51
|
Xu H, Liu S, Wang Y, Wu R, Yi T, Wang T, Zhu Y, Fang J, Xie Y, Zhao Q, Song X, Chen J, Rajagopaplan S, Brook RD, Li J, Cao J, Huang W. The mediating role of vascular inflammation in traffic-related air pollution associated changes in insulin resistance in healthy adults. Int J Hyg Environ Health 2021; 239:113878. [PMID: 34757311 DOI: 10.1016/j.ijheh.2021.113878] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 02/06/2023]
Abstract
AIM The precise pathophysiologic pathway linking traffic-related air pollution (TRAP) to diabetes mellitus is not well elucidated. We aimed to investigate whether activation of vascular inflammation can be a mechanistic linkage between ambient TRAP and insulin resistance. METHODS Study outcomes were determined by assessing a series of circulating biomarkers indicative of insulin resistance and vascular inflammation among 73 healthy adults who underwent repeated clinical visits in Beijing, China, 2014-2016. Concomitantly, concentrations of ambient TRAP indices, including particulate matter in diameter <2.5 μm (PM2.5), particles in size fractions of 5-560 nm, black carbon, carbon monoxide, nitrogen dioxide, and oxides of nitrogen, were continuously monitored. RESULTS Participants experienced extremely high levels of TRAP exposures, with mean (standard deviation) PM2.5 concentrations of 91.8 (48.3) μg/m3, throughout the study. We found that interquartile range increases in exposure to moving average concentrations of various TRAP indices at prior up to 7 days were associated with significant elevations of 8.9-49.6% in insulin levels. Higher pollutant levels were also related to worsening metrics of insulin resistance (soluble insulin receptor ectodomain, adipokines, and homeostasis model assessment of insulin resistance) and heightened vascular inflammatory responses, particularly disruptions of the receptor activator of nuclear factor κB ligand/osteoprotegerin system balance and elevations of monocyte/macrophage and T cell activation markers. Mediation analyses showed that activation of vascular inflammation could explain up to 66% of the alterations in metrics of insulin resistance attributable to air pollution. CONCLUSION Our results suggest that ambient traffic pollution exposure was capable of promoting insulin resistance possibly via generating vascular inflammation.
Collapse
Affiliation(s)
- Hongbing Xu
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, And Peking University Institute of Environmental Medicine, Beijing, China; Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Beijing, China
| | - Shengcong Liu
- Division of Cardiology, Peking University First Hospital, Beijing, China; Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Beijing, China
| | - Yang Wang
- Department of Prevention and Health Care, Hospital of Health Science Center, Peking University, Beijing, China
| | - Rongshan Wu
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, And Peking University Institute of Environmental Medicine, Beijing, China; State Key Laboratory of Environmental Criteria and Risk Assessment, State Environmental Protection Key Laboratory of Ecological Effect and Risk Assessment of Chemicals, Chinese Research Academy of Environmental Sciences, Beijing, China
| | - Tieci Yi
- Division of Cardiology, Peking University First Hospital, Beijing, China; Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Beijing, China
| | - Tong Wang
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, And Peking University Institute of Environmental Medicine, Beijing, China; Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Beijing, China
| | - Yutong Zhu
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, And Peking University Institute of Environmental Medicine, Beijing, China; Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Beijing, China
| | - Jiakun Fang
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, And Peking University Institute of Environmental Medicine, Beijing, China; Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Beijing, China
| | - Yunfei Xie
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, And Peking University Institute of Environmental Medicine, Beijing, China; Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Beijing, China
| | - Qian Zhao
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, And Peking University Institute of Environmental Medicine, Beijing, China; Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Beijing, China
| | - Xiaoming Song
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, And Peking University Institute of Environmental Medicine, Beijing, China; Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Beijing, China
| | - Jie Chen
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, And Peking University Institute of Environmental Medicine, Beijing, China; Institute for Risk Assessment Sciences, University Medical Centre Utrecht, University of Utrecht, the Netherlands
| | - Sanjay Rajagopaplan
- Division of Cardiovascular Medicine, Case Western Reserve University, Ohio, USA
| | - Robert D Brook
- Division of Cardiovascular Medicine, University of Michigan, Michigan, USA
| | - Jianping Li
- Division of Cardiology, Peking University First Hospital, Beijing, China; Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Beijing, China
| | - Junji Cao
- Institute of Atmospheric Physics Chinese Academy of Sciences, Beijing, China.
| | - Wei Huang
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, And Peking University Institute of Environmental Medicine, Beijing, China; Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Beijing, China.
| |
Collapse
|
52
|
Bernardi O, Estienne A, Reverchon M, Bigot Y, Froment P, Dupont J. Adipokines in metabolic and reproductive functions in birds: An overview of current knowns and unknowns. Mol Cell Endocrinol 2021; 534:111370. [PMID: 34171419 DOI: 10.1016/j.mce.2021.111370] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 06/04/2021] [Accepted: 06/14/2021] [Indexed: 01/09/2023]
Abstract
Adipose tissue is now recognized as an active endocrine organ, which synthesizes and secretes numerous peptides factors called adipokines. In mammals, they exert pleiotropic effects affecting energy metabolism but also fertility. In mammals, secretion of adipokines is altered in adipose tissue dysfunctions and may participate to obesity-associated disorders. Thus, adipokines are promising candidates both for novel pharmacological treatment strategies and as diagnostic tools. As compared to mammals, birds exhibit several unique physiological features, which make them an interesting model for comparative studies on endocrine control of metabolism and adiposity and reproductive functions. Some adipokines such as leptin and visfatin may have different roles in avian species as compared to mammals. In addition, some of them found in mammals such as CCL2 (chemokine ligand 2), resistin, omentin and FGF21 (Fibroblast Growth factor 21) have not yet been mapped to the chicken genome model and among its annotated gene models. This brief review aims to summarize data (structure, metabolic and reproductive roles and molecular mechanisms involved) related to main avian adipokines (leptin, adiponectin, visfatin, and chemerin) and we will briefly discuss the adipokines that are still lacking in avian species.
Collapse
Affiliation(s)
- Ophélie Bernardi
- CNRS, IFCE, INRAE, Université de Tours, PRC, F-37380, Nouzilly, France; SYSAAF-Syndicat des Sélectionneurs Avicoles et Aquacoles Français, Centre INRA Val de Loire, F-37380, Nouzilly, France
| | - Anthony Estienne
- CNRS, IFCE, INRAE, Université de Tours, PRC, F-37380, Nouzilly, France
| | - Maxime Reverchon
- SYSAAF-Syndicat des Sélectionneurs Avicoles et Aquacoles Français, Centre INRA Val de Loire, F-37380, Nouzilly, France
| | - Yves Bigot
- CNRS, IFCE, INRAE, Université de Tours, PRC, F-37380, Nouzilly, France
| | - Pascal Froment
- CNRS, IFCE, INRAE, Université de Tours, PRC, F-37380, Nouzilly, France
| | - Joëlle Dupont
- CNRS, IFCE, INRAE, Université de Tours, PRC, F-37380, Nouzilly, France.
