1
|
Gonzalez-Sanchez FA, Sanchez-Huerta TM, Huerta-Gonzalez A, Sepulveda-Villegas M, Altamirano J, Aguilar-Aleman JP, Garcia-Varela R. Diabetes current and future translatable therapies. Endocrine 2024; 86:865-881. [PMID: 38971945 DOI: 10.1007/s12020-024-03944-8] [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: 04/19/2024] [Accepted: 06/23/2024] [Indexed: 07/08/2024]
Abstract
Diabetes is one of the major diseases and concerns of public health systems that affects over 200 million patients worldwide. It is estimated that 90% of these patients suffer from diabetes type 2, while 10% present diabetes type 1. This type of diabetes and certain types of diabetes type 2, are characterized by dysregulation of blood glycemic levels due to the total or partial depletion of insulin-secreting pancreatic β-cells. Different approaches have been proposed for long-term treatment of insulin-dependent patients; amongst them, cell-based approaches have been the subject of basic and clinical research since they allow blood glucose level sensing and in situ insulin secretion. The current gold standard for insulin-dependent patients is on-demand exogenous insulin application; cell-based therapies aim to remove this burden from the patient and caregivers. In recent years, protocols to isolate and implant pancreatic islets from diseased donors have been developed and tested in clinical trials. Nevertheless, the shortage of donors, along with the need of immunosuppressive companion therapies, have pushed researchers to focus their attention and efforts to overcome these disadvantages and develop alternative strategies. This review discusses current tested clinical approaches and future potential alternatives for diabetes type 1, and some diabetes type 2, insulin-dependent patients. Additionally, advantages and disadvantages of these discussed methods.
Collapse
Affiliation(s)
- Fabio Antonio Gonzalez-Sanchez
- Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Departamento de Bioingeniería y Biotecnología, Av. General Ramon Corona No 2514, Colonia Nuevo Mexico, CP 45201, Zapopan, Jalisco, México
| | - Triana Mayra Sanchez-Huerta
- Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Departamento de Bioingeniería y Biotecnología, Av. General Ramon Corona No 2514, Colonia Nuevo Mexico, CP 45201, Zapopan, Jalisco, México
| | - Alexandra Huerta-Gonzalez
- Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Departamento de Bioingeniería y Biotecnología, Av. General Ramon Corona No 2514, Colonia Nuevo Mexico, CP 45201, Zapopan, Jalisco, México
| | - Maricruz Sepulveda-Villegas
- Departamento de Medicina Genómica y Hepatología, Hospital Civil de Guadalajara, "Fray Antonio Alcalde", Guadalajara, 44280, Jalisco, Mexico
- Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, 44100, Jalisco, Mexico
| | - Julio Altamirano
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Epigmenio González 500, San Pablo, 76130, Santiago de Queretaro, Qro, México
| | - Juan Pablo Aguilar-Aleman
- Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Departamento de Ingenieria Biomedica, Av. General Ramon Corona No 2514, Colonia Nuevo Mexico, CP 45201, Zapopan, Jalisco, México
| | - Rebeca Garcia-Varela
- Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Departamento de Bioingeniería y Biotecnología, Av. General Ramon Corona No 2514, Colonia Nuevo Mexico, CP 45201, Zapopan, Jalisco, México.
- Carbone Cancer Center, University of Wisconsin - Madison, 1111 Highland Ave, Wisconsin, 53705, Madison, USA.
| |
Collapse
|
2
|
Kim DS, Song L, Gou W, Kim J, Liu B, Wei H, Muise-Helmericks RC, Li Z, Wang H. GRP94 is an IGF-1R chaperone and regulates beta cell death in diabetes. Cell Death Dis 2024; 15:374. [PMID: 38811543 PMCID: PMC11137047 DOI: 10.1038/s41419-024-06754-y] [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: 06/15/2023] [Revised: 05/10/2024] [Accepted: 05/16/2024] [Indexed: 05/31/2024]
Abstract
High workload-induced cellular stress can cause pancreatic islet β cell death and dysfunction, or β cell failure, a hallmark of type 2 diabetes mellitus. Thus, activation of molecular chaperones and other stress-response genes prevents β cell failure. To this end, we have shown that deletion of the glucose-regulated protein 94 (GRP94) in Pdx1+ pancreatic progenitor cells led to pancreas hypoplasia and reduced β cell mass during pancreas development in mice. Here, we show that GRP94 was involved in β cell adaption and compensation (or failure) in islets from leptin receptor-deficient (db/db) mice in an age-dependent manner. GRP94-deficient cells were more susceptible to cell death induced by various diabetogenic stress conditions. We also identified a new client of GRP94, insulin-like growth factor-1 receptor (IGF-1R), a critical factor for β cell survival and function that may mediate the effect of GRP94 in the pathogenesis of diabetes. This study has identified essential functions of GRP94 in β cell failure related to diabetes.
Collapse
Affiliation(s)
- Do-Sung Kim
- Department of Surgery, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Lili Song
- Department of Surgery, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Wenyu Gou
- Department of Surgery, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Jisun Kim
- Microbiology and Immunology, Medical University of South Carolina, Charleson, SC, 29425, USA
| | - Bei Liu
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center-James, Columbus, OH, 43210, USA
| | - Hua Wei
- Department of Surgery, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Robin C Muise-Helmericks
- Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Zihai Li
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center-James, Columbus, OH, 43210, USA
| | - Hongjun Wang
- Department of Surgery, Medical University of South Carolina, Charleston, SC, 29425, USA.
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC, USA.
| |
Collapse
|
3
|
Mattke J, Darden CM, Vasu S, Lawrence MC, Kirkland J, Kane RR, Naziruddin B. Inhibition of Toll-like Receptor 4 Using Small Molecule, TAK-242, Protects Islets from Innate Immune Responses. Cells 2024; 13:416. [PMID: 38474380 PMCID: PMC10931053 DOI: 10.3390/cells13050416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 02/23/2024] [Accepted: 02/24/2024] [Indexed: 03/14/2024] Open
Abstract
Islet transplantation is a therapeutic option to replace β-cell mass lost during type 1 or type 3c diabetes. Innate immune responses, particularly the instant blood-mediated inflammatory reaction and activation of monocytes, play a major role in the loss of transplanted islet tissue. In this study, we aimed to investigate the inhibition of toll-like receptor 4 (TLR4) on innate inflammatory responses. We first demonstrate a significant loss of graft function shortly after transplant through the assessment of miR-375 and miR-200c in plasma as biomarkers. Using in vitro models, we investigate how targeting TLR4 mitigates islet damage and immune cell activation during the peritransplant period. The results of this study support the application of TAK-242 as a therapeutic agent to reduce inflammatory and innate immune responses to islets immediately following transplantation into the hepatic portal vein. Therefore, TLR4 may serve as a target to improve islet transplant outcomes in the future.
Collapse
Affiliation(s)
- Jordan Mattke
- Institute of Biomedical Studies, Baylor University, Waco, TX 76706, USA; (J.M.); (R.R.K.)
| | - Carly M. Darden
- Annette C. and Harold C. Simmons Transplant Institute, Baylor University Medical Center, Dallas, TX 75204, USA; (C.M.D.); (J.K.)
| | - Srividya Vasu
- Islet Cell Laboratory, Baylor Scott and White Research Institute, Dallas, TX 75204, USA; (S.V.); (M.C.L.)
| | - Michael C. Lawrence
- Islet Cell Laboratory, Baylor Scott and White Research Institute, Dallas, TX 75204, USA; (S.V.); (M.C.L.)
| | - Jeffrey Kirkland
- Annette C. and Harold C. Simmons Transplant Institute, Baylor University Medical Center, Dallas, TX 75204, USA; (C.M.D.); (J.K.)
| | - Robert R. Kane
- Institute of Biomedical Studies, Baylor University, Waco, TX 76706, USA; (J.M.); (R.R.K.)
| | - Bashoo Naziruddin
- Annette C. and Harold C. Simmons Transplant Institute, Baylor University Medical Center, Dallas, TX 75204, USA; (C.M.D.); (J.K.)
| |
Collapse
|
4
|
Duan K, Liu J, Zhang J, Chu T, Liu H, Lou F, Liu Z, Gao B, Wei S, Wei F. Advancements in innate immune regulation strategies in islet transplantation. Front Immunol 2024; 14:1341314. [PMID: 38288129 PMCID: PMC10823010 DOI: 10.3389/fimmu.2023.1341314] [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: 11/20/2023] [Accepted: 12/28/2023] [Indexed: 01/31/2024] Open
Abstract
As a newly emerging organ transplantation technique, islet transplantation has shown the advantages of minimal trauma and high safety since it was first carried out. The proposal of the Edmonton protocol, which has been widely applied, was a breakthrough in this method. However, direct contact between islets and portal vein blood will cause a robust innate immune response leading to massive apoptosis of the graft, and macrophages play an essential role in the innate immune response. Therefore, therapeutic strategies targeting macrophages in the innate immune response have become a popular research topic in recent years. This paper will summarize and analyze recent research on strategies for regulating innate immunity, primarily focusing on macrophages, in the field of islet transplantation, including drug therapy, optimization of islet preparation process, islet engineering and Mesenchymal stem cells cotransplantation. We also expounded the heterogeneity, plasticity and activation mechanism of macrophages in islet transplantation, providing a theoretical basis for further research.
Collapse
Affiliation(s)
- Kehang Duan
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Jiao Liu
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Jian Zhang
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Tongjia Chu
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Huan Liu
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Fengxiang Lou
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Ziyu Liu
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Bing Gao
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Shixiong Wei
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Feng Wei
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
5
|
Karimi A, Yaghobi R, Roozbeh J, Rahimi Z, Afshari A, Akbarpoor Z, Heidari M. Study the mRNA level of IL-27/IL-27R pathway molecules in kidney transplant rejection. Arch Ital Urol Androl 2023; 95:11691. [PMID: 38193229 DOI: 10.4081/aiua.2023.11691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 10/07/2023] [Indexed: 01/10/2024] Open
Abstract
BACKGROUND Renal transplantation stands as the sole remedy for individuals afflicted with end-stage renal diseases, and safeguarding them from transplant rejection represents a vital, life-preserving endeavor posttransplantation. In this context, the impact of cytokines, notably IL-27, assumes a critical role in managing immune responses aimed at countering rejection. Consequently, this investigation endeavors to explore the precise function of IL-27 and its associated cytokines in the context of kidney transplant rejection. METHODS The study involved the acquisition of blood samples from a cohort of participants, consisting of 61 individuals who had undergone kidney transplantation (comprising 32 nonrejected patients and 29 rejected patients), and 33 healthy controls. The expression levels of specific genes were examined using SYBR Green Real-time PCR. Additionally, the evaluation encompassed the estimation of the ROC curve, the assessment of the relationship between certain blood factors, and the construction of protein-protein interaction networks for the genes under investigation. RESULTS Significant statistical differences in gene expression levels were observed between the rejected group and healthy controls, encompassing all the genes examined, except for TLR3 and TLR4 genes. Moreover, the analysis of the Area Under the Curve (AUC) revealed that IL-27, IL-27R, TNF-α, and TLR4 exhibited greater significance in discriminating between the two patient groups. These findings highlight the potential importance of IL-27, IL-27R, TNF-α, and TLR4 as key factors for distinguishing between individuals in the rejected group and those in the healthy control group. CONCLUSIONS In the context of kidney rejections occurring within the specific timeframe of 2 weeks to 2 months post-transplantation, it is crucial to emphasize the significance of cytokines mRNA level, including IL-27, IL-27R, TNF-α, and TLR4, in elucidating and discerning the diverse immune system responses. The comprehensive examination of these cytokines' mRNA level assumes considerable importance in understanding the intricate mechanisms underlying kidney rejection processes during this critical period.
