1
|
Wang S, Zhang Y, Hu S, Bai X, Zhu J, Hao R, Cao Y, Shi Z. AGDMP1 alleviates insulin resistance by modulating heat shock protein 60-mediated IRS-1/AKT/GLUT4 pathway and adipose inflammation: A potential therapeutic peptide for gestational diabetes mellitus. FASEB J 2025; 39:e70339. [PMID: 39854096 PMCID: PMC11776800 DOI: 10.1096/fj.202402886r] [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: 11/21/2024] [Revised: 01/06/2025] [Accepted: 01/10/2025] [Indexed: 01/26/2025]
Abstract
Gestational Diabetes Mellitus (GDM) is the most frequent complication during pregnancy. Pharmacological interventions, such as peptide drugs that focused on improving the insulin sensitivity might be promising in the prevention and treatment of GDM. In this study, we aimed to investigate the role and mechanism of a novel peptide, named AGDMP1 (Anti-GDM peptide 1), which we previously identified lower in the serum of GDM patients using mass spectrometry, on the adipose insulin resistance in GDM. We found that AGDMP1 had a high affinity for adipose tissues in vivo. AGDMP1 markedly improved glucose homeostasis and insulin resistance in GDM mice. This was associated with reduced inflammation and upregulated AKT/GLUT4 signaling pathway in white adipose tissue. In vitro, AGDMP1 could increase glucose uptake and insulin sensitivity of adipocytes by activating the IRS-1/AKT/GLUT4 signaling pathway under basal, insulin-stimulated, and insulin-resistant conditions, respectively. Mechanistically, we found that AGDMP1 could bind to HSP60 to dampen its effects on AKT signaling and pro-inflammatory response, which was reversed in the present of recombinant HSP60 protein. AGDMP1 ameliorated adipose insulin resistance and inflammation by targeting HSP60 and activating the IRS-1/AKT/GLUT4 signaling pathway. These data supported AGDMP1 with therapeutic potential in GDM and associated pathologies.
Collapse
Affiliation(s)
- Shanshan Wang
- Changzhou Maternal and Child Health Care Hospital, Changzhou Medical CenterNanjing Medical UniversityChangzhouJiangsuChina
- Changzhou Key Laboratory of Maternal and Child Health MedicineChangzhouJiangsuChina
| | - Yuting Zhang
- State Key Laboratory of Reproductive Medicine and Offspring HealthNanjing Medical UniversityNanjingJiangsuChina
| | - Shiman Hu
- Changzhou Maternal and Child Health Care Hospital, Changzhou Medical CenterNanjing Medical UniversityChangzhouJiangsuChina
- Changzhou Key Laboratory of Maternal and Child Health MedicineChangzhouJiangsuChina
| | - Xueqi Bai
- Changzhou Maternal and Child Health Care Hospital, Changzhou Medical CenterNanjing Medical UniversityChangzhouJiangsuChina
- Changzhou Key Laboratory of Maternal and Child Health MedicineChangzhouJiangsuChina
| | - Jiamin Zhu
- State Key Laboratory of Reproductive Medicine and Offspring HealthNanjing Medical UniversityNanjingJiangsuChina
| | - Runrun Hao
- Changzhou Maternal and Child Health Care Hospital, Changzhou Medical CenterNanjing Medical UniversityChangzhouJiangsuChina
- Changzhou Key Laboratory of Maternal and Child Health MedicineChangzhouJiangsuChina
| | - Yan Cao
- Nanjing Women and Children's Healthcare Institute, Women's Hospital of Nanjing Medical UniversityNanjing Women and Children's Healthcare HospitalNanjingJiangsuChina
| | - Zhonghua Shi
- Changzhou Maternal and Child Health Care Hospital, Changzhou Medical CenterNanjing Medical UniversityChangzhouJiangsuChina
- Changzhou Key Laboratory of Maternal and Child Health MedicineChangzhouJiangsuChina
- State Key Laboratory of Reproductive Medicine and Offspring HealthNanjing Medical UniversityNanjingJiangsuChina
| |
Collapse
|
2
|
Singh MK, Shin Y, Han S, Ha J, Tiwari PK, Kim SS, Kang I. Molecular Chaperonin HSP60: Current Understanding and Future Prospects. Int J Mol Sci 2024; 25:5483. [PMID: 38791521 PMCID: PMC11121636 DOI: 10.3390/ijms25105483] [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: 04/24/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Molecular chaperones are highly conserved across evolution and play a crucial role in preserving protein homeostasis. The 60 kDa heat shock protein (HSP60), also referred to as chaperonin 60 (Cpn60), resides within mitochondria and is involved in maintaining the organelle's proteome integrity and homeostasis. The HSP60 family, encompassing Cpn60, plays diverse roles in cellular processes, including protein folding, cell signaling, and managing high-temperature stress. In prokaryotes, HSP60 is well understood as a GroEL/GroES complex, which forms a double-ring cavity and aids in protein folding. In eukaryotes, HSP60 is implicated in numerous biological functions, like facilitating the folding of native proteins and influencing disease and development processes. Notably, research highlights its critical involvement in sustaining oxidative stress and preserving mitochondrial integrity. HSP60 perturbation results in the loss of the mitochondria integrity and activates apoptosis. Currently, numerous clinical investigations are in progress to explore targeting HSP60 both in vivo and in vitro across various disease models. These studies aim to enhance our comprehension of disease mechanisms and potentially harness HSP60 as a therapeutic target for various conditions, including cancer, inflammatory disorders, and neurodegenerative diseases. This review delves into the diverse functions of HSP60 in regulating proteo-homeostasis, oxidative stress, ROS, apoptosis, and its implications in diseases like cancer and neurodegeneration.
Collapse
Affiliation(s)
- Manish Kumar Singh
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.K.S.); (Y.S.); (S.H.); (J.H.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Centre for Genomics, SOS Zoology, Jiwaji University, Gwalior 474011, India;
| | - Yoonhwa Shin
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.K.S.); (Y.S.); (S.H.); (J.H.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sunhee Han
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.K.S.); (Y.S.); (S.H.); (J.H.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Joohun Ha
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.K.S.); (Y.S.); (S.H.); (J.H.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Pramod K. Tiwari
- Centre for Genomics, SOS Zoology, Jiwaji University, Gwalior 474011, India;
| | - Sung Soo Kim
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.K.S.); (Y.S.); (S.H.); (J.H.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Insug Kang
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.K.S.); (Y.S.); (S.H.); (J.H.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
3
|
Singh MK, Shin Y, Ju S, Han S, Choe W, Yoon KS, Kim SS, Kang I. Heat Shock Response and Heat Shock Proteins: Current Understanding and Future Opportunities in Human Diseases. Int J Mol Sci 2024; 25:4209. [PMID: 38673794 PMCID: PMC11050489 DOI: 10.3390/ijms25084209] [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: 03/15/2024] [Revised: 04/03/2024] [Accepted: 04/05/2024] [Indexed: 04/28/2024] Open
Abstract
The heat shock response is an evolutionarily conserved mechanism that protects cells or organisms from the harmful effects of various stressors such as heat, chemicals toxins, UV radiation, and oxidizing agents. The heat shock response triggers the expression of a specific set of genes and proteins known as heat shock genes/proteins or molecular chaperones, including HSP100, HSP90, HSP70, HSP60, and small HSPs. Heat shock proteins (HSPs) play a crucial role in thermotolerance and aiding in protecting cells from harmful insults of stressors. HSPs are involved in essential cellular functions such as protein folding, eliminating misfolded proteins, apoptosis, and modulating cell signaling. The stress response to various environmental insults has been extensively studied in organisms from prokaryotes to higher organisms. The responses of organisms to various environmental stressors rely on the intensity and threshold of the stress stimuli, which vary among organisms and cellular contexts. Studies on heat shock proteins have primarily focused on HSP70, HSP90, HSP60, small HSPs, and ubiquitin, along with their applications in human biology. The current review highlighted a comprehensive mechanism of heat shock response and explores the function of heat shock proteins in stress management, as well as their potential as therapeutic agents and diagnostic markers for various diseases.
Collapse
Affiliation(s)
- Manish Kumar Singh
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.K.S.); (Y.S.); (S.J.); (S.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yoonhwa Shin
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.K.S.); (Y.S.); (S.J.); (S.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Songhyun Ju
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.K.S.); (Y.S.); (S.J.); (S.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sunhee Han
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.K.S.); (Y.S.); (S.J.); (S.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Wonchae Choe
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.K.S.); (Y.S.); (S.J.); (S.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Kyung-Sik Yoon
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.K.S.); (Y.S.); (S.J.); (S.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sung Soo Kim
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.K.S.); (Y.S.); (S.J.); (S.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Insug Kang
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.K.S.); (Y.S.); (S.J.); (S.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
4
|
Timofeev YS, Kiselev AR, Dzhioeva ON, Drapkina OM. Heat Shock Proteins (HSPs) and Cardiovascular Complications of Obesity: Searching for Potential Biomarkers. Curr Issues Mol Biol 2023; 45:9378-9389. [PMID: 38132434 PMCID: PMC10742314 DOI: 10.3390/cimb45120588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 11/07/2023] [Accepted: 11/21/2023] [Indexed: 12/23/2023] Open
Abstract
Heat shock proteins (HSPs), a family of proteins that support cellular proteostasis and perform a protective function under various stress conditions, such as high temperature, intoxication, inflammation, or tissue hypoxia, constitute a promising group of possible biochemical markers for obesity and cardiovascular diseases. HSP27 is involved in essential cellular processes occurring in conditions of obesity and its cardiometabolic complications; it has protective properties, and its secretion may indicate a cellular response to stress. HSP40 plays a controversial role in the pathogenesis of obesity. HSP60 is involved in various pathological processes of the cardiovascular, immune, excretory, and nervous systems and is associated with obesity and concomitant diseases. The hypersecretion of HSP60 is associated with poor prognosis; hence, this protein may become a target for further research on obesity and its cardiovascular complications. According to most studies, intracellular HSP70 is an obesity-promoting factor, whereas extracellular HSP70 exhibited inconsistent dynamics across different patient groups and diagnoses. HSPs are involved in the pathogenesis of cardiovascular pathology. However, in the context of cardiovascular and metabolic pathology, these proteins require further investigation.
Collapse
Affiliation(s)
| | - Anton R. Kiselev
- National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia
| | | | - Oxana M. Drapkina
- National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia
| |
Collapse
|
5
|
Esmaeilzadeh A, Mohammadi V, Elahi R, Rezakhani N. The role of heat shock proteins (HSPs) in type 2 diabetes mellitus pathophysiology. J Diabetes Complications 2023; 37:108564. [PMID: 37852076 DOI: 10.1016/j.jdiacomp.2023.108564] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 07/05/2023] [Accepted: 07/21/2023] [Indexed: 10/20/2023]
Abstract
Type 2 diabetes mellitus (T2DM) is a metabolic disorder characterized by sustained hyperglycemia caused by impaired insulin signaling and secretion. Metabolic stress, caused by an inappropriate diet, is one of the major hallmarks provoking inflammation, endoplasmic reticulum (ER) stress, and mitochondrial dysfunction. Heat shock proteins (HSPs) are a group of highly conserved proteins that have a crucial role in chaperoning damaged and misfolded proteins to avoid disruption of cellular homeostasis under stress conditions. To do this, HSPs interact with diverse intra-and extracellular pathways among which are the insulin signaling, insulin secretion, and apoptosis pathways. Therefore, HSP dysfunction, e.g. HSP70, may lead to disruption of the pathways responsible for insulin secretion and uptake. Consistently, the altered expression of other HSPs and genetic polymorphisms in HSP-producing genes in diabetic subjects has made HSPs hot research in T2DM. This paper provides a comprehensive overview of the role of different HSPs in T2DM pathogenesis, affected cellular pathways, and the potential therapeutic strategies targeting HSPs in T2DM.
Collapse
Affiliation(s)
- Abdolreza Esmaeilzadeh
- Department of Immunology, Zanjan University of Medical Sciences, Zanjan, Iran; Cancer Gene Therapy Research Center (CGRC), Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Vahid Mohammadi
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Reza Elahi
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Negin Rezakhani
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
6
|
Yıldırım Ö, Tatar E. The Roles of Heat Shock Protein-60 and 70 and Inflammation in Obesity-Related Kidney Disease. Cureus 2022; 14:e28675. [PMID: 36062294 PMCID: PMC9436442 DOI: 10.7759/cureus.28675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2022] [Indexed: 11/30/2022] Open
Abstract
Introduction The exact mechanisms of obesity-related kidney disease (ORKD) are not fully known. Heat shock proteins (HSPs) may play a role in ORKD mechanisms because of their role in cell apoptosis, cytoprotection, and inflammatory processes. We aimed to determine the role of circulating serum HSP-60 and HSP-70 levels as a biomarker for ORKD. Materials and methods This study included 40 ORKD patients, 40 obese age-matched and sex-matched controls with similar body mass index (BMI), and 40 healthy controls. Their serum biochemical and hemogram parameters as well as HSP-60 and HSP-70 levels were evaluated and compared. Their neutrophil-to-lymphocyte ratio (NLR) and C-reactive protein levels were assessed to define inflammation. Results The patients had significantly higher HSP-60 levels than the obese and healthy controls (537.58 ± 170.35, 430.80 ± 110.61, and 371.85 ± 76.34, respectively; p<0.00). The results revealed that the 24-hour urinary protein levels had a positive correlation (r= 0.544), whereas the glomerular filtration rate had a negative correlation (r = 0.38) with the serum HSP-60 level. According to the regression analysis performed on the HSP-60 and 24-hour urinary protein excretion levels, an increase in the HSP-60 level significantly increased the 24-hour urinary protein excretion rate (r=0.15; p<0.005). The HSP-60 levels were correlated with inflammatory markers Conclusion The serum HSP-60 levels increased in patients with ORKD. This increase was correlated with 24-hour urinary protein excretion. Increased circulating levels of HSP-60 may play a role in the initiation and/or progression of renal damage and inflammation. HSP-60 is a potential biomarker for ORKD. However, additional information and studies are required to further elucidate this finding.
