1
|
Kim YJ, Lee JR, Kim MR, Jeong JA, Kim JJ, Jeong KW. Protein kinase-mediated inhibition of autophagy by palmitic acid in hepatocytes. Eur J Pharmacol 2025; 998:177528. [PMID: 40113068 DOI: 10.1016/j.ejphar.2025.177528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/03/2025] [Accepted: 03/18/2025] [Indexed: 03/22/2025]
Abstract
Steatosis is characterized by an increase in free fatty acids, such as palmitic acid (PA), in hepatocytes and the accumulation of triglycerides in the liver. However, the role of intracellular autophagy in PA accumulation-induced hepatotoxicity is not clearly understood. Therefore, in this study, we investigated the effects of PA on autophagy in hepatocytes and its underlying mechanism of action. Treatment of HepG2 cells with PA induced a significant increase in intracellular p62 and LC3-II levels, suggesting inhibition of autophagy. Furthermore, PA inhibited autophagic flux in HepG2 cells, as monitored using GFP-RFP-LC3. Mechanistically, PA increased the phosphorylation of the Ser12 and Thr29 residues of LC3, which are autophagy inhibition markers, through protein kinase A (PKA) and protein kinase C (PKC) signaling. Finally, PKA and PKC inhibitors restored PA-induced autophagic flux inhibition, reduced intracellular lipid accumulation, and rescued the altered expression of lipogenic genes, such as SREBP-1c, in HepG2 cells. Thus, our study demonstrates the mechanism of autophagy inhibition by PA in hepatocytes and provides a potential therapeutic approach for preventing and treating hepatic steatosis.
Collapse
Affiliation(s)
- Yeon Jeong Kim
- College of Pharmacy, Gachon Research Institute of Pharmaceutical Sciences, Gachon University, Incheon, 21936, Republic of Korea
| | - Jae Rim Lee
- College of Pharmacy, Gachon Research Institute of Pharmaceutical Sciences, Gachon University, Incheon, 21936, Republic of Korea
| | - Myeong Ryeo Kim
- College of Pharmacy, Gachon Research Institute of Pharmaceutical Sciences, Gachon University, Incheon, 21936, Republic of Korea
| | - Jin Ah Jeong
- College of Pharmacy, Gachon Research Institute of Pharmaceutical Sciences, Gachon University, Incheon, 21936, Republic of Korea
| | - Jung Ju Kim
- Autophagy Sciences Inc., Seoul, 08376, Republic of Korea
| | - Kwang Won Jeong
- College of Pharmacy, Gachon Research Institute of Pharmaceutical Sciences, Gachon University, Incheon, 21936, Republic of Korea.
| |
Collapse
|
2
|
Pierre L, Juszczak F, Delmotte V, Decarnoncle M, Ledoux B, Bultot L, Bertrand L, Boonen M, Renard P, Arnould T, Declèves AE. AMPK protects proximal tubular epithelial cells from lysosomal dysfunction and dedifferentiation induced by lipotoxicity. Autophagy 2025; 21:860-880. [PMID: 39675352 PMCID: PMC11925112 DOI: 10.1080/15548627.2024.2435238] [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: 03/18/2024] [Revised: 11/13/2024] [Accepted: 11/25/2024] [Indexed: 12/17/2024] Open
Abstract
Renal proximal tubules are a primary site of injury in metabolic diseases. In obese patients and animal models, proximal tubular epithelial cells (PTECs) display dysregulated lipid metabolism, organelle dysfunctions, and oxidative stress that contribute to interstitial inflammation, fibrosis and ultimately end-stage renal failure. Our research group previously pointed out AMP-activated protein kinase (AMPK) decline as a driver of obesity-induced renal disease. Because PTECs display high macroautophagic/autophagic activity and rely heavily on their endo-lysosomal system, we investigated the effect of lipid stress on autophagic flux and lysosomes in these cells. Using a model of highly differentiated primary PTECs challenged with palmitate, our data placed lysosomes at the cornerstone of the lipotoxic phenotype. As soon as 6 h after palmitate exposure, cells displayed impaired lysosomal acidification subsequently leading to autophagosome accumulation and activation of lysosomal biogenesis. We also showed the inability of lysosomal quality control to restore acidic pH which finally drove PTECs dedifferentiation. When palmitate-induced AMPK activity decline was prevented by AMPK activators, lysosomal acidification and the differentiation profile of PTECs were preserved. Our work provided key insights on the importance of lysosomes in PTECs homeostasis and lipotoxicity and demonstrated the potential of AMPK in protecting the organelle from lipid stress.Abbreviation: ACAC: acetyl-CoA carboxylase; ACTB: actin beta; AICAR: 5-aminoimidazole-4-carboxamide-1-β-D-ribofuranoside; AMPK: AMP-activated protein kinase; APQ1: aquaporin 1 (Colton blood group); BSA: bovine serum albumin; CDH16: cadherin 16; CKD: chronic kidney disease; CTSB: cathepsin B; CTSD: cathepsin D; EPB41L5: erythrocyte membrane protein band 4.1 like 5; EIF4EBP1: eukaryotic translation initiation factor 4E binding protein 1; EMT: epithelial-to-mesenchymal transition; FA: fatty acid; FCCP: carbonyl cyanide 4-(trifluoromethoxy)phenylhydrazone; GFP: green fluorescent protein; GUSB: glucuronidase beta; HEXB: hexosaminidase subunit beta; LAMP: lysosomal associated membrane protein; LD: lipid droplet; LGALS3: galectin 3; LLOMe: L-leucyl-L-leucine methyl ester hydrobromide; LMP: lysosomal membrane permeabilization; LRP2: LDL receptor related protein 2; LSD: lysosomal storage disorder; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MCOLN1: mucolipin TRP cation channel 1; MG132: N-benzyloxycarbonyl-L-leucyl-L-leucyl-L-leucinal; MmPTECs: Mus musculus (mouse) proximal tubular epithelial cells; MTORC1: mechanistic target of rapamycin kinase complex 1; OA: oleate; PA: palmitate; PIKFYVE: phosphoinositide kinase, FYVE-type zinc finger containing; PTs: proximal tubules; PTECs: proximal tubular epithelial cells; PRKAA: protein kinase AMP-activated catalytic subunit alpha; RFP: red fluorescent protein; RPS6KB: ribosomal protein S6 kinase B; SLC5A2: solute carrier family 5 member 2; SOX9: SRY-box transcription factor 9; SQSTM1: sequestosome 1; TFEB: transcription factor EB; Ub: ubiquitin; ULK1: unc-51 like autophagy activating kinase 1; VIM: vimentin.
Collapse
Affiliation(s)
- Louise Pierre
- Laboratory of Biochemistry and Cell Biology, Namur Research Institute for Life Sciences (NARILIS), University of Namur, Namur, Belgium
- Laboratory of Metabolic and Molecular Biochemistry, Faculty of Medicine and Pharmacy, Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
| | - Florian Juszczak
- Laboratory of Metabolic and Molecular Biochemistry, Faculty of Medicine and Pharmacy, Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
| | - Valentine Delmotte
- Laboratory of Biochemistry and Cell Biology, Namur Research Institute for Life Sciences (NARILIS), University of Namur, Namur, Belgium
| | - Morgane Decarnoncle
- Laboratory of Metabolic and Molecular Biochemistry, Faculty of Medicine and Pharmacy, Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
| | - Benjamin Ledoux
- Laboratory of Biochemistry and Cell Biology, Namur Research Institute for Life Sciences (NARILIS), University of Namur, Namur, Belgium
| | - Laurent Bultot
- Pole of Cardiovascular Research, Experimental and Clinical Research Institute (CARD), UCLouvain, Brussels, Belgium
| | - Luc Bertrand
- Pole of Cardiovascular Research, Experimental and Clinical Research Institute (CARD), UCLouvain, Brussels, Belgium
- WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Marielle Boonen
- URPhyM, Intracellular Trafficking Biology, NARILIS, University of Namur, Namur, Belgium
| | - Patricia Renard
- Laboratory of Biochemistry and Cell Biology, Namur Research Institute for Life Sciences (NARILIS), University of Namur, Namur, Belgium
| | - Thierry Arnould
- Laboratory of Biochemistry and Cell Biology, Namur Research Institute for Life Sciences (NARILIS), University of Namur, Namur, Belgium
| | - Anne-Emilie Declèves
- Laboratory of Metabolic and Molecular Biochemistry, Faculty of Medicine and Pharmacy, Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
| |
Collapse
|
3
|
Eckel JC, Seidemann L, Albadry M, Schicht G, Skvoznikova M, Nickel S, Hänsel R, Seehofer D, Hiller GGR, Tautenhahn HM, Dahmen U, Damm G. Application of rapid evaporative ionization mass spectrometry in preclinical and clinical analyses of steatotic liver tissues and cells. Sci Rep 2025; 15:9226. [PMID: 40097529 PMCID: PMC11914079 DOI: 10.1038/s41598-025-93305-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 03/05/2025] [Indexed: 03/19/2025] Open
Abstract
Rapid evaporative ionization mass spectrometry (REIMS) shows promise as a preparation-free tissue analysis tool with the prospect for real-time diagnostics. Given that hepatic steatosis is characterized by shifts in lipid species and abundance, we selected it as basis for method development, as REIMS specifically measures lipidomic profiles. However, further validation and protocol refinement are necessary to establish its clinical utility. In this study, we applied REIMS to steatotic human liver tissues, focusing on its ability to differentiate varying degrees of steatosis. We established standardized protocols for tissue handling and lipid analysis, which were essential for reliable data interpretation. Notably, our findings revealed that tissue size impacts REIMS sensitivity, with smaller samples yielding lower total ion counts and altered lipid profiles. Through principal component analysis, we identified key lipid classes, namely triacylglycerides, fatty acids, and glycerophospholipids. Despite a missing link between triacylglyceride abundance and degree of steatosis, we successfully identified condition-specific lipid patterns, with ceramides emerging as markers of advanced steatosis. Our study provides a protocol for the measurements of lipid standards showing the detailed degradation of specific lipids using iKnife-coupled REIMS. It highlights the pitfalls and limitations and provides critical recommendations for REIMS use. It also emphasizes the need for standardized biobanking and tissue preparation to ensure accurate lipid profiling, laying the groundwork for future protocol adjustments required for clinical application.
