1
|
Wu Y, Ma J, Chen J, Liu X, Wang Z, Luo L, Sun C. Ablation of CD44 Attenuates Adipogenesis in White Adipocytes via the Tryptophan 5-Hydroxylase 2/5-Hydroxytryptamine Axis to Protect Mice from High-Fat Diet-Induced Obesity. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:247-264. [PMID: 39476955 DOI: 10.1016/j.ajpath.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 10/09/2024] [Accepted: 10/16/2024] [Indexed: 11/11/2024]
Abstract
CD44 is a transmembrane protein that plays an essential role in transducing extracellular stimuli into intracellular signaling cascades, especially in cancer cells. Recent studies have shown that CD44 contributes to metabolic regulation. However, the effect of CD44 on adipogenesis in white adipose tissue (WAT) remains unclear. Herein, the expression of CD44 was largely increased in the inguinal and epididymal WAT of obese mice. Ablation or neutralization of CD44 inhibited adipogenesis in cultured adipocytes. CD44-deficient mice were resistant to high-fat diet-induced obesity and metabolic dysfunction. RNA-sequencing, together with functional studies, revealed that reduced expression of tryptophan 5-hydroxylase 2 (Tph2) in WAT was responsible for the repressed adipogenesis in the absence of CD44. The application of 5-hydroxytryptamine, a product of TPH2, rescued the repressed adipogenesis induced by CD44 neutralization. Moreover, the inhibition of TPH2 by p-chlorophenylalanine recapitulated the beneficial phenotypes observed in CD44-deficient mice. Taken together, these data indicate that CD44 plays a pivotal role in adipogenesis in WAT. In this regard, CD44 and its downstream target TPH2 may hold great therapeutic potential for treating excessive adiposity-related metabolic disorders, such as obesity, insulin resistance, and type 2 diabetes.
Collapse
Affiliation(s)
- Yuting Wu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Jinyu Ma
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Jing Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Xiaoyu Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Zhe Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Lan Luo
- Department of Geriatrics, Affiliated Hospital of Nantong University, Nantong, China.
| | - Cheng Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China.
| |
Collapse
|
2
|
Nagy N, Czepiel KS, Kaber G, Stefanovski D, Hargil A, Pennetzdorfer N, Targ R, Reghupaty SC, Wight TN, Vernon RB, Hull-Meichle RL, Marshall P, Medina CO, Martinez H, Kalinowski A, Paladini RD, Garantziotis S, Knowles JW, Bollyky PL. Hymecromone Promotes Longevity and Insulin Sensitivity in Mice. Cells 2024; 13:1727. [PMID: 39451245 PMCID: PMC11506560 DOI: 10.3390/cells13201727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 09/12/2024] [Accepted: 09/27/2024] [Indexed: 10/26/2024] Open
Abstract
Given that the extracellular matrix polymer hyaluronan (HA) has been implicated in longevity, we asked whether 4-methylumbelliferone (4-MU), an inhibitor of HA synthesis, impacts lifespan in mice. We designed a prospective study of long-term administration of 4-MU with conventional C57BL/6J mice. We find that 4-MU extends median survival from 122 weeks (control) to 154 weeks (4-MU), an increase of 32 weeks (p < 0.0001 by Log-rank Mantel Cox test). The maximum lifespan of 4-MU treated mice increased from 159 to 194 weeks. In tandem with these effects, 4-MU enhances insulin sensitivity, a metabolic parameter known to regulate lifespan, as measured by insulin tolerance testing (ITT) as well as frequent sampling intra venous glucose tolerance tests (FSIVGTTs). We further observed that 4-MU treated mice weigh less while consuming the same amount of food, indicating that 4-MU treatment alters energy expenditure. However, we do not observe changes in tissue HA content in this model. We conclude that 4-MU promotes insulin sensitivity and longevity but that the underlying mechanism, and the contribution of HA is unclear. 4-MU, already approved in various countries for hepatobiliary conditions, is currently under investigation and clinical development as a therapy for several chronic inflammatory conditions. These data suggest that the beneficial effects of 4-MU on tissue metabolism may include effects on longevity.
Collapse
Affiliation(s)
- Nadine Nagy
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA 94305, USA; (N.N.); (K.S.C.); (A.H.)
| | - Kathryn S. Czepiel
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA 94305, USA; (N.N.); (K.S.C.); (A.H.)
| | - Gernot Kaber
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA 94305, USA; (N.N.); (K.S.C.); (A.H.)
| | - Darko Stefanovski
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Kennett Square, PA 19348, USA;
| | - Aviv Hargil
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA 94305, USA; (N.N.); (K.S.C.); (A.H.)
| | - Nina Pennetzdorfer
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA 94305, USA; (N.N.); (K.S.C.); (A.H.)
| | - Robert Targ
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA 94305, USA; (N.N.); (K.S.C.); (A.H.)
| | - Saranya C. Reghupaty
- Department of Pathology, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Thomas N. Wight
- Benaroya Research Institute, 1201 9th Ave, Seattle, WA 98101, USA (R.B.V.)
| | - Robert B. Vernon
- Benaroya Research Institute, 1201 9th Ave, Seattle, WA 98101, USA (R.B.V.)
| | - Rebecca L. Hull-Meichle
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, VA Puget Sound Health Care System and University of Washington, Seattle, WA 98108, USA;
| | - Payton Marshall
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA 94305, USA; (N.N.); (K.S.C.); (A.H.)
| | - Carlos O. Medina
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA 94305, USA; (N.N.); (K.S.C.); (A.H.)
| | - Hunter Martinez
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA 94305, USA; (N.N.); (K.S.C.); (A.H.)
| | - Anissa Kalinowski
- Halo Biosciences, 125 University St., Palo Alto, CA 94301, USA (R.D.P.)
| | | | - Stavros Garantziotis
- Immunity, Inflammation and Disease Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC 27709, USA;
| | - Joshua W. Knowles
- Cardiovascular Medicine and Cardiovascular Institute, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Paul L. Bollyky
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA 94305, USA; (N.N.); (K.S.C.); (A.H.)
| |
Collapse
|
3
|
Gao M, Wang H, Li N, Qiao Y, Liu H, Li W, Wang S, Li J, Yu Z, Hu G, Leng J, Yang X. Serum CD44 levels in early pregnancy and its genetic variants for increased risk of gestational diabetes mellitus in Chinese pregnant women. J Proteomics 2024; 307:105268. [PMID: 39097228 DOI: 10.1016/j.jprot.2024.105268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 08/05/2024]
Abstract
This study aimed to explore associations of serum cluster of differentiation 44 (CD44) levels and its genetic variants in early pregnancy with gestational diabetes mellitus (GDM). We conducted a 1:1 case-control study (n = 414) nested in a prospective cohort of 22,302 pregnant women recruited from 2010 to 2012 in Tianjin, China. Blood samples were collected at the first antenatal care visit (at a median of 10th gestational week). Binary conditional logistic regressions were performed to examine associations of serum CD44 levels and its genetic variants with increased risk of GDM. In this study, we found that serum CD44 levels in early pregnancy was associated with GDM risk in a U-shaped manner. High serum CD44 levels and its genetic risk score in early pregnancy were associated with markedly increased risk of GDM after adjustment for traditional confounders (OR: 1.95, 95%CI: 1.12-3.40 & 1.95, 1.05-3.61). Furthermore, after adjustment for serum CD44 levels, the OR of CD44 genetic risk score for GDM was slightly attenuated but not significant (1.84, 0.98-3.48). In conclusion, serum CD44 levels and its genetic variants in early pregnancy were associated with GDM risk in Chinese pregnant women, with the effect of CD44 genetic variants being accounted for by serum CD44. SIGNIFICANCE: Recent studies suggested that pregnant women with GDM may have abnormal levels of CD44 and abnormal expression of CD44 gene, but it is uncertain whether abnormal CD44 plays a causal role in occurrence of GDM. Specifically, it remains unknown whether serum CD44 levels in early pregnancy and its genetic variants can predict the later occurrence of GDM. In this study, we found that high serum CD44 levels in early pregnancy and its genetic variants were associated with markedly increased risk of GDM in Chinese pregnant women, with the effect of CD44 genetic variants being largely accounted for by serum CD44 levels. Our study is the first reporting that serum CD44 levels and its genetic variants were associated with markedly increased risk of GDM. These multi-omics risk markers may be useful for identification of women at high risk of GDM in early pregnancy. Our findings also provide new insights into the disease mechanisms.
Collapse
Affiliation(s)
- Ming Gao
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin 300070, China
| | - Hui Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin 300070, China; Tianjin Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China
| | - Ninghua Li
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin 300070, China
| | - Yijuan Qiao
- Department of Children's Health, Tianjin Women and Children's Health Center, Tianjin 300070, China
| | - Hongyan Liu
- Department of Children's Health, Tianjin Women and Children's Health Center, Tianjin 300070, China
| | - Weiqin Li
- Department of Children's Health, Tianjin Women and Children's Health Center, Tianjin 300070, China
| | - Shuting Wang
- Department of Children's Health, Tianjin Women and Children's Health Center, Tianjin 300070, China
| | - Jing Li
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin 300070, China; Tianjin Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China
| | - Zhijie Yu
- Population Cancer Research Program and Department of Pediatrics, Dalhousie University, Halifax 15000, Canada
| | - Gang Hu
- Chronic Disease Epidemiology Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana 70808, USA
| | - Junhong Leng
- Department of Children's Health, Tianjin Women and Children's Health Center, Tianjin 300070, China.
| | - Xilin Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin 300070, China; Tianjin Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China.
| |
Collapse
|
4
|
Weng X, Jiang H, Walker DJ, Zhou H, Lin D, Wang J, Kang L. Deletion of CD44 promotes adipogenesis by regulating PPARγ and cell cycle-related pathways. J Endocrinol 2024; 262:e240079. [PMID: 38692289 PMCID: PMC11227036 DOI: 10.1530/joe-24-0079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 05/01/2024] [Indexed: 05/03/2024]
Abstract
CD44, a cell surface adhesion receptor and stem cell biomarker, is recently implicated in chronic metabolic diseases. Ablation of CD44 ameliorates adipose tissue inflammation and insulin resistance in obesity. Here, we investigated cell type-specific CD44 expression in human and mouse adipose tissue and further studied how CD44 in preadipocytes regulates adipocyte function. Using Crispr Cas9-mdediated gene deletion and lentivirus-mediated gene re-expression, we discovered that deletion of CD44 promotes adipocyte differentiation and adipogenesis, whereas re-expression of CD44 abolishes this effect and decreases insulin responsiveness and adiponectin secretion in 3T3-L1 cells. Mechanistically, CD44 does so via suppressing Pparg expression. Using quantitative proteomics analysis, we further discovered that cell cycle-regulated pathways were mostly decreased by deletion of CD44. Indeed, re-expression of CD44 moderately restored expression of proteins involved in all phases of the cell cycle. These data were further supported by increased preadipocyte proliferation rates in CD44-deficient cells and re-expression of CD44 diminished this effect. Our data suggest that CD44 plays a crucial role in regulating adipogenesis and adipocyte function possibly through regulating PPARγ and cell cycle-related pathways. This study provides evidence for the first time that CD44 expressed in preadipocytes plays key roles in regulating adipocyte function outside immune cells where CD44 is primarily expressed. Therefore, targeting CD44 in (pre)adipocytes may provide therapeutic potential to treat obesity-associated metabolic complications.
Collapse
Affiliation(s)
- Xiong Weng
- Division of Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee, Scotland, UK
| | - Hao Jiang
- Gene Expression and Regulation, School of Life Sciences, University of Dundee, Dundee, Scotland, UK
| | - David J Walker
- Division of Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee, Scotland, UK
| | - Houjiang Zhou
- MRC Protein Phosphorylation Unit, School of Life Sciences, Dundee, Scotland, UK
| | - De Lin
- Drug Discovery Unit, School of Life Sciences, University of Dundee, Dundee, Scotland, UK
| | - Jing Wang
- Science for Life Laboratory, Department of Biomedical and Clinical Sciences (BKV), Linköping University, Linköping, Sweden
| | - Li Kang
- Division of Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee, Scotland, UK
| |
Collapse
|
5
|
Zhang Z, Sun G, Wang Y, Wang N, Lu Y, Chen Y, Xia F. Integrated Bioinformatics Analysis Revealed Immune Checkpoint Genes Relevant to Type 2 Diabetes. Diabetes Metab Syndr Obes 2024; 17:2385-2401. [PMID: 38881696 PMCID: PMC11179640 DOI: 10.2147/dmso.s458030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 05/30/2024] [Indexed: 06/18/2024] Open
Abstract
Objective Chronic low-grade inflammation of the pancreatic islets is the characteristic of type 2 diabetes (T2D), and some of the immune checkpoints may play important roles in the pancreatic islet inflammation. Thus, we aim to explore the immune checkpoint genes (ICGs) associated with T2D, thereby revealing the role of ICGs in the pathogenesis of T2D based on bioinformatic analyses. Methods Differentially expressed genes (DEGs) and immune checkpoint genes (ICGs) of islets between T2D and control group were screened from datasets of the Gene Expression Omnibus (GEO). A risk model was built based on the coefficients of ICGs calculated by ridge regression. Functional enrichment analysis and immune cell infiltration estimation were conducted. Correlations between ICGs and hub genes, T2D-related disease genes, insulin secretion genes, and beta cell function-related genes were analyzed. Finally, we conducted RT-PCR to verify the expression of these ICGs. Results In total, pancreatic islets from 19 cases of T2D and 84 healthy subjects were included. We identified 458 DEGs. Six significantly upregulated ICGs (CD44, CD47, HAVCR2, SIRPA, TNFSF9, and VTCN1) in T2D were screened out. These ICGs were significantly correlated with several hub genes and T2D-related genes; furthermore, they were correlated with insulin secretion and β cell function-related genes. The analysis of immune infiltration showed that the concentrations of eosinophils, T cells CD4 naive, and T cells regulatory (Tregs) were significantly higher, but CD4 memory resting T cells and monocytes were lower in islets of T2D patients. The infiltrated immune cells in T2D pancreatic islet were associated with these six ICGs. Finally, the expression levels of four ICGs were confirmed by RT-PCR, and three ICGs were validated in another independent dataset. Conclusion In conclusion, the identified ICGs may play an important role in T2D. Identification of these differential genes may provide new clues for the diagnosis and treatment of T2D.
Collapse
Affiliation(s)
- Ziteng Zhang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Guoting Sun
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yuying Wang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Ningjian Wang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yingli Lu
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yi Chen
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Fangzhen Xia
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
6
|
Han J, Lee C, Jung Y. Current Evidence and Perspectives of Cluster of Differentiation 44 in the Liver's Physiology and Pathology. Int J Mol Sci 2024; 25:4749. [PMID: 38731968 PMCID: PMC11084344 DOI: 10.3390/ijms25094749] [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: 03/24/2024] [Revised: 04/21/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Cluster of differentiation 44 (CD44), a multi-functional cell surface receptor, has several variants and is ubiquitously expressed in various cells and tissues. CD44 is well known for its function in cell adhesion and is also involved in diverse cellular responses, such as proliferation, migration, differentiation, and activation. To date, CD44 has been extensively studied in the field of cancer biology and has been proposed as a marker for cancer stem cells. Recently, growing evidence suggests that CD44 is also relevant in non-cancer diseases. In liver disease, it has been shown that CD44 expression is significantly elevated and associated with pathogenesis by impacting cellular responses, such as metabolism, proliferation, differentiation, and activation, in different cells. However, the mechanisms underlying CD44's function in liver diseases other than liver cancer are still poorly understood. Hence, to help to expand our knowledge of the role of CD44 in liver disease and highlight the need for further research, this review provides evidence of CD44's effects on liver physiology and its involvement in the pathogenesis of liver disease, excluding cancer. In addition, we discuss the potential role of CD44 as a key regulator of cell physiology.