Collapse
|
7
|
Caillet C, Stofberg ML, Muleya V, Shonhai A, Zininga T. Host cell stress response as a predictor of COVID-19 infectivity and disease progression. Front Mol Biosci 2022; 9:938099. [PMID: 36032680 PMCID: PMC9411049 DOI: 10.3389/fmolb.2022.938099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/15/2022] [Indexed: 11/13/2022] Open
Abstract
The coronavirus disease (COVID-19) caused by a coronavirus identified in December 2019 has caused a global pandemic. COVID-19 was declared a pandemic in March 2020 and has led to more than 6.3 million deaths. The pandemic has disrupted world travel, economies, and lifestyles worldwide. Although vaccination has been an effective tool to reduce the severity and spread of the disease there is a need for more concerted approaches to fighting the disease. COVID-19 is characterised as a severe acute respiratory syndrome . The severity of the disease is associated with a battery of comorbidities such as cardiovascular diseases, cancer, chronic lung disease, and renal disease. These underlying diseases are associated with general cellular stress. Thus, COVID-19 exacerbates outcomes of the underlying conditions. Consequently, coronavirus infection and the various underlying conditions converge to present a combined strain on the cellular response. While the host response to the stress is primarily intended to be of benefit, the outcomes are occasionally unpredictable because the cellular stress response is a function of complex factors. This review discusses the role of the host stress response as a convergent point for COVID-19 and several non-communicable diseases. We further discuss the merits of targeting the host stress response to manage the clinical outcomes of COVID-19.
Collapse
Affiliation(s)
- Celine Caillet
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa
| | | | - Victor Muleya
- Department of Biochemistry, Midlands State University, Gweru, Zimbabwe
| | - Addmore Shonhai
- Department of Biochemistry and Microbiology, University of Venda, Thohoyandou, South Africa
| | - Tawanda Zininga
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
8
|
Liu J, Lai F, Hou Y, Zheng R. Leptin signaling and leptin resistance. MEDICAL REVIEW (BERLIN, GERMANY) 2022; 2:363-384. [PMID: 37724323 PMCID: PMC10388810 DOI: 10.1515/mr-2022-0017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 07/12/2022] [Indexed: 09/20/2023]
Abstract
With the prevalence of obesity and associated comorbidities, studies aimed at revealing mechanisms that regulate energy homeostasis have gained increasing interest. In 1994, the cloning of leptin was a milestone in metabolic research. As an adipocytokine, leptin governs food intake and energy homeostasis through leptin receptors (LepR) in the brain. The failure of increased leptin levels to suppress feeding and elevate energy expenditure is referred to as leptin resistance, which encompasses complex pathophysiological processes. Within the brain, LepR-expressing neurons are distributed in hypothalamus and other brain areas, and each population of the LepR-expressing neurons may mediate particular aspects of leptin effects. In LepR-expressing neurons, the binding of leptin to LepR initiates multiple signaling cascades including janus kinase (JAK)-signal transducers and activators of transcription (STAT) phosphatidylinositol 3-kinase (PI3K)-protein kinase B (AKT), extracellular regulated protein kinase (ERK), and AMP-activated protein kinase (AMPK) signaling, etc., mediating leptin actions. These findings place leptin at the intersection of metabolic and neuroendocrine regulations, and render leptin a key target for treating obesity and associated comorbidities. This review highlights the main discoveries that shaped the field of leptin for better understanding of the mechanism governing metabolic homeostasis, and guides the development of safe and effective interventions to treat obesity and associated diseases.
Collapse
Affiliation(s)
- Jiarui Liu
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China
| | - Futing Lai
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China
| | - Yujia Hou
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China
| | - Ruimao Zheng
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China
- Neuroscience Research Institute, Peking University, Beijing, China
- Key Laboratory for Neuroscience of Ministry of Education, Peking University, Beijing, China
- Key Laboratory for Neuroscience of National Health Commission, Peking University, Beijing 100191, China
| |
Collapse
|
9
|
Heat Shock Proteins Alterations in Rheumatoid Arthritis. Int J Mol Sci 2022; 23:ijms23052806. [PMID: 35269948 PMCID: PMC8911505 DOI: 10.3390/ijms23052806] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/01/2022] [Accepted: 03/01/2022] [Indexed: 02/06/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory and autoimmune disease characterized by the attack of the immune system on the body's healthy joint lining and degeneration of articular structures. This disease involves an increased release of inflammatory mediators in the affected joint that sensitize sensory neurons and create a positive feedback loop to further enhance their release. Among these mediators, the cytokines and neuropeptides are responsible for the crippling pain and the persistent neurogenic inflammation associated with RA. More importantly, specific proteins released either centrally or peripherally have been shown to play opposing roles in the pathogenesis of this disease: an inflammatory role that mediates and increases the severity of inflammatory response and/or an anti-inflammatory and protective role that modulates the process of inflammation. In this review, we will shed light on the neuroimmune function of different members of the heat shock protein (HSPs) family and the complex manifold actions that they exert during the course of RA. Specifically, we will focus our discussion on the duality in the mechanism of action of Hsp27, Hsp60, Hsp70, and Hsp90.
Collapse
|
10
|
Manfredi LH. Overheating or overcooling: heat transfer in the spot to fight against the pandemic obesity. Rev Endocr Metab Disord 2021; 22:665-680. [PMID: 33000381 DOI: 10.1007/s11154-020-09596-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/17/2020] [Indexed: 12/25/2022]
Abstract
The prevalence of obesity has nearly doubled worldwide over the past three and a half decades, reaching pandemic status. Obesity is associated with decreased life expectancy and with an increased risk of metabolic, cardiovascular, nervous system diseases. Hence, understanding the mechanisms involved in the onset and development of obesity is mandatory to promote planned health actions to revert this scenario. In this review, common aspects of cold exposure, a process of heat generation, and exercise, a process of heat dissipation, will be discussed as two opposite mechanisms of obesity, which can be oversimplified as caloric conservation. A common road between heat generation and dissipation is the mobilization of Free Faty Acids (FFA) and Carbohydrates (CHO). An increase in energy expenditure (immediate effect) and molecular/metabolic adaptations (chronic effect) are responses that depend on SNS activity in both conditions of heat transfer. This cycle of using and removing FFA and CHO from blood either for heat or force generation disrupt the key concept of obesity: energy accumulation. Despite efforts in making the anti-obesity pill, maybe it is time to consider that the world's population is living at thermoneutrality since temperature-controlled places and the lack of exercise are favoring caloric accumulation.
Collapse
Affiliation(s)
- Leandro Henrique Manfredi
- Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, Santa Catarina, Brazil.
| |
Collapse
|
11
|
Cruciani S, Garroni G, Pala R, Cossu ML, Ginesu GC, Ventura C, Maioli M. Metformin and Vitamin D Modulate Inflammation and Autophagy during Adipose-Derived Stem Cell Differentiation. Int J Mol Sci 2021; 22:6686. [PMID: 34206506 PMCID: PMC8269127 DOI: 10.3390/ijms22136686] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/17/2021] [Accepted: 06/18/2021] [Indexed: 12/21/2022] Open
Abstract
Adipose-derived stem cells (ADSCs) came out from the regenerative medicine landscape for their ability to differentiate into several phenotypes, contributing to tissue regeneration both in vitro and in vivo. Dysregulation in stem cell recruitment and differentiation during adipogenesis is linked to a chronic low-grade inflammation and macrophage infiltration inside the adipose tissue, insulin resistance, cardiovascular disease and obesity. In the present paper we aimed to evaluate the role of metformin and vitamin D, alone or in combination, in modulating inflammation and autophagy in ADSCs during adipogenic commitment. ADSCs were cultured for 21 days in the presence of a specific adipogenic differentiation medium, together with metformin, or vitamin D, or both. We then analyzed the expression of FoxO1 and Heat Shock Proteins (HSP) and the secretion of proinflammatory cytokines IL-6 and TNF-α by ELISA. Autophagy was also assessed by specific Western blot analysis of ATG12, LC3B I, and LC3B II expression. Our results showed the ability of the conditioned media to modulate adipogenic differentiation, finely tuning the inflammatory response and autophagy. We observed a modulation in HSP mRNA levels, and a significant downregulation in cytokine secretion. Taken together, our findings suggest the possible application of these molecules in clinical practice to counteract uncontrolled lipogenesis and prevent obesity and obesity-related metabolic disorders.
Collapse
Affiliation(s)
- Sara Cruciani
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; (S.C.); (G.G.); (R.P.)
| | - Giuseppe Garroni
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; (S.C.); (G.G.); (R.P.)
| | - Renzo Pala
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; (S.C.); (G.G.); (R.P.)
| | - Maria Laura Cossu
- General Surgery Unit 2 “Clinica Chirurgica”, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Viale San Pietro 8, 07100 Sassari, Italy; (M.L.C.); (G.C.G.)
| | - Giorgio Carlo Ginesu
- General Surgery Unit 2 “Clinica Chirurgica”, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Viale San Pietro 8, 07100 Sassari, Italy; (M.L.C.); (G.C.G.)
| | - Carlo Ventura
- Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems Eldor Lab, Innovation Accelerator, Consiglio Nazionale delle Ricerche, 40129 Bologna, Italy;
| | - Margherita Maioli
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; (S.C.); (G.G.); (R.P.)
- Center for Developmental Biology and Reprogramming (CEDEBIOR), Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy
| |
Collapse
|
12
|
HSP60 reduction protects against diet-induced obesity by modulating energy metabolism in adipose tissue. Mol Metab 2021; 53:101276. [PMID: 34153520 PMCID: PMC8319365 DOI: 10.1016/j.molmet.2021.101276] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/06/2021] [Accepted: 06/15/2021] [Indexed: 11/29/2022] Open
Abstract
Objective Insulin regulates mitochondrial function, thereby propagating an efficient metabolism. Conversely, diabetes and insulin resistance are linked to mitochondrial dysfunction with a decreased expression of the mitochondrial chaperone HSP60. The aim of this investigation was to determine the effect of a reduced HSP60 expression on the development of obesity and insulin resistance. Methods Control and heterozygous whole-body HSP60 knockout (Hsp60+/−) mice were fed a high-fat diet (HFD, 60% calories from fat) for 16 weeks and subjected to extensive metabolic phenotyping. To understand the effect of HSP60 on white adipose tissue, microarray analysis of gonadal WAT was performed, ex vivo experiments were performed, and a lentiviral knockdown of HSP60 in 3T3-L1 cells was conducted to gain detailed insights into the effect of reduced HSP60 levels on adipocyte homeostasis. Results Male Hsp60+/− mice exhibited lower body weight with lower fat mass. These mice exhibited improved insulin sensitivity compared to control, as assessed by Matsuda Index and HOMA-IR. Accordingly, insulin levels were significantly reduced in Hsp60+/− mice in a glucose tolerance test. However, Hsp60+/− mice exhibited an altered adipose tissue metabolism with elevated insulin-independent glucose uptake, adipocyte hyperplasia in the presence of mitochondrial dysfunction, altered autophagy, and local insulin resistance. Conclusions We discovered that the reduction of HSP60 in mice predominantly affects adipose tissue homeostasis, leading to beneficial alterations in body weight, body composition, and adipocyte morphology, albeit exhibiting local insulin resistance. Mice with reduced HSP60 levels are protected from diet-induced obesity. Hsp60+/− mice exhibit altered adipose tissue energy metabolism. WAT of Hsp60+/− mice exhibit elevated insulin-independent glucose uptake. Hsp60+/− mice show improved global, but impaired WAT insulin action.
Collapse
|
13
|
Ghadimi D, Nielsen A, Hassan MFY, Fölster-Holst R, Ebsen M, Frahm SO, Röcken C, de Vrese M, Heller KJ. Modulation of Proinflammatory Bacteria- and Lipid-Coupled Intracellular Signaling Pathways in a Transwell Triple Co-Culture Model by Commensal Bifidobacterium Animalis R101-8. Antiinflamm Antiallergy Agents Med Chem 2021; 20:161-181. [PMID: 33135616 DOI: 10.2174/1871523019999201029115618] [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: 06/08/2020] [Revised: 09/22/2020] [Accepted: 09/30/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND AND AIMS Following a fat-rich diet, alterations in gut microbiota contribute to enhanced gut permeability, metabolic endotoxemia, and low grade inflammation-associated metabolic disorders. To better understand whether commensal bifidobacteria influence the expression of key metaflammation-related biomarkers (chemerin, MCP-1, PEDF) and modulate the pro-inflammatory bacteria- and lipid-coupled intracellular signaling pathways, we aimed at i) investigating the influence of the establishment of microbial signaling molecules-based cell-cell contacts on the involved intercellular communication between enterocytes, immune cells, and adipocytes, and ii) assessing their inflammatory mediators' expression profiles within an inflamed adipose tissue model. MATERIAL AND METHODS Bifidobacterium animalis R101-8 and Escherichia coli TG1, respectively, were added to the apical side of a triple co-culture model consisting of intestinal epithelial HT-29/B6 cell line, human monocyte-derived macrophage cells, and adipose-derived stem cell line in the absence or presence of LPS or palmitic acid. mRNA expression levels of key lipid metabolism genes HILPDA, MCP-1/CCL2, RARRES2, SCD, SFRP2 and TLR4 were determined using TaqMan qRT-PCR. Protein expression levels of cytokines (IL-1β, IL-6, and TNF-α), key metaflammation-related biomarkers including adipokines (chemerin and PEDF), chemokine (MCP- 1) as well as cellular triglycerides were assessed by cell-based ELISA, while those of p-ERK, p-JNK, p-p38, NF-κB, p-IκBα, pc-Fos, pc-Jun, and TLR4 were assessed by Western blotting. RESULTS B. animalis R101-8 inhibited LPS- and palmitic acid-induced protein expression of inflammatory cytokines IL-1β, IL-6, TNF-α concomitant with decreases in chemerin, MCP-1, PEDF, and cellular triglycerides, and blocked NF-kB and AP-1 activation pathway through inhibition of p- IκBα, pc-Jun, and pc-Fos phosphorylation. B. animalis R101-8 downregulated mRNA and protein levels of HILPDA, MCP-1/CCL2, RARRES2, SCD and SFRP2 and TLR4 following exposure to LPS and palmitic acid. CONCLUSION B. animalis R101-8 improves biomarkers of metaflammation through at least two molecular/signaling mechanisms triggered by pro-inflammatory bacteria/lipids. First, B. animalis R101-8 modulates the coupled intracellular signaling pathways via metabolizing saturated fatty acids and reducing available bioactive palmitic acid. Second, it inhibits NF-kB's and AP-1's transcriptional activities, resulting in the reduction of pro-inflammatory markers. Thus, the molecular basis may be formed by which commensal bifidobacteria improve intrinsic cellular tolerance against excess pro-inflammatory lipids and participate in homeostatic regulation of metabolic processes in vivo.
