1
|
Wang M, Guo Z, Zeng J, Liu L, Wang Y, Wang J, Lu H, Zhang H, Jiang H, Wang X. Bio-assembled smart nanocapsules for targeted delivery of KRAS shRNA and cancer cell bioimage. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.06.074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
2
|
Deficiency of two-pore segment channel 2 contributes to systemic lupus erythematosus via regulation of apoptosis and cell cycle. Chin Med J (Engl) 2022; 135:447-455. [PMID: 35194006 PMCID: PMC8869567 DOI: 10.1097/cm9.0000000000001893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Background: Methods: Results: Conclusion:
Collapse
|
3
|
Yang Y, Qiu W, Meng Q, Liu M, Lin W, Yang H, Wang R, Dong J, Yuan N, Zhou Z, He F. GRB10 rs1800504 Polymorphism Is Associated With the Risk of Coronary Heart Disease in Patients With Type 2 Diabetes Mellitus. Front Cardiovasc Med 2021; 8:728976. [PMID: 34651026 PMCID: PMC8505721 DOI: 10.3389/fcvm.2021.728976] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 09/02/2021] [Indexed: 12/14/2022] Open
Abstract
Diabetic vascular complications are one of the main causes of death and disability. Previous studies have reported that genetic variation is associated with diabetic vascular complications. In this study, we aimed to investigate the association between GRB10 polymorphisms and susceptibility to type 2 diabetes mellitus (T2DM) vascular complications. Eight single nucleotide polymorphisms (SNPs) in the GRB10 gene were genotyped by MassARRAY system and 934 patients with type 2 diabetes mellitus (T2DM) were included for investigation. We found that GRB10 rs1800504 CC+CT genotypes were significantly associated with increased risk of coronary heart disease (CHD) compared with TT genotype (OR = 2.24; 95%CI: 1.36-3.70, p = 0.002). Consistently, levels of cholesterol (CHOL) (CC+CT vs. TT, 4.44 ± 1.25 vs. 4.10 ± 1.00 mmol/L; p = 0.009) and low density lipoprotein cholesterin (LDL-CH) (CC+CT vs. TT, 2.81 ± 1.07 vs. 2.53 ± 0.82 mmol/L; p = 0.01) in T2DM patients with TT genotype were significant lower than those of CC+CT genotypes. We further validated in MIHA cell that the total cholesterol (TC) level in GRB10-Mut was significantly reduced compared with GRB10-WT; p = 0.0005. Likewise, the reversed palmitic acid (PA) induced lipid droplet formation in GRB10-Mut was more effective than in GRB10-WT. These results suggest that rs1800504 of GRB10 variant may be associated with the blood lipids and then may also related to the risk of CHD in patients with T2DM.
Collapse
Affiliation(s)
- Yang Yang
- Department of Pharmacy, Zhuhai People's Hospital (Zhuhai Hospital Affiliated With Jinan University), Zhuhai, China
| | - Wentao Qiu
- Department of Pharmacy, Zhuhai People's Hospital (Zhuhai Hospital Affiliated With Jinan University), Zhuhai, China.,College of Pharmacy, Jinan University, Guangzhou, China
| | - Qian Meng
- Department of Pharmacy, Zhuhai People's Hospital (Zhuhai Hospital Affiliated With Jinan University), Zhuhai, China
| | - Mouze Liu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Weijie Lin
- Department of Pharmacy, Zhuhai People's Hospital (Zhuhai Hospital Affiliated With Jinan University), Zhuhai, China
| | - Haikui Yang
- Department of Pharmacy, Zhuhai People's Hospital (Zhuhai Hospital Affiliated With Jinan University), Zhuhai, China
| | - Ruiqi Wang
- Department of Pharmacy, Zhuhai People's Hospital (Zhuhai Hospital Affiliated With Jinan University), Zhuhai, China
| | - Jiamei Dong
- Department of Pharmacy, Zhuhai People's Hospital (Zhuhai Hospital Affiliated With Jinan University), Zhuhai, China
| | - Ningning Yuan
- Department of Pharmacy, Zhuhai People's Hospital (Zhuhai Hospital Affiliated With Jinan University), Zhuhai, China
| | - Zhiling Zhou
- Department of Pharmacy, Zhuhai People's Hospital (Zhuhai Hospital Affiliated With Jinan University), Zhuhai, China
| | - Fazhong He
- Department of Pharmacy, Zhuhai People's Hospital (Zhuhai Hospital Affiliated With Jinan University), Zhuhai, China
| |
Collapse
|
4
|
Wang Q, Wang H, Jing Q, Yang Y, Xue D, Hao C, Zhang W. Regulation of Pancreatic Fibrosis by Acinar Cell-Derived Exosomal miR-130a-3p via Targeting of Stellate Cell PPAR-γ. J Inflamm Res 2021; 14:461-477. [PMID: 33658824 PMCID: PMC7917364 DOI: 10.2147/jir.s299298] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 02/10/2021] [Indexed: 12/20/2022] Open
Abstract
Introduction As endogenous miRNA carriers, exosomes play a role in the pathophysiological processes of various diseases. However, their functions and regulation mechanisms in pancreatic fibrosis remain unclear. Methods In this study, an RNA microarray was used to detect differentially expressed exosomal miR-130a-3p in AR42J cells before and after taurolithocholate (TLC) treatment. mRNA-seq was used to screen differentially expressed genes before and after pancreatic stellate cell (PSC) activation. We used the STRING database to construct a protein-protein interaction (PPI) network for differentially expressed genes, used CytoNCA to analyze the centrality of the PPI network, and identified 10 essential proteins in the biological network. Then, the TargetScan and miRanda databases were used to predict the target genes of miR-130a-3p. The intersections of the target genes and the mRNAs encoding the 10 essential proteins were identified to construct miR-130a-3p/peroxisome proliferator-activated receptor gamma (PPAR-γ) pairs. Fluorescence labeling of exosomes and dynamic tracing showed that exosomes can fuse with the cell membranes of PSCs and transport miR-130a-3p into PSCs. A luciferase reporter gene assay was used to confirm that miR-130a-3p can bind to PPAR-γ to inhibit PPAR-γ expression. In vitro and in vivo functional experiments were performed for gain-of-function studies and loss-of-function studies, respectively. Results The studies showed that acinar cell-derived exosomal miR-130a-3p promotes PSC activation and collagen formation through targeting of stellate cellular PPAR-γ. Knockdown of miR-130a-3p significantly improved pancreatic fibrosis. Notably, miR-130a-3p knockdown reduced serum levels of hyaluronic acid (HA) and β-amylase and increased the C-peptide level to protect endocrine and exocrine pancreatic functions and the function of endothelial cells. Conclusion This study revealed that the exosomal miR-130a-3p/PPAR-γ axis participates in PSC activation and the mechanism of chronic pancreatitis (CP) with fibrosis, thus providing a potential new target for the treatment of chronic pancreatic fibrosis.