Collapse
Affiliation(s)
- Aftab Karimi
- Zarghan branch, Islamic Azad University, Zarghan.
| | - Ramin Yaghobi
- Shiraz Transplant Research Center, Shiraz University of Medical Sciences, Shiraz.
| | - Jamshid Roozbeh
- Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz.
| | - Zahra Rahimi
- Zarghan branch, Islamic Azad University, Zarghan.
| | - Afsoon Afshari
- Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz.
| | | | - Mojdeh Heidari
- Shiraz Transplant Research Center, Shiraz University of Medical Sciences, Shiraz.
| |
Collapse
|
6
|
Behl T, Sharma E, Sehgal A, Kaur I, Kumar A, Arora R, Pal G, Kakkar M, Kumar R, Bungau S. Expatiating the molecular approaches of HMGB1 in diabetes mellitus: Highlighting signalling pathways via RAGE and TLRs. Mol Biol Rep 2021; 48:1869-1881. [PMID: 33479829 DOI: 10.1007/s11033-020-06130-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 12/24/2020] [Indexed: 12/16/2022]
Abstract
Diabetes mellitus (DM) has become one of the major healthcare challenges worldwide in the recent times and inflammation being one of its key pathogenic process/mechanism affect several body parts including the peripheral and central nervous system. High-mobility group box 1 (HMGB1) is one of the major non-histone proteins that plays a key role in triggering the inflammatory response. Upon its release into the extracellular milieu, HMGB1 acts as an "alarmin" for the immune system to initiate tissue repair as a component of the host defense system. Furthermore, HMGB1 along with its downstream receptors like Toll-like receptors (TLRs) and receptors for advanced glycation end products (RAGE) serve as the suitable target for DM. The forthcoming research in the field of diabetes would potentially focus on the development of alternative approaches to target the centre of inflammation that is primarily mediated by HMGB1 to improve diabetic-related complications. This review covers the therapeutic actions of HMGB1 protein, which acts by activating the RAGE and TLR molecules to constitute a functional tripod system, in turn activating NF-κB pathway that contributes to the production of mediators for pro-inflammatory cytokines associated with DM. The interaction between TLR2 and TLR4 with ligands present in the host and the activation of RAGE stimulates various immune and metabolic responses that contribute to diabetes. This review emphasizes to elucidate the role of HMGB1 in the initiation and progression of DM and control over the inflammatory tripod as a promising therapeutic approach in the management of DM.
Collapse
Affiliation(s)
- Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Eshita Sharma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Ishnoor Kaur
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Arun Kumar
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Rashmi Arora
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Giridhari Pal
- Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India
| | - Munish Kakkar
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Ravinder Kumar
- Cardiovascular Research Institute, Icahn School of Medicine, New York, USA
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
7
|
Yehualashet AS. Toll-like Receptors as a Potential Drug Target for Diabetes Mellitus and Diabetes-associated Complications. Diabetes Metab Syndr Obes 2020; 13:4763-4777. [PMID: 33311992 PMCID: PMC7724365 DOI: 10.2147/dmso.s274844] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 11/03/2020] [Indexed: 12/15/2022] Open
Abstract
Diabetes mellitus (DM) is a chronic endocrine disease distinguished by hyperglycemia due to disturbance in carbohydrate or lipid metabolism or insulin function. To date, diabetes, and its complications, is established as a global cause of morbidity and mortality. The intended aim during the management of diabetes is to maintain blood glucose close to normal because the majority of patients have poor control of their elevated blood glucose and are highly prone to severe macrovascular and microvascular complications. To decrease the burden of the disease and its complications, scientists from various disciplines are working intensively to identify novel and promising drug targets for diabetes and its complications. Increased and ongoing investigations on mechanisms relating to diabetes and associated complications could potentially consider inflammatory cascades as a promising component of the strategy in the prevention and control of diabetes and its complications. The potential of targeting mediators of inflammation like toll-like receptors (TLRs) are part of current investigation by the scientific community. Hence, the aim of the present review is to discuss the role of TLRs as a potential drug target for diabetes and diabetes associated complications.
Collapse
Affiliation(s)
- Awgichew Shewasinad Yehualashet
- Pharmacology and Toxicology Unit, Department of Pharmacy, College of Health Sciences, Debre Berhan University, Debre Berhan, Ethiopia
- Correspondence: Awgichew Shewasinad Yehualashet Pharmacology and Toxicology Unit, Department of Pharmacy, College of Health Sciences, Debre Berhan University, Debre Berhan, EthiopiaTel +251935450290 Email
| |
Collapse
|
8
|
A global perspective on the crosstalk between saturated fatty acids and Toll-like receptor 4 in the etiology of inflammation and insulin resistance. Prog Lipid Res 2019; 77:101020. [PMID: 31870728 DOI: 10.1016/j.plipres.2019.101020] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 11/15/2019] [Accepted: 11/25/2019] [Indexed: 12/16/2022]
Abstract
Obesity is featured by chronic systemic low-grade inflammation that eventually contributes to the development of insulin resistance. Toll-like receptor 4 (TLR4) is an important mediator that triggers the innate immune response by activating inflammatory signaling cascades. Human, animal and cell culture studies identified saturated fatty acids (SFAs), the dominant non-esterified fatty acid (NEFA) in the circulation of obese subjects, as non-microbial agonists that trigger the inflammatory response via activating TLR4 signaling, which acts as an important causative link between fatty acid overload, chronic low-grade inflammation and the related metabolic aberrations. The interaction between SFAs and TLR4 may be modulated through the myeloid differentiation primary response gene 88-dependent and independent signaling pathway. Greater understanding of the crosstalk between dietary SFAs and TLR4 signaling in the pathogenesis of metabolic imbalance may facilitate the design of a more efficient pharmacological strategy to alleviate the risk of developing chronic diseases elicited in part by fatty acid overload. The current review discusses recent advances in the impact of crosstalk between SFAs and TLR4 on inflammation and insulin resistance in multiple cell types, tissues and organs in the context of metabolic dysregulation.
Collapse
|
9
|
Lee T, Yun S, Jeong JH, Jung TW. Asprosin impairs insulin secretion in response to glucose and viability through TLR4/JNK-mediated inflammation. Mol Cell Endocrinol 2019; 486:96-104. [PMID: 30853600 DOI: 10.1016/j.mce.2019.03.001] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 03/02/2019] [Accepted: 03/04/2019] [Indexed: 01/02/2023]
Abstract
Severe inflammation in the islets is observed in obese patients with type 2 diabetes. Inflammation in the islets is caused by obesity-induced serum free fatty acids. Asprosin is a fasting-induced adipokine, which contributes to hepatic glucose production. However, the effects of asprosin on inflammation and cellular dysfunction in pancreatic β-cells remain to be elucidated. Here, we demonstrated that treatment of mouse insulinoma MIN6 cells and human primary islets containing β-cells with palmitate increased asprosin expression and secretion. Treatment of MIN6 cells and human primary islets with palmitate increased phosphorylation of the inflammatory marker nuclear factor-kappa B (NFκB) and the release of pro-inflammatory cytokines including TNF and MCP-1 and decreased glucose-stimulated insulin secretion and cell viability. However, siRNA-mediated suppression of asprosin reversed these changes. Recombinant asprosin treatment of MIN6 cells and human primary islets augmented the inflammation response, cellular dysfunction, and apoptosis in a dose-dependent manner. Asprosin induced toll-like receptor (TLR) 4 expression and JNK phosphorylation. siRNA for TLR4 or JNK mitigated the effects of asprosin on inflammation and cellular dysfunction. These results suggest that palmitate-derived asprosin secretion from β-cells results in their inflammation and dysfunction through a TLR4/JNK-mediated pathway. This report suggests asprosin as a novel therapeutic target for the treatment of type 2 diabetes through preservation of β-cell function.
Collapse
Affiliation(s)
- Taeseung Lee
- Department of Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, South Korea; Department of Surgery, Seoul National University College of Medicine, Seoul, South Korea
| | - Subin Yun
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, South Korea
| | - Tae Woo Jung
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, South Korea.
| |
Collapse
|
10
|
Kim DS, Song L, Wang J, Wu H, Gou W, Cui W, Kim JS, Wang H. Carbon Monoxide Inhibits Islet Apoptosis via Induction of Autophagy. Antioxid Redox Signal 2018; 28:1309-1322. [PMID: 28826228 PMCID: PMC5905947 DOI: 10.1089/ars.2016.6979] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
AIM Carbon monoxide (CO) functions as a therapeutic molecule in various disease models because of its anti-inflammatory and antiapoptotic properties. We investigated the capacity of CO to reduce hypoxia-induced islet cell death and dysfunction in human and mouse models. RESULTS Culturing islets in CO-saturated medium protected them from hypoxia-induced apoptosis and preserved β cell function by suppressing expression of proapoptotic (Bim, PARP, Cas-3), proinflammatory (TNF-α), and endoplasmic reticulum (ER) stress (glucose-regulated protein 94, grp94, CHOP) proteins. The prosurvival effects of CO on islets were attenuated when autophagy was blocked by specific inhibitors or when either ATG7 or ATG16L1, two essential factors for autophagy, was downregulated by siRNA. In vivo, CO exposure reduced both inflammation and cell death in grafts immediately after transplantation, and enhanced long-term graft survival of CO-treated human and mouse islet grafts in streptozotocin-induced diabetic non-obese diabetic severe combined immunodeficiency (NOD-SCID) or C57BL/6 recipients. INNOVATION These findings underline that pretreatment with CO protects islets from hypoxia and stress-induced cell death via upregulation of ATG16L1-mediated autophagy. CONCLUSION Our results suggested that CO exposure may provide an effective means to enhance survival of grafts in clinical islet cell transplantation, and may be beneficial in other diseases in which inflammation and cell death pose impediments to achieving optimal therapeutic effects. Antioxid. Redox Signal. 28, 1309-1322.
