1
|
Wang Z, Yu ZW, Zhang Y, Wang WH, Wu XY, Liu SZ, Bin YL, Cai BP, Huang SY, Fang MJ, Qi R, Li MY, Qiu YK. Hinokione: an abietene diterpene with pancreatic β cells regeneration and hypoglycemic activity, and other derivatives with novel structures from the woods of Agathis dammara. J Nat Med 2024; 78:849-862. [PMID: 38724866 DOI: 10.1007/s11418-024-01816-z] [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: 01/08/2024] [Accepted: 04/16/2024] [Indexed: 08/31/2024]
Abstract
In this study, 14 abietene and pimarene diterpenoids were isolated from the woods of Agathis dammara. Among them, 4 new compounds, dammarone A-C and dammaric acid A (1-4), were firstly reported, respectively. The structure of the new compounds was determined by HR ESI-MS and 1D/2D NMR spectroscopy, and their absolute configuration was determined by electronic circular dichroism (ECD) exciton chirality method. The hypoglycemic effect of all compounds was evaluated by transgenic zebrafish model, and the structure-activity relationship was discussed. Hinokione (7, HO) has low toxicity and significant hypoglycemic effects on zebrafish, the mechanism is mainly by promoting the differentiation of zebrafish pancreatic endocrine precursor cells (PEP cells) into β cells, thereby promoting the regeneration of pancreatic β cells.
Collapse
Affiliation(s)
- Zheng Wang
- Zhongshan Hospital Affiliated to Xiamen University, Xiamen, 361001, China
| | - Zhe-Wei Yu
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
- School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Yi Zhang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Wen-Hui Wang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Xin-Yi Wu
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Shun-Zhi Liu
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Yan-Lin Bin
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Bang-Ping Cai
- Xiamen Botanical Garden, Xiamen, 361003, Fujian, China
| | - Shi-Yan Huang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Mei-Juan Fang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Rong Qi
- School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Ming-Yu Li
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China.
| | - Ying-Kun Qiu
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China.
| |
Collapse
|
2
|
Diane A, Allouch A, Mu-U-Min RBA, Al-Siddiqi HH. Endoplasmic reticulum stress in pancreatic β-cell dysfunctionality and diabetes mellitus: a promising target for generation of functional hPSC-derived β-cells in vitro. Front Endocrinol (Lausanne) 2024; 15:1386471. [PMID: 38966213 PMCID: PMC11222326 DOI: 10.3389/fendo.2024.1386471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 06/10/2024] [Indexed: 07/06/2024] Open
Abstract
Diabetes mellitus (DM), is a chronic disorder characterized by impaired glucose homeostasis that results from the loss or dysfunction of pancreatic β-cells leading to type 1 diabetes (T1DM) and type 2 diabetes (T2DM), respectively. Pancreatic β-cells rely to a great degree on their endoplasmic reticulum (ER) to overcome the increased secretary need for insulin biosynthesis and secretion in response to nutrient demand to maintain glucose homeostasis in the body. As a result, β-cells are potentially under ER stress following nutrient levels rise in the circulation for a proper pro-insulin folding mediated by the unfolded protein response (UPR), underscoring the importance of this process to maintain ER homeostasis for normal β-cell function. However, excessive or prolonged increased influx of nascent proinsulin into the ER lumen can exceed the ER capacity leading to pancreatic β-cells ER stress and subsequently to β-cell dysfunction. In mammalian cells, such as β-cells, the ER stress response is primarily regulated by three canonical ER-resident transmembrane proteins: ATF6, IRE1, and PERK/PEK. Each of these proteins generates a transcription factor (ATF4, XBP1s, and ATF6, respectively), which in turn activates the transcription of ER stress-inducible genes. An increasing number of evidence suggests that unresolved or dysregulated ER stress signaling pathways play a pivotal role in β-cell failure leading to insulin secretion defect and diabetes. In this article we first highlight and summarize recent insights on the role of ER stress and its associated signaling mechanisms on β-cell function and diabetes and second how the ER stress pathways could be targeted in vitro during direct differentiation protocols for generation of hPSC-derived pancreatic β-cells to faithfully phenocopy all features of bona fide human β-cells for diabetes therapy or drug screening.
Collapse
Affiliation(s)
- Abdoulaye Diane
- Diabetes Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | | | | | | |
Collapse
|
3
|
Lariviere D, Craig SJC, Paul IM, Hohman EE, Savage JS, Wright RO, Chiaromonte F, Makova KD, Reimherr ML. Methylation profiles at birth linked to early childhood obesity. J Dev Orig Health Dis 2024; 15:e7. [PMID: 38660759 PMCID: PMC11268442 DOI: 10.1017/s2040174424000060] [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: 04/26/2024]
Abstract
Childhood obesity represents a significant global health concern and identifying its risk factors is crucial for developing intervention programs. Many "omics" factors associated with the risk of developing obesity have been identified, including genomic, microbiomic, and epigenomic factors. Here, using a sample of 48 infants, we investigated how the methylation profiles in cord blood and placenta at birth were associated with weight outcomes (specifically, conditional weight gain, body mass index, and weight-for-length ratio) at age six months. We characterized genome-wide DNA methylation profiles using the Illumina Infinium MethylationEpic chip, and incorporated information on child and maternal health, and various environmental factors into the analysis. We used regression analysis to identify genes with methylation profiles most predictive of infant weight outcomes, finding a total of 23 relevant genes in cord blood and 10 in placenta. Notably, in cord blood, the methylation profiles of three genes (PLIN4, UBE2F, and PPP1R16B) were associated with all three weight outcomes, which are also associated with weight outcomes in an independent cohort suggesting a strong relationship with weight trajectories in the first six months after birth. Additionally, we developed a Methylation Risk Score (MRS) that could be used to identify children most at risk for developing childhood obesity. While many of the genes identified by our analysis have been associated with weight-related traits (e.g., glucose metabolism, BMI, or hip-to-waist ratio) in previous genome-wide association and variant studies, our analysis implicated several others, whose involvement in the obesity phenotype should be evaluated in future functional investigations.
Collapse
Affiliation(s)
- Delphine Lariviere
- Department of Biochemistry and Molecular Biology, Penn State University, University Park, PA, USA
| | - Sarah J C Craig
- Department of Biology, Penn State University, University Park, PA, USA
- Center for Medical Genomics, Penn State University, University Park, PA, USA
| | - Ian M Paul
- Center for Medical Genomics, Penn State University, University Park, PA, USA
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Emily E Hohman
- Center for Childhood Obesity Research, Penn State University, University Park, PA, USA
| | - Jennifer S Savage
- Center for Childhood Obesity Research, Penn State University, University Park, PA, USA
- Nutrition Department, Penn State University, University Park, PA, USA
| | | | - Francesca Chiaromonte
- Center for Medical Genomics, Penn State University, University Park, PA, USA
- Department of Statistics, Penn State University, University Park, PA, USA
- L'EMbeDS, Sant'Anna School of Advanced Studies, Piazza Martiri della Libertà, Pisa, Italy
| | - Kateryna D Makova
- Department of Biology, Penn State University, University Park, PA, USA
- Center for Medical Genomics, Penn State University, University Park, PA, USA
| | - Matthew L Reimherr
- Center for Medical Genomics, Penn State University, University Park, PA, USA
- Department of Statistics, Penn State University, University Park, PA, USA
| |
Collapse
|
4
|
Lariviere D, Craig SJC, Paul IM, Hohman EE, Savage JS, Wright RO, Chiaromonte F, Makova KD, Reimherr ML. Methylation profiles at birth linked to early childhood obesity. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.12.24301172. [PMID: 38260407 PMCID: PMC10802761 DOI: 10.1101/2024.01.12.24301172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Childhood obesity represents a significant global health concern and identifying risk factors is crucial for developing intervention programs. Many 'omics' factors associated with the risk of developing obesity have been identified, including genomic, microbiomic, and epigenomic factors. Here, using a sample of 48 infants, we investigated how the methylation profiles in cord blood and placenta at birth were associated with weight outcomes (specifically, conditional weight gain, body mass index, and weight-for-length ratio) at age six months. We characterized genome-wide DNA methylation profiles using the Illumina Infinium MethylationEpic chip, and incorporated information on child and maternal health, and various environmental factors into the analysis. We used regression analysis to identify genes with methylation profiles most predictive of infant weight outcomes, finding a total of 23 relevant genes in cord blood and 10 in placenta. Notably, in cord blood, the methylation profiles of three genes (PLIN4, UBE2F, and PPP1R16B) were associated with all three weight outcomes, which are also associated with weight outcomes in an independent cohort suggesting a strong relationship with weight trajectories in the first six months after birth. Additionally, we developed a Methylation Risk Score (MRS) that could be used to identify children most at risk for developing childhood obesity. While many of the genes identified by our analysis have been associated with weight-related traits (e.g., glucose metabolism, BMI, or hip-to-waist ratio) in previous genome-wide association and variant studies, our analysis implicated several others, whose involvement in the obesity phenotype should be evaluated in future functional investigations.