Collapse
Affiliation(s)
- Qiang Wang
- Department of General Surgery, Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Hao Wang
- Department of General Surgery, Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Qingxu Jing
- Department of General Surgery, Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Yang Yang
- Department of General Surgery, Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Dongbo Xue
- Department of General Surgery, Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Chenjun Hao
- Department of General Surgery, Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Weihui Zhang
- Department of General Surgery, Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| |
Collapse
|
5
|
Darbey A, Rebourcet D, Curley M, Kilcoyne K, Jeffery N, Reed N, Milne L, Roesl C, Brown P, Smith LB. A comparison of in vivo viral targeting systems identifies adeno-associated virus serotype 9 (AAV9) as an effective vector for genetic manipulation of Leydig cells in adult mice. Andrology 2020; 9:460-473. [PMID: 32996275 DOI: 10.1111/andr.12915] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/01/2020] [Accepted: 09/24/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Despite the increasing popularity of deliverable transgenics, a robust and fully validated method for targeting Leydig cells, capable of delivering long-term transgene expression, is yet to be defined. OBJECTIVES We compared three viral vector systems in terms of their cell targeting specificity, longevity of gene expression and impact on targeted cell types when delivered to the interstitial compartment of the mouse testis. MATERIALS & METHODS We delivered lentiviral, adenoviral and adeno-associated (AAV) viral particles to the interstitial compartment of adult mouse testis. Immunolocalization and stereology were performed to characterize ability of vectors to target and deliver transgenes to Leydig cells. RESULTS Viral vectors utilized in this study were found to specifically target Leydig cells when delivered interstitially. Transgene expression in lentiviral-targeted Leydig cells was detected for 7 days post-injection before Leydig cells underwent apoptosis. Adenoviral-delivered transgene expression was detected for 10 days post-injection with no evidence of targeted cell apoptosis. We found serotype differences in AAV injected testis with AAV serotype 9 targeting a significant proportion of Leydig cells. Targeting efficiency increased to an average of 59.63% (and a maximum of 80%) of Leydig cells with the addition of neuraminidase during injection. In AAV injected testis sections, transgene expression was detectable for up to 50 days post-injection. DISCUSSION & CONCLUSION Lentivirus, Adenovirus and Adeno-Associated virus delivery to the testis resulted in key variances in targeting efficiency of Leydig cells and in longevity of transgene expression, but identified AAV9 + Neuraminidase as an efficient vector system for transgene delivery and long-term expression. Simple viral delivery procedures and the commercial availability of viral vectors suggests AAV9 + Neuraminidase will be of significant utility to researchers investigating the genetics underpinning Leydig cell function and holds promise to inform the development of novel therapeutics for the treatment of male reproductive disorders.
Collapse
Affiliation(s)
- Annalucia Darbey
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK.,School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW, Australia
| | - Diane Rebourcet
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW, Australia
| | - Michael Curley
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - Karen Kilcoyne
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - Nathan Jeffery
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - Natalie Reed
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW, Australia
| | - Laura Milne
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - Cornelia Roesl
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - Pamela Brown
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - Lee B Smith
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK.,School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW, Australia
| |
Collapse
|
6
|
Edick AM, Auclair O, Burgos SA. Role of Grb10 in mTORC1-dependent regulation of insulin signaling and action in human skeletal muscle cells. Am J Physiol Endocrinol Metab 2020; 318:E173-E183. [PMID: 31794259 DOI: 10.1152/ajpendo.00025.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Growth factor receptor-bound protein 10 (Grb10) is an adaptor protein that binds to the insulin receptor, upon which insulin signaling and action are thought to be inhibited. Grb10 is also a substrate for the mechanistic target of rapamycin complex 1 (mTORC1) that mediates its feedback inhibition on phosphatidylinositide 3-kinase (PI3K)/Akt signaling. To characterize the function of Grb10 and its regulation by mTORC1 in human muscle, primary skeletal muscle cells were isolated from healthy lean young men and then induced to differentiate into myotubes. Knockdown of Grb10 enhanced insulin-induced PI3K/Akt signaling and glucose uptake in myotubes, reinforcing the notion underlying its function as a negative regulator of insulin action in human muscle. The increased insulin responsiveness in Grb10-silenced myotubes was associated with a higher abundance of the insulin receptor. Furthermore, insulin and amino acids independently and additively stimulated phosphorylation of Grb10 at Ser476. However, acute inhibition of mTORC1 with rapamycin blocked Grb10 Ser476 phosphorylation and repressed a negative-feedback loop on PI3K/Akt signaling that increased myotube responsiveness to insulin. Chronic rapamycin treatment reduced Grb10 protein abundance in conjunction with increased insulin receptor protein levels. Based on these findings, we propose that mTORC1 controls PI3K/Akt signaling through modulation of insulin receptor abundance by Grb10. These findings have potential implications for obesity-linked insulin resistance, as well as clinical use of mTORC1 inhibitors.
