1
|
Luo H, Wu P, Chen X, Wang B, Chen G, Su X. Novel insights into the relationship between α-1 anti-trypsin with the pathological development of cardio-metabolic disorders. Int Immunopharmacol 2022; 111:109077. [PMID: 35907338 DOI: 10.1016/j.intimp.2022.109077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 07/09/2022] [Accepted: 07/18/2022] [Indexed: 11/05/2022]
Abstract
According to the previous studies, chronic low-grade systemic inflammatory response has been shown to be significantly associated with the pathological development of cardio-metabolic disorder diseases, including atherosclerosis, type 2 diabetes mellitus, and non-alcoholic fatty liver disease (NAFLD). On the other hand, auto-immunity process could also facilitate the pathogenesis of type 1 diabetes mellitus importantly. Concerning on this notion, the anti-inflammatory therapeutic strategy is demonstrated to embrace an essential function in those cardio-metabolic disorders in clinical practice. The α-1 anti-trypsin, also named Serpin-A1 and as an acute phase endogenous protein, has been verified to have several modulatory effects such as anti-inflammatory response, anti-apoptosis, and immunomodulatory functions. In addition, it is also used for therapeutic strategy of a rare genetic disease caused by the deficiency of α-1 anti-trypsin. Recent emerging evidence has indicated that the serum concentrations of α-1 anti-trypsin levels and its biological activity are significantly changed in those inflammatory and immune related cardio-metabolic disorder diseases. Nevertheless, the underlying mechanism is still not elucidated. In the current review, the basic experiments and clinical trials which provided the evidence revealing the potential therapeutic function of the α-1 anti-trypsin in cardio-metabolic disorder diseases were well-summarized. Furthermore, the results which indicated that the α-1 anti-trypsin presented the possibility as a novel serum biomarker in humans to predict those cardio-metabolic disorder diseases were also elucidated.
Collapse
Affiliation(s)
- Haizhen Luo
- Department of Cardiology, the Fuding Hospital of Fujian University of Traditional Chinese Medicine, Fuding, Fujian, China
| | - Penglong Wu
- Department of Cardiology, the Xiamen Cardiovascular Hospital of Xiamen University, Xiamen, Fujian, China
| | - Xiang Chen
- Department of Cardiology, the Xiamen Cardiovascular Hospital of Xiamen University, Xiamen, Fujian, China
| | - Bin Wang
- Department of Cardiology, the Xiamen Cardiovascular Hospital of Xiamen University, Xiamen, Fujian, China
| | - Geng Chen
- Department of Cardiology, the Fuding Hospital of Fujian University of Traditional Chinese Medicine, Fuding, Fujian, China.
| | - Xin Su
- Department of Cardiology, the Xiamen Cardiovascular Hospital of Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
2
|
Zhang X, Ostrov DA, Tian H. Alpha-1 antitrypsin: A novel biomarker and potential therapeutic approach for metabolic diseases. Clin Chim Acta 2022; 534:71-76. [PMID: 35810800 DOI: 10.1016/j.cca.2022.07.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/02/2022] [Accepted: 07/05/2022] [Indexed: 02/05/2023]
Abstract
It is well recognized that chronic low-grade systemic inflammation and autoimmunity contribute to the pathogenesis of metabolic syndrome, its associated diseases (e.g. type 2 diabetes, non-alcoholic fatty liver disease) and type 1 diabetes, respectively. Consequently, anti-inflammatory agents might play a role in managing these immune associated metabolic diseases. Alpha-1 antitrypsin (AAT), an endogenous acute phase protein being used for treatment of AAT deficiency (a rare genetic disease), has multiple functions including anti-inflammatory, immunomodulatory, anti-apoptosis and cytoprotective effects. In this review, we summarized basic and clinical studies that reported potential therapeutic role of AAT in metabolic syndrome associated diseases and type 1 diabetes. Studies that demonstrated AAT had the possibility to be used as a novel biomarker to predict these immune associated metabolic diseases were also included.
Collapse
Affiliation(s)
- Xiaojuan Zhang
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu 610041, China
| | - David A Ostrov
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL 32611, USA
| | - Haoming Tian
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
3
|
Lior Y, Shtriker E, Kahremany S, Lewis EC, Gruzman A. Development of anti-inflammatory peptidomimetics based on the structure of human alpha1-antitrypsin. Eur J Med Chem 2021; 228:113969. [PMID: 34763945 DOI: 10.1016/j.ejmech.2021.113969] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 10/29/2021] [Accepted: 10/30/2021] [Indexed: 01/10/2023]
Abstract
Human α1-antitrypsin (hAAT) has two distinguishing functions: anti-protease activity and regulation of the immune system. In the present study we hypothesized that those two protein functions are mediated by different structural domains on the hAAT surface. Indeed, such biologically active immunoregulatory sites (not associated with canonical anti-protease activity) on the surface of hAAT were identified by in silico methods. Several peptides were derived from those immunoregulatory sites. Four peptides exhibited impressive biological effects in pharmacological concentration ranges. Peptidomimetic (14) was developed, based on the structure of the most druggable and active peptide. The compound exhibited a potent anti-inflammatory activity in vitro and in vivo. Such a compound could be used as a basis for developing novel anti-inflammatory drug candidates and as a research tool for better understanding hAAT functions.
Collapse
Affiliation(s)
- Yotam Lior
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Efrat Shtriker
- Department of Chemistry, Faculty of Exact Sciences, Bar-Ilan University, Ramat Gan, 5290002, Israel
| | - Shirin Kahremany
- Department of Chemistry, Faculty of Exact Sciences, Bar-Ilan University, Ramat Gan, 5290002, Israel; The Skin Research Institute, The Dead Sea and Arava Science Center, 86910, Masada, Israel
| | - Eli C Lewis
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Arie Gruzman
- Department of Chemistry, Faculty of Exact Sciences, Bar-Ilan University, Ramat Gan, 5290002, Israel.
| |
Collapse
|
4
|
Finotti P, Pagetta A. A mutant α1antitrypsin in complex with heat shock proteins as the primary antigen in type 1 diabetes in silico investigation. Sci Rep 2021; 11:3002. [PMID: 33542414 PMCID: PMC7862655 DOI: 10.1038/s41598-021-82730-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 01/25/2021] [Indexed: 11/09/2022] Open
Abstract
Based on previous results demonstrating that complexes of a mutant α1-antitrypsin with the heat shock proteins (HSP)70 and glucose-regulated protein94 (Grp94) circulate in the blood of patients with type 1 diabetes, we raised the hypothesis that these complexes could represent the primary antigen capable of triggering the autoimmune reactions leading to overt diabetes. As a first approach to this issue, we searched whether A1AT and HSPs had a sequence similarity to major islet antigen proteins so as to identify among the similar sequences those with potential relevance for the pathogenesis of diabetes. A thorough in silico analysis was performed to establish the score of similarity of the human proteins: A1AT, pro-insulin (INS), GAD65, IAPP, IA-2, ICA69, Grp94, HSP70 and HSP60. The sequences of A1AT and HSPs with the highest score of similarity to the islet peptides reported in the literature as the main autoantigens in human diabetes were recorded. At variance with other HSPs, also including HSP90 and Grp78, Grp94 contained the highest number and the longest sequences with structural similarity to A1AT and to well-known immunogenic peptides/epitopes of INS, GAD65, and IA-2. The similarity of A1AT with Grp94 and that of Grp94 with INS also suggested a functional relationship among the proteins. Specific sequences were identified in A1AT, Grp94 and HSP70, with the highest score of cross-similarity to a pattern of eight different islet protein epitopes. The similarity also involved recently discovered autoantigens in type 1 diabetes such as a hybrid peptides of insulin and the defective ribosomal insulin gene product. The significant similarity displayed by specific sequences of Grp94 and A1AT to the islet peptides considered main antigens in human diabetes, is a strong indication for testing these sequences as new peptides of immunogenic relevance in diabetes.
