1
|
Ježek P. Physiological Fatty Acid-Stimulated Insulin Secretion and Redox Signaling Versus Lipotoxicity. Antioxid Redox Signal 2025; 42:566-622. [PMID: 39834189 DOI: 10.1089/ars.2024.0799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Significance: Type 2 diabetes as a world-wide epidemic is characterized by the insulin resistance concomitant to a gradual impairment of β-cell mass and function (prominently declining insulin secretion) with dysregulated fatty acids (FAs) and lipids, all involved in multiple pathological development. Recent Advances: Recently, redox signaling was recognized to be essential for insulin secretion stimulated with glucose (GSIS), branched-chain keto-acids, and FAs. FA-stimulated insulin secretion (FASIS) is a normal physiological event upon postprandial incoming chylomicrons. This contrasts with the frequent lipotoxicity observed in rodents. Critical Issues: Overfeeding causes FASIS to overlap with GSIS providing repeating hyperinsulinemia, initiates prediabetic states by lipotoxic effects and low-grade inflammation. In contrast the protective effects of lipid droplets in human β-cells counteract excessive lipids. Insulin by FASIS allows FATP1 recruitment into adipocyte plasma membranes when postprandial chylomicrons come late at already low glycemia. Future Directions: Impaired states of pancreatic β-cells and peripheral organs at prediabetes and type 2 diabetes should be revealed, including the inter-organ crosstalk by extracellular vesicles. Details of FA/lipid molecular physiology are yet to be uncovered, such as complex phenomena of FA uptake into cells, postabsorptive inactivity of G-protein-coupled receptor 40, carnitine carrier substrate specificity, the role of carnitine-O-acetyltransferase in β-cells, and lipid droplet interactions with mitochondria. Antioxid. Redox Signal. 42, 566-622.
Collapse
Affiliation(s)
- Petr Ježek
- Department of Mitochondrial Physiology, No.75, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
2
|
Alarfaj SJ, Bahaa MM, Elmasry TA, Elberri EI, El-Khateeb E, Hamouda AO, Salahuddin MM, Kamal M, Gadallah ANAA, Eltantawy N, Yasser M, Negm WA, Hamouda MA, Alsegiani AS, Alrubia S, Eldesoqui M, Abdallah MS. Fenofibrate as an Adjunct Therapy for Ulcerative Colitis: Targeting Inflammation via SIRT1, NLRP3, and AMPK Pathways: A Randomized Controlled Pilot Study. Drug Des Devel Ther 2024; 18:5239-5253. [PMID: 39575188 PMCID: PMC11578921 DOI: 10.2147/dddt.s490772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 11/08/2024] [Indexed: 11/24/2024] Open
Abstract
Background Ulcerative colitis (UC) is an idiopathic chronic inflammation of colonic and rectal mucosa. The peroxisome proliferator-activated receptor α (PPARα) has been identified as having protective effects in UC. Aim The study aimed to investigate the efficacy of fenofibrate, a PPARα agonist, in UC. Methods A total of 70 patients with mild to moderate UC were allocated randomly and assigned to two groups (n = 35 each) from Gastroenterology Department, Faculty of Medicine, Menoufia University. The mesalamine group received a placebo along with 1 g of mesalamine three times daily, while the fenofibrate group received 1 g of mesalamine three times and fenofibrate 160 mg once daily. The study duration was for six months. A gastroenterologist assessed patients by non-invasive Partial Mayo Score (PMS) and the Inflammatory Bowel Disease Questionnaire (IBDQ) to evaluate clinical response and remission. The serum levels of silent information regulator 1 (SIRT1), NOD-like receptor protein 3 (NLRP3), and adenosine monophosphate activated protein kinase (AMPK), as well as fecal calprotectin levels were examined to determine the biological effect of fenofibrate. Results After treatment, the fenofibrate group showed statistically significant reductions in PMS (p = 0.044) and improved digestive domain of IBDQ (p = 0.023). Additionally, there were significant decreases in serum NLRP3 (p = 0.041) and fecal calprotectin (p = 0.035), along with significant increases in SIRT1 (p = 0.002) and AMPK (p = 0.0003). The fenofibrate group also had higher response and remission rates compared to the mesalamine group. Conclusion Fenofibrate may be a promising adjunct for improving clinical outcomes, quality of life, and modulating inflammation in mild to moderate patients with UC. Trial Registration Identifier NCT05781698.
Collapse
Affiliation(s)
- Sumaiah J Alarfaj
- Department of Pharmacy Practice, College of Pharmacy, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Mostafa M Bahaa
- Pharmacy Practice Department, Faculty of Pharmacy, Horus University, New Damietta, Egypt
| | - Thanaa A Elmasry
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Eman I Elberri
- Clinical Pharmacy Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Eman El-Khateeb
- Clinical Pharmacy Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Amir O Hamouda
- Department of Biochemistry and Pharmacology, Faculty of Pharmacy, Horus University, New Damietta, Egypt
| | - Muhammed M Salahuddin
- Department of Biochemistry and Pharmacology, Faculty of Pharmacy, Horus University, New Damietta, Egypt
| | - Marwa Kamal
- Department of Clinical Pharmacy, Faculty of Pharmacy, Fayoum University, Fayoum, Egypt
| | | | - Nashwa Eltantawy
- Department of Pharmacy Practice, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University, Cairo, Egypt
| | - Mohamed Yasser
- Department of Pharmaceutics, Faculty of Pharmacy, Port Said University, Port Said, Egypt
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Horus University, New Damietta, Egypt
- Department of Pharmaceutics, Faculty of Pharmacy, East Port Said National University, Port Said, Egypt
| | - Walaa A Negm
- Pharmacognosy Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Manal A Hamouda
- Department of Clinical Pharmacy, Faculty of Pharmacy, Menofia University, Menofia, Egypt
| | - Amsha S Alsegiani
- Pharmaceutical Chemistry Department, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Sarah Alrubia
- Pharmaceutical Chemistry Department, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mamdouh Eldesoqui
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh, Saudi Arabia
| | - Mahmoud S Abdallah
- Department of Clinical Pharmacy, Faculty of Pharmacy, University of Sadat City (USC), Sadat City, Menoufia, Egypt
- Department of PharmD, Faculty of Pharmacy, Jadara University, Irbid, Jordan
| |
Collapse
|
3
|
Wu S, Zhou Y, Liang J, Ying P, Situ Q, Tan X, Zhu J. Upregulation of NF-κB by USP24 aggravates ferroptosis in diabetic cardiomyopathy. Free Radic Biol Med 2024; 210:352-366. [PMID: 38056575 DOI: 10.1016/j.freeradbiomed.2023.11.032] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/06/2023] [Accepted: 11/27/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND Recent investigations have proposed a potential causal association between the occurrence of ferroptosis, nuclear factor kappa B (NF-κB) and ubiquitin-specific protease 24 (USP24). Nevertheless, the mechanism of USP24 and NF-κB regulation of ferroptosis in the context of diabetic cardiomyopathy (DCM) remain unclear. METHODS In this study, a high-fat diet and a streptozotocin-induced mouse DCM model were established, and high glucose and palmitic acid treatment of H9c2 cells and neonatal mouse primary cardiomyocytes (NMPCs) was used as an in vitro DCM models. Utilizing both the in vivo and in vitro DCM models, we assessed of USP24, NF-κB, and ferroptosis levels, and explored the relationship among them. RESULTS In in vivo and in vitro DCM models, increased expression of USP24, NF-κB, phosphorylated NF-κB (p-NF-κB) and fatty acid-CoA ligase 4 (FACL4) were detected, along with accumulated iron, as well as reduced ferritin heavy chain 1 (FTH1), solute carrier family 7 member 11 (SLC7A11) and antioxidant capacity. Knockdown of USP24 resulted in a reduction of NF-κB levels, while knockdown of NF-κB did not lead to a decrease in USP24 expression. Moreover, in H9c2 cells, knockdown of USP24 and NF-κB separately resulted in reduced levels of FACL4, increased levels of SLC7A11 and FTH1, as well as improved antioxidant capacity and cell viability. In shUSP24 knockdown H9c2 cells, administration of phorbol 12-myristate 13-acetate (PMA) activated NF-κB, subsequently reversing the previously observed effect caused by USP24 knockdown. CONCLUSIONS These findings show that USP24 upregulates NF-κB to promote ferroptosis in DCM.
Collapse
Affiliation(s)
- Shenglin Wu
- Institute of Clinical Electrocardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Yueran Zhou
- Institute of Clinical Electrocardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Jiaquan Liang
- Institute of Clinical Electrocardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Pengxiang Ying
- Institute of Clinical Electrocardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Qiwei Situ
- Institute of Clinical Electrocardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Xuerui Tan
- Clinical Research Center, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Jinxiu Zhu
- Institute of Clinical Electrocardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China; Longgang Maternity and Child Institute of Shantou University Medical College (Longgang District Maternity & Child Healthcare Hospital of Shenzhen City), Shenzhen, Guangdong 518172, China.
| |
Collapse
|
4
|
Mostallino R, Santoni M, Sagheddu C, Serra V, Orrù V, Pistis M, Castelli MP. The PPARα agonist fenofibrate reduces the cytokine imbalance in a maternal immune activation model of schizophrenia. Eur J Pharmacol 2023; 961:176172. [PMID: 37939988 DOI: 10.1016/j.ejphar.2023.176172] [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: 07/28/2023] [Revised: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 11/10/2023]
Abstract
Maternal infections during pregnancy may increase the risk of psychiatric disorders in offspring. We recently demonstrated that activation of peroxisome proliferator-activate receptor-α (PPARα), with the clinically available agonist fenofibrate (FEN), attenuates the neurodevelopmental disturbances induced by maternal immune activation (MIA) in rat offspring. We hypothesized that fenofibrate might reduce MIA-induced cytokine imbalance using a MIA model based on the viral mimetic polyriboinosinic-polyribocytidilic acid [poly (I:C)]. By using the Bio-Plex Multiplex-Immunoassay-System, we measured cytokine/chemokine/growth factor levels in maternal serum and in the fetal brain of rats treated with fenofibrate, at 6 and 24 h after poly (I:C). We found that MIA induced time-dependent changes in the levels of several cytokines/chemokines/colony-stimulating factors (CSFs). Specifically, the maternal serum of the poly (I:C)/control (CTRL) group showed increased levels of (i) proinflammatory chemokine macrophage inflammatory protein 1-alpha (MIP-1α), (ii) tumor necrosis factor-alpha (TNF-α), the monocyte chemoattractant protein-1 (MCP-1), the macrophage (M-CSF) and granulocyte-macrophage colony-stimulating factor (GM-CSF). Conversely, in the fetal brain of the poly (I:C)/CTRL group, interleukin 12p70 and MIP-1α levels were lower than in vehicle (veh)/CTRL group. Notably, MIP-1α, TNF-α, keratinocyte derived chemokine (GRO/KC), GM-CSF, and M-CSF levels were lower in the poly (I:C)/FEN than in poly (I:C)/CTRL rats, suggesting the protective role of the PPARα agonist. PPARα might represent a therapeutic target to attenuate MIA-induced inflammation.
