1
|
Roshdy M, Zaky DA, Abbas SS, Abdallah DM. Niacin, an innovative protein kinase-C-dependent endoplasmic reticulum stress reticence in murine Parkinson's disease. Life Sci 2024; 351:122865. [PMID: 38914304 DOI: 10.1016/j.lfs.2024.122865] [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: 02/18/2024] [Revised: 06/08/2024] [Accepted: 06/20/2024] [Indexed: 06/26/2024]
Abstract
AIMS Niacin (NIA) supplementation showed effectiveness against Parkinson's disease (PD) in clinical trials. The depletion of NAD and endoplasmic reticulum stress response (ERSR) are implicated in the pathogenesis of PD, but the potential role for NAD precursors on ERSR is not yet established. This study was undertaken to decipher NIA molecular mechanisms against PD-accompanied ERSR, especially in relation to PKC. METHODS Alternate-day-low-dose-21 day-subcutaneous exposure to rotenone (ROT) in rats induced PD. Following the 5th ROT injection, rats received daily doses of either NIA alone or preceded by the PKC inhibitor tamoxifen (TAM). Extent of disease progression was assessed by behavioral, striatal biochemical and striatal/nigral histopathological/immunohistochemical analysis. KEY FINDINGS Via activating PKC/LKB1/AMPK stream, NIA post-treatment attenuated the ERSR reflected by the decline in ATF4, ATF6 and XBP1s to downregulate the apoptotic markers, CHOP/GADD153, p-JNK and active caspase-3. Such amendments congregated in motor activity/coordination improvements in open field and rotarod tasks, enhanced grid test latency and reduced overall PD scores, while boosting nigral/striatal tyrosine hydroxylase immunoreactivity and increasing intact neurons (Nissl stain) in both SNpc and striatum that showed less neurodegeneration (H&E stain). To different extents, TAM reverted all the NIA-related actions to prove PKC as a fulcrum in conveying the drug neurotherapeutic potential. SIGNIFICANCE PKC activation is a pioneer mechanism in the drug ERSR inhibitory anti-apoptotic modality to clarify NIA promising clinical and potent preclinical anti-PD efficacy. This kinase can be tagged as a druggable target for future add-on treatments that can assist dopaminergic neuronal aptitude against this devastating neurodegenerative disease.
Collapse
Affiliation(s)
- Merna Roshdy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Misr International University, Ahmed Orabi District, Cairo 44971, Egypt
| | - Doaa A Zaky
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo 11562, Egypt.
| | - Samah S Abbas
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Misr International University, Ahmed Orabi District, Cairo 44971, Egypt
| | - Dalaal M Abdallah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo 11562, Egypt
| |
Collapse
|
2
|
AICAR Ameliorates Non-Alcoholic Fatty Liver Disease via Modulation of the HGF/NF-κB/SNARK Signaling Pathway and Restores Mitochondrial and Endoplasmic Reticular Impairments in High-Fat Diet-Fed Rats. Int J Mol Sci 2023; 24:ijms24043367. [PMID: 36834782 PMCID: PMC9959470 DOI: 10.3390/ijms24043367] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/16/2023] [Accepted: 01/25/2023] [Indexed: 02/10/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a global health problem characterized by altered lipid and redox homeostasis, mitochondrial dysfunction, and endoplasmic reticulum (ER) stress. The AMP-dependent kinase (AMPK) agonist 5-aminoimidazole-4-carboxamide ribonucleoside (AICAR) has been shown to improve the outcome of NAFLD in the context of AMPK activation, yet the underlying molecular mechanism remains obscure. This study investigated the potential mechanism(s) of AICAR to attenuate NAFLD by exploring AICAR's effects on the HGF/NF-κB/SNARK axis and downstream effectors as well as mitochondrial and ER derangements. High-fat diet (HFD)-fed male Wistar rats were given intraperitoneal AICAR at 0.7 mg/g body weight or left untreated for 8 weeks. In vitro steatosis was also examined. ELISA, Western blotting, immunohistochemistry and RT-PCR were used to explore AICAR's effects. NAFLD was confirmed by steatosis score, dyslipidemia, altered glycemic, and redox status. HGF/NF-κB/SNARK was downregulated in HFD-fed rats receiving AICAR with improved hepatic steatosis and reduced inflammatory cytokines and oxidative stress. Aside from AMPK dominance, AICAR improved hepatic fatty acid oxidation and alleviated the ER stress response. In addition, it restored mitochondrial homeostasis by modulating Sirtuin 2 and mitochondrial quality gene expression. Our results provide a new mechanistic insight into the prophylactic role of AICAR in the prevention of NAFLD and its complications.
Collapse
|
3
|
Norouzi Z, Zarezadeh R, Mehdizadeh A, Niafar M, Germeyer A, Fayyazpour P, Fayezi S. Free Fatty Acids from Type 2 Diabetes Mellitus Serum Remodel Mesenchymal Stem Cell Lipids, Hindering Differentiation into Primordial Germ Cells. Appl Biochem Biotechnol 2022; 195:3011-3026. [PMID: 36495376 DOI: 10.1007/s12010-022-04204-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2022] [Indexed: 12/14/2022]
Abstract
Type 2 diabetes mellitus (T2DM) adversely affects the essential characteristics of adipose tissue-derived mesenchymal stem cells (AdMSCs). Given that T2DM is associated with an altered serum free fatty acid (FFA) profile, we examined whether diabetic serum FFAs influence the viability, differentiation, and fatty acid composition of the major lipid fractions of human AdMSCs in vitro. Serum FFAs were isolated from 7 diabetic and 10 healthy nondiabetic female individuals. AdMSCs were cultured and differentiated into primordial germ cell-like cells (PGCLCs) in the presence of either diabetic or nondiabetic FFAs. Cell viability was assessed using trypan blue staining. Cell differentiation was evaluated by measuring the PGCLC transcriptional markers Blimp1 and Stella. Lipid fractionation and fatty acid quantification were performed using thin-layer chromatography and gas-liquid chromatography, respectively. Both diabetic and nondiabetic FFAs significantly reduced the viability of PGCLCs. The gene expression of both differentiation markers was significantly lower in cells exposed to diabetic FFAs than in those treated with nondiabetic FFAs. Saturated fatty acids were significantly increased and linoleic acid was significantly decreased in the cellular phospholipid fraction after exposure to diabetic FFAs. In contrast, monounsaturated fatty acids were reduced and linoleic acid was elevated in the cellular triglyceride fraction in response to diabetic FFAs. Such an altered serum FFA profile in patients with T2DM reduces the proliferation and differentiation potential of AdMSCs, presumably due to the aberrant distribution of fatty acids into cell phospholipids and triglycerides.
Collapse
|
4
|
Ovarian cancer cell fate regulation by the dynamics between saturated and unsaturated fatty acids. Proc Natl Acad Sci U S A 2022; 119:e2203480119. [PMID: 36197994 PMCID: PMC9564215 DOI: 10.1073/pnas.2203480119] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Fatty acids are an important source of energy and a key component of phospholipids in membranes and organelles. Saturated fatty acids (SFAs) are converted into unsaturated fatty acids (UFAs) by stearoyl Co-A desaturase (SCD), an enzyme active in cancer. Here, we studied how the dynamics between SFAs and UFAs regulated by SCD impacts ovarian cancer cell survival and tumor progression. SCD depletion or inhibition caused lower levels of UFAs vs. SFAs and altered fatty acyl chain plasticity, as demonstrated by lipidomics and stimulated Raman scattering (SRS) microscopy. Further, increased levels of SFAs resulting from SCD knockdown triggered endoplasmic reticulum (ER) stress response with brisk activation of IRE1α/XBP1 and PERK/eIF2α/ATF4 axes. Disorganized ER membrane was visualized by electron microscopy and SRS imaging in ovarian cancer cells in which SCD was knocked down. The induction of long-term mild ER stress or short-time severe ER stress by the increased levels of SFAs and loss of UFAs led to cell death. However, ER stress and apoptosis could be readily rescued by supplementation with UFAs and reequilibration of SFA/UFA levels. The effects of SCD knockdown or inhibition observed in vitro translated into suppression of intraperitoneal tumor growth in ovarian cancer xenograft models. Furthermore, a combined intervention using an SCD inhibitor and an SFA-enriched diet initiated ER stress in tumors growing in vivo and potently blocked their dissemination. In all, our data support SCD as a key regulator of the cancer cell fate under metabolic stress and point to treatment strategies targeting the lipid balance.
Collapse
|
5
|
Resolvin D3 improves the impairment of insulin signaling in skeletal muscle and nonalcoholic fatty liver disease through AMPK/autophagy-associated attenuation of ER stress. Biochem Pharmacol 2022; 203:115203. [DOI: 10.1016/j.bcp.2022.115203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/30/2022] [Accepted: 08/02/2022] [Indexed: 11/17/2022]
|
6
|
Wang H, Yang L, Gao P, Deng P, Yue Y, Tian L, Xie J, Chen M, Luo Y, Liang Y, Qing W, Zhou Z, Pi H, Yu Z. Fluoride exposure induces lysosomal dysfunction unveiled by an integrated transcriptomic and metabolomic study in bone marrow mesenchymal stem cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 239:113672. [PMID: 35617906 DOI: 10.1016/j.ecoenv.2022.113672] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 05/13/2022] [Accepted: 05/18/2022] [Indexed: 06/15/2023]
Abstract
Fluoride has received much attention for its predominant bone toxicity in the human body. However, the toxic mechanism of bone injury caused by fluoride exposure remains largely unclear. Bone marrow mesenchymal stem cells (BMSCs) are widely used as model cells for evaluating bone toxicity after environmental toxicant exposure. In this study, BMSCs were exposed to fluoride at 1, 2, and 4 mM for 24 h, and fluoride significantly inhibited cell viability at 2 and 4 mM. A multiomics analysis combining transcriptomics with metabolomics was employed to detect alterations in genes and metabolites in BMSCs treated with 2 mM fluoride. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis of transcriptomics profiles identified "lysosomes" as the top enriched pathway, which was severely damaged by fluoride exposure. Lysosomal damage was indicated by decreases in the expression of lysosomal associated membrane protein 2 (LAMP 2) and cathepsin B (CTSB) as well as an increase in pH. Upregulation of the lysosome-related genes Atp6v0b and Gla was observed, which may be attributed to a compensatory lysosomal biogenesis transcriptional response. Interestingly, inhibition of glutathione metabolism was observed in fluoride-treated BMSCs at the metabolomic level. Moreover, an integrative analysis between altered genes, metabolites and lysosome signaling pathways was conducted. Palmitic acid, prostaglandin C2, and prostaglandin B2 metabolites were positively associated with Atp6v0b, a lysosome-related gene. Overall, our results provide novel insights into the mechanism responsible for fluoride-induced bone toxicity.
Collapse
Affiliation(s)
- Hui Wang
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Lu Yang
- Hunan Province Prevention and Treatment Hospital for Occupational Diseases, Hunan, China
| | - Peng Gao
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Ping Deng
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Yang Yue
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Li Tian
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Jia Xie
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Mengyan Chen
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Yan Luo
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Yidan Liang
- School of Medicine, Guangxi University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Weijia Qing
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China; The 63710th Military Hospital of PLA, Xinzhou, Shanxi, China
| | - Zhou Zhou
- Department of Environmental Medicine, School of Public Health, and Department of Emergency Medicine, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Huifeng Pi
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China.
| | - Zhengping Yu
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China.
| |
Collapse
|
7
|
Kim JH, Kim KM, Yang JH, Cho SS, Lee JH, Ki SH. Regulated in Development and DNA Damage Response 1 Protects Hepatocytes Against Palmitate-induced Lipotoxicity. BIOTECHNOL BIOPROC E 2022. [DOI: 10.1007/s12257-021-0140-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
8
|
Oh H, Cho W, Abd El-Aty AM, Jeong JH, Jung TW. Resolvin D3 Improves the Impairment of Insulin Signaling in Skeletal Muscle and Nonalcoholic Fatty Liver Disease Through AMPK/Autophagy-Associated Attenuation of ER Stress. SSRN ELECTRONIC JOURNAL 2022. [DOI: 10.2139/ssrn.4149178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
9
|
Guo D, Zhao M, Xu W, He H, Li B, Hou T. Dietary interventions for better management of osteoporosis: An overview. Crit Rev Food Sci Nutr 2021; 63:125-144. [PMID: 34251926 DOI: 10.1080/10408398.2021.1944975] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Osteoporosis is a public health concern and a cause of bone loss, increased risk of skeletal fracture, and a heavy economic burden. It is common in postmenopausal women and the elderly and is impacted by dietary factors, lifestyle and some secondary factors. Although many drugs are available for the treatment of osteoporosis, these therapies are accompanied by subsequent side effects. Hence, dietary interventions are highly important to prevent osteoporosis. This review was aimed to provide a comprehensive understanding of the roles of dietary nutrients derived from natural foods and of common dietary patterns in the regulation of osteoporosis. Nutrients from daily diets, such as unsaturated fatty acids, proteins, minerals, peptides, phytoestrogens, and prebiotics, can regulate bone metabolism and reverse bone loss. Meanwhile, these nutrients generally existed in food groups and certain dietary patterns also play critical roles in skeletal health. Appropriate dietary interventions (nutrients and dietary patterns) could be primary and effective strategies to prevent and treat osteoporosis across the lifespan for the consumers and food enterprises.
