Charaka
- Department of Radiation Oncology, Weill Cornell Medical College, The Houston Methodist Hospital Research Institute, Houston, TX 77030, USA
| | - Sharmistha Chakraborty
- Department of Radiation Oncology, Weill Cornell Medical College, The Houston Methodist Hospital Research Institute, Houston, TX 77030, USA
| | - Dharmendra Kumar Singh
- Department of Radiation Oncology, Weill Cornell Medical College, The Houston Methodist Hospital Research Institute, Houston, TX 77030, USA
| | - Shashank Hambarde
- Department of Radiation Oncology, Weill Cornell Medical College, The Houston Methodist Hospital Research Institute, Houston, TX 77030, USA
| | - Walter N Hittelman
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Nobuo Horikoshi
- Department of Radiation Oncology, Weill Cornell Medical College, The Houston Methodist Hospital Research Institute, Houston, TX 77030, USA
| | - Clayton R Hunt
- Department of Radiation Oncology, Weill Cornell Medical College, The Houston Methodist Hospital Research Institute, Houston, TX 77030, USA
| | - Kum Kum Khanna
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| | | | - E Brian Butler
- Department of Radiation Oncology, Weill Cornell Medical College, The Houston Methodist Hospital Research Institute, Houston, TX 77030, USA
| | - Ferid Murad
- The George Washington University, Washington, DC 20037, USA
| | - Tej K Pandita
- Department of Radiation Oncology, Weill Cornell Medical College, The Houston Methodist Hospital Research Institute, Houston, TX 77030, USA.
| |
Collapse
|
21
|
Kulkarni NN, Adase CA, Zhang LJ, Borkowski AW, Li F, Sanford JA, Coleman DJ, Aguilera C, Indra AK, Gallo RL. IL-1 Receptor-Knockout Mice Develop Epidermal Cysts and Show an Altered Innate Immune Response after Exposure to UVB Radiation. J Invest Dermatol 2017; 137:2417-2426. [PMID: 28754339 DOI: 10.1016/j.jid.2017.07.814] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 07/05/2017] [Accepted: 07/06/2017] [Indexed: 12/31/2022]
Abstract
In this study, we observed that mice lacking the IL-1 receptor (IL-1R) (IL1r-/-) or deficient in IL1-β developed multiple epidermal cysts after chronic UVB exposure. Cysts that developed in IL1r-/- mice were characterized by the presence of the hair follicle marker Sox 9, keratins 10 and 14, and normal melanocyte distribution and retinoid X receptor-α expression. The increased incidence of cysts in IL1r-/- mice was associated with less skin inflammation as characterized by decreased recruitment of macrophages, and their skin also maintained epidermal barrier function compared with wild-type mice. Transcriptional analysis of the skin of IL1r-/- mice after UVB exposure showed decreased gene expression of proinflammatory cytokines such as tumor necrosis factor-α and IL-6. In vitro, primary keratinocytes derived from IL1r-/- mice were more resistant to UVB-triggered cell death compared with wild-type cells, and tumor necrosis factor-α release was completely blocked in the absence of IL-1R. These observations illustrate an unexpected yet prominent phenotype associated with the lack of IL-1R signaling in mice and support further investigation into the role of IL-1 ligands in epidermal repair and innate immune response after damaging UVB exposure.
Collapse
Affiliation(s)
- Nikhil N Kulkarni
- Department of Dermatology, University of California, San Diego, California, USA
| | - Christopher A Adase
- Department of Dermatology, University of California, San Diego, California, USA
| | - Ling-Juan Zhang
- Department of Dermatology, University of California, San Diego, California, USA
| | - Andrew W Borkowski
- Department of Dermatology, University of California, San Diego, California, USA
| | - Fengwu Li
- Department of Dermatology, University of California, San Diego, California, USA
| | - James A Sanford
- Department of Dermatology, University of California, San Diego, California, USA
| | - Daniel J Coleman
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon, USA; Molecular and Cellular Biology Program, Oregon State University, Corvallis, Oregon, USA
| | - Carlos Aguilera
- Department of Dermatology, University of California, San Diego, California, USA
| | - Arup K Indra
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon, USA; Molecular and Cellular Biology Program, Oregon State University, Corvallis, Oregon, USA; Department of Biochemistry and Biophysics, Oregon State University, Corvallis, Oregon, USA; Linus Pauling Science Center, Oregon State University, Corvallis, Oregon, USA; Department of Dermatology, Oregon Health and Science University, Portland, Oregon, USA; Knight Cancer Institute, Portland, Oregon, USA
| | - Richard L Gallo
- Department of Dermatology, University of California, San Diego, California, USA.