Collapse
Affiliation(s)
- Paola Finotti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Building "C", Largo E. Meneghetti, 2, 35131, Padua, Italy.
| | - Andrea Pagetta
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Building "C", Largo E. Meneghetti, 2, 35131, Padua, Italy
| |
Collapse
|
5
|
Belchamber KBR, Walker EM, Stockley RA, Sapey E. Monocytes and Macrophages in Alpha-1 Antitrypsin Deficiency. Int J Chron Obstruct Pulmon Dis 2020; 15:3183-3192. [PMID: 33311976 PMCID: PMC7725100 DOI: 10.2147/copd.s276792] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 10/14/2020] [Indexed: 12/14/2022] Open
Abstract
Alpha-1 antitrypsin deficiency (AATD) is a genetic condition characterised by low circulating levels of alpha-1 antitrypsin (AAT), a serine proteinase inhibitor. The most common deficiency variants are the S and Z mutations, which cause the accumulation of misfolded AAT in hepatocytes resulting in endoplasmic reticular stress and insufficient release of AAT into the circulation (<11μmol/L). This leads to liver disease, as well as an increased risk of emphysema due to unopposed proteolytic activity of neutrophil-derived serine proteinases in the lungs. AATD has been traditionally viewed as an inflammatory disorder caused directly by a proteinase-antiproteinase imbalance in the lung, but increasing evidence suggests that low AAT levels may affect other cellular functions. Recently, AAT polymers have been identified in both monocytes and macrophages from AATD patients and evidence is building that these cells may also play a role in the development of AATD lung disease. Alveolar macrophages are phagocytic cells that are important in the lung immune response but are also implicated in driving inflammation. This review explores the potential implications of monocyte and macrophage involvement in non-liver AAT synthesis and the pathophysiology of AATD lung disease.
Collapse
Affiliation(s)
- Kylie B R Belchamber
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Eloise M Walker
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Robert A Stockley
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Elizabeth Sapey
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- NIHR Clinical Research Facility Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| |
Collapse
|
6
|
Evidence for the important role of inflammation in xenotransplantation. JOURNAL OF INFLAMMATION-LONDON 2019; 16:10. [PMID: 31148951 PMCID: PMC6537172 DOI: 10.1186/s12950-019-0213-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 05/02/2019] [Indexed: 12/17/2022]
Abstract
There is increasing evidence of a sustained state of systemic inflammation after pig-to-nonhuman primate (NHP) xenotransplantation (that has been termed systemic inflammation in xenograft recipients [SIXR]). Increases in inflammatory markers, e.g., C-reactive protein, histones, serum amyloid A, D-dimer, cytokines, chemokines, and a decrease in free triiodothyronine, have been demonstrated in the recipient NHPs. The complex interactions between inflammation, coagulation, and the immune response are well-recognized, but the role of inflammation in xenograft recipients is not fully understood. The evidence suggests that inflammation can promote the activation of coagulation and the adaptive immune response, but the exact mechanisms remain uncertain. If prolonged xenograft survival is to be achieved, anti-inflammatory strategies (e.g., the administration of anti-inflammatory agents, and/or the generation of genetically-engineered organ-source pigs that are protected from the effect of inflammation) may be necessary to prevent, control, or negate the effect of the systemic inflammation that develops in xenograft recipients. This may allow for a reduction in the intensity of exogenous immunosuppressive therapy. If immunological tolerance to a xenograft is to be obtained, then control of inflammation may be essential.
Collapse
|
7
|
Dominguez Gutierrez G, Kim J, Lee AH, Tong J, Niu J, Gray SM, Wei Y, Ding Y, Ni M, Adler C, Murphy AJ, Gromada J, Xin Y. Gene Signature of the Human Pancreatic ε Cell. Endocrinology 2018; 159:4023-4032. [PMID: 30380031 PMCID: PMC6963699 DOI: 10.1210/en.2018-00833] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 10/17/2018] [Indexed: 12/31/2022]
Abstract
The ghrelin-producing ε cell represents the fifth endocrine cell type in human pancreatic islets. The abundance of ε cells in adult pancreas is extremely low, which has hampered the investigation on the molecular pathways regulating the development and the function of this cell type. In this study, we explored the molecular features defining the function of pancreatic ε cells isolated from adult nondiabetic donors using single-cell RNA sequencing technology. We focus on transcription factors, cell surface receptors, and genes involved in metabolic pathways that contribute to regulation of cellular function. Furthermore, the genes that separate ε cells from the other islet endocrine cell types are presented. This study expands prior knowledge about the genes important for ε cell functioning during development and provides a resource to interrogate the transcriptome of this rare human islet cell type.
Collapse
Affiliation(s)
| | - Jinrang Kim
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| | - Ann-Hwee Lee
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| | - Jenny Tong
- Division of Endocrinology, Metabolism and Nutrition, Duke Molecular Physiology Institute, Duke University, Durham, North Carolina
| | - JingJing Niu
- Division of Endocrinology, Metabolism and Nutrition, Duke Molecular Physiology Institute, Duke University, Durham, North Carolina
| | - Sarah M Gray
- Division of Endocrinology, Metabolism and Nutrition, Duke Molecular Physiology Institute, Duke University, Durham, North Carolina
| | - Yi Wei
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| | - Yueming Ding
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| | - Min Ni
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| | | | | | | | - Yurong Xin
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| |
Collapse
|
8
|
Song S. Alpha-1 Antitrypsin Therapy for Autoimmune Disorders. CHRONIC OBSTRUCTIVE PULMONARY DISEASES-JOURNAL OF THE COPD FOUNDATION 2018; 5:289-301. [PMID: 30723786 DOI: 10.15326/jcopdf.5.4.2018.0131] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Autoimmune diseases are conditions caused by an over reactive immune system that attacks self-tissues and organs. Although the pathogenesis of autoimmune disease is complex and multi-factorial, inflammation is commonly involved. Therefore, anti-inflammatory therapies hold potential for the treatment of autoimmune diseases. However, long-term control of inflammation is challenging and most of the currently used drugs have side effects. Alpha-1 antitrypsin (AAT) is an anti-inflammatory protein with a well-known safety profile. The therapeutic potential of AAT has been tested in several autoimmune disease models. The first study using a recombinant adeno-associated viral (rAAV) vector showed that AAT gene transfer prevented the development of type 1 diabetes (T1D) in the non-obese diabetic (NOD) mouse model. Subsequent studies showed that treatment with AAT protein prevented and reversed type 1 diabetes. The beneficial effects of AAT treatment have also been observed in other autoimmune disease models such as rheumatoid arthritis and systemic lupus erythematosus. This paper reviews the therapeutic application of AAT and discusses possible mechanisms of action in various autoimmune diseases.