Collapse
Affiliation(s)
- Jinsol Han
- Department of Integrated Biological Science, College of Natural Science, Pusan National University, Pusan 46241, Republic of Korea;
| | - Chanbin Lee
- Institute of Systems Biology, College of Natural Science, Pusan National University, Pusan 46241, Republic of Korea;
| | - Youngmi Jung
- Department of Integrated Biological Science, College of Natural Science, Pusan National University, Pusan 46241, Republic of Korea;
- Department of Biological Sciences, College of Natural Science, Pusan National University, Pusan 46241, Republic of Korea
| |
Collapse
|
7
|
Xu J, Xia Q, Wu T, Shao Y, Wang Y, Jin N, Tian P, Wu L, Lu X. Prophylactic treatment with Bacteroides uniformis and Bifidobacterium bifidum counteracts hepatic NK cell immune tolerance in nonalcoholic steatohepatitis induced by high fat diet. Gut Microbes 2024; 16:2302065. [PMID: 38196273 PMCID: PMC10793665 DOI: 10.1080/19490976.2024.2302065] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 01/02/2024] [Indexed: 01/11/2024] Open
Abstract
Hepatic immunity is one of the driving forces for the development of nonalcoholic steatohepatitis (NASH), and targeting gut microbiota is believed to affect the hepatic immune constitution. Here, we aimed to investigate the hepatic immunological state in NASH, with a specific emphasis on natural killer (NK) cells. In addition, we aimed to identify the contributing species that target hepatic immunity to provide new directions and support the feasibility of immunotherapy for NASH. A possible NASH population was determined by combination of long-term severe fatty liver, metabolic disorders and increased serum CK18 to detect serum immune factors and gut microbiota. NASH was induced in mice fed a high-fat diet to verify the prophylactic effect of the functional species on the immunopathology and development of NASH. Hepatic immunologic state was examined, and the effector functions of NK cells were detected. Hepatic transcriptome, proteomic, and fecal metagenome were performed. We observed a statistical increase in serum IL-10 (p < 0.001) and non-statistical decrease in interferon-γ and IL-6 in NASH population, hinting at the possibility of immune tolerance. Fecal Bacteroides uniformis and Bifidobacterium bifidum were abundant in healthy population but depleted in NASH patients. In NASH mice, hepatic CD8+T cells, macrophages, and dendritic cells were increased (p < 0.01), and NK cells were inhibited, which were identified with decreased granzyme B (p < 0.05). Bacteroides uniformis and Bifidobacterium bifidum improved hepatic pathological and metabolic cues, increased hepatic NK cells and reduced macrophages (p < 0.05). Bacteroides uniformis also restored hepatic NK cell function, which was identified as increased CD107a (p < 0.05). Transcriptional and translational profiling revealed that the functional species might restore the function of hepatic NK cells through multiple pathways, such as reduction of inhibitory molecules in NK cells. Bacteroides uniformis and Bifidobacterium bifidum are novel prophylactics for NASH that restore the impaired function of hepatic NK cells.
Collapse
Affiliation(s)
- Jingyuan Xu
- Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
- Department of Gastroenterology, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Department of Gastroenterology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Qiaoyun Xia
- Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Ting Wu
- Department of Citizen Health, Community Health Service Center of Jinxi Town, Kunshan, China
| | - Yong Shao
- Department of Citizen Health, Community Health Service Center of Jinxi Town, Kunshan, China
| | - Yatao Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Nuyun Jin
- Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Peiying Tian
- Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Longyun Wu
- Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Xiaolan Lu
- Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| |
Collapse
|
8
|
Wang J, Casimiro-Garcia A, Johnson BG, Duffen J, Cain M, Savary L, Wang S, Nambiar P, Lech M, Zhao S, Xi L, Zhan Y, Olson J, Stejskal JA, Lin H, Zhang B, Martinez RV, Masek-Hammerman K, Schlerman FJ, Dower K. A protein kinase C α and β inhibitor blunts hyperphagia to halt renal function decline and reduces adiposity in a rat model of obesity-driven type 2 diabetes. Sci Rep 2023; 13:16919. [PMID: 37805649 PMCID: PMC10560236 DOI: 10.1038/s41598-023-43759-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 09/28/2023] [Indexed: 10/09/2023] Open
Abstract
Type 2 diabetes (T2D) and its complications can have debilitating, sometimes fatal consequences for afflicted individuals. The disease can be difficult to control, and therapeutic strategies to prevent T2D-induced tissue and organ damage are needed. Here we describe the results of administering a potent and selective inhibitor of Protein Kinase C (PKC) family members PKCα and PKCβ, Cmpd 1, in the ZSF1 obese rat model of hyperphagia-induced, obesity-driven T2D. Although our initial intent was to evaluate the effect of PKCα/β inhibition on renal damage in this model setting, Cmpd 1 unexpectedly caused a marked reduction in the hyperphagic response of ZSF1 obese animals. This halted renal function decline but did so indirectly and indistinguishably from a pair feeding comparator group. However, above and beyond this food intake effect, Cmpd 1 lowered overall animal body weights, reduced liver vacuolation, and reduced inguinal adipose tissue (iWAT) mass, inflammation, and adipocyte size. Taken together, Cmpd 1 had strong effects on multiple disease parameters in this obesity-driven rodent model of T2D. Further evaluation for potential translation of PKCα/β inhibition to T2D and obesity in humans is warranted.
Collapse
Affiliation(s)
- Ju Wang
- Inflammation and Immunology, Pfizer Worldwide Research and Development, Cambridge, MA, USA.
| | | | - Bryce G Johnson
- Inflammation and Immunology, Pfizer Worldwide Research and Development, Cambridge, MA, USA
| | - Jennifer Duffen
- Inflammation and Immunology, Pfizer Worldwide Research and Development, Cambridge, MA, USA
| | - Michael Cain
- Inflammation and Immunology, Pfizer Worldwide Research and Development, Cambridge, MA, USA
- Mediar Therapeutics, Boston, MA, USA
| | - Leigh Savary
- Inflammation and Immunology, Pfizer Worldwide Research and Development, Cambridge, MA, USA
- Instem Life Science Systems Ltd, Mount Ida College, South Hadley, MA, USA
| | - Stephen Wang
- Pharmacokinetics and Drug Metabolism, Pfizer Worldwide Research and Development, Cambridge, MA, USA
- Novartis Gene Therapies, Novartis Institute for Biomedical Research, Cambridge, MA, USA
| | - Prashant Nambiar
- Drug Safety Research and Development, Pfizer Worldwide Research and Development, Cambridge, MA, USA
- Strand Therapeutics, Cambridge, MA, USA
| | - Matthew Lech
- Inflammation and Immunology, Pfizer Worldwide Research and Development, Cambridge, MA, USA
| | - Shanrong Zhao
- Clinical Genetics and Bioinformatics, Pfizer Worldwide Research and Development, Cambridge, MA, USA
- Amunix Pharmaceuticals, San Francisco, CA, USA
| | - Li Xi
- Early Clinical Development, Pfizer Worldwide Research and Development, Cambridge, MA, USA
| | - Yutian Zhan
- Drug Safety Research and Development, Pfizer Worldwide Research and Development, Cambridge, MA, USA
| | - Jennifer Olson
- Drug Safety Research and Development, Pfizer Worldwide Research and Development, Groton, CT, USA
| | - James A Stejskal
- Drug Safety Research and Development, Pfizer Worldwide Research and Development, Groton, CT, USA
- Charles River Laboratories, Shrewsbury, MA, USA
| | - Hank Lin
- Drug Safety Research and Development, Pfizer Worldwide Research and Development, Cambridge, MA, USA
- Sunovion Pharmaceuticals Inc., Marlborough, MA, USA
| | - Baohong Zhang
- Clinical Genetics and Bioinformatics, Pfizer Worldwide Research and Development, Cambridge, MA, USA
- Data Sciences, Biogen, Cambridge, MA, USA
| | - Robert V Martinez
- Inflammation and Immunology, Pfizer Worldwide Research and Development, Cambridge, MA, USA
- Center for Technological Innovation, Pfizer Worldwide Research and Development, San Francisco, CA, USA
| | | | - Franklin J Schlerman
- Inflammation and Immunology, Pfizer Worldwide Research and Development, Cambridge, MA, USA
| | - Ken Dower
- Inflammation and Immunology, Pfizer Worldwide Research and Development, Cambridge, MA, USA.
| |
Collapse
|
9
|
Wang S, Lu Y, Chi T, Zhang Y, Zhao Y, Guo H, Feng L. Identification of ferroptosis-related genes in type 2 diabetes mellitus based on machine learning. Immun Inflamm Dis 2023; 11:e1036. [PMID: 37904700 PMCID: PMC10566453 DOI: 10.1002/iid3.1036] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 09/05/2023] [Accepted: 09/17/2023] [Indexed: 11/01/2023] Open
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM), which has a high incidence and several harmful consequences, poses a severe danger to human health. Research on the function of ferroptosis in T2DM is increasing. This study uses bioinformatics techniques identify new diagnostic T2DM biomarkers associated with ferroptosis. METHODS To identify ferroptosis-related genes (FRGs) that are differentially expressed between T2DM patients and healthy individuals, we first obtained T2DM sequencing data and FRGs from the Gene Expression Omnibus (GEO) database and FerrDb database. Then, drug-gene interaction networks and competitive endogenous RNA (ceRNA) networks linked to the marker genes were built after marker genes were filtered by two machine learning algorithms (LASSO and SVM-RFE algorithms). Finally, to confirm the expression of marker genes, the GSE76895 dataset was utilized. The protein and RNA expression of some marker genes in T2DM and nondiabetic tissues was also examined by Western blotting, immunohistochemistry (IHC), immunofluorescence (IF) and quantitative real-time PCR (qRT-PCR). RESULTS We obtained 58 differentially expressed genes (DEGs) associated with ferroptosis. GO and KEGG enrichment analyses showed that these DEGs were significantly enriched in hypoxia and ferroptosis. Subsequently, eight marker genes (SCD, CD44, HIF1A, BCAT2, MTF1, HILPDA, NR1D2, and MYCN) were screened by LASSO and SVM-RFE machine learning algorithms, and a model was constructed based on these eight genes. This model also has high diagnostic power. In addition, based on these eight genes, we obtained 48 drugs and constructed a complex ceRNA network map. Finally, Western blotting, IHC, IF, and qRT-PCR results of clinical samples further confirmed the results of public databases. CONCLUSIONS The diagnosis and aetiology of T2DM can be greatly aided by eight FRGs, providing novel therapeutic avenues.
Collapse
Affiliation(s)
- Sen Wang
- Department of Medical Ultrasound, Shandong Medicine and Health Key Laboratory of Abdominal Medical Imaging, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qian Foshan HospitalShandong First Medical UniversityJinanShandongChina
| | - Yongpan Lu
- Department of Plastic Surgery, The First Clinical Medical College, Shandong University of Traditional Chinese MedicineThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qian Foshan HospitalJinanShandongChina
| | - Tingting Chi
- Department of Acupuncture and RehabilitationThe Affiliated Qingdao Hai Ci Hospital of Qingdao University (West Hospital Area)QingdaoShandongChina
| | - Yixin Zhang
- Department of Medical Ultrasound, Shandong Medicine and Health Key Laboratory of Abdominal Medical Imaging, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qian Foshan HospitalShandong First Medical UniversityJinanShandongChina
| | - Yuli Zhao
- Department of Medical Ultrasound, Shandong Medicine and Health Key Laboratory of Abdominal Medical Imaging, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qian Foshan HospitalShandong First Medical UniversityJinanShandongChina
| | - Huimin Guo
- Department of Medical Ultrasound, Shandong Medicine and Health Key Laboratory of Abdominal Medical Imaging, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qian Foshan HospitalShandong First Medical UniversityJinanShandongChina
| | - Li Feng
- Department of Medical Ultrasound, Shandong Medicine and Health Key Laboratory of Abdominal Medical Imaging, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qian Foshan HospitalShandong First Medical UniversityJinanShandongChina
| |
Collapse
|
10
|
Musale V, Wasserman DH, Kang L. Extracellular matrix remodelling in obesity and metabolic disorders. LIFE METABOLISM 2023; 2:load021. [PMID: 37383542 PMCID: PMC10299575 DOI: 10.1093/lifemeta/load021] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 05/07/2023] [Accepted: 05/17/2023] [Indexed: 06/30/2023]
Abstract
Obesity causes extracellular matrix (ECM) remodelling which can develop into serious pathology and fibrosis, having metabolic effects in insulin-sensitive tissues. The ECM components may be increased in response to overnutrition. This review will focus on specific obesity-associated molecular and pathophysiological mechanisms of ECM remodelling and the impact of specific interactions on tissue metabolism. In obesity, complex network of signalling molecules such as cytokines and growth factors have been implicated in fibrosis. Increased ECM deposition contributes to the pathogenesis of insulin resistance at least in part through activation of cell surface integrin receptors and CD44 signalling cascades. These cell surface receptors transmit signals to the cell adhesome which orchestrates an intracellular response that adapts to the extracellular environment. Matrix proteins, glycoproteins, and polysaccharides interact through ligand-specific cell surface receptors that interact with the cytosolic adhesion proteins to elicit specific actions. Cell adhesion proteins may have catalytic activity or serve as scaffolds. The vast number of cell surface receptors and the complexity of the cell adhesome have made study of their roles challenging in health and disease. Further complicating the role of ECM-cell receptor interactions is the variation between cell types. This review will focus on recent insights gained from studies of two highly conserved, ubiquitously axes and how they contribute to insulin resistance and metabolic dysfunction in obesity. These are the collagen-integrin receptor-IPP (ILK-PINCH-Parvin) axis and the hyaluronan-CD44 interaction. We speculate that targeting ECM components or their receptor-mediated cell signalling may provide novel insights into the treatment of obesity-associated cardiometabolic complications.
Collapse
Affiliation(s)
- Vishal Musale
- Division of Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, United Kingdom
| | - David H Wasserman
- Department of Molecular Physiology and Biophysics, Mouse Metabolic Phenotyping Center, Vanderbilt University, Nashville, TN 37235, United States
| | - Li Kang
- Division of Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, United Kingdom
| |
Collapse
|
11
|
Sarkar K, Bank S, Chatterjee A, Dutta K, Das A, Chakraborty S, Paul N, Sarkar J, De S, Ghosh S, Acharyya K, Chattopadhyay D, Das M. Hyaluronic acid-graphene oxide quantum dots nanoconjugate as dual purpose drug delivery and therapeutic agent in meta-inflammation. J Nanobiotechnology 2023; 21:246. [PMID: 37528408 PMCID: PMC10394801 DOI: 10.1186/s12951-023-02015-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 07/21/2023] [Indexed: 08/03/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) predominantly considered a metabolic disease is now being considered an inflammatory disease as well due to the involvement of meta-inflammation. Obesity-induced adipose tissue inflammation (ATI) is one of the earliest phenomena in the case of meta-inflammation, leading to the advent of insulin resistance (IR) and T2DM. The key events of ATI are orchestrated by macrophages, which aggravate the inflammatory state in the tissue upon activation, ultimately leading to systemic chronic low-grade inflammation and Non-Alcoholic Steatohepatitis (NASH) through the involvement of proinflammatory cytokines. The CD44 receptor on macrophages is overexpressed in ATI, NASH, and IR. Therefore, we developed a CD44 targeted Hyaluronic Acid functionalized Graphene Oxide Quantum Dots (GOQD-HA) nanocomposite for tissue-specific delivery of metformin. Metformin-loaded GOQD-HA (GOQD-HA-Met) successfully downregulated the expression of proinflammatory cytokines and restored antioxidant status at lower doses than free metformin in both palmitic acid-induced RAW264.7 cells and diet induced obese mice. Our study revealed that the GOQD-HA nanocarrier enhanced the efficacy of Metformin primarily by acting as a therapeutic agent apart from being a drug delivery platform. The therapeutic properties of GOQD-HA stem from both HA and GOQD having anti-inflammatory and antioxidant properties respectively. This study unravels the function of GOQD-HA as a targeted drug delivery option for metformin in meta-inflammation where the nanocarrier itself acts as a therapeutic agent.
Collapse
Affiliation(s)
- Kunal Sarkar
- Department of Zoology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, 700019, India
| | - Sarbashri Bank
- Department of Zoology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, 700019, India
| | - Arindam Chatterjee
- Department of Zoology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, 700019, India
| | - Koushik Dutta
- Department of Polymer Science and Technology, University of Calcutta, 92 A.P.C. Road, Kolkata, 700009, India
| | - Anwesha Das
- Department of Zoology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, 700019, India
| | - Santanu Chakraborty
- Department of Zoology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, 700019, India
| | - Nirvika Paul
- Department of Zoology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, 700019, India
| | - Jit Sarkar
- Molecular and Applied Mycology and Plant Pathology Laboratory, Department of Botany, University of Calcutta, Kolkata, 700019, India
| | - Sriparna De
- Department of Allied Health Sciences, Brainware University, Kolkata, 700129, India
| | - Sudakshina Ghosh
- Department of Zoology, Vidyasagar College for Women, Kolkata, 700006, India
| | - Krishnendu Acharyya
- Molecular and Applied Mycology and Plant Pathology Laboratory, Department of Botany, University of Calcutta, Kolkata, 700019, India
| | - Dipankar Chattopadhyay
- Department of Polymer Science and Technology, University of Calcutta, 92 A.P.C. Road, Kolkata, 700009, India
| | - Madhusudan Das
- Department of Zoology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, 700019, India.
| |
Collapse
|
12
|
Nagy N, Kaber G, Sunkari VG, Marshall PL, Hargil A, Kuipers HF, Ishak HD, Bogdani M, Hull RL, Grandoch M, Fischer JW, McLaughlin TL, Wight TN, Bollyky PL. Inhibition of hyaluronan synthesis prevents β-cell loss in obesity-associated type 2 diabetes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.28.530522. [PMID: 36909502 PMCID: PMC10002695 DOI: 10.1101/2023.02.28.530522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
Pancreatic β-cell dysfunction and death are central to the pathogenesis of type 2 diabetes (T2D). We have identified a novel role for the inflammatory extracellular matrix polymer hyaluronan (HA) in this pathophysiology. Low levels of HA are present in healthy pancreatic islets. However, HA substantially accumulates in cadaveric islets of human T2D and islets of the db/db mouse model of T2D in response to hyperglycemia. Treatment with 4-methylumbelliferone (4-MU), an inhibitor of HA synthesis, or the deletion of the major HA receptor CD44, preserve glycemic control and insulin levels in db/db mice despite ongoing weight gain, indicating a critical role for this pathway in T2D pathogenesis. 4-MU treatment and the deletion of CD44 likewise preserve glycemic control in other settings of β-cell injury including streptozotocin treatment and islet transplantation. Mechanistically, we find that 4-MU increases the expression of the apoptosis inhibitor survivin, a downstream transcriptional target of CD44 dependent on HA/CD44 signaling, on β-cells such that caspase 3 activation does not result in β-cell apoptosis. These data indicate a role for HA accumulation in diabetes pathogenesis and suggest that it may be a viable target to ameliorate β-cell loss in T2D. These data are particularly exciting, because 4-MU is already an approved drug (also known as hymecromone), which could accelerate translation of these findings to clinical studies.