Collapse
Affiliation(s)
- Julian Connor Eckel
- Department of Hepatobiliary Surgery and Visceral Transplantation, Clinic for Visceral, Transplant, Thoracic and Vascular Surgery, Leipzig University Medical Center, Leipzig, 04103, Germany
- Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, Leipzig, 04103, Germany
| | - Lena Seidemann
- Department of Hepatobiliary Surgery and Visceral Transplantation, Clinic for Visceral, Transplant, Thoracic and Vascular Surgery, Leipzig University Medical Center, Leipzig, 04103, Germany
- Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, Leipzig, 04103, Germany
- Comprehensive Cancer Center Central Germany (CCCG), Jena and Leipzig, Germany
| | - Mohamed Albadry
- Department of General, Visceral and Vascular Surgery, Experimental Transplantation Surgery, University Hospital Jena, Jena, 07747, Germany
- Department of Pathology, Faculty of Veterinary Medicine, Menoufia University, Shebin Elkom, Menoufia, 6131567, Egypt
| | - Gerda Schicht
- Department of Hepatobiliary Surgery and Visceral Transplantation, Clinic for Visceral, Transplant, Thoracic and Vascular Surgery, Leipzig University Medical Center, Leipzig, 04103, Germany
- Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, Leipzig, 04103, Germany
| | - Marija Skvoznikova
- Department of Hepatobiliary Surgery and Visceral Transplantation, Clinic for Visceral, Transplant, Thoracic and Vascular Surgery, Leipzig University Medical Center, Leipzig, 04103, Germany
- Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, Leipzig, 04103, Germany
| | - Sandra Nickel
- Department of Hepatobiliary Surgery and Visceral Transplantation, Clinic for Visceral, Transplant, Thoracic and Vascular Surgery, Leipzig University Medical Center, Leipzig, 04103, Germany
- Department of General, Visceral and Vascular Surgery, University Hospital Jena, Jena, 07747, Germany
- Comprehensive Cancer Center Central Germany (CCCG), Jena and Leipzig, Germany
| | - René Hänsel
- Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, Leipzig, 04103, Germany
- Institute for Medical Informatics, Statistics and Epidemiology (IMISE), Leipzig University, Leipzig, 04107, Germany
| | - Daniel Seehofer
- Department of Hepatobiliary Surgery and Visceral Transplantation, Clinic for Visceral, Transplant, Thoracic and Vascular Surgery, Leipzig University Medical Center, Leipzig, 04103, Germany
- Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, Leipzig, 04103, Germany
- Comprehensive Cancer Center Central Germany (CCCG), Jena and Leipzig, Germany
| | | | - Hans-Michael Tautenhahn
- Department of Hepatobiliary Surgery and Visceral Transplantation, Clinic for Visceral, Transplant, Thoracic and Vascular Surgery, Leipzig University Medical Center, Leipzig, 04103, Germany
- Department of General, Visceral and Vascular Surgery, Experimental Transplantation Surgery, University Hospital Jena, Jena, 07747, Germany
- Department of General, Visceral and Vascular Surgery, University Hospital Jena, Jena, 07747, Germany
- Comprehensive Cancer Center Central Germany (CCCG), Jena and Leipzig, Germany
| | - Uta Dahmen
- Department of General, Visceral and Vascular Surgery, Experimental Transplantation Surgery, University Hospital Jena, Jena, 07747, Germany
| | - Georg Damm
- Department of Hepatobiliary Surgery and Visceral Transplantation, Clinic for Visceral, Transplant, Thoracic and Vascular Surgery, Leipzig University Medical Center, Leipzig, 04103, Germany.
- Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, Leipzig, 04103, Germany.
- Comprehensive Cancer Center Central Germany (CCCG), Jena and Leipzig, Germany.
| |
Collapse
|
4
|
Tao M, Zhang LL, Zhou GH, Wang C, Luo X. Inhibition of metabotropic glutamate receptor-5 alleviates hepatic steatosis by enhancing autophagy via activation of the AMPK signaling pathway. World J Gastroenterol 2025; 31:98852. [PMID: 39991675 PMCID: PMC11755260 DOI: 10.3748/wjg.v31.i7.98852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 12/08/2024] [Accepted: 12/26/2024] [Indexed: 01/20/2025] Open
Abstract
BACKGROUND The global prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) has continued to increase annually. Recent studies have indicated that inhibition of metabotropic glutamate receptor 5 (mGluR5) may alleviate hepatic steatosis. However, the precise mechanism warrants further exploration. AIM To investigate the potential mechanism by which mGluR5 attenuates hepatocyte steatosis in vitro and in vivo. METHODS Free fatty acids (FFAs)-stimulated HepG2 cells were treated with the mGluR5 antagonist MPEP and the mGluR5 agonist CHPG. Oil Red O staining and a triglyceride assay kit were used to evaluate lipid content. Western blot analysis was conducted to detect the expression of the autophagy-associated proteins p62 and LC3-II, as well as the expression of the key signaling molecules AMPK and ULK1, in the treated cells. To further elucidate the contributions of autophagy and AMPK, we used chloroquine (CQ) to inhibit autophagy and compound C (CC) to inhibit AMPK activity. In parallel, wild-type mice and mGluR5 knockout (KO) mice fed a normal chow diet or a high-fat diet (HFD) were used to evaluate the effect of mGluR5 inhibition in vivo. RESULTS mGluR5 inhibition by MPEP attenuated hepatocellular steatosis and increased LC3-II and p62 protein expression. The autophagy inhibitor CQ reversed the effects of MPEP. In addition, MPEP promoted AMPK and ULK1 expression in HepG2 cells exposed to FFAs. MPEP treatment led to the nuclear translocation of transcription factor EB, which is known to promote p62 expression. This effect was negated by the AMPK inhibitor CC. mGluR5 KO mice presented reduced body weight, improved glucose tolerance and reduced hyperlipidemia when fed a HFD. Additionally, the livers of HFD-fed mGluR5 KO mice presented increases in LC3-II and p62. CONCLUSION Our results suggest that mGluR5 inhibition promoted autophagy and reduced hepatocyte steatosis through activation of the AMPK signaling pathway. These findings reveal a new functional mechanism of mGluR5 as a target in the treatment of MASLD.
Collapse
Affiliation(s)
- Min Tao
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Li-Li Zhang
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Guang-Hong Zhou
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Cong Wang
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Xie Luo
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
5
|
Wan K, Li J, Ma L, Chen T, Chen Y, Li Z, Zouboulis CC, Wang GL, Wang J. Camellia saponin modulates oleic acid/linoleic acid-induced lipogenesis in human sebocytes through lipophagy activation. Int J Cosmet Sci 2025. [PMID: 39844373 DOI: 10.1111/ics.13047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 12/26/2024] [Indexed: 01/24/2025]
Abstract
BACKGROUND Oily skin not only threatens people with aesthetic and hygienic discomfort but also confronts them with annoying skin problems. To explore new skin care ingredients from herbal or plant extracts and understand their underlying mechanism for sebum control would assist in the discovery of desirable sebosuppressive agents, though it is still a deserving and challenging task. AIM To explore the effect of Camellia saponin (CS) on modulating the lipogenesis of human sebocytes. Moreover, to explore the underlying mechanism of CS on oleic acid/linoleic acid (OL) mixture stimulated lipid accumulation. METHODS The lipid accumulation model of cells was constructed by OL-induction in vitro. The lipid synthesis in SZ95 sebocytes was detected by Oil Red O, Nile Red and BODIPY staining and the distribution of lipid droplets and autophagosomes were evaluated by transmission electron microscopy (TEM). Fluorescence staining, immunofluorescence and western blot (WB) were used to characterize the spatial localization of lipid droplets (LDs)/autophagosome/lysosome, the levels of LC3 and P62 proteins related to intracellular autophagy, as well as the pH of lysosome. RESULTS CS treatment significantly relieved OL-induced lipid accumulation in SZ95 sebocytes. Furthermore, CS maintained lysosomal acid environment to promote the fusion of autophagosome and lysosome, thus recovering the OL-induced blockage of autophagy flow. We also found that CS activated AMPK, and down-regulated mTOR in SZ95 sebocytes. CONCLUSION CS was able to relieve OL-stimulated sebum accumulation in cultured human SZ95 sebocytes through lipophagy, in which process CS maintained lysosomal acid environment and activated the AMPK/mTOR pathway.