Collapse
Affiliation(s)
- Sihong Song
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville
| |
Collapse
|
9
|
Dunlea DM, Fee LT, McEnery T, McElvaney NG, Reeves EP. The impact of alpha-1 antitrypsin augmentation therapy on neutrophil-driven respiratory disease in deficient individuals. J Inflamm Res 2018; 11:123-134. [PMID: 29618937 PMCID: PMC5875399 DOI: 10.2147/jir.s156405] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Alpha-1 antitrypsin (AAT) is the most abundant serine protease inhibitor circulating in the blood. AAT deficiency (AATD) is an autosomal codominant condition affecting an estimated 3.4 million individuals worldwide. The clinical disease associated with AATD can present in a number of ways including COPD, liver disease, panniculitis and antineutrophil cytoplasmic antibody vasculitis. AATD is the only proven genetic risk factor for the development of COPD, and deficient individuals who smoke are disposed to more aggressive disease. Principally, AAT is a serine protease inhibitor; however, over the past number of years, the assessment of AAT as simply an antiprotease has evolved, and it is now recognized that AAT has significant anti-inflammatory properties affecting a wide range of cells, including the circulating neutrophil.
Collapse
Affiliation(s)
- Danielle M Dunlea
- Irish Centre for Genetic Lung Disease, Department of Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Laura T Fee
- Irish Centre for Genetic Lung Disease, Department of Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Thomas McEnery
- Irish Centre for Genetic Lung Disease, Department of Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Noel G McElvaney
- Irish Centre for Genetic Lung Disease, Department of Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Emer P Reeves
- Irish Centre for Genetic Lung Disease, Department of Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| |
Collapse
|
10
|
Abstract
α1-Antitrypsin deficiency (A1ATD) is an inherited disorder caused by mutations in SERPINA1, leading to liver and lung disease. It is not a rare disorder but frequently goes underdiagnosed or misdiagnosed as asthma, chronic obstructive pulmonary disease (COPD) or cryptogenic liver disease. The most frequent disease-associated mutations include the S allele and the Z allele of SERPINA1, which lead to the accumulation of misfolded α1-antitrypsin in hepatocytes, endoplasmic reticulum stress, low circulating levels of α1-antitrypsin and liver disease. Currently, there is no cure for severe liver disease and the only management option is liver transplantation when liver failure is life-threatening. A1ATD-associated lung disease predominately occurs in adults and is caused principally by inadequate protease inhibition. Treatment of A1ATD-associated lung disease includes standard therapies that are also used for the treatment of COPD, in addition to the use of augmentation therapy (that is, infusions of human plasma-derived, purified α1-antitrypsin). New therapies that target the misfolded α1-antitrypsin or attempt to correct the underlying genetic mutation are currently under development.
Collapse
|
11
|
Barbieux C, Parnaud G, Lavallard V, Brioudes E, Meyer J, Alibashe Ahmed M, Berishvili E, Berney T, Bosco D. Asymmetrical distribution of δ and PP cells in human pancreatic islets. J Endocrinol 2016; 229:123-132. [PMID: 26931137 DOI: 10.1530/joe-15-0542] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 03/01/2016] [Indexed: 01/09/2023]
Abstract
The aim of this study was to evaluate the location of PP and δ cells in relation to the vascularization within human pancreatic islets. To this end, pancreas sections were analysed by immunofluorescence using antibodies against endocrine islet and endothelial cells. Staining in different islet areas corresponding to islet cells adjacent or not to peripheral or central vascular channels was quantified by computerized morphometry. As results, α, PP and δ cells were preferentially found adjacent to vessels. In contrast to α cells, which were evenly distributed between islet periphery and intraislet vascular channels, PP and δ cells had asymmetric and opposite distributions: PP staining was higher and somatostatin staining was lower in the islet periphery than in the area around intraislet vascular channels. Additionally, frequencies of PP and δ cells were negatively correlated in the islets. No difference was observed between islets from the head and the tail of the pancreas, and from type 2 diabetic and non-diabetic donors. In conclusion, the distribution of δ cells differs from that of PP cells in human islets, suggesting that vessels at the periphery and at the centre of islets drain different hormonal cocktails.
Collapse
Affiliation(s)
- Charlotte Barbieux
- Department of SurgeryCell Isolation and Transplantation Center, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Géraldine Parnaud
- Department of SurgeryCell Isolation and Transplantation Center, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Vanessa Lavallard
- Department of SurgeryCell Isolation and Transplantation Center, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Estelle Brioudes
- Department of SurgeryCell Isolation and Transplantation Center, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Jérémy Meyer
- Department of SurgeryCell Isolation and Transplantation Center, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Mohamed Alibashe Ahmed
- Department of SurgeryCell Isolation and Transplantation Center, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Ekaterine Berishvili
- Department of SurgeryCell Isolation and Transplantation Center, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Thierry Berney
- Department of SurgeryCell Isolation and Transplantation Center, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Domenico Bosco
- Department of SurgeryCell Isolation and Transplantation Center, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| |
Collapse
|
12
|
Wanner A, Sandhaus RA. Alpha-1 Antitrypsin as a Therapeutic Agent for Conditions not Associated with Alpha-1 Antitrypsin Deficiency. ALPHA-1 ANTITRYPSIN 2016. [PMCID: PMC7121596 DOI: 10.1007/978-3-319-23449-6_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Alpha-1 antitrypsin is a positive acute phase reactant whose serum level rises in response to inflammatory stress, presumably to balance pro-inflammatory processes. In addition to its serine protease inhibitory action, alpha-1 antitrypsin exhibits broader anti-inflammatory and immunomodulatory activity, and increasing its serum concentration by the administration of exogenous alpha-1 antitrypsin to above-normal levels potentially could be therapeutic in conditions other than alpha-1 antitrypsin deficiency. In vitro observations, studies in animal models and in some instances early human trials suggest that intravenous or inhaled alpha-1 antitrypsin has beneficial effects in type 1 diabetes, viral infections, graft-versus-host disease, cystic fibrosis, and alpha-1 antitrypsin-replete chronic obstructive pulmonary disease among others. While the results of pivotal clinical trials have not been reported to date, new indications for alpha-1 antitrypsin therapy are likely to emerge in the future based on currently available scientific data.