| |
Collapse
|
53
|
HGF can reduce accumulation of inflammation and regulate glucose homeostasis in T2D mice. J Physiol Biochem 2021; 77:613-624. [PMID: 34363605 DOI: 10.1007/s13105-021-00828-7] [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: 09/23/2020] [Accepted: 07/09/2021] [Indexed: 10/20/2022]
Abstract
Hepatocyte growth factor (HGF) has been studied as a protective factor for the survival of islet β cells and regulatory glucose transport and metabolism in many studies. The addition of exogenous HGF to cells or mice is the most common way to study HGF, but the persistence and stability of the administered HGF are unclear. In this experiment, wild-type C57BL6 (WT) mice and HGF-overexpressing transgenic (HGF-Tg) mice were divided into a normal diet (ND) group and an HFD group. The HGF protein level in the liver, kidney, spleen, pancreas, and VAT of HGF-Tg-ND mice was upregulated compared to that of WT-ND mice, and it was also upregulated in HGF-Tg-HFD mice compared to that in WT-HFD mice. In the ND group, though the HGF-Tg-ND mice showed higher fasted blood glucose levels and larger integrated density (IOD) of glucagon-positive cells than WT-ND mice, we found that HGF-Tg-ND mice can still maintain normal glucose tolerance based on an intraperitoneal glucose tolerance test (IPGTT) and an intraperitoneal insulin tolerance test (IPITT). In the HFD group, the HGF-Tg-HFD mice showed insulin sensitivity in IPGTT and IPITT and had larger areas and higher IOD values of islet β cells and smaller areas and IOD values of islet α cells than the WT-HFD mice. HGF-Tg-HFD mice had lower level of serum insulin than WT-HFD mice. The HGF-Tg-HFD mice showed inhibited accumulation of CD4+ T cells, CD8+ T cells, Ly6G+ neutrophils, and F4/80+ macrophages in the blood and tissues and protected liver and kidney functions. Oil Red O-stained liver sections revealed that WT-HFD mice had larger areas and higher IOD values of Oil Red O-positive cells than HGF-Tg-HFD mice, and WT-HFD mice had higher score of NASH. PAS-stained kidney sections found WT-HFD has higher mesangial area/glomerular area × 100% than HGF-Tg-HFD mice. Comparative analyses demonstrated that HGF reduces the proportions of inflammatory cells in the blood and tissues, and protects liver and kidney tissues by regulating glucose homeostasis of type 2 diabetic mice.
Collapse
|
54
|
Li L, Lee J, Cho A, Kim JH, Ju W, An JN, Park JH, Zhu SM, Lee J, Yu SS, Lim CS, Kim DK, Kim YS, Yang SH, Lee JP. cMet agonistic antibody prevents acute kidney injury to chronic kidney disease transition by suppressing Smurf1 and activating Smad7. Clin Sci (Lond) 2021; 135:1427-1444. [PMID: 34061176 DOI: 10.1042/cs20210013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 05/19/2021] [Accepted: 06/01/2021] [Indexed: 11/17/2022]
Abstract
We aimed to investigate the role of cMet agonistic antibody (cMet Ab) in preventing kidney fibrosis during acute kidney injury (AKI) to chronic kidney disease (CKD) transition. Additionally, we explored the effect of cMet Ab on TGF-β1/Smad pathway during the pathogenesis of kidney fibrosis. A unilateral ischemia-reperfusion injury (UIRI) mouse model was established to induce AKI-to-CKD transition. Furthermore, we incubated human proximal tubular epithelial cells (hPTECs) under hypoxic conditions as in vitro model of kidney fibrosis. We analyzed the soluble plasma cMet level in patients with AKI requiring dialysis. Patients who did not recover kidney function and progressed to CKD presented a higher increase in the cMet level. The kidneys of mice treated with cMet Ab showed fewer contractions and weighed more than the controls. The mice in the cMet Ab-treated group showed reduced fibrosis and significantly decreased expression of fibronectin and α-smooth muscle actin. cMet Ab treatment decreased inflammatory markers (MCP-1, TNF-α, and IL-1β) expression, reduced Smurf1 and Smad2/3 level, and increased Smad7 expressions. cMet Ab treatment increased cMet expression and reduced the hypoxia-induced increase in collagen-1 and ICAM-1 expression, thereby reducing apoptosis in the in vitro cell model. After cMet Ab treatment, hypoxia-induced expression of Smurf1, Smad2/3, and TGF-β1 was reduced, and suppressed Smad7 was activated. Down-regulation of Smurf1 resulted in suppression of hypoxia-induced fibronectin expression, whereas treatment with cMet Ab showed synergistic effects. cMet Ab can successfully prevent fibrosis response in UIRI models of kidney fibrosis by decreasing inflammatory response and inhibiting the TGF-β1/Smad pathway.
Collapse
Affiliation(s)
- Lilin Li
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Intensive Care Unit, Yanbian University Hospital, Yanji, Jilin, China
| | - Jeonghwan Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Ara Cho
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Jin Hyuk Kim
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Wonmin Ju
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jung Nam An
- Department of Internal Medicine, Hallym Sacred Heart Hospital, Anyang, Gyeonggi-do, Republic of Korea
| | - Jeong Hwan Park
- Department of Pathology, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Shi Mao Zhu
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Junghun Lee
- R&D Center of Innovative Medicines, Helixmith Co., Ltd., Seoul, Republic of Korea
| | - Seung-Shin Yu
- R&D Center of Innovative Medicines, Helixmith Co., Ltd., Seoul, Republic of Korea
| | - Chun Soo Lim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Dong Ki Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Yon Su Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Seung Hee Yang
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
- Seoul National University Kidney Research Institute, Seoul, Republic of Korea
| | - Jung Pyo Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
- Seoul National University Kidney Research Institute, Seoul, Republic of Korea
| |
Collapse
|
55
|
Guebel DV, Torres NV, Acebes Á. Mapping the transcriptomic changes of endothelial compartment in human hippocampus across aging and mild cognitive impairment. Biol Open 2021; 10:bio057950. [PMID: 34184731 PMCID: PMC8181899 DOI: 10.1242/bio.057950] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 04/07/2021] [Indexed: 12/17/2022] Open
Abstract
Compromise of the vascular system has important consequences on cognitive abilities and neurodegeneration. The identification of the main molecular signatures present in the blood vessels of human hippocampus could provide the basis to understand and tackle these pathologies. As direct vascular experimentation in hippocampus is problematic, we achieved this information by computationally disaggregating publicly available whole microarrays data of human hippocampal homogenates. Three conditions were analyzed: 'Young Adults', 'Aged', and 'aged with Mild Cognitive Impairment' (MCI). The genes identified were contrasted against two independent data-sets. Here we show that the endothelial cells from the Younger Group appeared in an 'activated stage'. In turn, in the Aged Group, the endothelial cells showed a significant loss of response to shear stress, changes in cell adhesion molecules, increased inflammation, brain-insulin resistance, lipidic alterations, and changes in the extracellular matrix. Some specific changes in the MCI group were also detected. Noticeably, in this study the features arisen from the Aged Group (high tortuosity, increased bifurcations, and smooth muscle proliferation), pose the need for further experimental verification to discern between the occurrence of arteriogenesis and/or vascular remodeling by capillary arterialization. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Daniel V. Guebel
- Program Agustín de Betancourt, Universidad de La Laguna, Tenerife 38200, Spain
- Department of Biochemistry, Cellular Biology and Genetics, Institute of Biomedical Technologies, Universidad de La Laguna, Tenerife 38200, Spain
| | - Néstor V. Torres
- Department of Biochemistry, Cellular Biology and Genetics, Institute of Biomedical Technologies, Universidad de La Laguna, Tenerife 38200, Spain
| | - Ángel Acebes
- Department of Basic Medical Sciences, Institute of Biomedical Technologies, University of La Laguna, Tenerife 38200, Spain
| |
Collapse
|
56
|
Maeta N, Tamura K, Ezuka F, Takemitsu H. Comparative analysis of canine mesenchymal stem cells and bone marrow-derived mononuclear cells. Vet World 2021; 14:1028-1037. [PMID: 34083956 PMCID: PMC8167527 DOI: 10.14202/vetworld.2021.1028-1037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 03/16/2021] [Indexed: 12/16/2022] Open
Abstract
Background and aim: Mesenchymal stem cells (MSCs), which have multi-lineage differentiation potentials, are a promising source for regenerative medicine. However, the focus of study of MSCs is shifting from the characterization of the differentiation potential to their secretion potential for cell transplantation. Tissue regeneration and the attenuation of immune responses are thought to be affected by the secretion of multiple growth factors and cytokines by MSCs. However, the secretion potential of MSCs profiling remains incompletely characterized. In this study, we focused on the secretion ability related and protein mRNA expression of dog adipose tissue-derived MSCs (AT-MSC), bone marrow (BM)-derived MSCs, and BM-derived mononuclear cells (BM-MNC). Materials and Methods: Real-time polymerase chain reaction analyses revealed mRNA expression of nine growth factors and seven interleukins in these types of cells and three growth factors protein expression were determined using Enzyme-linked immunosorbent assay. Results: For the BM-MNC growth factors, the mRNA expression of transforming growth factor-β (TGF-β) was the highest. For the BM-derived MSC (BM-MSC) and AT-MSC growth factors, the mRNA expression of vascular endothelial growth factor (VEGF) was highest. BM-MSCs and AT-MSCs showed similar expression profiles. In contrast, BM-MNCs showed unique expression profiles for hepatocyte growth factor and epidermal growth factor. The three types of cells showed a similar expression of TGF-β. Conclusion: We conclude that expression of cytokine proteins and mRNAs suggests involvement in tissue repair and protection.