Collapse
Affiliation(s)
- Kaibo Wan
- School of Chemical and Material Engineering, Jiangnan University, Wuxi, China
| | - Jian Li
- School of Chemical and Material Engineering, Jiangnan University, Wuxi, China
| | - Ling Ma
- Adolph Innovation Laboratory, Guangzhou AOGU Cosmetics Co., Ltd., Guangzhou, China
| | - Timson Chen
- Adolph Innovation Laboratory, Guangzhou AOGU Cosmetics Co., Ltd., Guangzhou, China
| | - Ya Chen
- Adolph Innovation Laboratory, Guangzhou AOGU Cosmetics Co., Ltd., Guangzhou, China
| | - Zhizhen Li
- Adolph Innovation Laboratory, Guangzhou AOGU Cosmetics Co., Ltd., Guangzhou, China
| | - Christos C Zouboulis
- Departments of Dermatology, Venereology, Allergology and Immunology, Dessau Staedtisches Klinikum, Brandenburg Medical School Theodore Fontane, Dessau, Germany
| | - Guang-Li Wang
- School of Chemical and Material Engineering, Jiangnan University, Wuxi, China
| | - Jing Wang
- School of Chemical and Material Engineering, Jiangnan University, Wuxi, China
| |
Collapse
|
6
|
Jin S, Li Y, Xia T, Liu Y, Zhang S, Hu H, Chang Q, Yan M. Mechanisms and therapeutic implications of selective autophagy in nonalcoholic fatty liver disease. J Adv Res 2025; 67:317-329. [PMID: 38295876 PMCID: PMC11725165 DOI: 10.1016/j.jare.2024.01.027] [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: 12/03/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/08/2024] Open
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) has become the most common chronic liver disease worldwide, whereas there is no approved drug therapy due to its complexity. Studies are emerging to discuss the role of selective autophagy in the pathogenesis of NAFLD, because the specificity among the features of selective autophagy makes it a crucial process in mitigating hepatocyte damage caused by aberrant accumulation of dysfunctional organelles, for which no other pathway can compensate. AIM OF REVIEW This review aims to summarize the types, functions, and dynamics of selective autophagy that are of particular importance in the initiation and progression of NAFLD. And on this basis, the review outlines the therapeutic strategies against NAFLD, in particular the medications and potential natural products that can modulate selective autophagy in the pathogenesis of this disease. KEY SCIENTIFIC CONCEPTS OF REVIEW The critical roles of lipophagy and mitophagy in the pathogenesis of NAFLD are well established, while reticulophagy and pexophagy are still being identified in this disease due to the insufficient understanding of their molecular details. As gradual blockage of autophagic flux reveals the complexity of NAFLD, studies unraveling the underlying mechanisms have made it possible to successfully treat NAFLD with multiple pharmacological compounds that target associated pathways. Overall, it is convinced that the continued research into selective autophagy occurring in NAFLD will further enhance the understanding of the pathogenesis and uncover novel therapeutic targets.
Collapse
Affiliation(s)
- Suwei Jin
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| | - Yujia Li
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Tianji Xia
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| | - Yongguang Liu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| | - Shanshan Zhang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| | - Hongbo Hu
- College of Food Science and Nutritional Engineering, China Agricultural University, China.
| | - Qi Chang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, China.
| | - Mingzhu Yan
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, China.
| |
Collapse
|
7
|
Amer AE, Ghoneim HA, Abdelaziz RR, Shehatou GSG, Suddek GM. L-carnitine attenuates autophagic flux, apoptosis, and necroptosis in rats with dexamethasone-induced non-alcoholic steatohepatitis. BMC Pharmacol Toxicol 2024; 25:102. [PMID: 39736705 DOI: 10.1186/s40360-024-00820-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 11/27/2024] [Indexed: 01/01/2025] Open
Abstract
BACKGROUND UpToDate, no drugs have been approved to treat nonalcoholic steatohepatitis, the advanced stage of the most prevalent liver disease, non-alcoholic fatty liver disease. The present study was conducted to explore the potential influences of L-carnitine on the pathomechanisms of hepatic injury that mediate progression to non-alcoholic steatohepatitis in dexamethasone-toxified rats. METHODS Male Wistar rats were allocated as follows: dexamethasone group, rats received dexamethasone (8 mg/kg/day, intraperitoneally) for 6 days; DEXA-LCAR300, DEXA-LCAR500, and DEXA-MET groups, rats administered L-carnitine (300 or 500 mg/kg/day, IP) or metformin (500 mg/kg/day, orally) one week prior to dexamethasone injection (8 mg/kg/day, IP) and other six days alongside dexamethasone administration. Two groups of age-matched normal rats received either the drug vehicle (the control group) or the higher dose of L-carnitine (the drug-control group). At the end of the experiment, sets of biochemical, histological, and immunohistochemical examinations were performed. RESULTS L-carnitine (mainly at the dose of 500 mg/kg/day) markedly abolished dexamethasone-induced alterations in glucose tolerance, hepatic histological features, and serum parameters of hepatic function and lipid profile. Moreover, it significantly ameliorated dexamethasone-induced elevations of hepatic oxidative stress, SREBP-1 and p-MLKL protein levels, and nuclear FOXO1, LC3, P62, and caspase-3 immunohistochemical expression. Furthermore, it markedly diminished dexamethasone-induced suppression of hepatic Akt phosphorylation and Bcl2 immunohistochemical expression. The effects of L-carnitine (500 mg/kg/day) were comparable to those of metformin in most assessments and better than its corresponding lower dose. CONCLUSION These findings introduce L-carnitine as a potential protective drug that may mitigate the rate of disease progression in non-alcoholic fatty liver disease patients with early stages or those at the highest risks.
Collapse
Affiliation(s)
- Ahmed E Amer
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, International Coastal Road, Gamasa City, Dakahliya, 35712, Egypt.
| | - Hamdy A Ghoneim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Rania R Abdelaziz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - George S G Shehatou
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, International Coastal Road, Gamasa City, Dakahliya, 35712, Egypt
| | - Ghada M Suddek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| |
Collapse
|
8
|
Giacco A, Petito G, Silvestri E, Scopigno N, Vigliotti M, Mercurio G, de Lange P, Lombardi A, Moreno M, Goglia F, Lanni A, Senese R, Cioffi F. Comparative effects of 3,5-diiodo-L-thyronine and 3,5,3'-triiodo-L-thyronine on mitochondrial damage and cGAS/STING-driven inflammation in liver of hypothyroid rats. Front Endocrinol (Lausanne) 2024; 15:1432819. [PMID: 39301315 PMCID: PMC11410700 DOI: 10.3389/fendo.2024.1432819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 08/07/2024] [Indexed: 09/22/2024] Open
Abstract
Maintaining a well-functioning mitochondrial network through the mitochondria quality control (MQC) mechanisms, including biogenesis, dynamics and mitophagy, is crucial for overall health. Mitochondrial dysfunction caused by oxidative stress and further exacerbated by impaired quality control can trigger inflammation through the release of the damage-associated molecular patterns (mtDAMPs). mtDAMPs act by stimulating the cyclic GMP-AMP synthase (cGAS) stimulator of interferon genes (STING) pathway. Recently, aberrant signalling of the cGAS-STING axis has been recognised to be closely associated with several sterile inflammatory diseases (e.g. non-alcoholic fatty liver disease, obesity). This may fit the pathophysiology of hypothyroidism, an endocrine disorder characterised by the reduction of thyroid hormone production associated with impaired metabolic fluxes, oxidative balance and inflammatory status. Both 3,5,3'-triiodo-L-tyronine (T3) and its derivative 3,5-diiodo-L-thyronine (3,5-T2), are known to mitigate processes targeting mitochondria, albeit the underlying mechanisms are not yet fully understood. Therefore, we used a chemically induced hypothyroidism rat model to investigate the effect of 3,5-T2 or T3 administration on inflammation-related factors (inflammatory cytokines, hepatic cGAS-STING pathway), oxidative stress, antioxidant defence enzymes, mitochondrial DNA (mtDNA) damage, release and repair, and the MQC system in the liver. Hypothyroid rats showed: i) increased oxidative stress, ii) accumulation of mtDNA damage, iii) high levels of circulating cytokines, iv) hepatic activation of cGAS-STING pathways and v) impairment of MQC mechanisms and autophagy. Both iodothyronines restored oxidative balance by enhancing antioxidant defence, preventing mtDNA damage through the activation of mtDNA repair mechanisms (OGG1, APE1, and POLγ) and promoting autophagy progression. Concerning MQC, both iodothyronines stimulated mitophagy and dynamics, with 3,5-T2 activating fusion and T3 modulating both fusion and fission processes. Moreover, only T3 enhanced mitochondrial biogenesis. Notably, 3,5-T2, but not T3, reversed the hypothyroidism-induced activation of the cGAS-STING inflammatory cascade. In addition, it is noteworthy that 3,5-T2 seems more effective than T3 in reducing circulating pro-inflammatory cytokines IL-6 and IL-1B and in stimulating the release of IL-10, a known anti-inflammatory cytokine. These findings reveal novel molecular mechanisms of hepatic signalling pathways involved in hypothyroidism, which could be targeted by natural iodothyronines, particularly 3,5-T2, paving the way for the development of new treatment strategies for inflammatory diseases.