Collapse
Affiliation(s)
- Danjun Guo
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China.,College of Food Science & Engineering, Wuhan Polytechnic University, Wuhan, China
| | - Mengge Zhao
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Wei Xu
- College of Food Science & Engineering, Wuhan Polytechnic University, Wuhan, China
| | - Hui He
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Bin Li
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Tao Hou
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
10
|
Qiao J, Wu Y, Ren Y. The impact of a high fat diet on bones: potential mechanisms. Food Funct 2021; 12:963-975. [PMID: 33443523 DOI: 10.1039/d0fo02664f] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
High-fat diet led to bone loss via gut microbiota and fatty acid imbalances, immune disorder and adipose tissue accumulation inside and outside the bone marrow.
Collapse
Affiliation(s)
- Jie Qiao
- Department of Endocrinology and Metabolism
- the Second Affiliated Hospital of Zhejiang University School of Medicine
- Hangzhou
- 310009
- China
| | - Yiwen Wu
- Department of Neurosurgery
- Ningbo Hospital
- Zhejiang University School of Medicine
- Ningbo 315010
- China
| | - Yuezhong Ren
- Department of Endocrinology and Metabolism
- the Second Affiliated Hospital of Zhejiang University School of Medicine
- Hangzhou
- 310009
- China
| |
Collapse
|
11
|
Chen Y, Griffiths A, Wang J, Zhang T, Song Q, Song Z. Inositol-requiring enzyme 1α links palmitate-induced mTOR activation and lipotoxicity in hepatocytes. Am J Physiol Cell Physiol 2020; 319:C1130-C1140. [PMID: 33052067 PMCID: PMC7792676 DOI: 10.1152/ajpcell.00165.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 10/05/2020] [Accepted: 10/05/2020] [Indexed: 02/06/2023]
Abstract
Hepatic lipotoxicity, hepatocyte dysfunction/cell death induced by saturated fatty acids (SFA), plays a central role in the pathogenesis of nonalcoholic fatty liver disease (NAFLD); however, the underlying mechanisms remain unclear. Palmitate is the most abundant SFA in the circulation. In this study, via a small-scale screening of chemical inhibitors using AML12 hepatocytes, we identified mechanistic target of rapamycin (mTOR) complex 1 (mTORC1) to be a culprit in palmitate-induced cell death in hepatocytes in that mTOR inhibition is protective against palmitate-induced cell death. The protective effect of mTORC1 inhibition is independent of autophagy induction, as autophagy inhibition failed to ablate the mTORC1 inhibitor-conferred protection. We have previously reported that the endonuclease activity of inositol-requiring enzyme 1α (IRE1α), one of three canonical signaling pathways of endoplasmic reticulum (ER) stress, was implicated in palmitate-induced cell death in hepatocytes. The continuous mechanistic investigation in this study uncovered that IRE1α is a downstream target of mTORC1 activation upon palmitate exposure and the inhibition of either its endonuclease activity or kinase activity protects against the lipotoxic effect of palmitate. Our research further revealed that protein palmitoylation is potentially involved in palmitate-induced mTORC1 activation and lipotoxicity in hepatocytes. 2-Bromopalmitate, a protein palmitoylation inhibitor, ameliorated palmitate-triggered mTORC1 activation, concomitant with the protection of lipotoxicity in hepatocytes. Collectively, our data have identified that mTORC1 and ER stress are coordinately implicated in hepatocyte cell death in response to palmitate exposure and suggest that this pathway may potentially serve as a therapeutic target for the treatment of NAFLD as well as other metabolic disorders involving lipotoxicity.
Collapse
Affiliation(s)
- Yingli Chen
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois
- College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, Heilongjiang, People's Republic of China
| | - Alexandra Griffiths
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois
| | - Jun Wang
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois
- Department of Gastroenterology, Tongji Medical College and Central Hospital of Wuhan, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Tingting Zhang
- Department of Urology, Daqing Oilfield General Hospital, Daqing, Heilongjiang, People's Republic of China
| | - Qing Song
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois
| | - Zhenyuan Song
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
12
|
Fan C, Feng J, Tang C, Zhang Z, Feng Y, Duan W, Zhai M, Yan Z, Zhu L, Feng L, Zhu H, Luo E. Melatonin suppresses ER stress-dependent proapoptotic effects via AMPK in bone mesenchymal stem cells during mitochondrial oxidative damage. Stem Cell Res Ther 2020; 11:442. [PMID: 33059742 PMCID: PMC7560057 DOI: 10.1186/s13287-020-01948-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 09/23/2020] [Indexed: 12/12/2022] Open
Abstract
Background Bone marrow mesenchymal stem cells (BMSCs) have been used as important cell-based tools for clinical applications. Oxidative stress-induced apoptosis causes a low survival rate after transplantation, and the underlying mechanisms remain unknown. The endoplasmic reticulum (ER) and mitochondria are vital organelles regulated by adenosine monophosphate (AMP)-activated protein kinase (AMPK), especially during oxidative stress injury. Melatonin exerts an antioxidant effect by scavenging free radicals. Here, we aimed to explore whether cytoprotective melatonin relieves ER stress-mediated mitochondrial dysfunction through AMPK in BMSCs after oxidative stress injury. Methods Mouse BMSCs were isolated and exposed to H2O2 in the absence or presence of melatonin. Thereafter, cell damage, oxidative stress levels, mitochondrial function, AMPK activity, ER stress-related proteins, and apoptotic markers were measured. Additionally, the involvement of AMPK and ER stress in the melatonin-mediated protection of BMSCs against H2O2-induced injury was investigated using pharmacologic agonists and inhibitors. Results Melatonin improved cell survival and restored mitochondrial function. Moreover, melatonin intimately regulated the phosphorylation of AMPK and molecules associated with ER stress pathways. AMPK activation and ER stress inhibition following melatonin administration improved the mitochondrial membrane potential (MMP), reduced mitochondria-initiated oxidative damage, and ultimately suppressed apoptotic signaling pathways in BMSCs. Cotreatment with N-acetyl-l-cysteine (NAC) significantly enhanced the antioxidant effect of melatonin. Importantly, pharmacological AMPK activation/ER stress inhibition promoted melatonin-induced cytoprotection, while pharmacological AMPK inactivation/ER stress induction conferred resistance to the effect of melatonin against H2O2 insult. Conclusions Our data also reveal a new, potentially therapeutic mechanism by which melatonin protects BMSCs from oxidative stress-mediated mitochondrial apoptosis, possibly by regulating the AMPK-ER stress pathway.
Collapse
Affiliation(s)
- Chongxi Fan
- Department of Military Biomedical Engineering, Air Force Medical University, 169 Changle West Road, Xi'an, 710032, China. .,Department of Oncology, Air Force Medical Center of PLA, 30 Fucheng Road, Beijing, 100142, China.
| | - Jianyu Feng
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Medical University, 127 Changle West Road, Xi'an, 710032, China
| | - Chi Tang
- Department of Military Biomedical Engineering, Air Force Medical University, 169 Changle West Road, Xi'an, 710032, China
| | - Zhengbin Zhang
- Department of Geriatrics, The 8th Medical Center of Chinese PLA General Hospital, 17 Heishanhu Street, Beijing, 100091, China
| | - Yingtong Feng
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, 1 Xinsi Road, Xi'an, 710038, China
| | - Weixun Duan
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Medical University, 127 Changle West Road, Xi'an, 710032, China
| | - Mingming Zhai
- Department of Military Biomedical Engineering, Air Force Medical University, 169 Changle West Road, Xi'an, 710032, China
| | - Zedong Yan
- Department of Military Biomedical Engineering, Air Force Medical University, 169 Changle West Road, Xi'an, 710032, China
| | - Liwen Zhu
- Department of Cardiology, The First Affiliated Hospital of Xi'an Medical University, 277 Yanta West Road, Xi'an, 710077, China
| | - Lele Feng
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Medical University, 127 Changle West Road, Xi'an, 710032, China
| | - Hanzhao Zhu
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Medical University, 127 Changle West Road, Xi'an, 710032, China
| | - Erping Luo
- Department of Military Biomedical Engineering, Air Force Medical University, 169 Changle West Road, Xi'an, 710032, China.
| |
Collapse
|
13
|
Fan C, Feng J, Tang C, Zhang Z, Feng Y, Duan W, Zhai M, Yan Z, Zhu L, Feng L, Zhu H, Luo E. Melatonin suppresses ER stress-dependent proapoptotic effects via AMPK in bone mesenchymal stem cells during mitochondrial oxidative damage. Stem Cell Res Ther 2020. [PMID: 33059742 DOI: 10.1186/s13287-020-01948-5.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Bone marrow mesenchymal stem cells (BMSCs) have been used as important cell-based tools for clinical applications. Oxidative stress-induced apoptosis causes a low survival rate after transplantation, and the underlying mechanisms remain unknown. The endoplasmic reticulum (ER) and mitochondria are vital organelles regulated by adenosine monophosphate (AMP)-activated protein kinase (AMPK), especially during oxidative stress injury. Melatonin exerts an antioxidant effect by scavenging free radicals. Here, we aimed to explore whether cytoprotective melatonin relieves ER stress-mediated mitochondrial dysfunction through AMPK in BMSCs after oxidative stress injury. METHODS Mouse BMSCs were isolated and exposed to H2O2 in the absence or presence of melatonin. Thereafter, cell damage, oxidative stress levels, mitochondrial function, AMPK activity, ER stress-related proteins, and apoptotic markers were measured. Additionally, the involvement of AMPK and ER stress in the melatonin-mediated protection of BMSCs against H2O2-induced injury was investigated using pharmacologic agonists and inhibitors. RESULTS Melatonin improved cell survival and restored mitochondrial function. Moreover, melatonin intimately regulated the phosphorylation of AMPK and molecules associated with ER stress pathways. AMPK activation and ER stress inhibition following melatonin administration improved the mitochondrial membrane potential (MMP), reduced mitochondria-initiated oxidative damage, and ultimately suppressed apoptotic signaling pathways in BMSCs. Cotreatment with N-acetyl-L-cysteine (NAC) significantly enhanced the antioxidant effect of melatonin. Importantly, pharmacological AMPK activation/ER stress inhibition promoted melatonin-induced cytoprotection, while pharmacological AMPK inactivation/ER stress induction conferred resistance to the effect of melatonin against H2O2 insult. CONCLUSIONS Our data also reveal a new, potentially therapeutic mechanism by which melatonin protects BMSCs from oxidative stress-mediated mitochondrial apoptosis, possibly by regulating the AMPK-ER stress pathway.
Collapse
Affiliation(s)
- Chongxi Fan
- Department of Military Biomedical Engineering, Air Force Medical University, 169 Changle West Road, Xi'an, 710032, China. .,Department of Oncology, Air Force Medical Center of PLA, 30 Fucheng Road, Beijing, 100142, China.
| | - Jianyu Feng
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Medical University, 127 Changle West Road, Xi'an, 710032, China
| | - Chi Tang
- Department of Military Biomedical Engineering, Air Force Medical University, 169 Changle West Road, Xi'an, 710032, China
| | - Zhengbin Zhang
- Department of Geriatrics, The 8th Medical Center of Chinese PLA General Hospital, 17 Heishanhu Street, Beijing, 100091, China
| | - Yingtong Feng
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, 1 Xinsi Road, Xi'an, 710038, China
| | - Weixun Duan
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Medical University, 127 Changle West Road, Xi'an, 710032, China
| | - Mingming Zhai
- Department of Military Biomedical Engineering, Air Force Medical University, 169 Changle West Road, Xi'an, 710032, China
| | - Zedong Yan
- Department of Military Biomedical Engineering, Air Force Medical University, 169 Changle West Road, Xi'an, 710032, China
| | - Liwen Zhu
- Department of Cardiology, The First Affiliated Hospital of Xi'an Medical University, 277 Yanta West Road, Xi'an, 710077, China
| | - Lele Feng
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Medical University, 127 Changle West Road, Xi'an, 710032, China
| | - Hanzhao Zhu
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Medical University, 127 Changle West Road, Xi'an, 710032, China
| | - Erping Luo
- Department of Military Biomedical Engineering, Air Force Medical University, 169 Changle West Road, Xi'an, 710032, China.
| |
Collapse
|
14
|
Lei L, Bai G, Wang X, Liu S, Xia J, Wu S, Huan Y, Shen Z. Histone deacetylase 3-selective inhibitor RGFP966 ameliorates impaired glucose tolerance through β-cell protection. Toxicol Appl Pharmacol 2020; 406:115189. [PMID: 32800772 DOI: 10.1016/j.taap.2020.115189] [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: 04/23/2020] [Revised: 08/04/2020] [Accepted: 08/11/2020] [Indexed: 01/18/2023]
Abstract
The potential therapeutic effect of histone deacetylase 3 (HDAC3) pharmacologic inhibition on diabetes has been focused recently. RGFP966, as a highly-selective HDAC3 inhibitor, its possible roles and underlying mechanism in the treatment of diabetes needs to be clarified. In this study, low-dose streptozotocin (STZ)-induced pre-diabetic mice were used to test the regulatory ability of RGFP966 in blood glucose and insulin. We isolated the islets both from normal C57BL/6 J mice and KKAy mice with spontaneous type 2 diabetes to determine the potency of RGFP966 on glucose-stimulated insulin secretion. NIT-1 pancreatic β-cells induced by sodium palmitate (PA) were applied to identify the protective effects of RGFP966 against β-cell apoptosis. The results showed that administration of RGFP966 in the pre-diabetic mice not only significantly reduced hyperglycemia, promoted phase I insulin secretion, improved morphology of islets, but also increased glucose infusion rate (GIR) during hyperglycemic clamp test. When treated in vitro, RGFP966 enhanced insulin secretion and synthesis in islets of normal C57BL/6J mice and diabetic KKAy mice. In addition, it partially attenuated PA-induced apoptosis in NIT-1 cells. Therefore, our research suggests that RGFP966, probably through selective inhibition of HDAC3, might serve as a novel potential preventive and therapeutic candidate for diabetes.