| |
Collapse
|
22
|
ATM-ROS-iNOS axis regulates nitric oxide mediated cellular senescence. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:177-190. [DOI: 10.1016/j.bbamcr.2016.11.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Revised: 10/20/2016] [Accepted: 11/10/2016] [Indexed: 12/18/2022]
|
23
|
Oleson BJ, Broniowska KA, Naatz A, Hogg N, Tarakanova VL, Corbett JA. Nitric Oxide Suppresses β-Cell Apoptosis by Inhibiting the DNA Damage Response. Mol Cell Biol 2016; 36:2067-77. [PMID: 27185882 PMCID: PMC4946431 DOI: 10.1128/mcb.00262-16] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 05/12/2016] [Indexed: 12/15/2022] Open
Abstract
Nitric oxide, produced in pancreatic β cells in response to proinflammatory cytokines, plays a dual role in the regulation of β-cell fate. While nitric oxide induces cellular damage and impairs β-cell function, it also promotes β-cell survival through activation of protective pathways that promote β-cell recovery. In this study, we identify a novel mechanism in which nitric oxide prevents β-cell apoptosis by attenuating the DNA damage response (DDR). Nitric oxide suppresses activation of the DDR (as measured by γH2AX formation and the phosphorylation of KAP1 and p53) in response to multiple genotoxic agents, including camptothecin, H2O2, and nitric oxide itself, despite the presence of DNA damage. While camptothecin and H2O2 both induce DDR activation, nitric oxide suppresses only camptothecin-induced apoptosis and not H2O2-induced necrosis. The ability of nitric oxide to suppress the DDR appears to be selective for pancreatic β cells, as nitric oxide fails to inhibit DDR signaling in macrophages, hepatocytes, and fibroblasts, three additional cell types examined. While originally described as the damaging agent responsible for cytokine-induced β-cell death, these studies identify a novel role for nitric oxide as a protective molecule that promotes β-cell survival by suppressing DDR signaling and attenuating DNA damage-induced apoptosis.
Collapse
Affiliation(s)
- Bryndon J Oleson
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | | | - Aaron Naatz
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Neil Hogg
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Vera L Tarakanova
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - John A Corbett
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
24
|
STAT3 modulates β-cell cycling in injured mouse pancreas and protects against DNA damage. Cell Death Dis 2016; 7:e2272. [PMID: 27336716 PMCID: PMC5143397 DOI: 10.1038/cddis.2016.171] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 05/02/2016] [Accepted: 05/03/2016] [Indexed: 12/22/2022]
Abstract
Partial pancreatic duct ligation (PDL) of mouse pancreas induces a doubling of the β-cell mass mainly through proliferation of pre-existing and newly formed β-cells. The molecular mechanism governing this process is still largely unknown. Given the inflammatory nature of PDL and inflammation-induced signaling via the signal transducer and activator of transcription 3 (STAT3), the activation and the role of STAT3 in PDL-induced β-cell proliferation were investigated. Duct ligation stimulates the expression of several cytokines that can act as ligands inducing STAT3 signaling and phosphorylation in β-cells. β-Cell cycling increased by conditional β-cell-specific Stat3 knockout and decreased by STAT3 activation through administration of interleukin-6. In addition, the level of DNA damage in β-cells of PDL pancreas increased after deletion of Stat3. These data indicate a role for STAT3 in maintaining a steady state in the β-cell, by modulating its cell cycle and protection from DNA damage.