Collapse
|
13
|
Kohlhepp MS, Liu H, Tacke F, Guillot A. The contradictory roles of macrophages in non-alcoholic fatty liver disease and primary liver cancer-Challenges and opportunities. Front Mol Biosci 2023; 10:1129831. [PMID: 36845555 PMCID: PMC9950415 DOI: 10.3389/fmolb.2023.1129831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 01/31/2023] [Indexed: 02/12/2023] Open
Abstract
Chronic liver diseases from varying etiologies generally lead to liver fibrosis and cirrhosis. Among them, non-alcoholic fatty liver disease (NAFLD) affects roughly one-quarter of the world population, thus representing a major and increasing public health burden. Chronic hepatocyte injury, inflammation (non-alcoholic steatohepatitis, NASH) and liver fibrosis are recognized soils for primary liver cancer, particularly hepatocellular carcinoma (HCC), being the third most common cause for cancer-related deaths worldwide. Despite recent advances in liver disease understanding, therapeutic options on pre-malignant and malignant stages remain limited. Thus, there is an urgent need to identify targetable liver disease-driving mechanisms for the development of novel therapeutics. Monocytes and macrophages comprise a central, yet versatile component of the inflammatory response, fueling chronic liver disease initiation and progression. Recent proteomic and transcriptomic studies performed at singular cell levels revealed a previously overlooked diversity of macrophage subpopulations and functions. Indeed, liver macrophages that encompass liver resident macrophages (also named Kupffer cells) and monocyte-derived macrophages, can acquire a variety of phenotypes depending on microenvironmental cues, and thus exert manifold and sometimes contradictory functions. Those functions range from modulating and exacerbating tissue inflammation to promoting and exaggerating tissue repair mechanisms (i.e., parenchymal regeneration, cancer cell proliferation, angiogenesis, fibrosis). Due to these central functions, liver macrophages represent an attractive target for the treatment of liver diseases. In this review, we discuss the multifaceted and contrary roles of macrophages in chronic liver diseases, with a particular focus on NAFLD/NASH and HCC. Moreover, we discuss potential therapeutic approaches targeting liver macrophages.
Collapse
|
14
|
Differential effects of temperature and mTOR and Wnt-planar cell polarity pathways on syndecan-4 and CD44 expression in growth-selected turkey satellite cell populations. PLoS One 2023; 18:e0281350. [PMID: 36735684 PMCID: PMC9897570 DOI: 10.1371/journal.pone.0281350] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 01/23/2023] [Indexed: 02/04/2023] Open
Abstract
Satellite cells (SCs) comprise a heterogeneous population of muscle stem cells. Thermal stress during the first week after hatch alters proliferation, myogenesis, and adipogenesis of SCs of turkey pectoralis major (p. major) muscle via mechanistic target of rapamycin (mTOR) and wingless-type mouse mammary tumor virus integration site family/planar cell polarity (Wnt/PCP) pathways. Pivotal genes in mTOR and Wnt/PCP pathways are mTOR and frizzled-7 (Fzd7), respectively. The objective of this study was to determine the differential effects of thermal stress on SDC4 and CD44 expression in turkey p. major muscle SCs and how the expression of SDC4 and CD44 is modulated by the mTOR and Wnt/PCP pathways. Satellite cells were isolated from the p. major muscle of 1-week-old faster-growing modern-commercial (NC) turkeys and slower-growing historic Randombred Control Line 2 (RBC2) turkeys, and were challenged with hot (43°C) and cold (33°C) thermal stress for 72 h of proliferation followed by 48 h of differentiation. The NC line SCs were found to contain a lower proportion of SDC4 positive and CD44 negative (SDC4+CD44-) cells and a greater proportion of SDC4 negative and CD44 positive (SDC4-CD44+) cells compared to the RBC2 line at the control temperature (38°C) at both 72 h of proliferation and 48 h of differentiation. In general, at 72 h of proliferation, the proportion of SDC4+CD44- cells decreased with heat stress (43°C) and increased with cold stress (33°C) relative to the control temperature (38°C) in both lines, whereas the proportion of SDC4-CD44+ cells increased with heat stress and decreased with cold stress. In general, the expression of SDC4 and CD44 in the NC SCs showed greater response to both hot and cold thermal stress compared to the RBC2 cells. Knockdown of mTOR or Fzd7 expression increased the proportion of SDC4+CD44- cells while the proportion of SDC4-CD44+ cells decreased during differentiation with line differences being specific to treatment temperatures. Thus, differential composition of p. major muscle SCs in growth-selected commercial turkey may be resulted, in part, from the alteration in SDC4 and CD44 expression. Results indicate differential temperature sensitivity and mTOR and Wnt/PCP pathway responses of growth-selected SC populations and this may have long-lasting effect on muscle development and growth.
Collapse
|
15
|
Drygalski K, Lecoutre S, Clément K, Dugail I. Hyaluronan in Adipose Tissue, Metabolic Inflammation, and Diabetes: Innocent Bystander or Guilty Party? Diabetes 2023; 72:159-169. [PMID: 36668999 DOI: 10.2337/db22-0676] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 11/03/2022] [Indexed: 01/21/2023]
Abstract
Hyaluronic acid, or hyaluronan (HA), is a nonsulfated glucosaminoglycan that has long been recognized for its hydrophilic properties and is widely used as a dermal filler. Despite much attention given to the study of other extracellular matrix (ECM) components, in the field of ECM properties and their contribution to tissue fibroinflammation, little is known of HA's potential role in the extracellular milieu. However, recent studies suggest that it is involved in inflammatory response, diet-induced insulin resistance, adipogenesis, and autoimmunity in type 1 diabetes. Based on its unique physical property as a regulator of osmotic pressure, we emphasize underestimated implications in adipose tissue function, adipogenesis, and obesity-related dysfunction.
Collapse
Affiliation(s)
- Krzysztof Drygalski
- Nutrition and Obesities: Systemic Approaches Research Group, NutriOmics, Sorbonne Université, INSERM, Paris, France
- Clinical Research Center, Medical University of Bialystok, Bialystok, Poland
| | - Simon Lecoutre
- Nutrition and Obesities: Systemic Approaches Research Group, NutriOmics, Sorbonne Université, INSERM, Paris, France
| | - Karine Clément
- Nutrition and Obesities: Systemic Approaches Research Group, NutriOmics, Sorbonne Université, INSERM, Paris, France
- Nutrition Department, Assistance Publique Hôpitaux de Paris, Centre de Recherche en Nutrition Humaine Ile-de-France, Pitié-Salpêtrière Hospital, Paris, France
| | - Isabelle Dugail
- Nutrition and Obesities: Systemic Approaches Research Group, NutriOmics, Sorbonne Université, INSERM, Paris, France
| |
Collapse
|
16
|
Wiering L, Tacke F. Treating inflammation to combat non-alcoholic fatty liver disease. J Endocrinol 2023; 256:JOE-22-0194. [PMID: 36259984 DOI: 10.1530/joe-22-0194] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 10/19/2022] [Indexed: 11/05/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) with its more progressive form non-alcoholic steatohepatitis (NASH) has become the most common chronic liver disease, thereby representing a great burden for patients and healthcare systems. Specific pharmacological therapies for NAFLD are still missing. Inflammation is an important driver in the pathogenesis of NASH, and the mechanisms underlying inflammation in NAFLD represent possible therapeutic targets. In NASH, various intra- and extrahepatic triggers involved in the metabolic injury typically lead to the activation of different immune cells. This includes hepatic Kupffer cells, i.e. liver-resident macrophages, which can adopt an inflammatory phenotype and activate other immune cells by releasing inflammatory cytokines. As inflammation progresses, Kupffer cells are increasingly replaced by monocyte-derived macrophages with a distinct lipid-associated and scar-associated phenotype. Many other immune cells, including neutrophils, T lymphocytes - such as auto-aggressive cytotoxic as well as regulatory T cells - and innate lymphoid cells balance the progression and regression of inflammation and subsequent fibrosis. The detailed understanding of inflammatory cell subsets and their activation pathways prompted preclinical and clinical exploration of potential targets in NAFLD/NASH. These approaches to target inflammation in NASH include inhibition of immune cell recruitment via chemokine receptors (e.g. cenicriviroc), neutralization of CD44 or galectin-3 as well as agonism to nuclear factors like peroxisome proliferator-activated receptors and farnesoid X receptor that interfere with the activation of immune cells. As some of these approaches did not demonstrate convincing efficacy as monotherapies, a rational and personalized combination of therapeutic interventions may be needed for the near future.
Collapse
Affiliation(s)
- Leke Wiering
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Junior Clinician Scientist Program, Berlin, Germany
| | - Frank Tacke
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| |
Collapse
|
17
|
Jia X, Toda K, He L, Miao D, Yamada S, Yu L, Kodama K. Expression-based Genome-wide Association Study Links OPN and IL1-RA With Newly Diagnosed Type 1 Diabetes in Children. J Clin Endocrinol Metab 2022; 107:1825-1832. [PMID: 35460250 PMCID: PMC9391606 DOI: 10.1210/clinem/dgac256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Indexed: 11/19/2022]
Abstract
CONTEXT Islet autoantibodies (IAbs) are currently the most reliable indicators of islet autoimmunity. However, IAbs do not fully meet the need for the prediction and intervention of type 1 diabetes (T1D). Serological proteins should be great sources for biomarkers. OBJECTIVE This work aimed to identify new proteomic biomarkers with the technology of an expression-based genome-wide association study (eGWAS) in children newly diagnosed with T1D. METHODS In an attempt to identify additional biomarkers, we performed an eGWAS using microarray data from 169 arrays of the pancreatic islets of T1D rodents (78 T1D cases and 91 controls). We ranked all 16 099 protein-coding genes by the likelihood of differential expression in the pancreatic islets. Our top 20 secreted proteins were screened in 170 children including 100 newly diagnosed T1D, and 50 type 2 diabetes (T2D) and 20 age-matched healthy children. With 6 proteins showing significance, we further conducted a validation study using the second independent set of 400 samples from children including 200 newly diagnosed with T1D, 100 T2D, and 100 age-matched controls. RESULTS We identified 2 serum proteins that were significantly changed in T1D vs both control and T2D, and 5 serum proteins were significantly changed both in T1D and T2D vs control. Serum osteopontin (OPN) levels were uniquely higher in T1D (T1D vs controls, P = 1.29E-13 ~ 9.38E-7, T1D vs T2D, P = 2.65E-8 ~ 1.58E-7) with no difference between T2D and healthy control individuals. Serum interleukin 1 receptor antagonist (IL-1RA) levels were lower in T1D compared both with T2D (P = 3.36E-9~0.0236) and healthy participants (P = 1.09E-79 ~ 2.00E-12). CONCLUSION Our results suggest that OPN and IL1-RA could be candidates for useful biomarkers for T1D in children.
Collapse
Affiliation(s)
- Xiaofan Jia
- Barbara Davis Center for Diabetes, University of Colorado Denver, Aurora, Colorado 80045, USA
| | - Kyoko Toda
- Biomedical Research Center, Kitasato Institute Hospital, Kitasato University, Tokyo 108-8642, Japan
| | - Ling He
- Barbara Davis Center for Diabetes, University of Colorado Denver, Aurora, Colorado 80045, USA
| | - Dongmei Miao
- Barbara Davis Center for Diabetes, University of Colorado Denver, Aurora, Colorado 80045, USA
| | - Satoru Yamada
- Diabetes Center, Kitasato Institute Hospital, Kitasato University, Tokyo 108-8642, Japan
| | - Liping Yu
- Liping Yu, MD, Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, 1775 Aurora Ct, Aurora, CO 80045, USA.
| | - Keiichi Kodama
- Correspondence: Keiichi Kodama, MD, Health Promotion Team, ORIX Group Health Insurance Society, ORIX Corp, 2-4-1 Hamamatsuchou, Minato-ku, Tokyo 105-6135, Japan.
| |
Collapse
|
18
|
Weng X, Maxwell-Warburton S, Hasib A, Ma L, Kang L. The membrane receptor CD44: novel insights into metabolism. Trends Endocrinol Metab 2022; 33:318-332. [PMID: 35249813 DOI: 10.1016/j.tem.2022.02.002] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/04/2022] [Accepted: 02/07/2022] [Indexed: 12/17/2022]
Abstract
CD44, a cell-surface glycoprotein, has long been studied as a cancer molecule due to its essential role in physiological activities in normal cells and pathological activities in cancer cells, such as cell proliferation, adhesion, and migration; angiogenesis; inflammation; and cytoskeleton rearrangement. Yet, recent evidence suggests a role of CD44 in metabolism, especially insulin resistance in obesity and diabetes. In line with the current concept of fibroinflammation in obesity and insulin resistance, CD44 as the main receptor of the extracellular matrix component, hyaluronan (HA), has been shown to regulate diet-induced insulin resistance in muscle and other insulin-sensitive tissues. In this review, we integrate current evidence for a role of CD44 in regulating glucose and lipid homeostasis and speculate about its involvement in the pathogenesis of chronic metabolic diseases, including obesity and diabetes. We summarize the current development of CD44-targeted therapies and discuss its potential for the use in treating metabolic diseases.
Collapse
Affiliation(s)
- Xiong Weng
- Division of Systems Medicine, School of Medicine, University of Dundee, Dundee, UK
| | | | - Annie Hasib
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Lifeng Ma
- School of Medicine, Xizang Minzhu University, Xianyang, Shaanxi, China
| | - Li Kang
- Division of Systems Medicine, School of Medicine, University of Dundee, Dundee, UK.
| |
Collapse
|
19
|
VerHague M, Albright J, Barron K, Kim M, Bennett BJ. Obesogenic and diabetic effects of CD44 in mice are sexually dimorphic and dependent on genetic background. Biol Sex Differ 2022; 13:14. [PMID: 35410390 PMCID: PMC8996418 DOI: 10.1186/s13293-022-00426-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 03/29/2022] [Indexed: 11/30/2022] Open
Abstract
Introduction CD44 is a candidate gene for obesity and diabetes development and may be a critical mediator of a systemic inflammation associated with obesity and diabetes. Methods We investigated the relationship of CD44 with obesity in CD44-deficient mice challenged with a high-fat diet. Results In mice fed a diet high in fat, cholesterol, and sucrose for 12 weeks fat mass accumulation was reduced in CD44-deficient mice bred onto both a C57BL/6J and the naturally TLR deficient C3H/HeJ background. Reduced fat mass could not be attributed to lower food intake or an increase in energy expenditure as measured by indirect calorimetry. However, we observed a 40–60% lower mRNA expression of the inflammation markers, F4/80, CD11b, TNF-α, and CD14, in adipose tissue of CD44-deficient mice on the C57BL/6J background but not the C3H/HeJ background, perhaps indicating that alternative factors may be affecting adiposity in this model. Measures of hepatic steatosis and insulin sensitivity were improved in CD44-deficient mice on a C57BL/6J but not in the C3H/HeJ mice. These results were highly sexually dimorphic as there were no detectable effects of CD44 inactivation in female mice on a C57BL/6 J or C3H/HeJ background. Conclusion CD44 was associated with adiposity, liver fat, and glucose in male mice. However, the effects of CD44 on obesity may be independent of TLR4 signaling. Supplementary Information The online version contains supplementary material available at 10.1186/s13293-022-00426-2.
Collapse
Affiliation(s)
- Melissa VerHague
- Nutrition Research Institute, University of North Carolina Kannapolis, Kannapolis, NC, 28081, USA
| | - Jody Albright
- Nutrition Research Institute, University of North Carolina Kannapolis, Kannapolis, NC, 28081, USA
| | - Keri Barron
- Nutrition Research Institute, University of North Carolina Kannapolis, Kannapolis, NC, 28081, USA
| | - Myungsuk Kim
- Department of Nutrition, University of California, Davis, 95616, USA.,Korea Institute of Science and Technology (KIST), Gangneung, Gangwon-do, Republic of Korea
| | - Brian J Bennett
- Obesity and Metabolism Research Unit USDA, ARS Western Human Nutrition Research Center, 430 W Health Sciences Drive, Davis, CA, 95616, USA. .,Department of Nutrition, University of California, Davis, 95616, USA.
| |
Collapse
|
20
|
Shu J, Li N, Wei W, Zhang L. Detection of molecular signatures and pathways shared by Alzheimer's disease and type 2 diabetes. Gene 2022; 810:146070. [PMID: 34813915 DOI: 10.1016/j.gene.2021.146070] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 10/21/2021] [Accepted: 11/16/2021] [Indexed: 01/12/2023]
Abstract
Alzheimer's disease (AD) and type 2 diabetes (T2D) are common in the general elderly population, conferring heavy individual, social, and economic stresses on families and society. Accumulating evidence indicates T2D to be a risk factor for AD. However, the underlying mechanisms for this association are largely unknown. This study aimed to identify the shared molecular signatures between AD and T2D through integrated analysis of temporal cortex gene expression data. Gene Ontology (GO) and pathway enrichment analysis, protein over-representation analysis, protein-protein interaction, DEG-transcription factor interactions, DEG-microRNA interactions, protein-drug interactions, gene-disease association analysis, and protein subcellular localization analysis of the common DEGs were performed. We identified 16 common DEGs between the two datasets, which were mainly enriched in the biological processes of apoptosis, autophagy, inflammation, and hemostasis. We also identified five hub proteins encoded by the DEGs, five central regulatory transcription factors, and six microRNAs. Protein-drug interaction analysis showed C1QB to be associated with different drugs. Gene-disease association analysis revealed that hub genes, SFN and ITGB2, were actively engaged in other diseases. Collectively, these findings provide new insights into shared molecular mechanisms between AD and T2D and provide novel candidate targets for therapeutic intervention.