Collapse
Affiliation(s)
- Adam Wanner
- University of Miami Miller School of Medicine, Miami, Florida USA
| | | |
Collapse
|
13
|
Zhao H, Liu H, Chai L, Xu P, Hua L, Guan XY, Duan B, Huang YL, Li YS. Plasma α1-antitrypsin: a neglected predictor of angiographic severity in patients with stable angina pectoris. Chin Med J (Engl) 2015; 128:755-61. [PMID: 25758268 PMCID: PMC4833978 DOI: 10.4103/0366-6999.152485] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Background: As an acute phase protein, α1-antitrypsin (AAT) has been extensively studied in acute coronary syndrome, but it is unclear whether a relationship exists between AAT and stable angina pectoris (SAP). The purpose of the present study was to investigate the association between AAT plasma levels and SAP. Methods: Overall, 103 SAP patients diagnosed by coronary angiography and clinical manifestations and 118 control subjects matched for age and gender were enrolled in this case-control study. Plasma levels of AAT, high-sensitivity C-reactive protein (hsCRP), lipid profiles and other clinical parameters were assayed for all participants. The severity of coronary lesions was evaluated based on the Gensini score (GS) assessed by coronary angiography. Results: Positively correlated with the GS (r = 0.564, P < 0.001), the plasma AAT level in the SAP group was significantly higher than that in the control group (142.08 ± 19.61 mg/dl vs. 125.50 ± 19.67 mg/dl, P < 0.001). The plasma AAT level was an independent predictor for both SAP (odds ratio [OR] = 1.037, 95% confidence interval [CI]: 1.020–1.054, P < 0.001) and a high GS (OR = 1.087, 95% CI: 1.051–1.124, P < 0.001) in a multivariate logistic regression model. In the receiver operating characteristic curve analysis, plasma AAT level was found to have a larger area under the curve (AUC) for predicting a high GS (AUC = 0.858, 95% CI: 0.788–0.929, P < 0.001) than that of hsCRP (AUC = 0.665, 95% CI: 0.557–0.773, P = 0.006; Z = 2.9363, P < 0.001), with an optimal cut-off value of 137.85 mg/dl (sensitivity: 94.3%, specificity: 68.2%). Conclusions: Plasma AAT levels correlate with both the presence and severity of coronary stenosis in patients with SAP, suggesting that it could be a potential predictive marker of severe stenosis in SAP patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yi-Shi Li
- Key Laboratory of Clinical Trial Research in Cardiovascular Drugs, Ministry of Health, State Key Laboratory of Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| |
Collapse
|
14
|
Fleixo-Lima G, Ventura H, Medini M, Bar L, Strauss P, Lewis EC. Mechanistic evidence in support of alpha1-antitrypsin as a therapeutic approach for type 1 diabetes. J Diabetes Sci Technol 2014; 8:1193-203. [PMID: 25155845 PMCID: PMC4455465 DOI: 10.1177/1932296814547096] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Utilizing endogenous molecules as a therapeutic approach is almost unequivocally superior to engineered or synthetic molecules. However, one rarely encounters an anti-inflammatory, cytoprotective, immunomodulatory and wound-healing molecule that has been available for use for decades. α1-antitrypsin (AAT), a circulating protein that rises more than 4-fold during acute-phase responses, has been administered for a rare genetic deficiency at large doses, for life. Aside from advances in insulin therapy, medical research in type 1 diabetes (T1D) has predominantly focused on autoimmunity--controlling the adaptive immune response. However, it is now appreciated that one may need to extend therapeutic targets to incorporate immune responses to cellular injury, as well as promote selective control over excessive inflammation and early tissue repair. Recent data suggest that tissue damage related to lung and renal ischemia-reperfusion injury, stroke, and ischemic heart disease is markedly reduced by AAT. AAT was also shown to protect pancreatic islet β cells at multiple levels. Unlike classic immunosuppressive and anti-inflammatory approaches, AAT exerts some antiviral and antibacterial activities. Based on these and other reports, AAT is under evaluation for treatment of T1D patients in multiple clinical trials. Initial results suggest that AAT therapy could potentially improve insulin production without adverse effects. Up to 50% of individuals displayed improved islet function. It is a rare occurrence in T1D research that a therapy is offered that holds a safety profile equal or superior to that of insulin alone. While placebo-controlled trials are ongoing, the mechanism(s) behind these favorable activities of AAT are still being explored.
Collapse
Affiliation(s)
- Gabriella Fleixo-Lima
- Department of Clinical Biochemistry & Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Hilla Ventura
- Department of Clinical Biochemistry & Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Michal Medini
- Department of Clinical Biochemistry & Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | | | | | - Eli C Lewis
- Department of Clinical Biochemistry & Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| |
Collapse
|
15
|
Salem HH, Trojanowski B, Fiedler K, Maier HJ, Schirmbeck R, Wagner M, Boehm BO, Wirth T, Baumann B. Long-term IKK2/NF-κB signaling in pancreatic β-cells induces immune-mediated diabetes. Diabetes 2014; 63:960-75. [PMID: 24296718 DOI: 10.2337/db13-1037] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Type 1 diabetes is a multifactorial inflammatory disease in genetically susceptible individuals characterized by progressive autoimmune destruction of pancreatic β-cells initiated by yet unknown factors. Although animal models of type 1 diabetes have substantially increased our understanding of disease pathogenesis, heterogeneity seen in human patients cannot be reflected by a single model and calls for additional models covering different aspects of human pathophysiology. Inhibitor of κB kinase (IKK)/nuclear factor-κB (NF-κB) signaling is a master regulator of inflammation; however, its role in diabetes pathogenesis is controversially discussed by studies using different inhibition approaches. To investigate the potential diabetogenic effects of NF-κB in β-cells, we generated a gain-of-function model allowing conditional IKK2/NF-κB activation in β-cells. A transgenic mouse model that expresses a constitutively active mutant of human IKK2 dependent on Pdx-1 promoter activity (IKK2-CA(Pdx-1)) spontaneously develops full-blown immune-mediated diabetes with insulitis, hyperglycemia, and hypoinsulinemia. Disease development involves a gene expression program mimicking virus-induced diabetes and allergic inflammatory responses as well as increased major histocompatibility complex class I/II expression by β-cells that could collectively promote diabetes development. Potential novel diabetes candidate genes were also identified. Interestingly, animals successfully recovered from diabetes upon transgene inactivation. Our data give the first direct evidence that β-cell-specific IKK2/NF-κB activation is a potential trigger of immune-mediated diabetes. Moreover, IKK2-CA(Pdx-1) mice provide a novel tool for studying critical checkpoints in diabetes pathogenesis and mechanisms governing β-cell degeneration/regeneration.
Collapse
Affiliation(s)
- Heba H Salem
- Institute of Physiological Chemistry, Ulm University, Ulm, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Yarmohammadi ME, Hassan ZM, Mostafaie A, Ebtekar M, Yaraee R, Pourfarzam S, Jalali-Nadoushan M, Faghihzadeh S, Vaez-Mahdavi MR, Soroush MR, Khamesipour A, Faghihzadeh E, Sharifnia Z, Naghizadeh MM, Ghazanfari T. Salivary levels of secretary IgA, C5a and alpha 1-antitrypsin in sulfur mustard exposed patients 20 years after the exposure, Sardasht-Iran Cohort Study (SICS). Int Immunopharmacol 2013; 17:952-7. [PMID: 23375936 DOI: 10.1016/j.intimp.2012.12.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Revised: 11/17/2012] [Accepted: 12/27/2012] [Indexed: 12/24/2022]
Abstract
Sulfur mustard (SM) is a strong toxic agent that causes acute and chronic health effects on a myriad of organs following exposure. Although the primary targets of inhaled mustard gas are the epithelia of the upper respiratory tract, the lower respiratory tract is the focus of the current study, and upper tract complications remain obscure. To our knowledge there is no study addressing the secretory IgA (S-IgA), C5a, alpha 1 antitrypsin (A1AT) in the saliva of SM-exposed victims. In this study, as many as 500 volunteers, including 372 SM-exposed cases and 128 control volunteers were recruited. A 3 ml sample of saliva was collected from each volunteer, and the level of secretory IgA, C5a, and alpha 1 antitrypsin in the samples were compared between the two groups. The SM-exposed group showed a significantly higher amount of salivary alpha 1 antitrypsin and secretary IgA compared to the control group (p<.006 and p<.018 respectively). The two groups showed no significant difference (p=0.192) in the level of C5a. The results also showed that the level of salivary A1AT is more than that of IgA in severely injured cases. The findings presented here provide valuable insight for both researchers and practitioners dealing with victims of the chemical warfare agent, sulfur mustard. This research indicates that certain branches of the inflammatory processes mandate serious attention in therapeutic interventions.
