1
|
Paik S, Kim JK, Shin HJ, Park EJ, Kim IS, Jo EK. Updated insights into the molecular networks for NLRP3 inflammasome activation. Cell Mol Immunol 2025; 22:563-596. [PMID: 40307577 DOI: 10.1038/s41423-025-01284-9] [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: 12/24/2024] [Accepted: 03/17/2025] [Indexed: 05/02/2025] Open
Abstract
Over the past decade, significant advances have been made in our understanding of how NACHT-, leucine-rich-repeat-, and pyrin domain-containing protein 3 (NLRP3) inflammasomes are activated. These findings provide detailed insights into the transcriptional and posttranslational regulatory processes, the structural-functional relationship of the activation processes, and the spatiotemporal dynamics of NLRP3 activation. Notably, the multifaceted mechanisms underlying the licensing of NLRP3 inflammasome activation constitute a focal point of intense research. Extensive research has revealed the interactions of NLRP3 and its inflammasome components with partner molecules in terms of positive and negative regulation. In this Review, we provide the current understanding of the complex molecular networks that play pivotal roles in regulating NLRP3 inflammasome priming, licensing and assembly. In addition, we highlight the intricate and interconnected mechanisms involved in the activation of the NLRP3 inflammasome and the associated regulatory pathways. Furthermore, we discuss recent advances in the development of therapeutic strategies targeting the NLRP3 inflammasome to identify potential therapeutics for NLRP3-associated inflammatory diseases. As research continues to uncover the intricacies of the molecular networks governing NLRP3 activation, novel approaches for therapeutic interventions against NLRP3-related pathologies are emerging.
Collapse
Affiliation(s)
- Seungwha Paik
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- System Network Inflammation Control Research Center, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Biomedical Research Institute, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Jin Kyung Kim
- Department of Microbiology, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Hyo Jung Shin
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Department of Biochemistry and Cell Biology, Eulji University School of Medicine, Daejeon, Republic of Korea
- Brain Research Institute, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Eun-Jin Park
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - In Soo Kim
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Biomedical Research Institute, Chungnam National University Hospital, Daejeon, Republic of Korea
- Department of Pharmacology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea.
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea.
- Biomedical Research Institute, Chungnam National University Hospital, Daejeon, Republic of Korea.
| |
Collapse
|
2
|
Datta S, Koka S, Boini KM. Understanding the Role of Adipokines in Cardiometabolic Dysfunction: A Review of Current Knowledge. Biomolecules 2025; 15:612. [PMID: 40427505 PMCID: PMC12109550 DOI: 10.3390/biom15050612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/13/2025] [Accepted: 04/19/2025] [Indexed: 05/29/2025] Open
Abstract
Cardiometabolic risk and associated dysfunctions contribute largely to the recent rise in mortality globally. Advancements in multi-omics in recent years promise a better understanding of potential biomarkers that enable an early diagnosis of cardiometabolic dysfunction. However, the molecular mechanisms driving the onset and progression of cardiometabolic disorders remain poorly understood. Adipokines are adipocyte-specific cytokines that are central to deleterious cardiometabolic alterations. They exhibit both pro-inflammatory and anti-inflammatory effects, complicating their association with cardiometabolic disturbances. Thus, understanding the cardiometabolic association of adipokines from a molecular and signaling perspective assumes great importance. This review presents a comprehensive outline of the most prominent adipokines exhibiting pro-inflammatory and/or anti-inflammatory functions in cardiometabolic dysfunction. The review also presents an insight into the pathophysiological implications of such adipokines in different cardiometabolic dysfunction conditions, the status of adipokine druggability, and future studies that can be undertaken to address the existing scientific gap. A clear understanding of the functional and mechanistic role of adipokines can potentially improve our understanding of cardiovascular disease pathophysiology and enhance our current therapeutic regimen in the years to come.
Collapse
Affiliation(s)
- Sayantap Datta
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, 4349 Martin Luther King Blvd., Houston, TX 77204, USA;
| | - Saisudha Koka
- Department of Pharmaceutical Sciences, College of Pharmacy, Texas A & M University, Kingsville, TX 78363, USA
| | - Krishna M. Boini
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, 4349 Martin Luther King Blvd., Houston, TX 77204, USA;
| |
Collapse
|
3
|
Kosheleva L, Koshelev D, Lagunas-Rangel FA, Levit S, Rabinovitch A, Schiöth HB. Disease-modifying pharmacological treatments of type 1 diabetes: Molecular mechanisms, target checkpoints, and possible combinatorial treatments. Pharmacol Rev 2025; 77:100044. [PMID: 40014914 PMCID: PMC11964952 DOI: 10.1016/j.pharmr.2025.100044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 01/10/2025] [Indexed: 03/01/2025] Open
Abstract
After a century of extensive scientific investigations, there is still no curative or disease-modifying treatment available that can provide long-lasting remission for patients diagnosed with type 1 diabetes (T1D). Although T1D has historically been regarded as a classic autoimmune disorder targeting and destroying pancreatic islet β-cells, significant research has recently demonstrated that β-cells themselves also play a substantial role in the disease's progression, which could explain some of the unfavorable clinical outcomes. We offer a thorough review of scientific and clinical insights pertaining to molecular mechanisms behind pathogenesis and the different therapeutic interventions in T1D covering over 20 possible pharmaceutical intervention treatments. The interventions are categorized as immune therapies, treatments targeting islet endocrine dysfunctions, medications with dual modes of action in immune and islet endocrine cells, and combination treatments with a broader spectrum of activity. We suggest that these collective findings can provide a valuable platform to discover new combinatorial synergies in search of the curative disease-modifying intervention for T1D. SIGNIFICANCE STATEMENT: This research delves into the underlying causes of T1D and identifies critical mechanisms governing β-cell function in both healthy and diseased states. Thus, we identify specific pathways that could be manipulated by existing or new pharmacological interventions. These interventions fall into several categories: (1) immunomodifying therapies individually targeting immune cell processes, (2) interventions targeting β-cells, (3) compounds that act simultaneously on both immune cell and β-cell pathways, and (4) combinations of compounds simultaneously targeting immune and β-cell pathways.
Collapse
Affiliation(s)
- Liudmila Kosheleva
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala, Sweden
| | - Daniil Koshelev
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala, Sweden
| | - Francisco Alejandro Lagunas-Rangel
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala, Sweden; Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia
| | - Shmuel Levit
- Diabetes and Metabolism Institute, Assuta Medical Centers, Tel Aviv, Israel
| | | | - Helgi B Schiöth
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala, Sweden; Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia.
| |
Collapse
|
4
|
Jeun R. Immunotherapies for prevention and treatment of type 1 diabetes. Immunotherapy 2025; 17:201-210. [PMID: 40033931 PMCID: PMC11951698 DOI: 10.1080/1750743x.2025.2473311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 02/25/2025] [Indexed: 03/05/2025] Open
Abstract
Type 1 diabetes (T1D) is characterized by the autoimmune destruction of insulin-producing β-cells of the pancreatic islets necessitating lifelong insulin therapy. Despite significant advancements in diabetes technology with increasingly sophisticated methods of insulin delivery and glucose monitoring, people with T1D remain at risk of severe complications like hypoglycemia and diabetic ketoacidosis. There has long been an interest in altering the immune response in T1D to prevent or cure T1D across its various stages with limited efficacy. This review highlights immunomodulatory approaches over the years including the anti-CD3 monoclonal antibody teplizumab which is now approved to delay onset of T1DM and other interventions under current investigation.
Collapse
Affiliation(s)
- Rebecca Jeun
- Division of Endocrinology, Diabetes & Metabolism, University of Louisville, Louisville, KY, USA
| |
Collapse
|
5
|
Inalkac Gemici Y, Ekici C, Batum M, Akbostanci C, Koc A, Mavioglu H. Familial Mediterranean fever gene variations could trigger VPS16-associated early-onset dystonia and diabetes mellitus: clinical identification of a family with MEFV and VPS16 genetic variation association. Rheumatol Adv Pract 2024; 8:rkae083. [PMID: 39055541 PMCID: PMC11272166 DOI: 10.1093/rap/rkae083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 06/17/2024] [Indexed: 07/27/2024] Open
Abstract
Objectives We describe the clinical pictures of an index case with dystonia and his family resulting from VPS16 and MEFV genetic variations based on previously published data and discuss the mechanisms that may have brought out the clinical findings. Methods A 17-year-old male had generalized dystonia that started at age 6 years, non-febrile abdominal pain attacks and was diagnosed with type 1 diabetes at age 14 years. Meanwhile, his 13-year-old sister had the same clinical presentation. His father was diabetic and his mother was asymptomatic. There was no consanguinity between the parents. Genetic variations were detected with whole exome sequencing. Results VPS16 c.1513C>T/p.Arg505* (likely pathogenic), MEFV c.2080A>G p.Met694val (pathogenic) and MEFV c.1772T>C p.Ile591Thr (unknown significance) heterozygous variants were detected in his siblings. The father had VPS16 c.1513C>T/p.Arg505* and MEFV c.2080A>G p Met694val variations and the mother had MEFV c.1772T>C p.Ile591Thr variations. Conclusions The occurrence of these diseases in siblings but their absence in the parents suggests the idea that the coexistence of two separate variations in the VPS16 and MEFV genes determines the phenotype. In addition, the increase in MEFV variation load in this family and the fact that DM occurs at an earlier age suggest that inflammation may cause an early diabetic clinical presentation.
Collapse
Affiliation(s)
- Yagmur Inalkac Gemici
- Department of Neurology, Manisa Celal Bayar University School of Medicine, Manisa, Turkey
| | - Cemal Ekici
- Department of Medical Genetics, Inonu University School of Medicine, Malatya, Turkey
| | - Melike Batum
- Department of Neurology, Manisa Celal Bayar University School of Medicine, Manisa, Turkey
| | - Cenk Akbostanci
- Department of Neurology, Ankara University School of Medicine, Ankara, Turkey
| | - Ahmet Koc
- Department of Medical Genetics, Inonu University School of Medicine, Malatya, Turkey
| | - Hatice Mavioglu
- Department of Neurology, Manisa Celal Bayar University School of Medicine, Manisa, Turkey
| |
Collapse
|
6
|
Heidari E, Shafiee A, Noorian S, Rafiei MA, Abbasi M, Amini MJ, Safari O, Aghamahdi F, Bakhtiyari M. Efficacy of teplizumab for treatment of type 1 diabetes: A meta-analysis of randomized controlled trials. Diabetes Metab Res Rev 2024; 40:e3806. [PMID: 38757421 DOI: 10.1002/dmrr.3806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 04/17/2024] [Accepted: 04/22/2024] [Indexed: 05/18/2024]
Abstract
BACKGROUND The management of Type 1 Diabetes Mellitus (T1DM) is a significant clinical challenge. This study evaluated the efficacy of teplizumab, an immunomodulatory drug, in patients with T1DM, using a systematic review and meta-analysis approach. METHODS We systematically searched multiple databases including Medline, Scopus, and others up to 10 January 2024, without language or regional restrictions. We included randomized controlled trials (RCTs) comparing teplizumab with placebo in T1DM patients. RESULTS Our analysis incorporated 8 RCTs, predominantly involving participants aged 7-35 years, diagnosed with T1DM and treated with 14-day courses of teplizumab. The primary outcomes included insulin use, C-peptide levels, and HbA1c levels. We observed a significant reduction in insulin use in the teplizumab group standardised mean difference of -0.50 (95% Confidence Interval [CI]: -0.76 to -0.23, p < 0.001; I2 = 49%). C-peptide levels were consistently higher in the teplizumab group, indicating improved endogenous insulin production. However, no significant change was noted in HbA1c levels between the groups. Quality assessment indicated a low risk of bias in most studies. CONCLUSIONS Teplizumab has a significant impact on reducing insulin dependence and enhancing endogenous insulin production in T1DM patients. However, its effect on long-term glycaemic control, as indicated by HbA1c levels, remains inconclusive.