Collapse
Affiliation(s)
- Darab Ghadimi
- Department of Microbiology and Biotechnology, Max Rubner-Institute, Hermann-Weigmann-Str 1, D-24103 Kiel, Germany
| | - Annegret Nielsen
- Department of Microbiology and Biotechnology, Max Rubner-Institute, Hermann-Weigmann-Str 1, D-24103 Kiel, Germany
| | | | - Regina Fölster-Holst
- Clinic of Dermatology, University Hospital Schleswig-Holstein, Schittenhelmstr. 7, D-24105 Kiel, Germany
| | - Michael Ebsen
- Department of Pathology, Städtisches MVZ Kiel GmbH (Kiel City Hospital), Chemnitzstr.33, 24116 Kiel, Germany
| | - Sven Olaf Frahm
- Medizinisches Versorgungszentrum (MVZ), Pathology and Laboratory Medicine Dr. Rabenhorst, Prüner Gang 7, 24103 Kiel, Germany
| | - Christoph Röcken
- Institute of Pathology, Kiel University, University Hospital, Schleswig-Holstein, Arnold-Heller-Straße 3/14, D-24105 Kiel, Germany
| | - Michael de Vrese
- Department of Microbiology and Biotechnology, Max Rubner-Institute, Hermann-Weigmann-Str 1, D-24103 Kiel, Germany
| | - Knut J Heller
- Department of Microbiology and Biotechnology, Max Rubner-Institute, Hermann-Weigmann-Str 1, D-24103 Kiel, Germany
| |
Collapse
|
14
|
Olea-Flores M, Juárez-Cruz JC, Zuñiga-Eulogio MD, Acosta E, García-Rodríguez E, Zacapala-Gomez AE, Mendoza-Catalán MA, Ortiz-Ortiz J, Ortuño-Pineda C, Navarro-Tito N. New Actors Driving the Epithelial-Mesenchymal Transition in Cancer: The Role of Leptin. Biomolecules 2020; 10:E1676. [PMID: 33334030 PMCID: PMC7765557 DOI: 10.3390/biom10121676] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/12/2020] [Accepted: 12/13/2020] [Indexed: 12/24/2022] Open
Abstract
Leptin is a hormone secreted mainly by adipocytes; physiologically, it participates in the control of appetite and energy expenditure. However, it has also been linked to tumor progression in different epithelial cancers. In this review, we describe the effect of leptin on epithelial-mesenchymal transition (EMT) markers in different study models, including in vitro, in vivo, and patient studies and in various types of cancer, including breast, prostate, lung, and ovarian cancer. The different studies report that leptin promotes the expression of mesenchymal markers and a decrease in epithelial markers, in addition to promoting EMT-related processes such as cell migration and invasion and poor prognosis in patients with cancer. Finally, we report that leptin has the greatest biological relevance in EMT and tumor progression in breast, lung, prostate, esophageal, and ovarian cancer. This relationship could be due to the key role played by the enriched tumor microenvironment in adipose tissue. Together, these findings demonstrate that leptin is a key biomolecule that drives EMT and metastasis in cancer.
Collapse
Affiliation(s)
- Monserrat Olea-Flores
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39090, Mexico; (M.O.-F.); (J.C.J.-C.); (M.D.Z.-E.); (E.A.); (E.G.-R.)
| | - Juan C. Juárez-Cruz
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39090, Mexico; (M.O.-F.); (J.C.J.-C.); (M.D.Z.-E.); (E.A.); (E.G.-R.)
| | - Miriam D. Zuñiga-Eulogio
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39090, Mexico; (M.O.-F.); (J.C.J.-C.); (M.D.Z.-E.); (E.A.); (E.G.-R.)
| | - Erika Acosta
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39090, Mexico; (M.O.-F.); (J.C.J.-C.); (M.D.Z.-E.); (E.A.); (E.G.-R.)
| | - Eduardo García-Rodríguez
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39090, Mexico; (M.O.-F.); (J.C.J.-C.); (M.D.Z.-E.); (E.A.); (E.G.-R.)
| | - Ana E. Zacapala-Gomez
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39090, Mexico; (A.E.Z.-G.); (M.A.M.-C.); (J.O.-O.)
| | - Miguel A. Mendoza-Catalán
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39090, Mexico; (A.E.Z.-G.); (M.A.M.-C.); (J.O.-O.)
| | - Julio Ortiz-Ortiz
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39090, Mexico; (A.E.Z.-G.); (M.A.M.-C.); (J.O.-O.)
| | - Carlos Ortuño-Pineda
- Laboratorio de Ácidos Nucleicos y Proteinas, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39090, Mexico;
| | - Napoleón Navarro-Tito
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39090, Mexico; (M.O.-F.); (J.C.J.-C.); (M.D.Z.-E.); (E.A.); (E.G.-R.)
| |
Collapse
|
15
|
Qu J, Wang W, Zhang Q, Li S. Inhibition of Lipopolysaccharide-Induced Inflammation of Chicken Liver Tissue by Selenomethionine via TLR4-NF-κB-NLRP3 Signaling Pathway. Biol Trace Elem Res 2020; 195:205-214. [PMID: 31332706 DOI: 10.1007/s12011-019-01841-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 07/16/2019] [Indexed: 02/08/2023]
Abstract
Selenium (Se) is important in many physiological processes, such as antioxidant processes and inflammation. The aim of our experiments was to investigate the molecular mechanism that selenomethionine could reduce the lipopolysaccharide (LPS)-induced inflammation by inhibiting the TLR4-NF-κB-NLRP3 signaling pathway. Eighty broilers were randomly and evenly divided into two groups, giving normal Se content diets (Con group, 0.2 mg Se/kg diet) and Se-rich basal diets (Se group, 0.5 mg selenomethionine/kg diet) for 90 days. Se-rich basal diets were based on 0.2 mg/kg sodium selenite contained. Five hours before euthanized, 20 broilers were randomly selected from each group and given lipopolysaccharide (200 μg/kg BW) by intraperitoneal injection, Con+LPS group and Se+LPS group, respectively. The Con group and Se group were given equal saline by intraperitoneal injection. We observed the microscopic pathological changes of liver tissue detected oxidative stress by kit and detected the expression of inflammatory factors, heat shock protein (HSP), and nod-like receptor protein 3 (NLRP3)-related genes by qRT-PCR and Western blot. With the microscope, we found the Con+LPS group had obvious inflammatory lesions such as sinusoidal congestion, but the damage was significantly alleviated in the Se+LPS group. In the Con+LPS group, the activity of GSH-Px and the content of GSH were significantly decreased compared with those in the Con group; however, they are increased in the Se group and in the Se + LPS group. Inflammatory factors (MyD88, NF-κB, TNF-α, IL-1β, IL-6, IL-12, IL-18, iNOS, and COX-2), heat shock proteins (HSP27, HSP60, HSP70, and HSP90), and the expression of NLRP3 and caspase-1 increased in the Con+LPS group compared with those in the Con group, while they were lower in the Se+LPS group than in the Con+LPS group. We concluded that selenomethionine inhibits the LPS-induced inflammation of liver tissue via suppressing the TLR4-NF-κB-NLRP3 signaling pathway.
Collapse
Affiliation(s)
- Jingrui Qu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Wei Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Qiaojian Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Shu Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China.
- Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China.
| |
Collapse
|
16
|
Zhang D, Liu H, Zhang Y, Li J, Fu Y, Zheng Y, Wu J, Ma M, Wen Z, Wang C. Heat shock protein 60 (HSP60) modulates adiponectin signaling by stabilizing adiponectin receptor. Cell Commun Signal 2020; 18:60. [PMID: 32272950 PMCID: PMC7147001 DOI: 10.1186/s12964-020-00546-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/06/2020] [Indexed: 12/11/2022] Open
Abstract
Adiponectin, an adipokine produced and secreted by adipocytes, is involved in regulating the development and progression of insulin resistance, diabetes, and diabetic complications. Heat shock protein 60 (HSP60) is a molecular chaperone, most commonly presenting in mitochondria and participating in the maintenance of protein homeostasis. Accumulating studies have demonstrated that the elevated circulating HSP60 and the decreased intracellular HSP60 are closely associated with diabetic complications such as diabetic cardiomyopathy. However, the underlying mechanism remains poorly understood. In the present study, we reported that HSP60 interacted directly with adiponectin receptors. Its abundance was positively associated with adiponectin action. Furthermore, HSP60 depletion markedly mitigated the protective impacts of adiponectin on high glucose-induced oxidative stress and cell apoptosis in rat cardiac H9c2 cells. In addition, HSP60 knockdown significantly enhanced proteasome activity leading to the degradation of adiponectin receptor 1. Taken together, we showed for the first time that HSP60 interacted with adiponectin receptors and mediated adiponectin signaling through stabilizing adiponectin receptor. This in vitro study also provides an alternative explanation for mechanism by which adiponectin exerts its action. Video abstract
Collapse
Affiliation(s)
- Deling Zhang
- Department of Pathology & Pathophysiology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Hua Liu
- Department of Clinical Pathology, The First People's Hospital of Lianyungang, Lianyungang, 222061, China
| | - Yemin Zhang
- Department of Pathology & Pathophysiology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Junfeng Li
- Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yalin Fu
- Department of Pathology & Pathophysiology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Yuyang Zheng
- Department of Pathology & Pathophysiology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Jie Wu
- Department of Pathology & Pathophysiology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Mingke Ma
- Department of Pathology & Pathophysiology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Zhongyuan Wen
- Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Changhua Wang
- Department of Pathology & Pathophysiology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China. .,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| |
Collapse
|
17
|
Shi X, Wang W, Zheng S, Zhang Q, Xu S. Selenomethionine relieves inflammation in the chicken trachea caused by LPS though inhibiting the NF-κB pathway. Biol Trace Elem Res 2020; 194:525-535. [PMID: 31325027 DOI: 10.1007/s12011-019-01789-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 06/18/2019] [Indexed: 12/22/2022]
Abstract
Selenomethionine is able to relieve the effect of inflammation in various tissues and organs. However, there are few studies about the influences of organic selenium resisting inflammation induced by LPS in chicken trachea. Therefore, the purpose of this experiment is to explore the organic selenium (selenomethionine) can raise immune function and relieve the LPS-induced inflammation of chicken trachea via inhibiting the NF-κB pathway. To investigate the mechanism of organic selenium on chicken trachea, the supplement of selenomethionine and/or LPS-induced chicken models were established. One hundred 46-week-old isa chickens were randomly divided into four groups (n = 25). The four groups were the control group, the selenomethionine group (Se group), the LPS-induced group (LPS group), and the Se and LPS interaction group (Se + LPS group). Then, the expressions of inflammatory factors (including induced nitric oxide synthase (iNOS), nuclear factor-kappa B(NF-κB), tumor necrosis factor (TNF-α), cyclooxygenase-2 (COX-2), and prostaglandin E (PTGEs) synthase), inflammation-related cytokines (including interleukin (IL-2, IL-6, IL-8, IL-17) and immunoglobulin (IgA, IgM, IgY)), the marker of immune function (avian β-defensins (AvBD6, AvBD7)), heat shock proteins (including HSP60, HSP90), and selenoproteins (including Selo, Sels, Selm, Selh, Selu, Seli, SPS2, GPx1, GPx2, Dio1, Sepx1, Sep15, Sepp1, Txnrd1) were detected in our experiment. The above genes were significantly changed in different groups (p < 0.05). We can conclude that organic selenium can increase the function of immunity and the expression of selenoproteins, and mitigate the inflammation induced by LPS via suppression of the NF-κB pathway.
Collapse
Affiliation(s)
- Xu Shi
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Wei Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Shufang Zheng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Qiaojian Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Shiwen Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China.