Collapse
Affiliation(s)
- Noritaka Maeta
- Aikouishida Animal Hospital, Isehara, 1195-4 Takamori, Isehara, Kanagawa, 259-1114, Japan.,Faculty of Veterinary Medicine, Okayama University of Science, 1-3 Ikoinooka, Imabari, Ehime, 794-8555, Japan
| | - Katsutoshi Tamura
- Aikouishida Animal Hospital, Isehara, 1195-4 Takamori, Isehara, Kanagawa, 259-1114, Japan
| | - Fuuna Ezuka
- Science and Humanities Master's Programme, Graduate School of Science and the Humanities, Kurashiki University of Science and The Arts, 2640 Nishinoura Tsurajima Kurashiki Okayama, 712-8505, Japan
| | - Hiroshi Takemitsu
- Science and Humanities Master's Programme, Graduate School of Science and the Humanities, Kurashiki University of Science and The Arts, 2640 Nishinoura Tsurajima Kurashiki Okayama, 712-8505, Japan.,Department of Comparative Animal Science, College of Life Science, Kurashiki University of Science and The Arts, 2640 Nishinoura Tsurajima Kurashiki Okayama, 712-8505, Japan
| |
Collapse
|
57
|
Zhou M, Hou Y, Wu J, Li G, Cao P, Chen W, Hu L, Gan D. miR-93-5p promotes insulin resistance to regulate type 2 diabetes progression in HepG2 cells by targeting HGF. Mol Med Rep 2021; 23:329. [PMID: 33760164 PMCID: PMC7974269 DOI: 10.3892/mmr.2021.11968] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 01/04/2021] [Indexed: 12/12/2022] Open
Abstract
Insulin resistance is a common feature of type 2 diabetes mellitus (T2DM). However, the mechanisms underlying insulin resistance are not completely understood. The present study aimed to investigate the effect of microRNA (miR)-93-5p on insulin resistance in T2DM cells. Human hepatocellular carcinoma (HCC; HepG2) cells were cultured in medium with high glucose content (30 mM glucose) to establish an in vitro insulin-resistant cell model (IR group). Glucose consumption and glycogen synthesis assays were performed to assess glucose consumption and glycogen synthesis, respectively. By performing immunoprecipitation assays, the abundance of the Met-insulin receptor complex was detected in HepG2 cells. miR-93-5p and hepatocyte growth factor (HGF) mRNA expression levels were measured via reverse transcription-quantitative PCR, and HGF protein expression levels were measured via western blotting. A dual-luciferase reporter assay was conducted to investigate the interaction between miR-93-5p and HGF. Cell Counting Kit-8, BrdU and caspase-3 activity assays were performed to evaluate cell viability, proliferation and apoptosis, respectively, in insulin-resistant HepG2 cells following transfection with small interfering RNA-HGF, HGF overexpression vector, miR-93-5p mimic or miR-93-5p inhibitor. The results demonstrated that miR-93-5p expression was significantly increased and HGF expression was significantly decreased in HCC tissues isolated from patients with or without T2DM compared with adjacent healthy tissues isolated from patients without T2DM. Compared with the IR group, miR-93-5p overexpression significantly increased cell proliferation, glucose consumption and glycogen synthesis, but significantly inhibited apoptosis in insulin-resistant HepG2 cells. By contrast, compared with the IR group, HGF overexpression significantly inhibited cell proliferation, glucose consumption and glycogen synthesis, but significantly enhanced cell apoptosis in insulin-resistant HepG2 cells. Following co-transfection with HGF overexpression vector and miR-93-5p mimic, miR-93-5p mimic-mediated induction of HepG2 cell proliferation, glucose consumption and glycogen synthesis in insulin-resistant HepG2 cells was inhibited. Collectively, the results of the present study indicated that miR-93-5p enhanced insulin resistance to regulate T2DM progression in HepG2 cells by targeting HGF.
Collapse
Affiliation(s)
- Man Zhou
- Department of Endocrinology, Wuhan Third Hospital, Wuhan, Hubei 430060, P.R. China
| | - Yilin Hou
- Department of Endocrinology, Wuhan Third Hospital, Wuhan, Hubei 430060, P.R. China
| | - Jun Wu
- Department of Endocrinology, Wuhan Third Hospital, Wuhan, Hubei 430060, P.R. China
| | - Guangli Li
- Department of Endocrinology, Wuhan Third Hospital, Wuhan, Hubei 430060, P.R. China
| | - Ping Cao
- Department of Endocrinology, Wuhan Third Hospital, Wuhan, Hubei 430060, P.R. China
| | - Wan Chen
- Department of Endocrinology, Wuhan Third Hospital, Wuhan, Hubei 430060, P.R. China
| | - Lingli Hu
- Department of Endocrinology, Wuhan Third Hospital, Wuhan, Hubei 430060, P.R. China
| | - Dingyun Gan
- Department of Endocrinology, Wuhan Third Hospital, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
58
|
HGF/c-Met Signalling in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1270:31-44. [PMID: 33123991 DOI: 10.1007/978-3-030-47189-7_2] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Recently, it has become clearer that tumor plasticity increases the chance that cancer cells could acquire new mechanisms to escape immune surveillance, become resistant to conventional drugs, and spread to distant sites.Effectively, tumor plasticity drives adaptive response of cancer cells to hypoxia and nutrient deprivation leading to stimulation of neoangionesis or tumor escape. Therefore, tumor plasticity is believed to be a great contributor in recurrence and metastatic dissemination of cancer cells. Importantly, it could be an Achilles' heel of cancer if we could identify molecular mechanisms dictating this phenotype.The reactivation of stem-like signalling pathways is considered a great determinant of tumor plasticity; in addition, a key role has been also attributed to tumor microenvironment (TME). Indeed, it has been proved that cancer cells interact with different cells in the surrounding extracellular matrix (ECM). Interestingly, well-established communication represents a potential allied in maintenance of a plastic phenotype in cancer cells supporting tumor growth and spread. An important signalling pathway mediating cancer cell-TME crosstalk is represented by the HGF/c-Met signalling.Here, we review the role of the HGF/c-Met signalling in tumor-stroma crosstalk focusing on novel findings underlying its role in tumor plasticity, immune escape, and development of adaptive mechanisms.