Collapse
Affiliation(s)
- Do-Sung Kim
- 1 Department of Surgery, Medical University of South Carolina , Charleston, South Carolina
| | - Lili Song
- 1 Department of Surgery, Medical University of South Carolina , Charleston, South Carolina
| | - Jingjing Wang
- 1 Department of Surgery, Medical University of South Carolina , Charleston, South Carolina
| | - Hongju Wu
- 2 Department of Medicine, Tulane University , New Orleans, Louisiana
| | - Wenyu Gou
- 1 Department of Surgery, Medical University of South Carolina , Charleston, South Carolina
| | - Wanxing Cui
- 3 Medstar Georgetown University Hospital , Washington DC
| | - Jae-Sung Kim
- 4 Department of Surgery, University of Florida , Gainesville, Florida
| | - Hongjun Wang
- 1 Department of Surgery, Medical University of South Carolina , Charleston, South Carolina
| |
Collapse
|
11
|
Friedman O, Carmel N, Sela M, Abu Jabal A, Inbal A, Ben Hamou M, Krelin Y, Gur E, Shani N. Immunological and inflammatory mapping of vascularized composite allograft rejection processes in a rat model. PLoS One 2017; 12:e0181507. [PMID: 28746417 PMCID: PMC5528841 DOI: 10.1371/journal.pone.0181507] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 07/03/2017] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Hand and face vascularized composite allotransplantation (VCA) is an evolving and challenging field with great opportunities. During VCA, massive surgical damage is inflicted on both donor and recipient tissues, which may contribute to the high VCA rejection rates. To segregate between the damage-induced and rejection phase of post-VCA responses, we compared responses occurring up to 5 days following syngeneic versus allogeneic vascularized groin flap transplantations, culminating in transplant acceptance or rejection, respectively. METHODS The immune response elicited upon transplantation of a syngeneic versus allogeneic vascularized groin flap was compared at Post-operative days 2 or 5 by histology, immunohistochemistry and by broad-scope gene and protein analyses using quantitative real-time PCR and Multiplex respectively. RESULTS Immune cell infiltration began at the donor-recipient interface and paralleled expression of a large group of wound healing-associated genes in both allografts and syngrafts. By day 5 post-transplantation, cell infiltration spread over the entire allograft but remained confined to the wound site in the syngraft. This shift correlated with upregulation of IL-18, INFg, CXCL9, 10 and 11, CCL2, CCL5, CX3CL1 and IL-10 in the allograft only, suggesting their role in the induction of the anti-alloantigen adaptive immune response. CONCLUSIONS High resemblance between the cues governing VCA and solid organ rejection was observed. Despite this high resemblance we describe also, for the first time, a damage induced inflammatory component in VCA rejection as immune cell infiltration into the graft initiated at the surgical damage site spreading to the entire allograft only at late stage rejection. We speculate that the highly inflammatory setting created by the unique surgical damage during VCA may enhance acute allograft rejection.
Collapse
Affiliation(s)
- Or Friedman
- The Plastic Reconstructive Surgery Department, Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Narin Carmel
- The Plastic Reconstructive Surgery Department, Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Meirav Sela
- The Plastic Reconstructive Surgery Department, Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ameen Abu Jabal
- The Plastic Reconstructive Surgery Department, Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Amir Inbal
- The Plastic Reconstructive Surgery Department, Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Moshe Ben Hamou
- The Plastic Reconstructive Surgery Department, Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yakov Krelin
- The Plastic Reconstructive Surgery Department, Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eyal Gur
- The Plastic Reconstructive Surgery Department, Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nir Shani
- The Plastic Reconstructive Surgery Department, Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- * E-mail:
| |
Collapse
|
12
|
Wei X, Liu Q, Gao Y, Yang J, Wang B, Yang G, Zhang S, Zhou H. Two epitopes responsible for the catalytic activity of heme oxygenase-1 identified by phage display. FEBS Open Bio 2017; 7:719-726. [PMID: 28469983 PMCID: PMC5407895 DOI: 10.1002/2211-5463.12217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 03/07/2017] [Accepted: 03/09/2017] [Indexed: 12/16/2022] Open
Abstract
Heme oxygenase‐1 (HO‐1) catalyzes the oxidative degradation of heme. The catalytic mechanism of the HO‐1 reaction has been determined gradually by studies of its crystal structure and HO‐1 mutants. However, the neutralizing epitopes responsible for HO‐1 activity remain elusive. Screening of a phage display library revealed four epitopes that could interact with the polyclonal antibody prepared by immunizing rabbits with the purified HO‐1 protein. Two of these four epitopes are responsible for HO‐1 catalytic activity because their antibodies were able to neutralize HO‐1 activity. The results of the present study shed further light on the molecular character of HO‐1.
Collapse
Affiliation(s)
- Xuran Wei
- Beijing Key Laboratory of Blood Safety and Supply Technologies Beijing Institute of Transfusion Medicine China
| | - Qingjun Liu
- Beijing Key Laboratory of Blood Safety and Supply Technologies Beijing Institute of Transfusion Medicine China
| | - Yaping Gao
- Institute of Basic Medical Sciences Academy of Military Medical Sciences Beijing China
| | - Jun Yang
- Beijing Key Laboratory of Blood Safety and Supply Technologies Beijing Institute of Transfusion Medicine China
| | - Bo Wang
- Beijing Key Laboratory of Blood Safety and Supply Technologies Beijing Institute of Transfusion Medicine China
| | - Guang Yang
- Institute of Basic Medical Sciences Academy of Military Medical Sciences Beijing China
| | - Shihui Zhang
- Beijing Key Laboratory of Blood Safety and Supply Technologies Beijing Institute of Transfusion Medicine China
| | - Hong Zhou
- Beijing Key Laboratory of Blood Safety and Supply Technologies Beijing Institute of Transfusion Medicine China
| |
Collapse
|
13
|
Kennelly KP, Holmes TM, Wallace DM, O'Farrelly C, Keegan DJ. Early Subretinal Allograft Rejection Is Characterized by Innate Immune Activity. Cell Transplant 2017; 26:983-1000. [PMID: 28105976 DOI: 10.3727/096368917x694697] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Successful subretinal transplantation is limited by considerable early graft loss despite pharmacological suppression of adaptive immunity. We postulated that early innate immune activity is a dominant factor in determining graft survival and chose a nonimmunosuppressed mouse model of retinal pigment epithelial (RPE) cell transplantation to explore this. Expression of almost all measured cytokines by DH01 RPE cells increased significantly following graft preparation, and the neutrophil chemoattractant KC/GRO/CINC was most significantly increased. Subretinal allografts of DH01 cells (C57BL/10 origin) into healthy, nonimmunosuppressed C57BL/6 murine eyes were harvested and fixed at 1, 3, 7, and 28 days postoperatively and subsequently cryosectioned and stained. Graft cells were detected using SV40 large T antigen (SV40T) immunolabeling and apoptosis/necrosis by terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL). Sections were also immunolabeled for macrophage (CD11b and F4/80), neutrophil (Gr1 Ly-6G), and T-lymphocyte (CD3-ɛ) infiltration. Images captured with an Olympus FV1000 confocal microscope were analyzed using the Imaris software. The proportion of the subretinal bolus comprising graft cells (SV40T+) was significantly (p < 0.001) reduced between postoperative day (POD) 3 (90 ± 4%) and POD 7 (20 ± 7%). CD11b+, F4/80+, and Gr1 Ly-6G+ cells increased significantly (p < 0.05) from POD 1 and predominated over SV40T+ cells by POD 7. Colabeling confocal microscopic analysis demonstrated graft engulfment by neutrophils and macrophages at POD 7, and reconstruction of z-stacked confocal images confirmed SV40T inside Gr1 Ly-6G+ cells. Expression of CD3-ɛ was low and did not differ significantly between time points. By POD 28, no graft cells were detectable and few inflammatory cells remained. These studies reveal, for the first time, a critical role for innate immune mechanisms early in subretinal graft rejection. The future success of subretinal transplantation will require more emphasis on techniques to limit innate immune-mediated graft loss, rather than focusing exclusively on suppression of the adaptive immune response.
Collapse
|
14
|
Rehman K, Akash MSH. Mechanisms of inflammatory responses and development of insulin resistance: how are they interlinked? J Biomed Sci 2016; 23:87. [PMID: 27912756 PMCID: PMC5135788 DOI: 10.1186/s12929-016-0303-y] [Citation(s) in RCA: 370] [Impact Index Per Article: 41.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 11/24/2016] [Indexed: 02/06/2023] Open
Abstract
Background Insulin resistance (IR) is one of the major hallmark for pathogenesis and etiology of type 2 diabetes mellitus (T2DM). IR is directly interlinked with various inflammatory responses which play crucial role in the development of IR. Inflammatory responses play a crucial role in the pathogenesis and development of IR which is one of the main causative factor for the etiology of T2DM. Methods A comprehensive online English literature was searched using various electronic search databases. Different search terms for pathogenesis of IR, role of various inflammatory responses were used and an advanced search was conducted by combining all the search fields in abstracts, keywords, and titles. Results We summarized the data from the searched articles and found that inflammatory responses activate the production of various pro-inflammatory mediators notably cytokines, chemokines and adipocytokines through the involvement of various transcriptional mediated molecular pathways, oxidative and metabolic stress. Overnutrition is one of the major causative factor that contributes to induce the state of low-grade inflammation due to which accumulation of elevated levels of glucose and/or lipids in blood stream occur that leads to the activation of various transcriptional mediated molecular and metabolic pathways. This results in the induction of various pro-inflammatory mediators that are decisively involved to provoke the pathogenesis of tissue-specific IR by interfering with insulin signaling pathways. Once IR is developed, it increases oxidative stress in β-cells of pancreatic islets and peripheral tissues which impairs insulin secretion, and insulin sensitivity in β-cells of pancreatic islets and peripheral tissues, respectively. Moreover, we also summarized the data regarding various treatment strategies of inflammatory responses-induced IR. Conclusions In this article, we have briefly described that how pro-inflammatory mediators, oxidative stress, transcriptional mediated molecular and metabolic pathways are involved in the pathogenesis of tissues-specific IR. Moreover, based on recent investigations, we have also described that to counterfeit these inflammatory responses is one of the best treatment strategy to prevent the pathogenesis of IR through ameliorating the incidences of inflammatory responses.