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China.
| |
Collapse
|
18
|
Xiao T, Liang X, Liu H, Zhang F, Meng W, Hu F. Mitochondrial stress protein HSP60 regulates ER stress-induced hepatic lipogenesis. J Mol Endocrinol 2020; 64:67-75. [PMID: 31804966 PMCID: PMC6993205 DOI: 10.1530/jme-19-0207] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 12/04/2019] [Indexed: 01/15/2023]
Abstract
Endoplasmic reticulum (ER) stress and mitochondrial dysfunction are associated with hepatic steatosis and insulin resistance. Molecular mechanisms underlying ER stress and/or mitochondrial dysfunction that cause metabolic disorders and hepatic steatosis remain to be fully understood. Here, we found that a high fat diet (HFD) or chemically induced ER stress can stimulate mitochondrial stress protein HSP60 expression, impair mitochondrial respiration, and decrease mitochondrial membrane potential in mouse hepatocytes. HSP60 overexpression promotes ER stress and hepatic lipogenic protein expression and impairs insulin signaling in mouse hepatocytes. Mechanistically, HSP60 regulates ER stress-induced hepatic lipogenesis via the mTORC1-SREBP1 signaling pathway. These results suggest that HSP60 is an important ER and mitochondrial stress cross-talking protein and may control ER stress-induced hepatic lipogenesis and insulin resistance.
Collapse
Affiliation(s)
- Ting Xiao
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiuci Liang
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hailan Liu
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Feng Zhang
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wen Meng
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Correspondence should be addressed to F Hu: or to W Meng:
| | - Fang Hu
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Metabolic Diseases, Changsha, Hunan, China
- Metabolic Syndrome Research Center, Central South University, Changsha, Hunan, China
- Correspondence should be addressed to F Hu: or to W Meng:
| |
Collapse
|
19
|
Landa-Galvan HV, Rios-Castro E, Romero-Garcia T, Rueda A, Olivares-Reyes JA. Metabolic syndrome diminishes insulin-induced Akt activation and causes a redistribution of Akt-interacting proteins in cardiomyocytes. PLoS One 2020; 15:e0228115. [PMID: 31995605 PMCID: PMC6988918 DOI: 10.1371/journal.pone.0228115] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 01/07/2020] [Indexed: 12/31/2022] Open
Abstract
Metabolic syndrome (MetS) is a cluster of cardiometabolic risk factors, with insulin resistance as a critical component for its development. Insulin signaling in the heart leads to Akt (also known as PKB) activation, a serine/threonine protein kinase, which regulates cardiac glucose metabolism and growth. Cardiac metabolic inflexibility, characterized by impaired insulin-induced glucose uptake and oxidation, has been reported as an early and consistent change in the heart of different models of MetS and diabetes; however, the evaluation of Akt activation has yielded variable results. Here we report in cardiomyocytes of MetS rats, diminished insulin-induced glucose uptake and Akt activation, evaluated by its impaired mobilization towards the plasma membrane and phosphorylation, and reflected in a re-distribution of its interacting proteins, assessed by label-free mass spectrometry (data are available via ProteomeXchange with identifier PXD013260). We report 45 proteins with diminished abundance in Akt complex of MetS cardiomyocytes, mainly represented by energy metabolism-related proteins, and also, 31 Akt-interacting proteins with increased abundance, which were mainly related to contraction, endoplasmic reticulum stress, and Akt negative regulation. These results emphasize the relevance of Akt in the regulation of energy metabolism in the heart and highlight Akt-interacting proteins that could be involved in the detrimental effects of MetS in the heart.
Collapse
Affiliation(s)
| | - Emmanuel Rios-Castro
- Unidad de Genomica, Proteomica y Metabolomica (UGPM), LaNSE-Cinvestav-IPN, Mexico City, Mexico
| | | | - Angelica Rueda
- Departamento de Bioquimica, Cinvestav-IPN, Mexico City, Mexico
| | | |
Collapse
|
20
|
Sun Q, Fang L, Roth M, Tang X, Papakonstantinou E, Zhai W, Louis R, Heinen V, Schleich FN, Lu S, Savic S, Tamm M, Stolz D. Bronchial thermoplasty decreases airway remodelling by blocking epithelium-derived heat shock protein-60 secretion and protein arginine methyltransferase-1 in fibroblasts. Eur Respir J 2019; 54:13993003.00300-2019. [PMID: 31467116 DOI: 10.1183/13993003.00300-2019] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 08/13/2019] [Indexed: 02/04/2023]
Abstract
Bronchial thermoplasty (BT) is to date the only therapy that provides a lasting reduction in airway wall remodelling. However, the mechanism of action of BT is not well understood. This study aimed to characterise the changes of remodelling regulating signalling pathways by BT in asthma.Bronchoalveolar lavage fluid (BALF) was obtained from eight patients with severe asthma before and after BT. Primary bronchial epithelial cells were isolated from 23 patients before (n=66) and after (n=62) BT. Epithelial cell culture supernatant (Epi.S) was collected and applied to primary fibroblasts.Epithelial cells obtained from asthma patients after BT proliferated significantly faster compared with epithelial cells obtained before BT. In airway fibroblasts, BALF or Epi.S obtained before BT increased CCAAT enhancer-binding protein-β (C/EBPβ) expression, thereby downregulating microRNA-19a. This upregulated extracellular signal-regulated kinase-1/2 (ERK1/2) expression, protein arginine methyltransferase-1 (PRMT1) expression, cell proliferation and mitochondrial mass. BALF or Epi.S obtained after BT reduced the expression of C/EBPβ, ERK1/2, peroxisome proliferator-activated receptor-γ coactivator-1α (PGC1α), PRMT1 and mitochondrial mass in airway fibroblasts. Proteome and transcriptome analyses indicated that epithelial cell-derived heat shock protein-60 (HSP60) is the main mediator of BT effects on fibroblasts. Further analysis suggested that HSP60 regulated PRMT1 expression, which was responsible for the increased mitochondrial mass and α-smooth muscle actin expression by asthmatic fibroblasts. These effects were ablated after BT. These results imply that BT reduces fibroblast remodelling through modifying the function of epithelial cells, especially by reducing HSP60 secretion and subsequent signalling pathways that regulate PRMT1 expression.We therefore hypothesise that BT decreases airway remodelling by blocking epithelium-derived HSP60 secretion and PRMT1 in fibroblasts.
Collapse
Affiliation(s)
- Qingzhu Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling, China.,Pneumology and Pulmonary Cell Research, Depts of Internal Medicine and Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland.,These authors contributed equally to this work
| | - Lei Fang
- Pneumology and Pulmonary Cell Research, Depts of Internal Medicine and Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland.,These authors contributed equally to this work
| | - Michael Roth
- Pneumology and Pulmonary Cell Research, Depts of Internal Medicine and Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Xuemei Tang
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Eleni Papakonstantinou
- Pneumology and Pulmonary Cell Research, Depts of Internal Medicine and Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Weiqi Zhai
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Renaud Louis
- Dept of Pneumology, University of Liege, Liege, Belgium
| | | | | | - Shemin Lu
- Dept of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Centre, Xi'an, China
| | - Spasenjia Savic
- Dept of Pathology, University Hospital Basel, Basel, Switzerland
| | - Michael Tamm
- Pneumology and Pulmonary Cell Research, Depts of Internal Medicine and Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Daiana Stolz
- Pneumology and Pulmonary Cell Research, Depts of Internal Medicine and Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| |
Collapse
|
21
|
Cheng H, Qi T, Zhang X, Kong Q, Min X, Mao Q, Cao X, Liu L, Ding Z. Deficiency of heat shock protein A12A promotes browning of white adipose tissues in mice. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1451-1459. [DOI: 10.1016/j.bbadis.2019.02.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 01/30/2019] [Accepted: 02/19/2019] [Indexed: 02/08/2023]
|
22
|
Moura CS, Lollo PCB, Morato PN, Amaya-Farfan J. Dietary Nutrients and Bioactive Substances Modulate Heat Shock Protein (HSP) Expression: A Review. Nutrients 2018; 10:nu10060683. [PMID: 29843396 PMCID: PMC6024325 DOI: 10.3390/nu10060683] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 05/21/2018] [Accepted: 05/23/2018] [Indexed: 01/06/2023] Open
Abstract
Interest in the heat shock proteins (HSPs), as a natural physiological toolkit of living organisms, has ranged from their chaperone function in nascent proteins to the remedial role following cell stress. As part of the defence system, HSPs guarantee cell tolerance against a variety of stressors, including exercise, oxidative stress, hyper and hypothermia, hyper and hypoxia and improper diets. For the past couple of decades, research on functional foods has revealed a number of substances likely to trigger cell protection through mechanisms that involve the induction of HSP expression. This review will summarize the occurrence of the most easily inducible HSPs and describe the effects of dietary proteins, peptides, amino acids, probiotics, high-fat diets and other food-derived substances reported to induce HSP response in animals and humans studies. Future research may clarify the mechanisms and explore the usefulness of this natural alternative of defense and the modulating mechanism of each substance.
Collapse
Affiliation(s)
- Carolina Soares Moura
- Protein Resources Laboratory, Food and Nutrition Department, Faculty of Food Engineering, University of Campinas (UNICAMP), Campinas 13083-862 São Paulo, Brazil.
| | | | - Priscila Neder Morato
- School of Health Sciences, Federal University of Grande Dourados, Dourados 79825-070, Mato Grosso do Sul, Brazil.
| | - Jaime Amaya-Farfan
- Protein Resources Laboratory, Food and Nutrition Department, Faculty of Food Engineering, University of Campinas (UNICAMP), Campinas 13083-862 São Paulo, Brazil.
| |
Collapse
|
23
|
Alti D, Sambamurthy C, Kalangi SK. Emergence of Leptin in Infection and Immunity: Scope and Challenges in Vaccines Formulation. Front Cell Infect Microbiol 2018; 8:147. [PMID: 29868503 PMCID: PMC5954041 DOI: 10.3389/fcimb.2018.00147] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 04/20/2018] [Indexed: 01/01/2023] Open
Abstract
Deficiency of leptin (ob/ob) and/or desensitization of leptin signaling (db/db) and elevated expression of suppressor of cytokine signaling-3 (SOCS3) reported in obesity are also reported in a variety of pathologies including hypertriglyceridemia, insulin resistance, and malnutrition as the risk factors in host defense system. Viral infections cause the elevated SOCS3 expression, which inhibits leptin signaling. It results in immunosuppression by T-regulatory cells (Tregs). The host immunity becomes incompetent to manage pathogens' attack and invasion, which results in the accelerated infections and diminished vaccine-specific antibody response. Leptin was successfully used as mucosal vaccine adjuvant against Rhodococcus equi. Leptin induced the antibody response to Helicobacter pylori vaccination in mice. An integral leptin signaling in mucosal gut epithelial cells offered resistance against Clostridium difficile and Entameoba histolytica infections. We present in this review, the intervention of leptin in lethal diseases caused by microbial infections and propose the possible scope and challenges of leptin as an adjuvant tool in the development of effective vaccines.
Collapse
Affiliation(s)
- Dayakar Alti
- School of Life Sciences, University of Hyderabad, Hyderabad, India
| | | | - Suresh K Kalangi
- School of Life Sciences, University of Hyderabad, Hyderabad, India
| |
Collapse
|
24
|
Zhou C, Sun H, Zheng C, Gao J, Fu Q, Hu N, Shao X, Zhou Y, Xiong J, Nie K, Zhou H, Shen L, Fang H, Lyu J. Oncogenic HSP60 regulates mitochondrial oxidative phosphorylation to support Erk1/2 activation during pancreatic cancer cell growth. Cell Death Dis 2018; 9:161. [PMID: 29415987 PMCID: PMC5833694 DOI: 10.1038/s41419-017-0196-z] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 11/05/2017] [Accepted: 12/05/2017] [Indexed: 12/25/2022]
Abstract
HSP60 is a mitochondrial localized quality control protein responsible for maintaining mitochondrial function. Although HSP60 is considered both a tumor suppressor and promoter in different types of cancer, the role of HSP60 in human pancreatic ductal adenocarcinoma (PDAC) remains unknown. In this study, we demonstrated that HSP60 was aberrantly expressed in human pancreatic cancer tissues and cell lines. Analysis of the Cancer Genome Atlas database revealed that HSP60 expression is positively correlated with pancreatic cancer. Further, knockdown of HSP60 attenuated pancreatic ductal cancer cell proliferation and migration/invasion, whereas ectopic expression of HSP60 increased tumorigenesis. Using an in vivo tumorigenicity assay, we confirmed that HSP60 promoted the growth of pancreatic ductal cancer cells. Functional analyses demonstrated that HSP60 plays a key role in the regulation of mitochondrial function. Mechanistically, both HSP60 knockdown and oxidative phosphorylation (OXPHOS) inhibition by metformin decreased Erk1/2 phosphorylation and induced apoptosis and cell cycle arrest, whereas Erk1/2 reactivation with EGF promoted cell proliferation. Intriguingly, in vitro ATP supplementation partially restored Erk1/2 phosphorylation and promoted proliferation in PDAC cells with HSP60 knockdown and OXPHOS inhibition. These results suggest that mitochondrial ATP is an important sensor of Erk1/2 regulated apoptosis and the cell cycle in PDAC cells. Thus, our findings indicate for the first time that HSP60 may serve as a novel diagnostic target of human pancreatic cancer, and that inhibition of mitochondrial function using drugs such as metformin may be a beneficial therapeutic strategy targeting pancreatic cancer cells with aberrant function of the HSP60/OXPHOS/Erk1/2 phosphorylation axis.