Collapse
|
25
|
Mellado-Gil JM, Jiménez-Moreno CM, Martin-Montalvo A, Alvarez-Mercado AI, Fuente-Martin E, Cobo-Vuilleumier N, Lorenzo PI, Bru-Tari E, Herrera-Gómez IDG, López-Noriega L, Pérez-Florido J, Santoyo-López J, Spyrantis A, Meda P, Boehm BO, Quesada I, Gauthier BR. PAX4 preserves endoplasmic reticulum integrity preventing beta cell degeneration in a mouse model of type 1 diabetes mellitus. Diabetologia 2016; 59:755-65. [PMID: 26813254 PMCID: PMC4779135 DOI: 10.1007/s00125-016-3864-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 12/17/2015] [Indexed: 01/06/2023]
Abstract
AIMS/HYPOTHESIS A strategy to enhance pancreatic islet functional beta cell mass (BCM) while restraining inflammation, through the manipulation of molecular and cellular targets, would provide a means to counteract the deteriorating glycaemic control associated with diabetes mellitus. The aims of the current study were to investigate the therapeutic potential of such a target, the islet-enriched and diabetes-linked transcription factor paired box 4 (PAX4), to restrain experimental autoimmune diabetes (EAD) in the RIP-B7.1 mouse model background and to characterise putative cellular mechanisms associated with preserved BCM. METHODS Two groups of RIP-B7.1 mice were genetically engineered to: (1) conditionally express either PAX4 (BPTL) or its diabetes-linked mutant variant R129W (mutBPTL) using doxycycline (DOX); and (2) constitutively express luciferase in beta cells through the use of RIP. Mice were treated or not with DOX, and EAD was induced by immunisation with a murine preproinsulin II cDNA expression plasmid. The development of hyperglycaemia was monitored for up to 4 weeks following immunisation and alterations in the BCM were assessed weekly by non-invasive in vivo bioluminescence intensity (BLI). In parallel, BCM, islet cell proliferation and apoptosis were evaluated by immunocytochemistry. Alterations in PAX4- and PAX4R129W-mediated islet gene expression were investigated by microarray profiling. PAX4 preservation of endoplasmic reticulum (ER) homeostasis was assessed using thapsigargin, electron microscopy and intracellular calcium measurements. RESULTS PAX4 overexpression blunted EAD, whereas the diabetes-linked mutant variant PAX4R129W did not convey protection. PAX4-expressing islets exhibited reduced insulitis and decreased beta cell apoptosis, correlating with diminished DNA damage and increased islet cell proliferation. Microarray profiling revealed that PAX4 but not PAX4R129W targeted expression of genes implicated in cell cycle and ER homeostasis. Consistent with the latter, islets overexpressing PAX4 were protected against thapsigargin-mediated ER-stress-related apoptosis. Luminal swelling associated with ER stress induced by thapsigargin was rescued in PAX4-overexpressing beta cells, correlating with preserved cytosolic calcium oscillations in response to glucose. In contrast, RNA interference mediated repression of PAX4-sensitised MIN6 cells to thapsigargin cell death. CONCLUSIONS/INTERPRETATION The coordinated regulation of distinct cellular pathways particularly related to ER homeostasis by PAX4 not achieved by the mutant variant PAX4R129W alleviates beta cell degeneration and protects against diabetes mellitus. The raw data for the RNA microarray described herein are accessible in the Gene Expression Omnibus database under accession number GSE62846.
