1
|
Shao Y, Zhang Y, Zou S, Wang J, Li X, Qin M, Sun L, Yin W, Chang X, Wang S, Han X, Wu T, Chen F. (-)-Epigallocatechin 3-gallate protects pancreatic β-cell against excessive autophagy-induced injury through promoting FTO degradation. Autophagy 2024; 20:2460-2477. [PMID: 38910554 PMCID: PMC11572200 DOI: 10.1080/15548627.2024.2370751] [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: 11/09/2023] [Revised: 06/08/2024] [Accepted: 06/17/2024] [Indexed: 06/25/2024] Open
Abstract
Excessive macroautophagy/autophagy leads to pancreatic β-cell failure that contributes to the development of diabetes. Our previous study proved that the occurrence of deleterious hyperactive autophagy attributes to glucolipotoxicity-induced NR3C1 activation. Here, we explored the potential protective effects of (-)-epigallocatechin 3-gallate (EGCG) on β-cell-specific NR3C1 overexpression mice in vivo and NR3C1-enhanced β cells in vitro. We showed that EGCG protects pancreatic β cells against NR3C1 enhancement-induced failure through inhibiting excessive autophagy. RNA demethylase FTO (FTO alpha-ketoglutarate dependent dioxygenase) caused diminished m6A modifications on mRNAs of three pro-oxidant genes (Tlr4, Rela, Src) and, hence, oxidative stress occurs; by contrast, EGCG promotes FTO degradation by the ubiquitin-proteasome system in NR3C1-enhanced β cells, which alleviates oxidative stress, and thereby prevents excessive autophagy. Moreover, FTO overexpression abolishes the beneficial effects of EGCG on β cells against NR3C1 enhancement-induced damage. Collectively, our results demonstrate that EGCG protects pancreatic β cells against NR3C1 enhancement-induced excessive autophagy through suppressing FTO-stimulated oxidative stress, which provides novel insights into the mechanisms for the anti-diabetic effect of EGCG.Abbreviation 3-MA: 3-methyladenine; AAV: adeno-associated virus; Ad: adenovirus; ALD: aldosterone; AUC: area under curve; βNR3C1 mice: pancreatic β-cell-specific NR3C1 overexpression mice; Ctrl: control; CHX: cycloheximide; DEX: dexamethasone; DHE: dihydroethidium; EGCG: (-)-epigallocatechin 3-gallate; FTO: FTO alpha-ketoglutarate dependent dioxygenase; GSIS: glucose-stimulated insulin secretion; HFD: high-fat diet; HG: high glucose; i.p.: intraperitoneal; IOD: immunofluorescence optical density; KSIS: potassium-stimulated insulin secretion; m6A: N6-methyladenosine; MeRIP-seq: methylated RNA immunoprecipitation sequencing; NO: nitric oxide; NR3C1/GR: nuclear receptor subfamily 3, group C, member 1; NR3C1-Enhc.: NR3C1-enhancement; NAC: N-acetylcysteine; NC: negative control; PBS: phosphate-buffered saline; PI: propidium iodide; OCR: oxygen consumption rate; Palm.: palmitate; RELA: v-rel reticuloendotheliosis viral oncogene homolog A (avian); RNA-seq: RNA sequencing; O2.-: superoxide anion; SRC: Rous sarcoma oncogene; ROS: reactive oxygen species; T2D: type 2 diabetes; TEM: transmission electron microscopy; TLR4: toll-like receptor 4; TUNEL: terminal dUTP nick-end labeling; UTR: untranslated region; WT: wild-type.
Collapse
Affiliation(s)
- Yixue Shao
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuhan Zhang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Suyun Zou
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jianan Wang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xirui Li
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Miaozhen Qin
- Jiangsu Province Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Liangjun Sun
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wenyue Yin
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaoai Chang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shusen Wang
- Organ Transplant Center, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Xiao Han
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Tijun Wu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Fang Chen
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
2
|
Langlois A, Pinget M, Kessler L, Bouzakri K. Islet Transplantation: Current Limitations and Challenges for Successful Outcomes. Cells 2024; 13:1783. [PMID: 39513890 PMCID: PMC11544954 DOI: 10.3390/cells13211783] [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: 09/26/2024] [Revised: 10/22/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Islet transplantation is a promising approach for treating patients with unstable T1DM. However, it is confronted with numerous obstacles throughout the various stages of the transplantation procedure. Significant progress has been made over the last 25 years in understanding the mechanisms behind the loss of functional islet mass and in developing protective strategies. Nevertheless, at present, two to three pancreases are still needed to treat a single patient, which limits the maximal number of patients who can benefit from islet transplantation. Thus, this publication provides an overview of recent scientific findings on the various issues affecting islet transplantation. Specifically, we will focus on the understanding of the mechanisms involved and the strategies developed to alleviate these problems from the isolation stage to the post-transplantation phase. Finally, we hope that this review will highlight new avenues of action, enabling us to propose pancreatic islet transplantation to a maximum number of patients with T1DM.
Collapse
Affiliation(s)
- Allan Langlois
- UR «Diabète et Thérapeutiques», Centre Européen d’Étude du Diabète, Université de Strasbourg, Boulevard René Leriche, 67200 Strasbourg, France; (A.L.); (M.P.)
| | - Michel Pinget
- UR «Diabète et Thérapeutiques», Centre Européen d’Étude du Diabète, Université de Strasbourg, Boulevard René Leriche, 67200 Strasbourg, France; (A.L.); (M.P.)
| | - Laurence Kessler
- Department of Endocrinology, Diabetes and Nutrition, University Hospital of Strasbourg, 67200 Strasbourg, France;
- Inserm UMR 1260, Nanomédicine Regenerative, University of Strasbourg, 67085 Strasbourg, France
| | - Karim Bouzakri
- UR «Diabète et Thérapeutiques», Centre Européen d’Étude du Diabète, Université de Strasbourg, Boulevard René Leriche, 67200 Strasbourg, France; (A.L.); (M.P.)
| |
Collapse
|
3
|
Bochenek MA, Walters B, Zhang J, Fenton OS, Facklam A, Kroneková Z, Pelach M, Engquist EN, Leite NC, Morgart A, Lacík I, Langer R, Anderson DG. Enhancing the Functionality of Immunoisolated Human SC-βeta Cell Clusters through Prior Resizing. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307464. [PMID: 38212275 DOI: 10.1002/smll.202307464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 12/10/2023] [Indexed: 01/13/2024]
Abstract
The transplantation of immunoisolated stem cell derived beta cell clusters (SC-β) has the potential to restore physiological glycemic control in patients with type I diabetes. This strategy is attractive as it uses a renewable β-cell source without the need for systemic immune suppression. SC-β cells have been shown to reverse diabetes in immune compromised mice when transplanted as ≈300 µm diameter clusters into sites where they can become revascularized. However, immunoisolated SC-β clusters are not directly revascularized and rely on slower diffusion of nutrients through a membrane. It is hypothesized that smaller SC-β cell clusters (≈150 µm diameter), more similar to islets, will perform better within immunoisolation devices due to enhanced mass transport. To test this, SC-β cells are resized into small clusters, encapsulated in alginate spheres, and coated with a biocompatible A10 polycation coating that resists fibrosis. After transplantation into diabetic immune competent C57BL/6 mice, the "resized" SC-β cells plus the A10 biocompatible polycation coating induced long-term euglycemia in the mice (6 months). After retrieval, the resized A10 SC-β cells exhibited the least amount of fibrosis and enhanced markers of β-cell maturation. The utilization of small SC-β cell clusters within immunoprotection devices may improve clinical translation in the future.