Collapse
Affiliation(s)
- Chao Zhou
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Department of Clinical Laboratory, Children's Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Hongwei Sun
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chen Zheng
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jing Gao
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qingzi Fu
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Nianqi Hu
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaoli Shao
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yingying Zhou
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jingting Xiong
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ke Nie
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Huaibin Zhou
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lijun Shen
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hezhi Fang
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Jianxin Lyu
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China. .,Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
25
|
Khadir A, Kavalakatt S, Cherian P, Warsame S, Abubaker JA, Dehbi M, Tiss A. Physical Exercise Enhanced Heat Shock Protein 60 Expression and Attenuated Inflammation in the Adipose Tissue of Human Diabetic Obese. Front Endocrinol (Lausanne) 2018; 9:16. [PMID: 29467719 PMCID: PMC5808138 DOI: 10.3389/fendo.2018.00016] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 01/15/2018] [Indexed: 12/17/2022] Open
Abstract
Heat shock protein 60 (HSP60) is a key protein in the crosstalk between cellular stress and inflammation. However, the status of HSP60 in diabetes and obesity is unclear. In the present study, we investigated the hypothesis that HSP60 expression levels in the adipose tissue of human obese adults with and without diabetes are different and physical exercise might affect these levels. Subcutaneous adipose tissue (SAT) and blood samples were collected from obese adults with and without diabetes (n = 138 and n = 92, respectively, at baseline; n = 43 for both groups after 3 months of physical exercise). Conventional RT-PCR, immunohistochemistry, immunofluorescence, and ELISA were used to assess the expression and secretion of HSP60. Compared with obese adults without diabetes, HSP60 mRNA and protein levels were decreased in SAT in diabetic obese together with increased inflammatory marker expression and glycemic levels but lower VO2 Max. More interestingly, a 3-month physical exercise differentially affected HSP60 expression and the heat shock response but attenuated inflammation in both groups, as reflected by decreased endogenous levels of IL-6 and TNF-α. Indeed, HSP60 expression levels in SAT were significantly increased by exercise in the diabetes group, whereas they were decreased in the non-diabetes group. These results were further confirmed using immunofluorescence microscopy and anti-HSP60 antibody in SAT. Exercise had only marginal effects on HSP60 secretion and HSP60 autoantibody levels in plasma in both obese with and without diabetes. Physical exercise differentially alleviates cellular stress in obese adults with and without diabetes despite concomitant attenuation of the inflammatory response.
Collapse
Affiliation(s)
- Abdelkrim Khadir
- Research Division, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Sina Kavalakatt
- Research Division, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Preethi Cherian
- Research Division, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Samia Warsame
- Research Division, Dasman Diabetes Institute, Kuwait City, Kuwait
| | | | - Mohammed Dehbi
- Diabetes Research Centre, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Ali Tiss
- Research Division, Dasman Diabetes Institute, Kuwait City, Kuwait
| |
Collapse
|
26
|
Petrus P, Bialesova L, Checa A, Kerr A, Naz S, Bäckdahl J, Gracia A, Toft S, Dahlman-Wright K, Hedén P, Dahlman I, Wheelock CE, Arner P, Mejhert N, Gao H, Rydén M. Adipocyte Expression of SLC19A1 Links DNA Hypermethylation to Adipose Tissue Inflammation and Insulin Resistance. J Clin Endocrinol Metab 2018; 103:710-721. [PMID: 29121255 DOI: 10.1210/jc.2017-01382] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Accepted: 11/02/2017] [Indexed: 02/07/2023]
Abstract
CONTEXT Insulin resistance (IR) is promoted by a chronic low-grade inflammation in white adipose tissue (WAT). The latter might be regulated through epigenetic mechanisms such as DNA methylation. The one carbon cycle (1CC) is a central metabolic process governing DNA methylation. OBJECTIVE To identify adipocyte-expressed 1CC genes linked to WAT inflammation, IR, and their causal role. DESIGN Cohort study. SETTING Outpatient academic clinic. PARTICIPANTS Obese and nonobese subjects. METHODS Gene expression and DNA methylation arrays were performed in subcutaneous WAT and isolated adipocytes. In in vitro differentiated human adipocytes, gene knockdown was achieved by small interfering RNA, and analyses included microarray, quantitative polymerase chain reaction, DNA methylation by enzyme-linked immunosorbent assay and pyrosequencing, protein secretion by enzyme-linked immunosorbent assay, targeted metabolomics, and luciferase reporter and thermal shift assays. MAIN OUTCOME MEASURES Effects on adipocyte inflammation. RESULTS In adipocytes from obese individuals, global DNA hypermethylation was associated positively with gene expression of proinflammatory pathways. Among the 1CC genes, IR in vivo and proinflammatory gene expression in WAT were most strongly and inversely associated with SLC19A1, a gene encoding a membrane folate carrier. SLC19A1 knockdown in human adipocytes perturbed intracellular 1CC metabolism, induced global DNA hypermethylation, and increased expression of proinflammatory genes. Several CpG loci linked SLC19A1 to inflammation; validation studies were focused on the chemokine C-C motif chemokine ligand 2 (CCL2) in which methylation in the promoter (cg12698626) regulated CCL2 expression and CCL2 secretion through altered transcriptional activity. CONCLUSIONS Reduced SLC19A1 expression in human adipocytes induces DNA hypermethylation, resulting in increased expression of specific proinflammatory genes, including CCL2. This constitutes an epigenetic mechanism that might link dysfunctional adipocytes to WAT inflammation and IR.
Collapse
Affiliation(s)
- Paul Petrus
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lucia Bialesova
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Antonio Checa
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Alastair Kerr
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Shama Naz
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Jesper Bäckdahl
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ana Gracia
- Department of Nutrition and Food Science, University of Basque Country (UPV/EHU), Vitoria, Spain
- CIBER Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain
| | - Sofia Toft
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Karin Dahlman-Wright
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Per Hedén
- Department of Plastic Surgery, Akademikliniken, Stockholm, Sweden
| | - Ingrid Dahlman
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Craig E Wheelock
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Peter Arner
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Niklas Mejhert
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Hui Gao
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Mikael Rydén
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
27
|
Liu J, Yang X, Yu S, Zheng R. The Leptin Signaling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1090:123-144. [PMID: 30390288 DOI: 10.1007/978-981-13-1286-1_7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Leptin plays a critical role in the regulation of energy balance and metabolic homeostasis. Impairment of leptin signaling is closely involved in the pathogenesis of obesity and metabolic diseases, including diabetes, cardiovascular disease, etc. Leptin initiates its intracellular signaling in the leptin-receptor-expressing neurons in the central nervous system to exert physiological function, thereby leading to a suppression of appetite, a reduction of food intake, a promotion of mitochondrial oxidation, an enhancement of thermogenesis, and a decrease in body weight. In this review, the studies on leptin neural and cellular pathways are summarized with an emphasis on the progress made during the last 10 years, for better understanding the molecular mechanism of obesity and other metabolic diseases.
Collapse
Affiliation(s)
- Jiarui Liu
- Department of Anatomy, Histology and Embryology, Health Science Center, Peking University, Beijing, China.,Neuroscience Research Institute, Peking University, Beijing, China.,Key Laboratory for Neuroscience of Ministry of Education, Peking University, Beijing, China.,Key Laboratory for Neuroscience of National Health Commission, Peking University, Beijing, China
| | - Xiaoning Yang
- Department of Anatomy, Histology and Embryology, Health Science Center, Peking University, Beijing, China
| | - Siwang Yu
- Department of Molecular and Cellular Pharmacology, Peking University School of Pharmaceutical Sciences, Beijing, China
| | - Ruimao Zheng
- Department of Anatomy, Histology and Embryology, Health Science Center, Peking University, Beijing, China. .,Neuroscience Research Institute, Peking University, Beijing, China. .,Key Laboratory for Neuroscience of Ministry of Education, Peking University, Beijing, China. .,Key Laboratory for Neuroscience of National Health Commission, Peking University, Beijing, China.
| |
Collapse
|
28
|
Blasius E, Gülden E, Kolb H, Habich C, Burkart V. The Autoantigenic Proinsulin B-Chain Peptide B11-23 Synergises with the 70 kDa Heat Shock Protein DnaK in Macrophage Stimulation. J Diabetes Res 2018; 2018:4834673. [PMID: 30622969 PMCID: PMC6304834 DOI: 10.1155/2018/4834673] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Accepted: 10/10/2018] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Heat shock proteins (Hsp) act as intracellular chaperones and in addition are used as adjuvant in vaccines of peptides complexed with recombinant Hsp. By interacting with autologous peptides, Hsp may promote the induction of autoimmune reactivity. OBJECTIVE Here, we analysed whether the effect of Hsp on macrophages is modulated by insulin peptides known to interact with Hsp. RESULTS Combinations of the 70 kDa Hsp DnaK with peptide B11-23 from the core region of the proinsulin B-chain induced the release of the inflammatory mediators interleukin-6, tumor necrosis factor α, and interleukin-1β from cells of human and murine macrophage lines. In parallel, there was high-affinity binding of B11-23 to DnaK. DnaK mixed with peptides from other regions of the insulin molecule did not stimulate cytokine secretion. DnaK alone induced little cytokine production, and peptides alone induced none. CONCLUSION The macrophage-stimulating potential of Hsp70 family proteins when combined with the proinsulin B-chain peptide B11-23 may contribute to the immunodominance of this peptide in the development of beta cell-directed autoimmunity in type 1 diabetes.
Collapse
Affiliation(s)
- Elias Blasius
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, D-40225 Düsseldorf, Germany
| | - Elke Gülden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, D-40225 Düsseldorf, Germany
| | - Hubert Kolb
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, D-40225 Düsseldorf, Germany
- West German Center of Diabetes and Health, Düsseldorf Catholic Hospital Group, D-40591 Düsseldorf, Germany
| | - Christiane Habich
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, D-40225 Düsseldorf, Germany
| | - Volker Burkart
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, D-40225 Düsseldorf, Germany
- German Center for Diabetes Research (DZD), D-85764 München-Neuherberg, Germany
| |
Collapse
|
29
|
Sell H, Poitou C, Habich C, Bouillot JL, Eckel J, Clément K. Heat Shock Protein 60 in Obesity: Effect of Bariatric Surgery and its Relation to Inflammation and Cardiovascular Risk. Obesity (Silver Spring) 2017; 25:2108-2114. [PMID: 29024428 DOI: 10.1002/oby.22014] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 08/18/2017] [Accepted: 08/22/2017] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Heat shock protein 60 (Hsp60) is an adipokine, and its serum concentrations are higher in patients with obesity compared to lean patients. This study aimed to analyze the effect of bariatric surgery on circulating concentrations of Hsp60 in morbid obesity and their correlation with inflammation and metabolic and cardiovascular risk. METHODS Fifty-three females with morbid obesity undergoing bariatric surgery were enrolled. Serum parameters and anthropometric measures were obtained at baseline and 3 to 12 months post surgery. RESULTS During the 12-month observation period, Hsp60 decreased significantly from 31.6 ± 4.7 ng/mL at baseline to 22.3 ± 3.0 ng/mL (3 months), 26.5 ± 5.5 (6 months), and 21.1 ± 3.3 ng/mL (12 months). Preoperatively, Hsp60 concentrations correlated positively with total cholesterol, low-density lipoprotein cholesterol, and apolipoprotein B (ApoB) and negatively with adiponectin. At the end of the observation period, serum Hsp60 positively correlated with triglycerides, ApoB, HbA1c , and C-reactive protein (CRP). Patients in the highest quartile of serum Hsp60 were characterized by significantly elevated CRP and interleukin 6 independently of BMI, glycemia, and insulinemia. At baseline and 12 months after surgery, Hsp60 positively correlated with the ApoB/ApoA1 ratio and the cholesterol/high-density lipoprotein cholesterol ratio. CONCLUSIONS Hsp60 concentrations are elevated in morbid obesity and decreased after surgery-induced weight loss. Their correlation with inflammatory markers and cardiovascular risk might link obesity and cardiovascular disease.
Collapse
Affiliation(s)
- Henrike Sell
- Paul-Langerhaus-Group Integrative Physiology, German Diabetes Center, Düsseldorf, Germany
| | - Christine Poitou
- INSERM, U1166, team 6 Nutriomique, Université Pierre et Marie Curie-Paris 6, Paris, France
- Institute of Cardiometabolism and Nutrition (ICAN), Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Nutrition Department, Paris, France
| | - Christiane Habich
- Institute for Clinical Diabetology, German Diabetes Center, Düsseldorf, Germany
| | - Jean-Luc Bouillot
- Assistance Publique-Hôpitaux de Paris, Hôtel-Dieu Hospital, Surgery Department, Paris, France
| | - Jürgen Eckel
- Paul-Langerhaus-Group Integrative Physiology, German Diabetes Center, Düsseldorf, Germany
| | - Karine Clément
- INSERM, U1166, team 6 Nutriomique, Université Pierre et Marie Curie-Paris 6, Paris, France
- Institute of Cardiometabolism and Nutrition (ICAN), Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Nutrition Department, Paris, France
| |
Collapse
|
30
|
Madhu D, Hammad M, Kavalakatt S, Khadir A, Tiss A. GLP-1 Analogue, Exendin-4, Modulates MAPKs Activity but not the Heat Shock Response in Human HepG2 Cells. Proteomics Clin Appl 2017; 12. [PMID: 29105359 DOI: 10.1002/prca.201600169] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 09/17/2017] [Indexed: 02/04/2023]
Abstract
PURPOSE Glucagon-like peptide-1 (GLP-1) analogues reduce ER stress and inflammation in key metabolic organs, including the liver. However, their effects on heat shock response (HSR) and mitogen-activated protein kinases (MAPKs) have not yet been elucidated. In the present study, we investigate whether the GLP-1 analogue, exendin-4, triggers the expression of HSR and increases MAPK activity under metabolic stress. EXPERIMENTAL DESIGN The effects of exendin-4 in the presence or absence of palmitic acid (PA; 400 μm) or glucose (30 mm) in the HepG2 liver cell line are assessed using Western blots, quantitative real-time PCR, and label-free proteomics. RESULTS Heat shock proteins (HSP60, HSP72, HSP90, and GRP78) and other chaperones are not significantly affected by exendin-4 under the conditions tested. In contrast, the presence of exendin-4 alone increases the MAPK phosphorylation levels (JNK, ERK1/2, and p38). For short incubation periods, in the presence of PA or glucose, treatment with exendin-4 exhibits limited effects but significantly attenuates MAPK phosphorylation after a 24-h incubation. Interestingly, canonical signaling pathways, such as EIF2, ILK, PKA, and Rho, are modulated by exendin-4. CONCLUSION AND CLINICAL RELEVANCE Identifying new pathways modulated by GLP-1 analogues will provide further insights into their benefits beyond their currently recognized roles in glycemic control, such as MAPK activity, energy homeostasis, and body weight decrease.