Collapse
Affiliation(s)
- Antonia Giacco
- Department of Science and Technologies, University of Sannio, Benevento, Italy
| | - Giuseppe Petito
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "L. Vanvitelli", Caserta, Italy
| | - Elena Silvestri
- Department of Science and Technologies, University of Sannio, Benevento, Italy
| | - Nicla Scopigno
- Department of Science and Technologies, University of Sannio, Benevento, Italy
| | - Michela Vigliotti
- Department of Science and Technologies, University of Sannio, Benevento, Italy
| | - Giovanna Mercurio
- Department of Science and Technologies, University of Sannio, Benevento, Italy
| | - Pieter de Lange
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "L. Vanvitelli", Caserta, Italy
| | - Assunta Lombardi
- Department of Biology, University of Naples Federico II, Napoli, Italy
| | - Maria Moreno
- Department of Science and Technologies, University of Sannio, Benevento, Italy
| | - Fernando Goglia
- Department of Science and Technologies, University of Sannio, Benevento, Italy
| | - Antonia Lanni
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "L. Vanvitelli", Caserta, Italy
| | - Rosalba Senese
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "L. Vanvitelli", Caserta, Italy
| | - Federica Cioffi
- Department of Science and Technologies, University of Sannio, Benevento, Italy
| |
Collapse
|
9
|
Zheng Y, Zha X, Zhang B, Elsabagh M, Wang H, Wang M, Zhang H. The interaction of ER stress and autophagy in trophoblasts: navigating pregnancy outcome†. Biol Reprod 2024; 111:292-311. [PMID: 38678504 DOI: 10.1093/biolre/ioae066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 04/12/2024] [Accepted: 04/22/2024] [Indexed: 05/01/2024] Open
Abstract
The endoplasmic reticulum is a complex and dynamic organelle that initiates unfolded protein response and endoplasmic reticulum stress in response to the accumulation of unfolded or misfolded proteins within its lumen. Autophagy is a paramount intracellular degradation system that facilitates the transportation of proteins, cytoplasmic components, and organelles to lysosomes for degradation and recycling. Preeclampsia and intrauterine growth retardation are two common complications of pregnancy associated with abnormal trophoblast differentiation and placental dysfunctions and have a major impact on fetal development and maternal health. The intricate interplay between endoplasmic reticulum stress, and autophagy and their impact on pregnancy outcomes, through mediating trophoblast differentiation and placental development, has been highlighted in various reports. Autophagy controls trophoblast regulation through a variety of gene expressions and signaling pathways while excessive endoplasmic reticulum stress triggers downstream apoptotic signaling, culminating in trophoblast apoptosis. This comprehensive review delves into the intricacies of placental development and explores the underlying mechanisms of preeclampsia and intrauterine growth retardation. In addition, this review will elucidate the molecular mechanisms of endoplasmic reticulum stress and autophagy, both individually and in their interplay, in mediating placental development and trophoblast differentiation, particularly highlighting their roles in preeclampsia and intrauterine growth retardation development. This research seeks to the interplay between endoplasmic reticulum stress and impaired autophagy in the placental trophoderm, offering novel insights into their contribution to pregnancy complications.
Collapse
Affiliation(s)
- Yi Zheng
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, People's Repubic of China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, People's Republic of China
| | - Xia Zha
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, People's Repubic of China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, People's Republic of China
| | - Bei Zhang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, People's Repubic of China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, People's Republic of China
| | - Mabrouk Elsabagh
- Department of Animal Production and Technology, Faculty of Agricultural Sciences and Technologies, Niğde Ömer Halisdemir University, Nigde, Turkey
- Department of Nutrition and Clinical Nutrition, Faculty of Veterinary Medicine, Kafrelsheikh University, KafrelSheikh, Egypt
| | - Hongrong Wang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, People's Repubic of China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, People's Republic of China
| | - Mengzhi Wang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, People's Repubic of China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, People's Republic of China
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural Reclamation Science, Shihezi, P. R. China
| | - Hao Zhang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, People's Repubic of China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, People's Republic of China
| |
Collapse
|
10
|
Zhang S, Liu Y, Chai Y, Xing L, Li J. Effects of intermittent cold stimulation on growth performance, meat quality, antioxidant capacity and liver lipid metabolism in broiler chickens. Poult Sci 2024; 103:103442. [PMID: 38262335 PMCID: PMC10835453 DOI: 10.1016/j.psj.2024.103442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/30/2023] [Accepted: 01/04/2024] [Indexed: 01/25/2024] Open
Abstract
Intermittent cold stimulation (ICS) enhances broilers' resistance to cold stress. Nonetheless, further research is needed to investigate the underlying mechanisms that enhance cold stress resistance. A total of 160 one-day-old male Ross 308 broilers were randomly divided into 2 groups (CC and CS5), with the CC group managing temperature according to the standard for broiler growth stages, while the CS5 group were subjected to cold stimulation at a temperature 3℃ lower than the CC group for 5 h, every 2 d from 15 to 35 d. Sampling was conducted at 36 d (36D), 50 d (50D) and after acute cold stress for 24 h (Y24). First, we examined the effects of ICS on broiler growth performance, meat quality, antioxidant capacity, and lipid metabolism. The results demonstrated that ICS enhanced the performance of broilers to a certain degree. Specifically, the average weight gain in the CS5 group was significantly higher than that of the CC group, and the feed conversion ratio significantly decreased compared to CC at 4 W and 6 W (P ≤ 0.05). Compared with the CC group, cold stimulation significantly reduced drip loss, shearing force, and yellowness (a* value) of chicken meat, while significantly increased redness (b* value) (P ≤ 0.05). At Y24, the levels of T-AOC and GSH-PX in the serum of the CS5 group were significantly higher than those of the CC group, while the level of MDA was significantly lower (P ≤ 0.05). The content of TG, FFA, and VLDL in the serum of the CS5 group was significantly elevated, whereas the level of TC and HDL was significantly lower (P ≤ 0.05). In addition, we further explored whether AMPK-mTOR pathway is involved in the regulation of changes in lipid metabolism and the possible regulatory mechanisms downstream of the signaling pathway. The results showed that ICS significantly upregulated the expression levels of AMPK mRNA and protein in the liver of the CS5 group at 36D and Y24, while significantly down-regulating mTOR (P ≤ 0.05). Compared with the CC group, ICS significantly down-regulated the mRNA expression levels of lipid synthesis and endoplasmic reticulum stress-related genes (SREBP1c, FAS, SCD, ACC, GRP78 and PERK) at 36D and Y24, while significantly up-regulating the mRNA expression levels of lipid decomposition and autophagy-related genes (PPAR and LC3) (P ≤ 0.05). In addition, at Y24, the protein expression levels of endoplasmic reticulum stress-related genes (GRP78) in the CS5 group were significantly lower, while autophagy-related genes (LC3 and ATG7) were significantly higher (P ≤ 0.05). ICS can affect meat quality and lipid metabolism in broilers, and when broilers are subjected to acute cold stress, broilers trained with cold stimulation have stronger lipid metabolism capacity.
Collapse
Affiliation(s)
- Shijie Zhang
- College of Life Science, Northeast Agricultural University, Harbin, 150030, China
| | - Yuanyuan Liu
- College of Life Science, Northeast Agricultural University, Harbin, 150030, China
| | - Yiwen Chai
- College of Life Science, Northeast Agricultural University, Harbin, 150030, China
| | - Lu Xing
- College of Life Science, Northeast Agricultural University, Harbin, 150030, China
| | - Jianhong Li
- College of Life Science, Northeast Agricultural University, Harbin, 150030, China; Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, 150030, China.
| |
Collapse
|
11
|
Ren Q, Sun Q, Fu J. Dysfunction of autophagy in high-fat diet-induced non-alcoholic fatty liver disease. Autophagy 2024; 20:221-241. [PMID: 37700498 PMCID: PMC10813589 DOI: 10.1080/15548627.2023.2254191] [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: 05/01/2023] [Accepted: 08/24/2023] [Indexed: 09/14/2023] Open
Abstract
ABBREVIATIONS ACOX1: acyl-CoA oxidase 1; ADH5: alcohol dehydrogenase 5 (class III), chi polypeptide; ADIPOQ: adiponectin, C1Q and collagen domain containing; ATG: autophagy related; BECN1: beclin 1; CRTC2: CREB regulated transcription coactivator 2; ER: endoplasmic reticulum; F2RL1: F2R like trypsin receptor 1; FA: fatty acid; FOXO1: forkhead box O1; GLP1R: glucagon like peptide 1 receptor; GRK2: G protein-coupled receptor kinase 2; GTPase: guanosine triphosphatase; HFD: high-fat diet; HSCs: hepatic stellate cells; HTRA2: HtrA serine peptidase 2; IRGM: immunity related GTPase M; KD: knockdown; KDM6B: lysine demethylase 6B; KO: knockout; LAMP2: lysosomal associated membrane protein 2; LAP: LC3-associated phagocytosis; LDs: lipid droplets; Li KO: liver-specific knockout; LSECs: liver sinusoidal endothelial cells; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MAP3K5: mitogen-activated protein kinase kinase kinase 5; MED1: mediator complex subunit 1; MTOR: mechanistic target of rapamycin kinase; MTORC1: mechanistic target of rapamycin complex 1; NAFLD: non-alcoholic fatty liver disease; NASH: non-alcoholic steatohepatitis; NFE2L2: NFE2 like bZIP transcription factor 2; NOS3: nitric oxide synthase 3; NR1H3: nuclear receptor subfamily 1 group H member 3; OA: oleic acid; OE: overexpression; OSBPL8: oxysterol binding protein like 8; PA: palmitic acid; RUBCNL: rubicon like autophagy enhancer; PLIN2: perilipin 2; PLIN3: perilipin 3; PPARA: peroxisome proliferator activated receptor alpha; PRKAA2/AMPK: protein kinase AMP-activated catalytic subunit alpha 2; RAB: member RAS oncogene family; RPTOR: regulatory associated protein of MTOR complex 1; SCD: stearoyl-CoA desaturase; SIRT1: sirtuin 1; SIRT3: sirtuin 3; SNARE: soluble N-ethylmaleimide-sensitive factor attachment protein receptor; SQSTM1/p62: sequestosome 1; SREBF1: sterol regulatory element binding transcription factor 1;SREBF2: sterol regulatory element binding transcription factor 2; STING1: stimulator of interferon response cGAMP interactor 1; STX17: syntaxin 17; TAGs: triacylglycerols; TFEB: transcription factor EB; TP53/p53: tumor protein p53; ULK1: unc-51 like autophagy activating kinase 1; VMP1: vacuole membrane protein 1.