Collapse
Affiliation(s)
- Lei Lei
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Guoliang Bai
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xing Wang
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Shuainan Liu
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jie Xia
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Song Wu
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yi Huan
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Zhufang Shen
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
15
|
Tripathi AK, Rai D, Kothari P, Kushwaha P, Sinha S, Sardar A, Sashidhara KV, Trivedi R. Benzofuran pyran compound rescues rat and human osteoblast from lipotoxic effect of palmitate by inhibiting lipid biosynthesis and promoting stabilization of RUNX2. Toxicol In Vitro 2020; 66:104872. [DOI: 10.1016/j.tiv.2020.104872] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 04/06/2020] [Accepted: 04/20/2020] [Indexed: 02/06/2023]
|
16
|
Wang J, Chen Y, Song Q, Griffiths A, Song Z. mTORC1-IRE1α pathway activation contributes to palmitate-elicited triglyceride secretion and cell death in hepatocytes. Exp Biol Med (Maywood) 2020; 245:1268-1279. [PMID: 32436749 DOI: 10.1177/1535370220928276] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
IMPACT STATEMENT Lipotoxicity induced by saturated fatty acids (SFA) plays a pivotal role in the pathogenesis of a variety of obesity-related metabolic disorders; however, the exact mechanism(s) underlying lipotoxicity development remains elusive. The liver plays a central role in regulating intrahepatic and circulatory lipid homeostasis. In the current study, we identified that mammalian target of rapamycin complex 1 (mTORC1) activation plays an important role in regulating the detrimental effects of SFA palmitate in hepatocytes, in specific cell death, and TG overproduction. Furthermore, our data confirmed that palmitate-induced mTORC1 activation is attributable to its stimulatory effect on IRE1α, one of three canonical pathways activated during ER stress. Importantly, IRE1α inhibition prevented palmitate-triggered cell death and TG overproduction, suggesting mTORC1-IRE1α pathway is mechanistically implicated in palmitate lipotoxicity. The data obtained in the current investigation support future study to explore the therapeutic potential of targeting the mTORC1-IRE1α pathway as a novel clinical strategy for the treatment of metabolic disorders involving lipotoxicity.
Collapse
Affiliation(s)
- Jun Wang
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612, USA.,Department of Gastroenterology, Tongji Medical College and The Central Hospital of Wuhan, Huazhong University of Science and Technology, Wuhan 430000, PR. China
| | - Yingli Chen
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612, USA.,College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, Heilongjiang 163319, PR. China
| | - Qing Song
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Alexandra Griffiths
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Zhenyuan Song
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
17
|
Metabolomic Analysis Reveals That the Mechanism of Astaxanthin Improves the Osteogenic Differentiation Potential in Bone Marrow Mesenchymal Stem Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3427430. [PMID: 32308800 PMCID: PMC7132583 DOI: 10.1155/2020/3427430] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 01/09/2020] [Accepted: 02/25/2020] [Indexed: 01/06/2023]
Abstract
At present, little research has been done on the metabolic phenotype of the differentiation of mesenchymal stem cells (MSCs) into osteoblasts. In this study, the effect of astaxanthin on improving osteogenic differentiation potential of mesenchymal stem cells was studied by metabolomics. Results showed that L-methionine, L-tyrosine, and 2-hydroxycinnamic acid were upregulated in MSCs treated with astaxanthin, while L-lysine, L-pipecolic acid, L-histidine, L-arginine, D-fructose, and L-aspartic acid were downregulated in samples treated with astaxanthin. In addition, astaxanthin exhibited a significant dose-dependent relationship with these markers. Metabolic pathway enrichment analysis revealed that AST mainly regulated phenylalanine metabolism; phenylalanine, tyrosine, and tryptophan biosynthesis; and pantothenate and CoA biosynthesis during the process of osteogenic differentiation of MSCs. Furthermore, the staining results showed that astaxanthin could actively promote the osteogenic differentiation of mesenchymal stem cells. These findings clearly indicate that astaxanthin plays an important role in inducing osteogenic differentiation of mesenchymal stem cells. In addition, the changed metabolites can be used to monitor the differentiation process.
Collapse
|
18
|
Takeuchi T, Masuno K, Umeda M, Tanaka A, Tominaga K. Palmitate induces apoptosis and inhibits osteogenic differentiation of human periodontal ligament stem cells. Arch Oral Biol 2020; 112:104681. [PMID: 32070866 DOI: 10.1016/j.archoralbio.2020.104681] [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: 07/31/2019] [Revised: 01/22/2020] [Accepted: 02/10/2020] [Indexed: 10/25/2022]
Abstract
OBJECTIVE The aim of the present study was to investigate the effect of palmitate on human periodontal ligament stem cells (PDLSCs). DESIGN PDLSCs were isolated from the third molars of healthy adult donors, and cultured in normal or osteogenic medium supplemented with palmitate (0, 100, or 250 μM) for 21 days. Cell proliferation was evaluated by measuring the amount of formazan at 6, 24, 48, and 72 h. Apoptosis was detected by ELISA and terminal deoxynucleotidyl transferase dUTP nick end labeling assay at days 3 and 7. Osteogenic differentiation was evaluated by measuring the alkaline phosphatase (ALP) activity, production of procollagen type I C-peptide and osteocalcin, mineralization, and mRNA expression of Runx2 at days 3, 7, 14, and 21. In addition, mRNA expression of IL-6 and IL-8 was measured at day 3. RESULTS Palmitate inhibited the proliferation, ALP activity, production of procollagen type I C-peptide and osteocalcin, mineralization, and mRNA expression of Runx2 in the cultured PDLSCs. Palmitate also induced apoptosis and mRNA expression of IL-6 and IL-8 in the PDLSCs. CONCLUSIONS The results of the present study demonstrate that palmitate induces apoptosis and inhibits osteogenic differentiation of PDLSCs. These findings may help clarify the relationship between palmitate and periodontal tissue regeneration.
Collapse
Affiliation(s)
- Tomoki Takeuchi
- Department of Oral Pathology, Osaka Dental University, Osaka, Japan.
| | - Kazuya Masuno
- Department of Innovations in Dental Education, Osaka Dental University, Osaka, Japan
| | - Makoto Umeda
- Department of Periodontology, Osaka Dental University, Osaka, Japan
| | - Akio Tanaka
- Department of Pathology, Osaka Dental University, Osaka, Japan
| | - Kazuya Tominaga
- Department of Oral Pathology, Osaka Dental University, Osaka, Japan
| |
Collapse
|
19
|
Bao M, Zhang K, Wei Y, Hua W, Gao Y, Li X, Ye L. Therapeutic potentials and modulatory mechanisms of fatty acids in bone. Cell Prolif 2020; 53:e12735. [PMID: 31797479 PMCID: PMC7046483 DOI: 10.1111/cpr.12735] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 11/02/2019] [Accepted: 11/05/2019] [Indexed: 02/05/2023] Open
Abstract
Bone metabolism is a lifelong process that includes bone formation and resorption. Osteoblasts and osteoclasts are the predominant cell types associated with bone metabolism, which is facilitated by other cells such as bone marrow mesenchymal stem cells (BMMSCs), osteocytes and chondrocytes. As an important component in our daily diet, fatty acids are mainly categorized as long-chain fatty acids including polyunsaturated fatty acids (LCPUFAs), monounsaturated fatty acids (LCMUFAs), saturated fatty acids (LCSFAs), medium-/short-chain fatty acids (MCFAs/SCFAs) as well as their metabolites. Fatty acids are closely associated with bone metabolism and associated bone disorders. In this review, we summarized the important roles and potential therapeutic implications of fatty acids in multiple bone disorders, reviewed the diverse range of critical effects displayed by fatty acids on bone metabolism, and elucidated their modulatory roles and mechanisms on specific bone cell types. The evidence supporting close implications of fatty acids in bone metabolism and disorders suggests fatty acids as potential therapeutic and nutritional agents for the treatment and prevention of metabolic bone diseases.
Collapse
Affiliation(s)
- Minyue Bao
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Kaiwen Zhang
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Yangyini Wei
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Weihan Hua
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Yanzi Gao
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Xin Li
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Ling Ye
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesDepartment of Cariology and EndodonticsWest China Hospital of StomatologySichuan UniversityChengduChina
| |
Collapse
|
20
|
Casado-Díaz A, Dorado G, Quesada-Gómez JM. Influence of olive oil and its components on mesenchymal stem cell biology. World J Stem Cells 2019; 11:1045-1064. [PMID: 31875868 PMCID: PMC6904865 DOI: 10.4252/wjsc.v11.i12.1045] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 08/29/2019] [Accepted: 11/04/2019] [Indexed: 02/06/2023] Open
Abstract
Extra virgin olive oil is characterized by its high content of unsaturated fatty acid residues in triglycerides, mainly oleic acid, and the presence of bioactive and antioxidant compounds. Its consumption is associated with lower risk of suffering chronic diseases and unwanted processes linked to aging, due to the antioxidant capacity and capability of its components to modulate cellular signaling pathways. Consumption of olive oil can alter the physiology of mesenchymal stem cells (MSCs). This may explain part of the healthy effects of olive oil consumption, such as prevention of unwanted aging processes. To date, there are no specific studies on the action of olive oil on MSCs, but effects of many components of such food on cell viability and differentiation have been evaluated. The objective of this article is to review existing literature on how different compounds of extra virgin olive oil, including residues of fatty acids, vitamins, squalene, triterpenes, pigments and phenols, affect MSC maintenance and differentiation, in order to provide a better understanding of the healthy effects of this food. Interestingly, most studies have shown a positive effect of these compounds on MSCs. The collective findings support the hypothesis that at least part of the beneficial effects of extra virgin olive oil consumption on health may be mediated by its effects on MSCs.
Collapse
Affiliation(s)
- Antonio Casado-Díaz
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, Córdoba 14004, Spain
| | - Gabriel Dorado
- Departement Bioquímica y Biología Molecular, Campus Rabanales C6-1-E17, Campus de Excelencia Internacional Agroalimentario (ceiA3), Universidad de Córdoba, CIBERFES, Córdoba 14071, Spain
| | - José Manuel Quesada-Gómez
- Unidad de Gestión Clínica de Endocrinología y Nutrición, CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, Córdoba 14004, Spain.
| |
Collapse
|
21
|
Effects of palmitate and astaxanthin on cell viability and proinflammatory characteristics of mesenchymal stem cells. Int Immunopharmacol 2019; 68:164-170. [PMID: 30639962 DOI: 10.1016/j.intimp.2018.12.063] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 12/16/2018] [Accepted: 12/28/2018] [Indexed: 12/18/2022]
Abstract
Mesenchymal stem cells (MSCs) have broad immunomodulatory activities. These cells are a stable source of cytokine production such as interleukin-6 (IL6), monocyte chemoattractant protein-1 (MCP-1/CCL2) and vascular endothelial growth factor (VEGF). Fatty acid elevation in chronic metabolic diseases alters the microenvironment of MSCs and thereby, might affect their survival and cytokine production. In the present study, we investigated the effects of palmitate, the most abundant saturated free fatty acid (FFA) in plasma, and astaxanthin, a potent antioxidant, on cell viability and apoptosis in human bone marrow-driven mesenchymal stem cells. We also elucidated how palmitate and astaxanthin influence the inflammation in MSCs. Human mesenchymal stem cells were collected from an aspirate of the femurs and tibias marrow compartment. The effect of palmitate on cell viability, caspase activity and pro-inflammatory cytokines expression and secretion were evaluated. In addition, activation of the MAP kinases and NF-kB signaling pathways were investigated. The results showed that astaxanthin protected MSCs from palmitate-induced cell death. We found that palmitate significantly enhanced IL-6, VEGF and MCP-1 expression, and secretion in MSC cells. Increased cytokine expression was parallel to the enhanced phosphorylation of P38, ERK and IKKα-IKKβ. In addition, pretreatment with JNK, ERK, P38, and NF-kB inhibitors could correspondingly attenuate palmitate-induced expression of VEGF, IL-6, and MCP-1. Our results demonstrated that fatty acid exposure causes inflammatory responses in MSCs that can be alleviated favorably by astaxanthin treatment.
Collapse
|
22
|
Pino AM, Rodríguez JP. Is fatty acid composition of human bone marrow significant to bone health? Bone 2019; 118:53-61. [PMID: 29258874 DOI: 10.1016/j.bone.2017.12.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 12/15/2017] [Indexed: 12/31/2022]
Abstract
The bone marrow adipose tissue (BMAT) is a conserved component of the marrow microenvironment, providing storage and release of energy and stabilizing the marrow extent. Also, it is recognized both the amount and quality of BMAT are relevant to preserve the functional relationships between BMAT, bone, and blood cell production. In this article we ponder the information supporting the tenet that the quality of BMAT is relevant to bone health. In the human adult the distribution of BMAT is heterogeneous over the entire skeleton, and both BMAT accumulation and bone loss come about with aging in healthy populations. But some pathological conditions which increase BMAT formation lead to bone impairment and fragility. Analysis in vivo of the relative content of saturated and unsaturated fatty acids (FA) in BMAT indicates site-related bone marrow fat composition and an association between increased unsaturation index (UI) and bone health. With aging some impairment ensues in the regulation of bone marrow cells and systemic signals leading to local chronic inflammation. Most of the bone loss diseases which evolve altered BMAT composition have as common factors aging and/or chronic inflammation. Both saturated and unsaturated FAs originate lipid species which are active mediators in the inflammation process. Increased free saturated FAs may lead to lipotoxicity of bone marrow cells. The pro-inflammatory, anti-inflammatory or resolving actions of compounds derived from long chain poly unsaturated FAs (PUFA) on bone cells is varied, and depending on the metabolism of the parent n:3 or n:6 PUFAs series. Taking together the evidence substantiate that marrow adipocyte function is fundamental for an efficient link between systemic and marrow fatty acids to accomplish specific energy or regulatory needs of skeletal and marrow cells. Further, they reveal marrow requirements of PUFAs.