Collapse
Affiliation(s)
- Mohammad Ebrahim Yarmohammadi
- Immunoregulation Research Center, Shahed University, Tehran, Islamic Republic of Iran; Department of Otolaryngology, Shahed University, Tehran, Islamic Republic of Iran
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Bellacen K, Kalay N, Ozeri E, Shahaf G, Lewis EC. Revascularization of pancreatic islet allografts is enhanced by α-1-antitrypsin under anti-inflammatory conditions. Cell Transplant 2012; 22:2119-33. [PMID: 23050776 DOI: 10.3727/096368912x657701] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Pancreatic islets are a highly vascularized entity, and their transplantation into diabetic individuals requires optimal revascularization. In addition, β-cells in islets are extremely sensitive to inflammation. α-1-Antitrypsin (AAT), a circulating serine-protease inhibitor that is available for clinical use as an affinity-purified human product, has been shown to protect islets from graft failure in mouse transplantation models and to achieve readily vascularized islet grafts. AAT is known to induce vascular endothelial growth factor (VEGF) expression and release, as well as protect from proteolytic cleavage of VEGF by elastase, promote viability of endothelial cells, and enhance migration of myocytes. Our aim was to examine whether AAT enhances vasculogenesis toward islet grafts. We employed Matrigel-islet plugs as means to introduce islets in an explantable isolated compartment and examined vessel formation, vessel maturation, and inflammatory profile of explants 9 days after implantation. Also, we examined primary epithelial cell grafts that were prepared from lungs of mice that are transgenic for human AAT. In addition, aortic ring sprouting assay was performed, and HUVEC tube formation assays were studied in the presence of AAT. Our findings indicate that islet grafts exhibit mature vessels in the presence of AAT, as demonstrated by morphology, as well as expression of endothelial CD31, smooth muscle actin (SMA), and von Willebrand factor (vWF). Epithelial cells that express human AAT achieved a similar positive outcome. Aortic ring sprouting was enhanced in AAT-treated cultures and also in cultures that contained primary epithelial cells from human AAT transgenic animals in the absence of added AAT. According to the tube formation assay, HUVECs exhibited superior responses in the presence of AAT. We conclude that vasculogenesis toward islet grafts is enhanced in the presence of AAT. Together with the remarkable safety profile of AAT, the study supports its use in the relevant clinical setups.
Collapse
Affiliation(s)
- Keren Bellacen
- Faculty of Health Sciences, Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | | | | | | |
Collapse
|
18
|
Lewis EC. Expanding the clinical indications for α(1)-antitrypsin therapy. Mol Med 2012; 18:957-70. [PMID: 22634722 DOI: 10.2119/molmed.2011.00196] [Citation(s) in RCA: 127] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Accepted: 05/16/2012] [Indexed: 12/13/2022] Open
Abstract
α(1)-Antitrypsin (AAT) is a 52-kDa circulating serine protease inhibitor. Production of AAT by the liver maintains 0.9-1.75 mg/mL circulating levels. During acute-phase responses, circulating AAT levels increase more than fourfold. In individuals with one of several inherited mutations in AAT, low circulating levels increase the risk for lung, liver and pancreatic destructive diseases, particularly emphysema. These individuals are treated with lifelong weekly infusions of human plasma-derived AAT. An increasing amount of evidence appears to suggest that AAT possesses not only the ability to inhibit serine proteases, such as elastase and proteinase-3 (PR-3), but also to exert antiinflammatory and tissue-protective effects independent of protease inhibition. AAT modifies dendritic cell maturation and promotes T regulatory cell differentiation, induces interleukin (IL)-1 receptor antagonist and IL-10 release, protects various cell types from cell death, inhibits caspases-1 and -3 activity and inhibits IL-1 production and activity. Importantly, unlike classic immunosuppressants, AAT allows undeterred isolated T-lymphocyte responses. On the basis of preclinical and clinical studies, AAT therapy for nondeficient individuals may interfere with disease progression in type 1 and type 2 diabetes, acute myocardial infarction, rheumatoid arthritis, inflammatory bowel disease, cystic fibrosis, transplant rejection, graft versus host disease and multiple sclerosis. AAT also appears to be antibacterial and an inhibitor of viral infections, such as influenza and human immunodeficiency virus (HIV), and is currently evaluated in clinical trials for type 1 diabetes, cystic fibrosis and graft versus host disease. Thus, AAT therapy appears to have advanced from replacement therapy, to a safe and potential treatment for a broad spectrum of inflammatory and immune-mediated diseases.
Collapse
Affiliation(s)
- Eli C Lewis
- Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| |
Collapse
|
19
|
Alpha-1 antitrypsin: a potent anti-inflammatory and potential novel therapeutic agent. Arch Immunol Ther Exp (Warsz) 2012; 60:81-97. [PMID: 22349104 DOI: 10.1007/s00005-012-0162-5] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Accepted: 11/23/2011] [Indexed: 12/29/2022]
Abstract
Alpha-1 antitrypsin (AAT) has long been thought of as an important anti-protease in the lung where it is known to decrease the destructive effects of major proteases such as neutrophil elastase. In recent years, the perception of this protein in this simple one dimensional capacity as an anti-protease has evolved and it is now recognised that AAT has significant anti-inflammatory properties affecting a wide range of inflammatory cells, leading to its potential therapeutic use in a number of important diseases. This present review aims to discuss the described anti-inflammatory actions of AAT in modulating key immune cell functions, delineate known signalling pathways and specifically to identify the models of disease in which AAT has been shown to be effective as a therapy.
Collapse
|
20
|
Thorel F, Damond N, Chera S, Wiederkehr A, Thorens B, Meda P, Wollheim CB, Herrera PL. Normal glucagon signaling and β-cell function after near-total α-cell ablation in adult mice. Diabetes 2011; 60:2872-82. [PMID: 21926270 PMCID: PMC3198058 DOI: 10.2337/db11-0876] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE To evaluate whether healthy or diabetic adult mice can tolerate an extreme loss of pancreatic α-cells and how this sudden massive depletion affects β-cell function and blood glucose homeostasis. RESEARCH DESIGN AND METHODS We generated a new transgenic model allowing near-total α-cell removal specifically in adult mice. Massive α-cell ablation was triggered in normally grown and healthy adult animals upon diphtheria toxin (DT) administration. The metabolic status of these mice was assessed in 1) physiologic conditions, 2) a situation requiring glucagon action, and 3) after β-cell loss. RESULTS Adult transgenic mice enduring extreme (98%) α-cell removal remained healthy and did not display major defects in insulin counter-regulatory response. We observed that 2% of the normal α-cell mass produced enough glucagon to ensure near-normal glucagonemia. β-Cell function and blood glucose homeostasis remained unaltered after α-cell loss, indicating that direct local intraislet signaling between α- and β-cells is dispensable. Escaping α-cells increased their glucagon content during subsequent months, but there was no significant α-cell regeneration. Near-total α-cell ablation did not prevent hyperglycemia in mice having also undergone massive β-cell loss, indicating that a minimal amount of α-cells can still guarantee normal glucagon signaling in diabetic conditions. CONCLUSIONS An extremely low amount of α-cells is sufficient to prevent a major counter-regulatory deregulation, both under physiologic and diabetic conditions. We previously reported that α-cells reprogram to insulin production after extreme β-cell loss and now conjecture that the low α-cell requirement could be exploited in future diabetic therapies aimed at regenerating β-cells by reprogramming adult α-cells.