Collapse
Affiliation(s)
- Ehsan Heidari
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
- Student Research Committee, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Arman Shafiee
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
- Student Research Committee, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Shahab Noorian
- Department of Pediatric Endocrinology and Metabolism, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Mohammad Ali Rafiei
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Abbasi
- Student Research Committee, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Mohammad Javad Amini
- Student Research Committee, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Omid Safari
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Fatemeh Aghamahdi
- Department of Pediatric Endocrinology and Metabolism, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Mahmood Bakhtiyari
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| |
Collapse
|
7
|
Weston CS, Boehm BO, Pozzilli P. Type 1 diabetes: A new vision of the disease based on endotypes. Diabetes Metab Res Rev 2024; 40:e3770. [PMID: 38450851 DOI: 10.1002/dmrr.3770] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 01/02/2024] [Accepted: 01/22/2024] [Indexed: 03/08/2024]
Abstract
Diagnosis and management of type 1 diabetes (T1D) have remained largely unchanged for the last several years. The management of the disease remains primarily focused on its phenotypical presentation and less on endotypes, namely the specific biological mechanisms behind the development of the disease. Furthermore, the treatment of T1D is essentially universal and indiscriminate-with patients administering insulin at varying dosages and frequencies to maintain adequate glycaemic control. However, it is now well understood that T1D is a heterogeneous disease with many different biological mechanisms (i.e. endotypes) behind its complex pathophysiology. A range of factors, including age of onset, immune system regulation, rate of β-cell destruction, autoantibodies, body weight, genetics and the exposome are recognised to play a role in the development of the condition. Patients can be classified into distinct diabetic subtypes based on these factors, which can be used to categorise patients into specific endotypes. The classification of patients into endotypes allows for a greater understanding of the natural progression of the disease, giving rise to more accurate and patient-centred therapies and follow-up monitoring, specifically for other autoimmune diseases. This review proposes 6 unique endotypes of T1D based on the current literature. The recognition of these endotypes could then be used to direct therapeutic modalities based on patients' individual pathophysiology.
Collapse
Affiliation(s)
- Craig Sinclair Weston
- Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | | | - Paolo Pozzilli
- Endocrinology and Metabolic Diseases, Campus Bio-Medico of Rome, Rome, Italy
- Centre of Immunobiology, The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
8
|
Antar SA, Ashour NA, Sharaky M, Khattab M, Ashour NA, Zaid RT, Roh EJ, Elkamhawy A, Al-Karmalawy AA. Diabetes mellitus: Classification, mediators, and complications; A gate to identify potential targets for the development of new effective treatments. Biomed Pharmacother 2023; 168:115734. [PMID: 37857245 DOI: 10.1016/j.biopha.2023.115734] [Citation(s) in RCA: 86] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/13/2023] [Accepted: 10/13/2023] [Indexed: 10/21/2023] Open
Abstract
Nowadays, diabetes mellitus has emerged as a significant global public health concern with a remarkable increase in its prevalence. This review article focuses on the definition of diabetes mellitus and its classification into different types, including type 1 diabetes (idiopathic and fulminant), type 2 diabetes, gestational diabetes, hybrid forms, slowly evolving immune-mediated diabetes, ketosis-prone type 2 diabetes, and other special types. Diagnostic criteria for diabetes mellitus are also discussed. The role of inflammation in both type 1 and type 2 diabetes is explored, along with the mediators and potential anti-inflammatory treatments. Furthermore, the involvement of various organs in diabetes mellitus is highlighted, such as the role of adipose tissue and obesity, gut microbiota, and pancreatic β-cells. The manifestation of pancreatic Langerhans β-cell islet inflammation, oxidative stress, and impaired insulin production and secretion are addressed. Additionally, the impact of diabetes mellitus on liver cirrhosis, acute kidney injury, immune system complications, and other diabetic complications like retinopathy and neuropathy is examined. Therefore, further research is required to enhance diagnosis, prevent chronic complications, and identify potential therapeutic targets for the management of diabetes mellitus and its associated dysfunctions.
Collapse
Affiliation(s)
- Samar A Antar
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute, Virginia Tech, Roanoke, VA 24016, USA; Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Horus University, New Damietta 34518, Egypt
| | - Nada A Ashour
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| | - Marwa Sharaky
- Cancer Biology Department, Pharmacology Unit, National Cancer Institute (NCI), Cairo University, Cairo, Egypt
| | - Muhammad Khattab
- Department of Chemistry of Natural and Microbial Products, Division of Pharmaceutical and Drug Industries, National Research Centre, Cairo, Egypt
| | - Naira A Ashour
- Department of Neurology, Faculty of Physical Therapy, Horus University, New Damietta 34518, Egypt
| | - Roaa T Zaid
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ahram Canadian University, 6th of October City, Giza 12566, Egypt
| | - Eun Joo Roh
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science & Technology, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Ahmed Elkamhawy
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Republic of Korea; Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| | - Ahmed A Al-Karmalawy
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ahram Canadian University, 6th of October City, Giza 12566, Egypt; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta 34518, Egypt
| |
Collapse
|
9
|
Thakkar S, Chopra A, Nagendra L, Kalra S, Bhattacharya S. Teplizumab in Type 1 Diabetes Mellitus: An Updated Review. TOUCHREVIEWS IN ENDOCRINOLOGY 2023; 19:22-30. [PMID: 38187075 PMCID: PMC10769466 DOI: 10.17925/ee.2023.19.2.7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 07/01/2023] [Indexed: 01/09/2024]
Abstract
Type 1 diabetes mellitus (T1DM) is a chronic autoimmune condition characterized by the irreversible destruction of the β cells of the pancreas, which leads to a lifelong dependency on exogenous insulin. Despite the advancements in insulin delivery methods, the suboptimal outcomes of these methods have triggered the search for therapies that may prevent or reverse the disease. Given the autoimmune aetiology of T1DM, therapies counteracting the immune-mediated destruction of the β-cells are the obvious target. Although several treatment strategies have been attempted to target cellular, humoral and innate immunity, very few have had a clinically meaningful impact. Of all the available immunomodulatory agents, cluster of differentiation (CD) 3 antibodies have exhibited the most promising preclinical and clinical results. Muromonab-CD3, which also happened to be a murine CD3 antibody, was the first monoclonal antibody approved for clinical use and was primarily indicated for graft rejection. The adverse effects associated with muromonab-CD3 led to its withdrawal. Teplizumab, a newer CD3 antibody, has a better side-effect profile because of its humanized nature and non-Fc-receptor-binding domain. In November 2022, teplizumab became the first immunomodulatory agent to be licensed by the US Food and Drug Administration for delaying the onset of T1DM in high-risk adults and children over 8 years old. The mechanism seems to be enhancing regulatory T-cell activity and promoting immune tolerance. This article reviews the mechanism of action and the clinical trials of teplizumab in individuals with T1DM or at risk of developing the disease.
Collapse
Affiliation(s)
- Simran Thakkar
- Department of Endocrinology, Indraprastha Apollo Hospitals, New Delhi, India
| | - Aditi Chopra
- Department of Endocrinology, Manipal Hospital, Bengaluru, India
| | | | - Sanjay Kalra
- Department of Endocrinology, Bharti Hospital, Karnal, Haryana, India
| | | |
Collapse
|
10
|
Arefin A, Gage MC. Metformin, Empagliflozin, and Their Combination Modulate Ex-Vivo Macrophage Inflammatory Gene Expression. Int J Mol Sci 2023; 24:ijms24054785. [PMID: 36902218 PMCID: PMC10003317 DOI: 10.3390/ijms24054785] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 02/09/2023] [Accepted: 02/23/2023] [Indexed: 03/06/2023] Open
Abstract
Type-2 Diabetes Mellitus is a complex, chronic illness characterized by persistent high blood glucose levels. Patients can be prescribed anti-diabetes drugs as single agents or in combination depending on the severity of their condition. Metformin and empagliflozin are two commonly prescribed anti-diabetes drugs which reduce hyperglycemia, however their direct effects on macrophage inflammatory responses alone or in combination are unreported. Here, we show that metformin and empagliflozin elicit proinflammatory responses on mouse bone-marrow-derived macrophages with single agent challenge, which are modulated when added in combination. In silico docking experiments suggested that empagliflozin can interact with both TLR2 and DECTIN1 receptors, and we observed that both empagliflozin and metformin increase expression of Tlr2 and Clec7a. Thus, findings from this study suggest that metformin and empagliflozin as single agents or in combination can directly modulate inflammatory gene expression in macrophages and upregulate the expression of their receptors.
Collapse
Affiliation(s)
- Adittya Arefin
- Wolfson Institute for Biomedical Research, Division of Medicine, University College London, Gower Street, London WC1E 6BT, UK
| | - Matthew C. Gage
- Department of Comparative Biomedical Sciences, Royal Veterinary College, 4 Royal College Street, London NW1 0TU, UK
- Correspondence:
| |
Collapse
|
11
|
Wang J, Wang L, Zhang XJ, Zhang P, Cai J, She ZG, Li H. Recent updates on targeting the molecular mediators of NAFLD. J Mol Med (Berl) 2023; 101:101-124. [PMID: 36792729 DOI: 10.1007/s00109-022-02282-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/29/2022] [Accepted: 12/21/2022] [Indexed: 02/17/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is rapidly becoming the most common disease worldwide in an era of rapid economic growth. NAFLD is a multifactorial disease, involving multiple genetic, metabolic, and environmental factors, and is closely associated with metabolic syndrome, obesity, and cardiovascular disease. NAFLD can be classified into nonalcoholic fatty liver disease (NAFL) and nonalcoholic steatohepatitis (NASH), which can both progress to cirrhosis and even hepatocellular carcinoma (HCC). Due to the enormous burden of NAFLD and its complications, no FDA-approved drugs for the treatment of NAFLD are on the market, and therapeutic targets and drug therapies are being actively investigated. In view of the various pathological mechanisms of NAFLD, numbers of preclinical studies and clinical trials have made rapid progress. This review mainly summarizes the most recently characterized mechanisms and therapeutic targets in each mechanism of NAFLD, focusing on the mechanism and application potential.
Collapse
Affiliation(s)
- Jia Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Luojia Mount Wuchang, Wuhan, China
- Institute of Model Animal, Wuhan University, Wuhan, China
| | - Lei Wang
- Department of Neurosurgery, Huanggang Central Hospital of Yangtze University, Huanggang, China
- Translation Medicine Research Center, Yangtze University, Huanggang, China
| | - Xiao-Jing Zhang
- Institute of Model Animal, Wuhan University, Wuhan, China
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Peng Zhang
- Institute of Model Animal, Wuhan University, Wuhan, China
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Jingjing Cai
- Institute of Model Animal, Wuhan University, Wuhan, China
- Department of Cardiology, The Third Xiangya Hospital, Central South University, The Third Xiangya Hospital, Changsha, China
| | - Zhi-Gang She
- Department of Cardiology, Renmin Hospital of Wuhan University, Luojia Mount Wuchang, Wuhan, China.
- Institute of Model Animal, Wuhan University, Wuhan, China.
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Luojia Mount Wuchang, Wuhan, China.
- Institute of Model Animal, Wuhan University, Wuhan, China.
- Translation Medicine Research Center, Yangtze University, Huanggang, China.
| |
Collapse
|
12
|
Clare K, Dillon JF, Brennan PN. Reactive Oxygen Species and Oxidative Stress in the Pathogenesis of MAFLD. J Clin Transl Hepatol 2022; 10:939-946. [PMID: 36304513 PMCID: PMC9547261 DOI: 10.14218/jcth.2022.00067] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 05/22/2022] [Accepted: 06/06/2022] [Indexed: 12/04/2022] Open
Abstract
The pathogenesis of metabolic-associated fatty liver disease (MAFLD) is complex and thought to be dependent on multiple parallel hits on a background of genetic susceptibility. The evidence suggests that MAFLD progression is a dynamic two-way process relating to repetitive bouts of metabolic stress and inflammation interspersed with endogenous anti-inflammatory reparative responses. In MAFLD, excessive hepatic lipid accumulation causes the production of lipotoxins that induce mitochondrial dysfunction, endoplasmic reticular stress, and over production of reactive oxygen species (ROS). Models of MAFLD show marked disruption of mitochondrial function and reduced oxidative capacitance with impact on cellular processes including mitophagy, oxidative phosphorylation, and mitochondrial biogenesis. In excess, ROS modify insulin and innate immune signaling and alter the expression and activity of essential enzymes involved in lipid homeostasis. ROS can also cause direct damage to intracellular structures causing hepatocyte injury and death. In select cases, the use of anti-oxidants and ROS scavengers have been shown to diminish the pro-apoptopic effects of fatty acids. Given this link, endogenous anti-oxidant pathways have been a target of interest, with Nrf2 activation showing a reduction in oxidative stress and inflammation in models of MAFLD. Thyroid hormone receptor β (THRβ) agonists and nuclear peroxisome proliferation-activated receptor (PPAR) family have also gained interest in reducing hepatic lipotoxicity and restoring hepatic function in models of MAFLD. Unfortunately, the true interplay between the clinical and molecular components of MAFLD progression remain only partly understood. Most recently, multiomics-based strategies are being adopted for hypothesis-free analysis of the molecular changes in MAFLD. Transcriptome profiling maps the unique genotype-phenotype associations in MAFLD and with various single-cell transcriptome-based projects underway, there is hope of novel physiological insights to MAFLD progression and uncover therapeutic targets.