Collapse
Affiliation(s)
- Rafaela Mostallino
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy
| | - Michele Santoni
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy
| | - Claudia Sagheddu
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy
| | - Valentina Serra
- Institute for Genetic and Biomedical Research, National Research Council (CNR), Lanusei, Italy
| | - Valeria Orrù
- Institute for Genetic and Biomedical Research, National Research Council (CNR), Lanusei, Italy
| | - Marco Pistis
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy; Neuroscience Institute, National Research Council of Italy, Section of Cagliari, Italy; Unit of Clinical Pharmacology, University Hospital, Cagliari, Italy.
| | - M Paola Castelli
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy.
| |
Collapse
|
5
|
Souza-Tavares H, Miranda CS, Vasques-Monteiro IML, Sandoval C, Santana-Oliveira DA, Silva-Veiga FM, Fernandes-da-Silva A, Souza-Mello V. Peroxisome proliferator-activated receptors as targets to treat metabolic diseases: Focus on the adipose tissue, liver, and pancreas. World J Gastroenterol 2023; 29:4136-4155. [PMID: 37475842 PMCID: PMC10354577 DOI: 10.3748/wjg.v29.i26.4136] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/26/2023] [Accepted: 06/13/2023] [Indexed: 07/10/2023] Open
Abstract
The world is experiencing reflections of the intersection of two pandemics: Obesity and coronavirus disease 2019. The prevalence of obesity has tripled since 1975 worldwide, representing substantial public health costs due to its comorbidities. The adipose tissue is the initial site of obesity impairments. During excessive energy intake, it undergoes hyperplasia and hypertrophy until overt inflammation and insulin resistance turn adipocytes into dysfunctional cells that send lipotoxic signals to other organs. The pancreas is one of the organs most affected by obesity. Once lipotoxicity becomes chronic, there is an increase in insulin secretion by pancreatic beta cells, a surrogate for type 2 diabetes mellitus (T2DM). These alterations threaten the survival of the pancreatic islets, which tend to become dysfunctional, reaching exhaustion in the long term. As for the liver, lipotoxicity favors lipogenesis and impairs beta-oxidation, resulting in hepatic steatosis. This silent disease affects around 30% of the worldwide population and can evolve into end-stage liver disease. Although therapy for hepatic steatosis remains to be defined, peroxisome proliferator-activated receptors (PPARs) activation copes with T2DM management. Peroxisome PPARs are transcription factors found at the intersection of several metabolic pathways, leading to insulin resistance relief, improved thermogenesis, and expressive hepatic steatosis mitigation by increasing mitochondrial beta-oxidation. This review aimed to update the potential of PPAR agonists as targets to treat metabolic diseases, focusing on adipose tissue plasticity and hepatic and pancreatic remodeling.
Collapse
Affiliation(s)
| | | | | | - Cristian Sandoval
- Escuela de Tecnología Médica, Facultad de Salud, Universidad Santo Tomás, Osorno 5310431, Chile
- Departamento de Ciencias Preclínicas, Universidad de la Frontera, Temuco 4780000, Chile
| | | | | | | | - Vanessa Souza-Mello
- Department of Anatomy, Rio de Janeiro State University, Rio de Janeiro 20551030, Brazil
| |
Collapse
|
6
|
Alhirmizi IAO, Uysal F, Arslan SO, Özünlü SAÇ, Koç A, Parlar A, Bayram KK. Fenofibrate Attenuates Asthma Features in an Ovalbumin-induced Mouse Model Via Suppressing NF-κB Binding Activity. Respir Physiol Neurobiol 2023:104083. [PMID: 37295485 DOI: 10.1016/j.resp.2023.104083] [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: 02/24/2023] [Revised: 05/07/2023] [Accepted: 05/20/2023] [Indexed: 06/12/2023]
Abstract
BACKGROUND/AIM Asthma is a chronic inflammatory disease of the airways with a high prevalence. Asthma has a complex pathophysiology and about 5-10% of patients are not fully responsive to the currently available treatments. The aim of this study is to investigate the involvement of NF-κB in the effects of fenofibrate on a mouse model of allergic asthma. MATERIALS AND METHODS A total of 49 BALB/c mice were randomly distributed into 7 groups (n=7). Allergic asthma model was created by administering i.p. injections of ovalbumin on days 0, 14 and 21, followed by provocation with inhaled ovalbumin on days 28, 29 and 30. Fenofibrate was orally given in 3 different doses; 1, 10 and 30mg/kg through days 21 to 30 of the experiment. On day 31, pulmonary function test using whole body plethysmography was performed. The mice were sacrificed 24hours later. Blood samples were obtained, and serum of each sample was separated for IgE determination. Bronchoalveolar lavage fluid (BALF) and lung tissues were collected to measure IL-5 and IL-13 levels. Nuclear extracts of lung tissues were employed to assess nuclear factor kappa B (NF-κB) p65 binding activity. RESULTS Enhanced Pause (Penh) values were significantly increased in ovalbumin-sensitized and challenged mice (p<0.01). Administration of fenofibrate (10 and 30mg/kg) resulted in improved pulmonary function as shown by significantly lower Penh values (p<0.01). Interleukin (IL) -5 and IL-13 levels in BALF and lung tissues and immunoglobulin E (IgE) levels in serum were significantly elevated in the allergic mice. IL-5 levels in the lung tissues of mice treated with 1mg/kg fenofibrate (FEN1) group were significantly reduced (p<0.01). BALF and lung tissue IL-5 and IL-13 levels in mice treated with 10 and 30mg/kg fenofibrate, FEN10 and FEN30, respectively, were significantly diminished when compared to the ovalbumin-treated (OVA) group, whereas treatment with 1mg/kg fenofibrate resulted in insignificant changes. IgE levels in the serum of FEN30 group mice have shown a prominent reduction (p<0.01). NF-κB p65 binding activity was higher in mice sensitized and challenged with ovalbumin (p<0.01). NF-κB p65 binding activity was significantly reduced in allergic mice treated with 30mg/kg (p<0.01) fenofibrate. CONCLUSIONS In this study, we showed that administration of 10 and 30mg/kg fenofibrate effectively attenuated airway hyperresponsiveness and inflammation in a mouse model of allergic asthma, possibly through inhibition of NF-κB binding activity.
Collapse
Affiliation(s)
| | - Fatma Uysal
- Medical Pharmacology Department, Faculty of Medicine, Ankara Yıldırım Beyazıt University, Ankara, Türkiye
| | - Seyfullah Oktay Arslan
- Medical Pharmacology Department, Faculty of Medicine, Ankara Yıldırım Beyazıt University, Ankara, Türkiye.
| | - Saliha Ayşenur Çam Özünlü
- Medical Pharmacology, Ankara, Türkiye Department, Faculty of Medicine, Ankara Yıldırım Beyazıt University, Ankara, Türkiye
| | - Ayşegül Koç
- Ankara Yıldırım Beyazıt University, Ankara, Türkiye
| | - Ali Parlar
- Medical Pharmacology Department, Faculty of Medicine, University of Adıyaman, Adıyaman, Türkiye
| | - Keziban Korkmaz Bayram
- Medical Genetic Department, Faculty of Medicine, Ankara Yıldırım Beyazıt University, Ankara, Türkiye
| |
Collapse
|
7
|
Wu Z, Luan H, Huang J, Liao B, Xiao F. Migration inhibitory factor and cluster of differentiation 74-mediated dendritic cell apoptosis exacerbates acute acetaminophen-induced liver injury. Immun Inflamm Dis 2023; 11:e840. [PMID: 37102665 PMCID: PMC10108683 DOI: 10.1002/iid3.840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 03/09/2023] [Accepted: 03/29/2023] [Indexed: 04/28/2023] Open
Abstract
INTRODUCTION We investigated the role of macrophage migration inhibitory factor (MIF) on dendritic cells (DC) during acetaminophen (APAP)-induced acute liver injury (ALI) in mice. METHODS First, we randomly divided the mice into experimental (ALI model) and control groups, then intraperitoneally injected 600 mg/kg of APAP or phosphate-buffered saline, respectively. Then, we collected liver tissue and serum samples to evaluate liver inflammation using serum alanine aminotransferase level and hematoxylin and eosin (H&E) staining of liver tissues. Flow cytometry was used to identify changes in the quantity and percentage of DCs, as well as the expression of cluster of differentiation (CD) 74 and other apoptosis-related markers in the liver. Next, we randomly divided the mice into APAP-vehicles, APAP-bone marrow-derived dendritic cells (BMDCs), APAP-MIF, APAP-IgG (isotype immunoglobin G antibody) groups (four mice per group), after APAP injection, we injected control extracts, BMDCs, mouse recombinant MIF antibodies, or IgG antibodies into the tail vein. Lastly, the severity of the liver injury and the number of DCs were assessed. RESULTS The APAP-induced ALI mice had increased hepatic MIF expression but significantly lower amounts of hepatic DCs and apoptotic DCs than healthy mice; CD74 expression on the HDCs also increased markedly. Supplementing APAP-induced ALI mice with BMDCs or MIF antibodies significantly increased the number of hepatic DCs compared with the control mice, alleviating liver damage. CONCLUSION The MIF/CD74 signaling pathway may mediate hepatic DC apoptosis and promote liver damage.