Collapse
Affiliation(s)
- Qiannan Ren
- Department of Endocrinology, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Qiming Sun
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang, China
- Department of Biochemistry, and Department of Cardiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Junfen Fu
- Department of Endocrinology, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| |
Collapse
|
12
|
Amer AE, Ghoneim HA, Abdelaziz RR, Shehatou GSG, Suddek GM. Saroglitazar mitigated NASH-associated hepatic injury in dexamethasone-treated rats via modulating autophagy, apoptosis, and necroptosis. Toxicol Appl Pharmacol 2024; 482:116774. [PMID: 38040297 DOI: 10.1016/j.taap.2023.116774] [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: 08/28/2023] [Revised: 11/23/2023] [Accepted: 11/25/2023] [Indexed: 12/03/2023]
Abstract
This study aimed to evaluate the possible ameliorative effects of saroglitazar (SAR) on aspects of hepatic injury in dexamethasone (DEX)-induced nonalcoholic steatohepatitis (NASH) in rats. Wistar rats received SAR (2 or 4 mg/kg/day, orally) or metformin (MET, 500 mg/kg/day, orally) for one week before and concurrently with DEX administration (8 mg/kg/day, i.p., for 6 days. Control and drug control groups received vehicle or the higher dose of SAR, respectively. At the end of the experiment, an oral glucose tolerance test (OGTT) was conducted, serum hepatic function parameters and lipid profile were assessed, and hepatic histological changes were evaluated. Moreover, hepatic p-Akt/Akt ratios, malondialdehyde (MDA) content, SREBP-1, FOXO1, LC3, cleaved caspase-3, and p-MLKL protein levels were determined. Furthermore, hepatic immunohistochemical expressions of FOXO1, caspase-3, Bcl-2, LC3, and P62 were examined. SAR (mainly at 4 mg/kg/day) significantly improved Area under the OGTT curve (P < 0.0001), hepatic function parameters, lipid profile, and hepatic histopathological features in DEX-administered rats. Moreover, SAR significantly attenuated DEX-induced increases in hepatic MDA content (P < 0.05), SREBP-1 levels (P < 0.0001), and nuclear FOXO1, caspase-3, LC3, P62, and p-MLKL protein expressions (P < 0.0001). Furthermore, SAR significantly enhanced hepatic p-Akt/Akt ratio and Bcl-2 protein expression in DEX-administered rats (P < 0.0001). The higher dose of SAR showed greater hepatoprotective effects compared to its corresponding lower dose and MET in most assessments, approaching levels similar to the control group. SAR mitigated hepatic injury associated with DEX-induced NASH in rats, suggesting it might be a potential hepatoprotective drug for patients with or at high risk of NASH.
Collapse
Affiliation(s)
- Ahmed E Amer
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, International Coastal Road, Gamasa City, Dakahliya 11152, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Hamdy A Ghoneim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Rania R Abdelaziz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - George S G Shehatou
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, International Coastal Road, Gamasa City, Dakahliya 11152, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Ghada M Suddek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| |
Collapse
|
13
|
Kaul NL, Diebolt CM, Meier C, Tschernig T. Transient receptor potential channel 3 in human liver and gallbladder - An investigation in body donors. Ann Anat 2023; 250:152150. [PMID: 37633502 DOI: 10.1016/j.aanat.2023.152150] [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: 06/06/2023] [Revised: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 08/28/2023]
Abstract
Since the discovery of TRP proteins in 1969, during studies of the fruit fly Drosophila melanogaster, interest around them and the subfamily of TRPC channels has remained high. TRPC3 was able to be detected in a number of organs in rodents, such as rats and mice, and also in various human tissues. For the most part, these investigations were carried out using gene expression of TRPC3. Further work has already confirmed the relevance of TRPC3 in the context of neurodegenerative diseases, such as spinocerebellar ataxia, and carcinogenic entities, such as ovarian carcinoma. An association with TRPC3 has also been demonstrated for diseases that affect the liver. In order to confirm the expression of TRPC3 in the human liver, this study uses samples taken from eight (n = 8) fixated human body donors and analyzed with immunohistochemistry. In accordance with the macroscopic anatomy of the organs, six samples (n = 6) of liver tissue and three (n = 3) of gallbladder tissue were obtained. TRPC3 was clearly detected in all liver and gallbladder samples examined. Thus, it is not unlikely that TRPC3 plays a role in the extensive metabolic processes of the liver and could also serve as a target for pharmacological interventions in an imbalance of calcium homeostasis.
Collapse
Affiliation(s)
- Nele Leonie Kaul
- Institute of Anatomy and Cell Biology, Saarland University, Medical Campus, Homburg, Saar, Germany
| | - Coline M Diebolt
- Institute of Anatomy and Cell Biology, Saarland University, Medical Campus, Homburg, Saar, Germany
| | - Carola Meier
- Institute of Anatomy and Cell Biology, Saarland University, Medical Campus, Homburg, Saar, Germany
| | - Thomas Tschernig
- Institute of Anatomy and Cell Biology, Saarland University, Medical Campus, Homburg, Saar, Germany.
| |
Collapse
|
14
|
Wikan N, Tocharus J, Oka C, Sivasinprasasn S, Chaichompoo W, Denlumpai P, Suksamrarn A, Tocharus C. Pelargonic acid vanillylamide alleviates hepatic autophagy and ER stress in hepatic steatosis model. Food Chem Toxicol 2023; 180:113987. [PMID: 37611858 DOI: 10.1016/j.fct.2023.113987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 07/24/2023] [Accepted: 08/11/2023] [Indexed: 08/25/2023]
Abstract
Pelargonic acid vanillylamide (PAVA) has been shown to reduce hepatic lipid accumulation in an obese rat model, however the underlying mechanism responsible for regulating lipid metabolism remains unclear. This study investigated the molecular mechanisms invoked by PAVA in regulating lipogenesis, autophagy, and endoplasmic reticulum (ER) stress in obese rats. Male Sprague-Dawley rats were fed on a diet consisting of 65.26% fat (16 weeks) and HepG2 cells were incubated with 200 μM oleic acid (OA) plus 100 μM palmitic acid (PA) for 48 h. These treatments resulted in a steatosis model. PAVA was shown to reduce fat deposition in hepatocytes in HepG2 by reducing lipotoxicity, the triglyceride content, the expression of sterol regulatory element binding protein 1c (SREBP-1c) and fatty acid synthase (FASN). PAVA also significantly reduced the calcium level and the expression of calpain 2 and upregulated the expression of Atg7 in comparison to the HFD group. In addition, PAVA was shown to significantly decrease the expression of autophagy pathway-related proteins including LC3 and p62. Treatment with PAVA (1 mg/day) reduced the expressions of ER stress markers Bip, ATF6 (p50), p-IRE1/IRE1, p-eIF2α/eIF2α, pJNK, CHOP and cleaved CASP12. In conclusion, PAVA ameliorated obesity induced hepatic steatosis by attenuating defective autophagy and ER stress pathways.
Collapse
Affiliation(s)
- Naruemon Wikan
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Jiraporn Tocharus
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Chio Oka
- Laboratory of Functional Genomics and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Nara, Japan
| | | | - Waraluck Chaichompoo
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Ramkhamhaeng University, Bangkok, Thailand
| | - Panida Denlumpai
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Ramkhamhaeng University, Bangkok, Thailand
| | - Apichart Suksamrarn
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Ramkhamhaeng University, Bangkok, Thailand
| | - Chainarong Tocharus
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Radiation Research and Medical Imaging, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
15
|
Liu X, Li X, Su S, Yuan Y, Liu W, Zhu M, Zheng Q, Zeng X, Fu F, Lu Y, Chen Y. Oleic acid improves hepatic lipotoxicity injury by alleviating autophagy dysfunction. Exp Cell Res 2023:113655. [PMID: 37253404 DOI: 10.1016/j.yexcr.2023.113655] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 05/19/2023] [Accepted: 05/21/2023] [Indexed: 06/01/2023]
Abstract
Lipotoxicity caused by excess free fatty acids, particularly saturated fatty acids (SFAs) such as palmitic acid (PA), is one of the most important pathogenesis of nonalcoholic fatty liver disease (NAFLD). However, unsaturated fatty acids (UFAs), such as oleic acid (OA), are nontoxic and can combat SFA-induced toxicity through alleviation of cell apoptosis, endoplasmic reticulum stress (ER stress) and lipids metabolism disorder. However, whether OA is able to regulate autophagy is largely unknown. So, this study aims to investigate the mechanism underlying OA mediated modulation of autophagy in hepatocytes and mice with NAFLD. In vitro, human hepatoma cell line HepG2 cells, human normal liver cells L-02 and mouse normal liver cells AML12 were treated with palmitic acid (PA)/tunicamycin (TM) or/and OA for 48 h. In vivo, C57/BL6 mice were fed with high fat diet (HFD) to induce NAFLD. And the HFD was partial replaced by olive oil to observe the protective effects of olive oil. We demonstrated that PA/TM impaired cell viability and induced cellular apoptosis in HepG2 cells and L-02 cells. Moreover, PA/TM induced autophagy impairment by reducing the nuclear translocation of transcription factor EB (TFEB) and inhibiting the activity of CTSB. However, OA substantially alleviated PA/TM induced cellular apoptosis and autophagy dysfunction in hepatocytes. Additionally, restoring autophagy function is able to reduce ER stress. Similarly, HFD for 20 weeks successfully established NAFLD model in C57/BL6 mice, and significant autophagy impairment were observed in liver tissues. Noteworthily, 30% replacement of HFD with olive oil had profoundly reversed NAFLD. It significantly impoved steatosis, and reduced autophagy dysfunction, ER stress and apoptosis in liver tissue. Conclusively, these data demonstrated that OA is able to effectively impove autophagy dysfunction under the context of both PA and ER stress inducer induced lipotoxicity, and OA mediated regulation of lysosome dysfunction through TFEB plays an important role, suggesting that the regulation of ER stress-autophagy axis is a critical mechanism in OA driven protection in NAFLD.