Collapse
Affiliation(s)
- Ashlin M Edick
- Department of Animal Science, McGill University, Sainte-Anne-de-Bellevue, Quebec, Canada
| | - Olivia Auclair
- Department of Animal Science, McGill University, Sainte-Anne-de-Bellevue, Quebec, Canada
| | - Sergio A Burgos
- Department of Animal Science, McGill University, Sainte-Anne-de-Bellevue, Quebec, Canada
- Department of Medicine, McGill University, Montreal, Quebec, Canada
- Metabolic Disorders and Complications Program, Research Institute of McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
7
|
Zhang S, Macias-Garcia A, Ulirsch JC, Velazquez J, Butty VL, Levine SS, Sankaran VG, Chen JJ. HRI coordinates translation necessary for protein homeostasis and mitochondrial function in erythropoiesis. eLife 2019; 8:46976. [PMID: 31033440 PMCID: PMC6533081 DOI: 10.7554/elife.46976] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 04/26/2019] [Indexed: 12/05/2022] Open
Abstract
Iron and heme play central roles in the production of red blood cells, but the underlying mechanisms remain incompletely understood. Heme-regulated eIF2α kinase (HRI) controls translation by phosphorylating eIF2α. Here, we investigate the global impact of iron, heme, and HRI on protein translation in vivo in murine primary erythroblasts using ribosome profiling. We validate the known role of HRI-mediated translational stimulation of integratedstressresponse mRNAs during iron deficiency in vivo. Moreover, we find that the translation of mRNAs encoding cytosolic and mitochondrial ribosomal proteins is substantially repressed by HRI during iron deficiency, causing a decrease in cytosolic and mitochondrial protein synthesis. The absence of HRI during iron deficiency elicits a prominent cytoplasmic unfolded protein response and impairs mitochondrial respiration. Importantly, ATF4 target genes are activated during iron deficiency to maintain mitochondrial function and to enable erythroid differentiation. We further identify GRB10 as a previously unappreciated regulator of terminal erythropoiesis. Red blood cells use a molecule called hemoglobin to transport oxygen around the body. To make hemoglobin, cells require iron to build a component called heme. If an individual does not get enough iron in their diet, the body cannot produce enough red blood cells, or the cells lack hemoglobin. This condition is known as iron deficiency anemia, and it affects around one-third of the world’s population. Researchers did not know exactly how iron levels control red blood cell production, though several proteins had been identified to play important roles. Heme forms in the cell's mitochondria: the compartments in the cell that supply it with energy. When heme levels in a developing red blood cell are low, a protein called HRI reduces the production of many proteins, most importantly the proteins that make up hemoglobin. HRI also boosts the production of a protein called ATF4, which switches on a set of genes that help both the cell and its mitochondria to adapt to the lack of heme. In turn, HRI and ATF4 reduce the activity of a signaling pathway called mTORC1, which controls the production of proteins that help cells to grow and divide. To understand in more detail how iron and heme regulate the production of new red blood cells, Zhang et al. looked at immature red blood cells from the livers of developing mice. Some of the mice lacked the gene that produces HRI, and some experienced iron deficiency. Comparing gene activity in the different mice revealed that in the developing blood cells of iron-deficient mice, HRI largely reduces the rate of protein production in both the mitochondria and the wider cell. At the same time, the increased activity of ATF4 allows the mitochondria to carry on releasing energy and the cells to continue developing. Zhang et al. also found that a protein that inhibits the mTORC1 signaling pathway needs to be active for the new red blood cells to mature. Overall, the results suggest that drugs that activate HRI or block the activity of the mTORC1 pathway could help to treat anemia. The next step is to test the effects that such drugs have in anemic mice and cells from anemic people.
Collapse
Affiliation(s)
- Shuping Zhang
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, United States
| | - Alejandra Macias-Garcia
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, United States
| | - Jacob C Ulirsch
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, United States.,Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, United States.,Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, United States.,Program in Biological and Biomedical Sciences, Harvard University, Cambridge, United States
| | - Jason Velazquez
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, United States
| | - Vincent L Butty
- BioMicro Center, Massachusetts Institute of Technology, Cambridge, United States
| | - Stuart S Levine
- BioMicro Center, Massachusetts Institute of Technology, Cambridge, United States
| | - Vijay G Sankaran
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, United States.,Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, United States.,Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, United States
| | - Jane-Jane Chen
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, United States
| |
Collapse
|
8
|
Guo C, Li Y, Zhang R, Zhang Y, Zhao J, Yao J, Sun J, Dong J, Liao L. Protective Effect of Salidroside Against Diabetic Kidney Disease Through Inhibiting BIM-Mediated Apoptosis of Proximal Renal Tubular Cells in Rats. Front Pharmacol 2018; 9:1433. [PMID: 30564128 PMCID: PMC6289038 DOI: 10.3389/fphar.2018.01433] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 11/19/2018] [Indexed: 12/25/2022] Open
Abstract
Background: Accumulating evidences indicate that the apoptosis of proximal tubular epithelial cells (PTECs) play a vital role in the progression of the diabetic kidney disease (DKD). This study aimed to explore the therapeutic potential of salidroside (SAL) in DKD and its underlying mechanism in anti-apoptosis of PTECs. Methods: Twenty-eight male Wistar rats were allocated into four groups: sham-operated, uninephrectomy (unx), diabetes with uninephrectomy (DKD) and DKD treated with SAL (DKD + SAL). SAL (70 mg/kg) was gavage administered for 8 weeks. 24-h albuminuria and serum creatinine (SCr), blood urea nitrogen (BUN), renal histological changes were examined. The silico analysis was used to identify the main therapeutic targets and pathways of SAL involved in DKD treatment. Apoptosis was determined by TUNEL and Annexin V-FITC/PI double staining in vivo and in vitro, respectively. The expression of BIM, BAX, and cleaved caspase-3 were evaluated by western blot and immunostaining. Results: Treatment with SAL significantly attenuated diabetic kidney injury via inhibiting 24-h albuminuria, SCr, BUN, glomerular mesangial dilatation and tubular injury in DKD rats. The silico analysis identified the intrinsic apoptotic pathway as an important pathway responsible for the nephroprotective properties of SAL. Our data validated that SAL effectively inhibited the apoptosis of PTECs induced by high-glucose (HG), both in vitro and in vivo. Silence of BIM by shRNA in HK-2 cells prevented HG-induced apoptosis. The up-regulated BIM and its downstream targets (BAX and cleaved caspase-3) were also inhibited by SAL. Conclusion: In summary, SAL significantly relieved DKD. And the possible mechanisms might be partially attributed to inhibiting apoptosis of proximal renal tubular cells. The apoptotic protein BIM could be an important target of SAL in this process.
Collapse
Affiliation(s)
- Congcong Guo
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China.,Division of Endocrinology, Department of Internal Medicine, Shandong Provincial QianFoShan Hospital, Shandong University, Jinan, China
| | - Yun Li
- Department of General Health Care II, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Rui Zhang
- Division of Endocrinology, Department of Internal Medicine, Shandong Provincial QianFoShan Hospital, Shandong University, Jinan, China
| | - Yaqin Zhang
- Division of Endocrinology, Department of Internal Medicine, Shandong Provincial QianFoShan Hospital, Shandong University, Jinan, China
| | - Junyu Zhao
- Division of Endocrinology, Department of Internal Medicine, Shandong Provincial QianFoShan Hospital, Shandong University, Jinan, China
| | - Jinming Yao
- Division of Endocrinology, Department of Internal Medicine, Shandong Provincial QianFoShan Hospital, Shandong University, Jinan, China
| | - Jie Sun
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jianjun Dong
- Division of Endocrinology, Department of Internal Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Lin Liao
- Division of Endocrinology, Department of Internal Medicine, Shandong Provincial QianFoShan Hospital, Shandong University, Jinan, China
| |
Collapse
|
9
|
Doiron B, DeFronzo RA. A novel experimental model for human mixed acinar-ductal pancreatic cancer. Carcinogenesis 2018; 39:180-190. [PMID: 29106450 PMCID: PMC5862347 DOI: 10.1093/carcin/bgx119] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 10/27/2017] [Indexed: 12/14/2022] Open
Abstract
Pancreatic cancer has remained refractory to treatment. In large part, this results from the lack of an animal model that mimics pancreatic cancer in man. We describe a novel experimental model of pancreatic cancer that shares the genetic background, histologic features and natural history of human mixed acinar–ductal carcinoma. Adult wild-type mice received an injection into the pancreatic duct of lentivirus coding two molecules, KrasG12D mutation and shRNA p53, which recapitulate the mechanisms of pancreatic cancer in humans. The lentivirus constructs also co-expressed the luciferase gene for in vivo imaging by bioluminescence using the Xenogen IVIS imaging system. Weeks post-injection wild-type mice develop pancreatic cancer with the same histologic characteristics and metastases observed with human pancreatic mixed acinar–ductal carcinoma. This novel approach represents the first pancreatic cancer model that does not involve alterations of embryonic development, which is inherent with transgenic mice or knockout mice models. This novel experimental human pancreatic cancer model can be used to more effectively test new anti-cancer drug to inhibit tumor progression in situ and to retard metastases. Furthermore, our method of injecting lentivirus containing oncogenes and molecules implicated in the development of pancreatic can be employed in diabetic and obese mice, two common metabolic conditions characterized by an increased incidence of pancreatic cancer.