Collapse
Affiliation(s)
- Jun Shu
- Department of Neurology, Cognitive Disorders Center, Huadong Hospital, Fudan University, No. 221, West Yan An Road, Shanghai, China
| | - Nan Li
- Department of Neurology, Cognitive Disorders Center, Huadong Hospital, Fudan University, No. 221, West Yan An Road, Shanghai, China
| | - Wenshi Wei
- Department of Neurology, Cognitive Disorders Center, Huadong Hospital, Fudan University, No. 221, West Yan An Road, Shanghai, China.
| | - Li Zhang
- Department of Neurology, Cognitive Disorders Center, Huadong Hospital, Fudan University, No. 221, West Yan An Road, Shanghai, China.
| |
Collapse
|
21
|
Ugwoke CK, Cvetko E, Umek N. Skeletal Muscle Microvascular Dysfunction in Obesity-Related Insulin Resistance: Pathophysiological Mechanisms and Therapeutic Perspectives. Int J Mol Sci 2022; 23:ijms23020847. [PMID: 35055038 PMCID: PMC8778410 DOI: 10.3390/ijms23020847] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/09/2022] [Accepted: 01/11/2022] [Indexed: 02/04/2023] Open
Abstract
Obesity is a worrisomely escalating public health problem globally and one of the leading causes of morbidity and mortality from noncommunicable disease. The epidemiological link between obesity and a broad spectrum of cardiometabolic disorders has been well documented; however, the underlying pathophysiological mechanisms are only partially understood, and effective treatment options remain scarce. Given its critical role in glucose metabolism, skeletal muscle has increasingly become a focus of attention in understanding the mechanisms of impaired insulin function in obesity and the associated metabolic sequelae. We examined the current evidence on the relationship between microvascular dysfunction and insulin resistance in obesity. A growing body of evidence suggest an intimate and reciprocal relationship between skeletal muscle microvascular and glucometabolic physiology. The obesity phenotype is characterized by structural and functional changes in the skeletal muscle microcirculation which contribute to insulin dysfunction and disturbed glucose homeostasis. Several interconnected etiologic molecular mechanisms have been suggested, including endothelial dysfunction by several factors, extracellular matrix remodelling, and induction of oxidative stress and the immunoinflammatory phenotype. We further correlated currently available pharmacological agents that have deductive therapeutic relevance to the explored pathophysiological mechanisms, highlighting a potential clinical perspective in obesity treatment.
Collapse
|
22
|
Prabata A, Ikeda K, Rahardini EP, Hirata KI, Emoto N. GPNMB plays a protective role against obesity-related metabolic disorders by reducing macrophage inflammatory capacity. J Biol Chem 2021; 297:101232. [PMID: 34582891 PMCID: PMC8524194 DOI: 10.1016/j.jbc.2021.101232] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 09/14/2021] [Accepted: 09/21/2021] [Indexed: 01/30/2023] Open
Abstract
Obesity is a global health problem that is often related to cardiovascular and metabolic diseases. Chronic low-grade inflammation in white adipose tissue (WAT) is a hallmark of obesity. Previously, during a search for differentially expressed genes in WAT of obese mice, we identified glycoprotein nonmetastatic melanoma protein B (GPNMB), of which expression was robustly induced in pathologically expanded WAT. Here, we investigated the role of GPNMB in obesity-related metabolic disorders utilizing GPNMB-deficient mice. When fed a high-fat diet (HFD), GPNMB-deficient mice showed body weight and adiposity similar to those of wild-type (WT) mice. Nonetheless, insulin and glucose tolerance tests revealed significant obesity-related metabolic disorders in GPNMB-KO mice compared with WT mice fed with HFD. Chronic WAT inflammation was remarkably worsened in HFD-fed GPNMB-KO mice, accompanied by a striking increase in crown-like structures, typical hallmarks for diseased WAT. Macrophages isolated from GPNMB-KO mice were observed to produce more inflammatory cytokines than those of WT mice, a difference abolished by supplementation with recombinant soluble GPNMB extracellular domain. We demonstrated that GPNMB reduced the inflammatory capacity of macrophages by inhibiting NF-κB signaling largely through binding to CD44. Finally, we showed that macrophage depletion by addition of clodronate liposomes abolished the worsened WAT inflammation and abrogated the exacerbation of metabolic disorders in GPNMB-deficient mice fed on HFD. Our data reveal that GPNMB negatively regulates macrophage inflammatory capacities and ameliorates the WAT inflammation in obesity; therefore we conclude that GPNMB is a promising therapeutic target for the treatment of metabolic disorders associated with obesity.
Collapse
Affiliation(s)
- Adam Prabata
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, Kobe, Japan; Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Koji Ikeda
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, Kobe, Japan; Department of Epidemiology for Longevity and Regional Health, Kyoto Prefectural University of Medicine, Kyoto, Japan.
| | - Elda Putri Rahardini
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, Kobe, Japan
| | - Ken-Ichi Hirata
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Noriaki Emoto
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, Kobe, Japan; Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan. ,
| |
Collapse
|
23
|
Furihata T, Maekawa S, Takada S, Kakutani N, Nambu H, Shirakawa R, Yokota T, Kinugawa S. Premedication with pioglitazone prevents doxorubicin-induced left ventricular dysfunction in mice. BMC Pharmacol Toxicol 2021; 22:27. [PMID: 33962676 PMCID: PMC8103594 DOI: 10.1186/s40360-021-00495-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 04/26/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Doxorubicin (DOX) is widely used as an effective chemotherapeutic agent for cancers; however, DOX induces cardiac toxicity, called DOX-induced cardiomyopathy. Although DOX-induced cardiomyopathy is known to be associated with a high cumulative dose of DOX, the mechanisms of its long-term effects have not been completely elucidated. Pioglitazone (Pio) is presently contraindicated in patients with symptomatic heart failure owing to the side effects. The concept of drug repositioning led us to hypothesize the potential effects of Pio as a premedication before DOX treatment, and to analyze this hypothesis in mice. METHODS First, for the hyperacute (day 1) and acute (day 7) DOX-induced dysfunction models, mice were fed a standard diet with or without 0.02% (wt/wt) Pio for 5 days before DOX treatment (15 mg/kg body weight [BW] via intraperitoneal [i.p.] administration). The following 3 treatment groups were analyzed: standard diet + vehicle (Vehicle), standard diet + DOX (DOX), and Pio + DOX. Next, for the chronic model (day 35), the mice were administrated DOX once a week for 5 weeks (5 mg/kg BW/week, i.p.). RESULTS In the acute phase after DOX treatment, the percent fractional shortening of the left ventricle (LV) was significantly decreased in DOX mice. This cardiac malfunction was improved in Pio + DOX mice. In the chronic phase, we observed that LV function was preserved in Pio + DOX mice. CONCLUSIONS Our findings may provide a new pathophysiological explanation by which Pio plays a role in the treatment of DOX-induced cardiomyopathy, but the molecular links between Pio and DOX-induced LV dysfunction remain largely elusive.
Collapse
Affiliation(s)
- Takaaki Furihata
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo, 060-8638, Japan.
| | - Satoshi Maekawa
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Shingo Takada
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo, 060-8638, Japan
- Faculty of Lifelong Sport, Department of Sports Education, Hokusho University, Ebetsu, 069-8511, Japan
| | - Naoya Kakutani
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo, 060-8638, Japan
- Research Fellow of the Japan Society for the Promotion of Science, Tokyo, Japan
| | - Hideo Nambu
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Ryosuke Shirakawa
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Takashi Yokota
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Shintaro Kinugawa
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo, 060-8638, Japan
| |
Collapse
|
24
|
Wang XK, Peng ZG. Targeting Liver Sinusoidal Endothelial Cells: An Attractive Therapeutic Strategy to Control Inflammation in Nonalcoholic Fatty Liver Disease. Front Pharmacol 2021; 12:655557. [PMID: 33935770 PMCID: PMC8082362 DOI: 10.3389/fphar.2021.655557] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 03/10/2021] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), especially its advanced stage nonalcoholic steatohepatitis (NASH), has become a threatened public health problem worldwide. However, no specific drug has been approved for clinical use to treat patients with NASH, though there are many promising candidates against NAFLD in the drug development pipeline. Recently, accumulated evidence showed that liver sinusoidal endothelial cells (LSECs) play an essential role in the occurrence and development of liver inflammation in patients with NAFLD. LSECs, as highly specialized endothelial cells with unique structure and anatomical location, contribute to the maintenance of liver homeostasis and could be a promising therapeutic target to control liver inflammation of NAFLD. In this review, we outline the pathophysiological roles of LSECs related to inflammation of NAFLD, highlight the pro-inflammatory and anti-inflammatory effects of LSECs, and discuss the potential drug development strategies against NAFLD based on targeting to LSECs.
Collapse
Affiliation(s)
- Xue-Kai Wang
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zong-Gen Peng
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Biotechnology of Antibiotics, National Health and Family Planning Commission, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
25
|
Anti-inflammatory activity of CD44 antibodies in murine immune thrombocytopenia is mediated by Fcγ receptor inhibition. Blood 2021; 137:2114-2124. [PMID: 33662988 DOI: 10.1182/blood.2020009497] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 02/11/2021] [Indexed: 02/03/2023] Open
Abstract
Monoclonal immunoglobulin G (IgG) antibodies to CD44 (anti-CD44) are anti-inflammatory in numerous murine autoimmune models, but the mechanisms are poorly understood. Anti-CD44 anti-inflammatory activity shows complete therapeutic concordance with IV immunoglobulin (IVIg) in treating autoimmune disease models, making anti-CD44 a potential IVIg alternative. In murine immune thrombocytopenia (ITP), there is no mechanistic explanation for anti-CD44 activity, although anti-CD44 ameliorates disease similarly to IVIg. Here, we demonstrate a novel anti-inflammatory mechanism of anti-CD44 that explains disease amelioration by anti-CD44 in murine ITP. Macrophages treated with anti-CD44 in vitro had dramatically suppressed phagocytosis through FcγRs in 2 separate systems of IgG-opsonized platelets and erythrocytes. Phagocytosis inhibition by anti-CD44 was mediated by blockade of the FcγR IgG binding site without changing surface FcγR expression. Anti-CD44 of different subclasses revealed that FcγR blockade was specific to receptors that could be engaged by the respective anti-CD44 subclass, and Fc-deactivated anti-CD44 variants lost all FcγR-inhibiting activity. In vivo, anti-CD44 functioned analogously in the murine passive ITP model and protected mice from ITP when thrombocytopenia was induced through an FcγR that could be engaged by the CD44 antibody's subclass. Consistent with FcγR blockade, Fc-deactivated variants of anti-CD44 were completely unable to ameliorate ITP. Together, anti-CD44 inhibits macrophage FcγR function and ameliorates ITP consistent with an FcγR blockade mechanism. Anti-CD44 is a potential IVIg alternative and may be of particular benefit in ITP because of the significant role that FcγRs play in human ITP pathophysiology.
Collapse
|
26
|
Queiroz AL, Lessard SJ, Ouchida AT, Araujo HN, Gonçalves DA, Simões Fróes Guimarães DSP, Teodoro BG, So K, Espreafico EM, Hirshman MF, Alberici LC, Kettelhut IDC, Goodyear LJ, Silveira LR. The MicroRNA miR-696 is regulated by SNARK and reduces mitochondrial activity in mouse skeletal muscle through Pgc1α inhibition. Mol Metab 2021; 51:101226. [PMID: 33812060 PMCID: PMC8121711 DOI: 10.1016/j.molmet.2021.101226] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/16/2021] [Accepted: 03/25/2021] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE MicroRNAs (miRNA) are known to regulate the expression of genes involved in several physiological processes including metabolism, mitochondrial biogenesis, proliferation, differentiation, and cell death. METHODS Using "in silico" analyses, we identified 219 unique miRNAs that potentially bind to the 3'UTR region of a critical mitochondrial regulator, the peroxisome proliferator-activated receptor gamma coactivator (PGC) 1 alpha (Pgc1α). Of the 219 candidate miRNAs, miR-696 had one of the highest interactions at the 3'UTR of Pgc1α, suggesting that miR-696 may be involved in the regulation of Pgc1α. RESULTS Consistent with this hypothesis, we found that miR-696 was highly expressed in the skeletal muscle of STZ-induced diabetic mice and chronic high-fat-fed mice. C2C12 muscle cells exposed to palmitic acid also exhibited a higher expression of miR-696. This increased expression corresponded with a reduced expression of oxidative metabolism genes and reduced mitochondrial respiration. Importantly, reducing miR-696 reversed decreases in mitochondrial activity in response to palmitic acid. Using C2C12 cells treated with the AMP-activated protein kinase (AMPK) activator AICAR and skeletal muscle from AMPKα2 dominant-negative (DN) mice, we found that the signaling mechanism regulating miR-696 did not involve AMPK. In contrast, overexpression of SNF1-AMPK-related kinase (SNARK) in C2C12 cells increased miR-696 transcription while knockdown of SNARK significantly decreased miR-696. Moreover, muscle-specific transgenic mice overexpressing SNARK exhibited a lower expression of Pgc1α, elevated levels of miR-696, and reduced amounts of spontaneous activity. CONCLUSIONS Our findings demonstrate that metabolic stress increases miR-696 expression in skeletal muscle cells, which in turn inhibits Pgc1α, reducing mitochondrial function. SNARK plays a role in this process as a metabolic stress signaling molecule inducing the expression of miR-696.
Collapse
Affiliation(s)
- André L Queiroz
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, USP, Ribeirão Preto, Brazil; Research Division, Joslin Diabetes Center, and Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Sarah J Lessard
- Research Division, Joslin Diabetes Center, and Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Amanda T Ouchida
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, USP, Ribeirão Preto, Brazil
| | - Hygor N Araujo
- Obesity and Comorbidities Research Center, OCRC, IB, UNICAMP, Campinas, Brazil
| | - Dawit A Gonçalves
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, USP, Ribeirão Preto, Brazil
| | | | - Bruno G Teodoro
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, USP, Ribeirão Preto, Brazil; Department of Physics and Chemistry, Faculty of Pharmaceutical Science, USP, Ribeirão Preto, Brazil
| | - Kawai So
- Research Division, Joslin Diabetes Center, and Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Enilza M Espreafico
- Department of Cell Biology, Ribeirão Preto Medical School, USP, Ribeirão Preto, Brazil
| | - Michael F Hirshman
- Research Division, Joslin Diabetes Center, and Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Luciane C Alberici
- Department of Physics and Chemistry, Faculty of Pharmaceutical Science, USP, Ribeirão Preto, Brazil
| | - Isis do Carmo Kettelhut
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, USP, Ribeirão Preto, Brazil
| | - Laurie J Goodyear
- Research Division, Joslin Diabetes Center, and Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| | - Leonardo R Silveira
- Obesity and Comorbidities Research Center, OCRC, IB, UNICAMP, Campinas, Brazil.
| |
Collapse
|
27
|
Antihuman CD44 antibody BJ18 inhibits platelet phagocytosis by correcting aberrant FcɣR expression and M1 polarization in immune thrombocytopenia. Int Immunopharmacol 2021; 95:107502. [PMID: 33690000 DOI: 10.1016/j.intimp.2021.107502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND Immune thrombocytopenia (ITP) is an autoimmune hemorrhagic disease with a low platelet count. CD44 is a pivotal component involved in phagocytosis and inflammation, and monoclonal antibodies (mAbs) against CD44 have been shown to be beneficial in several autoimmune diseases. In the present study, we investigated the correlation between CD44 levels and disease severity in patients with ITP and explored the immunomodulatory mechanisms of the antihuman CD44 mAb BJ18 on platelet phagocytosis mediated by monocytes/macrophages. METHODS Plasma was collected from 45 participants to measure the circulating concentration of CD44 using ELISA. Peripheral blood mononuclear cells from patients and controls were isolated and induced to differentiate into monocytes/macrophages utilizing cytokines and drugs. CD44 expression on circulating cells and the effects of BJ18 on platelet phagocytosis, Fcɣ receptor (FcɣR) expression and M1/M2 polarization of macrophages were evaluated using flow cytometry and qPCR. RESULTS CD44 levels of both the soluble form found in plasma and the form expressed on the surface of circulating monocytes/macrophages were significantly elevated in ITP patients. Linear correlations were verified between the CD44 levels and major clinical characteristics. In an in vitro study, BJ18 successfully inhibited platelet phagocytosis by monocytes/macrophages obtained from ITP patients. Further studies indicated that BJ18 corrected abnormal FcγR expression on monocytes/macrophages. Moreover, the polarization of proinflammatory M1 macrophages could also be regulated by BJ18. CONCLUSIONS Our data indicated that the CD44 level has potential predictive value for disease severity and that the antihuman CD44 mAb BJ18 may be a promising therapy for ITP patients.