Collapse
|
59
|
Burganova G, Bridges C, Thorn P, Landsman L. The Role of Vascular Cells in Pancreatic Beta-Cell Function. Front Endocrinol (Lausanne) 2021; 12:667170. [PMID: 33981287 PMCID: PMC8109179 DOI: 10.3389/fendo.2021.667170] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 04/08/2021] [Indexed: 12/13/2022] Open
Abstract
Insulin-producing β-cells constitute the majority of the cells in the pancreatic islets. Dysfunction of these cells is a key factor in the loss of glucose regulation that characterizes type 2 diabetes. The regulation of many of the functions of β-cells relies on their close interaction with the intra-islet microvasculature, comprised of endothelial cells and pericytes. In addition to providing islet blood supply, cells of the islet vasculature directly regulate β-cell activity through the secretion of growth factors and other molecules. These factors come from capillary mural pericytes and endothelial cells, and have been shown to promote insulin gene expression, insulin secretion, and β-cell proliferation. This review focuses on the intimate crosstalk of the vascular cells and β-cells and its role in glucose homeostasis and diabetes.
Collapse
Affiliation(s)
- Guzel Burganova
- Department of Cell and Development Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Claire Bridges
- Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
| | - Peter Thorn
- Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
| | - Limor Landsman
- Department of Cell and Development Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- *Correspondence: Limor Landsman,
| |
Collapse
|
60
|
Acar MB, Ayaz-Güner Ş, Di Bernardo G, Güner H, Murat A, Peluso G, Özcan S, Galderisi U. Obesity induced by high-fat diet is associated with critical changes in biological and molecular functions of mesenchymal stromal cells present in visceral adipose tissue. Aging (Albany NY) 2020; 12:24894-24913. [PMID: 33361524 PMCID: PMC7803587 DOI: 10.18632/aging.202423] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 11/27/2020] [Indexed: 12/27/2022]
Abstract
The mesenchymal stromal cells (MSCs) residing within the stromal component of visceral adipose tissue appear to be greatly affected by obesity, with impairment of their functions and presence of senescence. To gain further insight into these phenomena, we analyzed the changes in total proteome content and secretome of mouse MSCs after a high-fat diet (HFD) treatment compared to a normal diet (ND). In healthy conditions, MSCs are endowed with functions mainly devoted to vesicle trafficking. These cells have an immunoregulatory role, affecting leukocyte activation and migration, acute inflammation phase response, chemokine signaling, and platelet activities. They also present a robust response to stress. We identified four signaling pathways (TGF-β, VEGFR2, HMGB1, and Leptin) that appear to govern the cells' functions. In the obese mice, MSCs showed a change in their functions. The immunoregulation shifted toward pro-inflammatory tasks with the activation of interleukin-1 pathway and of Granzyme A signaling. Moreover, the methionine degradation pathway and the processing of capped intronless pre-mRNAs may be related to the inflammation process. The signaling pathways we identified in ND MSCs were replaced by MET, WNT, and FGFR2 signal transduction, which may play a role in promoting inflammation, cancer, and aging.
Collapse
Affiliation(s)
- Mustafa Burak Acar
- Genome and Stem Cell Center (GENKÖK) Erciyes University, Kayseri, Turkey
| | - Şerife Ayaz-Güner
- Department of Molecular Biology and Genetics, Faculty of Life and Natural Science, Abdullah Gül University, Kayseri, Turkey
| | - Giovanni Di Bernardo
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, Naples, Italy
| | - Hüseyin Güner
- Department of Molecular Biology and Genetics, Faculty of Life and Natural Science, Abdullah Gül University, Kayseri, Turkey
| | - Ayşegül Murat
- Genome and Stem Cell Center (GENKÖK) Erciyes University, Kayseri, Turkey
| | | | - Servet Özcan
- Genome and Stem Cell Center (GENKÖK) Erciyes University, Kayseri, Turkey
- Department of Biology, Faculty of Science, Erciyes University, Kayseri, Turkey
| | - Umberto Galderisi
- Genome and Stem Cell Center (GENKÖK) Erciyes University, Kayseri, Turkey
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, Naples, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Temple University, Philadelphia, PA 19122, USA
| |
Collapse
|
61
|
Yang SY, Yang KC, Sumi S. Prevascularization-free Primary Subcutaneous Transplantation of Xenogeneic Islets Coencapsulated With Hepatocyte Growth Factor. Transplant Direct 2020; 6:e620. [PMID: 33134496 PMCID: PMC7587419 DOI: 10.1097/txd.0000000000001078] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 08/31/2020] [Accepted: 09/18/2020] [Indexed: 01/13/2023] Open
Abstract
Subcutaneous pouch is a potential site for islet transplantation. However, insufficient oxygen supply remains challenging. Pretreatment of neovascularization using basic fibroblast growth factor can solve this, but it needs 2× operations. We developed a device that contains rat islets in chitosan gel packed in a bag made of highly biocompatible ethylene vinyl alcohol copolymer porous membrane. This study investigated whether coencapsulation of hepatocyte growth factor (HGF) with islets in the device enables novel method of prevascularization-free primary subcutaneous transplantation. METHODS In vitro experiments examined slow release of HGF from the chitosan gel and islet-protection effect of HGF against hypoxia. In the latter, rat islets with/without HGF (200 ng/mL) was cultured in 1% oxygen. In in vivo experiment, fabricated device with/without HGF (10 μg/device) containing rat islets was primarily transplanted to streptozotocin-induced diabetic mice subcutaneously. RESULTS In vitro experiments showed sustained release of HGF for 28 d and alleviating effect of HGF on cell death and glucose-responsive insulin release after hypoxic culture. Islet + HGF mice, but not islet-alone mice, showed decreased nonfasting blood glucose and regained body weight after transplantation. In intraperitoneal glucose tolerance test, islet + HGF mice exhibited decreased fasting blood glucose (200 ± 55 mg/dL) and good blood glucose disappearance rate (K value) (0.817 ± 0.101) comparing to normal mice (123 ± 28 mg/dL and 1.074 ± 0.374, respectively). However, in islet-alone mice, fasting blood glucose was high (365 ± 172 mg/dL) and K value was indeterminable. Serum insulin in islet + HGF mice (1.58 ± 0.94 μg/L) was close to normal mice (1.66 ± 0.55 μg/L), whereas those in islet-alone mice (0.279 ± 0.076 μg/L) and diabetic mice (0.165 ± 0.079 μg/L) were low. Immunohistochemical examination showed intact insulin- and glucagon-positive islets in retrieved devices with HGF, but no intact islet was found in the device without HGF. CONCLUSIONS HGF could enhance islet survival in hypoxia and enhance in vivo function of encapsulated islets after primary subcutaneous transplantation.