Collapse
Affiliation(s)
- Delphine Lariviere
- Department of Biochemistry and Molecular Biology, Penn State University, University Park, PA
| | - Sarah J C Craig
- Department of Biology, Penn State University, University Park, PA
- Center for Medical Genomics, Penn State University, University Park, PA
| | - Ian M Paul
- Center for Medical Genomics, Penn State University, University Park, PA
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA
| | - Emily E Hohman
- Center for Childhood Obesity Research, Penn State University, University Park, PA
| | - Jennifer S Savage
- Center for Childhood Obesity Research, Penn State University, University Park, PA
- Nutrition Department, Penn State University, University Park, PA
| | | | - Francesca Chiaromonte
- Center for Medical Genomics, Penn State University, University Park, PA
- Department of Statistics, Penn State University, University Park, PA
- EMbeDS, Sant'Anna School of Advanced Studies, Piazza Martiri della Libertà, Pisa, Italy
| | - Kateryna D Makova
- Department of Biology, Penn State University, University Park, PA
- Center for Medical Genomics, Penn State University, University Park, PA
| | - Matthew L Reimherr
- Center for Medical Genomics, Penn State University, University Park, PA
- Department of Statistics, Penn State University, University Park, PA
| |
Collapse
|
5
|
Chen Y, Jia J, Zhao Q, Zhang Y, Huang B, Wang L, Tian J, Huang C, Li M, Li X. Novel Loss-of-Function Variant in HNF1a Induces β-Cell Dysfunction through Endoplasmic Reticulum Stress. Int J Mol Sci 2022; 23:ijms232113022. [PMID: 36361808 PMCID: PMC9656704 DOI: 10.3390/ijms232113022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/21/2022] [Accepted: 10/21/2022] [Indexed: 12/02/2022] Open
Abstract
Heterozygous variants in the hepatocyte nuclear factor 1a (HNF1a) cause MODY3 (maturity-onset diabetes of the young, type 3). In this study, we found a case of novel HNF1a p.Gln125* (HNF1a-Q125ter) variant clinically. However, the molecular mechanism linking the new HNF1a variant to impaired islet β-cell function remains unclear. Firstly, a similar HNF1a-Q125ter variant in zebrafish (hnf1a+/−) was generated by CRISPR/Cas9. We further crossed hnf1a+/− with several zebrafish reporter lines to investigate pancreatic β-cell function. Next, we introduced HNF1a-Q125ter and HNF1a shRNA plasmids into the Ins-1 cell line and elucidated the molecular mechanism. hnf1a+/− zebrafish significantly decreased the β-cell number, insulin expression, and secretion. Moreover, β cells in hnf1a+/− dilated ER lumen and increased the levels of ER stress markers. Similar ER-stress phenomena were observed in an HNF1a-Q125ter-transfected Ins-1 cell. Follow-up investigations demonstrated that HNF1a-Q125ter induced ER stress through activating the PERK/eIF2a/ATF4 signaling pathway. Our study found a novel loss-of-function HNF1a-Q125ter variant which induced β-cell dysfunction by activating ER stress via the PERK/eIF2a/ATF4 signaling pathway.
Collapse
Affiliation(s)
- Yinling Chen
- Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361005, China
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences and School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Jianxin Jia
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences and School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Qing Zhao
- Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361005, China
| | - Yuxian Zhang
- Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361005, China
- Fujian Province Key Laboratory of Diabetes Translational Medicine, Xiamen Diabetes Institute, Xiamen 361003, China
| | - Bingkun Huang
- Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361005, China
- Fujian Province Key Laboratory of Diabetes Translational Medicine, Xiamen Diabetes Institute, Xiamen 361003, China
| | - Likun Wang
- Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361005, China
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences and School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Juanjuan Tian
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences and School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Caoxin Huang
- Fujian Province Key Laboratory of Diabetes Translational Medicine, Xiamen Diabetes Institute, Xiamen 361003, China
| | - Mingyu Li
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences and School of Life Sciences, Xiamen University, Xiamen 361102, China
- Correspondence: (M.L.); (X.L.)
| | - Xuejun Li
- Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361005, China
- Fujian Province Key Laboratory of Diabetes Translational Medicine, Xiamen Diabetes Institute, Xiamen 361003, China
- Correspondence: (M.L.); (X.L.)
| |
Collapse
|
6
|
Mizutani H, Sato Y, Yamazaki M, Yoshizawa T, Ando Y, Ueda M, Yamagata K. SIRT7 Deficiency Protects against Aβ 42-Induced Apoptosis through the Regulation of NOX4-Derived Reactive Oxygen Species Production in SH-SY5Y Cells. Int J Mol Sci 2022; 23:ijms23169027. [PMID: 36012298 PMCID: PMC9408927 DOI: 10.3390/ijms23169027] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/11/2022] [Accepted: 08/11/2022] [Indexed: 11/16/2022] Open
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disease that is characterized by irreversible memory loss and cognitive decline. The deposition of amyloid-β (Aβ), especially aggregation-prone Aβ42, is considered to be an early event preceding neurodegeneration in AD. Sirtuins (SIRT1-7 in mammals) are nicotinamide adenine dinucleotide-dependent lysine deacetylases/deacylases, and several sirtuins play important roles in AD. However, the involvement of SIRT7 in AD pathogenesis is not known. Here, we demonstrate that SIRT7 mRNA expression is increased in the cortex, entorhinal cortex, and prefrontal cortex of AD patients. We also found that Aβ42 treatment rapidly increased NADPH oxidase 4 (NOX4) expression at the post-transcriptional level, and induced reactive oxygen species (ROS) production and apoptosis in neuronal SH-SY5Y cells. In contrast, SIRT7 knockdown inhibited Aβ42-induced ROS production and apoptosis by suppressing the upregulation of NOX4. Collectively, these findings suggest that the inhibition of SIRT7 may play a beneficial role in AD pathogenesis through the regulation of ROS production.
Collapse
Affiliation(s)
- Hironori Mizutani
- Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-0811, Japan
| | - Yoshifumi Sato
- Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
- Correspondence: (Y.S.); (K.Y.); Tel.: +81-96-373-5068 (Y.S. & K.Y.); Fax: +81-96-364-6940 (Y.S. & K.Y.)
| | - Masaya Yamazaki
- Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Tatsuya Yoshizawa
- Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Yukio Ando
- Department of Amyloidosis Research, Faculty of Pharmaceutical Sciences, Nagasaki International University, 2825-7 Huis Ten Bosch Sasebo, Nagasaki 859-3298, Japan
| | - Mitsuharu Ueda
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-0811, Japan
| | - Kazuya Yamagata
- Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
- Center for Metabolic Regulation of Healthy Aging, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
- Correspondence: (Y.S.); (K.Y.); Tel.: +81-96-373-5068 (Y.S. & K.Y.); Fax: +81-96-364-6940 (Y.S. & K.Y.)