Collapse
Affiliation(s)
- Dhanya Madhu
- Research Division, Functional Proteomics and Metabolomics Unit, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Maha Hammad
- Research Division, Functional Proteomics and Metabolomics Unit, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Sina Kavalakatt
- Research Division, Functional Proteomics and Metabolomics Unit, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Abdelkrim Khadir
- Research Division, Functional Proteomics and Metabolomics Unit, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Ali Tiss
- Research Division, Functional Proteomics and Metabolomics Unit, Dasman Diabetes Institute, Kuwait City, Kuwait
| |
Collapse
|
31
|
Zhu RM, Lin W, Zhang W, Ren JT, Su Y, He JR, Lin Y, Su FX, Xie XM, Tang LY, Ren ZF. Modification effects of genetic polymorphisms in FTO, IL-6, and HSPD1 on the associations of diabetes with breast cancer risk and survival. PLoS One 2017; 12:e0178850. [PMID: 28591216 PMCID: PMC5462388 DOI: 10.1371/journal.pone.0178850] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 05/21/2017] [Indexed: 12/29/2022] Open
Abstract
The contribution of diabetes to breast cancer remains uncertain among Chinese females, which may result from different genetic factors. We evaluated the associations of diabetes, combined with the polymorphisms in the genes of fat mass and obesity-associated gene (FTO), interleukin 6 (IL-6), and heat shock protein 60 (HSPD1), with breast cancer risk and survival in a Chinese Han population. The information on the history of diabetes was collected from 1551 incident breast cancer cases and 1605 age-frequency matched controls in Guangzhou, China. In total, 1168 cases were followed up. Diabetes was associated with both an increased risk of breast cancer [OR (95%CI): 1.67 (1.11, 2.52)] and a poor overall survival and progression free survival for breast cancer patients [HRs (95%CIs): 2.66 (1.10, 6.44) and 2.46 (1.29, 4.70), respectively]. IL-6 rs1800796 and HSPD1 rs2605039 had interactions with diabetes on breast cancer risk. Among women with CC genotype of IL-6 rs1800796 or GG genotype of HSPD1 rs2605039, diabetic individuals had a remarkably increased risk of breast cancer compared to non-diabetic women with ORs and 95%CIs of 2.53 (1.45, 4.41) and 6.40 (2.29, 17.87), respectively. GT/TT genotypes of HSPD1 rs2605039 was also associated with a better progression free survival for breast cancer patients [HR (95%CI): 0.70 (0.49, 0.99)]. Our results suggest that the contribution of diabetes to breast cancer risk might be modified by IL-6 rs1800796 and HSPD1 rs2605039. Diabetes and HSPD1 rs2605039 might also influence breast cancer prognosis.
Collapse
Affiliation(s)
- Rui-Mei Zhu
- Department of Statistics and Epidemiology, Guangzhou Key Laboratory of Environmental Pollution and Health Risk Assessment, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Wei Lin
- Department of Statistics and Epidemiology, Guangzhou Key Laboratory of Environmental Pollution and Health Risk Assessment, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Wei Zhang
- Department of Statistics and Epidemiology, Guangzhou Key Laboratory of Environmental Pollution and Health Risk Assessment, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Jun-Ting Ren
- Department of Statistics and Epidemiology, Guangzhou Key Laboratory of Environmental Pollution and Health Risk Assessment, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Yi Su
- Department of Statistics and Epidemiology, Guangzhou Key Laboratory of Environmental Pollution and Health Risk Assessment, School of Public Health, Sun Yat-Sen University, Guangzhou, China
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Jian-Rong He
- The Guangzhou Women and Children’s Medical Center, Guangzhou, China
| | - Ying Lin
- The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Feng-Xi Su
- The Second Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiao-Ming Xie
- The Cancer Center, Sun Yat-Sen University, Guangzhou, China
| | - Lu-Ying Tang
- The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- * E-mail: (LT); (ZR)
| | - Ze-Fang Ren
- Department of Statistics and Epidemiology, Guangzhou Key Laboratory of Environmental Pollution and Health Risk Assessment, School of Public Health, Sun Yat-Sen University, Guangzhou, China
- * E-mail: (LT); (ZR)
| |
Collapse
|
32
|
Şelli ME, Wick G, Wraith DC, Newby AC. Autoimmunity to HSP60 during diet induced obesity in mice. Int J Obes (Lond) 2016; 41:348-351. [PMID: 27899808 PMCID: PMC5300117 DOI: 10.1038/ijo.2016.216] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 10/18/2016] [Accepted: 11/13/2016] [Indexed: 02/06/2023]
Abstract
Adaptive immunity has been implicated in adipose tissue inflammation, obesity and its adverse metabolic consequences. No obesity-related autoantigen has yet been identified, although heat shock protein 60 (HSP60) has been implicated in other autoimmune diseases. We investigated whether feeding a high-fat diet to C57BL/6J mice would cause autoimmunity to HSP60 and whether immunomodulation with peptides from HSP60 would reverse the resulting obesity or metabolic dysfunction. Obese mice had higher circulating levels of HSP60 associated with increased T-lymphocyte proliferation responses and the emergence of circulating IgG1 and IgG2c antibody levels against HSP60. Treatment with escalating doses of a mixture of three proven immunomodulatory HSP60 peptides did not reduce weight but completely reversed the increase in VLDL/LDL levels and partially reversed the glucose intolerance in obese mice. Obese mice mount an autoimmune response to HSP60, which partly underlies the resulting metabolic disturbances.
Collapse
Affiliation(s)
- M E Şelli
- School of Clinical Sciences and Bristol Heart Institute, Bristol Royal Infirmary, University of Bristol, Bristol, UK
| | - G Wick
- Laboratory of Autoimmunity, Division for Experimental Pathology and Immunology, Biocenter Innsbruck Medical University, Innsbruck, Austria
| | - D C Wraith
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - A C Newby
- School of Clinical Sciences and Bristol Heart Institute, Bristol Royal Infirmary, University of Bristol, Bristol, UK
| |
Collapse
|
33
|
Kriebel J, Herder C, Rathmann W, Wahl S, Kunze S, Molnos S, Volkova N, Schramm K, Carstensen-Kirberg M, Waldenberger M, Gieger C, Peters A, Illig T, Prokisch H, Roden M, Grallert H. Association between DNA Methylation in Whole Blood and Measures of Glucose Metabolism: KORA F4 Study. PLoS One 2016; 11:e0152314. [PMID: 27019061 PMCID: PMC4809492 DOI: 10.1371/journal.pone.0152314] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 03/11/2016] [Indexed: 12/22/2022] Open
Abstract
Epigenetic regulation has been postulated to affect glucose metabolism, insulin sensitivity and the risk of type 2 diabetes. Therefore, we performed an epigenome-wide association study for measures of glucose metabolism in whole blood samples of the population-based Cooperative Health Research in the Region of Augsburg F4 study using the Illumina HumanMethylation 450 BeadChip. We identified a total of 31 CpG sites where methylation level was associated with measures of glucose metabolism after adjustment for age, sex, smoking, and estimated white blood cell proportions and correction for multiple testing using the Benjamini-Hochberg (B-H) method (four for fasting glucose, seven for fasting insulin, 25 for homeostasis model assessment-insulin resistance [HOMA-IR]; B-H-adjusted p-values between 9.2x10(-5) and 0.047). In addition, DNA methylation at cg06500161 (annotated to ABCG1) was associated with all the aforementioned phenotypes and 2-hour glucose (B-H-adjusted p-values between 9.2x10(-5) and 3.0x10(-3)). Methylation status of additional three CpG sites showed an association with fasting insulin only after additional adjustment for body mass index (BMI) (B-H-adjusted p-values = 0.047). Overall, effect strengths were reduced by around 30% after additional adjustment for BMI, suggesting that this variable has an influence on the investigated phenotypes. Furthermore, we found significant associations between methylation status of 21 of the aforementioned CpG sites and 2-hour insulin in a subset of samples with seven significant associations persisting after additional adjustment for BMI. In a subset of 533 participants, methylation of the CpG site cg06500161 (ABCG1) was inversely associated with ABCG1 gene expression (B-H-adjusted p-value = 1.5x10(-9)). Additionally, we observed an enrichment of the top 1,000 CpG sites for diabetes-related canonical pathways using Ingenuity Pathway Analysis. In conclusion, our study indicates that DNA methylation and diabetes-related traits are associated and that these associations are partially BMI-dependent. Furthermore, the interaction of ABCG1 with glucose metabolism is modulated by epigenetic processes.
Collapse
Affiliation(s)
- Jennifer Kriebel
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Muenchen-Neuherberg, Germany
| | - Christian Herder
- German Center for Diabetes Research (DZD), Muenchen-Neuherberg, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Wolfgang Rathmann
- Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Simone Wahl
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Muenchen-Neuherberg, Germany
| | - Sonja Kunze
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Neuherberg, Germany
| | - Sophie Molnos
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Muenchen-Neuherberg, Germany
| | - Nadezda Volkova
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Neuherberg, Germany
| | - Katharina Schramm
- Institute of Human Genetics, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Human Genetics, Technische Universitaet Muenchen, Munich, Germany
| | - Maren Carstensen-Kirberg
- German Center for Diabetes Research (DZD), Muenchen-Neuherberg, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Neuherberg, Germany
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Neuherberg, Germany
| | - Annette Peters
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Muenchen-Neuherberg, Germany
| | - Thomas Illig
- Hannover Unified Biobank, Hannover Medical School, Hanover, Germany
- Institute of Human Genetics, Hannover Medical School, Hanover, Germany
| | - Holger Prokisch
- Institute of Human Genetics, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Human Genetics, Technische Universitaet Muenchen, Munich, Germany
| | - Michael Roden
- German Center for Diabetes Research (DZD), Muenchen-Neuherberg, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Duesseldorf, Duesseldorf, Germany
- Department of Endocrinology and Diabetology, University Hospital Duesseldorf, Duesseldorf, Germany
| | - Harald Grallert
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Muenchen-Neuherberg, Germany
| |
Collapse
|
34
|
Abstract
Initial successful weight loss is often followed by weight regain after the dietary intervention. Compared with lean people, cellular stress in adipose tissue is increased in obese subjects. However, the relation between cellular stress and the risk for weight regain after weight loss is unclear. Therefore, we determined the expression levels of stress proteins during weight loss and weight maintenance in relation to weight regain. In vivo findings were compared with results from in vitro cultured human Simpson-Golabi-Behmel syndrome (SGBS) adipocytes. In total, eighteen healthy subjects underwent an 8-week diet programme with a 10-month follow-up. Participants were categorised as weight maintainers or weight regainers (WR) depending on their weight changes during the intervention. Abdominal subcutaneous adipose tissue biopsies were obtained before and after the diet and after the follow-up. In vitro differentiated SGBS adipocytes were starved for 96 h with low (0·55 mm) glucose. Levels of stress proteins were determined by Western blotting. WR showed increased expressions of β-actin, calnexin, heat shock protein (HSP) 27, HSP60 and HSP70. Changes of β-actin, HSP27 and HSP70 are linked to HSP60, a proposed key factor in weight regain after weight loss. SGBS adipocytes showed increased levels of β-actin and HSP60 after 96 h of glucose restriction. The increased level of cellular stress proteins in the adipose tissue of WR probably resides in the adipocytes as shown by in vitro experiments. Cellular stress accumulated in adipose tissue during weight loss may be a risk factor for weight regain.
Collapse
|
35
|
Habich C, Sell H. Heat shock proteins in obesity: links to cardiovascular disease. Horm Mol Biol Clin Investig 2015; 21:117-24. [PMID: 25781556 DOI: 10.1515/hmbci-2014-0040] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 01/28/2015] [Indexed: 01/16/2023]
Abstract
Adipose tissue expansion is associated with adipocyte dysfunction and increased inflammatory processes. In the obese state, adipose tissue is characterized by an impaired intracellular stress defense system and dysbalanced heat shock response. Several members of the heat shock protein (HSP) family have been identified as novel adipokines released upon cellular stress, which might be a molecular link from adipose tissue inflammation to the cardiovascular system. Therefore, this review aims at summarizing and discussing our recent knowledge on HSPs in relation to obesity and their potential links to cardiovascular disease. Of particular importance/interest are two members of the HSP family, HSP60 and heme oxygenase 1 (HO-1), which have been well described as adipokines, and studied in the context of obesity and cardiovascular disease. HSP60 is regarded as a novel molecular link between adipose tissue inflammation and obesity-associated insulin resistance. The role of HO-1 induction in the obese state is well-documented, but a causal relationship between increased HO-1 levels and obesity-associated metabolic diseases is still controversial. Both HSP60 and HO-1 are also forthcoming targets for the treatment of cardiovascular disease, and the current knowledge will also be discussed in this review.