Collapse
Affiliation(s)
- Xiaohong Liu
- Department of Clinical Nutrition and Key Laboratory of Transplant Engineering and Immunology, NHFPC, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, PR China
| | - Xiaoyu Li
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, PR China
| | - Shan Su
- Department of Clinical Nutrition and Key Laboratory of Transplant Engineering and Immunology, NHFPC, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, PR China
| | - Yujia Yuan
- Department of Clinical Nutrition and Key Laboratory of Transplant Engineering and Immunology, NHFPC, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, PR China
| | - Wen Liu
- Department of Clinical Nutrition and Key Laboratory of Transplant Engineering and Immunology, NHFPC, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, PR China
| | - Min Zhu
- Department of Clinical Nutrition and Key Laboratory of Transplant Engineering and Immunology, NHFPC, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, PR China
| | - Qing Zheng
- Department of Clinical Nutrition and Key Laboratory of Transplant Engineering and Immunology, NHFPC, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, PR China
| | - Xin Zeng
- Department of Clinical Nutrition and Key Laboratory of Transplant Engineering and Immunology, NHFPC, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, PR China
| | - Fudong Fu
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, PR China
| | - Yanrong Lu
- Department of Clinical Nutrition and Key Laboratory of Transplant Engineering and Immunology, NHFPC, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, PR China
| | - Younan Chen
- Department of Clinical Nutrition and Key Laboratory of Transplant Engineering and Immunology, NHFPC, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, PR China; Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, PR China.
| |
Collapse
|
16
|
Zuo Y, Chai Y, Liu X, Gao Z, Jin X, Wang F, Bai Y, Zheng Z. A ratiometric fluorescent probe based on spiropyran in situ switching for tracking dynamic changes of lysosomal autophagy and anticounterfeiting. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2023; 291:122338. [PMID: 36657288 DOI: 10.1016/j.saa.2023.122338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/28/2022] [Accepted: 01/06/2023] [Indexed: 06/17/2023]
Abstract
Autophagy is the controlled breakdown of cellular components that dysfunctional or nonessential, and the decomposition products are further recycled and synthesized for the normal physiological activities of cells. Lysosomal autophagy has been implicated in cancer, neurological disorders, Parkinson's disease, etc. Therefore, it is necessary to develop a fluorescent probe that can clearly describe the process of lysosomal autophagy. However, there are currently limited fluorescent probes for ratiometric monitoring of the autophagic process in dual channels. To solve this problem, a fluorescent probe based on spiropyran with lysosomal targeting and pH response for ratiometric monitoring the autophagy process of lysosomes were designed. The sensitive response of the probe to pH in vitro was verified by UV and fluorescence spectrum tests. Meanwhile, the probe demonstrated the ability to monitor the intracellular pH fluctuations. In addition, the application of Lyso-SD in the field of anti-counterfeiting has been proposed based on the obvious photoluminescence ability of Lyso-SD under UV irradiation.
Collapse
Affiliation(s)
- Yujing Zuo
- Tianjin Key Laboratory of Composite & Functional Materials, School of Materials Science and Engineering, Tianjin University, Tianjin 300072, China; Ningbo Yinzhou Chinaust Automobile Fittings Corp. Ltd., Ningbo 315142, China
| | - Yanfu Chai
- Tianjin Key Laboratory of Composite & Functional Materials, School of Materials Science and Engineering, Tianjin University, Tianjin 300072, China; School of Mechanical and Electrical Engineering, Shaoxing University, Shaoxing 312000, China; Ningbo Yinzhou Chinaust Automobile Fittings Corp. Ltd., Ningbo 315142, China.
| | - Xiaofei Liu
- Tianjin Key Laboratory of Composite & Functional Materials, School of Materials Science and Engineering, Tianjin University, Tianjin 300072, China
| | - Zhiming Gao
- Tianjin Key Laboratory of Composite & Functional Materials, School of Materials Science and Engineering, Tianjin University, Tianjin 300072, China
| | - Xiaofeng Jin
- Ningbo Yinzhou Chinaust Automobile Fittings Corp. Ltd., Ningbo 315142, China
| | - Feng Wang
- Ningbo Yinzhou Chinaust Automobile Fittings Corp. Ltd., Ningbo 315142, China
| | - Yongjie Bai
- Ningbo Yinzhou Chinaust Automobile Fittings Corp. Ltd., Ningbo 315142, China
| | - Zhijun Zheng
- Ningbo Yinzhou Chinaust Automobile Fittings Corp. Ltd., Ningbo 315142, China
| |
Collapse
|
17
|
Wang X, Cong P, Wang X, Wang Z, Liu B, Xue C, Xu J. Docosahexaenoic acid-acylated astaxanthin monoester ameliorates chronic high-fat diet-induced autophagy dysfunction via ULK1 pathway in the hypothalamus of mice. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2023; 103:2378-2388. [PMID: 36606564 DOI: 10.1002/jsfa.12429] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 11/21/2022] [Accepted: 01/06/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND Dietary astaxanthin (AST) exhibits the ability to resist lipid accumulation and stimulate hepatic autophagy. Natural AST predominantly exists in stable esterified forms. More importantly, in our previous study, docosahexaenoic acid-acylated AST monoester (AST-DHA) possessed better stability, bioavailability, and neuroprotective ability than AST in free and diester form. However, the AST-DHA mechanisms of action in regulating the obese phenotype and autophagy of the central nervous system remain unclear. RESULTS High-fat diet (HFD)-fed C57BL/6J mice were orally administered AST-DHA (50 mg/kg body weight/d) for 3 days or 8 weeks. AST-DHA supplementation alleviated HFD-induced abnormal body weight gain, significantly enhanced autophagy with an increased microtubule-associated protein light chain 3 II/I (LC3II/I) ratio, and reduced the accumulation of p62/sequestosome 1 (SQSTM1) in the hypothalamus rather than in the hippocampus. Mechanistically, AST-DHA effectively promoted autophagy and autophagosome formation, and most notably rescued the HFD-impaired autophagosome-lysosome fusion (indicated by the colocalization of LC3 and LAMP1) by regulating mTOR- and AMPK-induced phosphorylation of ULK1. Consequently, AST-DHA enhanced hypothalamic autophagy, leading to pro-opiomelanocortin (POMC) cleavage to produce alpha-melanocyte-stimulating hormone (α-MSH). CONCLUSIONS This study identified AST-DHA as an enhancer of autophagy that plays a beneficial role in restoring hypothalamic autophagy, and as a new potential therapeutic agent against HFD-induced obesity. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Xiaoxu Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
- College of Marine Life Sciences, Ocean University of China, Qingdao, China
| | - Peixu Cong
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Xincen Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Zhigao Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Bin Liu
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Changhu Xue
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
- Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Jie Xu
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| |
Collapse
|
18
|
Fatty Acids as Potent Modulators of Autophagy Activity in White Adipose Tissue. Biomolecules 2023; 13:biom13020255. [PMID: 36830623 PMCID: PMC9953325 DOI: 10.3390/biom13020255] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
A high-fat diet is one of the causative factors of obesity. The dietary profile of fatty acids is also an important variable in developing obesity, as saturated fatty acids are more obesogenic than monounsaturated and polyunsaturated fatty acids. Overweight and obesity are inseparably connected with the excess of adipose tissue in the body, characterized by hypertrophy and hyperplasia of fat cells, which increases the risk of developing metabolic syndrome. Changes observed within hypertrophic adipocytes result in elevated oxidative stress, unfolded protein accumulation, and increased endoplasmic reticulum (ER) stress. One of the processes involved in preservation of cellular homeostasis is autophagy, which is defined as an intracellular lysosome-dependent degradation system that serves to recycle available macromolecules and eliminate damaged organelles. In obesity, activation of autophagy is increased and the process appears to be regulated by different types of dietary fatty acids. This review describes the role of autophagy in adipose tissue and summarizes the current understanding of the effects of saturated and unsaturated fatty acids in autophagy modulation in adipocytes.