Collapse
Affiliation(s)
- Sin-Yu Yang
- Laboratory of Organ and Tissue Reconstruction, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Kai-Chiang Yang
- Laboratory of Organ and Tissue Reconstruction, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
- School of Dental Technology, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shoichiro Sumi
- Laboratory of Organ and Tissue Reconstruction, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
62
|
Ramne S, Drake I, Ericson U, Nilsson J, Orho-Melander M, Engström G, Sonestedt E. Identification of Inflammatory and Disease-Associated Plasma Proteins that Associate with Intake of Added Sugar and Sugar-Sweetened Beverages and Their Role in Type 2 Diabetes Risk. Nutrients 2020; 12:E3129. [PMID: 33066363 PMCID: PMC7602152 DOI: 10.3390/nu12103129] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/09/2020] [Accepted: 10/10/2020] [Indexed: 02/07/2023] Open
Abstract
It has been suggested that high intake of added sugar and sugar-sweetened beverages (SSBs) increase the level of circulating inflammatory proteins and that chronic inflammation plays a role in type 2 diabetes (T2D) development. We aim to examine how added sugar and SSB intake associate with 136 measured plasma proteins and C-reactive protein (CRP) in the Malmö Diet and Cancer-Cardiovascular Cohort (n = 4382), and examine if the identified added sugar- and SSB-associated proteins associate with T2D incidence. A two-step iterative resampling approach was used to internally replicate proteins that associated with added sugar and SSB intake. Nine proteins were identified to associate with added sugar intake, of which only two associated with T2D incidence (p < 0.00045). Seven proteins were identified to associate with SSB intake, of which six associated strongly with T2D incidence (p < 6.9 × 10-8). No significant associations were observed between added sugar and SSB intake and CRP concentrations. In summary, our elucidation of the relationship between plasma proteome and added sugar and SSB intake, in relation to future T2D risk, demonstrated that SSB intake, rather than the total intake of added sugar, was related to a T2D-pathological proteomic signature. However, external replication is needed to verify the findings.
Collapse
Affiliation(s)
- Stina Ramne
- Department of Clinical Sciences Malmö, Lund University, 214 28 Malmö, Sweden; (I.D.); (U.E.); (J.N.); (M.O.-M.); (G.E.); (E.S.)
| | | | | | | | | | | | | |
Collapse
|
63
|
Allen JN, Dey A, Cai J, Zhang J, Tian Y, Kennett M, Ma Y, Liang TJ, Patterson AD, Hankey-Giblin PA. Metabolic Profiling Reveals Aggravated Non-Alcoholic Steatohepatitis in High-Fat High-Cholesterol Diet-Fed Apolipoprotein E-Deficient Mice Lacking Ron Receptor Signaling. Metabolites 2020; 10:metabo10080326. [PMID: 32796650 PMCID: PMC7464030 DOI: 10.3390/metabo10080326] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/29/2020] [Accepted: 08/03/2020] [Indexed: 12/13/2022] Open
Abstract
Non-alcoholic steatohepatitis (NASH) represents the progressive sub-disease of non-alcoholic fatty liver disease that causes chronic liver injury initiated and sustained by steatosis and necroinflammation. The Ron receptor is a tyrosine kinase of the Met proto-oncogene family that potentially has a beneficial role in adipose and liver-specific inflammatory responses, as well as glucose and lipid metabolism. Since its discovery two decades ago, the Ron receptor has been extensively investigated for its differential roles on inflammation and cancer. Previously, we showed that Ron expression on tissue-resident macrophages limits inflammatory macrophage activation and promotes a repair phenotype, which can retard the progression of NASH in a diet-induced mouse model. However, the metabolic consequences of Ron activation have not previously been investigated. Here, we explored the effects of Ron receptor activation on major metabolic pathways that underlie the development and progression of NASH. Mice lacking apolipoprotein E (ApoE KO) and double knockout (DKO) mice that lack ApoE and Ron were maintained on a high-fat high-cholesterol diet for 18 weeks. We observed that, in DKO mice, the loss of ligand-dependent Ron signaling aggravated key pathological features in steatohepatitis, including steatosis, inflammation, oxidation stress, and hepatocyte damage. Transcriptional programs positively regulating fatty acid (FA) synthesis and uptake were upregulated in the absence of Ron receptor signaling, whereas lipid disposal pathways were downregulated. Consistent with the deregulation of lipid metabolism pathways, the DKO animals exhibited increased accumulation of FAs in the liver and decreased level of bile acids. Altogether, ligand-dependent Ron receptor activation provides protection from the deregulation of major metabolic pathways that initiate and aggravate non-alcoholic steatohepatitis.
Collapse
Affiliation(s)
- Joselyn N. Allen
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (J.N.A.); (A.D.); (J.C.); (J.Z.); (Y.T.); (M.K.)
| | - Adwitia Dey
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (J.N.A.); (A.D.); (J.C.); (J.Z.); (Y.T.); (M.K.)
| | - Jingwei Cai
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (J.N.A.); (A.D.); (J.C.); (J.Z.); (Y.T.); (M.K.)
| | - Jingtao Zhang
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (J.N.A.); (A.D.); (J.C.); (J.Z.); (Y.T.); (M.K.)
| | - Yuan Tian
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (J.N.A.); (A.D.); (J.C.); (J.Z.); (Y.T.); (M.K.)
| | - Mary Kennett
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (J.N.A.); (A.D.); (J.C.); (J.Z.); (Y.T.); (M.K.)
| | - Yanling Ma
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, The National Institutes of Health, Bethesda, MD 20814, USA; (Y.M.); (T.J.L.)
| | - T. Jake Liang
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, The National Institutes of Health, Bethesda, MD 20814, USA; (Y.M.); (T.J.L.)
| | - Andrew D. Patterson
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (J.N.A.); (A.D.); (J.C.); (J.Z.); (Y.T.); (M.K.)
- Correspondence: (A.D.P.); (P.A.H.-G.); Tel.: +1-814-867-4565; (A.D.P.); +1-814-863-0128 (P.A.H.-G.)
| | - Pamela A. Hankey-Giblin
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (J.N.A.); (A.D.); (J.C.); (J.Z.); (Y.T.); (M.K.)
- Correspondence: (A.D.P.); (P.A.H.-G.); Tel.: +1-814-867-4565; (A.D.P.); +1-814-863-0128 (P.A.H.-G.)
| |
Collapse
|
64
|
Hu X, Tang F, Liu P, Zhong T, Yuan F, He Q, von Itzstein M, Li H, Weng L, Yu X. Structural and Functional Insight Into the Glycosylation Impact Upon the HGF/c-Met Signaling Pathway. Front Cell Dev Biol 2020; 8:490. [PMID: 32626713 PMCID: PMC7314907 DOI: 10.3389/fcell.2020.00490] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 05/25/2020] [Indexed: 12/31/2022] Open
Abstract
Upon interactions with its specific ligand hepatocyte growth factor (HGF), the c-Met signal is relayed to series of downstream pathways, exerting essential biological roles. Dysregulation of the HGF-c-Met signaling pathway has been implicated in the onset, progression and metastasis of various cancers, making the HGF-c-Met axis a promising therapeutic target. Both c-Met and HGF undergo glycosylation, which appears to be biologically relevant to their function and structural integrity. Different types of glycoconjugates in the local cellular environment can also regulate HGF/c-Met signaling by distinct mechanisms. However, detailed knowledge pertaining to the glycosylation machinery of the HGF-c-Met axis as well as its potential applications in oncology research is yet to be established. This mini review highlights the significance of the HGF-c-Met signaling pathway in physiological and pathological context, and discusses the molecular mechanisms by which affect the glycosylation of the HGF-c-Met axis. Owing to the crucial role played by glycosylation in the regulation of HGF/c-Met activity, better understanding of this less exploited field may contribute to the development of novel therapeutics targeting glycoepitopes.