Collapse
Affiliation(s)
- Kathleen Clare
- Royal Alexandra Hospital, Paisley, NHS Greater Glasgow and Clyde, PA2 9PN, UK
| | - John F. Dillon
- University of Dundee, Ninewells Hospital and Medical School, Dundee, DD1 9SY, UK
| | - Paul N. Brennan
- University of Dundee, Ninewells Hospital and Medical School, Dundee, DD1 9SY, UK
- University of Edinburgh, Edinburgh BioQuarter, 5 Little France Drive, EH16 4UU, UK
- Correspondence to: Paul N. Brennan, University of Dundee, Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, Dundee, DD1 9SY, UK. ORCID: https://orcid.org/0000-0001-8368-1478. Tel: +44-7445308786, E-mail:
| |
Collapse
|
13
|
High Endogenously Synthesized N-3 Polyunsaturated Fatty Acids in Fat-1 Mice Attenuate High-Fat Diet-Induced Insulin Resistance by Inhibiting NLRP3 Inflammasome Activation via Akt/GSK-3β/TXNIP Pathway. Molecules 2022; 27:molecules27196384. [PMID: 36234919 PMCID: PMC9570616 DOI: 10.3390/molecules27196384] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/14/2022] [Accepted: 09/21/2022] [Indexed: 01/15/2023] Open
Abstract
High-fat (HF) diets and low-grade chronic inflammation contribute to the development of insulin resistance and type 2 diabetes (T2D), whereas n-3 polyunsaturated fatty acids (PUFAs), due to their anti-inflammatory effects, protect against insulin resistance. Interleukin (IL)-1β is implicated in insulin resistance, yet how n-3 PUFAs modulate IL-1β secretion and attenuate HF diet-induced insulin resistance remains elusive. In this study, a HF diet activated NLRP3 inflammasome via inducing reactive oxygen species (ROS) generation and promoted IL-1β production primarily from adipose tissue preadipocytes, but not from adipocytes and induced insulin resistance in wild type (WT) mice. Interestingly, endogenous synthesized n-3 polyunsaturated fatty acids (PUFAs) reversed this process in HF diet-fed fat-1 transgenic mice although the HF diet induced higher weight gain in fat-1 mice, compared with the control diet. Mechanistically, palmitic acid (PA), the main saturated fatty acid in an HF diet inactivated AMPK and led to decreased GSK-3β phosphorylation, at least partially through reducing Akt activity, which ultimately blocked the Nrf2/Trx1 antioxidant pathway and induced TXNIP cytoplasm translocation and NLRP3 inflammasome activation, whereas docosahexaenoic acid (DHA), the most abundant n-3 PUFA in fat-1 adipose tissue, reversed this process via inducing Akt activation. Our GSK-3β shRNA knockdown study further revealed that GSK-3β played a pivot role between the upstream AMPK/Akt pathway and downstream Nrf2/Trx1/TXNIP pathway. Given that NLRP3 inflammasome is implicated in the development of most inflammatory diseases, our results suggest the potential of n-3 PUFAs in the prevention or adjuvant treatment of NLRP3 inflammasome-driven diseases.
Collapse
|
14
|
Rohm TV, Meier DT, Olefsky JM, Donath MY. Inflammation in obesity, diabetes, and related disorders. Immunity 2022; 55:31-55. [PMID: 35021057 PMCID: PMC8773457 DOI: 10.1016/j.immuni.2021.12.013] [Citation(s) in RCA: 912] [Impact Index Per Article: 304.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 12/13/2021] [Accepted: 12/17/2021] [Indexed: 01/13/2023]
Abstract
Obesity leads to chronic, systemic inflammation and can lead to insulin resistance (IR), β-cell dysfunction, and ultimately type 2 diabetes (T2D). This chronic inflammatory state contributes to long-term complications of diabetes, including non-alcoholic fatty liver disease (NAFLD), retinopathy, cardiovascular disease, and nephropathy, and may underlie the association of type 2 diabetes with other conditions such as Alzheimer's disease, polycystic ovarian syndrome, gout, and rheumatoid arthritis. Here, we review the current understanding of the mechanisms underlying inflammation in obesity, T2D, and related disorders. We discuss how chronic tissue inflammation results in IR, impaired insulin secretion, glucose intolerance, and T2D and review the effect of inflammation on diabetic complications and on the relationship between T2D and other pathologies. In this context, we discuss current therapeutic options for the treatment of metabolic disease, advances in the clinic and the potential of immune-modulatory approaches.
Collapse
Affiliation(s)
- Theresa V. Rohm
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Daniel T. Meier
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, CH-4031 Basel, Switzerland.,Department of Biomedicine (DBM), University of Basel, University Hospital Basel, CH-4031 Basel, Switzerland
| | - Jerrold M. Olefsky
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Marc Y. Donath
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, CH-4031 Basel, Switzerland.,Department of Biomedicine (DBM), University of Basel, University Hospital Basel, CH-4031 Basel, Switzerland.,Correspondence:
| |
Collapse
|
15
|
Vallianou NG, Stratigou T, Geladari E, Tessier CM, Mantzoros CS, Dalamaga M. Diabetes type 1: Can it be treated as an autoimmune disorder? Rev Endocr Metab Disord 2021; 22:859-876. [PMID: 33730229 DOI: 10.1007/s11154-021-09642-4] [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: 03/03/2021] [Indexed: 02/06/2023]
Abstract
Type 1 Diabetes Mellitus (T1DM) is characterized by progressive autoimmune-mediated destruction of the pancreatic beta-cells leading to insulin deficiency and hyperglycemia. It is associated with significant treatment burden and necessitates life-long insulin therapy. The role of immunotherapy in the prevention and management of T1DM is an evolving area of interest which has the potential to alter the natural history of this disease.In this review, we give insight into recent clinical trials related to the use of immunotherapeutic approaches for T1DM, such as proinflammatory cytokine inhibition, cell-depletion and cell-therapy approaches, autoantigen-specific treatments and stem cell therapies. We highlight the timing of intervention, aspects of therapy including adverse effects and the emergence of a novel lymphocyte crucial in T1DM autoimmunity. We also discuss the role of cardiac autoimmunity and its link to excess CVD risk in T1DM.We conclude that significant advances have been made in development of immunotherapeutic targets and agents for the treatment and prevention of T1DM. These immune-based therapies promise preservation of beta-cells and decreasing insulin dependency. In their current state, immunotherapeutic approaches cannot yet halt the progression from a preclinical state to overt T1DM nor can they replace standard insulin therapy in existing T1DM. It remains to be seen whether immunotherapy will ultimately play a key role in the prevention of progression to overt T1DM and whether it may find a place in our therapeutic armamentarium to improve clinical outcomes and quality of life in established T1DM.
Collapse
Affiliation(s)
- Natalia G Vallianou
- Department of Internal Medicine, Evangelismos General Hospital, 45-47 Ipsilantou str, 10676, Athens, Greece
| | - Theodora Stratigou
- Department of Endocrinology, Diabetes and Metabolic Diseases, Evangelismos General Hospital, 45-47 Ipsilantou str, 10676, Athens, Greece
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias, 11527, Athens, Goudi, Greece
| | - Eleni Geladari
- Department of Internal Medicine, Evangelismos General Hospital, 45-47 Ipsilantou str, 10676, Athens, Greece
| | - Christopher M Tessier
- Endocrinology Section, VA Boston Healthcare System, 1400 VFW Parkway West Roxbury, Boston, MA, 02132, USA.
| | - Christos S Mantzoros
- Endocrinology Section, VA Boston Healthcare System, 1400 VFW Parkway West Roxbury, Boston, MA, 02132, USA
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias, 11527, Athens, Goudi, Greece
| |
Collapse
|
16
|
Tripolino C, Ciaffi J, Pucino V, Ruscitti P, van Leeuwen N, Borghi C, Giacomelli R, Meliconi R, Ursini F. Insulin Signaling in Arthritis. Front Immunol 2021; 12:672519. [PMID: 33995414 PMCID: PMC8119635 DOI: 10.3389/fimmu.2021.672519] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 04/12/2021] [Indexed: 12/23/2022] Open
Abstract
Inflammatory arthritis is burdened by an increased risk of metabolic disorders. Cytokines and other mediators in inflammatory diseases lead to insulin resistance, diabetes and hyperlipidemia. Accumulating evidence in the field of immunometabolism suggests that the cause-effect relationship between arthritis and metabolic abnormalities might be bidirectional. Indeed, the immune response can be modulated by various factors such as environmental agents, bacterial products and hormones. Insulin is produced by pancreatic cells and regulates glucose, fat metabolism and cell growth. The action of insulin is mediated through the insulin receptor (IR), localized on the cellular membrane of hepatocytes, myocytes and adipocytes but also on the surface of T cells, macrophages, and dendritic cells. In murine models, the absence of IR in T-cells coincided with reduced cytokine production, proliferation, and migration. In macrophages, defective insulin signaling resulted in enhanced glycolysis affecting the responses to pathogens. In this review, we focalize on the bidirectional cause-effect relationship between impaired insulin signaling and arthritis analyzing how insulin signaling may be involved in the aberrant immune response implicated in arthritis and how inflammatory mediators affect insulin signaling. Finally, the effect of glucose-lowering agents on arthritis was summarized.
Collapse
Affiliation(s)
- Cesare Tripolino
- Geriatric Medicine Unit, Department of Medical Functional Area, "San Giovanni di Dio" Hospital, Crotone, Italy
| | - Jacopo Ciaffi
- Medicine and Rheumatology Unit, IRCCS Istituto Ortopedico Rizzoli (IOR), Bologna, Italy
| | - Valentina Pucino
- Institute of Inflammation and Ageing, University of Birmingham and Queen Elizabeth Hospital, Birmingham, United Kingdom
| | - Piero Ruscitti
- Rheumatology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Nina van Leeuwen
- Rheumatology Department, Leiden University Medical Center, Leiden, Netherlands
| | - Claudio Borghi
- Unità Operativa Medicina Interna Cardiovascolare-IRCCS Azienda Ospedaliera-Universitaria, Bologna, Italy
| | - Roberto Giacomelli
- Rheumatology and Immunology Unit, Department of Medicine, University of Rome "Campus Biomedico", Rome, Italy
| | - Riccardo Meliconi
- Geriatric Medicine Unit, Department of Medical Functional Area, "San Giovanni di Dio" Hospital, Crotone, Italy.,Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Francesco Ursini
- Geriatric Medicine Unit, Department of Medical Functional Area, "San Giovanni di Dio" Hospital, Crotone, Italy.,Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum University of Bologna, Bologna, Italy
| |
Collapse
|
17
|
Farooq N, Chuan B, Mahmud H, El Khoudary SR, Nouraie SM, Evankovich J, Yang L, Dunlap D, Bain W, Kitsios G, Zhang Y, O’Donnell CP, McVerry BJ, Shah FA. Association of the systemic host immune response with acute hyperglycemia in mechanically ventilated septic patients. PLoS One 2021; 16:e0248853. [PMID: 33755703 PMCID: PMC7987165 DOI: 10.1371/journal.pone.0248853] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 03/07/2021] [Indexed: 12/13/2022] Open
Abstract
Hyperglycemia during sepsis is associated with increased organ dysfunction and higher mortality. The role of the host immune response in development of hyperglycemia during sepsis remains unclear. We performed a retrospective analysis of critically ill adult septic patients requiring mechanical ventilation (n = 153) to study the relationship between hyperglycemia and ten markers of the host injury and immune response measured on the first day of ICU admission (baseline). We determined associations between each biomarker and: (1) glucose, insulin, and c-peptide levels at the time of biomarker collection by Pearson correlation; (2) average glucose and glycemic variability in the first two days of ICU admission by linear regression; and (3) occurrence of hyperglycemia (blood glucose>180mg/dL) by logistic regression. Results were adjusted for age, pre-existing diabetes mellitus, severity of illness, and total insulin and glucocorticoid dose. Baseline plasma levels of ST2 and procalcitonin were positively correlated with average blood glucose and glycemic variability in the first two days of ICU admission in unadjusted and adjusted analyses. Additionally, higher baseline ST2, IL-1ra, procalcitonin, and pentraxin-3 levels were associated with increased risk of hyperglycemia. Our results suggest associations between the host immune response and hyperglycemia in critically ill septic patients particularly implicating the interleukin-1 axis (IL-1ra), the interleukin-33 axis (ST2), and the host response to bacterial infections (procalcitonin, pentraxin-3).