Collapse
Affiliation(s)
- Ana María Pino
- Laboratorio de Biología Celular, INTA, Universidad de Chile, Chile
| | | |
Collapse
|
23
|
Michalak M, Agellon LB. Stress Coping Strategies in the Heart: An Integrated View. Front Cardiovasc Med 2018; 5:168. [PMID: 30519562 PMCID: PMC6258784 DOI: 10.3389/fcvm.2018.00168] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 11/02/2018] [Indexed: 12/15/2022] Open
Abstract
The heart is made up of an ordered amalgam of cardiac cell types that work together to coordinate four major processes, namely energy production, electrical conductance, mechanical work, and tissue remodeling. Over the last decade, a large body of information has been amassed regarding how different cardiac cell types respond to cellular stress that affect the functionality of their elaborate intracellular membrane networks, the cellular reticular network. In the context of the heart, the manifestations of stress coping strategies likely differ depending on the coping strategy outcomes of the different cardiac cell types, and thus may underlie the development of distinct cardiac disorders. It is not clear whether all cardiac cell types have similar sensitivity to cellular stress, how specific coping response strategies modify their unique roles, and how their metabolic status is communicated to other cells within the heart. Here we discuss our understanding of the roles of specialized cardiac cells that together make the heart function as an organ with the ability to pump blood continuously and follow a regular rhythm.
Collapse
Affiliation(s)
- Marek Michalak
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Luis B Agellon
- School of Human Nutrition, McGill University, Ste. Anne de Bellevue, QC, Canada
| |
Collapse
|
24
|
Lu J, Zhu LF, Cai YM, Dong HY, Zhu L, Tan JM. Isolation and multipotential differentiation of mesenchymal stromal cell‑like progenitor cells from human bladder. Mol Med Rep 2018; 19:187-194. [PMID: 30431114 PMCID: PMC6297775 DOI: 10.3892/mmr.2018.9646] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 10/19/2018] [Indexed: 02/07/2023] Open
Abstract
Various types of mesenchymal stromal cells (MSCs) have been used in urological tissue engineering but to date the existence of MSCs has not been reported in the human bladder. The present study provided evidence that a small number of MSC‑like cells exist in the human bladder and designated this class of cells 'human bladder‑derived MSC‑like cells' (hBSCs). It was demonstrated that hBSCs can be cultured to yield a large population. These hBSCs expressed the surface markers of MSCs and exhibited the capacity for osteogenic, adipogenic and chondrogenic differentiation. On induction with appropriate media in vitro, hBSCs could differentiate into bladder‑associated cell types, including urothelial, endothelial and smooth muscle cell‑like lineages. In addition, the average telomerase activity of adult hBSCs was higher compared with adult human bone marrow‑derived MSCs, but lower than that of human umbilical cord Wharton's jelly‑derived MSCs. These findings may inspire future studies on the role of hBSCs in urological tissue engineering applications and in other fields.
Collapse
Affiliation(s)
- Jun Lu
- Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou General Hospital/ or Dongfang Hospital, Xiamen University, Fuzhou, Fujian 350025, P.R. China
| | - Ling-Feng Zhu
- Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou General Hospital/ or Dongfang Hospital, Xiamen University, Fuzhou, Fujian 350025, P.R. China
| | - Yuan-Ming Cai
- College of Basic Medical, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830001, P.R. China
| | - Hui-Yue Dong
- Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou General Hospital/ or Dongfang Hospital, Xiamen University, Fuzhou, Fujian 350025, P.R. China
| | - Ling Zhu
- Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou General Hospital/ or Dongfang Hospital, Xiamen University, Fuzhou, Fujian 350025, P.R. China
| | - Jian-Ming Tan
- Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou General Hospital/ or Dongfang Hospital, Xiamen University, Fuzhou, Fujian 350025, P.R. China
| |
Collapse
|
25
|
Yang L, Guan G, Lei L, Lv Q, Liu S, Zhan X, Jiang Z, Gu X. Palmitic acid induces human osteoblast-like Saos-2 cell apoptosis via endoplasmic reticulum stress and autophagy. Cell Stress Chaperones 2018; 23:1283-1294. [PMID: 30194633 PMCID: PMC6237680 DOI: 10.1007/s12192-018-0936-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 08/27/2018] [Accepted: 09/01/2018] [Indexed: 12/30/2022] Open
Abstract
Palmitic acid (PA) is the most common saturated long-chain fatty acid in food that causes cell apoptosis. However, little is known about the molecular mechanisms of PA toxicity. In this study, we explore the effects of PA on proliferation and apoptosis in human osteoblast-like Saos-2 cells and uncover the signaling pathways involved in the process. Our study showed that endoplasmic reticulum (ER) stress and autophagy are involved in PA-induced Saos-2 cell apoptosis. We found that PA inhibited the viability of Saos-2 cells in a dose- and time-dependent manner. At the same time, PA induced the expression of ER stress marker genes (glucose-regulated protein 78 (GRP78) and CCAAT/enhancer binding protein homologous protein (CHOP)), altered autophagy-related gene expression (microtubule-associated protein 1 light chain 3 (LC3), ATG5, p62, and Beclin), promoted apoptosis-related gene expression (Caspase 3 and BAX), and affected autophagic flux. Inhibiting ER stress with 4-PBA diminished the PA-induced cell apoptosis, activated autophagy, and increased the expression of Caspase 3 and BAX. Inhibiting autophagy with 3-MA attenuated the PA and ER stress-induced cell apoptosis and the apoptosis-related gene expression (Caspase 3 and BAX), but seemed to have no obvious effects on ER stress, although the CHOP expression was downregulated. Taken together, our results suggest that PA-induced Saos-2 cell apoptosis is activated via ER stress and autophagy, and the activation of autophagy depends on the ER stress during this process.
Collapse
Affiliation(s)
- Lei Yang
- College of Basic Medical, Jiujiang University, Jiujiang, 332000, Jiangxi, China.
- Key Laboratory of System Bio-medicine of Jiangxi Province, Jiujiang University, Jiujiang, 332000, Jiangxi, China.
| | - Gaopeng Guan
- Key Laboratory of System Bio-medicine of Jiangxi Province, Jiujiang University, Jiujiang, 332000, Jiangxi, China
- Affiliated Hospital of Jiujiang University, Jiujiang University, Jiujiang, 332000, Jiangxi, China
- Medicine Graduate School, Nanchang University, Nanchang, 330006, China
| | - Lanjie Lei
- Affiliated Hospital of Jiujiang University, Jiujiang University, Jiujiang, 332000, Jiangxi, China
| | - Qizhuang Lv
- College of Biology & Pharmacy, Yulin Normal University, Yulin, 537000, Guangxi, China
| | - Shengyuan Liu
- Key Laboratory of System Bio-medicine of Jiangxi Province, Jiujiang University, Jiujiang, 332000, Jiangxi, China
- Affiliated Hospital of Jiujiang University, Jiujiang University, Jiujiang, 332000, Jiangxi, China
- Medicine Graduate School, Nanchang University, Nanchang, 330006, China
| | - Xiuwen Zhan
- Key Laboratory of System Bio-medicine of Jiangxi Province, Jiujiang University, Jiujiang, 332000, Jiangxi, China
- Affiliated Hospital of Jiujiang University, Jiujiang University, Jiujiang, 332000, Jiangxi, China
| | - Zhenzhen Jiang
- Medicine Graduate School, Nanchang University, Nanchang, 330006, China
| | - Xiang Gu
- Key Laboratory of System Bio-medicine of Jiangxi Province, Jiujiang University, Jiujiang, 332000, Jiangxi, China.
- Affiliated Hospital of Jiujiang University, Jiujiang University, Jiujiang, 332000, Jiangxi, China.
| |
Collapse
|
26
|
Ni XQ, Lu WW, Zhang JS, Zhu Q, Ren JL, Yu YR, Liu XY, Wang XJ, Han M, Jing Q, Du J, Tang CS, Qi YF. Inhibition of endoplasmic reticulum stress by intermedin1-53 attenuates angiotensin II-induced abdominal aortic aneurysm in ApoE KO Mice. Endocrine 2018; 62:90-106. [PMID: 29943223 DOI: 10.1007/s12020-018-1657-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 06/15/2018] [Indexed: 12/23/2022]
Abstract
Endoplasmic reticulum stress (ERS) is involved in the development of abdominal aortic aneurysm (AAA). Since bioactive peptide intermedin (IMD)1-53 protects against AAA formation, here we investigated whether IMD1-53 attenuates AAA by inhibiting ERS. AAA model was induced by angiotensin II (AngII) in ApoE KO mouse background. AngII-treated mouse aortas showed increased ERS gene transcription of caspase12, eukaryotic translation initiation factor 2a (eIf2a) and activating transcription factor 4(ATF4).The protein level of ERS marker glucose regulated protein 94(GRP94), ATF4 and C/EBP homologous protein 10(CHOP) was also up-regulated by AngII. Increased ERS levels were accompanied by severe VSMC apoptosis in human AAA aorta. In vivo administration of IMD1-53 greatly reduced AngII-induced AAA and abrogated the activation of ERS. To determine whether IMD inhibited AAA by ameliorating ERS, we used 2 non-selective ERS inhibitors phenyl butyrate (4-PBA) and taurine (TAU). Similar to IMD, PBA, and TAU significantly reduced the incidence of AAA and AAA-related pathological disorders. In vitro, AngII infusion up-regulated CHOP, caspase12 expression and led to VSMC apoptosis. IMD siRNA aggravated the CHOP, caspase12-mediated VSMC apoptosis, which was abolished by ATF4 silencing. IMD infusion promoted the phosphorylation of adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) in aortas in ApoE KO mice, and the AMPK inhibitor compound C abolished the protective effect of IMD on VSMC ERS and apoptosis induced by AngII. In conclusion, IMD may protect against AAA formation by inhibiting ERS via activating AMPK phosphorylation.
Collapse
MESH Headings
- Adenylate Kinase/metabolism
- Angiotensin II
- Animals
- Aortic Aneurysm, Abdominal/chemically induced
- Aortic Aneurysm, Abdominal/drug therapy
- Aortic Aneurysm, Abdominal/metabolism
- Apolipoproteins E/genetics
- Apolipoproteins E/metabolism
- Endoplasmic Reticulum Stress/drug effects
- Mice
- Mice, Knockout
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Peptide Hormones/pharmacology
- Peptide Hormones/therapeutic use
- Phosphorylation/drug effects
Collapse
Affiliation(s)
- Xian-Qiang Ni
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, 100083, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, 100083, Beijing, China
- Department of Microbiology and Parasitology, School of Basic Medical Science, Peking University, 100083, Beijing, China
| | - Wei-Wei Lu
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, 100083, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, 100083, Beijing, China
- Department of Microbiology and Parasitology, School of Basic Medical Science, Peking University, 100083, Beijing, China
| | - Jin-Sheng Zhang
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, 100083, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, 100083, Beijing, China
- Department of Microbiology and Parasitology, School of Basic Medical Science, Peking University, 100083, Beijing, China
| | - Qing Zhu
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, 100083, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, 100083, Beijing, China
- Department of Microbiology and Parasitology, School of Basic Medical Science, Peking University, 100083, Beijing, China
| | - Jin-Ling Ren
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, 100083, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, 100083, Beijing, China
- Department of Microbiology and Parasitology, School of Basic Medical Science, Peking University, 100083, Beijing, China
| | - Yan-Rong Yu
- Department of Microbiology and Parasitology, School of Basic Medical Science, Peking University, 100083, Beijing, China
| | - Xiu-Ying Liu
- Key Laboratory of Genetic Network Biology, Collaborative Innovation Center of Genetics and Development, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Xiu-Jie Wang
- Key Laboratory of Genetic Network Biology, Collaborative Innovation Center of Genetics and Development, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Mei Han
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Hebei Medical University, 050017, Shijiazhuang, China
| | - Qing Jing
- Key Laboratory of Stem Cell Biology, Shanghai Institutes for Biological Science, Chinese Academy of Science, Shanghai, China
| | - Jie Du
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing An Zhen Hospital, Ministry of Education, Capital Medical University, 100029, Beijing, China
| | - Chao-Shu Tang
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, 100083, Beijing, China
| | - Yong-Fen Qi
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, 100083, Beijing, China.
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, 100083, Beijing, China.
| |
Collapse
|
27
|
Liu Y, Wang N, Zhang S, Liang Q. Autophagy protects bone marrow mesenchymal stem cells from palmitate‑induced apoptosis through the ROS‑JNK/p38 MAPK signaling pathways. Mol Med Rep 2018; 18:1485-1494. [PMID: 29901107 PMCID: PMC6072221 DOI: 10.3892/mmr.2018.9100] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 05/18/2018] [Indexed: 01/12/2023] Open
Abstract
In recent years, the association between saturated fatty acids (FA) and bone cells has received a high level of attention. Previous studies have shown that palmitate (PA), a common saturated FA, can cause apoptosis in bone marrow mesenchymal stem cells (BMSCs). However, whether PA can induce autophagy, an important intracellular protection mechanism that is closely associated with apoptosis, in BMSCs is still unknown; the association between autophagy and apoptosis is also unclear. The aim of the present study was to determine whether PA can induce autophagy in BMSCs. When BMSCs were treated with PA for >18 h, p62 began to accumulate, indicating that autophagic flux was impaired by prolonged exposure to PA. In addition, the proportion of apoptotic cells was increased when autophagy was inhibited by the autophagy inhibitor 3‑methyladenine. Furthermore, inducing autophagy by pretreating cells with rapamycin, a known inducer of autophagy, markedly reduced PA‑induced apoptosis, suggesting that autophagy may serve a protective role in PA‑induced apoptosis in BMSCs. PA also increased intracellular reactive oxygen species (ROS) production, which was decreased by the antioxidant N‑Acetyl‑cysteine, and promoted the activation of c‑Jun N‑terminal kinases (JNKs) and p38 mitogen‑activated protein kinase (MAPK). The addition of JNK and p38 MAPK inhibitors substantially reduced autophagy. Therefore, the results indicated that PA can induce autophagy in BMSCs and protect cells from PA‑induced apoptosis through the ROS‑JNK/p38 MAPK signaling pathways. These results may improve the general understanding of the mechanisms through which BMSCs adapt to PA‑induced apoptosis. The present study also provides a novel approach for the prevention and treatment of PA‑induced lipotoxicity.