| |
Collapse
|
7
|
Harvey KE, LaVigne EK, Dar MS, Salyer AE, Pratt EPS, Sample PA, Aryal UK, Gowher H, Hockerman GH. RyR2/IRBIT regulates insulin gene transcript, insulin content, and secretion in the insulinoma cell line INS-1. Sci Rep 2022; 12:7713. [PMID: 35562179 PMCID: PMC9095623 DOI: 10.1038/s41598-022-11276-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 03/31/2022] [Indexed: 12/01/2022] Open
Abstract
The role of ER Ca2+ release via ryanodine receptors (RyR) in pancreatic β-cell function is not well defined. Deletion of RyR2 from the rat insulinoma INS-1 (RyR2KO) enhanced IP3 receptor activity stimulated by 7.5 mM glucose, coincident with reduced levels of the protein IP3 Receptor Binding protein released with Inositol 1,4,5 Trisphosphate (IRBIT). Insulin content, basal (2.5 mM glucose) and 7.5 mM glucose-stimulated insulin secretion were reduced in RyR2KO and IRBITKO cells compared to controls. INS2 mRNA levels were reduced in both RyR2KO and IRBITKO cells, but INS1 mRNA levels were specifically decreased in RyR2KO cells. Nuclear localization of S-adenosylhomocysteinase (AHCY) was increased in RyR2KO and IRBITKO cells. DNA methylation of the INS1 and INS2 gene promotor regions was very low, and not different among RyR2KO, IRBITKO, and controls, but exon 2 of the INS1 and INS2 genes was more extensively methylated in RyR2KO and IRBITKO cells. Exploratory proteomic analysis revealed that deletion of RyR2 or IRBIT resulted in differential regulation of 314 and 137 proteins, respectively, with 41 in common. These results suggest that RyR2 regulates IRBIT levels and activity in INS-1 cells, and together maintain insulin content and secretion, and regulate the proteome, perhaps via DNA methylation.
Collapse
Affiliation(s)
- Kyle E Harvey
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
| | - Emily K LaVigne
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
- Interdisciplinary Life Sciences Program, Purdue University, West Lafayette, IN, USA
| | - Mohd Saleem Dar
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA
| | - Amy E Salyer
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
| | - Evan P S Pratt
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
- Interdisciplinary Life Sciences Program, Purdue University, West Lafayette, IN, USA
| | - Paxton A Sample
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
| | - Uma K Aryal
- Proteomics Facility, Bindley Bioscience Center, Purdue University, West Lafayette, IN, USA
| | - Humaira Gowher
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA
| | - Gregory H Hockerman
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
8
|
Barth R, Ruoso C, Ferreira SM, de Ramos FC, Lima FB, Boschero AC, Santos GJD. Hepatocyte Nuclear Factor 4-α (HNF4α) controls the insulin resistance-induced pancreatic β-cell mass expansion. Life Sci 2022; 289:120213. [PMID: 34902439 DOI: 10.1016/j.lfs.2021.120213] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/17/2021] [Accepted: 12/01/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Regardless of the etiology, any type of DM presents a reduction of insulin-secreting cell mass, so it is important to investigate pathways that induce the increase of this cell mass. AIM Based on the fact that (1) HNF4α is crucial for β-cell proliferation, (2) DEX-induced IR promotes β-cell mass expansion, and (3) the stimulation of β-cell mass expansion may be an important target for DM therapies, we aimed to investigate whether DEX-induced proliferation of β pancreatic cells is dependent on HNF4α. METHODS We used WildType (WT) and Knockout (KO) mice for HNF4-α, treated or not with 100 mg/Kg/day of DEX, for 5 consecutive days. One day after the last injection of DEX the IR was confirmed by ipITT and the mice were euthanized for pancreas removal. RESULTS In comparison to WT, KO mice presented increased glucose tolerance, lower fasting glucose and increased glucose-stimulates insulin secretion (GSIS). DEX induced IR in both KO and WT mice. In addition, DEX-induced β-cell mass expansion and an increase in the Ki67 immunostaining were observed only in WT mice, evidencing that IR-induced β-cell mass expansion is dependent on HNF4α. Also, we observed that DEX-treatment, in an HNF4α-dependent way, promoted an increase in PDX1, PAX4 and NGN3 gene expression. CONCLUSIONS Our results strongly suggest that DEX-induced IR promotes β-cell mass expansion through processes of proliferation and neogenesis that depend on the HNF4α activity, pointing to HNF4α as a possible therapeutic target in DM treatment.
Collapse
Affiliation(s)
- Robson Barth
- Islet Biology and Metabolism Lab - I.B.M. Lab, Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina - UFSC, Florianópolis 88040-900, Santa Catarina, Brazil; Multicenter Graduate Program in Physiological Sciences, Federal University of Santa Catarina (UFSC), 88040-900, Florianópolis, SC, Brazil
| | - Carolina Ruoso
- Islet Biology and Metabolism Lab - I.B.M. Lab, Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina - UFSC, Florianópolis 88040-900, Santa Catarina, Brazil; Multicenter Graduate Program in Physiological Sciences, Federal University of Santa Catarina (UFSC), 88040-900, Florianópolis, SC, Brazil
| | - Sandra Mara Ferreira
- Laboratory of endocrine pancreas and metabolism - LAPEM, Department of Structural and Functional Biology, Institute of Biology, State University of Campinas - UNICAMP, 13083-862 Campinas, Brazil
| | - Francieli Caroline de Ramos
- Islet Biology and Metabolism Lab - I.B.M. Lab, Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina - UFSC, Florianópolis 88040-900, Santa Catarina, Brazil; Multicenter Graduate Program in Physiological Sciences, Federal University of Santa Catarina (UFSC), 88040-900, Florianópolis, SC, Brazil
| | - Fernanda Barbosa Lima
- Islet Biology and Metabolism Lab - I.B.M. Lab, Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina - UFSC, Florianópolis 88040-900, Santa Catarina, Brazil; Multicenter Graduate Program in Physiological Sciences, Federal University of Santa Catarina (UFSC), 88040-900, Florianópolis, SC, Brazil
| | - Antônio Carlos Boschero
- Laboratory of endocrine pancreas and metabolism - LAPEM, Department of Structural and Functional Biology, Institute of Biology, State University of Campinas - UNICAMP, 13083-862 Campinas, Brazil
| | - Gustavo Jorge Dos Santos
- Islet Biology and Metabolism Lab - I.B.M. Lab, Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina - UFSC, Florianópolis 88040-900, Santa Catarina, Brazil; Multicenter Graduate Program in Physiological Sciences, Federal University of Santa Catarina (UFSC), 88040-900, Florianópolis, SC, Brazil.
| |
Collapse
|
9
|
Yong J, Johnson JD, Arvan P, Han J, Kaufman RJ. Therapeutic opportunities for pancreatic β-cell ER stress in diabetes mellitus. Nat Rev Endocrinol 2021; 17:455-467. [PMID: 34163039 PMCID: PMC8765009 DOI: 10.1038/s41574-021-00510-4] [Citation(s) in RCA: 107] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/06/2021] [Indexed: 02/06/2023]
Abstract
Diabetes mellitus is characterized by the failure of insulin-secreting pancreatic β-cells (or β-cell death) due to either autoimmunity (type 1 diabetes mellitus) or failure to compensate for insulin resistance (type 2 diabetes mellitus; T2DM). In addition, mutations of critical genes cause monogenic diabetes. The endoplasmic reticulum (ER) is the primary site for proinsulin folding; therefore, ER proteostasis is crucial for both β-cell function and survival under physiological and pathophysiological challenges. Importantly, the ER is also the major intracellular Ca2+ storage organelle, generating Ca2+ signals that contribute to insulin secretion. ER stress is associated with the pathogenesis of diabetes mellitus. In this Review, we summarize the mutations in monogenic diabetes that play causal roles in promoting ER stress in β-cells. Furthermore, we discuss the possible mechanisms responsible for ER proteostasis imbalance with a focus on T2DM, in which both genetics and environment are considered important in promoting ER stress in β-cells. We also suggest that controlled insulin secretion from β-cells might reduce the progression of a key aspect of the metabolic syndrome, namely nonalcoholic fatty liver disease. Finally, we evaluate potential therapeutic approaches to treat T2DM, including the optimization and protection of functional β-cell mass in individuals with T2DM.