Collapse
Affiliation(s)
- Nauman Farooq
- Division of General Internal Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Byron Chuan
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Hussain Mahmud
- Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Samar R. El Khoudary
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Seyed Mehdi Nouraie
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - John Evankovich
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Libing Yang
- School of Medicine, Tsinghua University, Haidian District, Beijing, China
| | - Daniel Dunlap
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - William Bain
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, United States of America
| | - Georgios Kitsios
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Center for Medicine and the Microbiome, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Yingze Zhang
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Christopher P. O’Donnell
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Bryan J. McVerry
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Center for Medicine and the Microbiome, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Faraaz Ali Shah
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
18
|
Wang CR, Tsai HW. Anti- and non-tumor necrosis factor-α-targeted therapies effects on insulin resistance in rheumatoid arthritis, psoriatic arthritis and ankylosing spondylitis. World J Diabetes 2021; 12:238-260. [PMID: 33758645 PMCID: PMC7958474 DOI: 10.4239/wjd.v12.i3.238] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/07/2021] [Accepted: 01/22/2021] [Indexed: 02/06/2023] Open
Abstract
In addition to β-cell failure with inadequate insulin secretion, the crucial mechanism leading to establishment of diabetes mellitus (DM) is the resistance of target cells to insulin, i.e. insulin resistance (IR), indicating a requirement of beyond-normal insulin concentrations to maintain euglycemic status and an ineffective strength of transduction signaling from the receptor, downstream to the substrates of insulin action. IR is a common feature of most metabolic disorders, particularly type II DM as well as some cases of type I DM. A variety of human inflammatory disorders with increased levels of proinflammatory cytokines, including tumor necrosis factor (TNF)-α, interleukin (IL)-6 and IL-1β, have been reported to be associated with an increased risk of IR. Autoimmune-mediated arthritis conditions, including rheumatoid arthritis (RA), psoriatic arthritis (PsA) and ankylosing spondylitis (AS), with the involvement of proinflammatory cytokines as their central pathogenesis, have been demonstrated to be associated with IR, especially during the active disease state. There is an increasing trend towards using biologic agents and small molecule-targeted drugs to treat such disorders. In this review, we focus on the effects of anti-TNF-α- and non-TNF-α-targeted therapies on IR in patients with RA, PsA and AS. Anti-TNF-α therapy, IL-1 blockade, IL-6 antagonist, Janus kinase inhibitor and phospho-diesterase type 4 blocker can reduce IR and improve diabetic hyper-glycemia in autoimmune-mediated arthritis.
Collapse
Affiliation(s)
- Chrong-Reen Wang
- Department of Internal Medicine, National Cheng Kung University Hospital, Tainan 70403, Taiwan
| | - Hung-Wen Tsai
- Department of Pathology, National Cheng Kung University Hospital, Tainan 70403, Taiwan
| |
Collapse
|
19
|
AlZaim I, Hammoud SH, Al-Koussa H, Ghazi A, Eid AH, El-Yazbi AF. Adipose Tissue Immunomodulation: A Novel Therapeutic Approach in Cardiovascular and Metabolic Diseases. Front Cardiovasc Med 2020; 7:602088. [PMID: 33282920 PMCID: PMC7705180 DOI: 10.3389/fcvm.2020.602088] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 10/22/2020] [Indexed: 12/12/2022] Open
Abstract
Adipose tissue is a critical regulator of systemic metabolism and bodily homeostasis as it secretes a myriad of adipokines, including inflammatory and anti-inflammatory cytokines. As the main storage pool of lipids, subcutaneous and visceral adipose tissues undergo marked hypertrophy and hyperplasia in response to nutritional excess leading to hypoxia, adipokine dysregulation, and subsequent low-grade inflammation that is characterized by increased infiltration and activation of innate and adaptive immune cells. The specific localization, physiology, susceptibility to inflammation and the heterogeneity of the inflammatory cell population of each adipose depot are unique and thus dictate the possible complications of adipose tissue chronic inflammation. Several lines of evidence link visceral and particularly perivascular, pericardial, and perirenal adipose tissue inflammation to the development of metabolic syndrome, insulin resistance, type 2 diabetes and cardiovascular diseases. In addition to the implication of the immune system in the regulation of adipose tissue function, adipose tissue immune components are pivotal in detrimental or otherwise favorable adipose tissue remodeling and thermogenesis. Adipose tissue resident and infiltrating immune cells undergo metabolic and morphological adaptation based on the systemic energy status and thus a better comprehension of the metabolic regulation of immune cells in adipose tissues is pivotal to address complications of chronic adipose tissue inflammation. In this review, we discuss the role of adipose innate and adaptive immune cells across various physiological and pathophysiological states that pertain to the development or progression of cardiovascular diseases associated with metabolic disorders. Understanding such mechanisms allows for the exploitation of the adipose tissue-immune system crosstalk, exploring how the adipose immune system might be targeted as a strategy to treat cardiovascular derangements associated with metabolic dysfunctions.
Collapse
Affiliation(s)
- Ibrahim AlZaim
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Safaa H. Hammoud
- Department of Pharmacology and Therapeutics, Beirut Arab University, Beirut, Lebanon
| | - Houssam Al-Koussa
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon
| | - Alaa Ghazi
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon
| | - Ali H. Eid
- Department of Pharmacology and Therapeutics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Basic Medical Sciences, College of Medicine, Qatar University, Doha, Qatar
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Ahmed F. El-Yazbi
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
20
|
Banerjee A, Singh J. Remodeling adipose tissue inflammasome for type 2 diabetes mellitus treatment: Current perspective and translational strategies. Bioeng Transl Med 2020; 5:e10150. [PMID: 32440558 PMCID: PMC7237149 DOI: 10.1002/btm2.10150] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 11/07/2019] [Accepted: 12/03/2019] [Indexed: 12/14/2022] Open
Abstract
Obesity-associated type 2 diabetes mellitus (T2DM) is characterized by low-grade chronic systemic inflammation that arises primarily from the white adipose tissue. The interplay between various adipose tissue-derived chemokines drives insulin resistance in T2DM and has therefore become a subject of rigorous investigation. The adipocytokines strongly associated with glucose homeostasis include tumor necrosis factor-α, various interleukins, monocyte chemoattractant protein-1, adiponectin, and leptin, among others. Remodeling the adipose tissue inflammasome in obesity-associated T2DM is likely to treat the underlying cause of the disease and bring significant therapeutic benefit. Various strategies have been adopted or are being investigated to modulate the serum/tissue levels of pro- and anti-inflammatory adipocytokines to improve glucose homeostasis in T2DM. These include use of small molecule agonists/inhibitors, mimetics, antibodies, gene therapy, and other novel formulations. Here, we discuss adipocytokines that are strongly associated with insulin activity and therapies that are under investigation for modulation of their levels in the treatment of T2DM.
Collapse
Affiliation(s)
- Amrita Banerjee
- Department of Pharmaceutical SciencesNorth Dakota State UniversityFargoNorth Dakota
| | - Jagdish Singh
- Department of Pharmaceutical SciencesNorth Dakota State UniversityFargoNorth Dakota
| |
Collapse
|
21
|
Lu J, Liu J, Li L, Lan Y, Liang Y. Cytokines in type 1 diabetes: mechanisms of action and immunotherapeutic targets. Clin Transl Immunology 2020; 9:e1122. [PMID: 32185024 PMCID: PMC7074462 DOI: 10.1002/cti2.1122] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 01/31/2020] [Accepted: 03/01/2020] [Indexed: 12/17/2022] Open
Abstract
Cytokines play crucial roles in orchestrating complex multicellular interactions between pancreatic β cells and immune cells in the development of type 1 diabetes (T1D) and are thus potential immunotherapeutic targets for this disorder. Cytokines that can induce regulatory functions-for example, IL-10, TGF-β and IL-33-are thought to restore immune tolerance and prevent β-cell damage. By contrast, cytokines such as IL-6, IL-17, IL-21 and TNF, which promote the differentiation and function of diabetogenic immune cells, are thought to lead to T1D onset and progression. However, targeting these dysregulated cytokine networks does not always result in consistent effects because anti-inflammatory or proinflammatory functions of cytokines, responsible for β-cell destruction, are context dependent. In this review, we summarise the current knowledge on the involvement of well-known cytokines in both the initiation and destruction phases of T1D and discuss advances in recently discovered roles of cytokines. Additionally, we emphasise the complexity and implications of cytokine modulation therapy and discuss the ways in which this strategy has been translated into clinical trials.
Collapse
Affiliation(s)
- Jingli Lu
- Department of Pharmacy The First Affiliated Hospital of Zhengzhou University Zhengzhou China.,Henan Key Laboratory of Precision Clinical Pharmacy Zhengzhou University Zhengzhou China
| | - Jiyun Liu
- Department of Pharmacy The First Affiliated Hospital of Zhengzhou University Zhengzhou China.,Henan Key Laboratory of Precision Clinical Pharmacy Zhengzhou University Zhengzhou China
| | - Lulu Li
- Department of Pharmacy Wuhan No.1 Hospital Wuhan China
| | - Yan Lan
- Department of Pharmacy Huangshi Center Hospital Huangshi China
| | - Yan Liang
- Department of Pharmacy The First Affiliated Hospital of Zhengzhou University Zhengzhou China.,Henan Key Laboratory of Precision Clinical Pharmacy Zhengzhou University Zhengzhou China
| |
Collapse
|
22
|
Inhibition of IL-1beta improves Glycaemia in a Mouse Model for Gestational Diabetes. Sci Rep 2020; 10:3035. [PMID: 32080229 PMCID: PMC7033251 DOI: 10.1038/s41598-020-59701-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 01/29/2020] [Indexed: 01/13/2023] Open
Abstract
Gestational diabetes mellitus (GDM) is one of the most common diseases associated with pregnancy, however, the underlying mechanisms remain unclear. Based on the well documented role of inflammation in type 2 diabetes, the aim was to investigate the role of inflammation in GDM. We established a mouse model for GDM on the basis of its two major risk factors, obesity and aging. In these GDM mice, we observed increased Interleukin-1β (IL-1β) expression in the uterus and the placenta along with elevated circulating IL-1β concentrations compared to normoglycemic pregnant mice. Treatment with an anti-IL-1β antibody improved glucose-tolerance of GDM mice without apparent deleterious effects for the fetus. Finally, IL-1β antagonism showed a tendency for reduced plasma corticosterone concentrations, possibly explaining the metabolic improvement. We conclude that IL-1β is a causal driver of impaired glucose tolerance in GDM.
Collapse
|
23
|
Kietsiriroje N, Pearson S, Campbell M, Ariëns RAS, Ajjan RA. Double diabetes: A distinct high-risk group? Diabetes Obes Metab 2019; 21:2609-2618. [PMID: 31373146 DOI: 10.1111/dom.13848] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/23/2019] [Accepted: 07/29/2019] [Indexed: 01/06/2023]
Abstract
The term double diabetes (DD) has been used to refer to individuals with type 1 diabetes (T1D) who are overweight, have a family history of type 2 diabetes and/or clinical features of insulin resistance. Several pieces of evidence indicate that individuals who display features of DD are at higher risk of developing future diabetes complications, independently of average glucose control, measured as glycated haemoglobin (HbA1c) concentration. Given the increased prevalence of individuals with features of DD, pragmatic criteria are urgently required to identify and stratify this group, which will help with subsequent implementation of more effective personalized interventions. In this review, we discuss the potential criteria for the clinical identification of individuals with DD, highlighting the strengths and weaknesses of each definition. We also cover potential mechanisms of DD and how these contribute to increased risk of diabetes complications. Special emphasis is placed on the role of estimated glucose disposal rate (eGDR) in the diagnosis of DD, which can be easily incorporated into clinical practice and is predictive of adverse clinical outcome. In addition to the identification of individuals with DD, eGDR has potential utility in monitoring response to different interventions. T1D is a more heterogeneous condition than initially envisaged, and those with features of DD represent a subgroup at higher risk of complications. Pragmatic criteria for the diagnosis of individuals with DD will help with risk stratification, allowing a more personalized and targeted management strategy to improve outcome and quality of life in this population.