Collapse
|
19
|
Guo J, Nie J, Chen Z, Wang X, Hu H, Xu J, Lu J, Ma L, Ji H, Yuan J, Xu B. Cold exposure-induced endoplasmic reticulum stress regulates autophagy through the SIRT2/FoxO1 signaling pathway. J Cell Physiol 2022; 237:3960-3970. [PMID: 35938526 DOI: 10.1002/jcp.30856] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 07/21/2022] [Accepted: 07/27/2022] [Indexed: 01/14/2023]
Abstract
Cold is a factor affecting health in humans and animals. The liver, a major metabolic center, is highly susceptible to ambient air temperature. Recent studies have shown that endoplasmic reticulum (ER) stress is associated with the liver, and regulates the occurrence and development of liver injury and autophagy. However, the mechanism underlying the relationship between cold exposure and ER stress in the liver is not well understood. In this study, we investigated the effect of ER stress on liver autophagy and its mechanism under cold exposure. AML12 cells were treated with Tg to construct an ER stress model, and the level of autophagy increased. To further explore the mechanism through which ER stress regulates autophagy, we knocked down SIRT2 with shRNA in Tg-treated AML12 cells. Knockdown of SIRT2 significantly increased ER stress and autophagy, increased FoxO1 acetylation, and promoted its entry into the nucleus. To further verify the results of in vitro experiments, we exposed mice to 4°C for 3 h per day for 3 weeks to exacerbate the burden on the liver after cold exposure. Cold exposure damaged the structure and function of the liver and promoted the inflammatory response. It also activated ER stress and promoted autophagy. In addition, cold exposure inhibited the expression of SIRT2, promoted FoxO1 acetylation, and enhanced the interaction with autophagy. Our findings indicated that cold exposure induces liver damage, ER stress, and autophagy through the SIRT2/FoxO1 pathway. These findings suggest that SIRT2 may be a potential target for regulating health under cold exposure.
Collapse
Affiliation(s)
- Jingru Guo
- National Experimental Teaching Demonstration Center of Animal Medicine Foundation, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Junshu Nie
- National Experimental Teaching Demonstration Center of Animal Medicine Foundation, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Zhuo Chen
- National Experimental Teaching Demonstration Center of Animal Medicine Foundation, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Xian Wang
- National Experimental Teaching Demonstration Center of Animal Medicine Foundation, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Huijie Hu
- National Experimental Teaching Demonstration Center of Animal Medicine Foundation, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Jing Xu
- National Experimental Teaching Demonstration Center of Animal Medicine Foundation, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Jingjing Lu
- National Experimental Teaching Demonstration Center of Animal Medicine Foundation, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Li Ma
- National Experimental Teaching Demonstration Center of Animal Medicine Foundation, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Hong Ji
- National Experimental Teaching Demonstration Center of Animal Medicine Foundation, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Jianbin Yuan
- National Experimental Teaching Demonstration Center of Animal Medicine Foundation, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Bin Xu
- National Experimental Teaching Demonstration Center of Animal Medicine Foundation, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| |
Collapse
|
20
|
Morishita H, Komatsu M. Role of autophagy in liver diseases. CURRENT OPINION IN PHYSIOLOGY 2022. [DOI: 10.1016/j.cophys.2022.100594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
21
|
Wang Y, Wang M, Liu Y, Tao H, Banerjee S, Srinivasan S, Nemeth E, Czaja MJ, He P. Integrated regulation of stress responses, autophagy and survival by altered intracellular iron stores. Redox Biol 2022; 55:102407. [PMID: 35853304 PMCID: PMC9294649 DOI: 10.1016/j.redox.2022.102407] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/07/2022] [Accepted: 07/11/2022] [Indexed: 02/06/2023] Open
Abstract
Iron is a mineral essential for blood production and a variety of critical cellular functions. Altered iron metabolism has been increasingly observed in many diseases and disorders, but a comprehensive and mechanistic understanding of the cellular impact of impaired iron metabolism is still lacking. We examined the effects of iron overload or iron deficiency on cellular stress responses and autophagy which collectively regulate cell homeostasis and survival. Acute iron loading led to increased mitochondrial ROS (mtROS) production and damage, lipid peroxidation, impaired autophagic flux, and ferroptosis. Iron-induced mtROS overproduction is the mechanism of increased lipid peroxidation, impaired autophagy, and the induction of ferroptosis. Iron excess-induced ferroptosis was cell-type dependent and regulated by activating transcription factor 4 (ATF4). Upregulation of ATF4 mitigated iron-induced autophagic dysfunction and ferroptosis, whereas silencing of ATF4 expression impaired autophagy and resulted in increased mtROS production and ferroptosis. Employing autophagy-deficient hepatocytes and different autophagy inhibitors, we further showed that autophagic impairment sensitized cells to iron-induced ferroptosis. In contrast, iron deficiency activated the endoplasmic reticulum (ER) stress response, decreased autophagy, and induced apoptosis. Decreased autophagy associated with iron deficiency was due to ER stress, as reduction of ER stress by 4-phenylbutyric acid (4-PBA) improved autophagic flux. The mechanism of decreased autophagy in iron deficiency is a disruption in lysosomal biogenesis due to impaired posttranslational maturation of lysosomal membrane proteins. In conclusion, iron excess and iron deficiency cause different forms of cell stress and death in part through the common mechanism of impaired autophagic function.
Collapse
Affiliation(s)
- Yunyang Wang
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Mo Wang
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Yunshan Liu
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Hui Tao
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Somesh Banerjee
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Shanthi Srinivasan
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA; Gastroenterology Research, Atlanta VA Health Care System, Decatur, GA, USA
| | - Elizabeta Nemeth
- Department of Medicine, Center for Iron Disorders, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Mark J Czaja
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Peijian He
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
22
|
Chen X, Zhang L, Zheng L, Tuo B. Role of Ca 2+ channels in non-alcoholic fatty liver disease and their implications for therapeutic strategies (Review). Int J Mol Med 2022; 50:113. [PMID: 35796003 PMCID: PMC9282635 DOI: 10.3892/ijmm.2022.5169] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 06/07/2022] [Indexed: 01/10/2023] Open
Affiliation(s)
- Xingyue Chen
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Li Zhang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Liming Zheng
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Biguang Tuo
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| |
Collapse
|
23
|
Yue K, Pu X, Loor JJ, Jiang Q, Dong J, Shen T, Li G, Gao W, Lei L, Du X, Song Y, Liu G, Li X. Impaired autophagy aggravates oxidative stress in mammary gland of dairy cows with clinical ketosis. J Dairy Sci 2022; 105:6030-6040. [PMID: 35637003 DOI: 10.3168/jds.2021-21234] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 03/21/2022] [Indexed: 01/03/2025]
Abstract
When ketosis occurs, supraphysiological levels of free fatty acids (FFA) can cause oxidative injury to the mammary gland and autophagy can regulate the cellular oxidative status. The aim of this study was to investigate the autophagy status of mammary tissue and its associations with oxidative stress in healthy and clinically ketotic dairy cows. Mammary tissue and blood samples were collected from healthy cows [n = 15, β-hydroxybutyrate (BHB) <0.6 mM] and clinically ketotic cows (n = 15, BHB >3.0 mM) at 3 to 15 (average = 7) days in milk. For in vitro study, bovine mammary epithelial cells (BMEC) isolated from healthy cows were treated with 0, 0.3, 0.6, or 1.2 mM FFA for 24 h. Furthermore, BMEC were pretreated with 100 nM rapamycin, an autophagy activator, for 4 h or 50 mM 3-methyladenine (3-MA), an autophagy inhibitor, for 1 h, followed by treatment with or without FFA (1.2 mM) for another 24 h. Oxidation indicators and autophagy-related protein abundance were measured. Compared with healthy cows, serum concentrations of FFA, BHB, and malondialdehyde were greater in clinically ketotic cows, but milk production (kg/d), milk protein (kg/d), activities of superoxide dismutase, catalase, and glutathione peroxidase were lower. Abundances of mRNA and protein of autophagy-related gene 5 (ATG5) and 7 (ATG7) were lower, but sequestosome-1 (SQSTM1, also called p62) greater in mammary tissue of clinically ketotic cows. The mRNA abundance of microtubule-associated protein 1 light chain 3 (MAP1LC3, also called LC3) and protein abundance of LC3-II were lower in mammary tissue of clinically ketotic cows. In vitro, exogenous FFA increased the content of malondialdehyde and reactive oxygen species, but decreased the activities of superoxide dismutase, catalase, and plasma glutathione peroxidase. Compared with the 0 mM FFA group, abundance of ATG5, ATG7, LC3-II was greater, but p62 was lower in the 0.6 mM FFA-treated cells. Similarly, abundance of ATG5, ATG7, and LC3-II was lower, but p62 greater in the 1.2 mM FFA-treated cells relative to 0 mM FFA group. Culture with rapamycin alleviated oxidative stress induced by 1.2 mM FFA, whereas 3-MA aggravated it. Overall, results indicated that a low concentration (0.6 mM) of FFA can induce oxidative stress and activate autophagy in BMEC. At higher concentrations of FFA (1.2 mM), autophagy is impaired and oxidative stress is aggravated. Autophagy is a mechanism for BMEC to counteract FFA-induced stress. As such, it could serve as a potential target for further development of novel strategies against oxidative stress.