Collapse
Affiliation(s)
- Kanwal Rehman
- Institute of Pharmacy, Physiology and Pharmacology, University of Agriculture, Faisalabad, Pakistan
| | | |
Collapse
|
15
|
Cheng Y, Xiong J, Chen Q, Xia J, Zhang Y, Yang X, Tao K, Zhang S, He S. Hypoxia/reoxygenation-induced HMGB1 translocation and release promotes islet proinflammatory cytokine production and early islet graft failure through TLRs signaling. Biochim Biophys Acta Mol Basis Dis 2016; 1863:354-364. [PMID: 27838489 DOI: 10.1016/j.bbadis.2016.11.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 10/20/2016] [Accepted: 11/08/2016] [Indexed: 02/05/2023]
Abstract
High-mobility group box 1 (HMGB1) translocation and release, which is involved in several tissue types of ischemia-reperfusion injuries, activate innate immunity by inducing proinflammatory cytokine production through its interaction with toll-like receptors (TLRs). Our objective was to determine the role of HMGB1 and the degree of activation of TLR-related signal transduction pathways in hypoxia/reoxygenation (H/R)-induced proinflammatory cytokine production and intra-islet graft inflammation. After islets are exposed to hypoxia-reoxygenation for 24h, TLR2/4 expression and TLR-mediated signaling was up-regulated in islets, and HMGB1 was translocated from the nucleus to the cytoplasm and released to the extracellular space. With H/R exposure, proinflammatory cytokine production (IL-1β and TNF-α) and islet injury were significantly increased, and these effects depend on TLR2/4 signaling pathways. Exogenous HMGB1 also induces islet inflammation and increases the phosphorylation of STAT3, p38 and IκBα in wild-type islets. TLR2 deficiency in TLR2-KO islets resulted in the inhibition of IL-1β production and STAT3/p38 phosphorylation after HMGB1 exposure. TLR4 deficiency in TLR4-KO islets resulted in the inhibition of TNF-α production and IκBα phosphorylation after HMGB1 exposure. Pre-incubation of the STAT3, p38, or NF-κB inhibitors significantly inhibited HMGB1-induced IL-1β or TNF-α production in islets, but the effect of HMGB1 or H/R-induced islet injury was not counteracted by a separate treatment of the STAT3 inhibitor, p38 inhibitor, or NF-κB inhibitors. HMGB1 inhibition by ethyl pyruvate or blockade by neutralizing antibodies significantly decreased the phosphorylation of STAT3, p38 and IκBα, the production of IL-1β and TNF-α, and the islet injury in wild-type islets after exposure to H/R and significantly improved early islet graft failure. Thus, our results suggest that HMGB1 released from H/R induced islets works in an autocrine manner to up-regulate STAT or p38 and augment IL-1β production via TLR2, and up-regulate NF-κB and augment TNF-α production via TLR4 in intra-islet, which are associated with H/R-induced islet injury and early graft failure.
Collapse
Affiliation(s)
- Yao Cheng
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, P.R. China; Department of Hepatobiliary Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Junjie Xiong
- Department of Pancreatic Surgery, West China Hospital, Sichuan University, Chengdu 610041, P.R. China
| | - Quan Chen
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Jie Xia
- Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yi Zhang
- Department of Pancreatic Surgery, West China Hospital, Sichuan University, Chengdu 610041, P.R. China
| | - Xiaoyan Yang
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Kun Tao
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Shuang Zhang
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Sirong He
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, P.R. China.
| |
Collapse
|
16
|
Dong H, Zhang Y, Song L, Kim DS, Wu H, Yang L, Li S, Morgan KA, Adams DB, Wang H. Cell-Permeable Peptide Blocks TLR4 Signaling and Improves Islet Allograft Survival. Cell Transplant 2016; 25:1319-29. [PMID: 26771084 DOI: 10.3727/096368916x690449] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Toll-like receptor 4 (TLR4) activation in pancreatic β cells activates aberrant islet graft cellular pathways and contributes to immune rejection in allogeneic islet transplantation. As an approach to overcoming this problem, we determined the capacity of a 33-amino acid peptide consisting of a protein transduction domain (PTD) from the Hph-1 virus and a fragment of the intracellular domain of TLR4 from the C3H mice (PTD-dnTLR4) to block TLR4 signaling and improve allogeneic islet survival in vitro and after transplantation. The efficacy of PTD-dnTLR4 in blocking TLR4 signaling was assessed in the Raw264.7 macrophage line, in the islets, and the βTC3 cell line. In Raw264.7 cells, preculture with the peptide reduced LPS-induced NF-κB activation and production of proinflammatory cytokines (IL-1β, TNF-α, iNOS, and IL-6). In islets and β cells, preincubation with PTD-dnTLR4 suppressed LPS-induced TNF-α expression via inhibition of NF-κB activation and protected them from stress-induced cell death. In vivo, preincubation of BALB/c (H-2(d)) islets with PTD-dnTLR4 resulted in significantly longer survival than control islets in a streptozotocin-induced diabetes model (two of seven grafts survived long term >100 days). PTD-dnTLR4-treated grafts exhibited reduced expression of TNF-α and iNOS and reduced macrophage infiltration posttransplant. The data indicate that PTD-dnTLR4 blocked TLR4 signaling in both macrophages and β cells, and prolonged allograft survival at least in part by suppressing inflammation and macrophage infiltration. This strategy for blocking TLR4 activity has potential utilization in the treatment of diseases where excessive TLR4 activation contributes to the pathologic cellular pathways such as islet transplantation.
Collapse
Affiliation(s)
- Huansheng Dong
- Department of Surgery, Medical University of South Carolina, Charleston, SC, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Giovannoni L, Muller YD, Lacotte S, Parnaud G, Borot S, Meier RP, Lavallard V, Bédat B, Toso C, Daubeuf B, Elson G, Shang L, Morel P, Kosco-Vilbois M, Bosco D, Berney T. Enhancement of Islet Engraftment and Achievement of Long-Term Islet Allograft Survival by Toll-Like Receptor 4 Blockade. Transplantation 2015; 99:29-35. [DOI: 10.1097/tp.0000000000000468] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
18
|
Cao Y, Bao S, Yang W, Zhang J, Li L, Shan Z, Teng W. Epigallocatechin gallate prevents inflammation by reducing macrophage infiltration and inhibiting tumor necrosis factor-α signaling in the pancreas of rats on a high-fat diet. Nutr Res 2014; 34:1066-74. [PMID: 25453543 DOI: 10.1016/j.nutres.2014.10.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 09/30/2014] [Accepted: 10/10/2014] [Indexed: 01/26/2023]
|
19
|
Wali JA, Gurzov EN, Fynch S, Elkerbout L, Kay TW, Masters SL, Thomas HE. Activation of the NLRP3 inflammasome complex is not required for stress-induced death of pancreatic islets. PLoS One 2014; 9:e113128. [PMID: 25405767 PMCID: PMC4236141 DOI: 10.1371/journal.pone.0113128] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 10/20/2014] [Indexed: 01/09/2023] Open
Abstract
Loss of pancreatic beta cells is a feature of type-2 diabetes. High glucose concentrations induce endoplasmic reticulum (ER) and oxidative stress-mediated apoptosis of islet cells invitro. ER stress, oxidative stress and high glucose concentrations may also activate the NLRP3 inflammasome leading to interleukin (IL)-1β production and caspase-1 dependent pyroptosis. However, whether IL-1β or intrinsic NLRP3 inflammasome activation contributes to beta cell death is controversial. This possibility was examined in mouse islets. Exposure of islets lacking functional NLRP3 or caspase-1 to H2O2, rotenone or thapsigargin induced similar cell death as in wild-type islets. This suggests that oxidative or ER stress do not cause inflammasome-mediated cell death. Similarly, deficiency of NLRP3 inflammasome components did not provide any protection from glucose, ribose or gluco-lipotoxicity. Finally, genetic activation of NLRP3 specifically in beta cells did not increase IL-1β production or cell death, even in response to glucolipotoxicity. Overall, our results show that glucose-, ER stress- or oxidative stress-induced cell death in islet cells is not dependent on intrinsic activation of the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Jibran A. Wali
- St Vincent’s Institute, Fitzroy, Victoria, Australia
- The University of Melbourne Department of Medicine, St. Vincent’s Hospital, Fitzroy, Victoria, Australia
| | - Esteban N. Gurzov
- St Vincent’s Institute, Fitzroy, Victoria, Australia
- The University of Melbourne Department of Medicine, St. Vincent’s Hospital, Fitzroy, Victoria, Australia
| | - Stacey Fynch
- St Vincent’s Institute, Fitzroy, Victoria, Australia
| | | | - Thomas W. Kay
- St Vincent’s Institute, Fitzroy, Victoria, Australia
- The University of Melbourne Department of Medicine, St. Vincent’s Hospital, Fitzroy, Victoria, Australia
| | - Seth L. Masters
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Helen E. Thomas
- St Vincent’s Institute, Fitzroy, Victoria, Australia
- The University of Melbourne Department of Medicine, St. Vincent’s Hospital, Fitzroy, Victoria, Australia
- * E-mail:
| |
Collapse
|
20
|
Vivot K, Langlois A, Bietiger W, Dal S, Seyfritz E, Pinget M, Jeandidier N, Maillard E, Gies JP, Sigrist S. Pro-inflammatory and pro-oxidant status of pancreatic islet in vitro is controlled by TLR-4 and HO-1 pathways. PLoS One 2014; 9:e107656. [PMID: 25343247 PMCID: PMC4208733 DOI: 10.1371/journal.pone.0107656] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 08/13/2014] [Indexed: 01/09/2023] Open
Abstract
Since their isolation until implantation, pancreatic islets suffer a major stress leading to the activation of inflammatory reactions. The maintenance of controlled inflammation is essential to preserve survival and function of the graft. Identification and targeting of pathway(s) implicated in post-transplant detrimental inflammatory events, is mandatory to improve islet transplantation success. We sought to characterize the expression of the pro-inflammatory and pro-oxidant mediators during islet culture with a focus on Heme oxygenase (HO-1) and Toll-like receptors-4 signaling pathways. Rat pancreatic islets were isolated and pro-inflammatory and pro-oxidant status were evaluated after 0, 12, 24 and 48 hours of culture through TLR-4, HO-1 and cyclooxygenase-2 (COX-2) expression, CCL-2 and IL-6 secretion, ROS (Reactive Oxygen Species) production (Dihydroethidine staining, DHE) and macrophages migration. To identify the therapeutic target, TLR4 inhibition (CLI-095) and HO-1 activation (cobalt protoporphyrin,CoPP) was performed. Activation of NFκB signaling pathway was also investigated. After isolation and during culture, pancreatic islet exhibited a proinflammatory and prooxidant status (increase levels of TLR-4, COX-2, CCL-2, IL-6, and ROS). Activation of HO-1 or inhibition of TLR-4 decreased inflammatory status and oxidative stress of islets. Moreover, the overexpression of HO-1 induced NFκB phosphorylation while the inhibition of TLR-4 had no effect NFκB activation. Finally, inhibition of pro-inflammatory pathway induced a reduction of macrophages migration. These data demonstrated that the TLR-4 signaling pathway is implicated in early inflammatory events leading to a pro-inflammatory and pro-oxidant status of islets in vitro. Moreover, these results provide the mechanism whereby the benefits of HO-1 target in TLR-4 signaling pathway. HO-1 could be then an interesting target to protect islets before transplantation.