Collapse
|
36
|
DNAJB3/HSP-40 cochaperone improves insulin signaling and enhances glucose uptake in vitro through JNK repression. Sci Rep 2015; 5:14448. [PMID: 26400768 PMCID: PMC4585859 DOI: 10.1038/srep14448] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 07/06/2015] [Indexed: 12/20/2022] Open
Abstract
Heat shock response (HSR) is an essential host-defense mechanism that is dysregulated in obesity-induced insulin resistance and type 2 diabetes (T2D). Our recent data demonstrated that DNAJB3 was downregulated in obese human subjects and showed negative correlation with inflammatory markers. Nevertheless, DNAJB3 expression pattern in diabetic subjects and its mode of action are not yet known. In this study, we showed reduction in DNAJB3 transcript and protein levels in PBMC and subcutaneous adipose tissue of obese T2D compared to obese non-diabetic subjects. Overexpression of DNAJB3 in HEK293 and 3T3-L1 cells reduced JNK, IRS-1 Ser-307 phosphorylation and enhanced Tyr-612 phosphorylation suggesting an improvement in IRS-1 signaling. Furthermore, DNAJB3 mediated the PI3K/AKT pathway activation through increasing AKT and AS160 phosphorylation. AS160 mediates the mobilization of GLUT4 transporter to the cell membrane and thereby improves glucose uptake. Using pre-adipocytes cells we showed that DNAJB3 overexpression caused a significant increase in the glucose uptake, possibly through its phosphorylation of AS160. In summary, our results shed the light on the possible role of DNAJB3 in improving insulin sensitivity and glucose uptake through JNK repression and suggest that DNAJB3 could be a potential target for therapeutic treatment of obesity-induced insulin resistance.
Collapse
|
37
|
Capuani B, Della-Morte D, Donadel G, Caratelli S, Bova L, Pastore D, De Canio M, D'Aguanno S, Coppola A, Pacifici F, Arriga R, Bellia A, Ferrelli F, Tesauro M, Federici M, Neri A, Bernardini S, Sbraccia P, Di Daniele N, Sconocchia G, Orlandi A, Urbani A, Lauro D. Liver protein profiles in insulin receptor-knockout mice reveal novel molecules involved in the diabetes pathophysiology. Am J Physiol Endocrinol Metab 2015; 308:E744-E755. [PMID: 25714671 DOI: 10.1152/ajpendo.00447.2014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 02/19/2015] [Indexed: 02/08/2023]
Abstract
Liver has a principal role in glucose regulation and lipids homeostasis. It is under a complex control by substrates such as hormones, nutrients, and neuronal impulses. Insulin promotes glycogen synthesis, lipogenesis, and lipoprotein synthesis and inhibits gluconeogenesis, glycogenolysis, and VLDL secretion by modifying the expression and enzymatic activity of specific molecules. To understand the pathophysiological mechanisms leading to metabolic liver disease, we analyzed liver protein patterns expressed in a mouse model of diabetes by proteomic approaches. We used insulin receptor-knockout (IR(-/-)) and heterozygous (IR(+/-)) mice as a murine model of liver metabolic dysfunction associated with diabetic ketoacidosis and insulin resistance. We evaluated liver fatty acid levels by microscopic examination and protein expression profiles by orthogonal experimental strategies using protein 2-DE MALDI-TOF/TOF and peptic nLC-MS/MS shotgun profiling. Identified proteins were then loaded into Ingenuity Pathways Analysis to find possible molecular networks. Twenty-eight proteins identified by 2-DE analysis and 24 identified by nLC-MS/MS shotgun were differentially expressed among the three genotypes. Bioinformatic analysis revealed a central role of high-mobility group box 1/2 and huntigtin never reported before in association with metabolic and related liver disease. A different modulation of these proteins in both blood and hepatic tissue further suggests their role in these processes. These results provide new insight into pathophysiology of insulin resistance and hepatic steatosis and could be useful in identifying novel biomarkers to predict risk for diabetes and its complications.
Collapse
Affiliation(s)
- Barbara Capuani
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - David Della-Morte
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; IRCCS San Raffaele Pisana, Rome, Italy; and
| | - Giulia Donadel
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Sara Caratelli
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Luca Bova
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Donatella Pastore
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Michele De Canio
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Rome, Italy; Laboratory of Proteomics and Metabonomics, S. Lucia Foundation-Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Simona D'Aguanno
- Laboratory of Proteomics and Metabonomics, S. Lucia Foundation-Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Andrea Coppola
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Francesca Pacifici
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Roberto Arriga
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Alfonso Bellia
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; Policlinico Tor Vergata Foundation, Rome, Italy
| | - Francesca Ferrelli
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Manfredi Tesauro
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; Policlinico Tor Vergata Foundation, Rome, Italy
| | - Massimo Federici
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; Policlinico Tor Vergata Foundation, Rome, Italy
| | - Anna Neri
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy; Policlinico Tor Vergata Foundation, Rome, Italy
| | - Sergio Bernardini
- Policlinico Tor Vergata Foundation, Rome, Italy; Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Rome, Italy
| | - Paolo Sbraccia
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; Policlinico Tor Vergata Foundation, Rome, Italy
| | - Nicola Di Daniele
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; Policlinico Tor Vergata Foundation, Rome, Italy
| | - Giuseppe Sconocchia
- Institute of Traslational Pharmacology, National Research Council, Rome, Italy
| | - Augusto Orlandi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Andrea Urbani
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Rome, Italy; Laboratory of Proteomics and Metabonomics, S. Lucia Foundation-Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Davide Lauro
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; Policlinico Tor Vergata Foundation, Rome, Italy;
| |
Collapse
|
38
|
Kim JH, Hong YC. HSP70-hom gene polymorphisms modify the association of diethylhexyl phthalates with insulin resistance. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2014; 55:727-34. [PMID: 25044062 DOI: 10.1002/em.21884] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 05/23/2014] [Indexed: 05/27/2023]
Abstract
Recent studies suggest that diethylhexyl phthalates (DEHP) could contribute to the development of insulin resistance (IR) through oxidative stress, and that heat shock protein (HSP) could be related with the association between DEHP and IR. Therefore, we evaluated the effect modification of genetic polymorphisms of HSP70-hom, an oxidative stress related gene, on the relation between exposure to DEHP and IR. We obtained repeated blood and urine samples from 414 elderly female participants and measured urinary levels of mono-(2-ethyl-5-hydroxyhexyl) phthalate (MEHHP) and mono-(2-ethyl-5-oxohexyl) phthalate (MEOHP) as metabolites of DEHP. We also measured serum levels of fasting glucose and insulin, derived the homeostatic model assessment (HOMA) index to assess IR, and genotyped two HSP70-hom polymorphisms (rs2227956 and rs2075800). A mixed effect model and penalized regression spline were used to estimate the associations between DEHP exposure and IR by genetic polymorphisms. The molar sum of MEHHP and MEOHP (∑DEHP) were significantly associated with HOMA (β = 0.30, P = 0.022). When stratified by genotype at rs2227956, the relationship between ∑DEHP and HOMA was statistically significant in participants with TT (β = 0.32, P = 0.048) or TC (β = 0.60, P = 0.008), while at rs2075800 there was a marginal association for the GA genotype (β = 0.33, P = 0.097). When haplotypes were constituted across the two HSP70-hom polymorphisms (rs2227956 and rs2075800), the association was apparent only in participants with the T-A haplotype (β = 0.39, P = 0.029). Our study suggests that HSP70-hom polymorphisms modify the association of DEHP with IR.
Collapse
Affiliation(s)
- Jin Hee Kim
- Department of Environmental Health, Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
| | | |
Collapse
|
39
|
Tiss A, Khadir A, Abubaker J, Abu-Farha M, Al-Khairi I, Cherian P, John J, Kavalakatt S, Warsame S, Al-Ghimlas F, Elkum N, Behbehani K, Dermime S, Dehbi M. Immunohistochemical profiling of the heat shock response in obese non-diabetic subjects revealed impaired expression of heat shock proteins in the adipose tissue. Lipids Health Dis 2014; 13:106. [PMID: 24986468 PMCID: PMC4085713 DOI: 10.1186/1476-511x-13-106] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2014] [Accepted: 06/17/2014] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Obesity is characterized by a chronic low-grade inflammation and altered stress responses in key metabolic tissues. Impairment of heat shock response (HSR) has been already linked to diabetes and insulin resistance as reflected by decrease in heat shock proteins (HSPs) expression. However, the status of HSR in non-diabetic human obese has not yet been elucidated. The aim of the current study was to investigate whether obesity triggers a change in the HSR pattern and the impact of physical exercise on this pattern at protein and mRNA levels. METHODS Two groups of adult non-diabetic human subjects consisting of lean and obese (n = 47 for each group) were enrolled in this study. The expression pattern of HSP-27, DNAJB3/HSP-40, HSP-60, HSC-70, HSP72, HSP-90 and GRP-94 in the adipose tissue was primarily investigated by immunohistochemistry and then complemented by western blot and qRT-PCR in Peripheral blood mononuclear cells (PBMCs). HSPs expression levels were correlated with various physical, clinical and biochemical parameters. We have also explored the effect of a 3-month moderate physical exercise on the HSPs expression pattern in obese subjects. RESULTS Obese subjects displayed increased expression of HSP-60, HSC-70, HSP-72, HSP-90 and GRP-94 and lower expression of DNAJB3/HSP-40 (P < 0.05). No differential expression was observed for HSP-27 between the two groups. Higher levels of HSP-72 and GRP-94 proteins correlated positively with the indices of obesity (body mass index and percent body fat) and circulating levels of IFN-gamma-inducible protein 10 (IP-10) and RANTES chemokines. This expression pattern was concomitant with increased inflammatory response in the adipose tissue as monitored by increased levels of Interleukin-6 (IL-6), Tumor necrosis factor-α (TNF-α), and RANTES (P < 0.05). Physical exercise reduced the expression of various HSPs in obese to normal levels observed in lean subjects with a parallel decrease in the endogenous levels of IL-6, TNF-α, and RANTES. CONCLUSION Taken together, these data indicate that obesity triggers differential regulation of various components of the HSR in non-diabetic subjects and a 3-month physical moderate exercise was sufficient to restore the normal expression of HSPs in the adipose tissue with concomitant attenuation in the inflammatory response.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Mohammed Dehbi
- Diabetes Research Centre, Qatar Biomedical Research Institute, Box: 5825, Doha, Qatar.
| |
Collapse
|
40
|
Moreno-Navarrete JM, Fernández-Real JM. The possible role of antimicrobial proteins in obesity-associated immunologic alterations. Expert Rev Clin Immunol 2014; 10:855-66. [DOI: 10.1586/1744666x.2014.911088] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
41
|
Prevalence of systemic inflammation and micronutrient imbalance in patients with complex abdominal hernias. J Gastrointest Surg 2014; 18:646-55. [PMID: 24356980 DOI: 10.1007/s11605-013-2431-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 12/02/2013] [Indexed: 01/31/2023]
Abstract
OBJECTIVE To determine the prevalence of low-grade inflammation, micronutrient imbalances and associated clinical profiles in patients being evaluated for complex abdominal hernia repair. METHODS Review of 127 consecutive adult patients for evaluation of complex ventral hernias from January 2012 to March 2013. Records were analyzed to determine the prevalence and correlations of clinical risk factors, attributes of hernias identified by computerized tomography, and laboratory indices of metabolism, inflammation and micronutrient imbalances. RESULTS Strong correlations (p < 0.001) were established for body mass index (BMI) with volume of hernia content and C-reactive protein (CRP) level. CRP levels correlated strongly with red cell distribution width and inversely with zinc (p < 0.01). Evidence of micronutrient imbalance (abnormal zinc or red cell distribution width [RDW]) was observed in 48%. CONCLUSIONS In this comorbidity-rich population with known variability in surgical outcomes, the prevalence of chronic inflammation and micronutrient deficiency are high enough to warrant systemic preoperative evaluation given their possible effect on wound healing and convalescence. Simple repletion is unlikely to improve outcomes without attention to the biological stresses that are associated with micronutrient imbalance.
Collapse
|
42
|
Murri M, Insenser M, Luque M, Tinahones FJ, Escobar-Morreale HF. Proteomic analysis of adipose tissue: informing diabetes research. Expert Rev Proteomics 2014; 11:491-502. [PMID: 24684164 DOI: 10.1586/14789450.2014.903158] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Diabetes, one of the most common endocrine diseases worldwide, results from complex pathophysiological mechanisms that are not fully understood. Adipose tissue is considered a major endocrine organ and plays a central role in the development of diabetes. The identification of the adipose tissue-derived factors that contribute to the onset and progression of diabetes will hopefully lead to the development of preventive and therapeutic interventions. Proteomic techniques may be useful tools for this purpose. In the present review, we have summarized the studies conducting adipose tissue proteomics in subjects with diabetes and insulin resistance, and discussed the proteins identified in these studies as candidates to exert important roles in these disorders.