Collapse
Affiliation(s)
- Yongyi Liu
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, Liaoning 110000, P.R. China
| | - Ning Wang
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, Liaoning 110000, P.R. China
| | - Shaokun Zhang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Qingwei Liang
- Department of Sports Medicine, The First Hospital of China Medical University, Shenyang, Liaoning 110000, P.R. China
| |
Collapse
|
28
|
Shen C, Ma W, Ding L, Li S, Dou X, Song Z. The TLR4-IRE1α pathway activation contributes to palmitate-elicited lipotoxicity in hepatocytes. J Cell Mol Med 2018; 22:3572-3581. [PMID: 29673059 PMCID: PMC6010797 DOI: 10.1111/jcmm.13636] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 03/10/2018] [Indexed: 02/06/2023] Open
Abstract
Lipotoxicity induced by saturated fatty acids (SFAs) plays a pathological role in the development of non‐alcoholic fatty liver disease (NAFLD); however, the exact mechanism(s) remain to be clearly elucidated. Toll‐like receptor (TLR) 4 plays a fundamental role in activating the innate immune system. Intriguingly, hepatocytes express TLR4 and machinery for TLR4 signalling pathway. That liver‐specific TLR4 knockout mice are protective against diet‐induced NAFLD suggests that hepatocyte TLR4 signalling pathway plays an important role in NAFLD pathogenesis. Herein, using cultured hepatocytes, we sought to directly examine the role of TLR4 signalling pathway in palmitate‐elicited hepatotoxicity and to elucidate underlying mechanism(s). Our data reveal that palmitate exposure up‐regulates TLR4 expression at both mRNA and protein levels in hepatocytes, which are associated with NF‐κB activation. The inhibition of TLR4 signalling pathway through both pharmacological and genetic approaches abolished palmitate‐induced cell death, suggesting that TLR4 signalling pathway activation contributes to palmitate‐induced hepatotoxicity. Mechanistic investigations demonstrate that inositol‐requiring enzyme 1α (IRE1α), one of three major signal transduction pathways activated during endoplasmic reticulum (ER) stress, is the downstream target of palmitate‐elicited TLR4 activation and mechanistically implicated in TLR4 activation‐triggered cell death in response to palmitate exposure. Collectively, our data identify that the TLR4‐IRE1α pathway activation contributes to palmitate‐elicited lipotoxicity in hepatocytes. Our findings suggest that targeting TLR4‐IRE1α pathway can be a potential therapeutic choice for the treatment of NAFLD as well as other metabolic disorders, with lipotoxicity being the principal pathomechanism.
Collapse
Affiliation(s)
- Chen Shen
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL, USA
| | - Wang Ma
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lei Ding
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Songtao Li
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| | - Xiaobing Dou
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhenyuan Song
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL, USA.,College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China.,Department of Pathology, University of Illinois, Medical Center, Chicago, IL, USA
| |
Collapse
|
29
|
IFN-γ and TNF-α Pre-licensing Protects Mesenchymal Stromal Cells from the Pro-inflammatory Effects of Palmitate. Mol Ther 2017; 26:860-873. [PMID: 29352647 DOI: 10.1016/j.ymthe.2017.12.013] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Revised: 12/12/2017] [Accepted: 12/14/2017] [Indexed: 12/17/2022] Open
Abstract
The use of mesenchymal stromal cell (MSC) therapy for the treatment of type 2 diabetes (T2D) and T2D complications is promising; however, the investigation of MSC function in the setting of T2D has not been thoroughly explored. In our current study, we investigated the phenotype and function of MSCs in a simulated in vitro T2D environment. We show that palmitate, but not glucose, exposure impairs MSC metabolic activity with moderate increases in apoptosis, while drastically affecting proliferation and morphology. In co-culture with peripheral blood mononuclear cells (PBMCs), we found that MSCs not only lose their normal suppressive ability in high levels of palmitate, but actively support and enhance inflammation, resulting in elevated PBMC proliferation and pro-inflammatory cytokine release. The pro-inflammatory effect of MSCs in palmitate was partially reversed via palmitate removal and fully reversed through pre-licensing MSCs with interferon-gamma and tumor necrosis factor alpha. Thus, palmitate, a specific metabolic factor enriched within the T2D environment, is a potent modulator of MSC immunosuppressive function, which may in part explain the depressed potency observed in MSCs isolated from T2D patients. Importantly, we have also identified a robust and durable pre-licensing regimen that protects MSC immunosuppressive function in the setting of T2D.
Collapse
|
30
|
Colvin BN, Longtine MS, Chen B, Costa ML, Nelson DM. Oleate attenuates palmitate-induced endoplasmic reticulum stress and apoptosis in placental trophoblasts. Reproduction 2017; 153:369-380. [PMID: 28159805 DOI: 10.1530/rep-16-0576] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 12/02/2016] [Accepted: 01/03/2017] [Indexed: 12/24/2022]
Abstract
Pre-pregnancy obesity is increasingly common and predisposes pregnant women and offspring to gestational diabetes, pre-eclampsia, fetal growth abnormalities and stillbirth. Obese women exhibit elevated levels of the two most common dietary fatty acids, palmitate and oleate, and the maternal blood containing these nutrients bathes the surface of trophoblasts of placental villi in vivo We test the hypothesis that the composition and concentration of free fatty acids modulate viability and function of primary human villous trophoblasts in culture. We found that palmitate increases syncytiotrophoblast death, specifically by caspase-mediated apoptosis, whereas oleate does not cause enhanced cell death. Importantly, exposure to both fatty acids in equimolar amounts yielded no increase in death or apoptosis, suggesting that oleate can protect syncytiotrophoblasts from palmitate-induced death. We further found that palmitate, but not oleate or oleate with palmitate, increases endoplasmic reticulum (ER) stress, signaling through the unfolded protein response, and yielding CHOP-mediated induction of apoptosis. Finally, we show that oleate or oleate plus palmitate both lead to increased lipid droplets in syncytiotrophoblasts, whereas palmitate does not. The data show palmitate is toxic to human syncytiotrophoblasts, through the induction of ER stress and apoptosis mediated by CHOP, whereas oleate is not toxic, abrogates palmitate toxicity and induces fat accumulation. We speculate that our in vitro results offer pathways by which the metabolic milieu of the obese pregnant woman can yield villous trophoblast dysfunction and sub-optimal placental function.
Collapse
Affiliation(s)
| | - Mark S Longtine
- Department of Obstetrics and GynecologyWashington University School of Medicine, St Louis, Missouri, USA
| | - Baosheng Chen
- Department of Obstetrics and GynecologyWashington University School of Medicine, St Louis, Missouri, USA
| | - Maria Laura Costa
- Department of Obstetrics and GynecologyWashington University School of Medicine, St Louis, Missouri, USA.,Department of Obstetrics and GynecologyUniversidade Estadual de Campinas, Cidade Universitaria Zeferino Vaz, Campinas, Brazil
| | - D Michael Nelson
- Department of Obstetrics and GynecologyWashington University School of Medicine, St Louis, Missouri, USA
| |
Collapse
|
31
|
Shinjo S, Jiang S, Nameta M, Suzuki T, Kanai M, Nomura Y, Goda N. Disruption of the mitochondria-associated ER membrane (MAM) plays a central role in palmitic acid–induced insulin resistance. Exp Cell Res 2017; 359:86-93. [DOI: 10.1016/j.yexcr.2017.08.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 07/10/2017] [Accepted: 08/02/2017] [Indexed: 12/19/2022]
|
32
|
Li S, Dou X, Ning H, Song Q, Wei W, Zhang X, Shen C, Li J, Sun C, Song Z. Sirtuin 3 acts as a negative regulator of autophagy dictating hepatocyte susceptibility to lipotoxicity. Hepatology 2017; 66:936-952. [PMID: 28437863 PMCID: PMC5570642 DOI: 10.1002/hep.29229] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 04/06/2017] [Accepted: 04/18/2017] [Indexed: 01/04/2023]
Abstract
UNLABELLED Lipotoxicity induced by saturated fatty acids (SFAs) plays a central role in the pathogenesis of nonalcoholic fatty liver disease (NAFLD); however, the exact mechanisms remain to be fully elucidated. Sirtuin 3 (SIRT3) is a nicotinamide adenine dinucleotide-dependent deacetylase located primarily inside mitochondria. In this study, we demonstrated that an SFA-rich high-fat diet (HFD) was more detrimental to the liver than an isocaloric unsaturated HFD rich in fatty acids. Unexpectedly, SIRT3 expression and activity were significantly elevated in the livers of mice exposed to the SFA-rich HFD. Using cultured HepG2 and AML-12 hepatocytes, we demonstrated that unlike monounsaturated fatty acids, SFAs up-regulate SIRT3 expression and activity. SIRT3 overexpression renders both the liver and hepatocytes susceptible to palmitate-induced cell death, which can be alleviated by SIRT3 small interfering RNA (siRNA) transfection. In contrast, SIRT3 suppression protects hepatocytes from palmitate cytotoxicity. Further studies revealed that SIRT3 acts as a negative regulator of autophagy, thereby enhancing the susceptibility of hepatocytes to SFA-induced cytotoxicity. Mechanistic investigations revealed that SIRT3 overexpression causes manganese superoxide dismutase deacetylation and activation, which depleted intracellular superoxide contents, leading to adenosine monophosphate-activated protein kinase (AMPK) inhibition and mammalian target of rapamycin C1 activation, resulting in autophagy suppression. In contrast, SIRT3 siRNA gene silencing enhanced autophagy flux. A similar result was observed in the liver tissue of SIRT3 knockout mice. CONCLUSION Our data indicate that SIRT3 is a negative regulator of autophagy whose activation by SFAs contributes to lipotoxicity in hepatocytes and suggest that restraining SIRT3 overactivation can be a potential therapeutic choice for the treatment of NAFLD as well as other metabolic disorders, with lipotoxicity being the principal pathomechanism. (Hepatology 2017;66:936-952).
Collapse
Affiliation(s)
- Songtao Li
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612,Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, 150086, P. R. China
| | - Xiaobing Dou
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612,College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, P.R. China
| | - Hua Ning
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, 150086, P. R. China,Research Institute of Food, Nutrition and Health, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, 150086, P. R. China
| | - Qing Song
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, 150086, P. R. China
| | - Wei Wei
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, 150086, P. R. China
| | - Ximei Zhang
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612
| | - Chen Shen
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612
| | - Jiaxin Li
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612
| | - Changhao Sun
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, 150086, P. R. China,Research Institute of Food, Nutrition and Health, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, 150086, P. R. China
| | - Zhenyuan Song
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612,College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, P.R. China,Department of Pathology, University of Illinois, Medical Center, Chicago, IL 60612
| |
Collapse
|
33
|
Lu J, Wang QH, Huang LH, Dong HY, Lin LJ, Tan JM. Correlation of CDC42 Activity with Cell Proliferation and Palmitate-Mediated Cell Death in Human Umbilical Cord Wharton's Jelly Derived Mesenchymal Stromal Cells. Stem Cells Dev 2017; 26:1283-1292. [PMID: 28548571 DOI: 10.1089/scd.2017.0032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
RHO GTPases regulate cell migration, cell-cycle progression, and cell survival in response to extracellular stimuli. However, the regulatory effects of RHO GTPases in mesenchymal stromal cells (MSCs) are unclear. Herein, we show that CDC42 acts as an essential factor in regulating cell proliferation and also takes part in lipotoxic effects of palmitate in human umbilical cord Wharton's jelly derived MSCs (hWJ-MSCs). Cultured human bone marrow, adipose tissue, and hWJ-MSC derived cells had varying pro-inflammatory cytokine secretion levels and cell death rates when treated by palmitate. Strikingly, the proliferation rate of these types of MSCs correlated with their sensitivity to palmitate. A glutathione-S-transferase pull-down assay demonstrated that hWJ-MSCs had the highest activation of CDC42, which was increased by palmitate treatment in a time-dependent manner. We demonstrated that palmitate-induced synthesis of pro-inflammatory cytokines and cell death was attenuated by shRNA against CDC42. In CDC42 depleted hWJ-MSCs, population-doubling levels were notably decreased, and phosphorylation of ERK1/2 and p38 MAPK was reduced. Our data therefore suggest a mechanistic role for CDC42 activity in hWJ-MSC proliferation and identified CDC42 activity as a promising pharmacological target for ameliorating lipotoxic cell dysfunction and death.