Collapse
Affiliation(s)
- Kevin Vivot
- DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle, Strasbourg, France
| | - Allan Langlois
- DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle, Strasbourg, France
| | - William Bietiger
- DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle, Strasbourg, France
| | - Stéphanie Dal
- DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle, Strasbourg, France
| | - Elodie Seyfritz
- DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle, Strasbourg, France
| | - Michel Pinget
- DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle, Strasbourg, France
- Structure d'Endocrinologie, Diabète –Nutrition et Addictologie, Pôle NUDE, Hôpitaux Universitaires de Strasbourg, (HUS), Strasbourg, France
| | - Nathalie Jeandidier
- DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle, Strasbourg, France
- Structure d'Endocrinologie, Diabète –Nutrition et Addictologie, Pôle NUDE, Hôpitaux Universitaires de Strasbourg, (HUS), Strasbourg, France
| | - Elisa Maillard
- DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle, Strasbourg, France
| | - Jean-Pierre Gies
- UMR 7034 CNRS, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France
| | - Séverine Sigrist
- DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle, Strasbourg, France
- * E-mail:
| |
Collapse
|
21
|
Fujisaki N, Nakao A, Osako T, Nishimura T, Yamada T, Kohama K, Sakata H, Ishikawa-Aoyama M, Kotani J. Can carbon monoxide-poisoned victims be organ donors? Med Gas Res 2014; 4:13. [PMID: 25097755 PMCID: PMC4121619 DOI: 10.1186/2045-9912-4-13] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 07/21/2014] [Indexed: 01/02/2023] Open
Abstract
The increasing demand for organ allografts to treat end-stage organ failure has driven changes in traditional donor criteria. Patients who have succumbed to carbon monoxide (CO) poisoning, a common cause of toxicological mortality, are usually rejected as organ donors. To fulfill the increasing demand, selection criteria must be expanded to include CO-poisoned donors. However, the use of allografts exposed to high CO concentrations is still under debate. Basic research and literature review data suggest that patients with brain death caused by CO poisoning should be considered appropriate organ donors. Accepting organs from CO-poisoned victims could increase the number of potential donors and lower the death rate of patients on the waiting lists. This review and reported cases may increase awareness among emergency department physicians, as well as transplant teams, that patients dying of CO exposure may be acceptable organ donors.
Collapse
Affiliation(s)
- Noritomo Fujisaki
- Department of Emergency, Disaster and Critical Care Medicine, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Atsunori Nakao
- Department of Emergency, Disaster and Critical Care Medicine, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Takaaki Osako
- Department of Emergency, Disaster and Critical Care Medicine, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Takeshi Nishimura
- Department of Emergency, Disaster and Critical Care Medicine, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Taihei Yamada
- Department of Emergency, Disaster and Critical Care Medicine, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Keisuke Kohama
- Department of Emergency, Disaster and Critical Care Medicine, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Hiroyuki Sakata
- Department of Emergency, Disaster and Critical Care Medicine, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Michiko Ishikawa-Aoyama
- Department of Emergency, Disaster and Critical Care Medicine, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Joji Kotani
- Department of Emergency, Disaster and Critical Care Medicine, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| |
Collapse
|
22
|
Cucak H, Mayer C, Tonnesen M, Thomsen LH, Grunnet LG, Rosendahl A. Macrophage contact dependent and independent TLR4 mechanisms induce β-cell dysfunction and apoptosis in a mouse model of type 2 diabetes. PLoS One 2014; 9:e90685. [PMID: 24594974 PMCID: PMC3940939 DOI: 10.1371/journal.pone.0090685] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 02/03/2014] [Indexed: 12/21/2022] Open
Abstract
Type 2 diabetes (T2D) is evolving into a global disease and patients have a systemic low-grade inflammation, yet the role of this inflammation is still not established. One plausible mechanism is enhanced expression and activity of the innate immune system. Therefore, we evaluated the expression and the function of the toll-like receptor 4 (TLR4) on pancreatic β-cells in primary mouse islets and on the murine β-cell line MIN6 in the presence or absence of macrophages. Diabetic islets have 40% fewer TLR4 positive β-cells, but twice the number of TLR4 positive macrophages as compared to healthy islets. Healthy and diabetic islets respond to a TLR4 challenge with enhanced production of cytokines (5–10-fold), while the TLR4 negative β-cell line MIN6 fails to produce cytokines. TLR4 stimulation induces β-cell dysfunction in mouse islets, measured as reduced glucose stimulated insulin secretion. Diabetic macrophages from 4-months old mice have acquired a transient enhanced capacity to produce cytokines when stimulated with LPS. Interestingly, this is lost in 6-months old diabetic mice. TLR4 activation alone does not induce apoptosis in islets or MIN-6 cells. In contrast, macrophages mediate TLR4-dependent cell-contact dependent (3-fold) as well as cell-contact independent (2-fold) apoptosis of both islets and MIN-6 cells. Importantly, diabetic macrophages have a significantly enhanced capacity to induce β-cell apoptosis compared to healthy macrophages. Taken together, the TLR4 responsiveness is elevated in the diabetic islets and mainly mediated by newly recruited macrophages. The TLR4 positive macrophages, in both a cell-contact dependent and independent manner, induce apoptosis of β-cells in a TLR4 dependent fashion and TLR4 activation directly induces β-cell dysfunction. Thus, targeting either the TLR4 pathway or the macrophages provides a novel attractive treatment regime for T2D.
Collapse
Affiliation(s)
- Helena Cucak
- Hagedorn Research Institute, Department of Diabetic Complication Biology, Måløv, Denmark
| | - Christopher Mayer
- Hagedorn Research Institute, Department of Diabetic Complication Biology, Måløv, Denmark
| | - Morten Tonnesen
- Hagedorn Research Institute, Department of Diabetic Complication Biology, Måløv, Denmark
| | - Lise Høj Thomsen
- Hagedorn Research Institute, Department of Diabetic Complication Biology, Måløv, Denmark
| | | | - Alexander Rosendahl
- Hagedorn Research Institute, Department of Diabetic Complication Biology, Måløv, Denmark
- * E-mail:
| |
Collapse
|
23
|
Abstract
The sensitive and broadly reactive character of the innate immune system makes it liable to activation by stress factors other than infection. Thermal and metabolic stresses experienced during the transplantation procedure are sufficient to trigger the innate immune response and also augment adaptive immunity in the presence of foreign antigen on the donor organ. The resulting inflammatory and immune reactions combine to form a potent effector response that can lead to graft rejection. Here we examine the evidence that the complement and toll-like receptor systems are central to these pathways of injury and present a formidable barrier to transplantation. We review extensive information about the effector mechanisms that are mediated by these pathways, and bring together what is known about the damage-associated molecular patterns that initiate this sequence of events. Finally, we refer to two ongoing therapeutic trials that are evaluating the validity of these concepts in man.
Collapse
Affiliation(s)
- Conrad A Farrar
- MRC Centre for Transplantation, Division of Transplantation Immunology and Mucosal Biology, King's College London School of Medicine at Guy's, King's College and St. Thomas' Hospitals, London SE1 9RT, United Kingdom
| | | | | |
Collapse
|
24
|
Ro H, Lee EW, Hong JH, Han KH, Yeom HJ, Kim HJ, Kim MG, Jung HS, Oh KH, Park KS, Ahn C, Yang J. Roles of Islet Toll-Like Receptors in Pig to Mouse Islet Xenotransplantation. Cell Transplant 2013; 22:1709-22. [DOI: 10.3727/096368912x657684] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Although innate immunity plays important roles in xenograft rejection, there have been few studies on the role of toll-like receptors (TLRs) in xenotransplantation. Furthermore, most studies focused on the recipient's TLRs. Therefore, we investigated whether TLRs in porcine islets can contribute to islet xenograft rejection. Adult porcine islets were isolated and stimulated by polyinosinic/polycytidylic acid (poly I:C) or lipopolysaccharide (LPS). Both poly I:C and LPS stimulation in porcine islets induced expression of chemokines (RANTES, MCP-1, IP-10, and IL-8), cytokines (IL-6 and type I interferons), and adhesion molecules (VCAM-1 and ICAM-1). Porcine islet supernatants stimulated by TLR agonists induced chemotaxis of human leukocytes. They also induced procoagulant activation (tissue factor and fgl-2). However, TLR stimulation did not influence insulin secretion. When porcine MyD88 was knocked down using shRNA lentivirus, TLR-mediated induction of proinflammatory mediators and procoagulants was attenuated. When LPS was injected to MyD88 or TLR4 knockout mice after porcine islet transplantation, LPS stimulation on donor islets interfered with islet xenograft tolerance induction by anti-CD154 antibodies. Inflammatory cell infiltration and expression of proinflammatory chemokines and cytokines in islet xenografts also increased. In conclusion, TLR activation in porcine islets induced both a proinflammatory and procoagulant response and thereby contributed to xenograft rejection.
Collapse
Affiliation(s)
- Han Ro
- Transplantation Center, Seoul National University Hospital, Seoul, Republic of Korea
- Transplantation Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Eun Won Lee
- Transplantation Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Joo Ho Hong
- Transplantation Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kyu Hyun Han
- Transplantation Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hye-Jung Yeom
- Transplantation Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hwa Jung Kim
- Transplantation Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Myung-Gyu Kim
- Transplantation Center, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hye Seung Jung
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kook-Hwan Oh
- Division of Nephrology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kyong Soo Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Curie Ahn
- Transplantation Center, Seoul National University Hospital, Seoul, Republic of Korea
- Transplantation Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
- Division of Nephrology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jaeseok Yang
- Transplantation Center, Seoul National University Hospital, Seoul, Republic of Korea
- Transplantation Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
25
|
Vasculotoxic and Proinflammatory Effects of Plasma Heme: Cell Signaling and Cytoprotective Responses. ACTA ACUST UNITED AC 2013; 2013. [PMID: 25506596 PMCID: PMC4261193 DOI: 10.1155/2013/831596] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The proinfammatory vasculotoxic effects of intravascular hemolysis are modulated by plasma hemoglobin and heme clearance via the haptoglobin/CD163 system and the hemopexin/CD91 system, respectively, and detoxification through the heme oxygenase/ferritin system. However, sudden or excessive hemolysis can overwhelm these protective systems leading to heme interacting with cells of the vasculature. Heme presents a damage-associated molecular pattern to the innate immune system. Heme is an extracellular inflammatory signaling molecule with strict binding specificity for TLR4 on monocyte/macrophages, endothelial, and other cells. The resulting TLR4 signaling cascade rapidly leads to intracellular oxidative stress and an inflammatory response. Heme also induces a cytoprotective response that includes Nrf2 responsive genes such as heme oxygenase-1, ferritin, haptoglobin, hemopexin, and other antioxidant response genes. It is the balance between the pro-inflammatory/vasculotoxic effects of plasma hemoglobin/heme and the cytoprotective responses that ultimately determines the pathophysiologic outcome in patients.