Collapse
Affiliation(s)
- Jing Yong
- Degenerative Diseases Program, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - James D Johnson
- Department of Cellular and Physiological Sciences & Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Peter Arvan
- Division of Metabolism Endocrinology & Diabetes, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Jaeseok Han
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan-si, Choongchungnam-do, Republic of Korea.
| | - Randal J Kaufman
- Degenerative Diseases Program, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, USA.
| |
Collapse
|
10
|
Decreased GLUT2 and glucose uptake contribute to insulin secretion defects in MODY3/HNF1A hiPSC-derived mutant β cells. Nat Commun 2021; 12:3133. [PMID: 34035238 PMCID: PMC8149827 DOI: 10.1038/s41467-021-22843-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 04/04/2021] [Indexed: 12/13/2022] Open
Abstract
Heterozygous HNF1A gene mutations can cause maturity onset diabetes of the young 3 (MODY3), characterized by insulin secretion defects. However, specific mechanisms of MODY3 in humans remain unclear due to lack of access to diseased human pancreatic cells. Here, we utilize MODY3 patient-derived human induced pluripotent stem cells (hiPSCs) to study the effect(s) of a causal HNF1A+/H126D mutation on pancreatic function. Molecular dynamics simulations predict that the H126D mutation could compromise DNA binding and gene target transcription. Genome-wide RNA-Seq and ChIP-Seq analyses on MODY3 hiPSC-derived endocrine progenitors reveal numerous HNF1A gene targets affected by the mutation. We find decreased glucose transporter GLUT2 expression, which is associated with reduced glucose uptake and ATP production in the MODY3 hiPSC-derived β-like cells. Overall, our findings reveal the importance of HNF1A in regulating GLUT2 and several genes involved in insulin secretion that can account for the insulin secretory defect clinically observed in MODY3 patients. Heterozygous HNF1A mutations can give rise to maturity onset diabetes of the young 3 (MODY3), characterized by insulin secretion defects. Here the authors show that MODY3-related HNF1A mutation in patient hiPSCderived pancreatic cells decreases glucose transporter GLUT2 expression due to compromised DNA binding.
Collapse
|
11
|
Wu W, He S, Shen Y, Zhang J, Wan Y, Tang X, Liu S, Yao X. Natural Product Luteolin Rescues THAP-Induced Pancreatic β-Cell Dysfunction through HNF4α Pathway. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2020; 48:1435-1454. [PMID: 32907363 DOI: 10.1142/s0192415x20500706] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Endoplasmic reticulum stress (ER stress) plays a main role in pancreatic [Formula: see text]-cell dysfunction and death because of intracellular Ca[Formula: see text] turbulence and inflammation activation. Although several drugs are targeting pancreatic [Formula: see text]-cell to improve [Formula: see text]-cell function, there still lacks agents to alleviate [Formula: see text]-cell ER stress conditions. Therefore we used thapsigargin (THAP) or high glucose (HG) to induce ER stress in [Formula: see text]-cell and aimed to screen natural molecules against ER stress-induced [Formula: see text]-cell dysfunction. Through screening the Traditional Chinese drug library ([Formula: see text] molecules), luteolin was finally discovered to improve [Formula: see text]-cell function. Cellular viability results indicated luteolin reduced the THAP or HG-induced [Formula: see text]-cell death and apoptosis through MTT and flow cytometry assay. Moreover, luteolin improved [Formula: see text]-cell insulin secretion ability under ER stress conditions. Also ER stress-induced intracellular Ca[Formula: see text] turbulence and inflammation activation were inhibited by luteolin treatment. Mechanically, luteolin inhibited HNF4[Formula: see text] signaling, which was induced by ER stress. Moreover, luteolin reduced the transcriptional level of HNF4[Formula: see text] downstream gene, such as Asnk4b and HNF1[Formula: see text]. Conversely HNF4[Formula: see text] knockdown abolished the effect of luteolin on [Formula: see text]-cell using siRNA. These results suggested the protective effect of luteolin on [Formula: see text]-cell was through HNF4[Formula: see text]/Asnk4b pathway. In conclusion, our study discovered that luteolin improved [Formula: see text]-cell function and disclosed the underlying mechanism of luteolin on [Formula: see text]-cell, suggesting luteolin is a promising agent against pancreatic dysfunction.
Collapse
Affiliation(s)
- Wenyu Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, P. R. China
| | - Shijun He
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern, Medical University, Guangzhou 510515, P. R. China
| | - Yuli Shen
- Nephrology Department, Longgang District People's Hospital of Shenzhen, Shenzhen 518172, P. R. China
| | - Jiawen Zhang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern, Medical University, Guangzhou 510515, P. R. China
| | - Yihong Wan
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern, Medical University, Guangzhou 510515, P. R. China
| | - Xiaodong Tang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern, Medical University, Guangzhou 510515, P. R. China
| | - Shuwen Liu
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern, Medical University, Guangzhou 510515, P. R. China.,Center of Pharmacy, Nanhai Hospital, Southern Medical University, Foshan 510080, P. R. China
| | - Xingang Yao
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern, Medical University, Guangzhou 510515, P. R. China.,Center of Clinical Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou 510515, P. R. China
| |
Collapse
|
12
|
Lin Q, Zhou S, Huang Y, Huo Z, Chen C, Luo X, He J, Liu C, Zhang P. ANKS4B Restricts Replication of Zika Virus by Downregulating the Autophagy. Front Microbiol 2020; 11:1745. [PMID: 32793175 PMCID: PMC7387654 DOI: 10.3389/fmicb.2020.01745] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/03/2020] [Indexed: 12/21/2022] Open
Abstract
Infection of Zika virus (ZIKV) has become a severe threaten to global health while no specific drug is available. In this study, we explored the relationship between ZIKV and a cellular protein, ankyrin repeat and sterile motif domain containing 4b (ANKS4B). Our data revealed that the expression of ANKS4B in cultured cells and in neonatal mice was downregulated by ZIKV infection. The reduction of ANKS4B upon ZIKV infection was caused by decrease of two hepatocyte nuclear factors HNF1α and HNF4α. Through CRISPR/Cas9 gene editing system, we generated two ANKS4B knockout (KO) cell clones in A549 and Huh7 cells respectively. In the ANKS4B-KO cells, the viral replication levels including viral RNA, protein, and titer were significantly enhanced, which was reversed by trans-complementation of ANKS4B. ANKS4B did not affect the viral entry step, but impaired the autophagy induced by ZIKV infection. Furthermore, our data showed that inhibition of autophagy led to similar replication levels of ZIKV in ANKS4B-sufficient and ANKS4B-deficient cells, suggesting the antiviral effect of ANKS4B relied on its modulation on the autophagy. Therefore, our work identified ANKS4B as a new restriction factor of ZIKV.
Collapse
Affiliation(s)
- Quanshi Lin
- Key Laboratory of Tropical Disease Control, Zhongshan School of Medicine, Sun Yat-sen University, Ministry of Education, Guangzhou, China.,Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Shili Zhou
- Key Laboratory of Tropical Disease Control, Zhongshan School of Medicine, Sun Yat-sen University, Ministry of Education, Guangzhou, China.,Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yanxia Huang
- Key Laboratory of Tropical Disease Control, Zhongshan School of Medicine, Sun Yat-sen University, Ministry of Education, Guangzhou, China.,Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhiting Huo
- Key Laboratory of Tropical Disease Control, Zhongshan School of Medicine, Sun Yat-sen University, Ministry of Education, Guangzhou, China.,Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Cancan Chen
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xin Luo
- Key Laboratory of Tropical Disease Control, Zhongshan School of Medicine, Sun Yat-sen University, Ministry of Education, Guangzhou, China.,Department of Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Junfang He
- Key Laboratory of Tropical Disease Control, Zhongshan School of Medicine, Sun Yat-sen University, Ministry of Education, Guangzhou, China.,Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Chao Liu
- Key Laboratory of Tropical Disease Control, Zhongshan School of Medicine, Sun Yat-sen University, Ministry of Education, Guangzhou, China.,Department of Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Ping Zhang
- Key Laboratory of Tropical Disease Control, Zhongshan School of Medicine, Sun Yat-sen University, Ministry of Education, Guangzhou, China.,Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
13
|
Sobuz SU, Sato Y, Yoshizawa T, Karim F, Ono K, Sawa T, Miyamoto Y, Oka M, Yamagata K. SIRT7 regulates the nuclear export of NF-κB p65 by deacetylating Ran. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2019; 1866:1355-1367. [PMID: 31075303 DOI: 10.1016/j.bbamcr.2019.05.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 04/19/2019] [Accepted: 05/01/2019] [Indexed: 12/25/2022]
Abstract
Sirtuin 7 (SIRT7) is an NAD+-dependent lysine deacetylase that regulates diverse biological processes. We recently observed that SIRT7 deficiency suppresses the nuclear accumulation of p65, which is a component of nuclear factor kappa B. However, the underlying molecular mechanism remains elusive. In this study, we demonstrated that SIRT7 interacts with a small GTPase, Ras-related nuclear antigen (Ran), and deacetylates Ran at K37. The nuclear export of p65 was facilitated in SIRT7-deficient fibroblast cells, while the nuclear export was inhibited in SIRT7-deficient cells expressing K37R-Ran (deacetylation-mimicking mutant). Additionally, the nuclear export of p65 in wild-type fibroblast cells was promoted by K37Q-Ran (acetylation-mimicking mutant). K37Q-Ran exhibited an increased ability to bind to chromosome region maintenance 1 (CRM1), which is a major nuclear receptor that mediates the export of cargo proteins, and enhanced the binding between p65 and CRM1. These data suggest that SIRT7 is a lysine deacetylase that targets the K37 residue of Ran to suppress the nuclear export of p65.