Collapse
Affiliation(s)
- Bruno Doiron
- Diabetes Division, University of Texas Health Science Center at San Antonio, USA
| | - Ralph A DeFronzo
- Diabetes Division, University of Texas Health Science Center at San Antonio, USA
| |
Collapse
|
10
|
Evaluation of vitrification protocol of mouse ovarian tissue by effect of DNA methyltransferase-1 and paternal imprinted growth factor receptor-binding protein 10 on signaling pathways. Cryobiology 2017; 80:89-95. [PMID: 29180273 DOI: 10.1016/j.cryobiol.2017.11.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 10/27/2017] [Accepted: 11/21/2017] [Indexed: 10/18/2022]
Abstract
Transplantation of cryopreserved ovarian tissue has been considered as a promising way of fertility preservation for women. however, this cryopreservation method is prone to post-resuscitation follicle proliferation and oocyte development stagnation, affecting late transplant survival. To evaluate current vitrification works, we investigated the critical pathway alternations in vitrified-warmed juvenile 10-day-old mouse ovary. We showed a significant decrease of protein kinase B (Akt) and Mitogen-activated protein kinase (Mapk) phosphorylation, during which serine/threonine kinases play central roles in coordinating follicle and oocyte development and stress response. Inhibition of Akt and Mapk activity were associated with one of the imprinted insulin pathway negative regulatory genes, Growth factor receptor-binding protein 10 (Grb10) which remarkably increased in vitrified-warmed juvenile mouse ovary than that of fresh group (p < 0.05). RNAi-induced Grb10 down-regulation reversed the decrease in Akt and Mapk phosphorylation. The increase of Grb10 expression was partially caused by the hyper-methylation of the promoter region, associated with the decrease of follicular DNA methyltransferase (Dnmt) 1 protein in different stages of vitrified-warmed group, compared to fresh group (p < 0.05). The mRNA and protein expression of Dnmt1 in ovary of vitrified-warmed juvenile mouse were remarkably lower than those in fresh group (p < 0.05). Dnmt1 overexpression dramatically reversed Grb10 up-regulation and Akt and Mapk phosphorylation reduction. Taken together, our findings suggest that Grb10 expression might be helpful in evaluation of effectiveness of vitrification, and considered as a potential target for further vitrification protocols improvement in the future.
Collapse
|
11
|
Quirin KA, Kwon JJ, Alioufi A, Factora T, Temm CJ, Jacobsen M, Sandusky GE, Shontz K, Chicoine LG, Clark KR, Mendell JT, Korc M, Kota J. Safety and Efficacy of AAV Retrograde Pancreatic Ductal Gene Delivery in Normal and Pancreatic Cancer Mice. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017; 8:8-20. [PMID: 29349096 PMCID: PMC5675991 DOI: 10.1016/j.omtm.2017.09.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 09/27/2017] [Indexed: 02/07/2023]
Abstract
Recombinant adeno-associated virus (rAAV)-mediated gene delivery shows promise to transduce the pancreas, but safety/efficacy in a neoplastic context is not well established. To identify an ideal AAV serotype, route, and vector dose and assess safety, we have investigated the use of three AAV serotypes (6, 8, and 9) expressing GFP in a self-complementary (sc) AAV vector under an EF1α promoter (scAAV.GFP) following systemic or retrograde pancreatic intraductal delivery. Systemic delivery of scAAV9.GFP transduced the pancreas with high efficiency, but gene expression did not exceed >45% with the highest dose, 5 × 1012 viral genomes (vg). Intraductal delivery of 1 × 1011 vg scAAV6.GFP transduced acini, ductal cells, and islet cells with >50%, ∼48%, and >80% efficiency, respectively, and >80% pancreatic transduction was achieved with 5 × 1011 vg. In a KrasG12D-driven pancreatic cancer mouse model, intraductal delivery of scAAV6.GFP targeted acini, epithelial, and stromal cells and exhibited persistent gene expression 5 months post-delivery. In normal mice, intraductal delivery induced a transient increase in serum amylase/lipase that resolved within a day of infusion with no sustained pancreatic inflammation or fibrosis. Similarly, in PDAC mice, intraductal delivery did not increase pancreatic intraepithelial neoplasia progression/fibrosis. Our study demonstrates that scAAV6 targets the pancreas/neoplasm efficiently and safely via retrograde pancreatic intraductal delivery.