Collapse
Affiliation(s)
- Matthew A Bochenek
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Ave, Boston, MA, 02115, USA
| | - Ben Walters
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Ave, Boston, MA, 02115, USA
| | - Jingping Zhang
- Harvard University, 7 Divinity Avenue, Cambridge, MA, 02138, USA
| | - Owen S Fenton
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Amanda Facklam
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Zuzana Kroneková
- Department for Biomaterials Research, Polymer Institute of the Slovak Academy of Sciences, Dubravska cesta 9, Bratislava, 845 41, Slovakia
| | - Michal Pelach
- Department for Biomaterials Research, Polymer Institute of the Slovak Academy of Sciences, Dubravska cesta 9, Bratislava, 845 41, Slovakia
| | - Elise N Engquist
- Harvard University, 7 Divinity Avenue, Cambridge, MA, 02138, USA
| | - Nayara C Leite
- Harvard University, 7 Divinity Avenue, Cambridge, MA, 02138, USA
| | - Alex Morgart
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Ave, Boston, MA, 02115, USA
| | - Igor Lacík
- Department for Biomaterials Research, Polymer Institute of the Slovak Academy of Sciences, Dubravska cesta 9, Bratislava, 845 41, Slovakia
| | - Robert Langer
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Ave, Boston, MA, 02115, USA
- Division of Health Science Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Daniel G Anderson
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Ave, Boston, MA, 02115, USA
- Division of Health Science Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| |
Collapse
|
4
|
Zhou R, Hu J, Qiu J, Lu S, Lin H, Huang R, Zhou S, Huang G, He J. Phenolic compound SG-1 from Balanophora harlandii and its derivatives exert anti-influenza A virus activity via activation of the Nrf2/HO-1 pathway. Biochem Pharmacol 2023; 210:115495. [PMID: 36918045 DOI: 10.1016/j.bcp.2023.115495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 03/03/2023] [Accepted: 03/06/2023] [Indexed: 03/14/2023]
Abstract
Influenza A virus (IAV) is one of the leading causes of respiratory illness and continues to cause pandemics around the world. Against this backdrop, drug resistance poses a challenge to existing antiviral drugs, and hence, there is an urgent need for developing new antiviral drugs. In this study, we obtained a phenolic compound SG-7, a derivative of natural compound 2-hydroxymethyl-1,4-hydroquinone, which exhibits inhibitory activity toward a panel of influenza viruses and has low cellular toxicity. Mechanistic studies have shown that SG-7 exerts its anti-IAV properties by acting on the virus itself and modulating host signaling pathways. Namely, SG-7 targets the HA2 subunit of hemagglutinin (HA) to block the fusion of viral-cellular membranes and inhibits IAV-induced oxidative stress and overexpression of pro-inflammatory factors by activating the Nrf2/HO-1 pathway and reducing NF-κB activation. In addition, SG-7 can enhance type I IFN antiviral response by inducing Nrf2 expression. Importantly, SG-7 showed the ability to inhibit viral replication in the lungs of IAV-infected mice and reduce their mortality. Therefore, SG-7 may be a promising lead compound for anti-influenza drug development.
Collapse
Affiliation(s)
- Runhong Zhou
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Jianan Hu
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Jingnan Qiu
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Shengsheng Lu
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Haixing Lin
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Ruifeng Huang
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Shaofen Zhou
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Guoqing Huang
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Jian He
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China.
| |
Collapse
|
5
|
Arleevskaya M, Takha E, Petrov S, Kazarian G, Renaudineau Y, Brooks W, Larionova R, Korovina M, Valeeva A, Shuralev E, Mukminov M, Kravtsova O, Novikov A. Interplay of Environmental, Individual and Genetic Factors in Rheumatoid Arthritis Provocation. Int J Mol Sci 2022; 23:ijms23158140. [PMID: 35897715 PMCID: PMC9329780 DOI: 10.3390/ijms23158140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 02/05/2023] Open
Abstract
In this review, we explore systemization of knowledge about the triggering effects of non-genetic factors in pathogenic mechanisms that contribute to the development of rheumatoid arthritis (RA). Possible mechanisms involving environmental and individual factors in RA pathogenesis were analyzed, namely, infections, mental stress, sleep deprivation ecology, age, perinatal and gender factors, eating habits, obesity and smoking. The non-genetic factors modulate basic processes in the body with the impact of these factors being non-specific, but these common challenges may be decisive for advancement of the disease in the predisposed body at risk for RA. The provocation of this particular disease is associated with the presence of congenital loci minoris resistentia. The more frequent non-genetic factors form tangles of interdependent relationships and, thereby, several interdependent external factors hit one vulnerable basic process at once, either provoking or reinforcing each other. Understanding the specific mechanisms by which environmental and individual factors impact an individual under RA risk in the preclinical stages can contribute to early disease diagnosis and, if the factor is modifiable, might be useful for the prevention or delay of its development.
Collapse
Affiliation(s)
- Marina Arleevskaya
- Central Research Laboratory, Kazan State Medical Academy, 420012 Kazan, Russia; (E.T.); (S.P.); (G.K.); (R.L.); (M.K.); (A.V.); (E.S.); (M.M.)
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia;
- Correspondence: ; Tel.: +7-89172-886-679; Fax: +7-843-238-5413
| | - Elena Takha
- Central Research Laboratory, Kazan State Medical Academy, 420012 Kazan, Russia; (E.T.); (S.P.); (G.K.); (R.L.); (M.K.); (A.V.); (E.S.); (M.M.)
| | - Sergey Petrov
- Central Research Laboratory, Kazan State Medical Academy, 420012 Kazan, Russia; (E.T.); (S.P.); (G.K.); (R.L.); (M.K.); (A.V.); (E.S.); (M.M.)
- Institute of Environmental Sciences, Kazan (Volga Region) Federal University, 420008 Kazan, Russia
| | - Gevorg Kazarian
- Central Research Laboratory, Kazan State Medical Academy, 420012 Kazan, Russia; (E.T.); (S.P.); (G.K.); (R.L.); (M.K.); (A.V.); (E.S.); (M.M.)
| | - Yves Renaudineau
- Department of Immunology, CHU Toulouse, INSERM U1291, CNRS U5051, University Toulouse IIII, 31000 Toulouse, France;
| | - Wesley Brooks
- Department of Chemistry, University of South Florida, Tampa, FL 33620, USA;
| | - Regina Larionova
- Central Research Laboratory, Kazan State Medical Academy, 420012 Kazan, Russia; (E.T.); (S.P.); (G.K.); (R.L.); (M.K.); (A.V.); (E.S.); (M.M.)
| | - Marina Korovina
- Central Research Laboratory, Kazan State Medical Academy, 420012 Kazan, Russia; (E.T.); (S.P.); (G.K.); (R.L.); (M.K.); (A.V.); (E.S.); (M.M.)
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia;
| | - Anna Valeeva
- Central Research Laboratory, Kazan State Medical Academy, 420012 Kazan, Russia; (E.T.); (S.P.); (G.K.); (R.L.); (M.K.); (A.V.); (E.S.); (M.M.)
| | - Eduard Shuralev
- Central Research Laboratory, Kazan State Medical Academy, 420012 Kazan, Russia; (E.T.); (S.P.); (G.K.); (R.L.); (M.K.); (A.V.); (E.S.); (M.M.)
- Institute of Environmental Sciences, Kazan (Volga Region) Federal University, 420008 Kazan, Russia
| | - Malik Mukminov
- Central Research Laboratory, Kazan State Medical Academy, 420012 Kazan, Russia; (E.T.); (S.P.); (G.K.); (R.L.); (M.K.); (A.V.); (E.S.); (M.M.)
- Institute of Environmental Sciences, Kazan (Volga Region) Federal University, 420008 Kazan, Russia
| | - Olga Kravtsova
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia;
| | - Andrey Novikov
- Mathematical Center, Sobolev Instiute of Mathematics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia;
| |
Collapse
|
6
|
Kim MJ, Hwang YH, Hwang JW, Alam Z, Lee DY. Heme oxygenase-1 gene delivery for altering high mobility group box-1 protein in pancreatic islet. J Control Release 2022; 343:326-337. [PMID: 35085698 DOI: 10.1016/j.jconrel.2022.01.031] [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: 10/24/2021] [Revised: 01/14/2022] [Accepted: 01/19/2022] [Indexed: 10/19/2022]
Abstract
Pancreatic islet transplantation is a promising strategy for the treatment of type I diabetes. High-mobility group box-1 (HMGB1), highly expressed in islet cells, is a potent immune stimulator in immune rejection. Heme oxygenase-1 (HO1) gene therapy can modulate the release of HMGB1 by altering intracellular molecules for successful cell transplantation. After delivery of the heme oxygenase-1 (HO1) gene to islet cells using an adeno-associated viral vector (AAV), it was evaluated the changes in cytoplasmic Ca2+ ions and calcineurin activity as well as histone acetyltransferase (HAT) and Poly(ADP) ribose polymerase-1 (PARP-1). Inhibition of HMGB1 release was evaluated through altering these intracellular molecules. Then, after transplantation of HO1-transduced islets, the therapeutic effect of them was evaluated through measuring blood glucose level to diabetic mice and through immunohistochemical analysis. The transduced HO1 gene significantly inhibited HMGB1 release in islets that was under the cell damage by hypoxia exposure. It was confirmed that this result was initially due to the decrease in cytoplasmic Ca2+ ion concentration and calcineurin activity. In addition, the delivered HO1 gene simultaneously reduced the activity of HAT and PARP-1, which are involved in the translocation of HMGB1 from the nucleus to the cytoplasm. As a result, when the HO1 gene-transduced islets were transplanted into diabetic mice, the treatment efficiency of diabetes was effectively improved by increasing the survival rate of the islets. Collectively, these results suggest that HO1 gene transfer can be used for successful islet transplantation by altering the activity of intracellular signal molecules and reducing HMGB1 release.