Collapse
Affiliation(s)
- Zezhou Wu
- Department of Infectious DiseasesThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - He Luan
- Department of Infectious DiseasesThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Jinghui Huang
- Department of Infectious DiseasesThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Boming Liao
- Department of Infectious DiseasesThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Fang Xiao
- Department of Infectious DiseasesThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| |
Collapse
|
8
|
Zheng S, Chen N, Kang X, Hu Y, Shi S. Irisin alleviates FFA induced β-cell insulin resistance and inflammatory response through activating PI3K/AKT/FOXO1 signaling pathway. Endocrine 2022; 75:740-751. [PMID: 34546489 DOI: 10.1007/s12020-021-02875-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/11/2021] [Indexed: 01/21/2023]
Abstract
PURPOSE Type 2 diabetes mellitus is characterized by insulin resistance and β-cell dysfunction. Elevated free fatty acids-induced lipotoxicity may play a vital role in the pathogenesis of β-cell insulin resistance. Exercise-stimulated myokine irisin has been reported to be closely related to T2DM. However, its function on β-cell insulin signaling and the underlying mechanisms are only partially elucidated as yet. METHODS High-fat diet-fed C57BL/6J mice and palmitic acid-treated MIN6 cell models were utilized as lipotoxic models. Factors associated with β-cell insulin signaling transduction and inflammatory responses were assessed in these models. Furthermore, the role of irisin in β-cells and the underlying mechanisms were also explored. RESULTS Irisin effectively decreased lipid levels in HFD mice, enhanced glucose-stimulated insulin secretion and nullified the expressions of inflammatory cytokines in vivo and in vitro experiments. Moreover, irisin improved PI3K/AKT insulin signaling pathway and inhibited TLR4/NF-κB inflammatory signaling pathway in both islets of HFD mice and PA-treated MIN6 cells. Mechanistic analysis indicated that FOXO1 might serve as a bridge between the two pathways. CONCLUSION Irisin alleviates lipotoxicity-induced β-cell insulin resistance and inflammatory response through the activation of PI3K/AKT/FOXO1 signaling pathways and the inhibition of TLR4/NF-κB signaling pathways. Irisin might provide a novel therapeutic strategy for T2DM.
Collapse
Affiliation(s)
- Shuang Zheng
- School of Medicine, Shanghai University, Shanghai, 200444, China
- Department of Endocrinology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
| | - Ningxin Chen
- Department of Endocrinology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
| | - Xingjian Kang
- School of Stomatology, Wenzhou Medical University, Wenzhou, 325000, China
| | - Yaomin Hu
- Department of Endocrinology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
| | - Sheng Shi
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China.
| |
Collapse
|
9
|
Cardelo MP, Alcala-Diaz JF, Gutierrez-Mariscal FM, Lopez-Moreno J, Villasanta-Gonzalez A, de Larriva APA, Cruz-Ares SDL, Delgado-Lista J, Rodriguez-Cantalejo F, Luque RM, Ordovas JM, Perez-Martinez P, Camargo A, Lopez-Miranda J. Diabetes remission is modulated by branched chain amino acids according to the diet consumed: from the CORDIOPREV study. Mol Nutr Food Res 2021; 66:e2100652. [PMID: 34863046 DOI: 10.1002/mnfr.202100652] [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: 07/07/2021] [Revised: 11/24/2021] [Indexed: 11/08/2022]
Abstract
SCOPE BCAA plasma levels may be differentially associated with type 2 diabetes mellitus (T2DM) remission through the consumption of the Mediterranean diet (Med) and a low-fat (LF) diet. METHODS 183 newly-diagnosed T2DM patients within the CORDIOPREV study were randomized to consume the Med or a LF diet. BCAA plasma levels (isoleucine, leucine and valine) were measured at fasting and after 120 min of an oral glucose tolerance test (OGTT) at the baseline of the study and after 5 y of the dietary intervention. RESULTS Isoleucine, leucine and valine plasma levels after 120 min of an OGTT in the Med diet (N = 80) were associated by COX analysis with T2DM remission: HR per SD (95%CI): 0.53 (0.37-0.77), 0.75 (0.52-1.08) and 0.61 (0.45-0.82), respectively; no association was found in patients who consumed a LF diet (N = 103). BCAA plasma levels combined in a score showed a HR of 3.33 (1.55-7.19) of T2DM remission for patients with a high score values in the Med diet, while in those with a LF diet no association was found. CONCLUSION Our study suggests that BCAA measurements potentially be used as a tool to select the most suitable diet to induce T2DM remission by nutritional strategies. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Magdalena P Cardelo
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University, Hospital, Cordoba, 14004, Spain.,Department of Medicine (Medicine, Dermatology and Otorhinolaryngology), University of Cordoba, 4004, Cordoba, Spain.,Maimonides Biomedical Research Institute of Cordoba (IMIBIC).,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain
| | - Juan F Alcala-Diaz
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University, Hospital, Cordoba, 14004, Spain.,Department of Medicine (Medicine, Dermatology and Otorhinolaryngology), University of Cordoba, 4004, Cordoba, Spain.,Maimonides Biomedical Research Institute of Cordoba (IMIBIC).,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain
| | - Francisco M Gutierrez-Mariscal
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University, Hospital, Cordoba, 14004, Spain.,Department of Medicine (Medicine, Dermatology and Otorhinolaryngology), University of Cordoba, 4004, Cordoba, Spain.,Maimonides Biomedical Research Institute of Cordoba (IMIBIC).,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain
| | - Javier Lopez-Moreno
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University, Hospital, Cordoba, 14004, Spain.,Department of Medicine (Medicine, Dermatology and Otorhinolaryngology), University of Cordoba, 4004, Cordoba, Spain.,Maimonides Biomedical Research Institute of Cordoba (IMIBIC).,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain
| | - Alejandro Villasanta-Gonzalez
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University, Hospital, Cordoba, 14004, Spain.,Department of Medicine (Medicine, Dermatology and Otorhinolaryngology), University of Cordoba, 4004, Cordoba, Spain.,Maimonides Biomedical Research Institute of Cordoba (IMIBIC).,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain
| | - Antonio P Arenas- de Larriva
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University, Hospital, Cordoba, 14004, Spain.,Department of Medicine (Medicine, Dermatology and Otorhinolaryngology), University of Cordoba, 4004, Cordoba, Spain.,Maimonides Biomedical Research Institute of Cordoba (IMIBIC)
| | - Silvia de la Cruz-Ares
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University, Hospital, Cordoba, 14004, Spain.,Department of Medicine (Medicine, Dermatology and Otorhinolaryngology), University of Cordoba, 4004, Cordoba, Spain.,Maimonides Biomedical Research Institute of Cordoba (IMIBIC).,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain
| | - Javier Delgado-Lista
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University, Hospital, Cordoba, 14004, Spain.,Department of Medicine (Medicine, Dermatology and Otorhinolaryngology), University of Cordoba, 4004, Cordoba, Spain.,Maimonides Biomedical Research Institute of Cordoba (IMIBIC)
| | - Fernando Rodriguez-Cantalejo
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University, Hospital, Cordoba, 14004, Spain.,Department of Medicine (Medicine, Dermatology and Otorhinolaryngology), University of Cordoba, 4004, Cordoba, Spain.,Maimonides Biomedical Research Institute of Cordoba (IMIBIC)
| | - Raul M Luque
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University, Hospital, Cordoba, 14004, Spain.,Department of Medicine (Medicine, Dermatology and Otorhinolaryngology), University of Cordoba, 4004, Cordoba, Spain.,Maimonides Biomedical Research Institute of Cordoba (IMIBIC).,Biochemical Laboratory, Reina Sofia University Hospital, Córdoba, Spain.,Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Reina Sofía University Hospital, Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain
| | - Jose M Ordovas
- Nutrition and Genomics Laboratory, J.M.-US Department of Agriculture Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts, USA.,IMDEA Alimentación, Madrid, Spain, CNIC, Madrid, Spain
| | - Pablo Perez-Martinez
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University, Hospital, Cordoba, 14004, Spain.,Department of Medicine (Medicine, Dermatology and Otorhinolaryngology), University of Cordoba, 4004, Cordoba, Spain.,Maimonides Biomedical Research Institute of Cordoba (IMIBIC).,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain
| | - Antonio Camargo
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University, Hospital, Cordoba, 14004, Spain.,Department of Medicine (Medicine, Dermatology and Otorhinolaryngology), University of Cordoba, 4004, Cordoba, Spain.,Maimonides Biomedical Research Institute of Cordoba (IMIBIC).,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain
| | - Jose Lopez-Miranda
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University, Hospital, Cordoba, 14004, Spain.,Department of Medicine (Medicine, Dermatology and Otorhinolaryngology), University of Cordoba, 4004, Cordoba, Spain.,Maimonides Biomedical Research Institute of Cordoba (IMIBIC).,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain
| |
Collapse
|
10
|
Wang H, Cao J, Su JB, Wang XQ, Wang X, Zhang DM, Wang XH. Serum fatty acid-binding protein 4 levels and responses of pancreatic islet β-cells and α-cells in patients with type 2 diabetes. Diabetol Metab Syndr 2021; 13:70. [PMID: 34174950 PMCID: PMC8234651 DOI: 10.1186/s13098-021-00690-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/11/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Serum fatty acid-binding protein 4 (FABP4), as an intracellular lipid chaperone and adipokine, was reported to be related to the incidence of type 2 diabetes (T2D) and diabetic complications, but its association with pancreatic islet β-cell and α-cell functions has not been fully elucidated. So the present study was to investigate the serum FABP4 levels and responses of islet β-cells and α-cells in patients with T2D. METHODS 115 patients with T2D and 89 healthy controls (HC), who received serum FABP4 levels test, were recruited to participate in this study. Moreover, 75-g oral glucose tolerance test (OGTT) was performed in T2D patients to evaluate islet β-cell and α-cell functions. Systemic insulin sensitivity and overall insulin secretion of islet β-cell function were assessed by Matsuda index using C peptide (ISIM-cp) and ratio of the area under the C peptide curve to the glucose curve (AUCcp/glu) during OGTT, respectively. Fasting glucagon (Gluca0min) and postchallenge glucagon assessed by the area under the glucagon curve (AUCgluca) were determined during OGTT to evaluate islet α-cell function. And other various clinical variables were also measured in all participants. Skewed variables were natural log-transformed (ln), such as lnFABP4. RESULTS The serum FABP4 levels in T2D patients were significantly higher than those in HC (p < 0.05). And after partially adjusting for fasting plasma glucose, serum lnFABP4 levels were negatively correlated with lnISIM-cp (r = - 0.332, p < 0.001) and positively correlated with lnAUCcp/glu (r = 0.324, p < 0.001), lnGluca0min (r = 0.200, p = 0.040) and lnAUCgluca (r = 0.311, p < 0.001), respectively, in patients with T2D. Furthermore, when multiple linear regression analyses were applied to adjust for other various clinical variables, serum lnFABP4 levels were found to remain associated with lnISIM-cp (β = - 0.296, t = - 2.900, p = 0.005), lnAUCcp/glu (β = 0.223, t = 2.038, p = 0.046), lnGluca0min (β = 0.272, t = 2.330, p = 0.024) and lnAUCgluca (β = 0.341, t = 3.065, p = 0.004), respectively. CONCLUSION Increased serum FABP4 levels were closely associated with blunted insulin sensitivity, increased insulin secretion, and elevated fasting and postchallenge glucagon levels in patients with T2D.