Collapse
Affiliation(s)
- Jun Lu
- Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou General Hospital/or Dongfang Hospital, Xiamen University , Fuzhou, China
| | - Qing-Hua Wang
- Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou General Hospital/or Dongfang Hospital, Xiamen University , Fuzhou, China
| | - Liang-Hu Huang
- Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou General Hospital/or Dongfang Hospital, Xiamen University , Fuzhou, China
| | - Hui-Yue Dong
- Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou General Hospital/or Dongfang Hospital, Xiamen University , Fuzhou, China
| | - Ling-Jing Lin
- Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou General Hospital/or Dongfang Hospital, Xiamen University , Fuzhou, China
| | - Jian-Ming Tan
- Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou General Hospital/or Dongfang Hospital, Xiamen University , Fuzhou, China
| |
Collapse
|
34
|
Zou R, Xue J, Huang Q, Dai Z, Xu Y. Involvement of receptor-interacting protein 140 in palmitate-stimulated macrophage infiltration of pancreatic beta cells. Exp Ther Med 2017; 14:483-494. [PMID: 28672957 PMCID: PMC5488400 DOI: 10.3892/etm.2017.4544] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 01/06/2017] [Indexed: 02/06/2023] Open
Abstract
Receptor-interacting protein 140 (RIP140) in macrophages stimulates the nuclear factor-κB subunit RelA to activate tumor necrosis factor (TNF)-α and interleukin (IL)-6 transcription. However, under lipotoxic conditions, the involvement of RIP140 in the infiltration of beta cells by macrophages remains unknown. In the present study, murine RAW264.7 macrophages were transfected with a RIP140 overexpression plasmid or siRNA prior to macrophage activation with 500 µM palmitate. Palmitate-free conditioned media was then collected and added to murine insulinoma MIN6 cells. Significant decreases were observed in cell viability (P<0.01), glucose-stimulated insulin secretion (P<0.01) and levels of peroxisome proliferator-activated receptor-γ coactivator-1α (P<0.05), phosphoenolpyruvate carboxykinase and proliferating cell nuclear antigen mRNA (P<0.01) in MIN6 cells. In addition, conditioned media from palmitate-treated and RIP140-upregulated macrophages significantly increased the levels of uncoupling protein-2 (P<0.01), inducible nitric oxide synthase 1 (P<0.01) and pancreatic and duodenal homeobox 1 (P<0.05) mRNA and levels of activated Jun N-terminal kinase (JNK) (P<0.01) and extracellular signal-regulated kinase (ERK) 1/2 (P<0.01). In turn, the conditioned media was found to be significantly enriched in TNF-α and IL-6 (both P<0.05). These results were the opposite of those obtained from MIN6 cells treated with conditioned media from palmitate-treated and RIP140-knockdown macrophages. MIN6 cells were transfected with RIP140 overexpression plasmid or siRNA prior to treatment with 500 µM palmitate and supernatant was collected for use in macrophage chemotaxis assays. In the palmitate-activated and RIP140-overexpressing MIN6 cells, TNF-α and IL-6 secretion increased significantly (both P<0.05) and macrophage chemotaxis towards MIN6 cells was enhanced. By contrast, downregulating RIP140 in MIN6 cells had the opposite effect. These data suggest that RIP140 in macrophages mediates the transcription of inflammatory cytokines when concentration of palmitate is high. Macrophage RIP140 may also impair beta cell function by activating the JNK and ERK1/2 signaling pathways and promoting specific gene transcription. Furthermore, expression of RIP140 in pancreatic beta cells may stimulate macrophage chemotaxis, thus triggering local low-grade inflammation.
Collapse
Affiliation(s)
- Runmei Zou
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China.,Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Junli Xue
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Qi Huang
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Zhe Dai
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Yancheng Xu
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
35
|
Graner AN, Hellwinkel JE, Lencioni AM, Madsen HJ, Harland TA, Marchando P, Nguyen GJ, Wang M, Russell LM, Bemis LT, Anchordoquy TJ, Graner MW. HSP90 inhibitors in the context of heat shock and the unfolded protein response: effects on a primary canine pulmonary adenocarcinoma cell line. Int J Hyperthermia 2016; 33:303-317. [PMID: 27829290 DOI: 10.1080/02656736.2016.1256503] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Agents targeting HSP90 and GRP94 are seldom tested in stressed contexts such as heat shock (HS) or the unfolded protein response (UPR). Tumor stress often activates HSPs and the UPR as pro-survival mechanisms. This begs the question of stress effects on chemotherapeutic efficacy, particularly with drugs targeting chaperones such as HSP90 or GRP94. We tested the utility of several HSP90 inhibitors, including PU-H71 (targeting GRP94), on a primary canine lung cancer line under HS/UPR stress compared to control conditions. METHODS We cultured canine bronchoalveolar adenocarcinoma cells that showed high endogenous HSP90 and GRP94 expression; these levels substantially increased upon HS or UPR induction. We treated cells with HSP90 inhibitors 17-DMAG, 17-AAG or PU-H71 under standard conditions, HS or UPR. Cell viability/survival was assayed. Antibody arrays measured intracellular signalling and apoptosis profiles. RESULTS HS and UPR had varying effects on cells treated with different HSP90 inhibitors; in particular, HS and UPR promoted resistance to inhibitors in short-term assays, but combinations of UPR stress and PU-H571 showed potent cytotoxic activity in longer-term assays. Array data indicated altered signalling pathways, with apoptotic and pro-survival implications. UPR induction + dual targeting of HSP90 and GRP94 swayed the balance toward apoptosis. CONCLUSION Cellular stresses, endemic to tumors, or interventionally inducible, can deflect or enhance chemo-efficacy, particularly with chaperone-targeting drugs. Stress is likely not held accountable when testing new pharmacologics or assessing currently-used drugs. A better understanding of stress impacts on drug activities should be critical in improving therapeutic targeting and in discerning mechanisms of drug resistance.
Collapse
Affiliation(s)
- Arin N Graner
- a Department of Neurosurgery , University of Colorado Denver , Aurora , CO , USA
| | - Justin E Hellwinkel
- a Department of Neurosurgery , University of Colorado Denver , Aurora , CO , USA.,b School of Medicine , University of Colorado School of Medicine , Aurora , CO , USA
| | - Alex M Lencioni
- a Department of Neurosurgery , University of Colorado Denver , Aurora , CO , USA.,c University of Arkansas for Medical Sciences , Little Rock , AR , USA
| | - Helen J Madsen
- a Department of Neurosurgery , University of Colorado Denver , Aurora , CO , USA.,b School of Medicine , University of Colorado School of Medicine , Aurora , CO , USA
| | - Tessa A Harland
- a Department of Neurosurgery , University of Colorado Denver , Aurora , CO , USA.,b School of Medicine , University of Colorado School of Medicine , Aurora , CO , USA
| | - Paul Marchando
- d Department of Chemical and Biological Engineering , University of Colorado Boulder , Boulder , CO , USA
| | - Ger J Nguyen
- a Department of Neurosurgery , University of Colorado Denver , Aurora , CO , USA
| | - Mary Wang
- a Department of Neurosurgery , University of Colorado Denver , Aurora , CO , USA
| | - Laura M Russell
- a Department of Neurosurgery , University of Colorado Denver , Aurora , CO , USA
| | - Lynne T Bemis
- e Department of Biomedical Sciences , University of Minnesota , Duluth , MN , USA
| | - Thomas J Anchordoquy
- f Skaggs School of Pharmacy and Pharmaceutical Sciences , University of Colorado Denver , Aurora , CO , USA
| | - Michael W Graner
- a Department of Neurosurgery , University of Colorado Denver , Aurora , CO , USA
| |
Collapse
|
36
|
Yang Y, Cheung HH, Tu J, Miu KK, Chan WY. New insights into the unfolded protein response in stem cells. Oncotarget 2016; 7:54010-54027. [PMID: 27304053 PMCID: PMC5288239 DOI: 10.18632/oncotarget.9833] [Citation(s) in RCA: 22] [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: 03/29/2016] [Accepted: 05/29/2016] [Indexed: 12/15/2022] Open
Abstract
The unfolded protein response (UPR) is an evolutionarily conserved adaptive mechanism to increase cell survival under endoplasmic reticulum (ER) stress conditions. The UPR is critical for maintaining cell homeostasis under physiological and pathological conditions. The vital functions of the UPR in development, metabolism and immunity have been demonstrated in several cell types. UPR dysfunction activates a variety of pathologies, including cancer, inflammation, neurodegenerative disease, metabolic disease and immune disease. Stem cells with the special ability to self-renew and differentiate into various somatic cells have been demonstrated to be present in multiple tissues. These cells are involved in development, tissue renewal and certain disease processes. Although the role and regulation of the UPR in somatic cells has been widely reported, the function of the UPR in stem cells is not fully known, and the roles and functions of the UPR are dependent on the stem cell type. Therefore, in this article, the potential significances of the UPR in stem cells, including embryonic stem cells, tissue stem cells, cancer stem cells and induced pluripotent cells, are comprehensively reviewed. This review aims to provide novel insights regarding the mechanisms associated with stem cell differentiation and cancer pathology.
Collapse
Affiliation(s)
- Yanzhou Yang
- Key Laboratory of Fertility Preservation and Maintenance, Ministry of Education, Key Laboratory of Reproduction and Genetics in Ningxia, Department of Histology and Embryology, Ningxia Medical University, Yinchuan, Ningxia, P.R. China
- The Chinese University of Hong Kong–Shandong University Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, HKSAR, China
| | - Hoi Hung Cheung
- The Chinese University of Hong Kong–Shandong University Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, HKSAR, China
| | - JiaJie Tu
- The Chinese University of Hong Kong–Shandong University Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, HKSAR, China
| | - Kai Kei Miu
- The Chinese University of Hong Kong–Shandong University Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, HKSAR, China
| | - Wai Yee Chan
- The Chinese University of Hong Kong–Shandong University Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, HKSAR, China
| |
Collapse
|
37
|
Bosquet A, Guaita-Esteruelas S, Saavedra P, Rodríguez-Calvo R, Heras M, Girona J, Masana L. Exogenous FABP4 induces endoplasmic reticulum stress in HepG2 liver cells. Atherosclerosis 2016; 249:191-9. [DOI: 10.1016/j.atherosclerosis.2016.04.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 03/21/2016] [Accepted: 04/13/2016] [Indexed: 01/22/2023]
|
38
|
Snodgrass RG, Huang S, Namgaladze D, Jandali O, Shao T, Sama S, Brüne B, Hwang DH. Docosahexaenoic acid and palmitic acid reciprocally modulate monocyte activation in part through endoplasmic reticulum stress. J Nutr Biochem 2016; 32:39-45. [PMID: 27142735 DOI: 10.1016/j.jnutbio.2016.01.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Revised: 01/05/2016] [Accepted: 01/23/2016] [Indexed: 12/22/2022]
Abstract
Palmitic acid (C16:0) and TLR2 ligand induce, but docosahexaenoic acid (DHA) inhibits monocyte activation. C16:0 and TLR2 or TLR4 ligand induce certain ER stress markers; thus, we determined whether ER stress induced by these agonists is sufficient to induce monocyte activation, and whether the ER stress is inhibited by DHA which is known to inhibit C16:0- or ligand-induced TLR activation. Monocyte activation and ER stress were assessed by TLR/inflammasome-induced IL-1β production, and phosphorylation of IRE-1 and eIF2 and expression of CHOP, respectively in THP-1 cells. TLR2 ligand Pam3CSK4 induced phosphorylation of eIF2, but not phosphorylation of IRE-1 and CHOP expression. LPS also induced phosphorylation of both IRE-1 and eIF2 but not CHOP expression suggesting that TLR2 or TLR4 ligand, or C16:0 induces different ER stress responses. C16:0-, Pam3CSK4-, or LPS-induced IL-1β production was inhibited by 4-phenylbutyric acid, an inhibitor of ER stress suggesting that IL-1β production induced by these agonists is partly mediated through ER stress. Among two ER stress-inducing molecules, thapsigargin but not tunicamycin led to the expression of pro-IL-1β and secretion of IL-1β. Thus, not all types of ER stress are sufficient to induce inflammasome-mediated IL-1β secretion in monocytes. Although both C16:0 and thapsigargin-induced IL-1β secretion was inhibited by DHA, only C16:0-mediated ER stress was responsive to DHA. These findings suggest that the anti-inflammatory effects of DHA are at least in part mediated through modulating ER homeostasis and that the propensity of ER stress can be differentially modulated by the types of dietary fat we consume.
Collapse
Affiliation(s)
- Ryan G Snodgrass
- U.S. Department of Agriculture, Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA 95616; Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | - Shurong Huang
- U.S. Department of Agriculture, Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA 95616
| | - Dmitry Namgaladze
- Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | - Ola Jandali
- U.S. Department of Agriculture, Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA 95616
| | - Tiffany Shao
- U.S. Department of Agriculture, Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA 95616
| | - Spandana Sama
- U.S. Department of Agriculture, Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA 95616
| | - Bernhard Brüne
- Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | - Daniel H Hwang
- U.S. Department of Agriculture, Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA 95616.
| |
Collapse
|
39
|
Danino H, Ben-Dror K, Birk R. Exocrine pancreas ER stress is differentially induced by different fatty acids. Exp Cell Res 2015; 339:397-406. [DOI: 10.1016/j.yexcr.2015.09.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Revised: 09/26/2015] [Accepted: 09/29/2015] [Indexed: 12/13/2022]
|
40
|
Gillet C, Spruyt D, Rigutto S, Dalla Valle A, Berlier J, Louis C, Debier C, Gaspard N, Malaisse WJ, Gangji V, Rasschaert J. Oleate Abrogates Palmitate-Induced Lipotoxicity and Proinflammatory Response in Human Bone Marrow-Derived Mesenchymal Stem Cells and Osteoblastic Cells. Endocrinology 2015; 156:4081-93. [PMID: 26327577 DOI: 10.1210/en.2015-1303] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Osteoporosis is a metabolic bone disease associated with unequilibrated bone remodeling resulting from decreased bone formation and/or increased bone resorption, leading to progressive bone loss. In osteoporotic patients, low bone mass is associated with an increase of bone marrow fat resulting from accumulation of adipocytes within the bone marrow. Marrow adipocytes are active secretory cells, releasing cytokines, adipokines and free fatty acids (FA) that influence the bone marrow microenvironment and alter the biology of neighboring cells. Therefore, we examined the effect of palmitate (Palm) and oleate (Ole), 2 highly prevalent FA in human organism and diet, on the function and survival of human mesenchymal stem cells (MSC) and MSC-derived osteoblastic cells. The saturated FA Palm exerted a cytotoxic action via initiation of endoplasmic reticulum stress and activation of the nuclear factor κB (NF-κB) and ERK pathways. In addition, Palm induced a proinflammatory response, as determined by the up-regulation of Toll-like receptor 4 expression as well as the increase of IL-6 and IL-8 expression and secretion. Moreover, we showed that MSC-derived osteoblastic cells were more sensitive to lipotoxicity than undifferentiated MSC. The monounsaturated FA Ole fully neutralized Palm-induced lipotoxicity by impairing activation of the pathways triggered by the saturated FA. Moreover, Ole promoted Palm detoxification by fostering its esterification into triglycerides and storage in lipid droplets. Altogether, our data showed that physiological concentrations of Palm and Ole differently modulated cell death and function in bone cells. We therefore propose that FA could influence skeletal health.