Collapse
Affiliation(s)
- José Manuel Mellado-Gil
- Pancreatic Islet Development and Regeneration Unit, Department of Stem Cells, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Avda Américo Vespucio, Parque Científico y Tecnológico Cartuja 93, 41092, Seville, Spain
| | - Carmen María Jiménez-Moreno
- Pancreatic Islet Development and Regeneration Unit, Department of Stem Cells, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Avda Américo Vespucio, Parque Científico y Tecnológico Cartuja 93, 41092, Seville, Spain
| | - Alejandro Martin-Montalvo
- Pancreatic Islet Development and Regeneration Unit, Department of Stem Cells, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Avda Américo Vespucio, Parque Científico y Tecnológico Cartuja 93, 41092, Seville, Spain
| | - Ana Isabel Alvarez-Mercado
- Pancreatic Islet Development and Regeneration Unit, Department of Stem Cells, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Avda Américo Vespucio, Parque Científico y Tecnológico Cartuja 93, 41092, Seville, Spain
| | - Esther Fuente-Martin
- Pancreatic Islet Development and Regeneration Unit, Department of Stem Cells, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Avda Américo Vespucio, Parque Científico y Tecnológico Cartuja 93, 41092, Seville, Spain
| | - Nadia Cobo-Vuilleumier
- Pancreatic Islet Development and Regeneration Unit, Department of Stem Cells, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Avda Américo Vespucio, Parque Científico y Tecnológico Cartuja 93, 41092, Seville, Spain
| | - Petra Isabel Lorenzo
- Pancreatic Islet Development and Regeneration Unit, Department of Stem Cells, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Avda Américo Vespucio, Parque Científico y Tecnológico Cartuja 93, 41092, Seville, Spain
| | - Eva Bru-Tari
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain
- Instituto de Bioingeniería, Universidad Miguel Hernandez, Elche, Spain
| | - Irene de Gracia Herrera-Gómez
- Pancreatic Islet Development and Regeneration Unit, Department of Stem Cells, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Avda Américo Vespucio, Parque Científico y Tecnológico Cartuja 93, 41092, Seville, Spain
| | - Livia López-Noriega
- Pancreatic Islet Development and Regeneration Unit, Department of Stem Cells, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Avda Américo Vespucio, Parque Científico y Tecnológico Cartuja 93, 41092, Seville, Spain
| | - Javier Pérez-Florido
- Medical Genome Project, Genomics & Bioinformatics Platform of Andalusia, Seville, Spain
| | - Javier Santoyo-López
- Medical Genome Project, Genomics & Bioinformatics Platform of Andalusia, Seville, Spain
- Edinburgh Genomics, University of Edinburgh, Edinburgh, UK
| | - Andreas Spyrantis
- Department of Internal Medicine, Ulm University Medical Centre, Ulm, Germany
| | - Paolo Meda
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Bernhard O Boehm
- Department of Internal Medicine, Ulm University Medical Centre, Ulm, Germany
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Republic of Singapore
- Imperial College, London, UK
| | - Ivan Quesada
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain
- Instituto de Bioingeniería, Universidad Miguel Hernandez, Elche, Spain
| | - Benoit R Gauthier
- Pancreatic Islet Development and Regeneration Unit, Department of Stem Cells, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Avda Américo Vespucio, Parque Científico y Tecnológico Cartuja 93, 41092, Seville, Spain.