Collapse
Affiliation(s)
- Kayla A Quirin
- Department of Medical and Molecular Genetics, Indiana University School of Medicine (IUSM), Indianapolis, IN 46202, USA
| | - Jason J Kwon
- Department of Medical and Molecular Genetics, Indiana University School of Medicine (IUSM), Indianapolis, IN 46202, USA
| | - Arafat Alioufi
- Department of Medical and Molecular Genetics, Indiana University School of Medicine (IUSM), Indianapolis, IN 46202, USA
| | - Tricia Factora
- Department of Medical and Molecular Genetics, Indiana University School of Medicine (IUSM), Indianapolis, IN 46202, USA
| | | | - Max Jacobsen
- Department of Pathology, IUSM, Indianapolis, IN 46202, USA
| | | | - Kim Shontz
- Center for Gene Therapy, Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Louis G Chicoine
- Center for Gene Therapy, Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - K Reed Clark
- Dimension Therapeutics, Cambridge, MA 02139, USA
| | - Joshua T Mendell
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Murray Korc
- The Melvin and Bren Simon Cancer Center, IUSM, Indianapolis, IN 46202, USA.,Pancreatic Cancer Signature Center, Indiana University and Purdue University-Indianapolis (IUPUI), Indianapolis, IN 46202, USA.,Department of Biochemistry and Molecular Biology, IUSM, Indianapolis, IN 43202, USA.,Department of Medicine, IUSM, Indianapolis, IN 43202, USA
| | - Janaiah Kota
- Department of Medical and Molecular Genetics, Indiana University School of Medicine (IUSM), Indianapolis, IN 46202, USA.,The Melvin and Bren Simon Cancer Center, IUSM, Indianapolis, IN 46202, USA.,Pancreatic Cancer Signature Center, Indiana University and Purdue University-Indianapolis (IUPUI), Indianapolis, IN 46202, USA
| |
Collapse
|
12
|
Doiron B, Hu W, DeFronzo RA. Beta Cell Formation in vivo Through Cellular Networking, Integration and Processing (CNIP) in Wild Type Adult Mice. Curr Pharm Biotechnol 2016; 17:376-88. [PMID: 26696016 PMCID: PMC5421132 DOI: 10.2174/1389201017666151223124031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Revised: 11/18/2015] [Accepted: 11/26/2015] [Indexed: 11/22/2022]
Abstract
Insulin replacement therapy is essential in type 1 diabetic individuals and is required in ~40-50% of type 2 diabetics during their lifetime. Prior attempts at beta cell regeneration have relied upon pancreatic injury to induce beta cell proliferation, dedifferentiation and activation of the embryonic pathway, or stem cell replacement. We report an alternative method to transform adult non-stem (somatic) cells into pancreatic beta cells. The Cellular Networking, Integration and Processing (CNIP) approach targets cellular mechanisms involved in pancreatic function in the organ’s adult state and utilizes a synergistic mechanism that integrates three important levels of cellular regulation to induce beta cell formation: (i) glucose metabolism, (ii) membrane receptor function, and (iii) gene transcription. The aim of the present study was to induce pancreatic beta cell formation in vivo in adult animals without stem cells and without dedifferentiating cells to recapitulate the embryonic pathway as previously published (1-3). Our results employing CNIP demonstrate that: (i) insulin secreting cells can be generated in adult pancreatic tissue in vivo and circumvent the problem of generating endocrine (glucagon and somatostatin) cells that exert deleterious effects on glucose homeostasis, and (ii) long-term normalization of glucose tolerance and insulin secretion can be achieved in a wild type diabetic mouse model. The CNIP cocktail has the potential to be used as a preventative or therapeutic treatment or cure for both type 1 and type 2 diabetes.
Collapse
Affiliation(s)
- Bruno Doiron
- Diabetes Division University of Texas Health Science Center 7703 Floyd Curl Drive San Antonio, Texas, 78231.
| | | | | |
Collapse
|
13
|
Lu H, Hao L, Li S, Lin S, Lv L, Chen Y, Cui H, Zi T, Chu X, Na L, Sun C. Elevated circulating stearic acid leads to a major lipotoxic effect on mouse pancreatic beta cells in hyperlipidaemia via a miR-34a-5p-mediated PERK/p53-dependent pathway. Diabetologia 2016; 59:1247-57. [PMID: 26969487 DOI: 10.1007/s00125-016-3900-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 01/25/2016] [Indexed: 01/15/2023]
Abstract
AIMS/HYPOTHESIS Serum stearic acid (C18:0) is elevated in individuals with hyperlipidaemia and type 2 diabetes. However, the lipotoxicity induced by increased stearic acid in beta cells has not been well described. This study aimed to examine the adverse effects of stearic acid on beta cells and the potential mechanisms through which these are mediated. METHODS Three groups of C57BL/6 mice were fed a normal diet or a high-stearic-acid/high-palmitic-acid diet for 24 weeks, respectively. The microRNA (miR) profiles of islets were determined by microarray screening. Islet injury was detected with co-staining using the TUNEL assay and insulin labelling. A lentiviral vector expressing anti-miRNA-34a-5p oligonucleotide (AMO-34a-5p) was injected into mice via an intraductal pancreatic route. RESULTS In both mouse islets and cultured rat insulinoma INS-1 cells, stearic acid exhibited a stronger lipotoxic role than other fatty acids, owing to repression of B cell CLL/lymphoma 2 (BCL-2) and BCL-2-like 2 (BCL-W) by stearic acid stimulation of miR-34a-5p. The stearic-acid-induced lipotoxicity and reduction in insulin secretion were alleviated by AMO-34a-5p. Further investigations in INS-1 cells revealed that p53 was involved in stearic-acid-induced elevation of miR-34a-5p, owing in part to activation of protein kinase-like endoplasmic reticulum kinase (PERK). Conversely, silencing PERK alleviated stearic-acid-induced p53, miR-34a-5p and lipotoxicity. CONCLUSIONS/INTERPRETATION These findings provide new insight for understanding the molecular mechanisms underlying not only the deleterious impact of stearic-acid-induced lipotoxicity but also apoptosis in beta cells and progression to type 2 diabetes.
Collapse
Affiliation(s)
- Huimin Lu
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, Hei Longjiang Province, 150081, People's Republic of China
| | - Liuyi Hao
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, Hei Longjiang Province, 150081, People's Republic of China
| | - Songtao Li
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, Hei Longjiang Province, 150081, People's Republic of China
| | - Song Lin
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, Hei Longjiang Province, 150081, People's Republic of China
| | - Lin Lv
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, Hei Longjiang Province, 150081, People's Republic of China
| | - Yang Chen
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, Hei Longjiang Province, 150081, People's Republic of China
| | - Hongli Cui
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, Hei Longjiang Province, 150081, People's Republic of China
| | - Tianqi Zi
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, Hei Longjiang Province, 150081, People's Republic of China
| | - Xia Chu
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, Hei Longjiang Province, 150081, People's Republic of China
| | - Lixin Na
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, Hei Longjiang Province, 150081, People's Republic of China.
| | - Changhao Sun
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, Hei Longjiang Province, 150081, People's Republic of China.
- Research Institute of Food, Nutrition and Health, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, People's Republic of China.
| |
Collapse
|
14
|
Lyssenko V, Groop L, Prasad RB. Genetics of Type 2 Diabetes: It Matters From Which Parent We Inherit the Risk. Rev Diabet Stud 2016; 12:233-42. [PMID: 27111116 DOI: 10.1900/rds.2015.12.233] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Type 2 diabetes (T2D) results from a co-occurrence of genes and environmental factors. There are more than 120 genetic loci suggested to be associated with T2D, or with glucose and insulin levels in European and multi-ethnic populations. Risk of T2D is higher in the offspring if the mother rather than the father has T2D. Genetically, this can be associated with a unique parent-of-origin (PoO) transmission of risk alleles, and it relates to genetic programming during the intrauterine period, resulting in the inability to increase insulin secretion in response to increased demands imposed by insulin resistance later in life. Such PoO transmission is seen for variants in the KLF14, KCNQ1, GRB10, TCF7L2, THADA, and PEG3 genes. Here we describe T2D susceptibility genes associated with defects in insulin secretion, and thereby risk of overt T2D. This review emphasizes the need to consider distorted parental transmission of risk alleles by exploring the genetic risk of T2D.