Collapse
|
28
|
|
29
|
Rao N, Rho JG, Um W, EK PK, Nguyen VQ, Oh BH, Kim W, Park JH. Hyaluronic Acid Nanoparticles as Nanomedicine for Treatment of Inflammatory Diseases. Pharmaceutics 2020; 12:E931. [PMID: 33003609 PMCID: PMC7600604 DOI: 10.3390/pharmaceutics12100931] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 09/24/2020] [Accepted: 09/28/2020] [Indexed: 02/06/2023] Open
Abstract
Owing to their unique biological functions, hyaluronic acid (HA) and its derivatives have been explored extensively for biomedical applications such as tissue engineering, drug delivery, and molecular imaging. In particular, self-assembled HA nanoparticles (HA-NPs) have been used widely as target-specific and long-acting nanocarriers for the delivery of a wide range of therapeutic or diagnostic agents. Recently, it has been demonstrated that empty HA-NPs without bearing any therapeutic agent can be used therapeutically for the treatment of inflammatory diseases via modulating inflammatory responses. In this review, we aim to provide an overview of the significant achievements in this field and highlight the potential of HA-NPs for the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- N.Vijayakameswara Rao
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Korea; (N.V.R.); (W.U.); (P.K.E.); (V.Q.N.); (B.H.O.)
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei 10617, Taiwan
| | - Jun Gi Rho
- Department of Molecular Science & Technology, Ajou University, Suwon 16499, Korea;
| | - Wooram Um
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Korea; (N.V.R.); (W.U.); (P.K.E.); (V.Q.N.); (B.H.O.)
| | - Pramod Kumar EK
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Korea; (N.V.R.); (W.U.); (P.K.E.); (V.Q.N.); (B.H.O.)
| | - Van Quy Nguyen
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Korea; (N.V.R.); (W.U.); (P.K.E.); (V.Q.N.); (B.H.O.)
| | - Byeong Hoon Oh
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Korea; (N.V.R.); (W.U.); (P.K.E.); (V.Q.N.); (B.H.O.)
| | - Wook Kim
- Department of Molecular Science & Technology, Ajou University, Suwon 16499, Korea;
| | - Jae Hyung Park
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Korea; (N.V.R.); (W.U.); (P.K.E.); (V.Q.N.); (B.H.O.)
- Department Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea
| |
Collapse
|
30
|
Hounkpe BW, Benatti RDO, Carvalho BDS, De Paula EV. Identification of common and divergent gene expression signatures in patients with venous and arterial thrombosis using data from public repositories. PLoS One 2020; 15:e0235501. [PMID: 32780732 PMCID: PMC7418995 DOI: 10.1371/journal.pone.0235501] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 06/17/2020] [Indexed: 12/31/2022] Open
Abstract
STRENGTHS AND LIMITATIONS OF THIS STUDY Our results represent the first comparison of venous and arterial thrombosis at the transcriptomic level.Our main result was the demonstration that immunothrombosis pathways are important to the pathophysiology of these conditions, also at the transcriptomic level.A specific signature for venous and arterial thrombosis was described, and validated in independent cohorts.The limited number of public repositories with gene expression data from patients with venous thromboembolism limits the representation of these patients in our analyses.In order to gather a meaningful number of studies with gene expression data we had to include patients in different time-points since the index thrombotic event, which might have increased the heterogeneity of our population.
Collapse
Affiliation(s)
| | | | - Benilton de Sá Carvalho
- Department of Statistics, Institute of Mathematics, Statistics and Scientific Computing, University of Campinas, Campinas, SP, Brazil
| | - Erich Vinicius De Paula
- School of Medical Sciences, University of Campinas, Campinas, SP, Brazil
- Hematology and Hemotherapy Center, University of Campinas, Campinas, SP, Brazil
| |
Collapse
|
31
|
Wu L, Sun J, Liu L, Du X, Liu Y, Yan X, Kombo Osoro E, Zhang F, Feng L, Liang D, Li Y, Chen Q, Sun S, Zhang L, Lan X, Li D, Lu S. Anti-toll-like receptor 2 antibody ameliorates hepatic injury, inflammation, fibrosis and steatosis in obesity-related metabolic disorder rats via regulating MAPK and NF-κB pathways. Int Immunopharmacol 2020; 82:106368. [PMID: 32151955 DOI: 10.1016/j.intimp.2020.106368] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 02/28/2020] [Accepted: 02/29/2020] [Indexed: 02/06/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is one of the most common liver diseases worldwide, which includes a spectrum of histological liver changes. Non-alcoholic steatohepatitis (NASH) is considered to be the progressive subtype of NAFLD, which is characterized by lobular inflammation and cellular ballooning on the basis of steatosis. There is a critical need to develop novel and effective therapeutic approaches for NAFLD/NASH. The activation of toll-like receptor 2 (TLR2) signaling pathway plays a key role in high-fat-related inflammation, triggering the occurrence and development of NASH. Herein, the anti-TLR2 monoclonal antibody (TLR2 mAb) was prepared and investigated for its ability to ameliorate the inflammatory response in vivo and in vitro. The anti-inflammatory role of TLR2 mAb in vitro was examined in NR8383 macrophage cells and THP-1 derived macrophage cells. For confirmation in vivo, three groups of SD rats were treated for 20 weeks: rats in the control were fed with a standard diet; rates in the IgG and TLR2 mAb groups were fed with a high-fat diet and with IgG or TLR2 mAb, respectively. Liver tissue and serum were collected for further analysis. Results showed that after 4-week treatment with TLR2 mAb, metabolic parameters in rats were improved markedly (body weight, fasting blood glucose level, liver steatosis, inflammatory response and fibrosis). Moreover, western blotting demonstrated that the TLR2 mAb blocked MAPKs and NF-κB activation, and inhibited the expression of inflammatory factors in rat liver tissue. These effects suggested that TLR2 mAb could improve HFD-induced hepatic injury, inflammation, fibrosis and steatosis by suppressing inflammatory response and regulating the hepatic MAPKs and NF-κB signaling pathways. This suggests that TLR2 may be a novel therapeutic target for metabolic diseases especially NASH.
Collapse
Affiliation(s)
- Litao Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China
| | - Juan Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China
| | - Li Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China; Department of Pathology, Sunshine Union Hospital, Weifang, Shandong Province 261061, PR China
| | - Xiaojuan Du
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China
| | - Yan Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China
| | - Xiaofei Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China
| | - Ezra Kombo Osoro
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China
| | - Fujun Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China
| | - Lina Feng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China
| | - Dong Liang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China
| | - Yazhao Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China
| | - Qian Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China
| | - Sha Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China
| | - Lei Zhang
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Science, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, PR China
| | - Xi Lan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China.
| | - Dongmin Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China.
| | - Shemin Lu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China
| |
Collapse
|
32
|
Luci C, Bourinet M, Leclère PS, Anty R, Gual P. Chronic Inflammation in Non-Alcoholic Steatohepatitis: Molecular Mechanisms and Therapeutic Strategies. Front Endocrinol (Lausanne) 2020; 11:597648. [PMID: 33384662 PMCID: PMC7771356 DOI: 10.3389/fendo.2020.597648] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 11/03/2020] [Indexed: 12/11/2022] Open
Abstract
Non-Alcoholic Steatohepatitis (NASH) is the progressive form of Non-Alcoholic Fatty Liver Disease (NAFLD), the main cause of chronic liver complications. The development of NASH is the consequence of aberrant activation of hepatic conventional immune, parenchymal, and endothelial cells in response to inflammatory mediators from the liver, adipose tissue, and gut. Hepatocytes, Kupffer cells and liver sinusoidal endothelial cells contribute to the significant accumulation of bone-marrow derived-macrophages and neutrophils in the liver, a hallmark of NASH. The aberrant activation of these immune cells elicits harmful inflammation and liver injury, leading to NASH progression. In this review, we highlight the processes triggering the recruitment and/or activation of hepatic innate immune cells, with a focus on macrophages, neutrophils, and innate lymphoid cells as well as the contribution of hepatocytes and endothelial cells in driving liver inflammation/fibrosis. On-going studies and preliminary results from global and specific therapeutic strategies to manage this NASH-related inflammation will also be discussed.
Collapse
Affiliation(s)
- Carmelo Luci
- Université Côte d’Azur, INSERM, C3M, Nice, France
| | | | | | - Rodolphe Anty
- Université Côte d’Azur, CHU, INSERM, C3M, Nice, France
| | - Philippe Gual
- Université Côte d’Azur, INSERM, C3M, Nice, France
- *Correspondence: Philippe Gual,
| |
Collapse
|
33
|
Hasib A, Hennayake CK, Bracy DP, Bugler-Lamb AR, Lantier L, Khan F, Ashford MLJ, McCrimmon RJ, Wasserman DH, Kang L. CD44 contributes to hyaluronan-mediated insulin resistance in skeletal muscle of high-fat-fed C57BL/6 mice. Am J Physiol Endocrinol Metab 2019; 317:E973-E983. [PMID: 31550181 PMCID: PMC6957377 DOI: 10.1152/ajpendo.00215.2019] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Extracellular matrix hyaluronan is increased in skeletal muscle of high-fat-fed insulin-resistant mice, and reduction of hyaluronan by PEGPH20 hyaluronidase ameliorates diet-induced insulin resistance (IR). CD44, the main hyaluronan receptor, is positively correlated with type 2 diabetes. This study determines the role of CD44 in skeletal muscle IR. Global CD44-deficient (cd44-/-) mice and wild-type littermates (cd44+/+) were fed a chow diet or 60% high-fat diet for 16 wk. High-fat-fed cd44-/- mice were also treated with PEGPH20 to evaluate its CD44-dependent action. Insulin sensitivity was measured by hyperinsulinemic-euglycemic clamp (ICv). High-fat feeding increased muscle CD44 protein expression. In the absence of differences in body weight and composition, despite lower clamp insulin during ICv, the cd44-/- mice had sustained glucose infusion rate (GIR) regardless of diet. High-fat diet-induced muscle IR as evidenced by decreased muscle glucose uptake (Rg) was exhibited in cd44+/+ mice but absent in cd44-/- mice. Moreover, gastrocnemius Rg remained unchanged between genotypes on chow diet but was increased in high-fat-fed cd44-/- compared with cd44+/+ when normalized to clamp insulin concentrations. Ameliorated muscle IR in high-fat-fed cd44-/- mice was associated with increased vascularization. In contrast to previously observed increases in wild-type mice, PEGPH20 treatment in high-fat-fed cd44-/- mice did not change GIR or muscle Rg during ICv, suggesting a CD44-dependent action. In conclusion, genetic CD44 deletion improves muscle IR, and the beneficial effects of PEGPH20 are CD44-dependent. These results suggest a critical role of CD44 in promoting hyaluronan-mediated muscle IR, therefore representing a potential therapeutic target for diabetes.
Collapse
Affiliation(s)
- Annie Hasib
- Division of Systems Medicine, School of Medicine, University of Dundee, Dundee, Scotland, United Kingdom
| | - Chandani K Hennayake
- Division of Systems Medicine, School of Medicine, University of Dundee, Dundee, Scotland, United Kingdom
| | - Deanna P Bracy
- Department of Molecular Physiology and Biophysics and Mouse Metabolic Phenotyping Centre, Vanderbilt University, Nashville, Tennessee
| | - Aimée R Bugler-Lamb
- Division of Systems Medicine, School of Medicine, University of Dundee, Dundee, Scotland, United Kingdom
| | - Louise Lantier
- Department of Molecular Physiology and Biophysics and Mouse Metabolic Phenotyping Centre, Vanderbilt University, Nashville, Tennessee
| | - Faisel Khan
- Division of Systems Medicine, School of Medicine, University of Dundee, Dundee, Scotland, United Kingdom
| | - Michael L J Ashford
- Division of Systems Medicine, School of Medicine, University of Dundee, Dundee, Scotland, United Kingdom
| | - Rory J McCrimmon
- Division of Systems Medicine, School of Medicine, University of Dundee, Dundee, Scotland, United Kingdom
| | - David H Wasserman
- Department of Molecular Physiology and Biophysics and Mouse Metabolic Phenotyping Centre, Vanderbilt University, Nashville, Tennessee
| | - Li Kang
- Division of Systems Medicine, School of Medicine, University of Dundee, Dundee, Scotland, United Kingdom
| |
Collapse
|
34
|
Masotti M, Guo B, Wu B. Pleiotropy informed adaptive association test of multiple traits using genome-wide association study summary data. Biometrics 2019; 75:1076-1085. [PMID: 31021400 DOI: 10.1111/biom.13076] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 04/16/2019] [Indexed: 12/17/2022]
Abstract
Genetic variants associated with disease outcomes can be used to develop personalized treatment. To reach this precision medicine goal, hundreds of large-scale genome-wide association studies (GWAS) have been conducted in the past decade to search for promising genetic variants associated with various traits. They have successfully identified tens of thousands of disease-related variants. However, in total these identified variants explain only part of the variation for most complex traits. There remain many genetic variants with small effect sizes to be discovered, which calls for the development of (a) GWAS with more samples and more comprehensively genotyped variants, for example, the NHLBI Trans-Omics for Precision Medicine (TOPMed) Program is planning to conduct whole genome sequencing on over 100 000 individuals; and (b) novel and more powerful statistical analysis methods. The current dominating GWAS analysis approach is the "single trait" association test, despite the fact that many GWAS are conducted in deeply phenotyped cohorts including many correlated and well-characterized outcomes, which can help improve the power to detect novel variants if properly analyzed, as suggested by increasing evidence that pleiotropy, where a genetic variant affects multiple traits, is the norm in genome-phenome associations. We aim to develop pleiotropy informed powerful association test methods across multiple traits for GWAS. Since it is generally very hard to access individual-level GWAS phenotype and genotype data for those existing GWAS, due to privacy concerns and various logistical considerations, we develop rigorous statistical methods for pleiotropy informed adaptive multitrait association test methods that need only summary association statistics publicly available from most GWAS. We first develop a pleiotropy test, which has powerful performance for truly pleiotropic variants but is sensitive to the pleiotropy assumption. We then develop a pleiotropy informed adaptive test that has robust and powerful performance under various genetic models. We develop accurate and efficient numerical algorithms to compute the analytical P-value for the proposed adaptive test without the need of resampling or permutation. We illustrate the performance of proposed methods through application to joint association test of GWAS meta-analysis summary data for several glycemic traits. Our proposed adaptive test identified several novel loci missed by individual trait based GWAS meta-analysis. All the proposed methods are implemented in a publicly available R package.
Collapse
Affiliation(s)
- Maria Masotti
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, Minnesota
| | - Bin Guo
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, Minnesota
| | - Baolin Wu
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
35
|
Nagy N, Sunkari VG, Kaber G, Hasbun S, Lam DN, Speake C, Sanda S, McLaughlin TL, Wight TN, Long SR, Bollyky PL. Hyaluronan levels are increased systemically in human type 2 but not type 1 diabetes independently of glycemic control. Matrix Biol 2019; 80:46-58. [PMID: 30196101 PMCID: PMC6401354 DOI: 10.1016/j.matbio.2018.09.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 09/04/2018] [Accepted: 09/05/2018] [Indexed: 01/19/2023]
Abstract
Hyaluronan (HA), an extracellular matrix glycosaminoglycan, is implicated in the pathogenesis of both type 1 diabetes (T1D) as well as type 2 diabetes (T2D) and has been postulated to be increased in these diseases due to hyperglycemia. We have examined the serum and tissue distribution of HA in human subjects with T1D and T2D and in mouse models of these diseases and evaluated the relationship between HA levels and glycemic control. We found that serum HA levels are increased in T2D but not T1D independently of hemoglobin-A1c, C-peptide, body mass index, or time since diabetes diagnosis. HA is likewise increased in skeletal muscle in T2D subjects relative to non-diabetic controls. Analogous increases in serum and muscle HA are seen in diabetic db/db mice (T2D), but not in diabetic DORmO mice (T1D). Diabetes induced by the β-cell toxin streptozotozin (STZ) lead to an increase in blood glucose but not to an increase in serum HA. These data indicate that HA levels are increased in multiple tissue compartments in T2D but not T1D independently of glycemic control. Given that T2D but not T1D is associated with systemic inflammation, these patterns are consistent with inflammatory factors and not hyperglycemia driving increased HA. Serum HA may have value as a biomarker of systemic inflammation in T2D.