Collapse
Affiliation(s)
- Fabrizio Thorel
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Nicolas Damond
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Simona Chera
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Andreas Wiederkehr
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Bernard Thorens
- Department of Physiology and Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Paolo Meda
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Claes B. Wollheim
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Pedro L. Herrera
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Corresponding author: Pedro L. Herrera,
| |
Collapse
|
21
|
Janciauskiene SM, Bals R, Koczulla R, Vogelmeier C, Köhnlein T, Welte T. The discovery of α1-antitrypsin and its role in health and disease. Respir Med 2011; 105:1129-39. [PMID: 21367592 DOI: 10.1016/j.rmed.2011.02.002] [Citation(s) in RCA: 246] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Revised: 01/24/2011] [Accepted: 02/07/2011] [Indexed: 01/08/2023]
Abstract
α1-Antitrypsin (AAT) is the archetype member of the serine protease inhibitor (SERPIN) supergene family. The AAT deficiency is most often associated with the Z mutation, which results in abnormal Z AAT folding in the endoplasmic reticulum of hepatocytes during biogenesis. This causes intra-cellular retention of the AAT protein rather than efficient secretion with consequent deficiency of circulating AAT. The reduced serum levels of AAT contribute to the development of chronic obstructive pulmonary disease (COPD) and the accumulation of abnormally folded AAT protein increases risk for liver diseases. In this review we show that with the discovery of AAT deficiency in the early 60s as a genetically determined predisposition to the development of early-onset emphysema, intensive investigations of enzymatic mechanisms that produce lung destruction in COPD were pursued. To date, the role of AAT in other than lung and liver diseases has not been extensively examined. Current findings provide new evidence that, in addition to protease inhibition, AAT expresses anti-inflammatory, immunomodulatory and antimicrobial properties, and highlight the importance of this protein in health and diseases. In this review co-occurrence of several diseases with AAT deficiency is discussed.
Collapse
|
22
|
Ris F, Lepetit-Coiffe M, Meda P, Crowe LA, Toso C, Armanet M, Niclauss N, Parnaud G, Giovannoni L, Bosco D, Morel P, Vallee JP, Berney T. Assessment of human islet labeling with clinical grade iron nanoparticles prior to transplantation for graft monitoring by MRI. Cell Transplant 2010; 19:1573-85. [PMID: 20719068 DOI: 10.3727/096368910x515863] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Ex vivo labeling of islets with superparamagnetic iron oxide (SPIO) nanoparticles allows posttransplant MRI imaging of the graft. In the present study, we compare two clinical grade SPIOs (ferucarbotran and ferumoxide) in terms of toxicity, islet cellular uptake, and MRI imaging. Human islets (80-90% purity) were incubated for 24 h with various concentrations of SPIOs (14-280 μg/ml of iron). Static incubations were performed, comparing insulin response to basal (2.8 mM) or high glucose stimulation (16.7 mM), with or without cAMP stimulation. Insulin and Perl's (assessment of iron content) staining were performed. Electronic microscopy analysis was performed. Labeled islets were used for in vitro or in vivo imaging in MRI 1.5T. Liver section after organ removal was performed in the same plane as MRI imaging to get a correlation between histology and radiology. Postlabeling islet viability (80 ± 10%) and function (in vitro static incubation and in vivo engraftment of human islets in nude mice) were similar in both groups. Iron uptake assessed by electron microscopy showed iron inclusions within the islets with ferucarbotran, but not with ferumoxide. MRI imaging (1.5T) of phantoms and of human islets transplanted in rats, demonstrated a strong signal with ferucarbotran, but only a weak signal with ferumoxide. Signal persisted for >8 weeks in the absence of rejection. An excellent correlation was observed between radiologic images and histology. The hepatic clearance of intraportally injected ferucarbotran was faster than that of ferumoxide, generating less background. A rapid signal decrease was observed in rejecting xenogeneic islets. According to the present data, ferucarbotran is the most appropriate of available clinical grade SPIOs for human islet imaging.
Collapse
Affiliation(s)
- Frederic Ris
- Cell Isolation and Transplantation Center, Geneva University Hospitals and University of Geneva, School of Medicine, 4 rue Gabrielle-Perret-Gentil, Geneva 14, Switzerland.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Bosco D, Armanet M, Morel P, Niclauss N, Sgroi A, Muller YD, Giovannoni L, Parnaud G, Berney T. Unique arrangement of alpha- and beta-cells in human islets of Langerhans. Diabetes 2010; 59:1202-10. [PMID: 20185817 PMCID: PMC2857900 DOI: 10.2337/db09-1177] [Citation(s) in RCA: 317] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE It is generally admitted that the endocrine cell organization in human islets is different from that of rodent islets. However, a clear description of human islet architecture has not yet been reported. The aim of this work was to describe our observations on the arrangement of human islet cells. RESEARCH DESIGN AND METHODS Human pancreas specimens and isolated islets were processed for histology. Sections were analyzed by fluorescence microscopy after immunostaining for islet hormones and endothelial cells. RESULTS In small human islets (40-60 mum in diameter), beta-cells had a core position, alpha-cells had a mantle position, and vessels laid at their periphery. In bigger islets, alpha-cells had a similar mantle position but were found also along vessels that penetrate and branch inside the islets. As a consequence of this organization, the ratio of beta-cells to alpha-cells was constantly higher in the core than in the mantle part of the islets, and decreased with increasing islet diameter. This core-mantle segregation of islet cells was also observed in type 2 diabetic donors but not in cultured isolated islets. Three-dimensional analysis revealed that islet cells were in fact organized into trilaminar epithelial plates, folded with different degrees of complexity and bordered by vessels on both sides. In epithelial plates, most beta-cells were located in a central position but frequently showed cytoplasmic extensions between outlying non-beta-cells. CONCLUSIONS Human islets have a unique architecture allowing all endocrine cells to be adjacent to blood vessels and favoring heterologous contacts between beta- and alpha-cells, while permitting homologous contacts between beta-cells.
Collapse
Affiliation(s)
- Domenico Bosco
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland.