Collapse
Affiliation(s)
- Mora Murri
- Department of Endocrinology and Nutrition, Diabetes, Obesity and Human Reproduction Research Group, Hospital Universitario Ramón y Cajal and Universidad de Alcalá and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Centro de Investigación Biomédica en Red Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), E-28034 Madrid, Spain
| | | | | | | | | |
Collapse
|
43
|
Märker T, Kriebel J, Wohlrab U, Burkart V, Habich C. Adipocytes from New Zealand obese mice exhibit aberrant proinflammatory reactivity to the stress signal heat shock protein 60. J Diabetes Res 2014; 2014:187153. [PMID: 24672802 PMCID: PMC3941600 DOI: 10.1155/2014/187153] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2013] [Revised: 11/29/2013] [Accepted: 12/03/2013] [Indexed: 11/29/2022] Open
Abstract
Adipocytes release immune mediators that contribute to diabetes-associated inflammatory processes. As the stress protein heat shock protein 60 (Hsp60) induces proinflammatory adipocyte activities, we hypothesized that adipocytes of diabetes-predisposed mice exhibit an increased proinflammatory reactivity to Hsp60. Preadipocytes and mature adipocytes from nonobese diabetic (NOD), New Zealand obese (NZO), and C57BL/6J mice were analyzed for Hsp60 binding, Hsp60-activated signaling pathways, and Hsp60-induced release of the chemokine CXCL-1 (KC), interleukin 6 (IL-6), and macrophage chemoattractant protein-1 (MCP-1). Hsp60 showed specific binding to (pre-)adipocytes of NOD, NZO, and C57BL/6J mice. Hsp60 binding involved conserved binding structure(s) and Hsp60 epitopes and was strongest to NZO mouse-derived mature adipocytes. Hsp60 exposure induced KC, IL-6, and MCP-1 release from (pre-)adipocytes of all mouse strains with a pronounced increase of IL-6 release from NZO mouse-derived adipocytes. Compared to NOD and C57BL/6J mouse derived cells, Hsp60-induced formation of IL-6, KC, and MCP-1 from NZO mouse-derived (pre-)adipocytes strongly depended on NF κ B-activation. Increased Hsp60 binding and Hsp60-induced IL-6 release by mature adipocytes of NZO mice suggest that enhanced adipocyte reactivity to the stress signal Hsp60 contributes to inflammatory processes underlying diabetes associated with obesity and insulin resistance.
Collapse
Affiliation(s)
- Tina Märker
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, the Heinrich-Heine-University Düsseldorf, Auf'm Hennekamp 65, 40225 Düsseldorf, Germany
| | - Jennifer Kriebel
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, the Heinrich-Heine-University Düsseldorf, Auf'm Hennekamp 65, 40225 Düsseldorf, Germany
| | - Ulrike Wohlrab
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, the Heinrich-Heine-University Düsseldorf, Auf'm Hennekamp 65, 40225 Düsseldorf, Germany
| | - Volker Burkart
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, the Heinrich-Heine-University Düsseldorf, Auf'm Hennekamp 65, 40225 Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Düsseldorf, Germany
- *Volker Burkart:
| | - Christiane Habich
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, the Heinrich-Heine-University Düsseldorf, Auf'm Hennekamp 65, 40225 Düsseldorf, Germany
| |
Collapse
|
44
|
Lambrecht S, Juchtmans N, Elewaut D. Heat-shock proteins in stromal joint tissues: innocent bystanders or disease-initiating proteins? Rheumatology (Oxford) 2013; 53:223-32. [DOI: 10.1093/rheumatology/ket277] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
|
45
|
DNAJB3/HSP-40 cochaperone is downregulated in obese humans and is restored by physical exercise. PLoS One 2013; 8:e69217. [PMID: 23894433 PMCID: PMC3722167 DOI: 10.1371/journal.pone.0069217] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 06/04/2013] [Indexed: 12/17/2022] Open
Abstract
Obesity is a major risk factor for a myriad of disorders such as insulin resistance and diabetes. The mechanisms underlying these chronic conditions are complex but low grade inflammation and alteration of the endogenous stress defense system are well established. Previous studies indicated that impairment of HSP-25 and HSP-72 was linked to obesity, insulin resistance and diabetes in humans and animals while their induction was associated with improved clinical outcomes. In an attempt to identify additional components of the heat shock response that may be dysregulated by obesity, we used the RT2-Profiler PCR heat shock array, complemented with RT-PCR and validated by Western blot and immunohistochemistry. Using adipose tissue biopsies and PBMC of non-diabetic lean and obese subjects, we report the downregulation of DNAJB3 cochaperone mRNA and protein in obese that negatively correlated with percent body fat (P = 0.0001), triglycerides (P = 0.035) and the inflammatory chemokines IP-10 and RANTES (P = 0.036 and P = 0.02, respectively). DNAJB positively correlated with maximum oxygen consumption (P = 0.031). Based on the beneficial effect of physical exercise, we investigated its possible impact on DNAJB3 expression and indeed, we found that exercise restored the expression of DNAJB3 in obese subjects with a concomitant decrease of phosphorylated JNK. Using cell lines, DNAJB3 protein was reduced following treatment with palmitate and tunicamycin which is suggestive of the link between the expression of DNAJB3 and the activation of the endoplasmic reticulum stress. DNAJB3 was also shown to coimmunoprecipiate with JNK and IKKβ stress kinases along with HSP-72 and thus, suggesting its potential role in modulating their activities. Taken together, these data suggest that DNAJB3 can potentially play a protective role against obesity.
Collapse
|
46
|
Jaffe S, Doulaveris G, Orfanelli T, Arantes M, Damasceno D, Calderon I, Rudge MVC, Witkin SS. Induction of the 72 kDa heat shock protein by glucose ingestion in black pregnant women. Cell Stress Chaperones 2013; 18:527-30. [PMID: 23325573 PMCID: PMC3682015 DOI: 10.1007/s12192-013-0401-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Revised: 12/23/2012] [Accepted: 01/02/2013] [Indexed: 10/27/2022] Open
Abstract
Obese Black women are at increased risk for development of gestational diabetes mellitus and have worse perinatal outcomes than do obese women of other ethnicities. Since hsp72 has been associated with the regulation of obesity-induced insulin resistance, we evaluated associations between glucose ingestion, hsp72 release and insulin production in Black pregnant women. Specifically, the effect of a 50-g glucose challenge test (GCT) on heat shock protein and insulin levels in the circulation 1 h later was evaluated. Hsp27 and hsp60 levels remained unchanged. In contrast, serum levels of hsp72 markedly increased after glucose ingestion (p = 0.0054). Further analysis revealed that this increase was limited to women who were not obese (body mass index <30). Insulin levels pre-GCT were positively correlated with body mass index (p = 0.0189). Median insulin concentrations also increased post GCT in non-obese women but remained almost unchanged in obese women. Post-GCT serum hsp72 concentrations were inversely correlated with post GCT insulin concentrations (p = 0.0111). These observations suggest that glucose intake during gestation in Black women rapidly leads to an elevation in circulating hsp72 only in non-obese Black women. The release of hsp72 may regulate the extent of insulin production in response to a glucose challenge and, thereby, protect the mother and/or fetus from development of hyperglycemia, hyperinsulinemia, and/or immune system alterations.
Collapse
Affiliation(s)
- Shirlee Jaffe
- />Division of Immunology and Infectious Diseases, Department of Obstetrics and Gynecology, Weill Cornell Medical College, New York, NY USA
| | - Georgios Doulaveris
- />Division of Immunology and Infectious Diseases, Department of Obstetrics and Gynecology, Weill Cornell Medical College, New York, NY USA
| | - Theofano Orfanelli
- />Division of Immunology and Infectious Diseases, Department of Obstetrics and Gynecology, Weill Cornell Medical College, New York, NY USA
| | - Mariana Arantes
- />Department of Obstetrics and Gynecology, Sao Paulo State University (UNESP), Botucatu, Brazil
| | - Débora Damasceno
- />Department of Obstetrics and Gynecology, Sao Paulo State University (UNESP), Botucatu, Brazil
| | - Iracema Calderon
- />Department of Obstetrics and Gynecology, Sao Paulo State University (UNESP), Botucatu, Brazil
| | - Marilza V. C. Rudge
- />Department of Obstetrics and Gynecology, Sao Paulo State University (UNESP), Botucatu, Brazil
| | - Steven S. Witkin
- />Division of Immunology and Infectious Diseases, Department of Obstetrics and Gynecology, Weill Cornell Medical College, New York, NY USA
- />Department of Obstetrics and Gynecology, Weill Cornell Medical College, 525 East 68th Street, Box 35, New York, NY 10065 USA
| |
Collapse
|
47
|
Henderson B, Fares MA, Lund PA. Chaperonin 60: a paradoxical, evolutionarily conserved protein family with multiple moonlighting functions. Biol Rev Camb Philos Soc 2013; 88:955-87. [DOI: 10.1111/brv.12037] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Revised: 02/20/2013] [Accepted: 03/04/2013] [Indexed: 02/07/2023]
Affiliation(s)
- Brian Henderson
- Department of Microbial Diseases, UCL-Eastman Dental Institute; University College London; London WC1X 8LD U.K
| | - Mario A. Fares
- Department of Genetics; University of Dublin, Trinity College Dublin; Dublin 2 Ireland
- Department of Abiotic Stress; Instituto de Biologia Molecular y Celular de Plantas, Consejo Superior de Investigaciones Cientificas (CSIC-UPV); Valencia 46022 Spain
| | - Peter A. Lund
- School of Biosciences; University of Birmingham; Birmingham B15 2TT U.K
| |
Collapse
|
48
|
Ippoliti F, Canitano N, Businaro R. Stress and obesity as risk factors in cardiovascular diseases: a neuroimmune perspective. J Neuroimmune Pharmacol 2013; 8:212-26. [PMID: 23329173 DOI: 10.1007/s11481-012-9432-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Accepted: 12/28/2012] [Indexed: 01/09/2023]
Abstract
Obesity is now growing at an alarming rate reaching epidemic proportions worldwide thus increasing morbidity and mortality rates for chronic disease. But although we have ample information on the complications associated with obesity, precisely what causes obesity remains poorly understood. Some evidence attributes a major role to a low-grade chronic inflammatory state (neurogenic inflammation) induced in obesity by inflammatory mediators produced and secreted within the expanded activated adipocyte pool. Adipose tissue is an endocrine organ that secretes numerous adipose tissue-specific or enriched hormones, known as adipokines, cytokine-like molecules thought to play a pathogenic role in cardiovascular diseases. The imbalance between increased inflammatory stimuli and decreased anti-inflammatory mechanisms may depend on chronic stress. Hence the positive correlation found between stress, obesity and cardiovascular diseases. The chronic inflammatory state associated with insulin resistance and endothelial dysfunction is highly deleterious for vascular function. This review focuses on the proposed neuroimmunodulatory mechanisms linking chronic (psychological) stress, obesity and cardiovascular diseases.
Collapse
Affiliation(s)
- Flora Ippoliti
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy.
| | | | | |
Collapse
|
49
|
Chalubinski M, Wojdan K, Dorantowicz R, Jackowska P, Gorzelak P, Broncel M. Comprehensive insight into immune regulatory mechanisms and vascular wall determinants of atherogenesis - emerging perspectives of immunomodulation. Arch Med Sci 2013; 9:159-65. [PMID: 23515919 PMCID: PMC3598149 DOI: 10.5114/aoms.2013.33355] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Revised: 07/12/2012] [Accepted: 10/02/2012] [Indexed: 12/14/2022] Open
Abstract
For many years atherosclerosis was believed to be the passive accumulation of cholesterol in vessel walls. Today the picture is more complex, as immune processes occur in atherogenesis. Considerable attention is focused on the particular role of adaptive immune responses orchestrated by T cell subsets. Since the role of Th1/Th2 balance and Th1 cell domination in atherogenesis is already known, the involvement of regulatory T lymphocytes and recently described Th17 cells raises new concerns. On one hand, each of these cells may specifically drive responses of vascular wall tissues and immune cells; however, they are subject to the control of a plethora of tissue- and pathogen-derived agents. Due to ineffective tissue regeneration, remodeling of the vascular wall occurs. The understanding of the immune regulatory network gives perspectives of innovative immunomodulatory therapies of atherosclerosis and the prevention of its complications, such as coronary artery disease.
Collapse
Affiliation(s)
- Maciej Chalubinski
- Laboratory of Tissue Pharmacology, Department of Internal Medicine and Clinical Pharmacology, Medical University of Lodz, Poland
| | | | | | | | | | | |
Collapse
|
50
|
White SJ, Taylor MJ, Hurt RT, Jensen MD, Poland GA. Leptin-based adjuvants: an innovative approach to improve vaccine response. Vaccine 2013; 31:1666-72. [PMID: 23370154 DOI: 10.1016/j.vaccine.2013.01.032] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Revised: 01/04/2013] [Accepted: 01/15/2013] [Indexed: 12/22/2022]
Abstract
Leptin is a pleiotropic hormone with multiple direct and regulatory immune functions. Leptin deficiency or resistance hinders the immunologic, metabolic, and neuroendocrinologic processes necessary to thwart infections and their associated complications, and to possibly protect against infectious diseases following vaccination. Circulating leptin levels are proportional to body fat mass. High circulating leptin concentrations, as observed in obesity, are indicative of the development of leptin transport saturation/signaling desensitization. Leptin bridges nutritional status and immunity. Although its role in vaccine response is currently unknown, over-nutrition has been shown to suppress vaccine-induced immune responses. For instance, obesity (BMI ≥30 kg/m(2)) is associated with lower antigen-specific antibody titers following influenza, hepatitis B, and tetanus vaccinations. This suggests that obesity, and possibly saturable leptin levels, are contributing factors to poor vaccine immunogenicity. While leptin-based therapies have not been investigated as vaccine adjuvants thus far, leptin's role in immunity suggests that application of these therapies is promising and worth investigation to enhance vaccine response in people with leptin signaling impairments. This review will examine the possibility of using leptin as a vaccine adjuvant by: briefly reviewing the distribution and signal transduction of leptin and its receptors; discussing the physiology of leptin with emphasis on its immune functions; reviewing the causes of attenuation of leptin signaling; and finally, providing plausible inferences for the innovative use of leptin-based pharmacotherapies as vaccine adjuvants.
Collapse
Affiliation(s)
- Sarah J White
- Mayo Clinic Vaccine Research Group, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, United States
| | | | | | | | | |
Collapse
|