Collapse
Affiliation(s)
| | - Leif Groop
- Department of Clinical Sciences, Diabetes and Endocrinology, Clinical Research Centre, Lund University, Malmö, Sweden
| | - Rashmi B Prasad
- Department of Clinical Sciences, Diabetes and Endocrinology, Clinical Research Centre, Lund University, Malmö, Sweden
| |
Collapse
|
15
|
Mukhopadhyay A, Ravikumar G, Dwarkanath P, Meraaj H, Thomas A, Crasta J, Thomas T, Kurpad A, Sridhar T. Placental expression of the insulin receptor binding protein GRB10: Relation to human fetoplacental growth and fetal gender. Placenta 2015; 36:1225-30. [DOI: 10.1016/j.placenta.2015.09.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 08/13/2015] [Accepted: 09/08/2015] [Indexed: 11/27/2022]
|
16
|
Li L, Li X, Zhu Y, Zhang M, Yin D, Lu J, Liu F, Wang C, Jia W. Growth receptor binding protein 10 inhibits glucose-stimulated insulin release from pancreatic β-cells associated with suppression of the insulin/insulin-like growth factor-1 signalling pathway. Clin Exp Pharmacol Physiol 2014; 40:841-7. [PMID: 23937793 DOI: 10.1111/1440-1681.12160] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2013] [Revised: 07/22/2013] [Accepted: 08/08/2013] [Indexed: 01/30/2023]
Abstract
Growth receptor binding protein 10 (Grb10) is an adaptor protein that interacts with the insulin receptor and insulin-like growth factor (IGF)-1 receptor. Overexpression of Grb10 in muscle cells and adipocytes inhibits insulin signalling, and transgenic mice overexpressing Grb10 exhibit impaired glucose tolerance. However, the roles of Grb10 in β-cells remain unknown. The aim of the present study was to explore the effect of Grb10 on β-cell function. The effects of Grb10 on glucose-stimulated insulin secretion (GSIS) and the insulin/IGF-1 signalling pathway were investigated in rat islets and/or dispersed islet cells with Grb10 overexpresion by adenovirus transfection. Protein expression was detected by western blot analysis. We found that Grb10 was expressed in both human and rat pancreas. Expression of Grb10 was increased in islets isolated from rats fed a high-fat plus high-sugar diet compared with islets isolated from rats fed normal chow diet, as well as in INS 832/13 cells exposed to high levels of glucose (20 mmol/L), palmitate (1 mmol/L) and interleukin-1β (50 U/mL). Overexpression of Grb10 in INS 832/13 cells or rat islets impaired GSIS compared with the respective control (all P < 0.05). Moreover, inhibition of GSIS by Grb10 overexpression was associated with a decrease in insulin- and IGF-1-induced Akt and extracellular signal-regulated kinase 1/2 phosphorylation. The results of the present study demonstrate that Grb10 is an important negative regulator of insulin/IGF-1 signalling in pancreatic β-cells and a potential target to improve β-cell function.
Collapse
Affiliation(s)
- Ling Li
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China; Diabetes Institute, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory of Diabetes Mellitus, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Yin DD, You LH, Yuan QX, Liang XD, Wang N, Wang LT, Yuan L, Wang KM, De W. Mesothelin promotes cell proliferation in the remodeling of neonatal rat pancreas. World J Gastroenterol 2014; 20:6884-6896. [PMID: 24944479 PMCID: PMC4051928 DOI: 10.3748/wjg.v20.i22.6884] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 03/21/2014] [Accepted: 04/16/2014] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effect of mesothelin in the remodeling of the endocrine pancreas in neonatal rats.
METHODS: Overexpression or downregulation of mesothelin expression in INS-1 cells was carried out to investigate the effect of mesothelin during cell proliferation and cell apoptosis in vitro. Adenovirus-mediated RNA interference was performed to block mesothelin in vivo to directly assess the role of mesothelin in the remodeling of the endocrine pancreas in neonatal rats.
RESULTS: Exogenous overexpression of mesothelin promoted cell proliferation, cell colony formation and enhanced cell resistance to apoptosis of INS-1 cells. Down-regulation of mesothelin made no difference in cell proliferation and apoptosis compared with that in the control group. After an injection of adenovirus-mesothelin, a significantly increased number of small islets appeared, and the expression of PCNA was decreased on day 7 and day 14 compared with the Ad-EGFP group.
CONCLUSION: Mesothelin was able to promote β cell proliferation in the remodeling stage of neonatal rats. Mesothelin may have an important role in the remodeling of the endocrine pancreas in neonatal rats.
Collapse
|
18
|
Prokopenko I, Poon W, Mägi R, Prasad B R, Salehi SA, Almgren P, Osmark P, Bouatia-Naji N, Wierup N, Fall T, Stančáková A, Barker A, Lagou V, Osmond C, Xie W, Lahti J, Jackson AU, Cheng YC, Liu J, O'Connell JR, Blomstedt PA, Fadista J, Alkayyali S, Dayeh T, Ahlqvist E, Taneera J, Lecoeur C, Kumar A, Hansson O, Hansson K, Voight BF, Kang HM, Levy-Marchal C, Vatin V, Palotie A, Syvänen AC, Mari A, Weedon MN, Loos RJF, Ong KK, Nilsson P, Isomaa B, Tuomi T, Wareham NJ, Stumvoll M, Widen E, Lakka TA, Langenberg C, Tönjes A, Rauramaa R, Kuusisto J, Frayling TM, Froguel P, Walker M, Eriksson JG, Ling C, Kovacs P, Ingelsson E, McCarthy MI, Shuldiner AR, Silver KD, Laakso M, Groop L, Lyssenko V. A central role for GRB10 in regulation of islet function in man. PLoS Genet 2014; 10:e1004235. [PMID: 24699409 PMCID: PMC3974640 DOI: 10.1371/journal.pgen.1004235] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2013] [Accepted: 01/20/2014] [Indexed: 01/03/2023] Open
Abstract
Variants in the growth factor receptor-bound protein 10 (GRB10) gene were in a GWAS meta-analysis associated with reduced glucose-stimulated insulin secretion and increased risk of type 2 diabetes (T2D) if inherited from the father, but inexplicably reduced fasting glucose when inherited from the mother. GRB10 is a negative regulator of insulin signaling and imprinted in a parent-of-origin fashion in different tissues. GRB10 knock-down in human pancreatic islets showed reduced insulin and glucagon secretion, which together with changes in insulin sensitivity may explain the paradoxical reduction of glucose despite a decrease in insulin secretion. Together, these findings suggest that tissue-specific methylation and possibly imprinting of GRB10 can influence glucose metabolism and contribute to T2D pathogenesis. The data also emphasize the need in genetic studies to consider whether risk alleles are inherited from the mother or the father. In this paper, we report the first large genome-wide association study in man for glucose-stimulated insulin secretion (GSIS) indices during an oral glucose tolerance test. We identify seven genetic loci and provide effects on GSIS for all previously reported glycemic traits and obesity genetic loci in a large-scale sample. We observe paradoxical effects of genetic variants in the growth factor receptor-bound protein 10 (GRB10) gene yielding both reduced GSIS and reduced fasting plasma glucose concentrations, specifically showing a parent-of-origin effect of GRB10 on lower fasting plasma glucose and enhanced insulin sensitivity for maternal and elevated glucose and decreased insulin sensitivity for paternal transmissions of the risk allele. We also observe tissue-specific differences in DNA methylation and allelic imbalance in expression of GRB10 in human pancreatic islets. We further disrupt GRB10 by shRNA in human islets, showing reduction of both insulin and glucagon expression and secretion. In conclusion, we provide evidence for complex regulation of GRB10 in human islets. Our data suggest that tissue-specific methylation and imprinting of GRB10 can influence glucose metabolism and contribute to T2D pathogenesis. The data also emphasize the need in genetic studies to consider whether risk alleles are inherited from the mother or the father.