Collapse
Affiliation(s)
- Hong Wang
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University, and First People’s Hospital of Nantong City, No. 6, Haierxiang North Road, Nantong, 226001 China
| | - Jie Cao
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University, and First People’s Hospital of Nantong City, No. 6, Haierxiang North Road, Nantong, 226001 China
| | - Jian-bin Su
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University, and First People’s Hospital of Nantong City, No. 6, Haierxiang North Road, Nantong, 226001 China
| | - Xue-qin Wang
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University, and First People’s Hospital of Nantong City, No. 6, Haierxiang North Road, Nantong, 226001 China
| | - Xing Wang
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University, and First People’s Hospital of Nantong City, No. 6, Haierxiang North Road, Nantong, 226001 China
| | - Dong-mei Zhang
- Medical Research Center, Affiliated Hospital 2 of Nantong University, and First People’s Hospital of Nantong City, No. 6, Haierxiang North Road, Nantong, 226001 China
| | - Xiao-hua Wang
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University, and First People’s Hospital of Nantong City, No. 6, Haierxiang North Road, Nantong, 226001 China
| |
Collapse
|
11
|
Wu M, Yang Z, Zhang C, Shi Y, Han W, Song S, Mu L, Du C, Shi Y. Inhibition of NLRP3 inflammasome ameliorates podocyte damage by suppressing lipid accumulation in diabetic nephropathy. Metabolism 2021; 118:154748. [PMID: 33675822 DOI: 10.1016/j.metabol.2021.154748] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 02/18/2021] [Accepted: 02/27/2021] [Indexed: 01/19/2023]
Abstract
BACKGROUND Nucleotide leukin-rich polypeptide 3 (NLRP3) inflammasome is documented as a potent target for treating metabolic diseases and inflammatory disorders. Our recent work demonstrated that inhibition of NLRP3 inflammasome activation inhibits renal inflammation and fibrosis in diabetic nephropathy. This study was to investigate the effect of NLRP3 inflammasome on podocyte injury and the underlying mechanism in diabetic nephropathy. METHODS In vivo, db/db mice were treated with MCC950, a NLRP3 inflammasome specific inhibitor. NLRP3 knockout (NKO) mice were induced to diabetes by intraperitoneal injections of streptozotocin (STZ). We assessed renal function, albuminuria, podocyte injury and glomerular lipid accumulation in diabetic mice. In vitro, apoptosis, cytoskeleton change, lipid accumulation, NF-κB p65 activation and reactive oxygen species (ROS) generation were evaluated in podocytes interfered with NLRP3 siRNA or MCC950 under high glucose (HG) conditions. In addition, the effect and mechanism of IL-1β on lipid accumulation was explored in podocytes exposed to normal glucose (NG) or HG. RESULTS MCC950 treatment improved renal function, attenuated albuminuria, mesangial expansion, podocyte loss, as well as glomerular lipid accumulation in db/db mice. The diabetes-induced podocyte loss and glomerular lipid accumulation were reversed in NLRP3 knockout mice. The increased expression of sterol regulatory element-binding protein1 (SREBP1) and SREBP2, and decreased expression of ATP-binding cassette A1 (ABCA1) in podocytes were reversed by MCC950 treatment or NLRP3 knockout in diabetic mice. In vitro, NLRP3 siRNA or MCC950 treatment markedly inhibited HG-induced apoptosis, cytoskeleton change, lipid accumulation, NF-κB p65 activation, and mitochondrial ROS production in cultured podocytes. In addition, BAY11-7082 or tempol treatment inhibited HG-induced lipid accumulation in podocytes. Moreover, exposure of IL-1β to podocytes induced lipid accumulation, NF-κB p65 activation and mitochondrial ROS generation. CONCLUSION Inhibition of NLRP3 inflammasome protects against podocyte damage through suppression of lipid accumulation in diabetic nephropathy. IL-1β/ROS/NF-κB p65 mediates diabetes-associated lipid accumulation in podocytes. The suppression of NLRP3 inflammasome activation may be an effective therapeutic approach to diabetic nephropathy.
Collapse
Affiliation(s)
- Ming Wu
- Department of Pathology, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Kidney Disease, Shijiazhuang 050017, China
| | - Zhifen Yang
- Department of Pathology, Hebei Medical University, Shijiazhuang 050017, China
| | - Chengyu Zhang
- Department of Pathology, Hebei Medical University, Shijiazhuang 050017, China
| | - Yu Shi
- Department of Pathology, Hebei Medical University, Shijiazhuang 050017, China
| | - Weixia Han
- Department of Pathology, Hebei Medical University, Shijiazhuang 050017, China
| | - Shan Song
- Department of Pathology, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Kidney Disease, Shijiazhuang 050017, China; Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang 050017, China
| | - Lin Mu
- Department of Pathology, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Kidney Disease, Shijiazhuang 050017, China
| | - Chunyang Du
- Department of Pathology, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Kidney Disease, Shijiazhuang 050017, China; Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang 050017, China
| | - Yonghong Shi
- Department of Pathology, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Kidney Disease, Shijiazhuang 050017, China; Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang 050017, China.
| |
Collapse
|
12
|
Li R, Song P, Tang G, Wei J, Rao L, Ma L, Jiang M, Huang J, Xu Q, Wu J, Lv Q, Yao D, Xiao B, Huang H, Lei L, Feng J, Mo B. Osthole Attenuates Macrophage Activation in Experimental Asthma by Inhibitingthe NF-ĸB/MIF Signaling Pathway. Front Pharmacol 2021; 12:572463. [PMID: 33828480 PMCID: PMC8020258 DOI: 10.3389/fphar.2021.572463] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 01/11/2021] [Indexed: 01/19/2023] Open
Abstract
Inhibition of activated macrophages is an alternative therapeutic strategy for asthma. We investigated whether a coumarin compound, osthole, isolated from Cnidium monnieri (L.) Cuss, alleviated macrophage activation in vivo and in vitro. Osthole could reduce expression of a marker of activated macrophages, cluster of differentiation (CD)206, in an ovalbumin-challenge model of asthma in mice. Osthole could also inhibit infiltration of inflammatory cells, collagen deposition and production of proinflammatory cytokines [interleukin (IL)-1β, tumor necrosis factor-ɑ, macrophage migration inhibitory factor (MIF)] in asthmatic mice. In vitro, expression of phosphorylated-IĸBɑ, MIF and M2 cytokines (Ym-1, Fizz-1, arginase-1) in IL-4-induced macrophages decreased upon exposure to the NF-ĸB inhibitor MG-132. In our short hairpin (sh)RNA-MIF-knockdown model, reduced expression of M2 cytokines was detected in the IL-4 + shRNA-MIF group. Osthole could attenuate the proliferation and migration of an IL-4-induced rat alveolar macrophages line (NR8383). Osthole could reduce IL-4-induced translocation of nuclear factor-kappa B (NF-ĸB) in NR8383 cells. Collectively, our results suggest that osthole ameliorates macrophage activation in asthma by suppressing the NF-ĸB/MIF signaling pathway, and might be a potential agent for treating asthma.