Collapse
Affiliation(s)
- Shihab U Sobuz
- Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Yoshifumi Sato
- Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Tatsuya Yoshizawa
- Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Fazlul Karim
- Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Katsuhiko Ono
- Department of Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Tomohiro Sawa
- Department of Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Yoichi Miyamoto
- Laboratory of Nuclear Transport Dynamics, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki-shi, Osaka 567-0085, Japan
| | - Masahiro Oka
- Laboratory of Nuclear Transport Dynamics, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki-shi, Osaka 567-0085, Japan
| | - Kazuya Yamagata
- Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan; Center for Metabolic Regulation of Healthy Aging (CMHA), Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan.
| |
Collapse
|
14
|
Camolotto SA, Belova VK, Snyder EL. The role of lineage specifiers in pancreatic ductal adenocarcinoma. J Gastrointest Oncol 2018; 9:1005-1013. [PMID: 30603119 DOI: 10.21037/jgo.2018.05.04] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Over the last decade, multiple genomics studies have led to the identification of discrete molecular subtypes of pancreatic ductal adenocarcinoma. A general theme has emerged that most pancreatic ductal adenocarcinoma (PDAC) can be grouped into two major subtypes based on cancer cell autonomous properties: classical/pancreatic progenitor and basal-like/squamous. The classical/progenitor subtype expresses higher levels of lineage specifiers that regulate endodermal differentiation than the basal-like/squamous subtype. The basal-like/squamous subtype confers a worse prognosis, raising the possibility that loss of these lineage specifiers might enhance the malignant potential of PDAC. Here, we discuss several of these differentially expressed lineage specifiers and examine the evidence that they might play a functional role in PDAC biology.
Collapse
Affiliation(s)
| | - Veronika K Belova
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Eric L Snyder
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.,Department of Pathology, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
15
|
Chen HY, Chang JTC, Chien KY, Lee YS, You GR, Cheng AJ. The Endogenous GRP78 Interactome in Human Head and Neck Cancers: A Deterministic Role of Cell Surface GRP78 in Cancer Stemness. Sci Rep 2018; 8:536. [PMID: 29323121 PMCID: PMC5765009 DOI: 10.1038/s41598-017-14604-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 10/09/2017] [Indexed: 12/04/2022] Open
Abstract
Cell surface glucose regulated protein 78 (GRP78), an endoplasmic reticulum (ER) chaperone, was suggested to be a cancer stem cell marker, but the influence of this molecule on cancer stemness is poorly characterized. In this study, we developed a mass spectrometry platform to detect the endogenous interactome of GRP78 and investigated its role in cancer stemness. The interactome results showed that cell surface GRP78 associates with multiple molecules. The influence of cell population heterogeneity of head and neck cancer cell lines (OECM1, FaDu, and BM2) according to the cell surface expression levels of GRP78 and the GRP78 interactome protein, Progranulin, was investigated. The four sorted cell groups exhibited distinct cell cycle distributions, asymmetric/symmetric cell divisions, and different relative expression levels of stemness markers. Our results demonstrate that cell surface GRP78 promotes cancer stemness, whereas drives cells toward a non-stemlike phenotype when it chaperones Progranulin. We conclude that cell surface GRP78 is a chaperone exerting a deterministic influence on cancer stemness.
Collapse
Affiliation(s)
- Hsin-Ying Chen
- Graduate Institute of Biomedical Sciences, Chang Gung University College of Medicine, Tao-Yuan, Taiwan
| | | | - Kun-Yi Chien
- Graduate Institute of Biomedical Sciences, Chang Gung University College of Medicine, Tao-Yuan, Taiwan.,Proteomics Core Laboratory, Molecular Medicine Research Center, Chang Gung University, Tao-Yuan, Taiwan
| | - Yun-Shien Lee
- Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan.,Department of Biotechnology, Ming Chuan University, Tao-Yuan, Taiwan
| | - Guo-Rung You
- Graduate Institute of Biomedical Sciences, Chang Gung University College of Medicine, Tao-Yuan, Taiwan
| | - Ann-Joy Cheng
- Graduate Institute of Biomedical Sciences, Chang Gung University College of Medicine, Tao-Yuan, Taiwan. .,Department of Radiation Oncology, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan. .,Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan.
| |
Collapse
|
16
|
Cnop M, Toivonen S, Igoillo-Esteve M, Salpea P. Endoplasmic reticulum stress and eIF2α phosphorylation: The Achilles heel of pancreatic β cells. Mol Metab 2017; 6:1024-1039. [PMID: 28951826 PMCID: PMC5605732 DOI: 10.1016/j.molmet.2017.06.001] [Citation(s) in RCA: 179] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 05/19/2017] [Accepted: 06/01/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Pancreatic β cell dysfunction and death are central in the pathogenesis of most if not all forms of diabetes. Understanding the molecular mechanisms underlying β cell failure is important to develop β cell protective approaches. SCOPE OF REVIEW Here we review the role of endoplasmic reticulum stress and dysregulated endoplasmic reticulum stress signaling in β cell failure in monogenic and polygenic forms of diabetes. There is substantial evidence for the presence of endoplasmic reticulum stress in β cells in type 1 and type 2 diabetes. Direct evidence for the importance of this stress response is provided by an increasing number of monogenic forms of diabetes. In particular, mutations in the PERK branch of the unfolded protein response provide insight into its importance for human β cell function and survival. The knowledge gained from different rodent models is reviewed. More disease- and patient-relevant models, using human induced pluripotent stem cells differentiated into β cells, will further advance our understanding of pathogenic mechanisms. Finally, we review the therapeutic modulation of endoplasmic reticulum stress and signaling in β cells. MAJOR CONCLUSIONS Pancreatic β cells are sensitive to excessive endoplasmic reticulum stress and dysregulated eIF2α phosphorylation, as indicated by transcriptome data, monogenic forms of diabetes and pharmacological studies. This should be taken into consideration when devising new therapeutic approaches for diabetes.