Collapse
Affiliation(s)
- Min Jun Kim
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul 04763, Republic of Korea
| | - Yong Hwa Hwang
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul 04763, Republic of Korea
| | - Jin Wook Hwang
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul 04763, Republic of Korea
| | - Zahid Alam
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul 04763, Republic of Korea
| | - Dong Yun Lee
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul 04763, Republic of Korea; Institute of Nano Science & Technology (INST), Hanyang University, Seoul 04763, Republic of Korea; Elixir Pharmatech Inc., Seoul 04763, Republic of Korea.
| |
Collapse
|
7
|
Verheij M, Zeerleder S, Voermans C. Heme oxygenase-1: Equally important in allogeneic hematopoietic stem cell transplantation and organ transplantation? Transpl Immunol 2021; 68:101419. [PMID: 34089821 DOI: 10.1016/j.trim.2021.101419] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 05/30/2021] [Accepted: 05/31/2021] [Indexed: 12/17/2022]
Abstract
The intracellular enzyme heme oxygenase-1 (HO-1) is responsible for the degradation of cell-free (cf) heme. Cfheme, released upon cell damage and cell death from hemoglobin, mitochondria and myoglobin, functions as a powerful damage-associated molecular pattern (DAMP). Indeed, cfheme plays a role in a myriad of diseases characterized by (systemic) inflammation, and its rapid degradation by HO-1 is pivotal to maintain homeostasis. In the past decade, HO-1 has been extensively studied for its potential protective role in different transplantation settings, including allogeneic hematopoietic stem cell transplantation (HSCT), solid organ transplantation and pancreatic islet transplantation. These studies have shown that HO-1 can be induced by a wide range of molecules, and that induction of HO-1 has the potential to significantly reduce the incidence and severity of transplantation-related complications such as graft-versus-host disease (GvHD) and ischemia/reperfusion injury (IRI). As such, further investigation into the use of HO-1-inducing agents in human transplantation settings to facilitate the potential use of these agents in the clinic is warranted. In this review, we summarize the literature of the past 10 years on the role of HO-1 in allogeneic HSCT, solid organ transplantation (focusing on kidney and liver) and pancreatic islet transplantation. Furthermore, we provide a hypothesis about the way that HO-1 is able to provide protection against acute GvHD after allogeneic HSCT. A total of 48 research articles and 17 review articles were included in this review.
Collapse
Affiliation(s)
- Myrddin Verheij
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands
| | - Sacha Zeerleder
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Switzerland; Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands; Department for Biomedical Research, University of Bern, Switzerland
| | - Carlijn Voermans
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands.
| |
Collapse
|
8
|
Li Y, Hou JG, Liu Z, Gong XJ, Hu JN, Wang YP, Liu WC, Lin XH, Wang Z, Li W. Alleviative effects of 20(R)-Rg3 on HFD/STZ-induced diabetic nephropathy via MAPK/NF-κB signaling pathways in C57BL/6 mice. JOURNAL OF ETHNOPHARMACOLOGY 2021; 267:113500. [PMID: 33091499 DOI: 10.1016/j.jep.2020.113500] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 10/14/2020] [Accepted: 10/15/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Diabetic nephropathy (DN) is a major complication of diabetes. The kidney disease develops in nearly 20%-40% of type 2 diabetes (T2D) patients. Ginseng is the root of Panax ginseng C. A. Meyer and has been used in prevention and treatment of diseases for more than 2000 years as a traditional oriental medicine. The 20(R)-ginsenoside Rg3, an active saponin isolated from ginseng, can prevent and treat many diseases. The object of this research was to explore the alleviative effects of 20(R)-Rg3 on DN in mice. MATERIALS AND METHODS The T2D animal model was induced by continuous access to a high fat diet (HFD) combined with a single injection of 100 mg/kg streptozotocin (STZ) in C57BL/6 mice. The mice were treated by oral gavage of the 20(R)-Rg3 (10, 20 mg/kg) for 8 weeks. Functional and histopathological analyses of the kidneys were then performed. Protein expression levels of MAPKs and NF-κB signal pathways in the kidney were evaluated by western blotting. The expressions of HO-1 and NF-κB in the kidney were measured by fluorescent labeling staining. Other assessments including fasting blood glucose (FBG) levels, blood lipids, oxidative indicators, and inflammatory factors were all performed. RESULTS Abnormally elevated FBG levels were observed in HFD/STZ mice, contributing significantly to the occurrence of DN. Simultaneously, HFD/STZ mice showed the rise of serum total cholesterol (TC), triglyceride (TG), and low-density lipoprotein cholesterol (LDL-C) levels, and the decrease in high density lipoprotein cholesterol (HDL-C). DN was evidenced by the overproduction of malondialdehyde (MDA), decreased levels of superoxide dismutase (SOD) and catalase (CAT) enzymatic activities, high levels of serum blood urea nitrogen (BUN) and creatinine (Cr). Simultaneously, the results of the immunofluorescence assay showed an increased expression level in NF-κB p65 while a decrease in antioxidant enzyme HO-1 was observed. Herein, 20(R)-Rg3 treatment for 8 weeks not only attenuated FBG levels and advanced glycation end products (AGEs) levels but also improved insulin (INS) level, blood lipids, oxidative stress, and renal function by regulating MAPKs and NF-κB signal pathways in DN mice. CONCLUSION Taken together, the findings from the present study explicitly confirmed that 20(R)-Rg3 exerted ameliorative effects on DN mice via improving anti-oxidative activity and reducing renal inflammation.
Collapse
Affiliation(s)
- Ying Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China; National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun, 130118, China
| | - Jin-Gang Hou
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China; Intelligent Synthetic Biology Center, Daejeon, 34141, Republic of Korea
| | - Zhi Liu
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China; National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun, 130118, China
| | - Xiao-Jie Gong
- College of Life Science, Dalian University, Dalian, 116600, China
| | - Jun-Nan Hu
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China; National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun, 130118, China
| | - Ying-Ping Wang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China; National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun, 130118, China
| | - Wen-Cong Liu
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China; National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun, 130118, China
| | - Xiang-Hui Lin
- Liaoning Xifeng Pharmaceutical Group Co., Ltd., Huanren, 117000, China
| | - Zi Wang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China; National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun, 130118, China.
| | - Wei Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China; National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun, 130118, China.
| |
Collapse
|
9
|
Haga J, Sato N, Anazawa T, Kimura T, Kenjo A, Gotoh M, Marubashi S. Comprehensive analysis of gene expression of isolated pancreatic islets during pretransplant culture. Fukushima J Med Sci 2021; 67:17-26. [PMID: 33597316 PMCID: PMC8075558 DOI: 10.5387/fms.2020-25] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background: The aim of this study was to investigate the effect of pretransplant culture on the survival of pancreatic islet grafts, and to determine the biological characteristics of isolated islets during pretransplant culture. Methods: The survival of islets from Wistar rats, transplanted to diabetic C57BL/B6 mice, was compared between fresh islets and cultured islets. A comprehensive gene expression analysis was employed to investigate biological processes during pretransplant culture, and in vitro validation studies were performed. Results: Survival of cultured xenografts was significantly prolonged as compared to that of fresh islets (fresh: 12.5 ± 1.9 days, 1-day cultured: 16.0 ± 1.3 days (p= 0.017), 3-day cultured: 17.0 ± 2.6 days (p= 0.014)). Comprehensive gene expression analysis identified significant upregulation of annotated functions associated with inflammation in cultured groups. Six proinflammatory genes, including heme oxygenase 1 (HO-1) and IL-6, were significantly upregulated during culture. Validation studies revealed significantly higher levels of IL-6 in the supernatant of cultured islets and HO-1 in the cultured islets when compared with fresh islets. Conclusion: Transplantation of cultured islets induced significant but minimal prolongation of graft survival in xenogeneic combinations. Comprehensive analysis of gene expression in cultured islets showed biological processes associated with proinflammation during culture.