Collapse
Affiliation(s)
- Ruyi Li
- Key Laboratory of National Clinical Research Center for Respiratory Disease, Department of Respiratory and Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Peng Song
- Key Laboratory of Prevention and Treatment for Chronic Diseases by Traditional Chinese Medicine, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China.,Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guofang Tang
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Jianghong Wei
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Lizong Rao
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Libing Ma
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Ming Jiang
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Jianwei Huang
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Qing Xu
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Jingjie Wu
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Qian Lv
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Dong Yao
- Laboratory of Pulmonary Diseases, Guilin Medical University, Guilin, China
| | - Bo Xiao
- Laboratory of Pulmonary Diseases, Guilin Medical University, Guilin, China
| | - Haiming Huang
- Laboratory of Pulmonary Diseases, Guilin Medical University, Guilin, China
| | - Liping Lei
- Laboratory of Pulmonary Diseases, Guilin Medical University, Guilin, China
| | - Juntao Feng
- Key Laboratory of National Clinical Research Center for Respiratory Disease, Department of Respiratory and Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Biwen Mo
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guilin Medical University, Guilin, China
| |
Collapse
|
13
|
Hu HQ, Qiao JT, Liu FQ, Wang JB, Sha S, He Q, Cui C, Song J, Zang N, Wang LS, Sun Z, Chen L, Hou XG. The STING-IRF3 pathway is involved in lipotoxic injury of pancreatic β cells in type 2 diabetes. Mol Cell Endocrinol 2020; 518:110890. [PMID: 32781250 DOI: 10.1016/j.mce.2020.110890] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 05/26/2020] [Accepted: 06/01/2020] [Indexed: 02/07/2023]
Abstract
Lipotoxic injury of pancreatic β cells is an important pathological feature in type 2 diabetes mellitus (T2DM). Stimulator of interferon genes (STING) can recognize its own DNA leaked into the cytoplasm from damaged mitochondria or nuclei of the host cell, thus activating its downstream factor interferon regulatory factor 3 (IRF3), causing inflammation and apoptosis. The STING-IRF3 signaling pathway is closely related to glycolipid metabolism, but its relationship with the lipotoxicity of pancreatic β cells has rarely been reported. Here, we investigated the role of the STING-IRF3 signaling pathway in lipotoxicity-induced inflammation, apoptosis, and dysfunction of pancreatic β cells. We examined the activation of STING and IRF3 in islets of db/db mice and identified the role of the STING-IRF3 signaling pathway in palmitic acid (PA)-induced lipotoxic injury of INS-1, a rat insulinoma cell line. STING and phosphorylated IRF3 including downstream interferon-β were upregulated in islets of db/db mice and PA-induced INS-1 cells. Gene silencing of STING or IRF3 ameliorated PA-induced INS-1 cell inflammation and apoptosis, and reversed impaired insulin synthesis. Additionally, PA induced downregulation of the phosphoinositide 3-kinase-AKT signaling pathway, and impaired high glucose-stimulated insulin secretion was reversed after knockdown of STING or IRF3. Our results suggest that activation of the STING-IRF3 pathway triggers inflammation and apoptosis of pancreatic β cells, leading to β-cell damage and dysfunction. Hence, inhibition of this signaling pathway may represent a novel approach for β-cell protection in T2DM.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Cells, Cultured
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Insulin-Secreting Cells/drug effects
- Insulin-Secreting Cells/physiology
- Interferon Regulatory Factor-3/physiology
- Male
- Membrane Proteins/physiology
- Mice
- Mice, Transgenic
- Palmitic Acid/pharmacology
- Palmitic Acid/toxicity
- Phosphatidylinositol 3-Kinases/metabolism
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- H Q Hu
- Department of Endocrine and Metabolism, Qilu Hospital of Shandong University, Jinan, Shandong, China; Institute of Endocrinology and Metabolism, Shandong University, Jinan, Shandong, China
| | - J T Qiao
- Department of Endocrine and Metabolism, Qilu Hospital of Shandong University, Jinan, Shandong, China; Institute of Endocrinology and Metabolism, Shandong University, Jinan, Shandong, China
| | - F Q Liu
- Department of Endocrine and Metabolism, Qilu Hospital of Shandong University, Jinan, Shandong, China; Institute of Endocrinology and Metabolism, Shandong University, Jinan, Shandong, China; Key Laboratory of Endocrine and Metabolic Diseases, Shandong Province Medicine & Health, Jinan 250012, China
| | - J B Wang
- Department of Endocrine and Metabolism, Qilu Hospital of Shandong University, Jinan, Shandong, China; Institute of Endocrinology and Metabolism, Shandong University, Jinan, Shandong, China
| | - S Sha
- Department of Endocrine and Metabolism, Qilu Hospital of Shandong University, Jinan, Shandong, China; Institute of Endocrinology and Metabolism, Shandong University, Jinan, Shandong, China
| | - Q He
- Department of Endocrine and Metabolism, Qilu Hospital of Shandong University, Jinan, Shandong, China; Institute of Endocrinology and Metabolism, Shandong University, Jinan, Shandong, China
| | - C Cui
- Department of Endocrine and Metabolism, Qilu Hospital of Shandong University, Jinan, Shandong, China; Institute of Endocrinology and Metabolism, Shandong University, Jinan, Shandong, China
| | - J Song
- Department of Endocrine and Metabolism, Qilu Hospital of Shandong University, Jinan, Shandong, China; Institute of Endocrinology and Metabolism, Shandong University, Jinan, Shandong, China
| | - N Zang
- Department of Endocrine and Metabolism, Qilu Hospital of Shandong University, Jinan, Shandong, China; Institute of Endocrinology and Metabolism, Shandong University, Jinan, Shandong, China
| | - L S Wang
- Department of Endocrine and Metabolism, Qilu Hospital of Shandong University, Jinan, Shandong, China; Institute of Endocrinology and Metabolism, Shandong University, Jinan, Shandong, China
| | - Z Sun
- Department of Endocrine and Metabolism, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - L Chen
- Department of Endocrine and Metabolism, Qilu Hospital of Shandong University, Jinan, Shandong, China; Institute of Endocrinology and Metabolism, Shandong University, Jinan, Shandong, China; Key Laboratory of Endocrine and Metabolic Diseases, Shandong Province Medicine & Health, Jinan 250012, China.
| | - X G Hou
- Department of Endocrine and Metabolism, Qilu Hospital of Shandong University, Jinan, Shandong, China; Institute of Endocrinology and Metabolism, Shandong University, Jinan, Shandong, China; Key Laboratory of Endocrine and Metabolic Diseases, Shandong Province Medicine & Health, Jinan 250012, China.
| |
Collapse
|
14
|
Li M, Rong ZJ, Cao Y, Jiang LY, Zhong D, Li CJ, Sheng XL, Hu JZ, Lu HB. Utx Regulates the NF-κB Signaling Pathway of Natural Stem Cells to Modulate Macrophage Migration during Spinal Cord Injury. J Neurotrauma 2020; 38:353-364. [PMID: 32977735 DOI: 10.1089/neu.2020.7075] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Neural stem cells (NSCs) play vital roles in the homeostasis of neurological function. Ubiquitously transcribed tetratricopeptide repeat, X chromosome (UTX) is an important regulator of stem cell phenotypes. In our current study, we aimed to investigate whether the conditional knockout of UTX on neural stem cells alters macrophage assembly in response to spinal cord injury (SCI). Conditional knockout Utx of NSC (Utx-KO) mice was used to generate SCI models by the modified Allen method. We reported that neurological function and scar hyperplasia significantly improved in Utx-KO mice after SCI, accompanied by significantly reduced assembly of macrophages. With a 45-fold pathway array and Western blot, we found that Utx-KO could significantly inhibit NF-κB signaling activation and promote the synthesis and secretion of macrophage migration inhibitory factor (MIF) in NSCs. Administration of the selective NF-κB p65 activator betulinic acid and the selective MIF inhibitor ISO-1 confirmed that the activation of NF-κB p65 phosphorylation or inhibition of MIF could eliminate the benefits of Utx-KO in SCI, such as inhibition of macrophage aggregation and reduction in scar proliferation. This study confirmed that UTX in NSCs could alter macrophage migration and improve neurological function recovery after SCI in mice.
Collapse
Affiliation(s)
- Miao Li
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, People's Republic of China.,Xiangya Hospital, Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, People's Republic of China
| | - Zi-Jie Rong
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, People's Republic of China.,Xiangya Hospital, Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, People's Republic of China
| | - Yong Cao
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, People's Republic of China.,Xiangya Hospital, Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, People's Republic of China
| | - Li-Yuan Jiang
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, People's Republic of China.,Xiangya Hospital, Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, People's Republic of China
| | - Dong Zhong
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, People's Republic of China.,Xiangya Hospital, Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, People's Republic of China
| | - Cheng-Jun Li
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, People's Republic of China.,Xiangya Hospital, Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, People's Republic of China
| | - Xiao-Long Sheng
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, People's Republic of China.,Xiangya Hospital, Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, People's Republic of China
| | - Jian-Zhong Hu
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, People's Republic of China.,Xiangya Hospital, Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, People's Republic of China
| | - Hong-Bin Lu
- Xiangya Hospital, Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, People's Republic of China.,Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| |
Collapse
|
15
|
Zhao T, Ma J, Li L, Teng W, Tian Y, Ma Y, Wang W, Yan W, Jiao P. MKP-5 Relieves Lipotoxicity-Induced Islet β-Cell Dysfunction and Apoptosis via Regulation of Autophagy. Int J Mol Sci 2020; 21:ijms21197161. [PMID: 32998359 PMCID: PMC7582937 DOI: 10.3390/ijms21197161] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 09/16/2020] [Accepted: 09/25/2020] [Indexed: 01/28/2023] Open
Abstract
Mitogen-activated protein kinase phosphatase-5 (MKP-5) is a regulator of extracellular signaling that is known to regulate lipid metabolism. In this study, we found that obesity caused by a high-fat diet (HFD) decreased the expression of MKP-5 in the pancreas and primary islet cells derived from mice. Then, we further investigated the role of MKP-5 in the protection of islet cells from lipotoxicity by modulating MKP-5 expression. As a critical inducer of lipotoxicity, palmitic acid (PA) was used to treat islet β-cells. We found that MKP-5 overexpression restored PA-mediated autophagy inhibition in Rin-m5f cells and protected these cells from PA-induced apoptosis and dysfunction. Consistently, a lack of MKP-5 aggravated the adverse effects of lipotoxicity. Islet cells from HFD-fed mice were infected using recombinant adenovirus expressing MKP-5 (Ad-MKP-5), and we found that Ad-MKP-5 was able to alleviate HFD-induced apoptotic protein activation and relieve the HFD-mediated inhibition of functional proteins. Notably, HFD-mediated impairments in autophagic flux were restored by Ad-MKP-5 transduction. Furthermore, the autophagy inhibitor 3-methyladenine (3-MA) was used to treat Rin-m5f cells, confirming that the MKP-5 overexpression suppressed apoptosis, dysfunction, inflammatory response, and oxidative stress induced by PA via improving autophagic signaling. Lastly, employing c-Jun amino-terminal kinas (JNK), P38, or extracellular-regulated kinase (ERK) inhibitors, we established that the JNK and P38 MAPK pathways were involved in the MKP-5-mediated apoptosis, dysfunction, and autophagic inhibition observed in islet β cells in response to lipotoxicity.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Weiqun Yan
- Correspondence: (W.Y.); (P.J.); Tel.: +86-431-8561-9289 (P.J.)