Collapse
Affiliation(s)
- Nadine Nagy
- Division of Infectious Diseases and Geographic Medicine, Dept. of Medicine, Stanford University School of Medicine, Beckman Center, 279 Campus Drive, Stanford, CA, 94305
| | - Vivekananda G. Sunkari
- Division of Infectious Diseases and Geographic Medicine, Dept. of Medicine, Stanford University School of Medicine, Beckman Center, 279 Campus Drive, Stanford, CA, 94305
| | - Gernot Kaber
- Division of Infectious Diseases and Geographic Medicine, Dept. of Medicine, Stanford University School of Medicine, Beckman Center, 279 Campus Drive, Stanford, CA, 94305
| | - Sonia Hasbun
- Department of Cardiology, Good Samaritan Regional Medical Center, 3600 NW Samaritan Dr, Corvallis, OR, 97330
| | - Dung N. Lam
- Division of Infectious Diseases and Geographic Medicine, Dept. of Medicine, Stanford University School of Medicine, Beckman Center, 279 Campus Drive, Stanford, CA, 94305
| | - Cate Speake
- Diabetes Clinical Research Program, Benaroya Research Institute, 1201 Ninth Ave, Seattle, WA, 98101
| | - Srinath Sanda
- Department of Pediatrics, UCSF School of Medicine, 513 Parnassus Avenue, San Francisco, CA, 94143
| | - Tracey L. McLaughlin
- Department of Medicine, Medicine – Endocrinology, Endocrine Clinic, Stanford School of Medicine, 300 Pasteur Drive, Stanford, CA, 94305
| | - Thomas N. Wight
- Matrix Biology Program, Benaroya Research Institute, 1201 Ninth Ave, Seattle, WA, 98101
| | - Steven R. Long
- Department of Pathology, Stanford University School of Medicine, Lane 235, 300 Pasteur Drive, Stanford, CA, 94305
| | - Paul L. Bollyky
- Division of Infectious Diseases and Geographic Medicine, Dept. of Medicine, Stanford University School of Medicine, Beckman Center, 279 Campus Drive, Stanford, CA, 94305
| |
Collapse
|
36
|
Gong XM, Li YF, Luo J, Wang JQ, Wei J, Wang JQ, Xiao T, Xie C, Hong J, Ning G, Shi XJ, Li BL, Qi W, Song BL. Gpnmb secreted from liver promotes lipogenesis in white adipose tissue and aggravates obesity and insulin resistance. Nat Metab 2019; 1:570-583. [PMID: 32694855 DOI: 10.1038/s42255-019-0065-4] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 04/05/2019] [Indexed: 12/13/2022]
Abstract
Metabolism in mammals is regulated by complex interplay among different organs. Fatty acid synthesis is increased in white adipose tissue (WAT) when it is inhibited in the liver. Here we identify glycoprotein non-metastatic melanoma protein B (Gpnmb) as one liver-WAT cross-talk factor involved in lipogenesis. Inhibition of the hepatic sterol regulatory element-binding protein pathway leads to increased transcription of Gpnmb and promotes processing of the membrane protein to a secreted form. Gpnmb stimulates lipogenesis in WAT and exacerbates diet-induced obesity and insulin resistance. In humans, Gpnmb is tightly associated with body mass index and is a strong risk factor for obesity. Gpnmb inhibition by a neutralizing antibody or liver-specific knockdown improves metabolic parameters, including weight gain reduction and increased insulin sensitivity, probably by promoting the beiging of WAT. These results suggest that Gpnmb is a liver-secreted factor regulating lipogenesis in WAT, and that Gpnmb inhibition may provide a therapeutic strategy in obesity and diabetes.
Collapse
Affiliation(s)
- Xue-Min Gong
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Institute for Advanced Studies, Wuhan University, Wuhan, China
- The State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yun-Feng Li
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Jie Luo
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Ji-Qiu Wang
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, State Key Laboratory of Medical Genomes, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Wei
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Ju-Qiong Wang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Ting Xiao
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Chang Xie
- The State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jie Hong
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, State Key Laboratory of Medical Genomes, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang Ning
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, State Key Laboratory of Medical Genomes, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiong-Jie Shi
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Bo-Liang Li
- The State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Wei Qi
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| | - Bao-Liang Song
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Institute for Advanced Studies, Wuhan University, Wuhan, China.
| |
Collapse
|
37
|
Hansel C, Erschfeld S, Baues M, Lammers T, Weiskirchen R, Trautwein C, Kroy DC, Drescher HK. The Inhibitory T Cell Receptors PD1 and 2B4 Are Differentially Regulated on CD4 and CD8 T Cells in a Mouse Model of Non-alcoholic Steatohepatitis. Front Pharmacol 2019; 10:244. [PMID: 30949049 PMCID: PMC6436071 DOI: 10.3389/fphar.2019.00244] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 02/26/2019] [Indexed: 12/26/2022] Open
Abstract
Infiltrating CD4 and CD8 T cells have been shown to worsen inflammatory liver damage in non-alcoholic steatohepatitis (NASH). Inhibitory T cell receptors such as the programmed cell death protein 1 (PD1) and the natural killer cell receptor 2B4 regulate the activity of CD4 and CD8 T cells and therefore play an important role in immune tolerance required in the liver. In this study, we investigated the expression profile of inhibitory T cell receptors on CD4 and CD8 T cells in a mouse model of NASH. Male B57BL/6J mice were fed a Western diet for 24 weeks. The expression levels of inhibitory receptors on the surface of intrahepatic and peripheral T cells were measured and correlated with markers of activation (CD107a, CD69, and CD44), metabolic disorder (serum triglycerides, serum cholesterol, γ-glutamyl transferase, hepatic triglycerides), inflammation (serum alanine aminotransferase and aspartate aminotransferase) and hepatic fibrosis (collagen 1A1, α-smooth muscle actin, hydroxyproline). Under Western diet, PD1 is exclusively upregulated on intrahepatic and peripheral CD8+ T cells, whereas the expression level on CD4 T cells is unaffected. In contrast, 2B4 is upregulated liver-specifically on both CD4 and CD8 T cells and unchanged on peripheral T cells. Upregulation of PD1 on CD8 T cells is restricted to CD8 effector memory T cells and correlates with lower levels of degranulation. Similarly, the inhibitory function of PD1 on intrahepatic CD4 T cells is shown by a lower CD69 and CD44 expression on PD1-positive CD4 T cells. In murine steatohepatitis, the upregulation of PD1 on CD8 T cells and 2B4 on CD4 and CD8 T cells potentially limits T cell-mediated liver damage. Therefore, these inhibitory T cell receptors could serve as promising targets of immune-modulatory NASH therapy.
Collapse
Affiliation(s)
- Cordula Hansel
- Department of Internal Medicine III, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Stephanie Erschfeld
- Department of Internal Medicine III, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Maike Baues
- Institute for Experimental Molecular Imaging, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy, and Clinical Chemistry (IFMPEGKC), University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Christian Trautwein
- Department of Internal Medicine III, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Daniela C Kroy
- Department of Internal Medicine III, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Hannah K Drescher
- Department of Internal Medicine III, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
38
|
Characterization of Growth, Fat Deposition, and Lipid Metabolism-Related Gene Expression in Lean and Obese Meat-Type Chickens. J Poult Sci 2019; 56:101-111. [PMID: 32055204 PMCID: PMC7005403 DOI: 10.2141/jpsa.0180064] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Excessive fat deposition adversely affects poultry production. In this study, we investigated growth, fat deposition, and hepatic mRNA expression of 13 lipid metabolism-related genes in three unique breeds of meat-type chickens with distinct breed origins and genetic relationships. One was Nagoya (NAG), a native Japanese breed, whereas the others were White Plymouth Rock (WPR) and White Cornish (WC), which have been used worldwide as the parental breeds of common broiler chickens. NAG chickens were phenotypically characterized by slow growth, lean body fat, and high gizzard and liver weights. In contrast, both WC and WPR chickens were characterized by rapid growth but high percentage of subcutaneous fat and abdominal fat weight, resulting from high feed intake. Among the three breeds, WC had the highest percentage of pectoral muscle weight, whereas WPR was the most obese. Among lipid metabolism-related genes, the expression of PPARA, PPARG, and CD36 was mostly associated with obesity. These results provide basic information for quantitative trait locus (QTL) analysis related to growth and fat traits in an F2 population of the lean NAG breed and the obese WPR breed of meat-type chickens in future.
Collapse
|
39
|
Kornicka K, Houston J, Marycz K. Dysfunction of Mesenchymal Stem Cells Isolated from Metabolic Syndrome and Type 2 Diabetic Patients as Result of Oxidative Stress and Autophagy may Limit Their Potential Therapeutic Use. Stem Cell Rev Rep 2018; 14:337-345. [PMID: 29611042 PMCID: PMC5960487 DOI: 10.1007/s12015-018-9809-x] [Citation(s) in RCA: 128] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Mesenchymal stem cells (MSC) have become a promising tool for therapeutic intervention. Their unique features, including self-renewal, multipotency and immunomodulatory properties draw the worldwide attention of researchers and physicians with respect to their application in disease treatment. However, the environment (so-called niche) from which MSCs are isolated may determine their usefulness. Many studies indicated the involvement of MSCs in ageing and disease. In this review, we have focused on how type 2 diabetes (T2D) and metabolic syndrome (MS) affect MSC properties, and thus limit their therapeutic potential. Herein, we mainly focus on apoptosis, autophagy and mitochondria deterioration processes that indirectly affect MSC fate. Based on the data presented, special attention should be paid when considering autologous MSC therapy in T2D or MS treatments, as their therapeutic potential may be restricted.
Collapse
Affiliation(s)
- Katarzyna Kornicka
- Department of Experimental Biology, The Faculty of Biology and Animal Science, Wroclaw University of Environmental and Life Sciences, Norwida 25, 50-375, Wrocław, Poland.
| | - Jenny Houston
- PferdePraxis Dr. Med. Vet. Daniel Weiss, Postmatte 14, CH-8807, Freienbach, Switzerland
| | - Krzysztof Marycz
- Department of Experimental Biology, The Faculty of Biology and Animal Science, Wroclaw University of Environmental and Life Sciences, Norwida 25, 50-375, Wrocław, Poland.,Wroclaw Research Centre EIT+, 54-066, Wroclaw, Poland
| |
Collapse
|
40
|
The Effect of Chronic Inflammation and Oxidative and Endoplasmic Reticulum Stress in the Course of Metabolic Syndrome and Its Therapy. Stem Cells Int 2018; 2018:4274361. [PMID: 30425746 PMCID: PMC6217741 DOI: 10.1155/2018/4274361] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 09/26/2018] [Accepted: 10/03/2018] [Indexed: 12/14/2022] Open
Abstract
Metabolic syndrome (MetS) is highly associated with a modern lifestyle. The prevalence of MetS has reached epidemic proportion and is still rising. The main cause of MetS and finally type 2 diabetes occurrence is excessive nutrient intake, lack of physical activity, and inflammatory cytokines secretion. These factors lead to redistribution of body fat and oxidative and endoplasmic reticulum (ER) stress occurrence, resulting in insulin resistance, increase adipocyte differentiation, and much elevated levels of proinflammatory cytokines. Cellular therapies, especially mesenchymal stem cell (MSC) transplantation, seem to be promising in the MetS and type 2 diabetes treatments, due to their immunomodulatory effect and multipotent capacity; adipose-derived stem cells (ASCs) play a crucial role in MSC-based cellular therapies. In this review, we focused on etiopathology of MetS, especially on the crosstalk between chronic inflammation, oxidative stress, and ER stress and their effect on MetS-related disease occurrence, as well as future perspectives of cellular therapies. We also provide an overview of therapeutic approaches that target endoplasmic reticulum and oxidative stress.
Collapse
|
41
|
Pierre C, Guillebaud F, Airault C, Baril N, Barbouche R, Save E, Gaigé S, Bariohay B, Dallaporta M, Troadec JD. Invalidation of Microsomal Prostaglandin E Synthase-1 (mPGES-1) Reduces Diet-Induced Low-Grade Inflammation and Adiposity. Front Physiol 2018; 9:1358. [PMID: 30333759 PMCID: PMC6176076 DOI: 10.3389/fphys.2018.01358] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 09/07/2018] [Indexed: 01/04/2023] Open
Abstract
Chronic low-grade inflammation is known to be linked to obesity, and to occur in the early stages of the disease. This mechanism is complex and involves numerous organs, cells, and cytokines. In this context, inflammation of white adipose tissue seems to play a key role in the development of obesity. Because of its properties, prostaglandin E2 (PGE2), an emblematic inflammatory mediator, has been proposed as an actor linking inflammation and obesity. Indeed, PGE2 is involved in mechanisms that are dysregulated in obesity such as lipolysis and adipogenesis. Microsomal prostaglandin E synthase-1 (mPGES-1) is an enzyme, which specifically catalyzes the final step of PGE2 biosynthesis. Interestingly, mPGES-1 invalidation dramatically alters the production of PGE2 during inflammation. In the present work, we sought to determine whether mPGES-1 could contribute to inflammation associated with obesity. To this end, we analyzed the energy metabolism of mPGES-1 deficient mice (mPGES-1-/-) and littermate controls, fed with a high-fat diet. Our data showed that mPGES-1-/- mice exhibited resistance to diet-induced obesity when compared to wild-type littermates. mPGES-1-/- mice fed with a high-fat diet, showed a lower body weight gain and a reduced adiposity, which were accompanied by a decrease in adipose tissues inflammation. We also observed an increase in energy expenditures in mPGES-1-/- mice fed with a high-fat diet without any changes in activity and browning process. Altogether, these data suggest that mPGES-1 inhibition may prevent diet-induced obesity.
Collapse
Affiliation(s)
- Clément Pierre
- Aix Marseille Université, CNRS, Laboratoire de Neurosciences Cognitives UMR 7291, Marseille, France.,Biomeostasis CRO, La Penne-sur-Huveaune, France
| | - Florent Guillebaud
- Aix Marseille Université, CNRS, Laboratoire de Neurosciences Cognitives UMR 7291, Marseille, France
| | - Coraline Airault
- Aix Marseille Université, CNRS, Laboratoire de Neurosciences Cognitives UMR 7291, Marseille, France
| | - Nathalie Baril
- CNRS, Fédération de Recherche 3C FR 3512, Aix-Marseille Université, Marseille, France
| | - Rym Barbouche
- Aix Marseille Université, CNRS, Laboratoire de Neurosciences Cognitives UMR 7291, Marseille, France
| | - Etienne Save
- Aix Marseille Université, CNRS, Laboratoire de Neurosciences Cognitives UMR 7291, Marseille, France
| | - Stéphanie Gaigé
- Aix Marseille Université, CNRS, Laboratoire de Neurosciences Cognitives UMR 7291, Marseille, France
| | | | - Michel Dallaporta
- Aix Marseille Université, CNRS, Laboratoire de Neurosciences Cognitives UMR 7291, Marseille, France
| | - Jean-Denis Troadec
- Aix Marseille Université, CNRS, Laboratoire de Neurosciences Cognitives UMR 7291, Marseille, France
| |
Collapse
|
42
|
Takahashi M, Hori M, Ishigamori R, Mutoh M, Imai T, Nakagama H. Fatty pancreas: A possible risk factor for pancreatic cancer in animals and humans. Cancer Sci 2018; 109:3013-3023. [PMID: 30099827 PMCID: PMC6172058 DOI: 10.1111/cas.13766] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 08/01/2018] [Accepted: 08/04/2018] [Indexed: 02/06/2023] Open
Abstract
Obesity, type 2 diabetes mellitus (T2DM) and aging are associated with pancreatic cancer risk, but the mechanisms of pancreatic cancer development caused by these factors are not clearly understood. Syrian golden hamsters are susceptible to N‐nitrosobis(2‐oxopropyl)amine (BOP)‐induced pancreatic carcinogenesis. Aging, BOP treatment and/or a high‐fat diet cause severe and scattered fatty infiltration (FI) of the pancreas with abnormal adipokine production and promote pancreatic ductal adenocarcinoma (PDAC) development. The KK‐Ay mouse, a T2DM model, also develops severe and scattered FI of the pancreas. Treatment with BOP induced significantly higher cell proliferation in the pancreatic ducts of KK‐Ay mice, but not in those of ICR and C57BL/6J mice, both of which are characterized by an absence of scattered FI. Thus, we hypothesized that severely scattered FI may be involved in the susceptibility to PDAC development. Indeed, severe pancreatic FI, or fatty pancreas, is observed in humans and is associated with age, body mass index (BMI) and DM, which are risk factors for pancreatic cancer. We analyzed the degree of FI in the non‐cancerous parts of PDAC and non‐PDAC patients who had undergone pancreatoduodenectomy by histopathology and demonstrated that the degree of pancreatic FI in PDAC cases is significantly higher than that in non‐PDAC controls. Moreover, the association with PDAC is positive, even after adjusting for BMI and the prevalence of DM. Accumulating evidence suggests that pancreatic FI is involved in PDAC development in animals and humans, and further investigations to clarify the genetic and environmental factors that cause pancreatic FI are warranted.