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Casimir M, Lasorsa FM, Rubi B, Caille D, Palmieri F, Meda P, Maechler P. Mitochondrial glutamate carrier GC1 as a newly identified player in the control of glucose-stimulated insulin secretion. J Biol Chem 2009; 284:25004-14. [PMID: 19584051 DOI: 10.1074/jbc.m109.015495] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The SLC25 carrier family mediates solute transport across the inner mitochondrial membrane, a process that is still poorly characterized regarding both the mechanisms and proteins implicated. This study investigated mitochondrial glutamate carrier GC1 in insulin-secreting beta-cells. GC1 was cloned from insulin-secreting cells, and sequence analysis revealed hydropathy profile of a six-transmembrane protein, characteristic of mitochondrial solute carriers. GC1 was found to be expressed at the mRNA and protein levels in INS-1E beta-cells and pancreatic rat islets. Immunohistochemistry showed that GC1 was present in mitochondria, and ultrastructural analysis by electron microscopy revealed inner mitochondrial membrane localization of the transporter. Silencing of GC1 in INS-1E beta-cells, mediated by adenoviral delivery of short hairpin RNA, reduced mitochondrial glutamate transport by 48% (p < 0.001). Insulin secretion at basal 2.5 mM glucose and stimulated either by intermediate 7.5 mM glucose or non-nutrient 30 mM KCl was not modified by GC1 silencing. Conversely, insulin secretion stimulated with optimal 15 mM glucose was reduced by 23% (p < 0.005) in GC1 knocked down cells compared with controls. Adjunct of cell-permeant glutamate (5 mM dimethyl glutamate) fully restored the secretory response at 15 mM glucose (p < 0.005). Kinetics of insulin secretion were investigated in perifused isolated rat islets. GC1 silencing in islets inhibited the secretory response induced by 16.7 mM glucose, both during first (-25%, p < 0.05) and second (-33%, p < 0.05) phases. This study demonstrates that insulin-secreting cells depend on GC1 for maximal glucose response, thereby assigning a physiological function to this newly identified mitochondrial glutamate carrier.
Collapse
Affiliation(s)
- Marina Casimir
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva 4, Switzerland
| | | | | | | | | | | | | |
Collapse
|
25
|
Serre-Beinier V, Bosco D, Zulianello L, Charollais A, Caille D, Charpantier E, Gauthier BR, Diaferia GR, Giepmans BN, Lupi R, Marchetti P, Deng S, Buhler L, Berney T, Cirulli V, Meda P. Cx36 makes channels coupling human pancreatic beta-cells, and correlates with insulin expression. Hum Mol Genet 2009; 18:428-39. [PMID: 19000992 PMCID: PMC2638800 DOI: 10.1093/hmg/ddn370] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Previous studies have documented that the insulin-producing beta-cells of laboratory rodents are coupled by gap junction channels made solely of the connexin36 (Cx36) protein, and have shown that loss of this protein desynchronizes beta-cells, leading to secretory defects reminiscent of those observed in type 2 diabetes. Since human islets differ in several respects from those of laboratory rodents, we have now screened human pancreas, and islets isolated thereof, for expression of a variety of connexin genes, tested whether the cognate proteins form functional channels for islet cell exchanges, and assessed whether this expression changes with beta-cell function in islets of control and type 2 diabetics. Here, we show that (i) different connexin isoforms are differentially distributed in the exocrine and endocrine parts of the human pancreas; (ii) human islets express at the transcript level different connexin isoforms; (iii) the membrane of beta-cells harbors detectable levels of gap junctions made of Cx36; (iv) this protein is concentrated in lipid raft domains of the beta-cell membrane where it forms gap junctions; (v) Cx36 channels allow for the preferential exchange of cationic molecules between human beta-cells; (vi) the levels of Cx36 mRNA correlated with the expression of the insulin gene in the islets of both control and type 2 diabetics. The data show that Cx36 is a native protein of human pancreatic islets, which mediates the coupling of the insulin-producing beta-cells, and contributes to control beta-cell function by modulating gene expression.
Collapse
Affiliation(s)
| | - Domenico Bosco
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals, Geneva, Switzerland
| | - Laurence Zulianello
- Department of Cell Physiology and Metabolism, University of Geneva School of Medicine, CMU 1, rue Michel-Servet, 1211 Geneva 4, CH, Switzerland
| | - Anne Charollais
- Department of Cell Physiology and Metabolism, University of Geneva School of Medicine, CMU 1, rue Michel-Servet, 1211 Geneva 4, CH, Switzerland
| | - Dorothée Caille
- Department of Cell Physiology and Metabolism, University of Geneva School of Medicine, CMU 1, rue Michel-Servet, 1211 Geneva 4, CH, Switzerland
| | - Eric Charpantier
- Department of Cell Physiology and Metabolism, University of Geneva School of Medicine, CMU 1, rue Michel-Servet, 1211 Geneva 4, CH, Switzerland
| | - Benoit R. Gauthier
- Department of Cell Physiology and Metabolism, University of Geneva School of Medicine, CMU 1, rue Michel-Servet, 1211 Geneva 4, CH, Switzerland
| | - Giuseppe R. Diaferia
- Islet Research Laboratory, The Whittier Institute for Diabetes, University of California San Diego, La Jolla, CA, USA
| | - Ben N. Giepmans
- Department of Cell Biology, University of Groningen, Groningen, The Netherlands
| | - Roberto Lupi
- Department of Endocrinology and Metabolism, University of Pisa, Pisa, Italy
| | - Piero Marchetti
- Department of Endocrinology and Metabolism, University of Pisa, Pisa, Italy
| | - Shaoping Deng
- Department of Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Léo Buhler
- Surgical Research Unit, Department of Surgery
| | - Thierry Berney
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals, Geneva, Switzerland
| | - Vincenzo Cirulli
- Islet Research Laboratory, The Whittier Institute for Diabetes, University of California San Diego, La Jolla, CA, USA
| | - Paolo Meda
- Department of Cell Physiology and Metabolism, University of Geneva School of Medicine, CMU 1, rue Michel-Servet, 1211 Geneva 4, CH, Switzerland
| |
Collapse
|
26
|
Li G, Luo R, Zhang J, Yeo KS, Lian Q, Xie F, Tan EKW, Caille D, Kon OL, Salto-Tellez M, Meda P, Lim SK. Generating mESC-derived insulin-producing cell lines through an intermediate lineage-restricted progenitor line. Stem Cell Res 2009; 2:41-55. [DOI: 10.1016/j.scr.2008.07.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2008] [Revised: 07/25/2008] [Accepted: 07/26/2008] [Indexed: 10/21/2022] Open
|
27
|
Mesenchymal stem cells derived from human exocrine pancreas express transcription factors implicated in beta-cell development. Pancreas 2008; 37:75-84. [PMID: 18580448 DOI: 10.1097/mpa.0b013e31815fcb1e] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVES Transplantation of in vitro generated islets or insulin-producing cells represents an attractive option to overcome organ shortage. The aim of this study was to isolate, expand, and characterize cells from human exocrine pancreas and analyze their potential to differentiate into beta cells. METHODS Fibroblast-like cells growing out of human exocrine tissue were characterized by flow cytometry and by their capacity to differentiate into mesenchymal cell lineages. During cell expansion and after differentiation toward beta cells, expression of transcription factors of endocrine pancreatic progenitors was analyzed by reverse transcription polymerase chain reaction. RESULTS Cells emerged from 14/18 human pancreatic exocrine fractions and were expanded up to 40 population doublings. These cells displayed surface antigens similar to mesenchymal stem cells from bone marrow. A culture of these cells in adipogenic and chondrogenic differentiation media allowed differentiation into adipocyte- and chondrocyte-like cells. During expansion, cells expressed transcription factors implicated in islet development such as Isl1, Nkx2.2, Nkx6.1, nestin, Ngn3, Pdx1, and NeuroD. Activin A and hepatocyte growth factor induced an expression of insulin, glucagon, and glucokinase. CONCLUSIONS Proliferating cells with characteristics of mesenchymal stem cells and endocrine progenitors were isolated from exocrine tissue. Under specific conditions, these cells expressed little insulin. Human pancreatic exocrine tissue might thus be a source of endocrine cell progenitors.