| | - Ping Jiao
- Correspondence: (W.Y.); (P.J.); Tel.: +86-431-8561-9289 (P.J.)
| |
Collapse
|
16
|
Puchałowicz K, Rać ME. The Multifunctionality of CD36 in Diabetes Mellitus and Its Complications-Update in Pathogenesis, Treatment and Monitoring. Cells 2020; 9:cells9081877. [PMID: 32796572 PMCID: PMC7465275 DOI: 10.3390/cells9081877] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/04/2020] [Accepted: 08/09/2020] [Indexed: 02/08/2023] Open
Abstract
CD36 is a multiligand receptor contributing to glucose and lipid metabolism, immune response, inflammation, thrombosis, and fibrosis. A wide range of tissue expression includes cells sensitive to metabolic abnormalities associated with metabolic syndrome and diabetes mellitus (DM), such as monocytes and macrophages, epithelial cells, adipocytes, hepatocytes, skeletal and cardiac myocytes, pancreatic β-cells, kidney glomeruli and tubules cells, pericytes and pigment epithelium cells of the retina, and Schwann cells. These features make CD36 an important component of the pathogenesis of DM and its complications, but also a promising target in the treatment of these disorders. The detrimental effects of CD36 signaling are mediated by the uptake of fatty acids and modified lipoproteins, deposition of lipids and their lipotoxicity, alterations in insulin response and the utilization of energy substrates, oxidative stress, inflammation, apoptosis, and fibrosis leading to the progressive, often irreversible organ dysfunction. This review summarizes the extensive knowledge of the contribution of CD36 to DM and its complications, including nephropathy, retinopathy, peripheral neuropathy, and cardiomyopathy.
Collapse
|
17
|
Liang DS, Huang AR, Lin MM, Xia XJ, Jin YM. Pyrrolidine dithiocarbamate and dexamethasone are novel treatments of Acute Exogenous Lipoid Pneumonia. Cytokine 2020; 133:155122. [PMID: 32446224 DOI: 10.1016/j.cyto.2020.155122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 04/29/2020] [Accepted: 05/06/2020] [Indexed: 11/18/2022]
Abstract
BACKGROUND Acute exogenous lipoid pneumonia (AELP) is characterized by pulmonary inflammation. This mainly occur in children who have ingested sewing machine oil or other mineral oils accidentally. Despite emerging evidences revealing that inhibiting inflammation improves acute exogenous lipoid pneumonia, the actual process of inhibiting inflammation remains unknown. This study aimed to evaluate the effects of PDTC and dexamethasone on AELP to gain insight into the mechanism of AELP. METHODS The experimental rats were randomly divided into 10 groups: NS control group (NS3 group, NS5 group), Oil inhalation group (AE3 group, AE5 group), PDTC intervention group (PDTC3 group, PDTC5 group), DXM intervention group (DXM3 group, DXM5 group), PDTC + DXM combined intervention group (PDTC + DXM3 group, PDTC + DXM 5 group). Enzyme-linked immunosorbent assay (ELISA) was used to determine concentrations of macrophage migration inhibitory factor (MIF), interleukin-6 (IL-6) and tumor necrosis factor-α (TNFα) in bronchoalveolar lavage fluid (BALF) and serum samples. On the other hand, western blotting was used to measure the expression levels of nuclear factor-κB p65 (NF-κB p65) and b-cell leukemia 2 (Bcl-2) in the lungs. Hematoxylin and Eosin (H&E) staining was performed to evaluate changes in the lung tissue. The wet-to-dry lung weight ratio was subsequently used to determine the pulmonary edema of the lungs. RESULTS There were increased MIF levels in both serum and BALF samples of the AE group. Pyrrolidine dithiocarbamate (PDTC) and dexamethasone (DXM) independently and in combination reduced pulmonary inflammation induced by the sewing machine oil by regulating MIF expression. TNF-α and IL-6 levels in serum and BALF samples of the AE group were higher than those of the NS control animals. However, their levels decreased after treatment with either PDTC, DXM or PDTC + DXM. Similarly, NF-κBp65 expression increased after oil inhalation but decreased after treatment with either PDTC, DXM or PDTC + DXM. PDTC, DXM and PDTC + DXM treatment significantly reduced pulmonary inflammation and pulmonary edema of the lung tissue following induction of acute exogenous lipoid pneumonia. CONCLUSIONS Individual or combined use of PDTC and DXM can ameliorate pulmonary inflammation induced by inhalation of sewing machine oil by inhibiting the NF-κB pathway in young rats. These findings provide novel insights that will greatly contribute in treatment of AELP.
Collapse
Affiliation(s)
- Dong-Shi Liang
- Department of Pediatrics, Second Affiliated Hospital &Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Ai-Rong Huang
- Department of Pediatrics, Second Affiliated Hospital &Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Miao-Miao Lin
- Department of Pediatrics, Second Affiliated Hospital &Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Xiao-Jiao Xia
- Department of Pediatrics, Second Affiliated Hospital &Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Yi-Mei Jin
- Department of Pediatrics, Second Affiliated Hospital &Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China.
| |
Collapse
|
18
|
Ježek P, Jabůrek M, Plecitá-Hlavatá L. Contribution of Oxidative Stress and Impaired Biogenesis of Pancreatic β-Cells to Type 2 Diabetes. Antioxid Redox Signal 2019; 31:722-751. [PMID: 30450940 PMCID: PMC6708273 DOI: 10.1089/ars.2018.7656] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 11/05/2018] [Indexed: 12/14/2022]
Abstract
Significance: Type 2 diabetes development involves multiple changes in β-cells, related to the oxidative stress and impaired redox signaling, beginning frequently by sustained overfeeding due to the resulting lipotoxicity and glucotoxicity. Uncovering relationships among the dysregulated metabolism, impaired β-cell "well-being," biogenesis, or cross talk with peripheral insulin resistance is required for elucidation of type 2 diabetes etiology. Recent Advances: It has been recognized that the oxidative stress, lipotoxicity, and glucotoxicity cannot be separated from numerous other cell pathology events, such as the attempted compensation of β-cell for the increased insulin demand and dynamics of β-cell biogenesis and its "reversal" at dedifferentiation, that is, from the concomitantly decreasing islet β-cell mass (also due to transdifferentiation) and low-grade islet or systemic inflammation. Critical Issues: At prediabetes, the compensation responses of β-cells, attempting to delay the pathology progression-when exaggerated-set a new state, in which a self-checking redox signaling related to the expression of Ins gene expression is impaired. The resulting altered redox signaling, diminished insulin secretion responses to various secretagogues including glucose, may lead to excretion of cytokines or chemokines by β-cells or excretion of endosomes. They could substantiate putative stress signals to the periphery. Subsequent changes and lasting glucolipotoxicity promote islet inflammatory responses and further pathology spiral. Future Directions: Should bring an understanding of the β-cell self-checking and related redox signaling, including the putative stress signal to periphery. Strategies to cure or prevent type 2 diabetes could be based on the substitution of the "wrong" signal by the "correct" self-checking signal.
Collapse
Affiliation(s)
- Petr Ježek
- Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Martin Jabůrek
- Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Lydie Plecitá-Hlavatá
- Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
19
|
Ren X, Chen N, Chen Y, Liu W, Hu Y. TRB3 stimulates SIRT1 degradation and induces insulin resistance by lipotoxicity via COP1. Exp Cell Res 2019; 382:111428. [PMID: 31125554 DOI: 10.1016/j.yexcr.2019.05.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 04/05/2019] [Accepted: 05/08/2019] [Indexed: 11/26/2022]
Abstract
Fatty acid-induced lipotoxicity plays an important role in the pathogenesis of diabetes mellitus. Our previous studies have documented that lipotoxicity contributes to the onset and development of diabetes via insulin resistance and/or compromised function of the pancreatic β-cells. However, the underlying molecular mechanisms associating lipotoxicity with insulin resistance remain to be fully elucidated. In this study, we explored the role of TRB3-COP1-SIRT1 in lipotoxicity leading to insulin resistance in hepatocytes. High fat diet (HFD)-fed mice and hepG2 cells stimulated with palmitate were utilized as models of lipid metabolism disorders. We analyzed the interactions of SIRT1 and COP1 with each other and with TRB3 using co-immunoprecipitation, western blotting. SIRT1 ubiquitination was also explored. Animal and cell experiments showed that lipotoxicity induced SIRT1 down-regulation at the protein level without altering the mRNA level, whereas, lipotoxicity led to up-regulation of TRB3 and COP1 at both the gene and protein levels. Mechanistic analysis indicated that COP1 functioned as an E3 Ub-ligase of SIRT1, responsible for its proteasomal degradation under lipotoxic conditions. TRB3 recruited COP1 to SIRT1 to promote its ubiquitination. Our data indicated for the first time that TRB3-COP1-SIRT1 pathway played an important role in lipotoxicity leading to insulin resistance in hepatocytes, and suggested that COP1 could be a potential therapeutic choice for the treatment of diabetes mellitus, with lipotoxicity being the important pathomechanism.