Collapse
Affiliation(s)
- Xinyue Hu
- College of Medicine, Hunan Normal University, Changsha, China
| | - Feiyu Tang
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Peilin Liu
- College of Medicine, Hunan Normal University, Changsha, China
| | - Taowei Zhong
- College of Medicine, Hunan Normal University, Changsha, China
| | - Fengyan Yuan
- College of Medicine, Hunan Normal University, Changsha, China
| | - Quanyuan He
- College of Medicine, Hunan Normal University, Changsha, China.,Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University, Changsha, China
| | - Mark von Itzstein
- Institute for Glycomics, Griffith University, Gold Coast, QLD, Australia
| | - Hao Li
- Biliary Tract Surgery Laboratory, Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China.,Hunan Research Center of Biliary Disease, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Liang Weng
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Molecular Radiation Oncology in Hunan Province, Central South University, Changsha, China
| | - Xing Yu
- College of Medicine, Hunan Normal University, Changsha, China.,Institute for Glycomics, Griffith University, Gold Coast, QLD, Australia
| |
Collapse
|
65
|
Wu SF, Noren Hooten N, Freeman DW, Mode NA, Zonderman AB, Evans MK. Extracellular vesicles in diabetes mellitus induce alterations in endothelial cell morphology and migration. J Transl Med 2020; 18:230. [PMID: 32517700 PMCID: PMC7285586 DOI: 10.1186/s12967-020-02398-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 06/01/2020] [Indexed: 02/08/2023] Open
Abstract
Background Inflammation-related atherosclerotic peripheral vascular disease is a major end organ complication of diabetes mellitus that results in devastating morbidity and mortality. Extracellular vesicles (EVs) are nano-sized particles that contain molecular cargo and circulate in the blood. Here, we examined EV protein cargo from diabetic individuals and whether these EVs cause functional changes in endothelial cells. Methods We quantified inflammatory protein levels in plasma-derived EVs from a longitudinal cohort of euglycemic and diabetic individuals and used in vitro endothelial cell biological assays to assess the functional effects of these EVs with samples from a cross-sectional cohort. Results We found several significant associations between EV inflammatory protein levels and diabetes status. The angiogenic factor, vascular endothelial growth factor A (VEGF-A), was associated with diabetes status in our longitudinal cohort. Those with diabetes mellitus had higher EV VEGF-A levels compared to euglycemic individuals. Additionally, EV levels of VEGF-A were significantly associated with homeostatic model assessment of insulin resistance (HOMA-IR) and β-cell function (HOMA-B). To test whether EVs with different inflammatory cargo can demonstrate different effects on endothelial cells, we performed cell migration and immunofluorescence assays. We observed that EVs from diabetic individuals increased cell lamellipodia formation and migration when compared to EVs from euglycemic individuals. Conclusions Higher levels of inflammatory proteins were found in EVs from diabetic individuals. Our data implicate EVs as playing important roles in peripheral vascular disease that occur in individuals with diabetes mellitus and suggest that EVs may serve as an informative diagnostic tool for the disease.
Collapse
Affiliation(s)
- Sharon F Wu
- Laboratory of Epidemiology and Population Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Nicole Noren Hooten
- Laboratory of Epidemiology and Population Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - David W Freeman
- Laboratory of Epidemiology and Population Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA.,University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Nicolle A Mode
- Laboratory of Epidemiology and Population Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Alan B Zonderman
- Laboratory of Epidemiology and Population Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Michele K Evans
- Laboratory of Epidemiology and Population Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA.
| |
Collapse
|
66
|
Ostróżka-Cieślik A, Dolińska B. The Role of Hormones and Trophic Factors as Components of Preservation Solutions in Protection of Renal Function before Transplantation: A Review of the Literature. Molecules 2020; 25:E2185. [PMID: 32392782 PMCID: PMC7248710 DOI: 10.3390/molecules25092185] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/29/2020] [Accepted: 05/05/2020] [Indexed: 12/12/2022] Open
Abstract
Transplantation is currently a routine method for treating end-stage organ failure. In recent years, there has been some progress in the development of an optimal composition of organ preservation solutions, improving the vital functions of the organ and allowing to extend its storage period until implantation into the recipient. Optimizations are mostly based on commercial solutions, routinely used to store grafts intended for transplantation. The paper reviews hormones with a potential nephroprotective effect, which were used to modify the composition of renal perfusion and preservation solutions. Their effectiveness as ingredients of preservation solutions was analysed based on a literature review. Hormones and trophic factors are innovative preservation solution supplements. They have a pleiotropic effect and affect normal renal function. The expression of receptors for melatonin, prolactin, thyrotropin, corticotropin, prostaglandin E1 and trophic factors was confirmed in the kidneys, which suggests that they are a promising therapeutic target for renal IR (ischemia-reperfusion) injury. They can have anti-inflammatory, antioxidant and anti-apoptotic effects, limiting IR injury.
Collapse
Affiliation(s)
- Aneta Ostróżka-Cieślik
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Kasztanowa 3, 41-200 Sosnowiec, Poland;
| | - Barbara Dolińska
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Kasztanowa 3, 41-200 Sosnowiec, Poland;
- “Biochefa” Pharmaceutical Research and Production Plant, Kasztanowa 3, 41-200 Sosnowiec, Poland
| |
Collapse
|
67
|
Tong X, Liang R, Jia Y, Qin W, Guo C, Wu X, Wang Z, Chen D, Tan N. Suhuang Antitussive Capsules-Ameliorative Effects on LPS-Induced Sputum Obstruction in Mice Through Promoting HGF Secretion. Front Pharmacol 2019; 10:1422. [PMID: 31920638 PMCID: PMC6930918 DOI: 10.3389/fphar.2019.01422] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 11/07/2019] [Indexed: 12/12/2022] Open
Abstract
Sputum obstruction is one of common cough complications, which is tightly associated with airway inflammation. Suhuang antitussive capsule (SH Capsule), a classic traditional Chinese medicine prescription, has been used for the treatment of post-cold cough and cough variant asthma in the long clinical application. This study aims to investigate the effects and underlying mechanisms of SH Capsule on LPS-induced sputum obstruction in mice. The results showed that SH Capsule effectively promoted the tracheal phenol red output and mucociliary clearance. SH Capsule also alleviated airway inflammation-mediated mucin 5AC (MUC5AC) level through EGFR-ERK signaling. A further in vivo analysis showed that HGF inhibitor SU11274 abrogated the effects of SH Capsule on MUC5AC, well demonstrating that HGF was required for the beneficial effects of SH Capsule on expectoration in vivo. Moreover, SH Capsule promoted HGF secretion in a colon-dependent manner, which reached lung tissues via blood circulation. Collectively, this study provided new pharmacological data for clinical use of SH Capsule, and proposed a novel mechanism by which SH Capsule was pharmacologically promising for treating sputum obstruction.