Collapse
|
26
|
Eldor R, Abel R, Sever D, Sadoun G, Peled A, Sionov R, Melloul D. Inhibition of nuclear factor-κB activation in pancreatic β-cells has a protective effect on allogeneic pancreatic islet graft survival. PLoS One 2013; 8:e56924. [PMID: 23437272 PMCID: PMC3578930 DOI: 10.1371/journal.pone.0056924] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 01/16/2013] [Indexed: 01/22/2023] Open
Abstract
Pancreatic islet transplantation, a treatment for type 1 diabetes, has met significant challenges, as a substantial fraction of the islet mass fails to engraft, partly due to death by apoptosis in the peri- and post-transplantation periods. Previous evidence has suggested that NF-κB activation is involved in cytokine-mediated β-cell apoptosis and regulates the expression of pro-inflammatory and chemokine genes. We therefore sought to explore the effects of β-cell-specific inhibition of NF-κB activation as a means of cytoprotection in an allogeneic model of islet transplantation. To this end, we used islets isolated from the ToI-β transgenic mouse, where NF-κB signalling can specifically and conditionally be inhibited in β-cells by expressing an inducible and non-degradable form of IκBα regulated by the tet-on system. Our results show that β-cell-specific blockade of NF-κB led to a prolonged islet graft survival, with a relative higher preservation of the engrafted endocrine tissue and reduced inflammation. Importantly, a longer delay in allograft rejection was achieved when mice were systemically treated with the proteasome inhibitor, Bortezomib. Our findings emphasize the contribution of NF-κB activation in the allograft rejection process, and suggest an involvement of the CXCL10/IP-10 chemokine. Furthermore, we suggest a potential, readily available therapeutic agent that may temper this process.
Collapse
Affiliation(s)
- Roy Eldor
- Department of Endocrinology, Hadassah University Hospital, Jerusalem, Israel
| | - Roy Abel
- Department of Endocrinology, Hadassah University Hospital, Jerusalem, Israel
| | - Dror Sever
- Department of Endocrinology, Hadassah University Hospital, Jerusalem, Israel
| | - Gad Sadoun
- Department of Endocrinology, Hadassah University Hospital, Jerusalem, Israel
| | - Amnon Peled
- Goldyne Savad Institute of Gene Therapy, Hadassah University Hospital, Jerusalem, Israel
| | - Ronit Sionov
- Department of Biochemistry and Molecular Biology, IMRIC, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Danielle Melloul
- Department of Endocrinology, Hadassah University Hospital, Jerusalem, Israel
- * E-mail:
| |
Collapse
|
27
|
Amano MT, Camara NOS. The immunomodulatory role of carbon monoxide during transplantation. Med Gas Res 2013; 3:1. [PMID: 23295066 PMCID: PMC3582539 DOI: 10.1186/2045-9912-3-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Accepted: 12/31/2012] [Indexed: 01/03/2023] Open
Abstract
The number of organ and tissue transplants has increased worldwide in recent decades. However, graft rejection, infections due to the use of immunosuppressive drugs and a shortage of graft donors remain major concerns. Carbon monoxide (CO) had long been regarded solely as a poisonous gas. Ultimately, physiological studies unveiled the endogenous production of CO, particularly by the heme oxygenase (HO)-1 enzyme, recognizing CO as a beneficial gas when used at therapeutic doses. The protective properties of CO led researchers to develop uses for it, resulting in devices and molecules that can deliver CO in vitro and in vivo. The resulting interest in clinical investigations was immediate. Studies regarding the CO/HO-1 modulation of immune responses and their effects on various immune disorders gave rise to transplantation research, where CO was shown to be essential in the protection against organ rejection in animal models. This review provides a perspective of how CO modulates the immune system to improve transplantation and suggests its use as a therapy in the field.
Collapse
Affiliation(s)
- Mariane Tami Amano
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil.
| | | |
Collapse
|
28
|
|
29
|
Wang H, Wu H, Rocuts F, Gu Z, Bach FH, Otterbein LE. Activation of Peroxisome Proliferator-Activated Receptor γ Prolongs Islet Allograft Survival. Cell Transplant 2012; 21:2111-8. [DOI: 10.3727/096368911x637399] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Exposing donor mice to carbon monoxide (CO) protects transplanted islet allografts from immune rejection after transplantation (referred as the “donor” effect). In an attempt to understand the mechanisms of the donor effect of CO, we found that donor treatment with CO upregulates expression of peroxisome proliferatoractivated receptor γ (PPARγ), a transcriptional regulator, in isolated islets. In this study, we evaluated whether PPARγ contributes to the survival and function of transplanted islets and whether PPARγ mediates the protective effect of CO in a major mismatch islet allogeneic transplantation model. BALB/c (H-2d) islets in which PPARγ activity was induced by its agonists, 15-deoxy-Δ12–14-prostaglandin J2 (15d-PGJ2) or troglitazone were transplanted into C57BL/6 (H-2b) recipients that had been rendered diabetic by streptozotocin (STZ). Blood glucose levels of recipients were monitored to determine the function of transplanted islets. Our data indicated that PPARγ activation in islets led to a high percentage of BALB/c islets survived long-term in C57BL/6 recipients. Activation of PPARγ in the donor suppresses expressions of proinflammatory cytokines including tumor necrosis factor-α (TNF-α) and inducible nitric oxide synthase (iNOS) in transplanted islets. Blocking PPARγ activity by its antagonist, GW9662, abrogated the donor effect of CO in vivo and in vitro. Our data demonstrate that PPARγ plays a critical role in the survival and function of transplanted islets after transplantation in the recipient. The protective effects of CO are at least in part mediated by PPARγ.
Collapse
Affiliation(s)
- Hongjun Wang
- Department of Surgery, Medical University of South Carolina, Charleston, SC, USA
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Hongju Wu
- Department of Obstetrics and Gynecology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Fredy Rocuts
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Zhuoying Gu
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Fritz H. Bach
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Leo E. Otterbein
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
30
|
Kreisel D, Goldstein DR. Innate immunity and organ transplantation: focus on lung transplantation. Transpl Int 2012; 26:2-10. [PMID: 22909350 DOI: 10.1111/j.1432-2277.2012.01549.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Ischemia reperfusion injury (IRI) that occurs with solid organ transplantation activates the innate immune system to induce inflammation. This leads to enhanced acute allograft rejection, impaired transplant tolerance and accelerated progression of chronic rejection. In this review, we discuss the innate immune signaling pathways that have been shown to play a role in organ transplantation. In particular, we focus on Toll-like receptor signaling pathways and how they have influenced outcomes after organ transplantation both experimentally and from clinical studies. Furthermore, we describe the substances that trigger the innate immune system after transplantation and several of the key cellular mediators of inflammation. We specifically point out unique aspects of activation of the innate immune system after lung transplantation. Finally, we discuss the areas that should be investigated in the future to more clearly understand the influence of the innate immune system after organ transplantation.
Collapse
Affiliation(s)
- Daniel Kreisel
- Department of Surgery, Washington University in St Louis, St. Louis, MO, USA
| | | |
Collapse
|
31
|
Saturated fatty acid and TLR signaling link β cell dysfunction and islet inflammation. Cell Metab 2012; 15:518-33. [PMID: 22465073 DOI: 10.1016/j.cmet.2012.01.023] [Citation(s) in RCA: 408] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Revised: 12/28/2011] [Accepted: 01/27/2012] [Indexed: 12/19/2022]
Abstract
Consumption of foods high in saturated fatty acids (FAs) as well as elevated levels of circulating free FAs are known to be associated with T2D. Though previous studies showed inflammation is crucially involved in the development of insulin resistance, how inflammation contributes to β cell dysfunction has remained unclear. We report here the saturated FA palmitate induces β cell dysfunction in vivo by activating inflammatory processes within islets. Through a combination of in vivo and in vitro studies, we show β cells respond to palmitate via the TLR4/MyD88 pathway and produce chemokines that recruit CD11b(+)Ly-6C(+) M1-type proinflammatory monocytes/macrophages to the islets. Depletion of M1-type cells protected mice from palmitate-induced β cell dysfunction. Islet inflammation also plays an essential role in β cell dysfunction in T2D mouse models. Collectively, these results demonstrate a clear mechanistic link between β cell dysfunction and inflammation mediated at least in part via the FFA-TLR4/MyD88 pathway.
Collapse
|
32
|
Adenovirus-mediated heme oxygenase-1 gene transfer to neonatal porcine islet-like cluster cells: the effects on gene expression and protection from cell stress. BIOCHIP JOURNAL 2012. [DOI: 10.1007/s13206-012-6108-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
33
|
Induction of protective genes leads to islet survival and function. J Transplant 2011; 2011:141898. [PMID: 22220267 PMCID: PMC3246756 DOI: 10.1155/2011/141898] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Accepted: 09/01/2011] [Indexed: 12/16/2022] Open
Abstract
Islet transplantation is the most valid approach to the treatment of type 1 diabetes. However, the function of transplanted islets is often compromised since a large number of β cells undergo apoptosis induced by stress and the immune rejection response elicited by the recipient after transplantation. Conventional treatment for islet transplantation is to administer immunosuppressive drugs to the recipient to suppress the immune rejection response mounted against transplanted islets. Induction of protective genes in the recipient (e.g., heme oxygenase-1 (HO-1), A20/tumor necrosis factor alpha inducible protein3 (tnfaip3), biliverdin reductase (BVR), Bcl2, and others) or administration of one or more of the products of HO-1 to the donor, the islets themselves, and/or the recipient offers an alternative or synergistic approach to improve islet graft survival and function. In this perspective, we summarize studies describing the protective effects of these genes on islet survival and function in rodent allogeneic and xenogeneic transplantation models and the prevention of onset of diabetes, with emphasis on HO-1, A20, and BVR. Such approaches are also appealing to islet autotransplantation in patients with chronic pancreatitis after total pancreatectomy, a procedure that currently only leads to 1/3 of transplanted patients being diabetes-free.