Collapse
Affiliation(s)
- Noppadol Kietsiriroje
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Hatyai, Songkhla, Thailand
| | - Sam Pearson
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Matthew Campbell
- School of Food Science and Nutrition, University of Leeds, Leeds, UK
| | - Robert A S Ariëns
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Ramzi A Ajjan
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| |
Collapse
|
24
|
Shahidi-Dadras M, Bahraini N, Rajabi F, Younespour S. Patients with alopecia areata show signs of insulin resistance. Arch Dermatol Res 2019; 311:529-533. [PMID: 31089876 DOI: 10.1007/s00403-019-01929-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 04/12/2019] [Accepted: 05/02/2019] [Indexed: 11/29/2022]
Abstract
Alopecia areata (AA) is an autoimmune disease associated with high levels of proinflammatory cytokines. Since chronic inflammation plays a major role in the pathogenesis of insulin resistance, AA can theoretically increase the risk of diabetes. We sought to investigate this theory by conducting a case-control study. Sixty patients with alopecia areata and 60 healthy volunteers (matched for age, sex, and body mass index) were evaluated. Fasting blood glucose (FBS), C-peptide, plasma insulin, and homeostasis model assessment for insulin resistance (HOMA-IR) were measured for each individual. Plasma levels of insulin [median (interquartile range IQR): 11.22 (7.28-18.15) µIU/ml vs. 4.80 (3.20-9.00), p < 0.0001)], C-peptide [median (IQR): 2.10 (1.61-3.00) ng/ml vs. 1.40 (1.20-1.88), p < 0.0001)] and HOMA-IR [median (IQR): 2.70 (1.58-3.96) µIU/ml vs. 1.01 (0.64-1.98, p < 0.0001)] were significantly higher in patients with AA compared to controls. The differences remained significant even after controlling for age, gender, and BMI. Patients with a more severe disease (alopecia totalis/universalis) had higher levels of insulin [median (IQR): 15.80 (9.68-21.55) vs. 9.30 (5.33-14.40), p = 0.02)] and HOMA-IR [median (IQR): 3.30 (2.20-4.84) vs. 2.15 (1.29-3.52), p = 0.01] compared to those with patchy hair loss. Our data suggest that individuals with AA are at a higher risk of developing insulin resistance. This may be due to common inflammatory pathogenesis or a shared genetic background.
Collapse
Affiliation(s)
| | - Negin Bahraini
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Fateme Rajabi
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Network of Dermatology Research (NDR), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Shima Younespour
- National Institute of Health Research, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
25
|
Tsalamandris S, Antonopoulos AS, Oikonomou E, Papamikroulis GA, Vogiatzi G, Papaioannou S, Deftereos S, Tousoulis D. The Role of Inflammation in Diabetes: Current Concepts and Future Perspectives. Eur Cardiol 2019; 14:50-59. [PMID: 31131037 PMCID: PMC6523054 DOI: 10.15420/ecr.2018.33.1] [Citation(s) in RCA: 809] [Impact Index Per Article: 134.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 02/18/2019] [Indexed: 02/06/2023] Open
Abstract
Diabetes is a complex metabolic disorder affecting the glucose status of the human body. Chronic hyperglycaemia related to diabetes is associated with end organ failure. The clinical relationship between diabetes and atherosclerotic cardiovascular disease is well established. This makes therapeutic approaches that simultaneously target diabetes and atherosclerotic disease an attractive area for research. The majority of people with diabetes fall into two broad pathogenetic categories, type 1 or type 2 diabetes. The role of obesity, adipose tissue, gut microbiota and pancreatic beta cell function in diabetes are under intensive scrutiny with several clinical trials to have been completed while more are in development. The emerging role of inflammation in both type 1 and type 2 diabetes (T1D and T1D) pathophysiology and associated metabolic disorders, has generated increasing interest in targeting inflammation to improve prevention and control of the disease. After an extensive review of the possible mechanisms that drive the metabolic pattern in T1D and T2D and the inflammatory pathways that are involved, it becomes ever clearer that future research should focus on a model of combined suppression for various inflammatory response pathways.
Collapse
Affiliation(s)
- Sotirios Tsalamandris
- First Cardiology Clinic, Hippokration General Hospital, National and Kapodistrian University of Athens, School of Medicine Athens, Greece
| | - Alexios S Antonopoulos
- First Cardiology Clinic, Hippokration General Hospital, National and Kapodistrian University of Athens, School of Medicine Athens, Greece
| | - Evangelos Oikonomou
- First Cardiology Clinic, Hippokration General Hospital, National and Kapodistrian University of Athens, School of Medicine Athens, Greece
| | - George-Aggelos Papamikroulis
- First Cardiology Clinic, Hippokration General Hospital, National and Kapodistrian University of Athens, School of Medicine Athens, Greece
| | - Georgia Vogiatzi
- First Cardiology Clinic, Hippokration General Hospital, National and Kapodistrian University of Athens, School of Medicine Athens, Greece
| | - Spyridon Papaioannou
- First Cardiology Clinic, Hippokration General Hospital, National and Kapodistrian University of Athens, School of Medicine Athens, Greece
| | - Spyros Deftereos
- First Cardiology Clinic, Hippokration General Hospital, National and Kapodistrian University of Athens, School of Medicine Athens, Greece
| | - Dimitris Tousoulis
- First Cardiology Clinic, Hippokration General Hospital, National and Kapodistrian University of Athens, School of Medicine Athens, Greece
| |
Collapse
|
26
|
Brennan FH, Popovich PG. Emerging targets for reprograming the immune response to promote repair and recovery of function after spinal cord injury. Curr Opin Neurol 2019; 31:334-344. [PMID: 29465433 DOI: 10.1097/wco.0000000000000550] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE OF REVIEW In adult mammals, a traumatic spinal cord injury (SCI) elicits a chronic unregulated neuroinflammatory response accompanied by seemingly paradoxical suppression of systemic immunity. These SCI-induced changes in immune function contribute to poor neurological outcomes and enhanced morbidity or mortality. Nonspecific anti-inflammatory or proinflammatory therapies are ineffective and can even worsen outcomes. Therefore, recent experimental SCI research has advanced the understanding of how neuroimmune cross-talk contributes to spinal cord and systemic pathology. RECENT FINDINGS It is now appreciated that the immune response caused by injury to the brain or spinal cord encompasses heterogeneous elements that can drive events on the spectrum between exacerbating pathology and promoting tissue repair, within the spinal cord and throughout the body. Recent novel discoveries regarding the role and regulation of soluble factors, monocytes/macrophages, microRNAs, lymphocytes and systemic immune function are highlighted in this review. SUMMARY A more nuanced understanding of how the immune system responds and reacts to nervous system injury will present an array of novel therapeutic opportunities for clinical SCI and other forms of neurotrauma.
Collapse
Affiliation(s)
- Faith H Brennan
- Center for Brain and Spinal Cord Repair, Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | | |
Collapse
|
27
|
Nopparat J, Nualla-ong A, Phongdara A. Ethanolic extracts of Pluchea indica (L.) leaf pretreatment attenuates cytokine-induced β-cell apoptosis in multiple low-dose streptozotocin-induced diabetic mice. PLoS One 2019; 14:e0212133. [PMID: 30779805 PMCID: PMC6380574 DOI: 10.1371/journal.pone.0212133] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 01/27/2019] [Indexed: 01/18/2023] Open
Abstract
Loss of β-cell mass and function is a fundamental feature of pathogenesis for type 1 and type 2 diabetes. Increasing evidence indicates that apoptosis is one of the main mechanisms of β-cell death in both types. Ethanolic extracts of Pluchea indica leaf (PILE) have been reported to possess blood glucose lowering actions in vivo. Nevertheless, further study is required to determine the underlying mechanisms. In this report, we have investigated the preventive effects of PILE on multiple low doses of streptozotocin (MLDS)-induced β-cell apoptosis. Mice were pre-treated with PILE at 50 mg/kg (PILE 50) or 100 mg/kg (PILE 100) for 2 weeks before streptozotocin (STZ) stimulation, and the treatment continued for 4 or 8 weeks. Results revealed that PILE 100 mice exhibited improved blood biochemistry, maintained a higher body weight, had decreased hyperglycemia, and restored islet architectures compared to non-treated STZ mice. Significantly, PILE 100 decreased levels of inflammatory response markers interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α), and interlukin1-β (IL-1β), concomitant with the inhibition of caspase-3, caspase-8, capsepase-9, phosphorylation of signal transducer and activator of transcription 1 (pSTAT1), nuclear factor-κBp65 (NF-κBp65), and inducible nitric oxide synthase (iNOS). Additionally, survival and proliferative ability of β-cells was mediated by up-regulated Bcl-2 and Ki67, respectively. These results provide strong evidence that pretreatment with PILE 100 effectively attenuated STZ-induced diabetes-related symptoms and these effects could be associated with the inhibition of cytokine-induced β-cell apoptosis.
Collapse
Affiliation(s)
- Jongdee Nopparat
- Department of Anatomy, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, Thailand
- * E-mail:
| | - Aekkaraj Nualla-ong
- Center for Genomics and Bioinformatics Research, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, Thailand
- Medical Technology Service Center, Faculty of Medical Technology, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| | - Amornrat Phongdara
- Center for Genomics and Bioinformatics Research, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, Thailand
- Faculty of Medical Technology, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| |
Collapse
|
28
|
Ruscitti P, Ursini F, Cipriani P, Greco M, Alvaro S, Vasiliki L, Di Benedetto P, Carubbi F, Berardicurti O, Gulletta E, De Sarro G, Giacomelli R. IL-1 inhibition improves insulin resistance and adipokines in rheumatoid arthritis patients with comorbid type 2 diabetes: An observational study. Medicine (Baltimore) 2019; 98:e14587. [PMID: 30762811 PMCID: PMC6408058 DOI: 10.1097/md.0000000000014587] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Recently, it has been shown that some well-known pathogenic mediators in rheumatoid arthritis (RA), such as interleukin-1β (IL-1β) and tumor necrosis factor (TNF), could play a pathogenic role in insulin resistance and (IR) and type 2 diabetes (T2D).In this 6-month longitudinal study, we aimed at investigating if the inhibition of IL-1 or TNF is associated with an improvement of IR in RA patients with comorbid T2D and the possible effects on selected serum adipokines. RA patients with comorbid T2D were recruited among those undergoing treatment with anakinra (ANA) or with TNF inhibitor (TNFi). The 1998-updated version of the Homeostasis Model Assessment (HOMA2) was used to calculate surrogate indexes of IR (HOMA2-IR) and steady-state beta cell function (%B) from fasting values of glucose and C-peptide. Glucagon, adiponectin, adipsin, leptin, and resistin were also measured. All these parameters were collected at baseline, after 3 and 6 months of treatment.ANA-treated patients showed a significant improvement in HOMA2-%β, HOMA2-IR, and glucagon. In TNFi-treated patients, no significant difference was observed analyzing these metabolic parameters. Adipsin and resistin decreased after 6 months in ANA-treated patients whereas, no difference was recognized analyzing adiponectin and leptin. In TNFi-treated patients, leptin and resistin significantly increased, whereas no difference was found analyzing adiponectin and adipsin, during the follow-up.Our data may suggest a beneficial effect of IL-1 inhibition on measures of metabolic derangement in RA-associated T2D. If further confirmed by larger studies, IL-1 targeting therapies may represent a tailored approach in these patients.