Collapse
Key Words
- ATF, activating transcription factor
- CHOP, C/EBP homologous protein
- CRISPR, clustered regularly interspaced short palindromic repeats
- CReP, constitutive repressor of eIF2α phosphorylation
- Diabetes
- ER, endoplasmic reticulum
- ERAD, ER-associated degradation
- Endoplasmic reticulum stress
- GCN2, general control non-derepressible-2
- GIP, glucose-dependent insulinotropic polypeptide
- GLP-1, glucagon-like peptide 1
- GWAS, genome-wide association study
- HNF1A, hepatocyte nuclear factor 1-α
- HRI, heme-regulated inhibitor kinase
- IAPP, islet amyloid polypeptide
- IER3IP1, immediate early response-3 interacting protein-1
- IRE1, inositol-requiring protein-1
- ISR, integrated stress response
- Insulin
- Islet
- MEHMO, mental retardation, epilepsy, hypogonadism and -genitalism, microcephaly and obesity
- MODY, maturity-onset diabetes of the young
- NRF2, nuclear factor, erythroid 2 like 2
- PBA, 4-phenyl butyric acid
- PERK, PKR-like ER kinase
- PKR, protein kinase RNA
- PP1, protein phosphatase 1
- PPA, phenylpropenoic acid glucoside
- Pancreatic β cell
- Pdx1, pancreatic duodenal homeobox 1
- RIDD, regulated IRE1-dependent decay
- RyR2, type 2 ryanodine receptor/Ca2+ release channel
- SERCA, sarcoendoplasmic reticulum Ca2+ ATPase
- TUDCA, taurine-conjugated ursodeoxycholic acid derivative
- UPR, unfolded protein response
- WFS, Wolfram syndrome
- XBP1, X-box binding protein 1
- eIF2, eukaryotic translation initiation factor 2
- eIF2α
- hESC, human embryonic stem cell
- hPSC, human pluripotent stem cell
- hiPSC, human induced pluripotent stem cell
- uORF, upstream open reading frame
Collapse
Affiliation(s)
- Miriam Cnop
- ULB Center for Diabetes Research, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
- Division of Endocrinology, Erasmus Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Sanna Toivonen
- ULB Center for Diabetes Research, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Mariana Igoillo-Esteve
- ULB Center for Diabetes Research, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Paraskevi Salpea
- ULB Center for Diabetes Research, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
17
|
Sato Y, Tsuyama T, Sato C, Karim MF, Yoshizawa T, Inoue M, Yamagata K. Hypoxia reduces HNF4α/MODY1 protein expression in pancreatic β-cells by activating AMP-activated protein kinase. J Biol Chem 2017; 292:8716-8728. [PMID: 28364040 DOI: 10.1074/jbc.m116.767574] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 03/29/2017] [Indexed: 12/12/2022] Open
Abstract
Hypoxia plays a role in the deterioration of β-cell function. Hepatocyte nuclear factor 4α (HNF4α) has an important role in pancreatic β-cells, and mutations of the human HNF4A gene cause a type of maturity-onset diabetes of the young (MODY1). However, it remains unclear whether hypoxia affects the expression of HNF4α in β-cells. Here, we report that hypoxia reduces HNF4α protein expression in β-cells. Hypoxia-inducible factor was not involved in the down-regulation of HNF4α under hypoxic conditions. The down-regulation of HNF4α was dependent on the activation of AMP-activated protein kinase (AMPK), and the reduction of HNF4α protein expression by metformin, an AMPK activator, and hypoxia was inhibited by the overexpression of a kinase-dead (KD) form of AMPKα2. In addition, hypoxia decreased the stability of the HNF4α protein, and the down-regulation of HNF4α was sensitive to proteasome inhibitors. Adenovirus-mediated overexpression of KD-AMPKα2 improved insulin secretion in metformin-treated islets, hypoxic islets, and ob/ob mouse islets. These results suggest that down-regulation of HNF4α could be of importance in β-cell dysfunction by hypoxia.
Collapse
Affiliation(s)
- Yoshifumi Sato
- From the Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University, Kumamoto, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556 and
| | - Tomonori Tsuyama
- From the Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University, Kumamoto, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556 and
| | - Chinami Sato
- From the Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University, Kumamoto, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556 and
| | - Md Fazlul Karim
- From the Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University, Kumamoto, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556 and
| | - Tatsuya Yoshizawa
- From the Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University, Kumamoto, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556 and
| | - Masahiro Inoue
- the Department of Biochemistry, Osaka Medical Center for Cancer and Cardiovascular Diseases, 1-3-3 Nakamichi, Higashinari-ku, Osaka 537-8511, Japan
| | - Kazuya Yamagata
- From the Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University, Kumamoto, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556 and
| |
Collapse
|
18
|
Moore BD, Khurana SS, Huh WJ, Mills JC. Hepatocyte nuclear factor 4α is required for cell differentiation and homeostasis in the adult mouse gastric epithelium. Am J Physiol Gastrointest Liver Physiol 2016; 311:G267-75. [PMID: 27340127 PMCID: PMC5007292 DOI: 10.1152/ajpgi.00195.2016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 06/17/2016] [Indexed: 01/31/2023]
Abstract
We have previously shown that the sequential transcription factors Xbp1→Mist1 (Bhlha15) govern the ultrastructural maturation of the secretory apparatus in enzyme-secreting zymogenic chief cells (ZCs) in the gastric unit. Here we sought to identify transcriptional regulators upstream of X-box binding protein 1 (XBP1) and MIST1. We used immunohistochemistry to characterize Hnf4α(flox/flox) adult mouse stomachs after tamoxifen-induced deletion of Hnf4α We used qRT-PCR, Western blotting, and chromatin immunoprecipitation to define the molecular interaction between hepatocyte nuclear factor 4 alpha (HNF4α) and Xbp1 in mouse stomach and human gastric cells. We show that HNF4α protein is expressed in pit (foveolar) cells, mucous neck cells, and zymogenic chief cells (ZCs) of the corpus gastric unit. Loss of HNF4α in adult mouse stomach led to reduced ZC size and ER content, phenocopying previously characterized effects of Xbp1 deletion. However, HNF4α(Δ/Δ) stomachs also exhibited additional phenotypes including increased proliferation in the isthmal stem cell zone and altered mucous neck cell migration, indicating a role of HNF4α in progenitor cells as well as in ZCs. HNF4α directly occupies the Xbp1 promoter locus in mouse stomach, and forced HNF4α expression increased abundance of XBP1 mRNA in human gastric cancer cells. Finally, as expected, loss of HNF4α caused decreased Xbp1 and Mist1 expression in mouse stomachs. We show that HNF4α regulates homeostatic proliferation in the gastric epithelium and is both necessary and sufficient for the upstream regulation of the Xbp1→Mist1 axis in maintenance of ZC secretory architecture.
Collapse
Affiliation(s)
- Benjamin D. Moore
- 1Division of Gastroenterology, Departments of Medicine, Pathology and Immunology, and Developmental Biology, Washington University, St. Louis, Missouri
| | - Shradha S. Khurana
- 1Division of Gastroenterology, Departments of Medicine, Pathology and Immunology, and Developmental Biology, Washington University, St. Louis, Missouri
| | - Won Jae Huh
- 1Division of Gastroenterology, Departments of Medicine, Pathology and Immunology, and Developmental Biology, Washington University, St. Louis, Missouri
| | - Jason C. Mills
- 1Division of Gastroenterology, Departments of Medicine, Pathology and Immunology, and Developmental Biology, Washington University, St. Louis, Missouri
| |
Collapse
|
19
|
Moore BD, Jin RU, Lo H, Jung M, Wang H, Battle MA, Wollheim CB, Urano F, Mills JC. Transcriptional Regulation of X-Box-binding Protein One (XBP1) by Hepatocyte Nuclear Factor 4α (HNF4Α) Is Vital to Beta-cell Function. J Biol Chem 2016; 291:6146-57. [PMID: 26792861 DOI: 10.1074/jbc.m115.685750] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Indexed: 12/20/2022] Open
Abstract
The transcription factor, X-box-binding protein-1 (XBP1), controls the development and maintenance of the endoplasmic reticulum (ER) in multiple secretory cell lineages. We show here that Hepatocyte Nuclear Factor 4α (HNF4α) directly induces XBP1 expression. Mutations in HNF4α cause Mature-Onset Diabetes of the Young I (MODYI), a subset of diabetes characterized by diminished GSIS. In mouse models, cell lines, and ex vivo islets, using dominant negative and human- disease-allele point mutants or knock-out and knockdown models, we show that disruption of HNF4α caused decreased expression of XBP1 and reduced cellular ER networks. GSIS depends on ER Ca(2+) signaling; we show that diminished XBP1 and/or HNF4α in β-cells led to impaired ER Ca(2+) homeostasis. Restoring XBP1 expression was sufficient to completely rescue GSIS in HNF4α-deficient β-cells. Our findings uncover a transcriptional relationship between HNF4α and Xbp1 with potentially broader implications about MODYI and the importance of transcription factor signaling in the regulation of secretion.