Collapse
Affiliation(s)
- Xingxing Ren
- Department of Endocrinology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
| | - Ningxin Chen
- Department of Endocrinology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
| | - Yawen Chen
- Department of Endocrinology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
| | - Wei Liu
- Department of Endocrinology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China.
| | - Yaomin Hu
- Department of Endocrinology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China.
| |
Collapse
|
20
|
GPR120 protects lipotoxicity-induced pancreatic β-cell dysfunction through regulation of PDX1 expression and inhibition of islet inflammation. Clin Sci (Lond) 2019; 133:101-116. [PMID: 30523046 DOI: 10.1042/cs20180836] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 11/29/2018] [Accepted: 12/05/2018] [Indexed: 12/23/2022]
Abstract
G-protein coupled receptor 120 (GPR120) has been shown to act as an omega-3 unsaturated fatty acid sensor and is involved in insulin secretion. However, the underlying mechanism in pancreatic β cells remains unclear. To explore the potential link between GPR120 and β-cell function, its agonists docosahexaenoic acid (DHA) and GSK137647A were used in palmitic acid (PA)-induced pancreatic β-cell dysfunction, coupled with GPR120 knockdown (KD) in MIN6 cells and GPR120 knockout (KO) mice to identify the underlying signaling pathways. In vitro and ex vivo treatments of MIN6 cells and islets isolated from wild-type (WT) mice with DHA and GSK137647A restored pancreatic duodenal homeobox-1 (PDX1) expression levels and β-cell function via inhibiting PA-induced elevation of proinflammatory chemokines and activation of nuclear factor κB, c-Jun amino (N)-terminal kinases1/2 and p38MAPK signaling pathways. On the contrary, these GPR120 agonism-mediated protective effects were abolished in GPR120 KD cells and islets isolated from GPR120 KO mice. Furthermore, GPR120 KO mice displayed glucose intolerance and insulin resistance relative to WT littermates, and β-cell functional related genes were decreased while inflammation was exacerbated in islets with increased macrophages in pancreas from GPR120 KO mice. DHA and GSK137647A supplementation ameliorated glucose tolerance and insulin sensitivity, as well as improved Pdx1 expression and islet inflammation in diet-induced obese WT mice, but not in GPR120 KO mice. These findings indicate that GPR120 activation is protective against lipotoxicity-induced pancreatic β-cell dysfunction, via the mediation of PDX1 expression and inhibition of islet inflammation, and that GPR120 activation may serve as a preventative and therapeutic target for obesity and diabetes.
Collapse
|
21
|
Daems C, Welsch S, Boughaleb H, Vanderroost J, Robert A, Sokal E, Lysy PA. Early Treatment with Empagliflozin and GABA Improves β-Cell Mass and Glucose Tolerance in Streptozotocin-Treated Mice. J Diabetes Res 2019; 2019:2813489. [PMID: 31467926 PMCID: PMC6701376 DOI: 10.1155/2019/2813489] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 05/31/2019] [Accepted: 06/23/2019] [Indexed: 12/27/2022] Open
Abstract
While the autoimmune character of T1D (type 1 diabetes) is being challenged, it is currently recognized that inflammation plays a key role in its development. We hypothesized that glucotoxicity could contribute to β-cell mass destruction through participation in islet inflammation. We evaluated the potential of empagliflozin (EMPA) and GABA (gamma-aminobutyric acid) to protect β-cell mass against glucotoxicity and to increase β-cell mass after diagnosis of T1D. Empagliflozin is a SGLT2 (sodium-dependent glucose cotransporter) inhibitor which thereby blocks glucose recapture by the kidney and promotes glucose excretion in urine. GABA is an inhibitory neurotransmitter, which stimulates α-to-β cell transdifferentiation. In streptozotocin-treated mice, empagliflozin and/or GABA were delivered for a period of five days or three weeks. As compared to untreated T1D mice, EMPA-treated T1D mice had decreased FFA (free fatty acid) levels and improved glucose homeostasis. EMPA-treated T1D mice had higher islet density, with preserved architecture, compared to T1D mice, and EMPA-treated T1D mice also differed from T1D mice by the total absence of immune cell infiltration within islets. Islets from EMPA-treated mice were also less subjected to ER (endoplasmic reticulum) stress and inflammation, as shown by qPCR analysis. Glucose homeostasis parameters and islet area/pancreas area ratio improved, as compared to diabetic controls, when T1D mice were treated for three weeks with GABA and EMPA. T1D EMPA+GABA mice had higher glucagon levels than T1D mice, without modifications of glucagon area/islet area ratios. In conclusion, empagliflozin and GABA, used in monotherapy in streptozotocin-induced diabetic mice, have positive effects on β-cell mass preservation or proliferation through an indirect effect on islet cell inflammation and ER stress. Further research is mandatory to evaluate whether empagliflozin and GABA may be a potential therapeutic target for the protection of β-cell mass after new-onset T1D.
Collapse
Affiliation(s)
- Caroline Daems
- Pôle PEDI, Institut de Recherche Expérimentale et Clinique, UCLouvain, Av. Hippocrate 10, B-1200 Brussels, Belgium
| | - Sophie Welsch
- Pôle PEDI, Institut de Recherche Expérimentale et Clinique, UCLouvain, Av. Hippocrate 10, B-1200 Brussels, Belgium
| | - Hasnae Boughaleb
- Pôle PEDI, Institut de Recherche Expérimentale et Clinique, UCLouvain, Av. Hippocrate 10, B-1200 Brussels, Belgium
| | - Juliette Vanderroost
- Pôle PEDI, Institut de Recherche Expérimentale et Clinique, UCLouvain, Av. Hippocrate 10, B-1200 Brussels, Belgium
| | - Annie Robert
- Pôle d'Epidémiologie et Biostatistique, Institut de Recherche Expérimentale et Clinique, UCLouvain, Av. Hippocrate 10, B-1200 Brussels, Belgium
| | - Etienne Sokal
- Pôle PEDI, Institut de Recherche Expérimentale et Clinique, UCLouvain, Av. Hippocrate 10, B-1200 Brussels, Belgium
| | - Philippe A. Lysy
- Pôle PEDI, Institut de Recherche Expérimentale et Clinique, UCLouvain, Av. Hippocrate 10, B-1200 Brussels, Belgium
| |
Collapse
|
22
|
Shen X, Fan B, Hu X, Luo L, Yan Y, Yang L. Metformin Reduces Lipotoxicity-Induced Meta-Inflammation in β-Cells through the Activation of GPR40-PLC-IP3 Pathway. J Diabetes Res 2019; 2019:7602427. [PMID: 31950065 PMCID: PMC6948338 DOI: 10.1155/2019/7602427] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 08/01/2019] [Accepted: 09/04/2019] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND AND PURPOSE Metformin, a widely used antidiabetic drug, has been shown to have anti-inflammatory properties; nevertheless, its influence on β-cell meta-inflammation remains unclear. The following study investigated the effects of metformin on meta-inflammatory in β-cells and whether the underlying mechanisms were associated with the G protein-coupled receptor 40-phospholipase C-inositol 1, 4, 5-trisphosphate (GPR40-PLC-IP3) pathway. MATERIALS AND METHODS Lipotoxicity-induced β-cells and the high-fat diet-induced obese rat model were used in the study. RESULTS Metformin-reduced lipotoxicity-induced β-cell meta-inflammatory injury was associated with the expression of GPR40. GPR40 was involved in metformin reversing metabolic inflammation key marker TLR4 activation-mediated β-cell injury. Furthermore, downstream signaling protein PLC-IP3 of GPR40 was involved in the protective effect of metformin on meta-inflammation, and the above process of metformin was partially regulated by AMPK activity. In addition, the anti-inflammatory effects of metformin were observed in obese rats. CONCLUSION Metformin can reduce lipotoxicity-induced meta-inflammation in β-cells through the regulation of the GPR40-PLC-IP3 pathway and partially via the regulation of AMPK activity.
Collapse
Affiliation(s)
- Ximei Shen
- Endocrinology Department, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005 Fujian, China
- Diabetes Research Institute of Fujian Province, Fuzhou, 350005 Fujian, China
| | - Beibei Fan
- Endocrinology Department, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005 Fujian, China
| | - Xin Hu
- Endocrinology Department, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005 Fujian, China
| | - Liufen Luo
- Endocrinology Department, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005 Fujian, China
| | - Yuanli Yan
- Endocrinology Department, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005 Fujian, China
| | - Liyong Yang
- Endocrinology Department, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005 Fujian, China
- Diabetes Research Institute of Fujian Province, Fuzhou, 350005 Fujian, China
| |
Collapse
|
23
|
The Protective Effects of Clams on Hypercholesterolemia in Late-Stage Triple-Transgenic Alzheimer's Diseased Mice Hearts. Mar Drugs 2018; 16:md16080263. [PMID: 30071640 PMCID: PMC6117677 DOI: 10.3390/md16080263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 07/18/2018] [Accepted: 07/28/2018] [Indexed: 12/17/2022] Open
Abstract
To investigate a high cholesterol diet in Alzheimer’s disease (AD) mice, they were fed with (2% cholesterol) in five groups with a control group, AD mice group, AD mice plus Meretrix lusoria group, AD mice plus Geloina eros group, and, AD mice plus Corbicula fluminea group for three months, and treated with the fatty acid profiles of clams by gas chromatography (GC). The results showed that treatment with clams for three months reduced Fas/L and Caspase-3 in the Meretrix lusoria and Geloina eros groups, but Fas-associated death domain (FADD) and Caspase-8 were strongly reduced in the Geloina eros group. For the mitochondria-dependent apoptotic pathway, the reduction of apoptosis proteins were observed in the hearts of clams-treated AD mice. BAK and Caspase-9 was reduced in the Meretrix lusoria group, but Caspase-3 and Cytochrome-c were reduced in Geloina eros group. Enhancement of survival proteins p-AKT, p-IGF1R, p-PI3K, Bcl-XL, Bcl2, and the longevity SIRT1 signaling proteins, p-AMPK-α, SIRT1, PGC1-α, p-FOXO3 were observed in clams-treated mice and even more strongly enhanced in the Meretrix lusoria, Geloina eros and Corbicula fluminea groups. This study observed that the ingestion of clams caused a reduction of apoptosis proteins and enhancement of survival and SIRT1 signaling proteins in the hearts.