Collapse
Affiliation(s)
- Mami Takahashi
- Central Animal Division, National Cancer Center Research Institute, Tokyo, Japan
| | - Mika Hori
- Department of Molecular Innovation in Lipidology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Rikako Ishigamori
- Central Animal Division, National Cancer Center Research Institute, Tokyo, Japan
| | - Michihiro Mutoh
- Epidemiology and Prevention Division, Research Center for Cancer Prevention and Screening, National Cancer Center, Tokyo, Japan
| | - Toshio Imai
- Central Animal Division, National Cancer Center Research Institute, Tokyo, Japan
| | | |
Collapse
|
43
|
Ben Hamda C, Sangeda R, Mwita L, Meintjes A, Nkya S, Panji S, Mulder N, Guizani-Tabbane L, Benkahla A, Makani J, Ghedira K. A common molecular signature of patients with sickle cell disease revealed by microarray meta-analysis and a genome-wide association study. PLoS One 2018; 13:e0199461. [PMID: 29979707 PMCID: PMC6034806 DOI: 10.1371/journal.pone.0199461] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 06/07/2018] [Indexed: 12/16/2022] Open
Abstract
A chronic inflammatory state to a large extent explains sickle cell disease (SCD) pathophysiology. Nonetheless, the principal dysregulated factors affecting this major pathway and their mechanisms of action still have to be fully identified and elucidated. Integrating gene expression and genome-wide association study (GWAS) data analysis represents a novel approach to refining the identification of key mediators and functions in complex diseases. Here, we performed gene expression meta-analysis of five independent publicly available microarray datasets related to homozygous SS patients with SCD to identify a consensus SCD transcriptomic profile. The meta-analysis conducted using the MetaDE R package based on combining p values (maxP approach) identified 335 differentially expressed genes (DEGs; 224 upregulated and 111 downregulated). Functional gene set enrichment revealed the importance of several metabolic pathways, of innate immune responses, erythrocyte development, and hemostasis pathways. Advanced analyses of GWAS data generated within the framework of this study by means of the atSNP R package and SIFT tool identified 60 regulatory single-nucleotide polymorphisms (rSNPs) occurring in the promoter of 20 DEGs and a deleterious SNP, affecting CAMKK2 protein function. This novel database of candidate genes, transcription factors, and rSNPs associated with SCD provides new markers that may help to identify new therapeutic targets.
Collapse
Affiliation(s)
- Cherif Ben Hamda
- Laboratory of Bioinformatics, Biomathematics and Biostatistics, Institute Pasteur of Tunis, Tunis, Tunisia
- University of Tunis El Manar, Tunis, Tunisia
- Faculty of Science of Bizerte, Jarzouna, University of Carthage, Tunisia
- * E-mail: (KG); (CBH)
| | - Raphael Sangeda
- Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Liberata Mwita
- Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | | | - Siana Nkya
- Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Sumir Panji
- University of Cape Town, Cape Town, South Africa
| | | | - Lamia Guizani-Tabbane
- University of Tunis El Manar, Tunis, Tunisia
- Laboratory of Medical Parasitology, Biotechnology and Biomolecules, Institute Pasteur of Tunis, Tunis, Tunisia
| | - Alia Benkahla
- Laboratory of Bioinformatics, Biomathematics and Biostatistics, Institute Pasteur of Tunis, Tunis, Tunisia
- University of Tunis El Manar, Tunis, Tunisia
| | - Julie Makani
- Faculty of Science of Bizerte, Jarzouna, University of Carthage, Tunisia
| | - Kais Ghedira
- Laboratory of Bioinformatics, Biomathematics and Biostatistics, Institute Pasteur of Tunis, Tunis, Tunisia
- University of Tunis El Manar, Tunis, Tunisia
- * E-mail: (KG); (CBH)
| | | |
Collapse
|
44
|
Abstract
The frequency of prediabetes is increasing as the prevalence of obesity rises worldwide. In prediabetes, hyperglycemia, insulin resistance, and inflammation and metabolic derangements associated with concomitant obesity cause endothelial vasodilator and fibrinolytic dysfunction, leading to increased risk of cardiovascular and renal disease. Importantly, the microvasculature affects insulin sensitivity by affecting the delivery of insulin and glucose to skeletal muscle; thus, endothelial dysfunction and extracellular matrix remodeling promote the progression from prediabetes to diabetes mellitus. Weight loss is the mainstay of treatment in prediabetes, but therapies that improved endothelial function and vasodilation may not only prevent cardiovascular disease but also slow progression to diabetes mellitus.
Collapse
Affiliation(s)
- David H Wasserman
- From the Departments of Molecular Physiology and Biophysics (D.H.W.) and Medicine (T.J.W., N.J.B.), Vanderbilt University Medical Center, Nashville, TN
| | - Thomas J Wang
- From the Departments of Molecular Physiology and Biophysics (D.H.W.) and Medicine (T.J.W., N.J.B.), Vanderbilt University Medical Center, Nashville, TN
| | - Nancy J Brown
- From the Departments of Molecular Physiology and Biophysics (D.H.W.) and Medicine (T.J.W., N.J.B.), Vanderbilt University Medical Center, Nashville, TN.
| |
Collapse
|
45
|
Rho JG, Han HS, Han JH, Lee H, Nguyen VQ, Lee WH, Kwon S, Heo S, Yoon J, Shin HH, Lee EY, Kang H, Yang S, Lee EK, Park JH, Kim W. Self-assembled hyaluronic acid nanoparticles: Implications as a nanomedicine for treatment of type 2 diabetes. J Control Release 2018; 279:89-98. [PMID: 29649530 DOI: 10.1016/j.jconrel.2018.04.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 03/28/2018] [Accepted: 04/05/2018] [Indexed: 01/13/2023]
Abstract
Self-assembled hyaluronic acid nanoparticles (HA-NPs) have been extensively investigated for biomedical and pharmaceutical applications owing to their biocompatibility and receptor-binding properties. Here, we report that an empty HA-NP itself not bearing any drug has therapeutic effects on adipose tissue inflammation and insulin resistance. HA-NPs inhibited not only the receptor-mediated internalization of low-molecular-weight (LMW) free HA but also LMW free HA-induced pro-inflammatory gene expression in mouse primary bone marrow-derived macrophages (BMDMs) isolated from wild-type mice, but not in CD44-null (CD44-/-) BMDMs. An in vivo biodistribution study showed the distribution of HA-NPs and their co-localization with CD44 in adipose tissues including epididymal white adipose tissues (eWATs), but these were rarely observed in the eWATs of CD44-/- mice. In addition, CD44 expression and HA-NP accumulation in the eWATs were increased in mice with diet-induced obesity (DIO) compared to lean mice. Interestingly, treatment with HA-NPs in DIO mice suppressed adipose tissue inflammation as indicated by reduced macrophage content, the production of proinflammatory cytokines and NLRP3 inflammasome activity in eWATs, leading to improved insulin sensitivity and normalized blood glucose levels. Collectively, these results suggest that an empty HA-NP itself can be a therapeutic agent for the treatment of type 2 diabetes.
Collapse
Affiliation(s)
- Jun Gi Rho
- Department of Molecular Science & Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Hwa Seung Han
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Ji Hye Han
- Department of Molecular Science & Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Hansang Lee
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Van Quy Nguyen
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Wang Hee Lee
- Department of Molecular Science & Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Seunglee Kwon
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Sungeun Heo
- Department of Molecular Science & Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Juhwan Yoon
- Department of Molecular Science & Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Han Ho Shin
- Department of Molecular Science & Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Eun-Young Lee
- Department of Molecular Science & Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Hoin Kang
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Siyoung Yang
- Department of Pharmacology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Eun Kyung Lee
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jae Hyung Park
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea; Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Wook Kim
- Department of Molecular Science & Technology, Ajou University, Suwon 16499, Republic of Korea.
| |
Collapse
|
46
|
Solimena M, Schulte AM, Marselli L, Ehehalt F, Richter D, Kleeberg M, Mziaut H, Knoch KP, Parnis J, Bugliani M, Siddiq A, Jörns A, Burdet F, Liechti R, Suleiman M, Margerie D, Syed F, Distler M, Grützmann R, Petretto E, Moreno-Moral A, Wegbrod C, Sönmez A, Pfriem K, Friedrich A, Meinel J, Wollheim CB, Baretton GB, Scharfmann R, Nogoceke E, Bonifacio E, Sturm D, Meyer-Puttlitz B, Boggi U, Saeger HD, Filipponi F, Lesche M, Meda P, Dahl A, Wigger L, Xenarios I, Falchi M, Thorens B, Weitz J, Bokvist K, Lenzen S, Rutter GA, Froguel P, von Bülow M, Ibberson M, Marchetti P. Systems biology of the IMIDIA biobank from organ donors and pancreatectomised patients defines a novel transcriptomic signature of islets from individuals with type 2 diabetes. Diabetologia 2018; 61:641-657. [PMID: 29185012 PMCID: PMC5803296 DOI: 10.1007/s00125-017-4500-3] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 08/29/2017] [Indexed: 01/25/2023]
Abstract
AIMS/HYPOTHESIS Pancreatic islet beta cell failure causes type 2 diabetes in humans. To identify transcriptomic changes in type 2 diabetic islets, the Innovative Medicines Initiative for Diabetes: Improving beta-cell function and identification of diagnostic biomarkers for treatment monitoring in Diabetes (IMIDIA) consortium ( www.imidia.org ) established a comprehensive, unique multicentre biobank of human islets and pancreas tissues from organ donors and metabolically phenotyped pancreatectomised patients (PPP). METHODS Affymetrix microarrays were used to assess the islet transcriptome of islets isolated either by enzymatic digestion from 103 organ donors (OD), including 84 non-diabetic and 19 type 2 diabetic individuals, or by laser capture microdissection (LCM) from surgical specimens of 103 PPP, including 32 non-diabetic, 36 with type 2 diabetes, 15 with impaired glucose tolerance (IGT) and 20 with recent-onset diabetes (<1 year), conceivably secondary to the pancreatic disorder leading to surgery (type 3c diabetes). Bioinformatics tools were used to (1) compare the islet transcriptome of type 2 diabetic vs non-diabetic OD and PPP as well as vs IGT and type 3c diabetes within the PPP group; and (2) identify transcription factors driving gene co-expression modules correlated with insulin secretion ex vivo and glucose tolerance in vivo. Selected genes of interest were validated for their expression and function in beta cells. RESULTS Comparative transcriptomic analysis identified 19 genes differentially expressed (false discovery rate ≤0.05, fold change ≥1.5) in type 2 diabetic vs non-diabetic islets from OD and PPP. Nine out of these 19 dysregulated genes were not previously reported to be dysregulated in type 2 diabetic islets. Signature genes included TMEM37, which inhibited Ca2+-influx and insulin secretion in beta cells, and ARG2 and PPP1R1A, which promoted insulin secretion. Systems biology approaches identified HNF1A, PDX1 and REST as drivers of gene co-expression modules correlated with impaired insulin secretion or glucose tolerance, and 14 out of 19 differentially expressed type 2 diabetic islet signature genes were enriched in these modules. None of these signature genes was significantly dysregulated in islets of PPP with impaired glucose tolerance or type 3c diabetes. CONCLUSIONS/INTERPRETATION These studies enabled the stringent definition of a novel transcriptomic signature of type 2 diabetic islets, regardless of islet source and isolation procedure. Lack of this signature in islets from PPP with IGT or type 3c diabetes indicates differences possibly due to peculiarities of these hyperglycaemic conditions and/or a role for duration and severity of hyperglycaemia. Alternatively, these transcriptomic changes capture, but may not precede, beta cell failure.
Collapse
Affiliation(s)
- Michele Solimena
- Paul Langerhans Institute Dresden (PLID) of the Helmholtz Center Munich at University Hospital Carl Gustav Carus and Faculty of Medicine, TU Dresden, Fetscherstrasse 74, 01307, Dresden, Germany.
- German Center for Diabetes Research (DZD), Munich Neuherberg, Germany.
- Max Planck Institute of Molecular Cell Biology and Genetics (MPI-CBG), 01307, Dresden, Germany.
| | - Anke M Schulte
- Sanofi-Aventis Deutschland GmbH, Diabetes Research, Industriepark Höchst, Building H821, 65926, Frankfurt am Main, Germany.
| | - Lorella Marselli
- Department of Clinical and Experimental Medicine, Cisanello University Hospital, University of Pisa, Via Paradisa 2, 56126, Pisa, Italy
| | - Florian Ehehalt
- Paul Langerhans Institute Dresden (PLID) of the Helmholtz Center Munich at University Hospital Carl Gustav Carus and Faculty of Medicine, TU Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
- German Center for Diabetes Research (DZD), Munich Neuherberg, Germany
- Department of Visceral-Thoracic-Vascular Surgery, University Hospital Carl Gustav Carus and Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Daniela Richter
- Paul Langerhans Institute Dresden (PLID) of the Helmholtz Center Munich at University Hospital Carl Gustav Carus and Faculty of Medicine, TU Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
- German Center for Diabetes Research (DZD), Munich Neuherberg, Germany
| | - Manuela Kleeberg
- Paul Langerhans Institute Dresden (PLID) of the Helmholtz Center Munich at University Hospital Carl Gustav Carus and Faculty of Medicine, TU Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
- German Center for Diabetes Research (DZD), Munich Neuherberg, Germany
- Department of Visceral-Thoracic-Vascular Surgery, University Hospital Carl Gustav Carus and Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Hassan Mziaut
- Paul Langerhans Institute Dresden (PLID) of the Helmholtz Center Munich at University Hospital Carl Gustav Carus and Faculty of Medicine, TU Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
- German Center for Diabetes Research (DZD), Munich Neuherberg, Germany
| | - Klaus-Peter Knoch
- Paul Langerhans Institute Dresden (PLID) of the Helmholtz Center Munich at University Hospital Carl Gustav Carus and Faculty of Medicine, TU Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
- German Center for Diabetes Research (DZD), Munich Neuherberg, Germany
| | - Julia Parnis
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, UK
| | - Marco Bugliani
- Department of Clinical and Experimental Medicine, Cisanello University Hospital, University of Pisa, Via Paradisa 2, 56126, Pisa, Italy
| | - Afshan Siddiq
- Queen Mary University of London, Dawson Hall, London, UK
- Department of Genomics of Common Disease, School of Public Health, Imperial College London, Hammersmith Hospital, London, UK
| | - Anne Jörns
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Frédéric Burdet
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, Quartier Sorge, bâtiment Génopode, 1015, Lausanne, Switzerland
| | - Robin Liechti
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, Quartier Sorge, bâtiment Génopode, 1015, Lausanne, Switzerland
| | - Mara Suleiman
- Department of Clinical and Experimental Medicine, Cisanello University Hospital, University of Pisa, Via Paradisa 2, 56126, Pisa, Italy
| | - Daniel Margerie
- Sanofi-Aventis Deutschland GmbH, Diabetes Research, Industriepark Höchst, Building H821, 65926, Frankfurt am Main, Germany
| | - Farooq Syed
- Department of Clinical and Experimental Medicine, Cisanello University Hospital, University of Pisa, Via Paradisa 2, 56126, Pisa, Italy
| | - Marius Distler
- Department of Visceral-Thoracic-Vascular Surgery, University Hospital Carl Gustav Carus and Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Robert Grützmann
- Department of Surgery, University Hospital of Erlangen, Erlangen, Germany
| | - Enrico Petretto
- Medical Research Council (MRC), Institute of Medical Sciences, Imperial College London, London, UK
- Duke-NUS Medical School, Singapore, Republic of Singapore
| | - Aida Moreno-Moral
- Medical Research Council (MRC), Institute of Medical Sciences, Imperial College London, London, UK
- Duke-NUS Medical School, Singapore, Republic of Singapore
| | - Carolin Wegbrod
- Paul Langerhans Institute Dresden (PLID) of the Helmholtz Center Munich at University Hospital Carl Gustav Carus and Faculty of Medicine, TU Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
- German Center for Diabetes Research (DZD), Munich Neuherberg, Germany
| | - Anke Sönmez
- Paul Langerhans Institute Dresden (PLID) of the Helmholtz Center Munich at University Hospital Carl Gustav Carus and Faculty of Medicine, TU Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
- German Center for Diabetes Research (DZD), Munich Neuherberg, Germany
| | - Katja Pfriem
- Paul Langerhans Institute Dresden (PLID) of the Helmholtz Center Munich at University Hospital Carl Gustav Carus and Faculty of Medicine, TU Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
- German Center for Diabetes Research (DZD), Munich Neuherberg, Germany
| | - Anne Friedrich
- Paul Langerhans Institute Dresden (PLID) of the Helmholtz Center Munich at University Hospital Carl Gustav Carus and Faculty of Medicine, TU Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
- German Center for Diabetes Research (DZD), Munich Neuherberg, Germany
| | - Jörn Meinel
- Department of Pathology, University Hospital Carl Gustav Carus and Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Claes B Wollheim
- Department of Cell Physiology and Metabolism, Geneva University Medical Center, Geneva, Switzerland
| | - Gustavo B Baretton
- Department of Pathology, University Hospital Carl Gustav Carus and Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Raphael Scharfmann
- INSERM, U1016, Institut Cochin, Faculté de Médecine, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Everson Nogoceke
- F. Hoffmann-La Roche Ltd, Roche Innovation Center Basel, Basel, Switzerland
| | - Ezio Bonifacio
- Paul Langerhans Institute Dresden (PLID) of the Helmholtz Center Munich at University Hospital Carl Gustav Carus and Faculty of Medicine, TU Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
- German Center for Diabetes Research (DZD), Munich Neuherberg, Germany
- Center for Regenerative Therapies Dresden (CRTD), TU Dresden, Dresden, Germany
| | - Dorothée Sturm
- Paul Langerhans Institute Dresden (PLID) of the Helmholtz Center Munich at University Hospital Carl Gustav Carus and Faculty of Medicine, TU Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
- German Center for Diabetes Research (DZD), Munich Neuherberg, Germany
- Department of Visceral-Thoracic-Vascular Surgery, University Hospital Carl Gustav Carus and Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Birgit Meyer-Puttlitz
- Sanofi-Aventis Deutschland GmbH, Diabetes Research, Industriepark Höchst, Building H821, 65926, Frankfurt am Main, Germany
| | - Ugo Boggi
- Department of Clinical and Experimental Medicine, Cisanello University Hospital, University of Pisa, Via Paradisa 2, 56126, Pisa, Italy
| | - Hans-Detlev Saeger
- Paul Langerhans Institute Dresden (PLID) of the Helmholtz Center Munich at University Hospital Carl Gustav Carus and Faculty of Medicine, TU Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
- German Center for Diabetes Research (DZD), Munich Neuherberg, Germany
- Department of Visceral-Thoracic-Vascular Surgery, University Hospital Carl Gustav Carus and Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Franco Filipponi
- Department of Clinical and Experimental Medicine, Cisanello University Hospital, University of Pisa, Via Paradisa 2, 56126, Pisa, Italy
| | | | - Paolo Meda
- Department of Cell Physiology and Metabolism, Geneva University Medical Center, Geneva, Switzerland
| | - Andreas Dahl
- Biotechnology Center, TU Dresden, Dresden, Germany
| | - Leonore Wigger
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, Quartier Sorge, bâtiment Génopode, 1015, Lausanne, Switzerland
| | - Ioannis Xenarios
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, Quartier Sorge, bâtiment Génopode, 1015, Lausanne, Switzerland
| | - Mario Falchi
- Department of Genomics of Common Disease, School of Public Health, Imperial College London, Hammersmith Hospital, London, UK
| | - Bernard Thorens
- Centre for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Jürgen Weitz
- Paul Langerhans Institute Dresden (PLID) of the Helmholtz Center Munich at University Hospital Carl Gustav Carus and Faculty of Medicine, TU Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
- German Center for Diabetes Research (DZD), Munich Neuherberg, Germany
- Department of Visceral-Thoracic-Vascular Surgery, University Hospital Carl Gustav Carus and Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Krister Bokvist
- Lilly Research Laboratories, Eli Lilly, Indianapolis, IN, USA
| | - Sigurd Lenzen
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, UK
| | - Philippe Froguel
- Department of Genomics of Common Disease, School of Public Health, Imperial College London, Hammersmith Hospital, London, UK
- CNRS-UMR8199, Lille Pasteur Institute, Lille, France
- Lille University Hospital, Lille, France
- European Genomic Institute for Diabetes (EGID), Lille, France
| | - Manon von Bülow
- Sanofi-Aventis Deutschland GmbH, Diabetes Research, Industriepark Höchst, Building H821, 65926, Frankfurt am Main, Germany
| | - Mark Ibberson
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, Quartier Sorge, bâtiment Génopode, 1015, Lausanne, Switzerland.