Collapse
|
34
|
Vivot K, Langlois A, Jeandidier N, Bietiger W, Pinget M, Gies J, Sigrist S. Instant Blood-mediated Inflammatory Reaction During Islet Transplantation: The Role of Toll-like Receptors Signaling Pathways. Transplant Proc 2011; 43:3192-4. [DOI: 10.1016/j.transproceed.2011.09.056] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
35
|
Jahansouz C, Jahansouz C, Kumer SC, Brayman KL. Evolution of β-Cell Replacement Therapy in Diabetes Mellitus: Islet Cell Transplantation. J Transplant 2011; 2011:247959. [PMID: 22013505 PMCID: PMC3195999 DOI: 10.1155/2011/247959] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2011] [Accepted: 08/08/2011] [Indexed: 12/12/2022] Open
Abstract
Diabetes mellitus remains one of the leading causes of morbidity and mortality worldwide. According to the Centers for Disease Control and Prevention, approximately 23.6 million people in the United States are affected. Of these individuals, 5 to 10% have been diagnosed with Type 1 diabetes mellitus (T1DM), an autoimmune disease. Although it often appears in childhood, T1DM may manifest at any age, leading to significant morbidity and decreased quality of life. Since the 1960s, the surgical treatment for diabetes mellitus has evolved to become a viable alternative to insulin administration, beginning with pancreatic transplantation. While islet cell transplantation has emerged as another potential alternative, its role in the treatment of T1DM remains to be solidified as research continues to establish it as a truly viable alternative for achieving insulin independence. In this paper, the historical evolution, procurement, current status, benefits, risks, and ongoing research of islet cell transplantation are explored.
Collapse
Affiliation(s)
- Cyrus Jahansouz
- School of Medicine, University of Virginia, Charlottesville, VA 22102, USA
| | | | | | | |
Collapse
|
36
|
Kennelly KP, Wallace DM, Holmes TM, Hankey DJ, Grant TS, O'Farrelly C, Keegan DJ. Preparation of pre-confluent retinal cells increases graft viability in vitro and in vivo: a mouse model. PLoS One 2011; 6:e21365. [PMID: 21738643 PMCID: PMC3126823 DOI: 10.1371/journal.pone.0021365] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2011] [Accepted: 05/30/2011] [Indexed: 11/18/2022] Open
Abstract
PURPOSE Graft failure remains an obstacle to experimental subretinal cell transplantation. A key step is preparing a viable graft, as high levels of necrosis and apoptosis increase the risk of graft failure. Retinal grafts are commonly harvested from cell cultures. We termed the graft preparation procedure "transplant conditions" (TC). We hypothesized that culture conditions influenced graft viability, and investigated whether viability decreased following TC using a mouse retinal pigment epithelial (RPE) cell line, DH01. METHODS Cell viability was assessed by trypan blue exclusion. Levels of apoptosis and necrosis in vitro were determined by flow cytometry for annexin V and propidium iodide and Western blot analysis for the pro- and cleaved forms of caspases 3 and 7. Graft viability in vivo was established by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) and cleaved caspase 3 immunolabeling of subretinal allografts. RESULTS Pre-confluent cultures had significantly less nonviable cells than post-confluent cultures (6.6%±0.8% vs. 13.1%±0.9%, p<0.01). Cell viability in either group was not altered significantly following TC. Caspases 3 and 7 were not altered by levels of confluence or following TC. Pre-confluent cultures had low levels of apoptosis/necrosis (5.6%±1.1%) that did not increase following TC (4.8%±0.5%). However, culturing beyond confluence led to progressively increasing levels of apoptosis and necrosis (up to 16.5%±0.9%). Allografts prepared from post-confluent cultures had significantly more TUNEL-positive cells 3 hours post-operatively than grafts of pre-confluent cells (12.7%±3.1% vs. 4.5%±1.4%, p<0.001). Subretinal grafts of post-confluent cells also had significantly higher rates of cleaved caspase 3 than pre-confluent grafts (20.2%±4.3% vs. 7.8%±1.8%, p<0.001). CONCLUSION Pre-confluent cells should be used to maximize graft cell viability.
Collapse
Affiliation(s)
- Kevin P Kennelly
- Catherine McAuley Clinical Research Centre, University College Dublin, Dublin, Ireland.
| | | | | | | | | | | | | |
Collapse
|
37
|
Toll-like receptor 4 (TLR4) expression in human and murine pancreatic beta-cells affects cell viability and insulin homeostasis. BMC Immunol 2011; 12:18. [PMID: 21356084 PMCID: PMC3060152 DOI: 10.1186/1471-2172-12-18] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Accepted: 02/28/2011] [Indexed: 01/04/2023] Open
Abstract
Background Toll-like receptor 4 (TLR4) is widely recognized as an essential element in the triggering of innate immunity, binding pathogen-associated molecules such as Lipopolysaccharide (LPS), and in initiating a cascade of pro-inflammatory events. Evidence for TLR4 expression in non-immune cells, including pancreatic β-cells, has been shown, but, the functional role of TLR4 in the physiology of human pancreatic β-cells is still to be clearly established. We investigated whether TLR4 is present in β-cells purified from freshly isolated human islets and confirmed the results using MIN6 mouse insulinoma cells, by analyzing the effects of TLR4 expression on cell viability and insulin homeostasis. Results CD11b positive macrophages were practically absent from isolated human islets obtained from non-diabetic brain-dead donors, and TLR4 mRNA and cell surface expression were restricted to β-cells. A significant loss of cell viability was observed in these β-cells indicating a possible relationship with TLR4 expression. Monitoring gene expression in β-cells exposed for 48h to the prototypical TLR4 ligand LPS showed a concentration-dependent increase in TLR4 and CD14 transcripts and decreased insulin content and secretion. TLR4-positive MIN6 cells were also LPS-responsive, increasing TLR4 and CD14 mRNA levels and decreasing cell viability and insulin content. Conclusions Taken together, our data indicate a novel function for TLR4 as a molecule capable of altering homeostasis of pancreatic β-cells.
Collapse
|
38
|
Goldstein DR. Inflammation and transplantation tolerance. Semin Immunopathol 2011; 33:111-5. [DOI: 10.1007/s00281-011-0251-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2010] [Accepted: 01/13/2011] [Indexed: 12/01/2022]
|
39
|
Blancou P, Tardif V, Simon T, Rémy S, Carreño L, Kalergis A, Anegon I. Immunoregulatory properties of heme oxygenase-1. Methods Mol Biol 2011; 677:247-268. [PMID: 20941616 DOI: 10.1007/978-1-60761-869-0_18] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Heme oxygenase-1 (HO-1) is one of the three isoforms of the heme oxygenase enzyme that catabolyzes the degradation of heme into biliverdin with the production of free iron and CO. HO-1 is induced by its substrate and by other stimuli, including agents involved in oxidative stress and proinflammatory cytokines as well as several anti-inflammatory stimuli. A growing body of evidence points toward the capacity of this molecule to inhibit immune reactions and the pivotal role of HO-1 in inflammatory diseases. We will first review the physiological role of HO-1 as determined by the analysis of HO-1-deficient individuals. This will be followed by an examination of the effect of HO-1 within immunopathological contexts such as immune disorders (autoimmunity and allergy) or infections. A section will be devoted to the use of an HO-1 inducer as an immunosuppressive molecule in transplantation. Finally, we will review the molecular basis of HO-1 actions on different immune cells.
Collapse
|
40
|
Testro AG, Visvanathan K, Skinner N, Markovska V, Crowley P, Angus PW, Gow PJ. Acute allograft rejection in human liver transplant recipients is associated with signaling through toll-like receptor 4. J Gastroenterol Hepatol 2011; 26:155-63. [PMID: 21175809 DOI: 10.1111/j.1440-1746.2010.06324.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS Toll-like receptor (TLR) signaling is a crucial step in initiating adaptive immune responses. In addition to recognizing endotoxin, TLR4 also recognizes endogenous ligands ('damage-associated structures'), which are released into the circulation in the peri-transplantation period. TLR2 to a lesser extent also recognizes these endogenous ligands. Multiple studies involving solid organ transplants demonstrate a clear association between TLR4 and allograft rejection. In the present study we assessed whether an association exists between TLR4 and TLR2-dependent responses and acute liver allograft rejection. METHODS The sample included 26 liver transplant recipients. Blood was taken pre-transplant and at multiple points over the first 14 days post-transplant. Monocytes were stimulated with TLR4 and TLR2 ligands, lipopolysaccharide and Pam-3-Cys, respectively. Monocyte TLR expression was determined using flow cytometry; enzyme-linked immunosorbent assays measured tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) production. RESULTS Nine (34.6%) patients experienced rejection. No differences existed in age, sex, disease or immunosuppression between rejectors and non-rejectors. Baseline TLR4 expression was significantly higher in rejectors (1.36 vs 1.02, P=0.01). There was no difference in TLR2 expression. In rejectors, baseline TLR4- and TLR2-dependent production of TNF-α and IL-6 was also significantly increased. Post-transplant, the two groups differed with regard to TLR4-dependent TNF-α production, with rejectors demonstrating progressive downregulation over the first week. CONCLUSIONS Prior to liver transplantation, patients who subsequently experience rejection demonstrate robust TLR4-dependent immune responses, which are not seen in those who do not reject. This supports the theory that damage-associated structures signaling through TLR4 may be responsible for the early activation of alloimmune T-cells, favoring allograft rejection.
Collapse
Affiliation(s)
- Adam G Testro
- Department of Medicine, The University of Melbourne, Austin Health, Melbourne, Victoria, Australia.
| | | | | | | | | | | | | |
Collapse
|
41
|
Koliaraki V, Kollias G. A new role for myeloid HO-1 in the innate to adaptive crosstalk and immune homeostasis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 780:101-11. [PMID: 21842368 DOI: 10.1007/978-1-4419-5632-3_9] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Increasing evidence supports the presence of a dynamic crosstalk between innate and adaptive immunity with a pivotal role played by pathways governing innate immune responses. TLRs (Toll-like receptors) and RLHs (retinoic acid-inducible gene I [RIG-I]-like helicases) are known to play a key role in these processes. A molecule of high significance in the protection against innate and adaptive immune aberrations is heme oxygenase 1 (HO-1). HO-1 is a microsomal enzyme that catalyses the degradation of heme to iron, carbon monoxide and bilirubin. These by-products appear to be the key mediators of its anti--inflammatory and cytoprotective action, mainly through the downregulation of pro-inflammatory and upregulation of anti-inflammatory molecules. Recent data from our lab support the presence of an additional direct effect of myeloid HO-1 on innate immune conditioning, and more specifically on the TLR3/TLR4/RIG-I pathway. In myeloid cells, HO-1 forms a complex with the transcription factor IRF3 (Interferon regulating factor 3) and is required for IRF3 phosphorylation and consequent type-I interferon and chemokine gene induction. Myeloid HO-1-deficient mice show reduced expression of IRF3 target genes and altered responses to infectious and organ-specific auto-immune diseases. This new frame of understanding HO-1 function should also be important for the future design of novel interventions differentially targeting the enzymatic versus the IRF3 modulating properties of HO-1.