Collapse
Affiliation(s)
- C Gillet
- Laboratory of Bone and Metabolic Biochemistry (C.G., D.S., S.R., A.D.V., J.B., N.G., W.J.M., V.G., J.R.), Faculty of Medicine, Université libre de Bruxelles, B-1070 Brussels, Belgium; Institute of Life Sciences (C.L., C.D.), Université catholique de Louvain, 1348 Louvain-la-Neuve, Belgium; and Department of Rheumatology and Physical Medicine (V.G.), Erasme Hospital, B-1070 Brussels, Belgium
| | - D Spruyt
- Laboratory of Bone and Metabolic Biochemistry (C.G., D.S., S.R., A.D.V., J.B., N.G., W.J.M., V.G., J.R.), Faculty of Medicine, Université libre de Bruxelles, B-1070 Brussels, Belgium; Institute of Life Sciences (C.L., C.D.), Université catholique de Louvain, 1348 Louvain-la-Neuve, Belgium; and Department of Rheumatology and Physical Medicine (V.G.), Erasme Hospital, B-1070 Brussels, Belgium
| | - S Rigutto
- Laboratory of Bone and Metabolic Biochemistry (C.G., D.S., S.R., A.D.V., J.B., N.G., W.J.M., V.G., J.R.), Faculty of Medicine, Université libre de Bruxelles, B-1070 Brussels, Belgium; Institute of Life Sciences (C.L., C.D.), Université catholique de Louvain, 1348 Louvain-la-Neuve, Belgium; and Department of Rheumatology and Physical Medicine (V.G.), Erasme Hospital, B-1070 Brussels, Belgium
| | - A Dalla Valle
- Laboratory of Bone and Metabolic Biochemistry (C.G., D.S., S.R., A.D.V., J.B., N.G., W.J.M., V.G., J.R.), Faculty of Medicine, Université libre de Bruxelles, B-1070 Brussels, Belgium; Institute of Life Sciences (C.L., C.D.), Université catholique de Louvain, 1348 Louvain-la-Neuve, Belgium; and Department of Rheumatology and Physical Medicine (V.G.), Erasme Hospital, B-1070 Brussels, Belgium
| | - J Berlier
- Laboratory of Bone and Metabolic Biochemistry (C.G., D.S., S.R., A.D.V., J.B., N.G., W.J.M., V.G., J.R.), Faculty of Medicine, Université libre de Bruxelles, B-1070 Brussels, Belgium; Institute of Life Sciences (C.L., C.D.), Université catholique de Louvain, 1348 Louvain-la-Neuve, Belgium; and Department of Rheumatology and Physical Medicine (V.G.), Erasme Hospital, B-1070 Brussels, Belgium
| | - C Louis
- Laboratory of Bone and Metabolic Biochemistry (C.G., D.S., S.R., A.D.V., J.B., N.G., W.J.M., V.G., J.R.), Faculty of Medicine, Université libre de Bruxelles, B-1070 Brussels, Belgium; Institute of Life Sciences (C.L., C.D.), Université catholique de Louvain, 1348 Louvain-la-Neuve, Belgium; and Department of Rheumatology and Physical Medicine (V.G.), Erasme Hospital, B-1070 Brussels, Belgium
| | - C Debier
- Laboratory of Bone and Metabolic Biochemistry (C.G., D.S., S.R., A.D.V., J.B., N.G., W.J.M., V.G., J.R.), Faculty of Medicine, Université libre de Bruxelles, B-1070 Brussels, Belgium; Institute of Life Sciences (C.L., C.D.), Université catholique de Louvain, 1348 Louvain-la-Neuve, Belgium; and Department of Rheumatology and Physical Medicine (V.G.), Erasme Hospital, B-1070 Brussels, Belgium
| | - N Gaspard
- Laboratory of Bone and Metabolic Biochemistry (C.G., D.S., S.R., A.D.V., J.B., N.G., W.J.M., V.G., J.R.), Faculty of Medicine, Université libre de Bruxelles, B-1070 Brussels, Belgium; Institute of Life Sciences (C.L., C.D.), Université catholique de Louvain, 1348 Louvain-la-Neuve, Belgium; and Department of Rheumatology and Physical Medicine (V.G.), Erasme Hospital, B-1070 Brussels, Belgium
| | - W J Malaisse
- Laboratory of Bone and Metabolic Biochemistry (C.G., D.S., S.R., A.D.V., J.B., N.G., W.J.M., V.G., J.R.), Faculty of Medicine, Université libre de Bruxelles, B-1070 Brussels, Belgium; Institute of Life Sciences (C.L., C.D.), Université catholique de Louvain, 1348 Louvain-la-Neuve, Belgium; and Department of Rheumatology and Physical Medicine (V.G.), Erasme Hospital, B-1070 Brussels, Belgium
| | - V Gangji
- Laboratory of Bone and Metabolic Biochemistry (C.G., D.S., S.R., A.D.V., J.B., N.G., W.J.M., V.G., J.R.), Faculty of Medicine, Université libre de Bruxelles, B-1070 Brussels, Belgium; Institute of Life Sciences (C.L., C.D.), Université catholique de Louvain, 1348 Louvain-la-Neuve, Belgium; and Department of Rheumatology and Physical Medicine (V.G.), Erasme Hospital, B-1070 Brussels, Belgium
| | - J Rasschaert
- Laboratory of Bone and Metabolic Biochemistry (C.G., D.S., S.R., A.D.V., J.B., N.G., W.J.M., V.G., J.R.), Faculty of Medicine, Université libre de Bruxelles, B-1070 Brussels, Belgium; Institute of Life Sciences (C.L., C.D.), Université catholique de Louvain, 1348 Louvain-la-Neuve, Belgium; and Department of Rheumatology and Physical Medicine (V.G.), Erasme Hospital, B-1070 Brussels, Belgium
| |
Collapse
|
41
|
Octanoate in Human Albumin Preparations Is Detrimental to Mesenchymal Stromal Cell Culture. Stem Cells Int 2015; 2015:192576. [PMID: 26074972 PMCID: PMC4444585 DOI: 10.1155/2015/192576] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 03/29/2015] [Accepted: 04/15/2015] [Indexed: 12/17/2022] Open
Abstract
Cell therapies hold great promise as the next major advance in medical treatment. To enable safe, effective ex vivo culture whilst maintaining cell phenotype, growth media constituents must be carefully controlled. We have used a chemically defined mesenchymal stromal cell culture medium to investigate the influence of different preparations of human serum albumin. We examined two aspects of cell culture, growth rate as measured by population doubling time and colony forming ability which is a representative measure of the stemness of the cell population. Albumin preparations showed comparative differences in both of these criteria. Analysis of the albumin bound fatty acids also showed differences depending on the manufacturing procedure used. We demonstrated that octanoate, an additive used to stabilize albumin during pasteurization, slows growth and lowers colony forming ability during ex vivo culture. Further to this we also found the level of Na+/K+ ATPase, a membrane bound cation pump inhibited by octanoate, is increased in cells exposed to this compound. We conclude that the inclusion of human serum albumin in ex vivo growth media requires careful consideration of not only the source of albumin, but also the associated molecular cargo, for optimal cell growth and behavior.
Collapse
|
42
|
Fillmore N, Huqi A, Jaswal JS, Mori J, Paulin R, Haromy A, Onay-Besikci A, Ionescu L, Thébaud B, Michelakis E, Lopaschuk GD. Effect of fatty acids on human bone marrow mesenchymal stem cell energy metabolism and survival. PLoS One 2015; 10:e0120257. [PMID: 25768019 PMCID: PMC4358990 DOI: 10.1371/journal.pone.0120257] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 01/24/2015] [Indexed: 01/07/2023] Open
Abstract
Successful stem cell therapy requires the optimal proliferation, engraftment, and differentiation of stem cells into the desired cell lineage of tissues. However, stem cell therapy clinical trials to date have had limited success, suggesting that a better understanding of stem cell biology is needed. This includes a better understanding of stem cell energy metabolism because of the importance of energy metabolism in stem cell proliferation and differentiation. We report here the first direct evidence that human bone marrow mesenchymal stem cell (BMMSC) energy metabolism is highly glycolytic with low rates of mitochondrial oxidative metabolism. The contribution of glycolysis to ATP production is greater than 97% in undifferentiated BMMSCs, while glucose and fatty acid oxidation combined only contribute 3% of ATP production. We also assessed the effect of physiological levels of fatty acids on human BMMSC survival and energy metabolism. We found that the saturated fatty acid palmitate induces BMMSC apoptosis and decreases proliferation, an effect prevented by the unsaturated fatty acid oleate. Interestingly, chronic exposure of human BMMSCs to physiological levels of palmitate (for 24 hr) reduces palmitate oxidation rates. This decrease in palmitate oxidation is prevented by chronic exposure of the BMMSCs to oleate. These results suggest that reducing saturated fatty acid oxidation can decrease human BMMSC proliferation and cause cell death. These results also suggest that saturated fatty acids may be involved in the long-term impairment of BMMSC survival in vivo.
Collapse
Affiliation(s)
- Natasha Fillmore
- Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Alda Huqi
- Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Jagdip S. Jaswal
- Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Jun Mori
- Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Roxane Paulin
- Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Alois Haromy
- Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Arzu Onay-Besikci
- Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Lavinia Ionescu
- Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Bernard Thébaud
- Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Evangelos Michelakis
- Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Gary D. Lopaschuk
- Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
- * E-mail:
| |
Collapse
|
43
|
Lee KM, Yang SJ, Park S, Choi YD, Shin HK, Pak JH, Park CS, Kim I. Depletion of the cereblon gene activates the unfolded protein response and protects cells from ER stress-induced cell death. Biochem Biophys Res Commun 2015; 458:34-9. [PMID: 25619137 DOI: 10.1016/j.bbrc.2015.01.054] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 01/13/2015] [Indexed: 11/29/2022]
Abstract
Previous studies showed that cereblon (CRBN) binds to various cellular target proteins, implying that CRBN regulates a wide range of cell responses. In this study, we found that deletion of the Crbn gene desensitized mouse embryonic fibroblast cells to various cell death-promoting stimuli, including endoplasmic reticulum stress inducers. Mechanistically, deletion of Crbn activates pathways involved in the unfolded protein response prior to ER stress induction. Loss of Crbn activated PKR-like ER kinase (PERK) with enhanced phosphorylation of eIF2α. Following ER stress induction, loss of Crbn delayed dephosphorylation of eIF2α, while reconstitution of Crbn reversed enhanced phosphorylation of PERK and eIF2α. Lastly, we found that activation of the PERK/eIF2α pathway following Crbn deletion is caused by activation of AMP-activated protein kinase (AMPK). We propose that CRBN plays a role in cellular stress signaling, including the unfolded protein response, by controlling the activity of AMPK.