| |
Collapse
|
26
|
Tong X, Kono T, Evans-Molina C. Nitric oxide stress and activation of AMP-activated protein kinase impair β-cell sarcoendoplasmic reticulum calcium ATPase 2b activity and protein stability. Cell Death Dis 2015; 6:e1790. [PMID: 26086963 PMCID: PMC4669835 DOI: 10.1038/cddis.2015.154] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 04/13/2015] [Accepted: 04/20/2015] [Indexed: 11/15/2022]
Abstract
The sarcoendoplasmic reticulum Ca2+ ATPase 2b (SERCA2b) pump maintains a steep Ca2+ concentration gradient between the cytosol and ER lumen in the pancreatic β-cell, and the integrity of this gradient has a central role in regulated insulin production and secretion, maintenance of ER function and β-cell survival. We have previously demonstrated loss of β-cell SERCA2b expression under diabetic conditions. To define the mechanisms underlying this, INS-1 cells and rat islets were treated with the proinflammatory cytokine interleukin-1β (IL-1β) combined with or without cycloheximide or actinomycin D. IL-1β treatment led to increased inducible nitric oxide synthase (iNOS) gene and protein expression, which occurred concurrently with the activation of AMP-activated protein kinase (AMPK). IL-1β led to decreased SERCA2b mRNA and protein expression, whereas time-course experiments revealed a reduction in protein half-life with no change in mRNA stability. Moreover, SERCA2b protein but not mRNA levels were rescued by treatment with the NOS inhibitor l-NMMA (NG-monomethyl l-arginine), whereas the NO donor SNAP (S-nitroso-N-acetyl-d,l-penicillamine) and the AMPK activator AICAR (5-aminoimidazole-4-carboxamide ribonucleotide) recapitulated the effects of IL-1β on SERCA2b protein stability. Similarly, IL-1β-induced reductions in SERCA2b expression were rescued by pharmacological inhibition of AMPK with compound C or by transduction of a dominant-negative form of AMPK, whereas β-cell death was prevented in parallel. Finally, to determine a functional relationship between NO and AMPK signaling and SERCA2b activity, fura-2/AM (fura-2-acetoxymethylester) Ca2+ imaging experiments were performed in INS-1 cells. Consistent with observed changes in SERCA2b expression, IL-1β, SNAP and AICAR increased cytosolic Ca2+ and decreased ER Ca2+ levels, suggesting congruent modulation of SERCA activity under these conditions. In aggregate, these results show that SERCA2b protein stability is decreased under inflammatory conditions through NO- and AMPK-dependent pathways and provide novel insight into pathways leading to altered β-cell calcium homeostasis and reduced β-cell survival in diabetes.
Collapse
Affiliation(s)
- X Tong
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - T Kono
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - C Evans-Molina
- 1] Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA [2] Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA [3] Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA [4] The Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA [5] The Roudebush VA Medical Center, Indianapolis, IN 46202, USA
| |
Collapse
|
27
|
Kim EJ, Kim J, Lee MY, Sudhanva MS, Devakumar S, Jeon YJ. Silymarin Inhibits Cytokine-Stimulated Pancreatic Beta Cells by Blocking the ERK1/2 Pathway. Biomol Ther (Seoul) 2014; 22:282-7. [PMID: 25143805 PMCID: PMC4131525 DOI: 10.4062/biomolther.2014.072] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 06/19/2014] [Accepted: 06/24/2014] [Indexed: 12/11/2022] Open
Abstract
We show that silymarin, a polyphenolic flavonoid isolated from milk thistle (Silybum marianum), inhibits cytokine mixture (CM: TNF-α, IFN-γ, and IL-1β)-induced production of nitric oxide (NO) in the pancreatic beta cell line MIN6N8a. Immunostaining and Western blot analysis showed that silymarin inhibits iNOS gene expression. RT-PCR showed that silymarin inhibits iNOS gene expression in a dose-dependent manner. We also showed that silymarin inhibits extracellular signal-regulated protein kinase-1 and 2 (ERK1/2) phosphorylation. A MEK1 inhibitor abrogated CM-induced nitrite production, similar to silymarin. Treatment of MIN6N8a cells with silymarin also inhibited CM-stimulated activation of NF-κB, which is important for iNOS transcription. Collectively, we demonstrate that silymarin inhibits NO production in pancreatic beta cells, and silymarin may represent a useful anti-diabetic agent.
Collapse
Affiliation(s)
- Eun Jeong Kim
- Department of Pharmacology, School of Medicine, Chosun University, Gwangju 501-759, Republic of Korea
| | - Jeeho Kim
- Department of Pharmacology, School of Medicine, Chosun University, Gwangju 501-759, Republic of Korea
| | - Min Young Lee
- Department of Pharmacology, School of Medicine, Chosun University, Gwangju 501-759, Republic of Korea
| | | | | | - Young Jin Jeon
- Department of Pharmacology, School of Medicine, Chosun University, Gwangju 501-759, Republic of Korea
| |
Collapse
|