Collapse
|
42
|
|
43
|
Krüger B, Yin N, Zhang N, Yadav A, Coward W, Lal G, Zang W, S Heeger P, Bromberg JS, Murphy B, Schröppel B. Islet-expressed TLR2 and TLR4 sense injury and mediate early graft failure after transplantation. Eur J Immunol 2010; 40:2914-24. [PMID: 20809521 DOI: 10.1002/eji.201040601] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Although islet transplantation is an effective treatment for Type 1 diabetes, primary engraftment failure contributes to suboptimal outcomes. We tested the hypothesis that islet isolation and transplantation activate innate immunity through TLR expressed on islets. Murine islets constitutively express TLR2 and TLR4, and TLR activation with peptidoglycan or LPS upregulates islet production of cytokines and chemokines. Following transplantation into streptozotocin-induced diabetic, syngeneic mice, islets exposed to LPS or peptidoglycan had primary graft failure with intra- and peri-islet mononuclear cell inflammation. The use of knockout mice showed that recipient CD8(+) T cells caused engraftment failure and did so in the absence of islet-derived DC. To mimic physiological islet injury, islets were transplanted with exocrine debris. Transplantation of TLR2/4(-/-) islets reduced proinflammatory cytokine production and improved islet survival. Stressed islets released the alarmin high-mobility group box protein 1 (HMGB1) and recombinant HMGB1 (rHMGB1) induced NFkB activation. NFkB activation was prevented in the absence of both TLR2 and TLR4. rHMGB1 pretreatment also prevented primary engraftment through a TLR2/4-dependent pathway. Our results show that islet graft failure can be initiated by TLR2 and TLR4 signaling and suggest that HMGB1 is one likely early mediator. Subsequent downstream signaling results in intra-islet inflammation followed by T-cell-mediated graft destruction.
Collapse
Affiliation(s)
- Bernd Krüger
- Division of Nephrology, Mount Sinai School of Medicine, New York, NY 10029-6574, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
Heme oxygenase-1 (HO-1) is the rate-limiting enzyme in heme catabolism that converts heme to Fe++, carbon monoxide and biliverdin. HO-1 acts anti-inflammatory and modulates apoptosis in many pathological conditions. In transplantation, HO-1 is overexpressed in organs during brain death, when undergoing ischemic damage and rejection. However, intentionally induced, it ameliorates pathological processes like ischemia reperfusion injury, allograft, xenograft or islet rejection, facilitates donor specific tolerance and alleviates chronic allograft changes. We herein consistently summarize the huge amount of data on HO-1 and transplantation that have been generated in multiple laboratories during the last 15years and suggest possible clinical implications and applications for the near future.
Collapse
Affiliation(s)
- Robert Öllinger
- Department of Visceral, Transplant and Thoracic Surgery, Medical University Innsbruck, Innsbruck, Austria.
| | | |
Collapse
|
45
|
Abstract
Chronic inflammation is a key feature of insulin resistance and obesity. Toll-Like Receptor 4 (TLR4), involved in modulating innate immunity, is an important mediator of insulin resistance and its comorbidities. TLR4 contributes to the development of insulin resistance and inflammation through its activation by elevated exogenous ligands (e.g., dietary fatty acids and enteric lipopolysaccharide) and endogenous ligands (e.g., free fatty acids) which are elevated in obese states. TLR4, expressed in insulin target tissues, activates proinflammatory kinases JNK, IKK, and p38 that impair insulin signal transduction directly through inhibitory phosphorylation of insulin receptor substrate (IRS) on serine residues. TLR4 activation also leads to increased transcription of pro-inflammatory genes, resulting in elevation of cytokine, chemokine, reactive oxygen species, and eicosanoid levels that promote further insulin-desensitization within the target cell itself and in other cells via paracrine and systemic effects. Increased understanding of cell type-specific TLR4-mediated effects on insulin action present the opportunity and challenge of developing related therapeutic approaches for improving insulin sensitivity while preserving innate immunity.
Collapse
|
46
|
Rocuts F, Ma Y, Zhang X, Gao W, Yue Y, Vartanian T, Wang H. Carbon monoxide suppresses membrane expression of TLR4 via myeloid differentiation factor-2 in betaTC3 cells. THE JOURNAL OF IMMUNOLOGY 2010; 185:2134-9. [PMID: 20631306 DOI: 10.4049/jimmunol.0902782] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Islet allografts from donor mice exposed to CO are protected from immune rejection after transplantation via the suppression of membrane trafficking/activation of TLR4 in islets/beta cells. The molecular mechanisms of how CO suppresses TLR4 activation in beta cells remain unclear and are the focus of this study. Cells of the insulinoma cell line, betaTC3, were stably transfected with pcDNA3-TLR4-YFP and pDsRed-Monomer-Golgi plasmids and used to identify the subcellular distribution of TLR4 before and after LPS stimulation by confocal microscopy. Immunofluorescence analysis revealed that TLR4 mainly resides in the Golgi apparatus in betaTC3 cells when in a quiescent state. LPS stimulation led to a rapid trafficking of TLR4 from the Golgi to the cell membrane. Physical interaction between TLR4 and myeloid differentiation factor-2 (MD-2) was confirmed by immunoprecipitation. Depleting MD-2 using small interfering RNA or blocking the N-glycosylation of cells using tunicamycin blocked membrane trafficking of TLR4. Pre-exposing cells to CO at a concentration of 250 parts per million suppressed membrane trafficking of TLR4 via inhibiting its glycosylation and the interaction between TLR4 and MD-2. In conclusion, MD-2 is required for the glycosylation of TLR4 and its consequent membrane trafficking in betaTC3 cells. CO suppresses membrane activation of TLR4 via blocking its glycosylation and the physical interaction between TLR4 and MD-2.
Collapse
Affiliation(s)
- Fredy Rocuts
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Gao Q, Ma LL, Gao X, Yan W, Williams P, Yin DP. TLR4 mediates early graft failure after intraportal islet transplantation. Am J Transplant 2010; 10:1588-96. [PMID: 20642685 DOI: 10.1111/j.1600-6143.2010.03151.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
We have previously shown that islet emboli in the portal vein block blood flow and induce local inflammatory reaction, resulting in functional loss of islet grafts following intraportal transplantation. This study was designed to test whether Toll-like receptor (TLR) activation mediates early islet graft failure. Syngeneic islet grafts were transplanted into chemically induced diabetic mice, and TLR deficient mice were used as donors and/or recipients of islet grafts. Islet viability, proinflammatory cytokines, high-mobility group box-1 (HMGB1) and NF-kappaB activation were analyzed by bioluminesce imaging (BLI), quantitative RT-PCR (qRT-PCR) and histology. Early islet graft failure was observed in mice with intraportal islet engrafts with increased proinflammatory cytokines, HMGB1 expression, NF-kappaB activation, caspase-3 and TUNEL positive cells. Deficiency of TLR4 in donor, but not in recipient, inhibited NF-kappaB activation, reduced proinflammatory cytokines and improved viability of islet grafts. Blockade of HMGB1 with anti-HMGB1 monoclonal antibody (mAb, 2g7) inhibited inflammatory reactions, as evidenced by reduced TNFalpha and IL-1ss production, and improved islet viability. We conclude that TLR4 activation mediates early graft failure following intraportal islet transplantation. Inhibition of TLR4 activation represents a novel strategy to attenuate early graft failure following intraportal islet transplantation.
Collapse
Affiliation(s)
- Q Gao
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | | | | | | | | |
Collapse
|
48
|
Park SG, Kim JH, Oh JH, Lee HN, Park HS, Chung SS, Lee YJ, Lee YY, Jung HS, Park KS. Polymyxin B, scavenger of endotoxin, enhances isolation yield andin vivofunction of islets. Transpl Int 2010; 23:325-32. [DOI: 10.1111/j.1432-2277.2009.00987.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
49
|
Abstract
BACKGROUND Islet transplantation is a promising therapy for type 1 diabetes; however, most islet grafts fail within 5 years. Innate immunity has been suggested to play a role in islet allograft rejection, potentially mediated by toll-like receptors (TLRs), a class of innate immune receptors. Lack of TLR4, in particular, has been reported to improve allograft survival. Therefore, we hypothesized that TLRs may be involved in islet allograft rejection, and that deletion of TLR4 may improve islet graft survival. METHODS Islets were isolated from C57BL/10ScNJ (Tlr4(-/-)) and C57BL/10 (wild-type [WT]) animals and transplanted into Balb/cJ recipients with streptozotocin-induced diabetes. Blood glucose levels were used to determine graft viability and immunostaining to assess graft morphology and immune cell infiltration. The roles of the TLR4 adaptor molecules MyD88 and TLR adaptor molecule 1 (Ticam-1) were assessed using islets isolated from mice lacking MyD88 (MyD88(-/-)), Ticam-1 (Ticam-1(-/-)), or the combined double knockout (MyD88(-/-)/Ticam-1(-/-)). RESULTS Contrary to our hypothesis, Tlr4(-/-) and WT islet allografts had similar failure rates; grafts failed at 23.2+/-1.2 and 24.5+/-1.5 days posttransplant, respectively (P=NS). Syngeneic grafts of Tlr4(-/-) and WT islets maintained normoglycemia for up to 10 weeks posttransplant, indicating that failure of Tlr4(-/-) islet allografts could not be attributed to an intrinsic defect in Tlr4(-/-) islets. Similarly, islet allotransplants from MyD88(-/-), Ticam-1(-/-), and MyD88(-/-)/Ticam-1(-/-) donors did not have improved allograft survival compared with WT controls. CONCLUSIONS These findings indicate that islet allograft rejection in mice is independent of TLR4 and the TLR adaptor molecules MyD88 and Ticam-1, speaking against an essential role for TLR signaling in islet allograft rejection.
Collapse
|
50
|
Inhibition of TLR4 signaling prolongs Treg-dependent murine islet allograft survival. Immunol Lett 2009; 127:119-25. [PMID: 19879295 DOI: 10.1016/j.imlet.2009.10.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2009] [Revised: 09/18/2009] [Accepted: 10/20/2009] [Indexed: 11/20/2022]
Abstract
Toll-like receptors (TLRs) provide an important link between innate and adaptive immune system. We hypothesized that the recognition of endogenous TLR4 ligands is occurring at the time of transplantation, and these innate signals drive the inflammation and affect alloimmune responses. We confirmed that early after transplantation of allogenic islets, transcripts for TLR4 as well as potential ligands were released or up-regulated. In an allogenic islet transplantation model, genetic disruption of TLR4 on donor islets had no effect on allograft survival, whereas TLR4 deficiency in recipients lead to prolonged graft survival. Low dose rapamycin-treatment of TLR4(-/-) recipients induced permanent engraftment of 45% islet graft (p=0.005) compared to WT recipients. This prolonged graft survival was dependent on the presence of CD4(+)CD25(+)Foxp3(+) Treg. Naïve CD4(+)CD25(-) T cells cultured with the TLR4 ligand lipopolysaccharide showed enhanced IL-4, IL-6, IL-17, IFN gamma secretion and inhibited TGFbeta induced Foxp3(+)Treg generation. Thus, inhibition of recipient TLR4 activation at the time of transplantation decreases proinflammatory signals and allows Treg generation.
Collapse
|