Collapse
Affiliation(s)
- Benjamin D Moore
- From the Division of Gastroenterology, Departments of Medicine, Pathology & Immunology, and Developmental Biology, Washington University, St. Louis, Missouri 63110
| | - Ramon U Jin
- From the Division of Gastroenterology, Departments of Medicine, Pathology & Immunology, and Developmental Biology, Washington University, St. Louis, Missouri 63110
| | - Heiyong Lo
- From the Division of Gastroenterology, Departments of Medicine, Pathology & Immunology, and Developmental Biology, Washington University, St. Louis, Missouri 63110
| | - Min Jung
- From the Division of Gastroenterology, Departments of Medicine, Pathology & Immunology, and Developmental Biology, Washington University, St. Louis, Missouri 63110
| | - Haiyan Wang
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, CH-4070 Basel, Switzerland
| | - Michele A Battle
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Claes B Wollheim
- Lund University Diabetes Center, Department of Clinical Sciences, Diabetes & Endocrinology, Skåne University Hospital, Lund University, Malmö 20502, Sweden, Department of Cell Physiology and Metabolism, Université de Genève, University Medical Centre, 1 rue Michel-Servet, Geneva 4 1211, Switzerland, and
| | - Fumihiko Urano
- Division of Endocrinology, Departments of Medicine, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Jason C Mills
- From the Division of Gastroenterology, Departments of Medicine, Pathology & Immunology, and Developmental Biology, Washington University, St. Louis, Missouri 63110,
| |
Collapse
|
20
|
Jin L, Wang K, Liu H, Chen T, Yang Y, Ma X, Wang J, Li Y, Du D, Zhao Y, He Y. Genomewide Histone H3 Lysine 9 Acetylation Profiling in CD4+ T Cells Revealed Endoplasmic Reticulum Stress Deficiency in Patients with Acute‐on‐chronic Liver Failure. Scand J Immunol 2015; 82:452-9. [PMID: 26173605 DOI: 10.1111/sji.12341] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 07/07/2015] [Indexed: 12/22/2022]
Affiliation(s)
- L. Jin
- Department of Infectious Diseases the First Affiliated Hospital of Medical College Xi'an Jiaotong University Xi'an Shaanxi Province China
| | - K. Wang
- Department of Infectious Diseases the Third Affiliated Hospital Sun Yat‐sen University Guangzhou City Guangdong Province China
| | - H. Liu
- The Eighth Hospital Xi'an Shaanxi Province China
| | - T. Chen
- Department of Infectious Diseases the First Affiliated Hospital of Medical College Xi'an Jiaotong University Xi'an Shaanxi Province China
| | - Y. Yang
- Department of Infectious Diseases the First Affiliated Hospital of Medical College Xi'an Jiaotong University Xi'an Shaanxi Province China
| | - X. Ma
- The Eighth Hospital Xi'an Shaanxi Province China
| | - J. Wang
- Department of Infectious Diseases the First Affiliated Hospital of Medical College Xi'an Jiaotong University Xi'an Shaanxi Province China
| | - Y. Li
- Xi'an children's hospital Xi'an Shaanxi Province China
| | - D. Du
- Department of Infectious Diseases the First Affiliated Hospital of Medical College Xi'an Jiaotong University Xi'an Shaanxi Province China
| | - Y. Zhao
- Department of Infectious Diseases the First Affiliated Hospital of Medical College Xi'an Jiaotong University Xi'an Shaanxi Province China
| | - Y. He
- Department of Infectious Diseases the First Affiliated Hospital of Medical College Xi'an Jiaotong University Xi'an Shaanxi Province China
| |
Collapse
|
21
|
Roles of hepatic glucokinase in intertissue metabolic communication: Examination of novel liver-specific glucokinase knockout mice. Biochem Biophys Res Commun 2015; 460:727-32. [PMID: 25817793 DOI: 10.1016/j.bbrc.2015.03.097] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 03/17/2015] [Indexed: 11/21/2022]
Abstract
Glucokinase is expressed principally in pancreatic β-cells and hepatocytes, and catalyzes the phosphorylation of glucose to glucose-6-phosphate, a rate-limiting step of glycolysis. To better understand the roles of hepatic glucokinase, we generated Gck knockout mice by ablating liver-specific exon 1b. The knockout mice exhibited impaired glucose tolerance, decreased hepatic glycogen content, and reduced Pklr and Fas gene expression in the liver, indicating that hepatic glucokinase plays important roles in glucose metabolism. It has also been reported that hepatic glucokinase regulates the expression of thermogenesis-related genes in brown adipose tissue (BAT) and insulin secretion in response to glucose. However, the liver-specific Gck knockout mice displayed neither altered expression of thermogenesis-related genes in BAT nor impaired insulin secretion by β-cells under a normal chow diet. These results suggest that chronic suppression of hepatic glucokinase has a small influence on intertissue (liver-to-BAT as well as liver-to-β-cell) metabolic communication.
Collapse
|
22
|
Yamagata K. Roles of HNF1α and HNF4α in pancreatic β-cells: lessons from a monogenic form of diabetes (MODY). VITAMINS AND HORMONES 2015; 95:407-23. [PMID: 24559927 DOI: 10.1016/b978-0-12-800174-5.00016-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mutations in the genes encoding hepatocyte nuclear factor (HNF)1α and HNF4α cause a monogenic form of diabetes mellitus known as maturity-onset diabetes of the young (MODY). The primary cause of MODY is an impairment of glucose-stimulated insulin secretion by pancreatic β-cells, indicating the important roles of HNF1α and HNF4α in β-cells. Large-scale genetic studies have clarified that the common variants of HNF1α and HNF4α genes are also associated with type 2 diabetes, suggesting that they are involved in the pathogenesis of both diseases. Recent experimental studies revealed that HNF1α controls both β-cell function and growth by regulating target genes such as glucose transporter 2, pyruvate kinase, collectrin, hepatocyte growth factor activator, and HNF4α. In contrast, HNF4α mainly regulates the function of β-cells. Although direct target genes of HNF4α in β-cells are largely unknown, we recently identified Anks4b as a novel target of HNF4α that regulates β-cell susceptibility to endoplasmic reticulum stress. Studies of MODY have led to a better understanding of the molecular mechanism of glucose-stimulated insulin secretion by pancreatic β-cells.
Collapse
Affiliation(s)
- Kazuya Yamagata
- Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
23
|
Li K, Han X. Endoplasmic reticulum stress is involved in the lidocaine-induced apoptosis in SH-SY5Y neuroblastoma cells. J Mol Neurosci 2014; 56:122-30. [PMID: 25522790 DOI: 10.1007/s12031-014-0473-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 11/18/2014] [Indexed: 01/20/2023]
Abstract
Lidocaine has been indicated to promote apoptosis and to promote endoplasmic reticulum (ER) stress. However, the mechanism underlining ER stress-mediated apoptosis is unclear. In the present study, we investigated the promotion to ER stress in the lidocaine-induced apoptosis in human neuroblastoma SH-SY5Y cells. Firstly, we confirmed that lidocaine treatment induced apoptosis in SH-SY5Y cells, time-dependently and dose-dependently, via MTT cell viability assay and annexin V/FITC apoptosis detection with a FACScan flow cytometer. And the anti-apoptosis Bcl-2 and Bcl-xL were downregulated, whereas the apoptosis-executive caspase 3 was promoted through Western blot assay and caspase 3 activity assay. Moreover, the ER stress-associated binding immunoglobulin protein (BiP), PKR-like ER kinase (PERK), activating transcription factor 4 (ATF4) and CCAAT/enhancer-binding protein homologous protein (CHOP) were also upregulated at both mRNA and protein levels by lidocaine treatment. On the other hand, downregulation of the ER stress-associated BiP by RNAi method not only blocked the lidocaine-promoted ER stress but also attenuated the lidocaine-induced SH-SY5Y cell apoptosis. In conclusion, the present study confirmed the involvement of ER stress in the lidocaine-induced apoptosis in human neuroblastoma SH-SY5Y cells. Our study provides a better understanding on the mechanism of lidocaine's neurovirulence.