Collapse
Affiliation(s)
- Inga Prokopenko
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom; Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom; Department of Genomics of Common Disease, School of Public Health, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Wenny Poon
- Department of Clinical Science, Diabetes & Endocrinology, Lund University Diabetes Centre, Malmö, Sweden
| | - Reedik Mägi
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom; Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Rashmi Prasad B
- Department of Clinical Science, Diabetes & Endocrinology, Lund University Diabetes Centre, Malmö, Sweden
| | - S Albert Salehi
- Department of Clinical Science, Diabetes & Endocrinology, Lund University Diabetes Centre, Malmö, Sweden
| | - Peter Almgren
- Department of Clinical Science, Diabetes & Endocrinology, Lund University Diabetes Centre, Malmö, Sweden
| | - Peter Osmark
- Department of Clinical Science, Diabetes & Endocrinology, Lund University Diabetes Centre, Malmö, Sweden
| | - Nabila Bouatia-Naji
- University of Lille Nord de France, Lille, France; CNRS UMR8199, Institut Pasteur de Lille, Lille, France; INSERM U970, Paris Cardiovascular Research Center PARCC, Paris, France
| | - Nils Wierup
- Department of Clinical Science, Neuroendocrine Cell Biology, Lund University Diabetes Centre, Malmö, Sweden
| | - Tove Fall
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Alena Stančáková
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Adam Barker
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Vasiliki Lagou
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom; Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Clive Osmond
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, United Kingdom
| | - Weijia Xie
- Peninsula College of Medicine and Dentistry, University of Exeter, Exeter, United Kingdom
| | - Jari Lahti
- Institute of Behavioural Sciences, University of Helsinki, Helsinki, Finland; Folkhälsan Research Centre, Helsinki, Finland
| | - Anne U Jackson
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Yu-Ching Cheng
- Division of Endocrinology Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Jie Liu
- Division of Endocrinology Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Jeffrey R O'Connell
- Division of Endocrinology Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Paul A Blomstedt
- Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland; Department of Mathematics, Åbo Akademi University, Turku, Finland
| | - Joao Fadista
- Department of Clinical Science, Diabetes & Endocrinology, Lund University Diabetes Centre, Malmö, Sweden
| | - Sami Alkayyali
- Department of Clinical Science, Diabetes & Endocrinology, Lund University Diabetes Centre, Malmö, Sweden
| | - Tasnim Dayeh
- Epigenetics and Diabetes Unit, Department of Clinical Sciences, Lund University, CRC, Scania University Hospital, Malmö, Sweden
| | - Emma Ahlqvist
- Department of Clinical Science, Diabetes & Endocrinology, Lund University Diabetes Centre, Malmö, Sweden
| | - Jalal Taneera
- Department of Clinical Science, Diabetes & Endocrinology, Lund University Diabetes Centre, Malmö, Sweden
| | - Cecile Lecoeur
- University of Lille Nord de France, Lille, France; CNRS UMR8199, Institut Pasteur de Lille, Lille, France
| | - Ashish Kumar
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom; Swiss Tropical and Public Health Institute, University of Basel, Basel, Switzerland
| | - Ola Hansson
- Department of Clinical Science, Diabetes & Endocrinology, Lund University Diabetes Centre, Malmö, Sweden
| | - Karin Hansson
- Department of Clinical Science, Diabetes & Endocrinology, Lund University Diabetes Centre, Malmö, Sweden
| | - Benjamin F Voight
- Department of Pharmacology and Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Hyun Min Kang
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Claire Levy-Marchal
- INSERM - Institut de Santé Publique, Paris, France; INSERM CIC EC 05, Hôpital Robert Debré, Paris, France
| | - Vincent Vatin
- University of Lille Nord de France, Lille, France; CNRS UMR8199, Institut Pasteur de Lille, Lille, France
| | - Aarno Palotie
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland; Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, United Kingdom; Department of Medicine, Helsinki University Central Hospital, Helsinki, Finland; Program in Medical and Population Genetics and Genetics Analysis Platform, The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusettes, United States of America
| | - Ann-Christine Syvänen
- Molecular Medicine, Department of Medical Sciences, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Andrea Mari
- CNR Institute of Biomedical Engineering, Padova, Italy
| | - Michael N Weedon
- Peninsula College of Medicine and Dentistry, University of Exeter, Exeter, United Kingdom
| | - Ruth J F Loos
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Ken K Ong
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Peter Nilsson
- Department of Clinical Science, Internal Medicine, Skåne University Hospital Malmö, Malmö, Sweden
| | - Bo Isomaa
- Folkhälsan Research Centre, Helsinki, Finland; Department of Social Service and Health Care, Jakobstad, Finland
| | - Tiinamaija Tuomi
- Folkhälsan Research Centre, Helsinki, Finland; Department of Medicine, Helsinki University Central Hospital, Helsinki, Finland
| | - Nicholas J Wareham
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Michael Stumvoll
- University of Leipzig, Department of Medicine, Leipzig, Germany; University of Leipzig, IFB Adiposity Diseases, Leipzig, Germany
| | - Elisabeth Widen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Timo A Lakka
- Institute of Biomedicine/Physiology, University of Eastern Finland, Kuopio, Finland; Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - Claudia Langenberg
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Anke Tönjes
- University of Leipzig, Department of Medicine, Leipzig, Germany; University of Leipzig, IFB Adiposity Diseases, Leipzig, Germany
| | - Rainer Rauramaa
- Kuopio Research Institute of Exercise Medicine, Kuopio, Finland; Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Johanna Kuusisto
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Timothy M Frayling
- Peninsula College of Medicine and Dentistry, University of Exeter, Exeter, United Kingdom
| | - Philippe Froguel
- Department of Genomics of Common Disease, School of Public Health, Imperial College London, Hammersmith Hospital, London, United Kingdom; University of Lille Nord de France, Lille, France; CNRS UMR8199, Institut Pasteur de Lille, Lille, France
| | - Mark Walker
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Johan G Eriksson