Collapse
|
28
|
Zimmerli SC, Masson F, Cancela J, Meda P, Hauser C. Cutting edge: Lack of evidence for connexin-43 expression in human epidermal Langerhans cells. THE JOURNAL OF IMMUNOLOGY 2007; 179:4318-21. [PMID: 17878326 DOI: 10.4049/jimmunol.179.7.4318] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A provocative study has shown that viral peptides may be transferred in vitro from epithelial cells to APC through connexin-43 gap junction channels. In support of this cross-presentation pathway, the study also reported that human dendritic cells, including Langerhans cells of skin, express connexin-43. In this report we show that if this was the case, the levels of connexin-43 are below those detectable by immunofluorescence, flow cytometry, quantitative PCR of purified CD1a+ cells, and electron microscopy, raising questions about the relevance of the connexin-43-dependent mechanism for Langerhans cells of noninflamed human skin.
Collapse
Affiliation(s)
- Simone C Zimmerli
- Division of Immunology and Allergy, Department of Internal Medicine, Geneva University Hospitals and Medical School, Switzerland.
| | | | | | | | | |
Collapse
|
29
|
Schmid GM, Meda P, Caille D, Wargent E, O'Dowd J, Hochstrasser DF, Cawthorne MA, Sanchez JC. Inhibition of insulin secretion by betagranin, an N-terminal chromogranin A fragment. J Biol Chem 2007; 282:12717-24. [PMID: 17289672 DOI: 10.1074/jbc.m700788200] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Betagranin, an N-terminal fragment of chromogranin A, results from a proteolytic processing, and is co-secreted with insulin. While other chromogranin A-derived peptides negatively modulate hormone secretion, the role of betagranin in pancreatic beta-cells is so far unknown. We have recently shown that pancreatic islet betagranin levels are down-regulated in obese, leptin-deficient mice. In the present study, we have investigated the distribution of betagranin in primary mouse islets and cells of the MIN6 line and have evaluated its effects on insulin secretion. We showed that betagranin co-localizes with insulin within secretory granules and strongly inhibited insulin secretion in response to both glucose and potassium, by blocking the influx of calcium. The data demonstrated a hitherto unknown inhibitory effect of betagranin on insulin secretion.
Collapse
Affiliation(s)
- Gerhard M Schmid
- Biomedical Proteomics Research Group (BPRG), Department of Structural Biology and Bioinformatics, Geneva University Medical Center, CH-1211 Geneva 4, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Schmechel DE, Browndyke J, Ghio A. Strategies for dissecting genetic-environmental interactions in neurodegenerative disorders. Neurotoxicology 2006; 27:637-57. [PMID: 16870258 DOI: 10.1016/j.neuro.2006.05.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2006] [Revised: 05/18/2006] [Accepted: 05/20/2006] [Indexed: 01/22/2023]
Abstract
Complex genetic and environmental interactions contribute to abnormal aging and neurodegenerative disorders. We present information from a series of 1136 consecutive patients presenting with cognitive disorders and show possible significant contribution of toxic environmental and occupational exposures to pathological aging (21% of patients) and interactions of these exposures with common polymorphisms that affect cell injury and inflammation. Such exposures may lower age of onset to same degree as APOE4/4. Common polymorphisms in apolipoprotein E (APOE), hemochromatosis gene (Hfe) and alpha-1-antitrypsin (AAT) are present in up to 40+% of patients and may partially account for differences in clinical syndrome, age of onset and rate of progression. Strategies for the study of these disorders must also consider the role and treatment of common co-morbid illnesses such as alcohol use, nutritional deficiencies, sleep disorders, and pre-existing affective disorder. APOE, Hfe, and AAT genes are expressed in liver tissue and in macrophages and are involved in the host innate immune response to stress, inflammation and infections. Hfe and AAT are involved in iron metabolism and their polymorphisms may contribute to hepatosteatosis and altered homeostasis of lipids (role of APOE), iron, and trace minerals. Some of these responses may be adaptive. Hfe and AAT modulate the apparent effects of toxic exposures on age of onset and progression rate. C282Y polymorphism paradoxically reverses APOE4/4 effect on age of onset. S and Z AAT polymorphisms may attenuate earlier age of onset in persons with toxic or environmental exposure. AAT S or Z polymorphisms are present in 25% of persons with anxiety disorder and 42% of persons with bipolar disorder compared to 10% of control group without pre-existing affective disorder. Common genetic polymorphisms that affect the response to inflammation and cell injury provide a beginning strategy for dissecting neurodegenerative disorders. The effects of APOE, Hfe, and AAT on glucose, lipid, iron and trace mineral homeostasis may affect normal development and aging of the nervous system in addition to their effects on outcome of toxic environmental and occupational exposures and susceptibility and outcome of neurodegenerative illnesses.
Collapse
Affiliation(s)
- Donald E Schmechel
- Joseph and Kathleen Bryan Alzheimer Disease Research Center, Department of Medicine (Neurology), Duke University Medical Center, Durham, NC 27710, USA.
| | | | | |
Collapse
|
31
|
Olsson R, Maxhuni A, Carlsson PO. Revascularization of Transplanted Pancreatic Islets Following Culture with Stimulators of Angiogenesis. Transplantation 2006; 82:340-7. [PMID: 16906031 DOI: 10.1097/01.tp.0000229418.60236.87] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Insufficient revascularization of transplanted islets may result in chronic hypoxia and loss of islet function. This study investigated whether simple culture of islets with angiogenic substances before transplantation could improve graft revascularization. METHODS Mouse islets were cultured with vascular endothelial growth factor (VEGF; 20 ng/ml), fibroblast growth factor 2 (FGF-2; 20 ng/ml) or matrix metalloproteinase 9 (MMP-9; 1 mug/ml). Thereafter, 250 islets were implanted beneath the renal capsule of syngeneic C57Bl/6 mice. One month posttransplantation, blood flow (laser-Doppler flowmetry), oxygen tension (Clark microelectrodes), and vascular density were measured and correlated to graft function. RESULTS Treatment of islets with VEGF during culture caused islet blood vessels to dilate, whereas FGF-2 treatment induced endothelial cell proliferation. However, the number of capillaries in both cases decreased during culture. When investigated one month posttransplantation, both VEGF and FGF-2 pretreated islets had similar or worse vascular engraftment when compared to transplanted control islets. MMP-9 pretreatment of islets increased vascular density, blood flow and oxygen tension within the grafts. Animals receiving MMP-9 pretreated islets returned, however, more slowly to normoglycemia than control animals, and performed worse than controls in a glucose tolerance test one month posttransplantation. CONCLUSIONS Treatment of islets during culture with VEGF or FGF-2 changed the islet vascular phenotype, but capillaries were still lost. Notably, the number of capillaries in the grafted islets one month posttransplantation was in all cases strikingly similar to that observed prior to transplantation. MMP-9 pretreatment of islets elicited an angiogenic response, which improved revascularization of the transplanted islets.
Collapse
Affiliation(s)
- Richard Olsson
- Department of Medical Cell Biology, Uppsala University, Sweden.
| | | | | |
Collapse
|