Collapse
Affiliation(s)
- Junichiro Haga
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Fukushima Medical University
| | - Naoya Sato
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Fukushima Medical University
| | - Takayuki Anazawa
- Department of Surgery, Graduate School of Medicine, Kyoto University
| | - Takashi Kimura
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Fukushima Medical University
| | - Akira Kenjo
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Fukushima Medical University
| | - Mitsukazu Gotoh
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Fukushima Medical University
| | - Shigeru Marubashi
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Fukushima Medical University
| |
Collapse
|
10
|
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
|
11
|
Saravanan PB, Vasu S, Yoshimatsu G, Darden CM, Wang X, Gu J, Lawrence MC, Naziruddin B. Differential expression and release of exosomal miRNAs by human islets under inflammatory and hypoxic stress. Diabetologia 2019; 62:1901-1914. [PMID: 31372667 DOI: 10.1007/s00125-019-4950-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 05/29/2019] [Indexed: 01/24/2023]
Abstract
AIMS/HYPOTHESIS Pancreatic islets produce non-coding microRNAs (miRNAs) that regulate islet cell function and survival. Our earlier investigations revealed that human islets undergo significant damage due to various types of stresses following transplantation and release miRNAs. Here, we sought to identify and validate exosomal miRNAs (exo-miRNAs) produced by human islets under conditions of cellular stress, preceding loss of cell function and death. We also aimed to identify islet stress signalling pathways targeted by exo-miRNAs to elucidate potential regulatory roles in islet cell stress. METHODS Human islets were subjected to proinflammatory cytokine and hypoxic cell stress and miRNA from exosomes was isolated for RNA sequencing and analysis. Stress-induced exo-miRNAs were evaluated for kinetics of expression and release by intact islets for up to 48 h exposure to cytokines and hypoxia. A subset of stress-induced exo-miRNAs were assessed for recovery and detection as biomarkers of islet cell stress in a diabetic nude mouse xenotransplant model and in patients undergoing total pancreatectomy with islet auto-transplantation (TPIAT). Genes and signalling pathways targeted by stress-induced exo-miRNAs were identified by Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis and direct interactions of miRNAs with downstream signalling targets were validated in human islet cells using the miRNA Tests for Read Analysis and Prediction (MirTrap) system. RESULTS Global exo-miRNA sequencing revealed that 879 miRNA species were released from human islets and 190 islet exo-miRNAs were differentially expressed in response to proinflammatory cytokines, hypoxia or both. Release of exo-miRNAs hsa-miR-29b-3p and hsa-miR-216a-5p was detected within 6 h of exposure to cytokines and hypoxia. The remaining subset of stress-induced exo-miRNAs, including hsa-miR-148a-3p and islet cell damage marker hsa-miR-375, showed delayed release at 24-48 h, correlating with apoptosis and cell death. Stress and damage exo-miRNAs were significantly elevated in the circulation in human-to-mouse xenotransplant models and in human transplant recipients. Elevated blood exo-miRNAs negatively correlated with post-transplant islet function based on comparisons of stress and damage exo-miRNA indices with Secretory Unit of Islet Transplant Objects (SUITO) indices. KEGG analysis and further validation of exo-miRNA targets by MirTrap analysis revealed significant enrichment of islet mRNAs involved in phosphoinositide 3-kinase/Akt and mitogen-activated protein kinase signalling pathways. CONCLUSIONS/INTERPRETATION The study identifies exo-miRNAs differentially expressed and released by islets in response to damage and stress. These exo-miRNAs could serve as potential biomarkers for assessing islet damage and predicting outcomes in islet transplantation. Notably, exo-miRNAs 29b-3p and 216a-5p could be detected in islets prior to damage-released miRNAs and indicators of cellular apoptosis and death. Thus, these stress-induced exo-miRNAs may have potential diagnostic value for detecting early islet stress prior to progressive loss of islet cell mass and function. Further investigations are warranted to investigate the utility of these exo-miRNAs as early indicators of islet cell stress during prediabetic conditions.
Collapse
Affiliation(s)
- Prathab Balaji Saravanan
- Division of Transplantation, Department of Surgery, Virginia Commonwealth University, Medical Center, Richmond, VA, USA
| | - Srividya Vasu
- Islet Cell Laboratory, Baylor Scott and White Research Institute, 3434 Live Oak Street, Dallas, TX, 75204, USA
| | - Gumpei Yoshimatsu
- Islet Cell Laboratory, Baylor Scott and White Research Institute, 3434 Live Oak Street, Dallas, TX, 75204, USA
| | - Carly M Darden
- Islet Cell Laboratory, Baylor Scott and White Research Institute, 3434 Live Oak Street, Dallas, TX, 75204, USA
| | - Xuan Wang
- Islet Cell Laboratory, Baylor Scott and White Research Institute, 3434 Live Oak Street, Dallas, TX, 75204, USA
| | - Jinghua Gu
- Islet Cell Laboratory, Baylor Scott and White Research Institute, 3434 Live Oak Street, Dallas, TX, 75204, USA
| | - Michael C Lawrence
- Islet Cell Laboratory, Baylor Scott and White Research Institute, 3434 Live Oak Street, Dallas, TX, 75204, USA.
| | - Bashoo Naziruddin
- Islet Cell Laboratory, Baylor Simmons Transplant Institute, 3410 Worth Street, Suite 950, Dallas, TX, 75246, USA.
| |
Collapse
|
12
|
Laporte C, Tubbs E, Cristante J, Gauchez AS, Pesenti S, Lamarche F, Cottet-Rousselle C, Garrel C, Moisan A, Moulis JM, Fontaine E, Benhamou PY, Lablanche S. Human mesenchymal stem cells improve rat islet functionality under cytokine stress with combined upregulation of heme oxygenase-1 and ferritin. Stem Cell Res Ther 2019; 10:85. [PMID: 30867050 PMCID: PMC6416979 DOI: 10.1186/s13287-019-1190-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 02/14/2019] [Accepted: 02/25/2019] [Indexed: 12/15/2022] Open
Abstract
Background Islets of Langerhans transplantation is a promising therapy for type 1 diabetes mellitus, but this technique is compromised by transplantation stresses including inflammation. In other tissues, co-transplantation with mesenchymal stem cells has been shown to reduce damage by improving anti-inflammatory and anti-oxidant defences. Therefore, we probed the protection afforded by bone marrow mesenchymal stem cells to islets under pro-inflammatory cytokine stress. Methods In order to evaluate the cytoprotective potential of mesenchymal stem cells on rat islets, co-cultures were exposed to the interleukin-1, tumour necrosis factor α and interferon γ cocktail for 24 h. Islet viability and functionality tests were performed. Reactive oxygen species and malondialdehyde were measured. Expression of stress-inducible genes acting as anti-oxidants and detoxifiers, such as superoxide dismutases 1 and 2, NAD(P)H quinone oxidoreductase 1, heme oxygenase-1 and ferritin H, was compared to non-stressed cells, and the corresponding proteins were measured. Data were analysed by a two-way ANOVA followed by a Holm-Sidak post hoc analysis. Results Exposure of rat islets to cytokines induces a reduction in islet viability and functionality concomitant with an oxidative status shift with an increase of cytosolic ROS production. Mesenchymal stem cells did not significantly increase rat islet viability under exposure to cytokines but protected islets from the loss of insulin secretion. A drastic reduction of the antioxidant factors heme oxygenase-1 and ferritin H protein levels was observed in islets exposed to the cytokine cocktail with a prevention of this effect by the presence of mesenchymal stem cells. Conclusions Our data evidenced that MSCs are able to preserve islet insulin secretion through a modulation of the oxidative imbalance mediated by heme and iron via heme oxygenase-1 and ferritin in a context of cytokine exposure. Electronic supplementary material The online version of this article (10.1186/s13287-019-1190-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Camille Laporte
- Laboratory of Fundamental and Applied Bioenergetics (LBFA), INSERM U 1055 and SFR Environmental and Systems Biology (BEeSy), University Grenoble Alpes, Grenoble, BP 53, F-38041, Grenoble Cedex, France.