- Folkhälsan Research Centre, Helsinki, Finland; Helsinki University, Department of General Practice and Primary Health Care, Helsinki, Finland; Helsinki University Central Hospital, Unit of General Practice, Helsinki, Finland
| | - Charlotte Ling
- Epigenetics and Diabetes Unit, Department of Clinical Sciences, Lund University, CRC, Scania University Hospital, Malmö, Sweden
| | - Peter Kovacs
- University of Leipzig, Department of Medicine, Leipzig, Germany; University of Leipzig, IFB Adiposity Diseases, Leipzig, Germany
| | - Erik Ingelsson
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom; Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Mark I McCarthy
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom; Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom; Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, United Kindom
| | - Alan R Shuldiner
- Division of Endocrinology Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States of America; Baltimore Geriatric Research, Education and Clinical Center, Baltimore, Maryland, United States of America
| | - Kristi D Silver
- Division of Endocrinology Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States of America; Baltimore Geriatric Research, Education and Clinical Center, Baltimore, Maryland, United States of America
| | - Markku Laakso
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Leif Groop
- Department of Clinical Science, Diabetes & Endocrinology, Lund University Diabetes Centre, Malmö, Sweden; Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Valeriya Lyssenko
- Department of Clinical Science, Diabetes & Endocrinology, Lund University Diabetes Centre, Malmö, Sweden; Steno Diabetes Center A/S, Gentofte, Denmark
| |
Collapse
|
19
|
Pan S, Wan J, Liu S, Zhang S, Xiong H, Zhou J, Xiong W, Yu K, Fu Y. Lentivirus carrying the Atoh1 gene infects normal rat cochlea. Neural Regen Res 2013; 8:1551-9. [PMID: 25206450 PMCID: PMC4145961 DOI: 10.3969/j.issn.1673-5374.2013.17.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 04/24/2013] [Indexed: 12/29/2022] Open
Abstract
Lentivirus carrying the Atoh1 gene can infect Corti's organ and express a hair-like cell surface marker in the supporting cell area. However, expression of the gene carried by adenovirus is instantaneous, which undoubtedly limits its clinical application. Lentivirus acts as a carrier that can stably and continuously express genes. In this study, the cochlear structure and hearing level were not affected, and Atoh1 gene carried by lentivirus promoted the production of hair-like cells in the cochlear supporting cell area. This led to expression of the hair-like cell surface marker myosin 7a 30 days after lentivirus carrying Atoh1 was microinjected into the cochlear round window of rats.
Collapse
Affiliation(s)
- Song Pan
- Department of Otolaryngology, Guangming New District People's Hospital, Shenzhen 518106, Guangdong Province, China
| | - Jingzhi Wan
- Department of Surgery, Hubei University of Science and Technology, Xianning 437100, Hubei Province, China
| | - Shaosheng Liu
- Department of Otolaryngology, Beilun Branch of the First Affiliated Hospital, College of Medicine, Zhejiang University, Ningbo 315800, Zhejiang Province, China
| | - Song Zhang
- Department of Otolaryngology, Guangming New District People's Hospital, Shenzhen 518106, Guangdong Province, China
| | - Hao Xiong
- Department of Otolaryngology, Second Affiliated Hospital, Sun Yat-sen University, Guangzhou 510120, Guangdong Province, China
| | - Jun Zhou
- Department of Otolaryngology, Guangming New District People's Hospital, Shenzhen 518106, Guangdong Province, China
| | - Wu Xiong
- Department of Otolaryngology, Guangming New District People's Hospital, Shenzhen 518106, Guangdong Province, China
| | - Kunfei Yu
- Department of Otolaryngology, Guangming New District People's Hospital, Shenzhen 518106, Guangdong Province, China
| | - Yong Fu
- Department of Otolaryngology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| |
Collapse
|
20
|
Desbuquois B, Carré N, Burnol AF. Regulation of insulin and type 1 insulin-like growth factor signaling and action by the Grb10/14 and SH2B1/B2 adaptor proteins. FEBS J 2013. [PMID: 23190452 DOI: 10.1111/febs.12080] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The effects of insulin and type 1 insulin-like growth factor (IGF-1) on metabolism, growth and survival are mediated by their association with specific receptor tyrosine kinases, which results in both receptor and substrate phosphorylation. Phosphotyrosine residues on receptors and substrates provide docking sites for signaling proteins containing SH2 (Src homology 2) domains, including molecular adaptors. This review focuses on the regulation of insulin/IGF-1 signaling and action by two adaptor families with a similar domain organization: the growth factor receptor-bound proteins Grb7/10/14 and the SH2B proteins. Both Grb10/14 and SH2B1/B2 associate with the activation loop of insulin/IGF-1 receptors through their SH2 domains, but association of Grb10/14 also involves their unique BPS domain. Consistent with Grb14 binding as a pseudosubstrate to the kinase active site, insulin/IGF-induced activation of receptors and downstream signaling pathways in cultured cells is inhibited by Grb10/14 adaptors, but is potentiated by SH2B1/B2 adaptors. Accordingly, Grb10 and Grb14 knockout mice show improved insulin/IGF sensitivity in vivo, and, for Grb10, overgrowth and increased skeketal muscle and pancreatic β-cell mass. Conversely, SH2B1-depleted mice display insulin and IGF-1 resistance, with peripheral depletion leading to reduced adiposity and neuronal depletion leading to obesity through associated leptin resistance. Grb10/14 and SH2B1 adaptors also modulate insulin/IGF-1 action by interacting with signaling components downstream of receptors and exert several tissue-specific effects. The identification of Grb10/14 and SH2B1 as physiological regulators of insulin signaling and action, together with observations that variants at their gene loci are associated with obesity and/or insulin resistance, highlight them as potential therapeutic targets for these conditions.
Collapse
Affiliation(s)
- Bernard Desbuquois
- Institut Cochin, Départment d'Endocrinologie, Métabolisme et Cancer, Université Paris-Descartes, Institut National de la Santé et de la Recherche Médicale, Unité 1016, et Centre National de la Recherche Scientifique, Unité Mixte de Recherche, Paris, France
| | | | | |
Collapse
|
21
|
Ward A. New role for Grb10 signaling in the pancreas. Diabetes 2012; 61:3066-7. [PMID: 23172954 PMCID: PMC3501884 DOI: 10.2337/db12-1044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Andrew Ward
- Department of Biology and Biochemistry and Centre for Regenerative Medicine, University of Bath, Bath, U.K.
| |
Collapse
|