Collapse
Affiliation(s)
- Piero Ruscitti
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila
| | - Francesco Ursini
- Department of Health Sciences, University of Catanzaro “Magna Graecia”, Catanzaro
| | - Paola Cipriani
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila
| | - Marta Greco
- Clinical Pathology Unit, University of Catanzaro “Magna Graecia”, Catanzaro, Italy
| | - Saverio Alvaro
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila
| | - Liakouli Vasiliki
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila
| | - Paola Di Benedetto
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila
| | - Francesco Carubbi
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila
| | - Onorina Berardicurti
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila
| | - Elio Gulletta
- Clinical Pathology Unit, University of Catanzaro “Magna Graecia”, Catanzaro, Italy
| | | | - Roberto Giacomelli
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila
| |
Collapse
|
29
|
Zhong L, Tran T, Baguley TD, Lee SJ, Henke A, To A, Li S, Yu S, Grieco FA, Roland J, Schultz PG, Eizirik DL, Rogers N, Chartterjee AK, Tremblay MS, Shen W. A novel inhibitor of inducible NOS dimerization protects against cytokine-induced rat beta cell dysfunction. Br J Pharmacol 2018; 175:3470-3485. [PMID: 29888783 PMCID: PMC6086989 DOI: 10.1111/bph.14388] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 05/14/2018] [Accepted: 05/28/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND AND PURPOSE Beta cell apoptosis is a major feature of type 1 diabetes, and pro-inflammatory cytokines are key drivers of the deterioration of beta cell mass through induction of apoptosis. Mitochondrial stress plays a critical role in mediating apoptosis by releasing cytochrome C into the cytoplasm, directly activating caspase-9 and its downstream signalling cascade. We aimed to identify new compounds that protect beta cells from cytokine-induced activation of the intrinsic (mitochondrial) pathway of apoptosis. EXPERIMENTAL APPROACH Diabetogenic media, composed of IL-1β, IFN-γ and high glucose, were used to induce mitochondrial stress in rat insulin-producing INS1E cells, and a high-content image-based screen of small molecule modulators of Casp9 pathway was performed. KEY RESULTS A novel small molecule, ATV399, was identified from a high-content image-based screen for compounds that inhibit cleaved caspase-9 activation and subsequent beta cell apoptosis induced by a combination of IL-1β, IFN-γ and high glucose, which together mimic the pathogenic diabetic milieu. Through medicinal chemistry optimization, potency was markedly improved (6-30 fold), with reduced inhibitory effects on CYP3A4. Improved analogues, such as CAT639, improved beta cell viability and insulin secretion in cytokine-treated rat insulin-producing INS1E cells and primary dispersed islet cells. Mechanistically, CAT639 reduced the production of NO by allosterically inhibiting dimerization of inducible NOS (iNOS) without affecting its mRNA levels. CONCLUSION AND IMPLICATIONS Taken together, these studies demonstrate a successful phenotypic screening campaign resulting in identification of an inhibitor of iNOS dimerization that protects beta cell viability and function through modulation of mitochondrial stress induced by cytokines.
Collapse
Affiliation(s)
- Linlin Zhong
- California Institute for Biomedical Research (Calibr)La JollaCA92037USA
| | - Tuan Tran
- California Institute for Biomedical Research (Calibr)La JollaCA92037USA
| | - Tyler D Baguley
- California Institute for Biomedical Research (Calibr)La JollaCA92037USA
| | - Sang Jun Lee
- California Institute for Biomedical Research (Calibr)La JollaCA92037USA
| | - Adam Henke
- California Institute for Biomedical Research (Calibr)La JollaCA92037USA
| | - Andrew To
- California Institute for Biomedical Research (Calibr)La JollaCA92037USA
| | - Sijia Li
- California Institute for Biomedical Research (Calibr)La JollaCA92037USA
| | - Shan Yu
- California Institute for Biomedical Research (Calibr)La JollaCA92037USA
| | - Fabio A Grieco
- ULB Center for Diabetes ResearchUniversite´ Libre de Bruxelles (ULB)Brussels1070Belgium
| | - Jason Roland
- California Institute for Biomedical Research (Calibr)La JollaCA92037USA
| | - Peter G Schultz
- California Institute for Biomedical Research (Calibr)La JollaCA92037USA
- Department of ChemistryThe Scripps Research InstituteLa JollaCA92037USA
| | - Decio L Eizirik
- ULB Center for Diabetes ResearchUniversite´ Libre de Bruxelles (ULB)Brussels1070Belgium
| | - Nikki Rogers
- California Institute for Biomedical Research (Calibr)La JollaCA92037USA
| | | | | | - Weijun Shen
- California Institute for Biomedical Research (Calibr)La JollaCA92037USA
| |
Collapse
|
30
|
Ratter JM, Tack CJ, Netea MG, Stienstra R. Environmental Signals Influencing Myeloid Cell Metabolism and Function in Diabetes. Trends Endocrinol Metab 2018; 29:468-480. [PMID: 29789206 DOI: 10.1016/j.tem.2018.04.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 04/18/2018] [Accepted: 04/19/2018] [Indexed: 12/13/2022]
Abstract
The environment induces metabolic reprogramming of immune cells via specific signaling pathways. Recent studies have revealed that changes in cell metabolism affect key immune cell functions including cytokine production and migration. In diabetes, these functions are either insufficiently or excessively activated, translating into diabetes-associated complications, including increased susceptibility to infection and accelerated cardiovascular disease. Diabetes alters the abundance of environmental signals, including glucose, insulin, and lipids. Subsequently, changes in environmental signals drive metabolic reprogramming, impair immune cell function, and ultimately contribute to diabetes-associated complications. We review here recent studies on changes in innate immune cell metabolism, especially in myeloid cells, that are driven by environmental signals relevant to diabetes, and discuss therapeutic perspectives of targeting metabolism of immune cells in diabetes.
Collapse
Affiliation(s)
- Jacqueline M Ratter
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands; Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - Cees J Tack
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mihai G Netea
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands; Department for Genomics and Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Rinke Stienstra
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands; Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands.
| |
Collapse
|
31
|
Gao N, Me R, Dai C, Seyoum B, Yu FSX. Opposing Effects of IL-1Ra and IL-36Ra on Innate Immune Response to Pseudomonas aeruginosa Infection in C57BL/6 Mouse Corneas. THE JOURNAL OF IMMUNOLOGY 2018; 201:688-699. [PMID: 29891552 DOI: 10.4049/jimmunol.1800046] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 05/16/2018] [Indexed: 12/22/2022]
Abstract
Pseudomonas aeruginosa keratitis is characterized by severe corneal ulceration and may lead to blindness if not treated properly in a timely manner. Although the roles of the IL-1 subfamily of cytokines are well established, as a newly discovered subfamily, IL-36 cytokine regulation, immunological relevance, and relation with IL-1 cytokines in host defense remain largely unknown. In this study, we showed that P. aeruginosa infection induces the expression of IL-36α and IL-36γ, as well as IL-1β and secreted IL-1Ra (sIL-1Ra), but not IL-36Ra. Downregulation of IL-1Ra increases, whereas downregulation of IL-36Ra decreases the severity of P. aeruginosa keratitis. IL-1R and IL-36Ra downregulation have opposing effects on the expression of IL-1β, sIL-1Ra, IL-36γ, S100A8, and CXCL10 and on the infiltration of innate immune cells. Administration of recombinant IL-1Ra improved, whereas IL-36Ra worsened the outcome of P. aeruginosa keratitis. Local application of IL-36γ stimulated the expression of innate defense molecules S100A9, mouse β-defensin 3, but suppressed IL-1β expression in B6 mouse corneas. IL-36γ diminished the severity of P. aeruginosa keratitis, and its protective effects were abolished in the presence of S100A9 neutralizing Ab and partially affected by CXCL10 and CXCR3 neutralizations. Thus, our data reveal that IL-1Ra and IL-36Ra have opposing effects on the outcome of P. aeruginosa keratitis and suggest that IL-36 agonists may be used as an alternative therapeutic to IL-1β-neutralizing reagents in controlling microbial keratitis and other mucosal infections.
Collapse
Affiliation(s)
- Nan Gao
- Department of Ophthalmology, Wayne State University School of Medicine, Detroit, MI 48201.,Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, MI 48201
| | - Rao Me
- Department of Ophthalmology, Wayne State University School of Medicine, Detroit, MI 48201.,Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, MI 48201
| | - Chenyang Dai
- Department of Ophthalmology, Wayne State University School of Medicine, Detroit, MI 48201.,Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, MI 48201.,Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan, Shandong Province 250014, China; and
| | - Berhane Seyoum
- Division of Endocrinology, Department of Internal Medicine, Wayne State University School of Medicine, Detroit, MI 48201
| | - Fu-Shin X Yu
- Department of Ophthalmology, Wayne State University School of Medicine, Detroit, MI 48201; .,Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, MI 48201
| |
Collapse
|
32
|
Mirea AM, Tack CJ, Chavakis T, Joosten LAB, Toonen EJM. IL-1 Family Cytokine Pathways Underlying NAFLD: Towards New Treatment Strategies. Trends Mol Med 2018; 24:458-471. [PMID: 29665983 PMCID: PMC5939989 DOI: 10.1016/j.molmed.2018.03.005] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 03/11/2018] [Accepted: 03/12/2018] [Indexed: 02/07/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common liver disease worldwide. Pathways responsible for the activation of IL-1 family cytokines are key in the development of NAFLD but underlying mechanisms are not fully understood. Many studies have focused on the inflammasome-caspase-1 pathway and have shown that this pathway is an important inducer of inflammation in NAFLD. However, this pathway is not solely responsible for the activation of proinflammatory cytokines. Also, neutrophil serine proteases (NSPs) are capable of activating cytokines and recent studies reported that these proteases also contribute to NAFLD. These studies provided, for the first time, evidence that this inflammasome-independent pathway is involved in NAFLD. In our opinion, these new insights open up new approaches for therapeutic intervention.
Collapse
Affiliation(s)
- Andreea-Manuela Mirea
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Cees J Tack
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Carl-Gustav-Carus, Technische Universität Dresden, Dresden, Germany
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Erik J M Toonen
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands; R&D Department, Hycult Biotech, Uden, The Netherlands.
| |
Collapse
|
33
|
Wang YH, Zhang YG. Amyloid and immune homeostasis. Immunobiology 2018; 223:288-293. [DOI: 10.1016/j.imbio.2017.10.038] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Accepted: 10/14/2017] [Indexed: 01/06/2023]
|
34
|
Kaminitz A, Ash S, Askenasy N. Neutralization Versus Reinforcement of Proinflammatory Cytokines to Arrest Autoimmunity in Type 1 Diabetes. Clin Rev Allergy Immunol 2018; 52:460-472. [PMID: 27677500 DOI: 10.1007/s12016-016-8587-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
As physiological pathways of intercellular communication produced by all cells, cytokines are involved in the pathogenesis of inflammatory insulitis as well as pivotal mediators of immune homeostasis. Proinflammatory cytokines including interleukins, interferons, transforming growth factor-β, tumor necrosis factor-α, and nitric oxide promote destructive insulitis in type 1 diabetes through amplification of the autoimmune reaction, direct toxicity to β-cells, and sensitization of islets to apoptosis. The concept that neutralization of cytokines may be of therapeutic benefit has been tested in few clinical studies, which fell short of inducing sustained remission or achieving disease arrest. Therapeutic failure is explained by the redundant activities of individual cytokines and their combinations, which are rather dispensable in the process of destructive insulitis because other cytolytic pathways efficiently compensate their deficiency. Proinflammatory cytokines are less redundant in regulation of the inflammatory reaction, displaying protective effects through restriction of effector cell activity, reinforcement of suppressor cell function, and participation in islet recovery from injury. Our analysis suggests that the role of cytokines in immune homeostasis overrides their contribution to β-cell death and may be used as potent immunomodulatory agents for therapeutic purposes rather than neutralized.