| | - Emily Tubbs
- Laboratory of Fundamental and Applied Bioenergetics (LBFA), INSERM U 1055 and SFR Environmental and Systems Biology (BEeSy), University Grenoble Alpes, Grenoble, BP 53, F-38041, Grenoble Cedex, France
| | - Justine Cristante
- Laboratory of Fundamental and Applied Bioenergetics (LBFA), INSERM U 1055 and SFR Environmental and Systems Biology (BEeSy), University Grenoble Alpes, Grenoble, BP 53, F-38041, Grenoble Cedex, France.,Grenoble University Hospital, Grenoble, France
| | - Anne-Sophie Gauchez
- Biology Institute, Grenoble Alpes University Hospital, CS 10217, 38043, Grenoble Cedex 9, France
| | - Sandra Pesenti
- Univ Lyon, CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, 69600, Oullins, France
| | - Frédéric Lamarche
- Laboratory of Fundamental and Applied Bioenergetics (LBFA), INSERM U 1055 and SFR Environmental and Systems Biology (BEeSy), University Grenoble Alpes, Grenoble, BP 53, F-38041, Grenoble Cedex, France
| | - Cécile Cottet-Rousselle
- Laboratory of Fundamental and Applied Bioenergetics (LBFA), INSERM U 1055 and SFR Environmental and Systems Biology (BEeSy), University Grenoble Alpes, Grenoble, BP 53, F-38041, Grenoble Cedex, France
| | - Catherine Garrel
- Biology Institute, Grenoble Alpes University Hospital, CS 10217, 38043, Grenoble Cedex 9, France
| | - Anaick Moisan
- Cell Therapy and Engineering Unit, EFS Auvergne-Rhône-Alpes, 464 Route de lancey - La Bâtie, 38330, Saint Ismier, France
| | - Jean-Marc Moulis
- Laboratory of Fundamental and Applied Bioenergetics (LBFA), INSERM U 1055 and SFR Environmental and Systems Biology (BEeSy), University Grenoble Alpes, Grenoble, BP 53, F-38041, Grenoble Cedex, France.,CEA-Grenoble, Bioscience and Biotechnology Institute (BIG), 38054, Grenoble, France
| | - Eric Fontaine
- Laboratory of Fundamental and Applied Bioenergetics (LBFA), INSERM U 1055 and SFR Environmental and Systems Biology (BEeSy), University Grenoble Alpes, Grenoble, BP 53, F-38041, Grenoble Cedex, France.,Grenoble University Hospital, Grenoble, France
| | - Pierre-Yves Benhamou
- Laboratory of Fundamental and Applied Bioenergetics (LBFA), INSERM U 1055 and SFR Environmental and Systems Biology (BEeSy), University Grenoble Alpes, Grenoble, BP 53, F-38041, Grenoble Cedex, France.,Grenoble University Hospital, Grenoble, France
| | - Sandrine Lablanche
- Laboratory of Fundamental and Applied Bioenergetics (LBFA), INSERM U 1055 and SFR Environmental and Systems Biology (BEeSy), University Grenoble Alpes, Grenoble, BP 53, F-38041, Grenoble Cedex, France.,Grenoble University Hospital, Grenoble, France
| |
Collapse
|
13
|
Mouré A, Bacou E, Bosch S, Jegou D, Salama A, Riochet D, Gauthier O, Blancho G, Soulillou J, Poncelet D, Olmos E, Bach J, Mosser M. Extracellular hemoglobin combined with an O
2
‐generating material overcomes O
2
limitation in the bioartificial pancreas. Biotechnol Bioeng 2019; 116:1176-1189. [DOI: 10.1002/bit.26913] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 12/05/2018] [Accepted: 12/26/2018] [Indexed: 12/29/2022]
Affiliation(s)
- Anne Mouré
- Immuno-Endocrinology Unit (IECM), Oniris, INRA, Université Bretagne LoireNantes France
| | - Elodie Bacou
- Immuno-Endocrinology Unit (IECM), Oniris, INRA, Université Bretagne LoireNantes France
| | - Steffi Bosch
- Immuno-Endocrinology Unit (IECM), Oniris, INRA, Université Bretagne LoireNantes France
| | - Dominique Jegou
- Immuno-Endocrinology Unit (IECM), Oniris, INRA, Université Bretagne LoireNantes France
| | - Apolline Salama
- Immuno-Endocrinology Unit (IECM), Oniris, INRA, Université Bretagne LoireNantes France
- Centre de Recherche en Transplantation et Immunologie UMR 1064INSERM, Université de NantesNantes France
| | - David Riochet
- Service de Pédiatrie des Maladies ChroniquesCHU de NantesNantes France
| | | | - Gilles Blancho
- Centre de Recherche en Transplantation et Immunologie UMR 1064INSERM, Université de NantesNantes France
- Institut de Transplantation Urologie Néphrologie (ITUN), CHU NantesNantes France
| | - Jean‐Paul Soulillou
- Centre de Recherche en Transplantation et Immunologie UMR 1064INSERM, Université de NantesNantes France
- Institut de Transplantation Urologie Néphrologie (ITUN), CHU NantesNantes France
| | - Denis Poncelet
- Department of Process Engineering for Environment and Food Laboratory (GEPEA)UMR CNRS 6144, OnirisNantes France
| | - Eric Olmos
- Laboratoire Réactions et Génie des Procédés (LRGP)Université de Lorraine, CNRSNancy France
| | - Jean‐Marie Bach
- Immuno-Endocrinology Unit (IECM), Oniris, INRA, Université Bretagne LoireNantes France
| | - Mathilde Mosser
- Immuno-Endocrinology Unit (IECM), Oniris, INRA, Université Bretagne LoireNantes France
| |
Collapse
|
14
|
Heng XP, Li XJ, Li L, Yang LQ, Wang ZT, Huang SP. Therapy to Obese Type 2 Diabetes Mellitus: How Far Will We Go Down the Wrong Road? Chin J Integr Med 2018; 26:62-71. [PMID: 30328570 DOI: 10.1007/s11655-018-3053-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2018] [Indexed: 01/19/2023]
Abstract
Traditional glucose-lowering chemical agents, including various types of insulin and insulin secretagogues, insulin sensitizers, gliptins, etc., are based on diabetic pathogenesis of insulin resistance (IR) and islet insufficiency. Numerous evidence-based medical studies have shown that these traditional hypoglycemic chemical agents do not provide cardiovascular benefit to patients with type 2 diabetes mellitus (T2DM) and may even increase the risk of all-cause mortality. Based on research evidence published to date, these studies show that overload of energy could increase the incidence and prevalence of T2DM, and reduction in the heat load can significantly reduce the incidence of T2DM. Therefore, the essence of T2DM is heat overload, meaning heat overload is the etiology of obese T2DM. At the same time, results of numerous studies show that heat overloading is the cause of IR. IR and islet dysfunction are protective factors in intervening with heat overload. These drugs, which are based on the mechanisms of IR and islet insufficiency, increase caloric reserve and cause or worsen obesity, which is equivalent to exacerbating the basic etiology and the cardiovascular risk factor of T2DM. Thus, a reasonable strategy for prevention and treatment of obese T2DM appears to promote the negative balance of calories and the elimination of caloric reserves. Chinese herbal medicines can promote negative balance of heat in many aspects, which can bring new hope for prevention and treatment of T2DM.
Collapse
Affiliation(s)
- Xian-Pei Heng
- Department of Endocrinology, People's Hospital Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou, 350004, China.
| | - Xiu-Jun Li
- West China Medical Center of Sichuan Medical University (West China University of Medical Science), Chengdu, 610041, China
| | - Liang Li
- Department of Endocrinology, People's Hospital Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou, 350004, China
| | - Liu-Qing Yang
- Department of Endocrinology, People's Hospital Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou, 350004, China
| | - Zi-Ta Wang
- Department of Endocrinology, People's Hospital Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou, 350004, China
| | - Su-Ping Huang
- Academy of Integrative Medicine Fujian, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| |
Collapse
|
15
|
He W, Rebello O, Savino R, Terracciano R, Schuster-Klein C, Guardiola B, Maedler K. TLR4 triggered complex inflammation in human pancreatic islets. Biochim Biophys Acta Mol Basis Dis 2018; 1865:86-97. [PMID: 30287405 DOI: 10.1016/j.bbadis.2018.09.030] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 09/06/2018] [Accepted: 09/20/2018] [Indexed: 02/08/2023]
Abstract
Type 2 Diabetes (T2D) is strongly associated with obesity and inflammation. Toll-like receptor-4 (TLR-4) is the major pro-inflammatory pathway with its ligands and downstream products increased systemically in T2D and in at-risk individuals. Detailed mechanisms of the complex proinflammatory response in pancreatic islets remain unknown. In isolated human islets LPS induced IL-1β, IL-6, IL-8 and TNF production in a TLR4-dependent manner and severely impaired β-cell survival and function. IL-6 antagonism improved β-cell function. IL-8, which was identified specifically in α-cells, initiated monocyte migration, a process fully blocked by IL-8 neutralization. The TLR4 response was potentiated in obese donors; with higher IL-1β, IL-6 and IL-8 expression than in non-obese donors. TLR4 activation leads to a complex multi-cellular inflammatory response in human islets, which involves β-cell failure, cytokine production and macrophage recruitment to islets. In obesity, the amplified TLR4 response may potentiate β-cell damage and accelerate diabetes progression.