| | - Piero Marchetti
- Department of Clinical and Experimental Medicine, Cisanello University Hospital, University of Pisa, Via Paradisa 2, 56126, Pisa, Italy.
| |
Collapse
|
47
|
Patouraux S, Rousseau D, Bonnafous S, Lebeaupin C, Luci C, Canivet CM, Schneck AS, Bertola A, Saint-Paul MC, Iannelli A, Gugenheim J, Anty R, Tran A, Bailly-Maitre B, Gual P. CD44 is a key player in non-alcoholic steatohepatitis. J Hepatol 2017; 67:328-338. [PMID: 28323124 DOI: 10.1016/j.jhep.2017.03.003] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 02/01/2017] [Accepted: 03/02/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Cluster of differentiation (CD)44 regulates adipose tissue inflammation in obesity and hepatic leukocyte recruitment in a lithogenic context. However, its role in hepatic inflammation in a mouse model of steatohepatitis and its relevance in humans have not yet been investigated. We aimed to evaluated the contribution of CD44 to non-alcoholic steatohepatitis (NASH) development and liver injury in mouse models and in patients at various stages of non-alcoholic fatty liver disease (NAFLD) progression. METHODS The role of CD44 was evaluated in CD44-/- mice and after injections of an αCD44 antibody in wild-type mice challenged with a methionine- and choline-deficient diet (MCDD). In obese patients, hepatic CD44 (n=30 and 5 NASH patients with a second liver biopsy after bariatric surgery) and serum sCD44 (n=64) were evaluated. RESULTS Liver inflammation (including inflammatory foci number, macrophage and neutrophil infiltration and CCL2/CCR2 levels), liver injury and fibrosis strongly decreased in CD44-/- mice compared to wild-type mice on MCDD. CD44 deficiency enhanced the M2 polarization and strongly decreased the activation of macrophages by lipopolysaccharide (LPS), hepatocyte damage-associated molecular patterns (DAMPs) and saturated fatty acids. Neutralization of CD44 in mice with steatohepatitis strongly decreased the macrophage infiltration and chemokine ligand (CCL)2 expression with a partial correction of liver inflammation and injury. In obese patients, hepatic CD44 was strongly upregulated in NASH patients (p=0.0008) and correlated with NAFLD activity score (NAS) (p=0.001), ballooning (p=0.003), alanine transaminase (p=0.005) and hepatic CCL2 (p<0.001) and macrophage marker CD68 (p<0.001) expression. Correction of NASH was associated with a strong decrease in liver CD44+ cells. Finally, the soluble form of CD44 increased with severe steatosis (p=0.0005) and NASH (p=0.007). CONCLUSION Human and experimental data suggest that CD44 is a marker and key player of hepatic inflammation and its targeting partially corrects NASH. LAY SUMMARY Human and experimental data suggest that CD44, a cellular protein mainly expressed in immune cells, is a marker and key player of non-alcoholic steatohepatitis (NASH). Indeed, CD44 enhances the non-alcoholic fatty liver (NAFL) (hepatic steatosis) to NASH progression by regulating hepatic macrophage polarization (pro-inflammatory phenotype) and infiltration (macrophage motility and the MCP1/CCL2/CCR2 system). Targeting CD44 partially corrects NASH, making it a potential therapeutic strategy.
Collapse
Affiliation(s)
- Stéphanie Patouraux
- INSERM, U1065, C3M, Team 8 "Hepatic Complications in Obesity", Nice, France; Université Côte d'Azur, Nice, France; CHU of Nice, Biological Center, Pasteur Hôpital, Nice, France
| | - Déborah Rousseau
- INSERM, U1065, C3M, Team 8 "Hepatic Complications in Obesity", Nice, France; Université Côte d'Azur, Nice, France
| | - Stéphanie Bonnafous
- INSERM, U1065, C3M, Team 8 "Hepatic Complications in Obesity", Nice, France; Université Côte d'Azur, Nice, France; CHU of Nice, Digestive Center, Nice, France
| | - Cynthia Lebeaupin
- INSERM, U1065, C3M, Team 8 "Hepatic Complications in Obesity", Nice, France; Université Côte d'Azur, Nice, France
| | - Carmelo Luci
- INSERM, U1065, C3M, Team 8 "Hepatic Complications in Obesity", Nice, France; Université Côte d'Azur, Nice, France
| | - Clémence M Canivet
- INSERM, U1065, C3M, Team 8 "Hepatic Complications in Obesity", Nice, France; Université Côte d'Azur, Nice, France; CHU of Nice, Digestive Center, Nice, France
| | - Anne-Sophie Schneck
- INSERM, U1065, C3M, Team 8 "Hepatic Complications in Obesity", Nice, France; Université Côte d'Azur, Nice, France; CHU of Nice, Digestive Center, Nice, France
| | - Adeline Bertola
- INSERM, U1065, C3M, Team 8 "Hepatic Complications in Obesity", Nice, France; Université Côte d'Azur, Nice, France
| | - Marie-Christine Saint-Paul
- INSERM, U1065, C3M, Team 8 "Hepatic Complications in Obesity", Nice, France; Université Côte d'Azur, Nice, France; CHU of Nice, Biological Center, Pasteur Hôpital, Nice, France
| | - Antonio Iannelli
- INSERM, U1065, C3M, Team 8 "Hepatic Complications in Obesity", Nice, France; Université Côte d'Azur, Nice, France; CHU of Nice, Digestive Center, Nice, France
| | - Jean Gugenheim
- INSERM, U1065, C3M, Team 8 "Hepatic Complications in Obesity", Nice, France; Université Côte d'Azur, Nice, France; CHU of Nice, Digestive Center, Nice, France
| | - Rodolphe Anty
- INSERM, U1065, C3M, Team 8 "Hepatic Complications in Obesity", Nice, France; Université Côte d'Azur, Nice, France; CHU of Nice, Digestive Center, Nice, France
| | - Albert Tran
- INSERM, U1065, C3M, Team 8 "Hepatic Complications in Obesity", Nice, France; Université Côte d'Azur, Nice, France; CHU of Nice, Digestive Center, Nice, France
| | - Béatrice Bailly-Maitre
- INSERM, U1065, C3M, Team 8 "Hepatic Complications in Obesity", Nice, France; Université Côte d'Azur, Nice, France
| | - Philippe Gual
- INSERM, U1065, C3M, Team 8 "Hepatic Complications in Obesity", Nice, France; Université Côte d'Azur, Nice, France.
| |
Collapse
|
48
|
Bahrami SB, Tolg C, Peart T, Symonette C, Veiseh M, Umoh JU, Holdsworth DW, McCarthy JB, Luyt LG, Bissell MJ, Yazdani A, Turley EA. Receptor for hyaluronan mediated motility (RHAMM/HMMR) is a novel target for promoting subcutaneous adipogenesis. Integr Biol (Camb) 2017; 9:223-237. [PMID: 28217782 DOI: 10.1039/c7ib00002b] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Hyaluronan, CD44 and the Receptor for Hyaluronan-Mediated Motility (RHAMM, gene name HMMR) regulate stem cell differentiation including mesenchymal progenitor differentiation. Here, we show that CD44 expression is required for subcutaneous adipogenesis, whereas RHAMM expression suppresses this process. We designed RHAMM function blocking peptides to promote subcutaneous adipogenesis as a clinical and tissue engineering tool. Adipogenic RHAMM peptides were identified by screening for their ability to promote adipogenesis in culture assays using rat bone marrow mesenchymal stem cells, mouse pre-adipocyte cell lines and primary human subcutaneous pre-adipocytes. Oil red O uptake into fat droplets and adiponectin production were used as biomarkers of adipogenesis. Positive peptides were formulated in either collagen I or hyaluronan (Orthovisc) gels then assessed for their adipogenic potential in vivo following injection into dorsal rat skin and mammary fat pads. Fat content was quantified and characterized using micro CT imaging, morphometry, histology, RT-PCR and ELISA analyses of adipogenic gene expression. Injection of screened peptides increased dorsal back subcutaneous fat pad area (208.3 ± 10.4 mm2versus control 84.11 ± 4.2 mm2; p < 0.05) and mammary fat pad size (45 ± 11 mg above control background, p = 0.002) in female rats. This effect lasted >5 weeks as detected by micro CT imaging and perilipin 1 mRNA expression. RHAMM expression suppresses while blocking peptides promote expression of PPARγ, C/EBP and their target genes. Blocking RHAMM function by peptide injection or topical application is a novel and minimally invasive method for potentially promoting subcutaneous adipogenesis in lipodystrophic diseases and a complementary tool to subcutaneous fat augmentation techniques.
Collapse
Affiliation(s)
- S B Bahrami
- Biological Systems and Engineering Division, BioSciences Area, Lawrence Berkeley National Laboratories, 977R225A, Berkeley, CA 94720, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Mansego ML, Garcia-Lacarte M, Milagro FI, Marti A, Martinez JA. DNA methylation of miRNA coding sequences putatively associated with childhood obesity. Pediatr Obes 2017; 12:19-27. [PMID: 26780939 DOI: 10.1111/ijpo.12101] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 12/04/2015] [Accepted: 12/09/2015] [Indexed: 01/08/2023]
Abstract
BACKGROUND Epigenetic mechanisms may be involved in obesity onset and its consequences. The aim of the present study was to evaluate whether DNA methylation status in microRNA (miRNA) coding regions is associated with childhood obesity. MATERIAL AND METHODS DNA isolated from white blood cells of 24 children (identification sample: 12 obese and 12 non-obese) from the Grupo Navarro de Obesidad Infantil study was hybridized in a 450 K methylation microarray. Several CpGs whose DNA methylation levels were statistically different between obese and non-obese were validated by MassArray® in 95 children (validation sample) from the same study. RESULTS Microarray analysis identified 16 differentially methylated CpGs between both groups (6 hypermethylated and 10 hypomethylated). DNA methylation levels in miR-1203, miR-412 and miR-216A coding regions significantly correlated with body mass index standard deviation score (BMI-SDS) and explained up to 40% of the variation of BMI-SDS. The network analysis identified 19 well-defined obesity-relevant biological pathways from the KEGG database. MassArray® validation identified three regions located in or near miR-1203, miR-412 and miR-216A coding regions differentially methylated between obese and non-obese children. CONCLUSIONS The current work identified three CpG sites located in coding regions of three miRNAs (miR-1203, miR-412 and miR-216A) that were differentially methylated between obese and non-obese children, suggesting a role of miRNA epigenetic regulation in childhood obesity.
Collapse
Affiliation(s)
- M L Mansego
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Spain.,Nutrition Research Center, University of Navarra, Pamplona, Spain.,CIBERobn, Fisiopatología de la Obesidad y la Nutrición, Carlos III Institute of Health, Madrid, Spain
| | - M Garcia-Lacarte
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Spain.,Nutrition Research Center, University of Navarra, Pamplona, Spain
| | - F I Milagro
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Spain.,Nutrition Research Center, University of Navarra, Pamplona, Spain.,CIBERobn, Fisiopatología de la Obesidad y la Nutrición, Carlos III Institute of Health, Madrid, Spain
| | - A Marti
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Spain.,CIBERobn, Fisiopatología de la Obesidad y la Nutrición, Carlos III Institute of Health, Madrid, Spain.,IdiSNA, Navarra's Health Research Institute, Pamplona, Spain
| | - J A Martinez
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Spain.,Nutrition Research Center, University of Navarra, Pamplona, Spain.,CIBERobn, Fisiopatología de la Obesidad y la Nutrición, Carlos III Institute of Health, Madrid, Spain.,IdiSNA, Navarra's Health Research Institute, Pamplona, Spain
| | | |
Collapse
|
50
|
Marycz K, Kornicka K, Marędziak M, Golonka P, Nicpoń J. Equine metabolic syndrome impairs adipose stem cells osteogenic differentiation by predominance of autophagy over selective mitophagy. J Cell Mol Med 2016; 20:2384-2404. [PMID: 27629697 PMCID: PMC5134411 DOI: 10.1111/jcmm.12932] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 05/28/2016] [Indexed: 12/12/2022] Open
Abstract
Adipose‐derived mesenchymal stem cells (ASC) hold great promise in the treatment of many disorders including musculoskeletal system, cardiovascular and/or endocrine diseases. However, the cytophysiological condition of cells, used for engraftment seems to be fundamental factor that might determine the effectiveness of clinical therapy. In this study we investigated growth kinetics, senescence, accumulation of oxidative stress factors, mitochondrial biogenesis, autophagy and osteogenic differentiation potential of ASC isolated from horses suffered from equine metabolic syndrome (EMS). We demonstrated that EMS condition impairs multipotency/pluripotency in ASCs causes accumulation of reactive oxygen species and mitochondria deterioration. We found that, cytochrome c is released from mitochondria to the cytoplasm suggesting activation of intrinsic apoptotic pathway in those cells. Moreover, we observed up‐regulation of p21 and decreased ratio of Bcl‐2/BAX. Deteriorations in mitochondria structure caused alternations in osteogenic differentiation of ASCEMS resulting in their decreased proliferation rate and reduced expression of osteogenic markers BMP‐2 and collagen type I. During osteogenic differentiation of ASCEMS, we observed autophagic turnover as probably, an alternative way to generate adenosine triphosphate and amino acids required to increased protein synthesis during differentiation. Downregulation of PGC1α, PARKIN and PDK4 in differentiated ASCEMS confirmed impairments in mitochondrial biogenesis and function. Hence, application of ASCEMS into endocrinological or ortophedical practice requires further investigation and analysis in the context of safeness of their application.
Collapse
Affiliation(s)
- Krzysztof Marycz
- Electron Microscopy Laboratory, The Faculty of Biology and Animal Science, University of Environmental and Life Sciences Wroclaw, Wroclaw, Poland.,Wroclaw Research Centre EIT+, Wrocław, Poland
| | - Katarzyna Kornicka
- Electron Microscopy Laboratory, The Faculty of Biology and Animal Science, University of Environmental and Life Sciences Wroclaw, Wroclaw, Poland.,Wroclaw Research Centre EIT+, Wrocław, Poland
| | - Monika Marędziak
- Department of Animal Physiology and Biostructure, Faculty of Veterinary Medicine, University of Environmental and Life Sciences Wroclaw, Wroclaw, Poland
| | | | - Jakub Nicpoń
- Department of Surgery, Faculty of Veterinary Medicine, University of Environmental and Life Sciences Wroclaw, Wroclaw, Poland
| |
Collapse
|