Collapse
Affiliation(s)
- Ayelet Kaminitz
- The Leah and Edward M. Frankel Laboratory of Experimental Bone Marrow Transplantation, 14 Kaplan Street, Petach Tikva, Israel, 49202
| | - Shifra Ash
- The Leah and Edward M. Frankel Laboratory of Experimental Bone Marrow Transplantation, 14 Kaplan Street, Petach Tikva, Israel, 49202
| | - Nadir Askenasy
- The Leah and Edward M. Frankel Laboratory of Experimental Bone Marrow Transplantation, 14 Kaplan Street, Petach Tikva, Israel, 49202.
| |
Collapse
|
35
|
Ursini F, Russo E, Ruscitti P, Giacomelli R, De Sarro G. The effect of non-TNF-targeted biologics and small molecules on insulin resistance in inflammatory arthritis. Autoimmun Rev 2018; 17:399-404. [PMID: 29452240 DOI: 10.1016/j.autrev.2017.11.030] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Accepted: 11/25/2017] [Indexed: 12/14/2022]
Abstract
Inflammatory arthritides are chronic diseases characterised by an increase in cardiovascular risk, largely attributable to the synergy between high-grade systemic inflammation and an elevated prevalence of traditional cardiovascular risk factors. Amongst the latter, insulin resistance and type 2 diabetes (T2D) play a key position. Previous studies demonstrated a potential insulin-sensitizing effect of anti-TNF biologic medications. For converse, less is known about the role of newer biologics or small molecules. For this reason, we performed a systematic review of the literature in order to identify the available data on the effect on insulin resistance of non-TNF targeting biologics and small molecules approved for the treatment of inflammatory arthritides. The search strategy initially retrieved 486 records of which only 10 articles were selected for inclusion in the final review. According to the available evidence, some of the newest molecules, in particular tocilizumab and abatacept, may have a role in improving insulin sensitivity; for converse, anakinra-mediated effect on glucose metabolism may exploit different facets of T2D pathophysiology, such as the preservation of beta-cell function. However, the data available on this issue are largely inconsistent and future, adequately designed studies are still needed to clarify the differential impact of novel therapeutics on individual pathophysiological features of T2D and other emerging cardiovascular risk factors.
Collapse
Affiliation(s)
- Francesco Ursini
- Department of Health Sciences, University of Catanzaro "Magna Graecia", Catanzaro, Italy; Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom.
| | - Emilio Russo
- Department of Health Sciences, University of Catanzaro "Magna Graecia", Catanzaro, Italy
| | - Piero Ruscitti
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Science, University of L'Aquila, L'Aquila, Italy
| | - Roberto Giacomelli
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Science, University of L'Aquila, L'Aquila, Italy
| | | |
Collapse
|
36
|
Comorbidity of Type 1 Diabetes Mellitus in Patients with Juvenile Idiopathic Arthritis. J Pediatr 2018; 192:196-203. [PMID: 29246341 DOI: 10.1016/j.jpeds.2017.07.050] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 06/02/2017] [Accepted: 07/26/2017] [Indexed: 12/26/2022]
Abstract
OBJECTIVES To determine the prevalence of type 1 diabetes mellitus (T1D) in patients with juvenile idiopathic arthritis (JIA) and to characterize patients having both. STUDY DESIGN Diabetes comorbidity was recorded in the National Pediatric Rheumatologic Database since 2012. Data from the North Rhine-Westphalian diabetes registry served as the reference population for the prevalence of diabetes in the general population. The National Pediatric Rheumatologic Database data were indirectly standardized for age and sex for comparison with the general population. The diabetes prevalence ratio was calculated using the Poisson regression model. RESULTS The analysis included 12 269 patients with JIA. A total of 58 patients had comorbid T1D, and the diabetes prevalence was 0.5%. The mean age was 11.6 years at the time of documentation, and the mean disease duration was 4.2 years. Compared with the general population, the prevalence of diabetes in patients with JIA was significantly increased (prevalence ratio 1.76 [95% CI 1.34; 2.28], P < .001). The onset of diabetes in patients with JIA was earlier than that reported in the reference data. Sixty-three percent of patients developed T1D before JIA. On average, diabetes onset was 56 months before the onset of JIA. Patients who first developed JIA developed T1D on average 40 months later. The majority of patients had not received disease-modifying antirheumatic drugs before diabetes onset. CONCLUSIONS T1D occurs more frequently in patients with JIA than in the general population. The likelihood of T1D occurrence appears to be slightly higher before JIA manifestation and without disease-modifying antirheumatic drug therapy after JIA onset.
Collapse
|
37
|
TRPV1: A Potential Therapeutic Target in Type 2 Diabetes and Comorbidities? Trends Mol Med 2017; 23:1002-1013. [DOI: 10.1016/j.molmed.2017.09.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 09/07/2017] [Accepted: 09/14/2017] [Indexed: 12/14/2022]
|
38
|
Harouki N, Nicol L, Remy-Jouet I, Henry JP, Dumesnil A, Lejeune A, Renet S, Golding F, Djerada Z, Wecker D, Bolduc V, Bouly M, Roussel J, Richard V, Mulder P. The IL-1β Antibody Gevokizumab Limits Cardiac Remodeling and Coronary Dysfunction in Rats With Heart Failure. JACC Basic Transl Sci 2017; 2:418-430. [PMID: 30062160 PMCID: PMC6034492 DOI: 10.1016/j.jacbts.2017.06.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 06/06/2017] [Accepted: 06/08/2017] [Indexed: 12/31/2022]
Abstract
Immediate IL-1β antibody gevokizumab administration reduces ischemia/reperfusion related infarct size. Immediate and late IL-1β antibody gevokizumab administration improves heart failure related left ventricular remodeling. IL-1β antibody gevokizumab improves heart failure related coronary dysfunction.
This study reports preclinical data showing that the interleukin (IL)-1β modulation is a new promising target in the pathophysiological context of heart failure. Indeed, in nondiabetic Wistar and diabetic Goto-Kakizaki rats with chronic heart failure induced by myocardial infarction, administration of the IL-1β antibody gevokizumab improves ‘surrogate’ markers of survival (i.e., left ventricular remodeling, hemodynamics, and function as well as coronary function). However, whether IL-1β modulation per se or in combination with standard treatments of heart failure improves long-term outcome in human heart failure remains to be determined.
Collapse
Key Words
- GK, Goto-Kakisaki
- I/R, ischemia/reperfusion
- IL, interleukin
- IL-1β
- LV, left ventricle/ventricular
- LVEDP, left ventricular end-diastolic pressure
- LVEDPV, left ventricular end-diastolic pressure–volume relationship
- LVESP, left ventricular end-systolic pressure
- LVESPVR, left ventricular end-systolic pressure–volume relationship
- ROS, reactive oxygen species
- SOD, superoxide dismutase
- cardiovascular function
- heart failure
- ischemia/reperfusion
Collapse
Affiliation(s)
- Najah Harouki
- INSERM U1096, Rouen, France.,Normandy University, IRIB, Rouen, France.,Unité de formation et de recherche de Médecine et Pharmacie, Rouen University, Rouen, France
| | - Lionel Nicol
- INSERM U1096, Rouen, France.,Normandy University, IRIB, Rouen, France.,Unité de formation et de recherche de Médecine et Pharmacie, Rouen University, Rouen, France.,Plateau d'Imagerie CardioThoracique de l'Université de Rouen, Rouen, France
| | - Isabelle Remy-Jouet
- INSERM U1096, Rouen, France.,Normandy University, IRIB, Rouen, France.,Unité de formation et de recherche de Médecine et Pharmacie, Rouen University, Rouen, France
| | - Jean-Paul Henry
- INSERM U1096, Rouen, France.,Normandy University, IRIB, Rouen, France.,Unité de formation et de recherche de Médecine et Pharmacie, Rouen University, Rouen, France
| | - Anais Dumesnil
- INSERM U1096, Rouen, France.,Normandy University, IRIB, Rouen, France.,Unité de formation et de recherche de Médecine et Pharmacie, Rouen University, Rouen, France
| | - Annie Lejeune
- INSERM U1096, Rouen, France.,Normandy University, IRIB, Rouen, France.,Unité de formation et de recherche de Médecine et Pharmacie, Rouen University, Rouen, France
| | - Sylvanie Renet
- INSERM U1096, Rouen, France.,Normandy University, IRIB, Rouen, France.,Unité de formation et de recherche de Médecine et Pharmacie, Rouen University, Rouen, France
| | - Francesca Golding
- INSERM U1096, Rouen, France.,Normandy University, IRIB, Rouen, France.,Unité de formation et de recherche de Médecine et Pharmacie, Rouen University, Rouen, France
| | - Zoubir Djerada
- INSERM U1096, Rouen, France.,Normandy University, IRIB, Rouen, France.,Unité de formation et de recherche de Médecine et Pharmacie, Rouen University, Rouen, France.,Pharmacology Department, Centre Hospitalier Universitaire de Reims, Reims, France
| | | | - Virginie Bolduc
- Institut de Recherches Internationales Servier, Suresnes, France
| | - Muriel Bouly
- Institut de Recherches Internationales Servier, Suresnes, France
| | - Jerome Roussel
- Institut de Recherches Internationales Servier, Suresnes, France
| | - Vincent Richard
- INSERM U1096, Rouen, France.,Normandy University, IRIB, Rouen, France.,Unité de formation et de recherche de Médecine et Pharmacie, Rouen University, Rouen, France
| | - Paul Mulder
- INSERM U1096, Rouen, France.,Normandy University, IRIB, Rouen, France.,Unité de formation et de recherche de Médecine et Pharmacie, Rouen University, Rouen, France.,Plateau d'Imagerie CardioThoracique de l'Université de Rouen, Rouen, France
| |
Collapse
|
39
|
Frumento D, Ben Nasr M, El Essawy B, D'Addio F, Zuccotti GV, Fiorina P. Immunotherapy for type 1 diabetes. J Endocrinol Invest 2017; 40:803-814. [PMID: 28260183 DOI: 10.1007/s40618-017-0641-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 02/13/2017] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Although many approaches have been tested to overcome the insulin dependence caused by the pancreatic β-cells destruction observed in individuals affected by type 1 diabetes (T1D), medical research has largely failed to halt the onset or to reverse T1D. METHODS In this work, the state of the art of immunotherapy will be examined, and the most important achievement in the field will be critically discussed. Particularly, we will focus on the clinical aspect, thus avoiding the tedious preclinical work done in NOD mice, which has been so poorly translated to the bedside. CONCLUSIONS Stem cell therapies achieved thus this far the most promising results, while immune ablation and standard immunosuppressants did not maintain the premises of preclinical results. The next step will be to generate a feasible and safe clinical approach in order to cure the thousands of patients affected by T1D.
Collapse
Affiliation(s)
| | - Moufida Ben Nasr
- International Center for T1D, Pediatric Clinical Research Center Fondazione Romeo e Enrica Invernizzi, Department of Biomedical and Clinical Science L. Sacco, University of Milan, Milan, Italy
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Enders Building 5th floor Room EN511, 300 Longwood Ave, Boston, MA, USA
| | | | - Francesca D'Addio
- DITID, San Raffaele Hospital, Milan, Italy
- International Center for T1D, Pediatric Clinical Research Center Fondazione Romeo e Enrica Invernizzi, Department of Biomedical and Clinical Science L. Sacco, University of Milan, Milan, Italy
| | - Gian Vincenzo Zuccotti
- Pediatric Clinical Research Center Fondazione Romeo ed Enrica Invernizzi, Department of Biomedical and Clinical Science L. Sacco, University of Milan, Milan, Italy
| | - Paolo Fiorina
- International Center for T1D, Pediatric Clinical Research Center Fondazione Romeo e Enrica Invernizzi, Department of Biomedical and Clinical Science L. Sacco, University of Milan, Milan, Italy.
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Enders Building 5th floor Room EN511, 300 Longwood Ave, Boston, MA, USA.
| |
Collapse
|
40
|
Landmann EC, Walker UA. Pharmacological treatment options for cryopyrin-associated periodic syndromes. Expert Rev Clin Pharmacol 2017; 10:855-864. [DOI: 10.1080/17512433.2017.1338946] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
| | - Ulrich A. Walker
- Department of Rheumatology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
41
|
Pafili K, Maltezos E, Papanas N. Dapagliflozin for the treatment of type 1 diabetes mellitus. Expert Opin Investig Drugs 2017; 26:873-881. [DOI: 10.1080/13543784.2017.1339788] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Kalliopi Pafili
- Diabetes Centre, Second Department of Internal Medicine, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Efstratios Maltezos
- Diabetes Centre, Second Department of Internal Medicine, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Nikolaos Papanas
- Diabetes Centre, Second Department of Internal Medicine, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece
| |
Collapse
|
42
|
Surendar J, Indulekha K, Hoerauf A, Hübner MP. Immunomodulation by helminths: Similar impact on type 1 and type 2 diabetes? Parasite Immunol 2017; 39. [PMID: 27862000 DOI: 10.1111/pim.12401] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 11/04/2016] [Indexed: 12/19/2022]
Abstract
The incidence of both type 1 (T1D) and type 2 diabetes (T2D) is drastically increasing, and it is predicted that the global prevalence of diabetes will reach almost 600 million cases by 2035. Even though the pathogenesis of both types of diabetes is distinct, the immune system is actively involved in both forms of the disease. Genetic and environmental factors determine the risk to develop T1D. On the other hand, sedentary life style, surplus of food intake and other lifestyle changes contribute to the increase of T2D incidence. Improved sanitation with high-quality medical treatment is such an environmental factor that has led to a continuous reduction of infectious diseases including helminth infections over the past decades. Recently, a growing body of evidence has implicated a negative association between helminth infections and diabetes in humans as well as animal models. In this review, we discuss studies that have provided evidence for the beneficial impact of helminth infections on T1D and T2D. Possible mechanisms are presented by which helminths prevent T1D onset by mitigating pancreatic inflammation and confer protection against T2D by improving insulin sensitivity, alleviating inflammation, augmenting browning of adipose tissue and improving lipid metabolism and insulin signalling.