Collapse
Affiliation(s)
- Kwang Min Lee
- School of Life Sciences and Cell Dynamics Research Center and National Leading Research Laboratory, Gwangju Institute Science and Technology (GIST), Gwangju 500-712, Republic of Korea; Department of Molecular Genetics, University of Texas South Western Medical Center, Dallas, TX 75390-9046, USA
| | - Seung-Joo Yang
- School of Life Sciences and Cell Dynamics Research Center and National Leading Research Laboratory, Gwangju Institute Science and Technology (GIST), Gwangju 500-712, Republic of Korea
| | - Sojung Park
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 138-736, Republic of Korea
| | - Yoo Duk Choi
- Department of Pathology, Chonnam National University Medical School, Gwangju 501-757, Republic of Korea
| | - Hwa Kyoung Shin
- Division of Meridian and Structural Medicine, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Jhang Ho Pak
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 138-736, Republic of Korea; Department of Medicine, University of Ulsan College of Medicine, Seoul 138-736, Republic of Korea
| | - Chul-Seung Park
- School of Life Sciences and Cell Dynamics Research Center and National Leading Research Laboratory, Gwangju Institute Science and Technology (GIST), Gwangju 500-712, Republic of Korea.
| | - Inki Kim
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 138-736, Republic of Korea; Department of Medicine, University of Ulsan College of Medicine, Seoul 138-736, Republic of Korea.
| |
Collapse
|
44
|
Alvarez-Garcia O, Rogers NH, Smith RG, Lotz MK. Palmitate has proapoptotic and proinflammatory effects on articular cartilage and synergizes with interleukin-1. Arthritis Rheumatol 2014; 66:1779-88. [PMID: 24591481 DOI: 10.1002/art.38399] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 02/04/2014] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Obesity is a major risk factor for the development of osteoarthritis (OA) that is associated with a state of low-grade inflammation and increased circulating levels of adipokines and free fatty acids (FFAs). The aim of this study was to analyze the effects of saturated (palmitate) and monounsaturated (oleate) FFAs on articular chondrocytes, synoviocytes, and cartilage. METHODS Human articular chondrocytes and fibroblast-like synoviocytes obtained from young healthy donors and OA chondrocytes from patients undergoing total knee replacement surgery were treated with palmitate or oleate alone or in combination with interleukin-1β (IL-1β). Cell viability, caspase activation, and gene expression of proinflammatory factors, extracellular matrix (ECM) proteins, and proteases were studied. In addition, chondrocyte viability, IL-6 production, and matrix damage were assessed in bovine and human articular cartilage explants cultured with FFAs in the presence or absence of IL-1β. RESULTS Palmitate, but not oleate, induced caspase activation and cell death in IL-1β-stimulated normal chondrocytes, and up-regulated the expression of IL-6 and cyclooxygenase 2 in chondrocytes and fibroblast-like synoviocytes through Toll-like receptor 4 (TLR-4) signaling. In cartilage explants, palmitate induced chondrocyte death, IL-6 release, and ECM degradation. Palmitate synergized with IL-1β in stimulating proapoptotic and proinflammatory cellular responses. Pharmacologic inhibition of caspases or TLR-4 signaling reduced palmitate and IL-1β induced cartilage damage. CONCLUSION Palmitate acts as a proinflammatory and catabolic factor that, in synergy with IL-1β, induces chondrocyte apoptosis and articular cartilage breakdown. Collectively, our data suggest that elevated levels of saturated FFAs that are often found in patients who are obese may contribute to the pathogenesis of OA.
Collapse
|
45
|
Hayashi H, Yamada R, Das SS, Sato T, Takahashi A, Hiratsuka M, Hirasawa N. Glucagon-like peptide-1 production in the GLUTag cell line is impaired by free fatty acids via endoplasmic reticulum stress. Metabolism 2014; 63:800-11. [PMID: 24680601 DOI: 10.1016/j.metabol.2014.02.012] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 02/15/2014] [Accepted: 02/17/2014] [Indexed: 12/16/2022]
Abstract
OBJECTS Glucagon-like peptide-1 (GLP-1) is secreted from intestinal L cells, enhances glucose-stimulated insulin secretion, and protects pancreas beta cells. However, few studies have examined hypernutrition stress in L cells and its effects on their function. Here, we demonstrated that a high-fat diet reduced glucose-stimulated secretion of GLP-1 and induced expression of an endoplasmic reticulum (ER) stress markers in the intestine of a diet-induced obesity mouse model. METHODS To clarify whether ER stress in L cells caused the attenuation of GLP-1 secretion, we treated the mouse intestinal L cell line, GLUTag cells with palmitate or oleate. RESULTS Palmitate, but not oleate caused ER stress and decreased the protein levels of prohormone convertase 1/3 (PC1/3), an essential enzyme in GLP-1 production. The same phenomena were observed in GLUTag cells treated with in ER stress inducer, thapsigargin. Moreover, oleate improved palmitate-induced ER stress, reduced protein and activity levels of PC1/3, and attenuated GLP-1 secretion from GLUTag cells. CONCLUSIONS/INTERPRETATION These results suggest that the intake of abundant saturated fatty acids induces ER stress in the intestine and decreases GLP-1 production.
Collapse
Affiliation(s)
- Hiroto Hayashi
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Ren Yamada
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Siddhartha Shankar Das
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Taiki Sato
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Aki Takahashi
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Masahiro Hiratsuka
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Noriyasu Hirasawa
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan.
| |
Collapse
|
46
|
Hui T, Wang C, Ye L. Comments on ethylenediaminetetraacetic acid contained in trypsin detaching cells for apoptosis detection. Stem Cells 2014; 32:1993-4. [PMID: 24677738 DOI: 10.1002/stem.1712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Revised: 01/16/2014] [Accepted: 01/18/2014] [Indexed: 02/05/2023]
Affiliation(s)
- Tianqian Hui
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | | | | |
Collapse
|
47
|
Shang L, Hua H, Foo K, Martinez H, Watanabe K, Zimmer M, Kahler DJ, Freeby M, Chung W, LeDuc C, Goland R, Leibel RL, Egli D. β-cell dysfunction due to increased ER stress in a stem cell model of Wolfram syndrome. Diabetes 2014; 63:923-33. [PMID: 24227685 PMCID: PMC3931392 DOI: 10.2337/db13-0717] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Wolfram syndrome is an autosomal recessive disorder caused by mutations in WFS1 and is characterized by insulin-dependent diabetes mellitus, optic atrophy, and deafness. To investigate the cause of β-cell failure, we used induced pluripotent stem cells to create insulin-producing cells from individuals with Wolfram syndrome. WFS1-deficient β-cells showed increased levels of endoplasmic reticulum (ER) stress molecules and decreased insulin content. Upon exposure to experimental ER stress, Wolfram β-cells showed impaired insulin processing and failed to increase insulin secretion in response to glucose and other secretagogues. Importantly, 4-phenyl butyric acid, a chemical protein folding and trafficking chaperone, restored normal insulin synthesis and the ability to upregulate insulin secretion. These studies show that ER stress plays a central role in β-cell failure in Wolfram syndrome and indicate that chemical chaperones might have therapeutic relevance under conditions of ER stress in Wolfram syndrome and other forms of diabetes.
Collapse
Affiliation(s)
- Linshan Shang
- The New York Stem Cell Foundation Research Institute, New York, NY
| | - Haiqing Hua
- The New York Stem Cell Foundation Research Institute, New York, NY
- Division of Molecular Genetics, Department of Pediatrics and Naomi Berrie Diabetes Center, Columbia University, New York, NY
| | - Kylie Foo
- Division of Molecular Genetics, Department of Pediatrics and Naomi Berrie Diabetes Center, Columbia University, New York, NY
| | - Hector Martinez
- The New York Stem Cell Foundation Research Institute, New York, NY
| | - Kazuhisa Watanabe
- Division of Molecular Genetics, Department of Pediatrics and Naomi Berrie Diabetes Center, Columbia University, New York, NY
| | - Matthew Zimmer
- The New York Stem Cell Foundation Research Institute, New York, NY
| | - David J. Kahler
- The New York Stem Cell Foundation Research Institute, New York, NY
| | - Matthew Freeby
- Division of Molecular Genetics, Department of Pediatrics and Naomi Berrie Diabetes Center, Columbia University, New York, NY
| | - Wendy Chung
- Division of Molecular Genetics, Department of Pediatrics and Naomi Berrie Diabetes Center, Columbia University, New York, NY
| | - Charles LeDuc
- Division of Molecular Genetics, Department of Pediatrics and Naomi Berrie Diabetes Center, Columbia University, New York, NY
| | - Robin Goland
- Division of Molecular Genetics, Department of Pediatrics and Naomi Berrie Diabetes Center, Columbia University, New York, NY
| | - Rudolph L. Leibel
- Division of Molecular Genetics, Department of Pediatrics and Naomi Berrie Diabetes Center, Columbia University, New York, NY
| | - Dieter Egli
- The New York Stem Cell Foundation Research Institute, New York, NY
- Corresponding author: Dieter Egli,
| |
Collapse
|
48
|
Jung TW, Lee SY, Hong HC, Choi HY, Yoo HJ, Baik SH, Choi KM. AMPK activator-mediated inhibition of endoplasmic reticulum stress ameliorates carrageenan-induced insulin resistance through the suppression of selenoprotein P in HepG2 hepatocytes. Mol Cell Endocrinol 2014; 382:66-73. [PMID: 24055274 DOI: 10.1016/j.mce.2013.09.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 08/11/2013] [Accepted: 09/10/2013] [Indexed: 11/23/2022]
Abstract
Carrageenan (CGN) has been shown to cause inflammation through toll-like receptor 4, which may play an important role in insulin resistance and type 2 diabetes mellitus. Selenoprotein P (SeP) has recently been identified as a novel hepatokine that causes insulin resistance. Here, we report that treatment of HepG2 cells with CGN increased both CCAAT enhancer binding protein homologous protein (CHOP) and SeP expression. Pretreatment with 4-phenylbutyrate (4-PBA), an endoplasmic reticulum stress inhibitor, and PD98059, a c-Jun N-terminal kinase (JNK) inhibitor, reversed CGN-induced SeP expression. Moreover, both 4-PBA and knock-down of SeP improved CGN-induced insulin resistance. In addition, we found that adenosine monophosphate-activated protein kinase (AMPK) activators ameliorated CGN-induced insulin resistance in addition to suppressing CHOP and SeP expression. In conclusion, CGN-induced ER stress increased the expression of SeP through the JNK pathway, while AMPK activators ameliorated CGN-induced insulin resistance via SeP inhibition through the AMPK-mediated alleviation of ER stress in hepatocytes.
Collapse
Affiliation(s)
- Tae Woo Jung
- The Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Korea University, Seoul, Republic of Korea
| | - So Young Lee
- The Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Ho Cheol Hong
- The Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Hae Yoon Choi
- The Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Hye Jin Yoo
- The Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Sei Hyun Baik
- The Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Kyung Mook Choi
- The Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Korea University, Seoul, Republic of Korea.
| |
Collapse
|
49
|
Li S, Li J, Shen C, Zhang X, Sun S, Cho M, Sun C, Song Z. tert-Butylhydroquinone (tBHQ) protects hepatocytes against lipotoxicity via inducing autophagy independently of Nrf2 activation. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1841:22-33. [PMID: 24055888 DOI: 10.1016/j.bbalip.2013.09.004] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 08/23/2013] [Accepted: 09/10/2013] [Indexed: 12/30/2022]
Abstract
Saturated fatty acids (SFAs) induce hepatocyte cell death, wherein oxidative stress is mechanistically involved. Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) is a master transcriptional regulator of cellular antioxidant defense enzymes. Therefore, Nrf2 activation is regarded as an effective strategy against oxidative stress-triggered cellular damage. In this study, tert-butylhydroquinone (tBHQ), a widely used Nrf2 activator, was initially employed to investigate the potential protective role of Nrf2 activation in SFA-induced hepatoxicity. As expected, SFA-induced hepatocyte cell death was prevented by tBHQ in both AML-12 mouse hepatocytes and HepG2 human hepatoma cells. However, the protective effect of tBHQ is Nrf2-independent, because the siRNA-mediated Nrf2 silencing did not abrogate tBHQ-conferred protection. Alternatively, our results revealed that autophagy activation was critically involved in the protective effect of tBHQ on lipotoxicity. tBHQ induced autophagy activation and autophagy inhibitors abolished tBHQ's protection. The induction of autophagy by tBHQ exposure was demonstrated by the increased accumulation of LC3 puncta, LC3-II conversion, and autophagic flux (LC3-II conversion in the presence of proteolysis inhibitors). Subsequent mechanistic investigation discovered that tBHQ exposure activated AMP-activated protein kinase (AMPK) and siRNA-mediated AMPK gene silencing abolished tBHQ-induced autophagy activation, indicating that AMPK is critically involved in tBHQ-triggered autophagy induction. Furthermore, our study provided evidence that tBHQ-induced autophagy activation is required for its Nrf2-activating property. Collectively, our data uncover a novel mechanism for tBHQ in protecting hepatocytes against SFA-induced lipotoxicity. tBHQ-triggered autophagy induction contributes not only to its hepatoprotective effect, but also to its Nrf2-activating property.
Collapse
Affiliation(s)
- Songtao Li
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612, USA; Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin 150081, PR China
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Wikstrom JD, Israeli T, Bachar-Wikstrom E, Swisa A, Ariav Y, Waiss M, Kaganovich D, Dor Y, Cerasi E, Leibowitz G. AMPK regulates ER morphology and function in stressed pancreatic β-cells via phosphorylation of DRP1. Mol Endocrinol 2013; 27:1706-23. [PMID: 23979843 DOI: 10.1210/me.2013-1109] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Experimental lipotoxicity constitutes a model for β-cell demise induced by metabolic stress in obesity and type 2 diabetes. Fatty acid excess induces endoplasmic reticulum (ER) stress, which is accompanied by ER morphological changes whose mechanisms and relevance are unknown. We found that the GTPase dynamin-related protein 1 (DRP1), a key regulator of mitochondrial fission, is an ER resident regulating ER morphology in stressed β-cells. Inhibition of DRP1 activity using a GTP hydrolysis-defective mutant (Ad-K38A) attenuated fatty acid-induced ER expansion and mitochondrial fission. Strikingly, stimulating the key energy-sensor AMP-activated protein kinase (AMPK) increased the phosphorylation at the anti-fission site Serine 637 and largely prevented the alterations in ER and mitochondrial morphology. Expression of a DRP1 mutant resistant to phosphorylation at this position partially prevented the recovery of ER and mitochondrial morphology by AMPK. Fatty acid-induced ER enlargement was associated with proinsulin retention in the ER, together with increased proinsulin/insulin ratio. Stimulation of AMPK prevented these alterations, as well as mitochondrial fragmentation and apoptosis. In summary, DRP1 regulation by AMPK delineates a novel pathway controlling ER and mitochondrial morphology, thereby modulating the response of β-cells to metabolic stress. DRP1 may thus function as a node integrating signals from stress regulators, such as AMPK, to coordinate organelle shape and function.
Collapse
Affiliation(s)
- Jakob D Wikstrom
- MD, Endocrinology and Metabolism Service, Department of Medicine, Hadassah-Hebrew University Medical Center, PO Box 12000, Jerusalem 91120.
| | | | | | | | | | | | | | | | | | | |
Collapse
|