Collapse
Affiliation(s)
- Xiyang Tong
- State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Rongyao Liang
- State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yuning Jia
- Yangtze River Pharmaceutical Group Beijing Haiyan Pharmaceutical Co., Ltd., Beijing, China.,Beijing University of Chemical Technology, Beijing, China
| | - Weiwei Qin
- State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Chao Guo
- State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xingdong Wu
- State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Zhen Wang
- State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Dong Chen
- Yangtze River Pharmaceutical Group Beijing Haiyan Pharmaceutical Co., Ltd., Beijing, China
| | - Ninghua Tan
- State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
68
|
Tarantino G, Citro V, Conforti P, Balsano C, Capone D. Is There a Link between Basal Metabolic Rate, Spleen Volume and Hepatic Growth Factor Levels in Patients with Obesity-Related NAFLD? J Clin Med 2019; 8:jcm8101510. [PMID: 31547124 PMCID: PMC6832562 DOI: 10.3390/jcm8101510] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/10/2019] [Accepted: 09/16/2019] [Indexed: 01/30/2023] Open
Abstract
Background: Recent pieces of research point to a link between basal metabolic rate (BMR) and non-alcoholic fatty liver disease (NAFLD) or hepatic steatosis (HS). The spleen in obese patients is associated with the cardiovascular system. Enlargement of the spleen is suggestive of nonalcoholic steatohepatitis (NASH). Patients with NASH present an increase in growth factor (HGF) as well as those with advanced heart failure. Interleukin-16 and interleukin-12p40 levels were found to correlate significantly with BMI, and waist circumference. Aim: We tried to find a relationship between BMR, spleen length and HGF. Methods: We analysed retrospective data from 80 obese patients with NAFLD. We evaluated indices of indirect calorimetry by the bioimpendance analysis; carotid intima-media thickness (IMT), spleen length (SLD) and HS by ultrasonography; serum HGF, IL-16, IL-12p40 and IL-6 concentrations by a magnetic bead-based multiplex immunoassays and the severity of NAFLD by BARD score > 2. Results: HGF levels of the obese were higher than those of controls, p < 0.001. At linear regression, BMR was foreseen by spleen length (p < 0.001), which was predicted by HGF (p = 0.04). BMR was predicted by IL-16 (p = 0.005), which predicted HGF, p = 0.034. Only fat mass, among other factors, predicted early atherosclerosis, p = 0.017; IL-12p40 did not predict IMT, HGF and BMR (p = 0.57, 0.09 and 0.59, respectively). The BARD score > 2 was negatively predicted by BMR and FFM (p =0.032 and 0.031, respectively), at the logistic regression. Interesting findings at the extended regression (mediation effect) were: IL-16 was likely causal in predicting BMR by HGF levels; HGF was influential in predicting BMR by SLD level. HS was predicted by SLD in males (p = 0.014), of advanced age (p < 0.001) and by BMR (p < 0.001). IL-6 concentrations, but not BMR were influential in the prediction of HS by SLD. Conclusion: These data reinforce the concept that the immune system is a sensor of the metabolic state, showing a link between HGF levels and BMR, which is mediated by IL-16 (cytokine inducing a cascade of inflammatory factors), and ascertaining the influential effect of the spleen, as main immune organ.
Collapse
Affiliation(s)
- Giovanni Tarantino
- Department of Clinical Medicine and Surgery, Federico II University Medical School of Naples, 80131 Napoli NA, Italy.
| | - Vincenzo Citro
- Department of General Medicine, "Umberto I" Hospital, Nocera Inferiore (Sa), 84014 Nocera Inferiore SA, Italy.
| | - Paolo Conforti
- "Federico II" University Medical School of Naples, 80131 Napoli NA, Italy.
| | - Clara Balsano
- Department of Clinical Medicine, Life, Health & Environmental Sciences-MESVA, University of L'Aquila, 67100 L'Aquila AQ, Italy.
| | - Domenico Capone
- Care Department of Public Health and Drug-Use, Section of Medical Pharmacology and Toxicology, "Federico II" University, 80131 Naples NA, Italy.
| |
Collapse
|
69
|
Henning SM, Yang J, Woo SL, Lee RP, Huang J, Rasmusen A, Carpenter CL, Thames G, Gilbuena I, Tseng CH, Heber D, Li Z. Hass Avocado Inclusion in a Weight-Loss Diet Supported Weight Loss and Altered Gut Microbiota: A 12-Week Randomized, Parallel-Controlled Trial. Curr Dev Nutr 2019; 3:nzz068. [PMID: 31367691 PMCID: PMC6658913 DOI: 10.1093/cdn/nzz068] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 06/04/2019] [Accepted: 06/05/2019] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Avocados contain fiber, lutein, and vitamin E, and they are a rich source of MUFAs. The effect of including an avocado daily as part of a hypocaloric weight-loss diet on weight loss is not known. OBJECTIVE The aim of this study was to determine the effect of daily avocado consumption as part of a hypocaloric diet on weight loss, body composition, satiety, biomarkers of inflammation, and intestinal microbiota composition. METHODS In this randomized, parallel-controlled, open-label, 2-arm intervention study, 51 healthy overweight/obese women and men were assigned to a hypocaloric diet with 1 Hass avocado daily (AVO; n = 24) or a hypocaloric diet (CTRL; n = 27) without daily avocado for 12 wk. Serum markers and intestinal microbiota were analyzed at baseline and week 12. RESULTS Both groups experienced significant weight loss, decrease in BMI (in kg/m2), total body fat, and visceral adipose tissue, respectively (AVO: -2.3 ± 2 kg, -0.8 ± 0.8, -1.1% ± 2%, and -81.2 ± 118 g; CTRL: -2.6 ± 3.6 kg, -0.9 ± 1, -1.5% ± 2%, and -87.4 ± 216 g). We observed a significant decrease in serum glucose over time in the control group compared with the AVO group. There was no change between the groups in serum triglyceride, but a significant decrease from baseline to 12 wk was observed in the AVO group. Serum hepatic growth factor (HGF) and relative proportion of bacterial phyla (Firmicutes and Bacteroidetes), family (Bacteroidaceae and Erysipelotrichaceae), and genus (Bacteroides, Clostridium, Methanosphaera, and Candidatus Soleaferrea) were significantly altered in the AVO group compared with the CTRL group. A trend to decrease in serum inflammatory factors IL-1β (P = 0.07) and C-reactive protein (P = 0.074) was observed in the AVO group compared with CTRL. CONCLUSIONS Daily Hass avocado consumption as part of a hypocaloric diet supported weight loss, a decrease in serum HGF, and an increase in the abundance of bacteria involved in plant polysaccharide fermentation. This trial was registered at clinicaltrials.gov as NCT02953158.