Collapse
Affiliation(s)
- Wei He
- University of Bremen, Center for Biomolecular Interactions Bremen, Germany.
| | - Osmond Rebello
- University of Bremen, Center for Biomolecular Interactions Bremen, Germany
| | - Rocco Savino
- Department of Health Sciences, Magna Graecia University, Catanzaro, Italy
| | - Rosa Terracciano
- Department of Health Sciences, Magna Graecia University, Catanzaro, Italy
| | | | | | - Kathrin Maedler
- University of Bremen, Center for Biomolecular Interactions Bremen, Germany.
| |
Collapse
|
16
|
Dai J, Gu L, Su Y, Wang Q, Zhao Y, Chen X, Deng H, Li W, Wang G, Li K. Inhibition of curcumin on influenza A virus infection and influenzal pneumonia via oxidative stress, TLR2/4, p38/JNK MAPK and NF-κB pathways. Int Immunopharmacol 2018; 54:177-187. [PMID: 29153953 DOI: 10.1016/j.intimp.2017.11.009] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 10/29/2017] [Accepted: 11/07/2017] [Indexed: 02/07/2023]
Abstract
Oxidative stress, Nrf2-HO-1 and TLR-MAPK/NF-κB signaling pathways have been proved to be involved in influenza A virus (IAV) replication and influenzal pneumonia. In the previous studies, we have performed several high-throughput drug screenings based on the TLR pathways. In the present study, through plaque inhibition test, luciferase reporter assay, TCID50, qRT-PCR, western blotting, ELISA and siRNA assays, we investigated the effect and mechanism of action of curcumin against IAV infection in vitro and in vivo. The results showed that curcumin could directly inactivate IAV, blocked IAV adsorption and inhibited IAV proliferation. As for the underlying mechanisms, we found that curcumin could significantly inhibit IAV-induced oxidative stress, increased Nrf2, HO-1, NQO1, GSTA3 and IFN-β production, and suppressed IAV-induced activation of TLR2/4/7, Akt, p38/JNK MAPK and NF-κB pathways. Suppression of Nrf2 via siRNA significantly abolished the stimulatory effect of curcumin on HO-1, NQO1, GSTA3 and IFN-β production and meanwhile blocked the inhibitory effect of curcumin on IAV M2 production. Oxidant H2O2 and TLR2/4, p38/JNK and NF-κB agonists could significantly antagonize the anti-IAV activity of curcumin in vitro. Additionally, curcumin significantly increased the survival rate of mice, reduced lung index, inflammatory cytokines and lung IAV titer, and finally improved pulmonary histopathological changes after IAV infection. In conclusion, curcumin can directly inactivate IAV, inhibits IAV adsorption and replication; and its inhibition on IAV replication may be via activating Nrf2 signal and inhibiting IAV-induced activation of TLR2/4, p38/JNK MAPK and NF-κB pathways.
Collapse
Affiliation(s)
- Jianping Dai
- Department of Microbiology and Immunology, Shantou University Medical College, 22 Xingling Rd, Shantou 515041, China.
| | - Liming Gu
- Department of Microbiology and Immunology, Shantou University Medical College, 22 Xingling Rd, Shantou 515041, China
| | - Yun Su
- Department of Microbiology and Immunology, Shantou University Medical College, 22 Xingling Rd, Shantou 515041, China
| | - Qianwen Wang
- Department of Microbiology and Immunology, Shantou University Medical College, 22 Xingling Rd, Shantou 515041, China
| | - Ying Zhao
- Department of Microbiology and Immunology, Shantou University Medical College, 22 Xingling Rd, Shantou 515041, China
| | - Xiaoxua Chen
- Department of Microbiology and Immunology, Shantou University Medical College, 22 Xingling Rd, Shantou 515041, China
| | - Huixiong Deng
- Department of Microbiology and Immunology, Shantou University Medical College, 22 Xingling Rd, Shantou 515041, China
| | - Weizhong Li
- Department of Veterinary Medicine, University of Maryland, College Park, Virginia-Maryland Regional College of Veterinary Medicine, 159 College Park Rd, MD 20742, USA
| | - Gefei Wang
- Department of Microbiology and Immunology, Shantou University Medical College, 22 Xingling Rd, Shantou 515041, China
| | - Kangsheng Li
- Department of Microbiology and Immunology, Shantou University Medical College, 22 Xingling Rd, Shantou 515041, China
| |
Collapse
|
17
|
Glade MJ, Meguid MM. A glance at…antioxidant and antiinflammatory properties of dietary cobalt. Nutrition 2017; 46:62-66. [PMID: 29290358 DOI: 10.1016/j.nut.2017.08.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 08/28/2017] [Indexed: 12/17/2022]
Affiliation(s)
| | - Michael M Meguid
- Professor Emeritus, Surgery, Neuroscience and Nutrition, Department of Surgery, University Hospital, Upstate Medical University, Syracuse, New York, USA
| |
Collapse
|
18
|
Ping Z, Aiqun M, Jiwu L, Liang S. TNF Receptor 1/2 Predict Heart Failure Risk in Type 2 Diabetes Mellitus Patients. Int Heart J 2017; 58:245-249. [PMID: 28367848 DOI: 10.1536/ihj.16-236] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Inflammation plays an important role in heart failure and diabetes mellitus. Traditional serum markers have limited predictive value in heart failure and diabetes. TNFR1 and TNFR2 (TNFR1/2) have been proven to be strongly associated with heart failure and diabetes complications. This study aimed to assess the association of sTNFR1 and sTNFR2 levels and incidental HF risk in diabetes patients.We detected the mRNA, protein, and serum expression of TNFR1/2, their downstream signaling pathway protein NF-kB, and JNK expression and some traditional serum inflammatory markers in a heart failure group without diabetes mellitus or abnormal glucose tolerance (n = 84), a diabetes mellitus group without heart failure (n = 86), and a heart failure with diabetes mellitus group (n = 86).TNFR1/2 were significantly higher in patients with heart failure and diabetes mellitus based on mRNA expression to protein expression and serum expression. However, there were no differences in mRNA, protein, and serum levels of TNFR1/2 between the HF group and DM group. Furthermore, there were no differences between the groups in some traditional serum inflammatory markers.This study demonstrated higher expressions of TNFR, NF-kB, and JNK in patients with heart failure and diabetes mellitus. Compared with traditional serum markers, TNFR1 and TNFR2 are associated with heart failure risk in type 2 diabetes mellitus patients.
Collapse
Affiliation(s)
- Zhang Ping
- Department of Geriatrics and Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University
| | | | | | | |
Collapse
|
19
|
Delaune V, Berney T, Lacotte S, Toso C. Intraportal islet transplantation: the impact of the liver microenvironment. Transpl Int 2017; 30:227-238. [DOI: 10.1111/tri.12919] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 11/09/2016] [Accepted: 01/16/2017] [Indexed: 12/20/2022]
Affiliation(s)
- Vaihere Delaune
- Hepatology and Transplantation Laboratory; Department of Surgery; Faculty of Medicine; University of Geneva; Geneva Switzerland
- Divisions of Abdominal and Transplantation Surgery; Department of Surgery; Geneva University Hospitals; Geneva Switzerland
| | - Thierry Berney
- Divisions of Abdominal and Transplantation Surgery; Department of Surgery; Geneva University Hospitals; Geneva Switzerland
- Cell Transplantation Laboratory; Department of Surgery; Faculty of Medicine; University of Geneva; Geneva Switzerland
| | - Stéphanie Lacotte
- Hepatology and Transplantation Laboratory; Department of Surgery; Faculty of Medicine; University of Geneva; Geneva Switzerland
| | - Christian Toso
- Hepatology and Transplantation Laboratory; Department of Surgery; Faculty of Medicine; University of Geneva; Geneva Switzerland
- Divisions of Abdominal and Transplantation Surgery; Department of Surgery; Geneva University Hospitals; Geneva Switzerland
| |
Collapse
|
20
|
Wang X, Ge QM, Bian F, Dong Y, Huang CM. Inhibition of TLR4 protects rat islets against lipopolysaccharide-induced dysfunction. Mol Med Rep 2016; 15:805-812. [PMID: 28101570 DOI: 10.3892/mmr.2016.6097] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 10/07/2016] [Indexed: 11/06/2022] Open
Abstract
Oxidative stress leads to dysfunction in pancreatic cells, causing a reduction in insulin secretion following exposure to glucose. Toll-like receptor 4 (TLR4) may be activated by exposure to lipopolysaccharide (LPS) stress. TLR4 may mediate the initiation of inflammatory and immune defense responses; however, the importance of the LPS/TLR4 interaction in apoptosis induced by oxidative stress in pancreatic β cells remains to be elucidated. The present study aimed to investigate the importance of TLR4 during LPS‑induced oxidative stress, apoptosis and dysfunction of insulin secretion in isolated islets of rats. LPS‑induced stimulation of TLR4 increased the production of reactive oxygen species and promoted apoptosis by upregulating the expression levels of caspase‑3, poly ADP ribose polymerase and altering the expression ratio of B‑cell lymphoma‑2 (Bcl‑2)/Bcl‑2 associated X protein. Additionally, the insulin secretion of islets cells was reduced. Anti‑TLR4 antibody and a knockdown of TLR4 by TLR4‑short hairpin RNA were used to inhibit TLR4 activity, which may reverse LPS‑induced events. The present study determined that in islets exposed to LPS oxidative stress, dysfunction may be partly mediated via the TLR4 pathway. Inhibition of TLR4 may prevent dysfunction of rat islets due to oxidative stress. The present study revealed that targeting the LPS/TLR4 signaling pathway and antioxidant therapy may be a novel treatment for the severely septic patients with hyperglycemia stress.