Collapse
|
24
|
Fatty Acid-Stimulated Insulin Secretion vs. Lipotoxicity. Molecules 2018; 23:molecules23061483. [PMID: 29921789 PMCID: PMC6100479 DOI: 10.3390/molecules23061483] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/13/2018] [Accepted: 06/15/2018] [Indexed: 12/29/2022] Open
Abstract
Fatty acid (FA)-stimulated insulin secretion (FASIS) is reviewed here in contrast to type 2 diabetes etiology, resulting from FA overload, oxidative stress, intermediate hyperinsulinemia, and inflammation, all converging into insulin resistance. Focusing on pancreatic islet β-cells, we compare the physiological FA roles with the pathological ones. Considering FAs not as mere amplifiers of glucose-stimulated insulin secretion (GSIS), but as parallel insulin granule exocytosis inductors, partly independent of the KATP channel closure, we describe the FA initiating roles in the prediabetic state that is induced by retardations in the glycerol-3-phosphate (glucose)-promoted glycerol/FA cycle and by the impaired GPR40/FFA1 (free FA1) receptor pathway, specifically in its amplification by the redox-activated mitochondrial phospholipase, iPLA2γ. Also, excessive dietary FAs stimulate intestine enterocyte incretin secretion, further elevating GSIS, even at low glucose levels, thus contributing to diabetic hyperinsulinemia. With overnutrition and obesity, the FA overload causes impaired GSIS by metabolic dysbalance, paralleled by oxidative and metabolic stress, endoplasmic reticulum stress and numerous pro-apoptotic signaling, all leading to decreased β-cell survival. Lipotoxicity is exerted by saturated FAs, whereas ω-3 polyunsaturated FAs frequently exert antilipotoxic effects. FA-facilitated inflammation upon the recruitment of excess M1 macrophages into islets (over resolving M2 type), amplified by cytokine and chemokine secretion by β-cells, leads to an inevitable failure of pancreatic β-cells.
Collapse
|
25
|
Nishihama K, Yasuma T, Yano Y, D' Alessandro-Gabazza CN, Toda M, Hinneh JA, Baffour Tonto P, Takeshita A, Totoki T, Mifuji-Moroka R, Kobayashi T, Iwasa M, Takei Y, Morser J, Cann I, Gabazza EC. Anti-apoptotic activity of human matrix metalloproteinase-2 attenuates diabetes mellitus. Metabolism 2018; 82:88-99. [PMID: 29366755 DOI: 10.1016/j.metabol.2018.01.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 01/05/2018] [Accepted: 01/18/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND Chronic progression of diabetes is associated with decreased pancreatic islet mass due to apoptosis of β-cells. Patients with diabetes have increased circulating matrix metalloproteinase-2 (MMP2); however, the physiological significance has remained elusive. This study tested the hypothesis that MMP2 inhibits cell apoptosis, including islet β-cells. METHODS Samples from diabetic patients and newly developed transgenic mice overexpressing human MMP2 (hMMP2) were harnessed, and diabetes was induced with streptozotocin. RESULTS Circulating hMMP2 was significantly increased in diabetic patients compared to controls and significantly correlated with the serum C-peptide levels. The diabetic hMMP2 transgenic mice showed significant improvements in glycemia, glucose tolerance and insulin secretion compared to diabetic wild type mice. Importantly, the increased hMMP2 levels in mice correlated with significant reduction in islet β-cell apoptosis compared to wild-type counterparts, and an inhibitor of hMMP2 reversed this mitigating activity against diabetes. The increased activation of Akt and BAD induced by hMMP2 in β-cells compared to controls, links this signaling pathway to the anti-apoptotic activity of hMMP2, a property that was reversible by both an hMMP2 inhibitor and antibody against integrin-β3. CONCLUSION Overall, this study demonstrates that increased expression of hMMP2 may attenuate the severity of diabetes by protecting islet β-cells from apoptosis through an integrin-mediated activation of the Akt/BAD pathway.
Collapse
Affiliation(s)
- Kota Nishihama
- Department of Diabetes, Metabolism and Endocrinology, Mie University Graduate School of Medicine, Edobashi 2-174, Tsu, Mie 514-8507, Japan
| | - Taro Yasuma
- Department of Diabetes, Metabolism and Endocrinology, Mie University Graduate School of Medicine, Edobashi 2-174, Tsu, Mie 514-8507, Japan; Department of Immunology, Mie University Graduate School of Medicine, Edobashi 2-174, Tsu, Mie 514-8507, Japan
| | - Yutaka Yano
- Department of Diabetes, Metabolism and Endocrinology, Mie University Graduate School of Medicine, Edobashi 2-174, Tsu, Mie 514-8507, Japan
| | - Corina N D' Alessandro-Gabazza
- Department of Immunology, Mie University Graduate School of Medicine, Edobashi 2-174, Tsu, Mie 514-8507, Japan; Microbiome Metabolic Engineering Theme, Carl R. Woese Biology Institute for Genomic Biology, Department of Animal Sciences, Department of Microbiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, United States
| | - Masaaki Toda
- Department of Immunology, Mie University Graduate School of Medicine, Edobashi 2-174, Tsu, Mie 514-8507, Japan
| | - Josephine A Hinneh
- Department of Immunology, Mie University Graduate School of Medicine, Edobashi 2-174, Tsu, Mie 514-8507, Japan
| | - Prince Baffour Tonto
- Department of Immunology, Mie University Graduate School of Medicine, Edobashi 2-174, Tsu, Mie 514-8507, Japan
| | - Atsuro Takeshita
- Department of Diabetes, Metabolism and Endocrinology, Mie University Graduate School of Medicine, Edobashi 2-174, Tsu, Mie 514-8507, Japan
| | - Toshiaki Totoki
- Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine, Edobashi 2-174, Tsu, Mie 514-8507, Japan
| | - Rumi Mifuji-Moroka
- Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine, Edobashi 2-174, Tsu, Mie 514-8507, Japan
| | - Tetsu Kobayashi
- Department of Pulmonary and Critical Care Medicine, Mie University Graduate School of Medicine, Edobashi 2-174, Tsu, Mie 514-8507, Japan
| | - Motoh Iwasa
- Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine, Edobashi 2-174, Tsu, Mie 514-8507, Japan
| | - Yoshiyuki Takei
- Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine, Edobashi 2-174, Tsu, Mie 514-8507, Japan
| | - John Morser
- Division of Hematology, Stanford School of Medicine, 269 Campus Drive, CCSR 1155, Stanford, CA 94305-5156, United States
| | - Isaac Cann
- Microbiome Metabolic Engineering Theme, Carl R. Woese Biology Institute for Genomic Biology, Department of Animal Sciences, Department of Microbiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, United States
| | - Esteban C Gabazza
- Department of Immunology, Mie University Graduate School of Medicine, Edobashi 2-174, Tsu, Mie 514-8507, Japan.
| |
Collapse
|
26
|
Khan S, Kowluru A. CD36 mediates lipid accumulation in pancreatic beta cells under the duress of glucolipotoxic conditions: Novel roles of lysine deacetylases. Biochem Biophys Res Commun 2017; 495:2221-2226. [PMID: 29274335 DOI: 10.1016/j.bbrc.2017.12.111] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 12/19/2017] [Indexed: 12/13/2022]
Abstract
The cluster of differentiation 36 (CD36) is implicated in the intake of long-chain fatty acids and fat storage in various cell types including the pancreatic beta cell, thus contributing to the pathogenesis of metabolic stress and diabetes. Recent evidence indicates that CD36 undergoes post-translational modifications such as acetylation-deacetylation. However, putative roles of such modifications in its functional activation and onset of beta cell dysregulation under the duress of glucolipotoxicity (GLT) remain largely unknown. Using pharmacological approaches, we validated, herein, the hypothesis that acetylation-deacetylation signaling steps are involved in CD36-mediated lipid accumulation and downstream apoptotic signaling in pancreatic beta (INS-1832/13) cells under GLT. Exposure of these cells to GLT resulted in significant lipid accumulation without affecting the CD36 expression. Sulfo-n-succinimidyl oleate (SSO), an irreversible inhibitor of CD36, significantly attenuated lipid accumulation under GLT conditions, thus implicating CD36 in this metabolic step. Furthermore, trichostatin A (TSA) or valproic acid (VPA), known inhibitors of lysine deacetylases, markedly suppressed GLT-associated lipid accumulation with no discernible effects on CD36 expression. Lastly, SSO or TSA prevented caspase 3 activation in INS-1832/13 cells exposed to GLT conditions. Based on these findings, we conclude that an acetylation-deacetylation signaling step might regulate CD36 functional activity and subsequent lipid accumulation and caspase 3 activation in pancreatic beta cells exposed to GLT conditions. Identification of specific lysine deacetylases that control CD36 function should provide novel clues for the prevention of beta-cell dysfunction under GLT.
Collapse
Affiliation(s)
- Sabbir Khan
- β-Cell Biochemistry Laboratory, John D. Dingell VA Medical Center, and Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI, 48201, USA
| | - Anjaneyulu Kowluru
- β-Cell Biochemistry Laboratory, John D. Dingell VA Medical Center, and Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI, 48201, USA.
| |
Collapse
|