Collapse
Affiliation(s)
- J Surendar
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital of Bonn, Bonn, Germany
| | - K Indulekha
- LIMES Institute, Membrane Biology & Lipid Biochemistry, University of Bonn, Bonn, Germany
| | - A Hoerauf
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital of Bonn, Bonn, Germany
- German Centre for Infection Research (DZIF), partner site Bonn-Cologne, Bonn, Germany
| | - M P Hübner
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital of Bonn, Bonn, Germany
| |
Collapse
|
43
|
Li Y, Lu Z, Huang Y, Lopes-Virella MF, Virella G. F(ab′)2 fragments of anti-oxidized LDL IgG attenuate vascular inflammation and atherogenesis in diabetic LDL receptor-deficient mice. Clin Immunol 2016; 173:50-56. [DOI: 10.1016/j.clim.2016.07.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 07/18/2016] [Accepted: 07/21/2016] [Indexed: 10/21/2022]
|
44
|
High Mobility Group Box-1: A Missing Link between Diabetes and Its Complications. Mediators Inflamm 2016; 2016:3896147. [PMID: 27847406 PMCID: PMC5099456 DOI: 10.1155/2016/3896147] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 10/03/2016] [Indexed: 12/13/2022] Open
Abstract
High mobility group box-1 (HMGB-1), a damage-associated molecular pattern, can be actively or passively released from various cells under different conditions and plays a pivotal role in the pathogenesis of inflammation and angiogenesis-dependent diseases. More and more evidence suggests that inflammation, in addition to its role in progression of diabetes, also promotes initiation and development of diabetic complications. In this review, we focus on the role of HMGB-1 in diabetes-related complications and the therapeutic strategies targeting HMGB-1 in diabetic complications.
Collapse
|
45
|
Seelig E, Timper K, Falconnier C, Stoeckli R, Bilz S, Oram R, McDonald TJ, Donath MY. Interleukin-1 antagonism in type 1 diabetes of long duration. DIABETES & METABOLISM 2016; 42:453-456. [PMID: 27720360 DOI: 10.1016/j.diabet.2016.08.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 08/26/2016] [Accepted: 08/29/2016] [Indexed: 01/12/2023]
Affiliation(s)
- E Seelig
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, Basel, Switzerland.
| | - K Timper
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, Basel, Switzerland
| | - C Falconnier
- Division of Endocrinology, Diabetology and Metabolism, Medical University Clinic, Kantonsspital Baselland, Liestal, Switzerland
| | - R Stoeckli
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, Basel, Switzerland
| | - S Bilz
- Clinic for Endocrinology, Diabetes, Bone Disease and Metabolism, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - R Oram
- NIHR Exeter Clinical Research Facility, University of Exeter, Exeter, UK
| | - T J McDonald
- NIHR Exeter Clinical Research Facility, University of Exeter, Exeter, UK
| | - M Y Donath
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
46
|
Pilla SJ, Quan AQ, Germain-Lee EL, Hellmann DB, Mathioudakis NN. Immune-Modulating Therapy for Rheumatologic Disease: Implications for Patients with Diabetes. Curr Diab Rep 2016; 16:91. [PMID: 27525682 PMCID: PMC6031126 DOI: 10.1007/s11892-016-0792-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Immune modulators used to treat rheumatologic disease have diverse endocrine effects in patients with diabetes. Providers should be aware of these effects given that diabetes and rheumatologic disease overlap in prevalence and cardiovascular morbidity. In patients with type 1 diabetes, clinical trials have demonstrated that immune modulators used early in the disease can improve pancreatic function, though their efficacy in adults with longstanding autoimmune diabetes is unknown. In patients with type 2 diabetes, hydroxychloroquine is an effective antihyperglycemic and may be preferred for rheumatologic use in patients with difficult glycemic control. In patients without diabetes, hydroxychloroquine and tumor necrosis factor (TNF) inhibitors have been found to decrease diabetes incidence in observational studies. Additionally, dapsone and sulfasalazine alter erythrocyte survival resulting in inaccurate HbA1c values. These multifaceted effects of immune modulators create a need for coordinated care between providers treating patients with diabetes to individualize medication selection and prevent hypoglycemic events. More research is needed to determine the long-term outcomes of immune modulators in patients with diabetes.
Collapse
Affiliation(s)
- Scott J Pilla
- General Internal Medicine, Johns Hopkins University, 2024 E. Monument St, Room 2-604A, Baltimore, MD, 21205, USA
| | - Amy Q Quan
- Johns Hopkins School of Medicine, 2202 E Fairmount Ave, Baltimore, MD, 21231, USA
| | - Emily L Germain-Lee
- Department of Pediatrics, Division of Pediatric Endocrinology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- Kennedy Krieger Institute, Broadway, Room 583, Baltimore, MD, 801 N, USA
| | - David B Hellmann
- Department of Medicine, Johns Hopkins University School of Medicine and Johns Hopkins, Bayview, Johns Hopkins Bayview Medical Center, Mason F. Lord Building, Center Tower, Room 322, 5200 Eastern Avenue, Baltimore, MD, 21224, USA
| | - Nestoras N Mathioudakis
- Endocrinology, Diabetes, and Metabolism, Johns Hopkins University, 1830 E. Monument Street, Suite 333, Baltimore, MD, 21287, USA.
| |
Collapse
|
47
|
Kusminski CM, Bickel PE, Scherer PE. Targeting adipose tissue in the treatment of obesity-associated diabetes. Nat Rev Drug Discov 2016; 15:639-660. [PMID: 27256476 DOI: 10.1038/nrd.2016.75] [Citation(s) in RCA: 518] [Impact Index Per Article: 57.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Adipose tissue regulates numerous physiological processes, and its dysfunction in obese humans is associated with disrupted metabolic homeostasis, insulin resistance and type 2 diabetes mellitus (T2DM). Although several US-approved treatments for obesity and T2DM exist, these are limited by adverse effects and a lack of effective long-term glucose control. In this Review, we provide an overview of the role of adipose tissue in metabolic homeostasis and assess emerging novel therapeutic strategies targeting adipose tissue, including adipokine-based strategies, promotion of white adipose tissue beiging as well as reduction of inflammation and fibrosis.
Collapse
Affiliation(s)
- Christine M Kusminski
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center
| | - Perry E Bickel
- Division of Endocrinology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center
| |
Collapse
|
48
|
Donath MY. Multiple benefits of targeting inflammation in the treatment of type 2 diabetes. Diabetologia 2016; 59:679-82. [PMID: 26868493 DOI: 10.1007/s00125-016-3873-z] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 12/30/2015] [Indexed: 01/21/2023]
Abstract
The association between the metabolic syndrome and a pathological activation of the innate immune system is now well established. Thus, defective insulin secretion and action are due, at least in part, to islet, liver and fat inflammation in type 2 diabetes. Furthermore, an inflammatory process also seems to be involved in the development of cardiovascular, renal and ophthalmological complications of this disease. Interestingly, several other inflammatory diseases are associated with the metabolic syndrome, such as psoriasis, gout and rheumatic arthritis. The aim of this review is to discuss the clinical progress of anti-inflammatory drugs in the treatment of type 2 diabetes and then speculate on the possible further development of these drugs, with the aim of using the drugs in combination in order to combat the multiple manifestations of inflammatory diseases. This review summarises a presentation given at the 'Islet inflammation in type 2 diabetes' symposium at the 2015 annual meeting of the EASD. It is accompanied by two other reviews on topics from this symposium (by Simone Baltrusch, DOI: 10.1007/s00125-016-3891-x , and Jerry Nadler and colleagues, DOI: 10.1007/s00125-016-3890-y ) and a commentary by the Session Chair, Piero Marchetti (DOI: 10.1007/s00125-016-3875-x ).
Collapse
Affiliation(s)
- Marc Y Donath
- Endocrinology, Diabetes & Metabolism and Department of Biomedicine, University Hospital Basel, CH-4031, Basel, Switzerland.
| |
Collapse
|
49
|
Cucak H, Hansen G, Vrang N, Skarsfeldt T, Steiness E, Jelsing J. The IL-1β Receptor Antagonist SER140 Postpones the Onset of Diabetes in Female Nonobese Diabetic Mice. J Diabetes Res 2016; 2016:7484601. [PMID: 26953152 PMCID: PMC4756207 DOI: 10.1155/2016/7484601] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Revised: 01/07/2016] [Accepted: 01/10/2016] [Indexed: 12/21/2022] Open
Abstract
The cytokine interleukin-1β (IL-1β) is known to stimulate proinflammatory immune responses and impair β-cell function and viability, all critical events in the pathogenesis of type 1 diabetes (T1D). Here we evaluate the effect of SER140, a small peptide IL-1β receptor antagonist, on diabetes progression and cellular pancreatic changes in female nonobese diabetic (NOD) mice. Eight weeks of treatment with SER140 reduced the incidence of diabetes by more than 50% compared with vehicle, decreased blood glucose, and increased plasma insulin. Additionally, SER140 changed the endocrine and immune cells dynamics in the NOD mouse pancreas. Together, the data suggest that SER140 treatment postpones the onset of diabetes in female NOD mice by interfering with IL-1β activated pathways.
Collapse
Affiliation(s)
| | | | - Niels Vrang
- Gubra ApS, Agern Alle 1, 2970 Hørsholm, Denmark
| | | | - Eva Steiness
- Serodus ASA, Gaustadalléen 21, 0349 Oslo, Norway
| | - Jacob Jelsing
- Gubra ApS, Agern Alle 1, 2970 Hørsholm, Denmark
- *Jacob Jelsing:
| |
Collapse
|
50
|
Herder C, Dalmas E, Böni-Schnetzler M, Donath MY. The IL-1 Pathway in Type 2 Diabetes and Cardiovascular Complications. Trends Endocrinol Metab 2015; 26:551-563. [PMID: 26412156 DOI: 10.1016/j.tem.2015.08.001] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 07/31/2015] [Accepted: 08/03/2015] [Indexed: 12/31/2022]
Abstract
Patients with type 2 diabetes (T2D) exhibit chronic activation of the innate immune system in pancreatic islets, in insulin-sensitive tissues, and at sites of diabetic complications. This results from a pathological response to overnutrition and physical inactivity seen in genetically predisposed individuals. Processes mediated by the proinflammatory cytokine interleukin-1 (IL-1) link obesity and dyslipidemia and have implicated IL-1β in T2D and related cardiovascular complications. Epidemiological, molecular, and animal studies have now assigned a central role for IL-1β in driving tissue inflammation during metabolic stress. Proof-of-concept clinical studies have validated IL-1β as a target to improve insulin production and action in patients with T2D. Large ongoing clinical trials will address the potential of IL-1 antagonism to prevent cardiovascular and other related complications.
Collapse
Affiliation(s)
- Christian Herder
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; German Center for Diabetes Research, Partner Düsseldorf, 40225 Düsseldorf, Germany
| | - Elise Dalmas
- Endocrinology, Diabetes, and Metabolism and the Department of Biomedicine, University Hospital Basel, CH-4031 Basel, Switzerland
| | - Marianne Böni-Schnetzler
- Endocrinology, Diabetes, and Metabolism and the Department of Biomedicine, University Hospital Basel, CH-4031 Basel, Switzerland
| | - Marc Y Donath
- Endocrinology, Diabetes, and Metabolism and the Department of Biomedicine, University Hospital Basel, CH-4031 Basel, Switzerland.
| |
Collapse
|