Collapse
Affiliation(s)
- Xiao Wang
- Department of Emergency, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, P.R. China
| | - Qin Min Ge
- Department of Emergency, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, P.R. China
| | - Fan Bian
- Department of Nephrology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, P.R. China
| | - Yan Dong
- Department of Endocrinology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, P.R. China
| | - Chun Mei Huang
- Department of Emergency, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, P.R. China
| |
Collapse
|
21
|
Langlois A, Dal S, Vivot K, Mura C, Seyfritz E, Bietiger W, Dollinger C, Peronet C, Maillard E, Pinget M, Jeandidier N, Sigrist S. Improvement of islet graft function using liraglutide is correlated with its anti-inflammatory properties. Br J Pharmacol 2016; 173:3443-3453. [PMID: 27515367 PMCID: PMC5120160 DOI: 10.1111/bph.13575] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 06/27/2016] [Accepted: 07/20/2016] [Indexed: 01/15/2023] Open
Abstract
Background and Purpose Liraglutide improves the metabolic control of diabetic animals after islet transplantation. However, the mechanisms underlying this effect remain unknown. The objective of this study was to evaluate the anti‐inflammatory and anti‐oxidative properties of liraglutide on rat pancreatic islets in vitro and in vivo. Experimental Approach In vitro, rat islets were incubated with 10 μmol·L−1 liraglutide for 12 and 24 h. Islet viability functionality was assessed. The anti‐inflammatory properties of liraglutide were evaluated by measuring CCL2, IL‐6 and IL‐10 secretion and macrophage chemotaxis. The anti‐oxidative effect of liraglutide was evaluated by measuring intracellular ROS and the total anti‐oxidative capacity. In vivo, 1000 islets were cultured for 24 h with or without liraglutide and then transplanted into the liver of streptozotocin‐induced diabetic Lewis rats with or without injections of liraglutide. Effects of liraglutide on metabolic control were evaluated for 1 month. Key Results Islet viability and function were preserved and enhanced with liraglutide treatment. Liraglutide decreased CCL2 and IL‐6 secretion and macrophage activation after 12 h of culture, while IL‐10 secretion was unchanged. However, intracellular levels of ROS were increased with liraglutide treatment at 12 h. This result was correlated with an increase of anti‐oxidative capacity. In vivo, liraglutide decreased macrophage infiltration and reduced fasting blood glucose in transplanted rats. Conclusions and Implications The beneficial effects of liraglutide on pancreatic islets appear to be linked to its anti‐inflammatory and anti‐oxidative properties. These findings indicated that analogues of glucagon‐like peptide‐1 could be used to improve graft survival.
Collapse
Affiliation(s)
- A Langlois
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - S Dal
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - K Vivot
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - C Mura
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - E Seyfritz
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - W Bietiger
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - C Dollinger
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - C Peronet
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - E Maillard
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - M Pinget
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.,Service d'Endocrinologie, Diabète, Maladies Métaboliques, Pôle NUDE, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - N Jeandidier
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.,Service d'Endocrinologie, Diabète, Maladies Métaboliques, Pôle NUDE, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - S Sigrist
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| |
Collapse
|
22
|
Liu J, Zhang BL, Sun CL, Wang J, Li S, Wang JF. High mobility group box1 protein is involved in acute inflammation induced by Clostridium difficile toxin A. Acta Biochim Biophys Sin (Shanghai) 2016; 48:554-62. [PMID: 27151296 DOI: 10.1093/abbs/gmw038] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 03/31/2016] [Indexed: 12/31/2022] Open
Abstract
High mobility group box1 (HMGB1), as a damage-associated inflammatory factor, contributes to the pathogenesis of numerous chronic inflammatory and autoimmune diseases. In this study, we explored the role of HMGB1 in CDI (Clostridium difficile infection) by in vivo and in vitro experiments. Our results showed that HMGB1 might play an important role in the acute inflammatory responses to C. difficile toxin A (TcdA), affect early inflammatory factors, and induce inflammation via the HMGB1-TLR4 pathway. Our study provides the essential information for better understanding the molecular mechanisms of CDI and the potential new therapeutic strategies for the treatment of this infection.
Collapse
Affiliation(s)
- Ji Liu
- School of Bioscience & Bioengineering, South China University of Technology, Guangzhou 510006, China
| | - Bei-Lei Zhang
- School of Bioscience & Bioengineering, South China University of Technology, Guangzhou 510006, China
| | - Chun-Li Sun
- School of Bioscience & Bioengineering, South China University of Technology, Guangzhou 510006, China
| | - Jun Wang
- Shenzhen Huada Gene Research Institute, Shenzhen 518083, China
| | - Shan Li
- School of Bioscience & Bioengineering, South China University of Technology, Guangzhou 510006, China Guangdong Province Key Laboratory of Fermentation and Enzyme Engineering, South China University of Technology, Guangzhou 510006, China
| | - Ju-Fang Wang
- School of Bioscience & Bioengineering, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|
23
|
Long-term exposure to ELF-MF ameliorates cognitive deficits and attenuates tau hyperphosphorylation in 3xTg AD mice. Neurotoxicology 2016; 53:290-300. [PMID: 26945731 DOI: 10.1016/j.neuro.2016.02.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 02/24/2016] [Accepted: 02/29/2016] [Indexed: 12/30/2022]
Abstract
Although numerous studies have reported the influence of extremely low frequency magnetic field (ELF-MF) exposure on human health, its effects on cognitive deficits in Alzheimer's disease (AD) have remained under debate. Moreover, the influence of ELF-MF on hyperphosphorylated tau, which is one of the most common pathological hallmarks of AD, has not been reported to date. Therefore, transgenic mice (3xTg) were used in the present study. 3xTg mice, which express an APP/PS1 mutation combined with a tau (P301L) mutation and that develop cognitive deficits at 6 months of age, were subjected to ELF-MF (50Hz, 500μT) exposure or sham exposure daily for 3 months. We discovered that ELF-MF exposure ameliorated cognitive deficits and increased synaptic proteins in 3xTg mice. The protective effects of ELF-MF exposure may have also been caused by the inhibition of apoptosis and/or decreased oxidative stress levels that were observed in the hippocampus tissues of treated mice. Furthermore, tau hyperphosphorylation was decreased in vivo because of ELF-MF exposure, and this decrease was induced by the inhibition of GSK3β and CDK5 activities and activation of PP2Ac. We are the first to report that exposure to ELF-MF can attenuate tau phosphorylation. These findings suggest that ELF-MF exposure could act as a valid therapeutic strategy for ameliorating cognitive deficits and attenuating tau hyperphosphorylation in AD.
Collapse
|
24
|
Rodriguez-Brotons A, Bietiger W, Peronet C, Magisson J, Sookhareea C, Langlois A, Mura C, Jeandidier N, Pinget M, Sigrist S, Maillard E. Impact of Pancreatic Rat Islet Density on Cell Survival during Hypoxia. J Diabetes Res 2016; 2016:3615286. [PMID: 26824040 PMCID: PMC4707363 DOI: 10.1155/2016/3615286] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 09/21/2015] [Accepted: 09/29/2015] [Indexed: 01/01/2023] Open
Abstract
In bioartificial pancreases (BP), the number of islets needed to restore normoglycaemia in the diabetic patient is critical. However, the confinement of a high quantity of islets in a limited space may impact islet survival, particularly in regard to the low oxygen partial pressure (PO2) in such environments. The aim of the present study was to evaluate the impact of islet number in a confined space under hypoxia on cell survival. Rat islets were seeded at three different concentrations (150, 300, and 600 Islet Equivalents (IEQ)/cm(2)) and cultured in normal atmospheric pressure (160 mmHg) as well as hypoxic conditions (15 mmHg) for 24 hours. Cell viability, function, hypoxia-induced changes in gene expression, and cytokine secretion were then assessed. Notably, hypoxia appeared to induce a decrease in viability and increasing islet density exacerbated the observed increase in cellular apoptosis as well as the loss of function. These changes were also associated with an increase in inflammatory gene transcription. Taken together, these data indicate that when a high number of islets are confined to a small space under hypoxia, cell viability and function are significantly impacted. Thus, in order to improve islet survival in this environment during transplantation, oxygenation is of critical importance.