Collapse
Affiliation(s)
- Kehan Li
- Department of Anesthesiology, The First Affiliated Hospital of Henan Science and Technology University, No. 24 Jinghua Road, Jianxi District, 471003, Luoyang, China,
| | | |
Collapse
|
24
|
Moderate hypoxia induces β-cell dysfunction with HIF-1-independent gene expression changes. PLoS One 2014; 9:e114868. [PMID: 25503986 PMCID: PMC4264765 DOI: 10.1371/journal.pone.0114868] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 11/14/2014] [Indexed: 01/09/2023] Open
Abstract
Pancreatic β-cell failure is central to the development and progression of type 2 diabetes. We recently demonstrated that β-cells become hypoxic under high glucose conditions due to increased oxygen consumption and that the pancreatic islets of diabetic mice but not those of control mice are moderately hypoxic. However, the impact of moderate hypoxia on β-cell number and function is unknown. In the present study, moderate hypoxia induced a hypoxic response in MIN6 cells, as evidenced by increased levels of HIF-1α protein and target genes. Under these conditions, a selective downregulation of Mafa, Pdx1, Slc2a2, Ndufa5, Kcnj11, Ins1, Wfs1, Foxa2, and Neurod1, which play important roles in β-cells, was also observed in both MIN6 cells and isolated pancreatic islets. Consistent with the altered expression of these genes, abnormal insulin secretion was detected in hypoxic MIN6 cells. Most of the hypoxia-induced gene downregulation in MIN6 cells was not affected by the suppression of HIF-1α, suggesting a HIF-1–independent mechanism. Moderate hypoxia also induced apoptosis in MIN6 cells. These results suggest that hypoxia is a novel stressor of β-cells and that hypoxic stress may play a role in the deterioration of β-cell function.
Collapse
|
25
|
Karim MF, Yoshizawa T, Sato Y, Sawa T, Tomizawa K, Akaike T, Yamagata K. Inhibition of H3K18 deacetylation of Sirt7 by Myb-binding protein 1a (Mybbp1a). Biochem Biophys Res Commun 2013; 441:157-63. [DOI: 10.1016/j.bbrc.2013.10.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Accepted: 10/07/2013] [Indexed: 11/29/2022]
|
26
|
He B, Luo B, Chen Q, Zhang L. Cigarette smoke extract induces the expression of GRP78 in A549 cells via the p38/MAPK pathway. Mol Med Rep 2013; 8:1683-8. [PMID: 24126384 DOI: 10.3892/mmr.2013.1724] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Accepted: 09/26/2013] [Indexed: 11/06/2022] Open
Abstract
Apoptosis of alveolar epithelial cells has been implicated in the pathogenesis of chronic obstructive pulmonary disease. To determine the involvement of glucose‑regulated protein 78 (GRP78) in the cigarette smoke extract (CSE)‑induced apoptosis of alveolar epithelial cells and the potential mechanisms underlying this effect, A549 cells that originate from alveolar type II epithelial cells were exposed to various CSE conditions in the present study. GRP78 expression and its effect on the apoptosis of A549 cells were investigated using techniques such as RT-PCR, western blot analysis, gene knockdown by GRP78 siRNA interference and the terminal deoxynucleotidyl transferase dUTP nick‑end labeling assay. The activity of the p38/mitogen‑activated protein kinase (MAPK) pathway and its involvement in GRP78 expression were also analyzed using SB203580, a p38/MAPK pathway inhibitor. It was demonstrated that GRP78 expression in the cells was significantly upregulated following CSE exposure and a 12‑h exposure of 5% CSE was the most efficient in inducing GRP78 expression. This CSE‑induced GRP78 expression was significantly attenuated by GRP78 siRNA or by the use of SB203580. The downregulation of GRP78 expression by GRP78 siRNA also led to the increased expression of caspase-3 and an increased apoptotic index (AI, P<0.05 vs. other groups). These results suggested that CSE induced GRP78 expression in A549 cells. This study demonstrated that upregulated GRP78 expression may be anti‑apoptotic effects and the p38/MAPK pathway was involved in the process of CSE‑induced GRP78 expression in A549 cells.
Collapse
Affiliation(s)
- Baimei He
- Department of Geriatric Medicine, Xiangya Hospital of Central South University, Changsha, Hunan 410008, P.R. China
| | | | | | | |
Collapse
|
27
|
The role of the unfolded protein response in diabetes mellitus. Semin Immunopathol 2013; 35:333-50. [PMID: 23529219 DOI: 10.1007/s00281-013-0369-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Accepted: 03/13/2013] [Indexed: 12/13/2022]
Abstract
The endoplasmic reticulum (ER) plays a key role in the synthesis and modification of secretory and membrane proteins in all eukaryotic cells. Under normal conditions, these proteins are correctly folded and assembled in the ER. However, when cells are exposed to environmental factors such as overproduction of ER proteins, viral infections, or glucose deprivation, the secretory and membrane proteins can accumulate in unfolded or misfolded forms in the lumen of the ER, and consequently, cause stress in the ER. To maintain cellular homeostasis, cells induce several responses to ER stress. In mammalian cells, ER stress responses are induced by a diversity of signal pathways. There are three ER-located transmembrane proteins that play important roles in mammalian ER stress responses: activating transcription factor 6, inositol-requiring protein 1, and protein kinase RNA-like endoplasmic reticulum kinase. ER stress is linked to various diseases, including diabetes. This review highlights the particular importance of ER stress-responsive molecules in insulin biosynthesis, glyconeogenesis, insulin resistance, glucose intolerance, and pancreatic β-cell apoptosis. An understanding of the pathogenic mechanism of diabetes from the aspect of ER stress is crucial in formulating therapeutic strategies.
Collapse
|
28
|
Abstract
Beta cell dysfunction and insulin resistance are inherently complex with their interrelation for triggering the pathogenesis of diabetes also somewhat undefined. Both pathogenic states induce hyperglycemia and therefore increase insulin demand. Beta cell dysfunction results from inadequate glucose sensing to stimulate insulin secretion therefore elevated glucose concentrations prevail. Persistently elevated glucose concentrations above the physiological range result in the manifestation of hyperglycemia. With systemic insulin resistance, insulin signaling within glucose recipient tissues is defective therefore hyperglycemia perseveres. Beta cell dysfunction supersedes insulin resistance in inducing diabetes. Both pathological states influence each other and presumably synergistically exacerbate diabetes. Preserving beta cell function and insulin signaling in beta cells and insulin signaling in the glucose recipient tissues will maintain glucose homeostasis.
Collapse
Affiliation(s)
- Marlon E. Cerf
- Diabetes Discovery Platform, South African Medical Research CouncilCape Town, South Africa
- *Correspondence: Marlon E. Cerf, Diabetes Discovery Platform, South African Medical Research Council, PO Box 19070, Tygerberg, Cape Town 7505, South Africa. e-mail:
| |
Collapse
|
29
|
Ohki T, Sato Y, Yoshizawa T, Yamamura KI, Yamada K, Yamagata K. Identification of hepatocyte growth factor activator (Hgfac) gene as a target of HNF1α in mouse β-cells. Biochem Biophys Res Commun 2012; 425:619-24. [PMID: 22877752 DOI: 10.1016/j.bbrc.2012.07.134] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 07/24/2012] [Indexed: 10/28/2022]
Abstract
HNF1α is a transcription factor that is expressed in pancreatic β-cells and mutations of the HNF1α gene cause a form of monogenic diabetes. To understand the role of HNF1α in pancreatic β-cells, we established the MIN6 β-cell line that stably expressed HNF1α-specific shRNA. Expression of the gene encoding hepatocyte growth factor (HGF) activator (Hgfac), a serine protease that efficiently activates HGF, was decreased in HNF1α KD-MIN6 cells. Down-regulation of Hgfac expression was also found in the islets of HNF1α (+/-) mice. Reporter gene analysis and the chromatin immunoprecipitation assay indicated that HNF1α directly regulates the expression of Hgfac in β-cells. It has been reported that HGF has an important influence on β-cell mass and β-cell function. Thus, HNF1α might regulate β-cell mass or function at least partly by modulating Hgfac expression.
Collapse
Affiliation(s)
- Tsuyoshi Ohki
- Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | | | | | | | | | | |
Collapse
|