Collapse
Affiliation(s)
- Susanne M Henning
- Department of Medicine, Center for Human Nutrition, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jieping Yang
- Department of Medicine, Center for Human Nutrition, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Shih Lung Woo
- Department of Medicine, Center for Human Nutrition, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ru-Po Lee
- Department of Medicine, Center for Human Nutrition, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jianjun Huang
- Department of Medicine, Center for Human Nutrition, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Anna Rasmusen
- Department of Medicine, Center for Human Nutrition, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Catherine L Carpenter
- Department of Medicine, Center for Human Nutrition, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Gail Thames
- Department of Medicine, Center for Human Nutrition, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Irene Gilbuena
- Department of Medicine, Center for Human Nutrition, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Chi-Hong Tseng
- Department of Statistics Core, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - David Heber
- Department of Medicine, Center for Human Nutrition, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Zhaoping Li
- Department of Medicine, Center for Human Nutrition, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
70
|
Wang J, Lai B, Nanayakkara G, Yang Q, Sun Y, Lu Y, Shao Y, Yu D, Yang WY, Cueto R, Fu H, Zeng H, Shen W, Wu S, Zhang C, Liu Y, Choi ET, Wang H, Yang X. Experimental Data-Mining Analyses Reveal New Roles of Low-Intensity Ultrasound in Differentiating Cell Death Regulatome in Cancer and Non-cancer Cells via Potential Modulation of Chromatin Long-Range Interactions. Front Oncol 2019; 9:600. [PMID: 31355136 PMCID: PMC6640725 DOI: 10.3389/fonc.2019.00600] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 06/18/2019] [Indexed: 12/17/2022] Open
Abstract
Background: The mechanisms underlying low intensity ultrasound (LIUS) mediated suppression of inflammation and tumorigenesis remain poorly determined. Methods: We used microarray datasets from NCBI GEO Dataset databases and conducted a comprehensive data mining analyses, where we studied the gene expression of 299 cell death regulators that regulate 13 different cell death types (cell death regulatome) in cells treated with LIUS. Results: We made the following findings: (1) LIUS exerts a profound effect on the expression of cell death regulatome in cancer cells and non-cancer cells. Of note, LIUS has the tendency to downregulate the gene expression of cell death regulators in non-cancer cells. Most of the cell death regulator genes downregulated by LIUS in non-cancer cells are responsible for mediating inflammatory signaling pathways; (2) LIUS activates different cell death transcription factors in cancer and non-cancer cells. Transcription factors TP-53 and SRF- were induced by LIUS exposure in cancer cells and non-cancer cells, respectively; (3) As two well-accepted mechanisms of LIUS, mild hyperthermia and oscillatory shear stress induce changes in the expression of cell death regulators, therefore, may be responsible for inducing LIUS mediated changes in gene expression patterns of cell death regulators in cells; (4) LIUS exposure may change the redox status of the cells. LIUS may induce more of antioxidant effects in non-cancer cells compared to cancer cells; and (5) The genes modulated by LIUS in cancer cells have distinct chromatin long range interaction (CLRI) patterns to that of non-cancer cells. Conclusions: Our analysis suggests novel molecular mechanisms that may be utilized by LIUS to induce tumor suppression and inflammation inhibition. Our findings may lead to development of new treatment protocols for cancers and chronic inflammation.
Collapse
Affiliation(s)
- Jiwei Wang
- Department of Pharmacology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Microbiology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Immunology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Ultrasound, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Bin Lai
- Department of Pharmacology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Microbiology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Immunology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Gayani Nanayakkara
- Department of Pharmacology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Microbiology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Immunology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
| | - Qian Yang
- Department of Pharmacology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Microbiology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Immunology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
| | - Yu Sun
- Department of Pharmacology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Microbiology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Immunology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
| | - Yifan Lu
- Department of Pharmacology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Microbiology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Immunology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
| | - Ying Shao
- Department of Pharmacology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Microbiology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Immunology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
| | - Daohai Yu
- Department of Clinical Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - William Y. Yang
- Department of Pharmacology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Microbiology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Immunology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
| | - Ramon Cueto
- Department of Pharmacology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Microbiology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Immunology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
| | - Hangfei Fu
- Department of Pharmacology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Microbiology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Immunology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
| | - Huihong Zeng
- Department of Pharmacology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Microbiology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Immunology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
| | - Wen Shen
- Department of Pharmacology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Microbiology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Immunology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
| | - Susu Wu
- Department of Pharmacology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Microbiology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Immunology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
| | - Chunquan Zhang
- Department of Ultrasound, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yanna Liu
- Department of Ultrasound, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Eric T. Choi
- Department of Pharmacology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Microbiology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Immunology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Division of Vascular and Endovascular Surgery, Department of Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Hong Wang
- Department of Pharmacology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Microbiology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Immunology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
| | - Xiaofeng Yang
- Department of Pharmacology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Microbiology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Immunology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
| |
Collapse
|
71
|
Ge MX, Shao RG, He HW. Advances in understanding the regulatory mechanism of cholesterol 7α-hydroxylase. Biochem Pharmacol 2019; 164:152-164. [DOI: 10.1016/j.bcp.2019.04.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 04/08/2019] [Indexed: 02/07/2023]
|
72
|
Schüssler-Fiorenza Rose SM, Contrepois K, Moneghetti KJ, Zhou W, Mishra T, Mataraso S, Dagan-Rosenfeld O, Ganz AB, Dunn J, Hornburg D, Rego S, Perelman D, Ahadi S, Sailani MR, Zhou Y, Leopold SR, Chen J, Ashland M, Christle JW, Avina M, Limcaoco P, Ruiz C, Tan M, Butte AJ, Weinstock GM, Slavich GM, Sodergren E, McLaughlin TL, Haddad F, Snyder MP. A longitudinal big data approach for precision health. Nat Med 2019; 25:792-804. [PMID: 31068711 PMCID: PMC6713274 DOI: 10.1038/s41591-019-0414-6] [Citation(s) in RCA: 278] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 03/06/2019] [Indexed: 12/31/2022]
Abstract
Precision health relies on the ability to assess disease risk at an individual level, detect early preclinical conditions and initiate preventive strategies. Recent technological advances in omics and wearable monitoring enable deep molecular and physiological profiling and may provide important tools for precision health. We explored the ability of deep longitudinal profiling to make health-related discoveries, identify clinically relevant molecular pathways and affect behavior in a prospective longitudinal cohort (n = 109) enriched for risk of type 2 diabetes mellitus. The cohort underwent integrative personalized omics profiling from samples collected quarterly for up to 8 years (median, 2.8 years) using clinical measures and emerging technologies including genome, immunome, transcriptome, proteome, metabolome, microbiome and wearable monitoring. We discovered more than 67 clinically actionable health discoveries and identified multiple molecular pathways associated with metabolic, cardiovascular and oncologic pathophysiology. We developed prediction models for insulin resistance by using omics measurements, illustrating their potential to replace burdensome tests. Finally, study participation led the majority of participants to implement diet and exercise changes. Altogether, we conclude that deep longitudinal profiling can lead to actionable health discoveries and provide relevant information for precision health.
Collapse
Affiliation(s)
- Sophia Miryam Schüssler-Fiorenza Rose
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Spinal Cord Injury Service, Veteran Affairs Palo Alto Health Care System, Palo Alto, CA, USA
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Kévin Contrepois
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Kegan J Moneghetti
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, St Vincent's Hospital, University of Melbourne, Melbourne, Australia
| | - Wenyu Zhou
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Tejaswini Mishra
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Samson Mataraso
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, Berkeley, CA, USA
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, USA
| | - Orit Dagan-Rosenfeld
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Ariel B Ganz
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Jessilyn Dunn
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Mobilize Center, Stanford University, Stanford, CA, USA
| | - Daniel Hornburg
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Shannon Rego
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Dalia Perelman
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Sara Ahadi
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - M Reza Sailani
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Yanjiao Zhou
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
- Department of Medicine, University of Connecticut Health, Farmington, CT, USA
| | - Shana R Leopold
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Jieming Chen
- Bakar Computational Health Sciences Institute and Department of Pediatrics, University of California, San Francisco, CA, USA
| | - Melanie Ashland
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Jeffrey W Christle
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Monika Avina
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Patricia Limcaoco
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Camilo Ruiz
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Marilyn Tan
- Division of Endocrinology, Stanford University School of Medicine, Stanford, CA, USA
| | - Atul J Butte
- Bakar Computational Health Sciences Institute and Department of Pediatrics, University of California, San Francisco, CA, USA
| | | | - George M Slavich
- Cousins Center for Psychoneuroimmunology and Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA, USA
| | - Erica Sodergren
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Tracey L McLaughlin
- Division of Endocrinology, Stanford University School of Medicine, Stanford, CA, USA
| | - Francois Haddad
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA.
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| | - Michael P Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|