Collapse
Affiliation(s)
- A. Rodriguez-Brotons
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg, Bld René Leriche, 67200 Strasbourg, France
| | - W. Bietiger
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg, Bld René Leriche, 67200 Strasbourg, France
| | - C. Peronet
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg, Bld René Leriche, 67200 Strasbourg, France
| | - J. Magisson
- Defymed, avenue Dante, 67200 Strasbourg, France
| | - C. Sookhareea
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg, Bld René Leriche, 67200 Strasbourg, France
| | - A. Langlois
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg, Bld René Leriche, 67200 Strasbourg, France
| | - C. Mura
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg, Bld René Leriche, 67200 Strasbourg, France
| | - N. Jeandidier
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg, Bld René Leriche, 67200 Strasbourg, France
- Structure d'Endocrinologie, Diabète-Nutrition et Addictologie, Pôle NUDE, Hôpitaux Universitaires de Strasbourg (HUS), 67000 Strasbourg, France
| | - M. Pinget
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg, Bld René Leriche, 67200 Strasbourg, France
- Structure d'Endocrinologie, Diabète-Nutrition et Addictologie, Pôle NUDE, Hôpitaux Universitaires de Strasbourg (HUS), 67000 Strasbourg, France
| | - S. Sigrist
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg, Bld René Leriche, 67200 Strasbourg, France
| | - E. Maillard
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg, Bld René Leriche, 67200 Strasbourg, France
- *E. Maillard:
| |
Collapse
|
25
|
Chen J, Lai J, Yang L, Ruan G, Chaugai S, Ning Q, Chen C, Wang DW. Trimetazidine prevents macrophage-mediated septic myocardial dysfunction via activation of the histone deacetylase sirtuin 1. Br J Pharmacol 2015; 173:545-61. [PMID: 26566260 DOI: 10.1111/bph.13386] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 10/13/2015] [Accepted: 10/18/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE Sepsis is a systemic inflammatory response accompanied by excessive production of inflammatory cytokines and cardiovascular dysfunction. Importantly, macrophage-derived pro-inflammatory agents play a key role in cardiovascular impairment in sepsis. Here we have investigated the effects of trimetazidine (TMZ) on pro-inflammatory responses of macrophages in endotoxin-induced myocardial dysfunction. EXPERIMENTAL APPROACH Mice pretreated with TMZ were injected i.p. with LPS and cardiac function evaluated. Levels of macrophage infiltration, macrophage inflammatory response and cardiomyocyte apoptosis were measured using immunohistochemical staining, elisa, real-time RT-PCR, Western blot, TUNEL and flow cytometry assays. KEY RESULTS Pretreatment with TMZ prevented LPS-induced myocardial dysfunction and apoptosis. TMZ also lowered levels of pro-inflammatory cytokines in serum and cardiac tissue and myocardial macrophage infiltration. Bone marrow transplantation indicated that TMZ alleviated LPS-induced myocardial dysfunction via decreasing macrophage infiltration. TMZ reduced expression of pro-inflammatory cytokines in LPS-stimulated cardiac and peritoneal macrophages. Co-culture of TMZ-pretreated macrophages with cardiomyocytes and conditioned media from TMZ-pretreated macrophages both decreased LPS-induced cardiomyocyte apoptosis. The anti-apoptosis effects of TMZ resulted from decrease of pro-inflammatory cytokines, partly due to normalizing the sirtuin 1 (Sirt1)/AMP-activated protein kinase (AMPK)/Nrf2/haem oxygenase-1 and Sirt1/PPARα pathways in macrophages. Cytokine secretion was also regulated by ROS, which were attenuated by TMZ via activation of Sirt1, AMPK and PPARα. CONCLUSIONS AND IMPLICATIONS TMZ protected against LPS-induced myocardial dysfunction and apoptosis, accompanied by inhibition of macrophage pro-inflammatory responses. Our studies suggest that TMZ might represent a novel therapeutic agent to prevent and treat sepsis-induced myocardial dysfunction.
Collapse
Affiliation(s)
- Jing Chen
- Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinsheng Lai
- Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Yang
- Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guoran Ruan
- Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sandip Chaugai
- Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qin Ning
- Department of Infectious Disease, Institute of Infectious Disease, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chen Chen
- Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dao Wen Wang
- Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
26
|
Abraham NG, Junge JM, Drummond GS. Translational Significance of Heme Oxygenase in Obesity and Metabolic Syndrome. Trends Pharmacol Sci 2015; 37:17-36. [PMID: 26515032 DOI: 10.1016/j.tips.2015.09.003] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 09/10/2015] [Accepted: 09/17/2015] [Indexed: 01/04/2023]
Abstract
The global epidemic of obesity continues unabated with sequelae of diabetes and metabolic syndrome. This review reflects the dramatic increase in research on the role of increased expression of heme oxygenase (HO)-1/HO-2, biliverdin reductase, and HO activity on vascular disease. The HO system engages with other systems to mitigate the deleterious effects of oxidative stress in obesity and cardiovascular disease (CVD). Recent reports indicate that HO-1/HO-2 protein expression and HO activity have several important roles in hemostasis and reactive oxygen species (ROS)-dependent perturbations associated with metabolic syndrome. HO-1 protects tissue during inflammatory stress in obesity through the degradation of pro-oxidant heme and the production of carbon monoxide (CO) and bilirubin, both of which have anti-inflammatory and anti-apoptotic properties. By contrast, repression of HO-1 is associated with increases of cellular heme and inflammatory conditions including hypertension, stroke, and atherosclerosis. HO-1 is a major focus in the development of potential therapeutic strategies to reverse the clinical complications of obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Nader G Abraham
- Departments of Medicine and Pharmacology, New York Medical College, School of Medicine, Valhalla, NY 10595, USA; Marshall University, Joan C. Edwards School of Medicine, Huntington, WV 25701, USA.
| | - Joshua M Junge
- Departments of Medicine and Pharmacology, New York Medical College, School of Medicine, Valhalla, NY 10595, USA
| | - George S Drummond
- Departments of Medicine and Pharmacology, New York Medical College, School of Medicine, Valhalla, NY 10595, USA
| |
Collapse
|
27
|
Zeng P, Pi RB, Li P, Chen RX, Lin LM, He H, Zhou SY. Fasudil hydrochloride, a potent ROCK inhibitor, inhibits corneal neovascularization after alkali burns in mice. Mol Vis 2015; 21:688-98. [PMID: 26120273 PMCID: PMC4463969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Accepted: 06/10/2015] [Indexed: 11/30/2022] Open
Abstract
PURPOSE To investigate the effects and mechanisms of fasudil hydrochloride (fasudil) on and in alkali burn-induced corneal neovascularization (CNV) in mice. METHODS To observe the effect of fasudil, mice with alkali-burned corneas were treated with either fasudil eye drops or phosphate-buffered saline (PBS) four times per day for 14 consecutive days. After injury, CNV and corneal epithelial defects were measured. The production of reactive oxygen species (ROS) and heme oxygenase-1(HO-1) was measured. The infiltration of polymorphonuclear neutrophils (PMNs) and the mRNA expressions of CNV-related genes were analyzed on day 14. RESULTS The incidence of CNV was significantly lower after treatment with 100 μM and 300 μM fasudil than with PBS, especially with 100 μM fasudil. Meanwhile, the incidences of corneal epithelial defects was lower (n=15, all p<0.01). After treatment with 100 μM fasudil, the intensity of DHE fluorescence was reduced in the corneal epithelium and stroma than with PBS treatment (n=5, all p<0.01), and the number of filtrated PMNs decreased. There were significant differences between the expressions of VEGF, TNF-a, MMP-8, and MMP-9 in the 100 μM fasudil group and the PBS group (n=8, all p<0.05). The production of HO-1 protein in the 100 μM fasudil group was 1.52±0.34 times more than in the PBS group (n=5 sample, p<0.05). CONCLUSIONS 100 μM fasudil eye drops administered four times daily can significantly inhibit alkali burn-induced CNV and promote the healing of corneal epithelial defects in mice. These effects are attributed to a decrease in inflammatory cell infiltration, reduction of ROS, and upregulation of HO-1 protein after fasudil treatment.
Collapse
Affiliation(s)
- Peng Zeng
- Zhongshan Ophthalmic Center of Sun Yat-sen University, The State Key Laboratory of Ophthalmology,Guangzhou, China
| | - Rong-biao Pi
- Department of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Peng Li
- Department of Ophthalmology, No.181 Hospital of PLA, Guangxi, Guilin, China
| | - Rong-xin Chen
- Zhongshan Ophthalmic Center of Sun Yat-sen University, The State Key Laboratory of Ophthalmology,Guangzhou, China
| | - Li-mian Lin
- Zhongshan Ophthalmic Center of Sun Yat-sen University, The State Key Laboratory of Ophthalmology,Guangzhou, China
| | - Hong He
- Hainan Eye Hospital of Zhongshan Ophthalmic Center, Haikou, Hainan Province, China
| | - Shi-you Zhou
- Zhongshan Ophthalmic Center of Sun Yat-sen University, The State Key Laboratory of Ophthalmology,Guangzhou, China
| |
Collapse
|