Collapse
Affiliation(s)
- Kaiming Yue
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, 130062, Jilin, China
| | - Xudong Pu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, 130062, Jilin, China
| | - Juan J Loor
- Mammalian NutriPhysioGenomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - Qianming Jiang
- Mammalian NutriPhysioGenomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - Jihong Dong
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, 130062, Jilin, China
| | - Taiyu Shen
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, 130062, Jilin, China
| | - Guojin Li
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, 130062, Jilin, China
| | - Wenwen Gao
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, 130062, Jilin, China
| | - Lin Lei
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, 130062, Jilin, China
| | - Xiliang Du
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, 130062, Jilin, China
| | - Yuxiang Song
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, 130062, Jilin, China
| | - Guowen Liu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, 130062, Jilin, China
| | - Xinwei Li
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, 130062, Jilin, China.
| |
Collapse
|
24
|
Ji S, Sun J, Bian C, Huang X, Ji H. PKA/ATGL signaling pathway is involved in ER stress-mediated lipolysis in adipocytes of grass carp (Ctenopharyngodon idella). FISH PHYSIOLOGY AND BIOCHEMISTRY 2022; 48:683-691. [PMID: 35460470 DOI: 10.1007/s10695-021-01032-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 11/13/2021] [Indexed: 06/14/2023]
Abstract
The relationship between endoplasmic reticulum stress (ER stress) and lipolysis in mammals has been widely studied, but it is relatively scarce in fish. The present study used grass carp Ctenopharyngodon idella as a model to investigate the effect of ER stress on lipolysis in adipocytes of fish. We found that ER stress evoked by tunicamycin (TM) treatment significantly induced lipolysis in adipocytes. Subsequently, in order to further investigate whether protein kinase A (PKA) is involved in ER stress-induced lipolysis, we treated adipocytes with PKA activator forskolin and inhibitor H89. The results showed that the mechanism was related to the activation of PKA, especially the catalytic subunit PRKACBa. Notably, we also found that PKA regulates lipolysis by targeting mRNA level and protein and enzyme activities of adipotriglyceride lipase (ATGL). Taken together, our findings suggest that PKA/ATGL signaling pathway is involved in ER stress-mediated lipolysis of grass carp adipocytes. It provides a theoretical basis for further study on the mechanism of lipolysis in fish and other vertebrates.
Collapse
Affiliation(s)
- Shanghong Ji
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, 712100, China
| | - Jian Sun
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, 712100, China
| | - Chenchen Bian
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, 712100, China
| | - Xiaocheng Huang
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, 712100, China
| | - Hong Ji
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, 712100, China.
| |
Collapse
|
25
|
Septin 9 and phosphoinositides regulate lysosome localization and their association with lipid droplets. iScience 2022; 25:104288. [PMID: 35573204 PMCID: PMC9097704 DOI: 10.1016/j.isci.2022.104288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 12/21/2021] [Accepted: 04/20/2022] [Indexed: 11/23/2022] Open
Abstract
The accumulation of lipid droplets (LDs) in the liver is a hallmark of steatosis, which is often associated with lysosomal dysfunction. Nevertheless, the underlying mechanisms remain unclear. Here, using Huh7 cells loaded with oleate as a model to study LD metabolism, we show that cellular content and distribution of LDs are correlated with those of the lysosome and regulated by oleate and septin 9. High expression of septin 9 promotes perinuclear clustering of lysosomes which co-localized with Golgi and not with their surrounding LDs. On the other hand, knockdown of septin 9 disperses the two organelles which colocalize at the cell periphery. The Rab7 is present around these peripheral LDs. PtdIns5P which binds septin 9 and MTMR3 which converts PtdIns(3,5)P2 into PtdIns(5) recapitulates the effects of septin 9. By contrast, PtdIns(3,5)P2 promotes LD/lysosome co-localization. Overall, our data reveal a phosphoinositide/septin 9-dependent mechanism that regulates LD behavior through the control of their association with lysosomes.
Septin 9 is regulates oleate-induced lysosome perinuclear clustering Septin 9 and MTs regulate oleate-induced lysosome co-localization with Golgi LDs with high septin 9 have less interaction with Rab7 and LAMP1 PIs have specific effects on LD and lysosome
Collapse
|
26
|
Restoration of ER proteostasis attenuates remote apoptotic cell death after spinal cord injury by reducing autophagosome overload. Cell Death Dis 2022; 13:381. [PMID: 35444186 PMCID: PMC9021197 DOI: 10.1038/s41419-022-04830-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 03/30/2022] [Accepted: 04/04/2022] [Indexed: 02/07/2023]
Abstract
The pathogenic mechanisms that underlie the progression of remote degeneration after spinal cord injury (SCI) are not fully understood. In this study, we examined the relationship between endoplasmic reticulum (ER) stress and macroautophagy, hereafter autophagy, and its contribution to the secondary damage and outcomes that are associated with remote degeneration after SCI. Using a rat model of spinal cord hemisection at the cervical level, we measured ER stress and autophagy markers in the axotomized neurons of the red nucleus (RN). In SCI animals, mRNA and protein levels of markers of ER stress, such as GRP78, CHOP, and GADD34, increased 1 day after the injury, peaking on Day 5. Notably, in SCI animals, the increase of ER stress markers correlated with a blockade in autophagic flux, as evidenced by the increase in microtubule-associated protein 2 light chain 3 (LC3-II) and p62/SQSTM1 (p62) and the decline in LAMP1 and LAMP2 levels. After injury, treatment with guanabenz protected neurons from UPR failure and increased lysosomes biogenesis, unblocking autophagic flux. These effects correlated with greater activation of TFEB and improved neuronal survival and functional recovery—effects that persisted after suspension of the treatment. Collectively, our results demonstrate that in remote secondary damage, impairments in autophagic flux are intertwined with ER stress, an association that contributes to the apoptotic cell death and functional damage that are observed after SCI.
Collapse
|
27
|
Wang X, Wang X, Cong P, Wu L, Ma Y, Wang Z, Jiang T, Xu J. Sea cucumber ether-phospholipids improve hepatic steatosis and enhance hypothalamic autophagy in high-fat diet-fed mice. J Nutr Biochem 2022; 106:109032. [DOI: 10.1016/j.jnutbio.2022.109032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 01/02/2022] [Accepted: 03/18/2022] [Indexed: 10/18/2022]
|
28
|
Lauzier A, Bossanyi MF, Larcher R, Nassari S, Ugrankar R, Henne WM, Jean S. Snazarus and its human ortholog SNX25 modulate autophagic flux. J Cell Sci 2022; 135:273525. [PMID: 34821359 DOI: 10.1242/jcs.258733] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 11/12/2021] [Indexed: 12/26/2022] Open
Abstract
Macroautophagy, the degradation and recycling of cytosolic components in the lysosome, is an important cellular mechanism. It is a membrane-mediated process that is linked to vesicular trafficking events. The sorting nexin (SNX) protein family controls the sorting of a large array of cargoes, and various SNXs impact autophagy. To improve our understanding of their functions in vivo, we screened all Drosophila SNXs using inducible RNA interference in the fat body. Significantly, depletion of Snazarus (Snz) led to decreased autophagic flux. Interestingly, we observed altered distribution of Vamp7-positive vesicles with Snz depletion, and the roles of Snz were conserved in human cells. SNX25, the closest human ortholog to Snz, regulates both VAMP8 endocytosis and lipid metabolism. Through knockout-rescue experiments, we demonstrate that these activities are dependent on specific SNX25 domains and that the autophagic defects seen upon SNX25 loss can be rescued by ethanolamine addition. We also demonstrate the presence of differentially spliced forms of SNX14 and SNX25 in cancer cells. This work identifies a conserved role for Snz/SNX25 as a regulator of autophagic flux and reveals differential isoform expression between paralogs.
Collapse
Affiliation(s)
- Annie Lauzier
- Faculté de Médecine et des Sciences de la Santé, Département d'immunologie et de biologie cellulaire, Université de Sherbrooke, 3201, Rue Jean Mignault, Sherbrooke, Québec, CanadaJ1E 4K8
| | - Marie-France Bossanyi
- Faculté de Médecine et des Sciences de la Santé, Département d'immunologie et de biologie cellulaire, Université de Sherbrooke, 3201, Rue Jean Mignault, Sherbrooke, Québec, CanadaJ1E 4K8
| | - Raphaëlle Larcher
- Faculté de Médecine et des Sciences de la Santé, Département d'immunologie et de biologie cellulaire, Université de Sherbrooke, 3201, Rue Jean Mignault, Sherbrooke, Québec, CanadaJ1E 4K8
| | - Sonya Nassari
- Faculté de Médecine et des Sciences de la Santé, Département d'immunologie et de biologie cellulaire, Université de Sherbrooke, 3201, Rue Jean Mignault, Sherbrooke, Québec, CanadaJ1E 4K8
| | - Rupali Ugrankar
- Department of Cell Biology, UT Southwestern Medical Center, 6000 Hary Lines Boulevard, Dallas, TX 75390, USA
| | - W Mike Henne
- Department of Cell Biology, UT Southwestern Medical Center, 6000 Hary Lines Boulevard, Dallas, TX 75390, USA
| | - Steve Jean
- Faculté de Médecine et des Sciences de la Santé, Département d'immunologie et de biologie cellulaire, Université de Sherbrooke, 3201, Rue Jean Mignault, Sherbrooke, Québec, CanadaJ1E 4K8
| |
Collapse
|
29
|
Park EJ, Jin SW, Kang MS, Yang MJ, Kim SH, Han HY, Kang JW. Pulmonary inflammation and cellular responses following exposure to benzalkonium chloride: Potential impact of disrupted pulmonary surfactant homeostasis. Toxicol Appl Pharmacol 2022; 440:115930. [PMID: 35202710 DOI: 10.1016/j.taap.2022.115930] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 10/19/2022]
Abstract
Benzalkonium chloride (BKC) is a prototypical quaternary ammonium disinfectant. Previously, we suggested a no lethal dose level (0.005%) and an LD50 range (0.5-0.05%) of BKC following a single pharyngeal aspiration. Herein, we exposed BKC repeatedly by pharyngeal aspiration for 14 days (0.005 and 0.01%, female mice, total five times with interval of two days, 5 mice/group) and 28 days (0, 0.001, 0.005, and 0.01%, male and female mice, weekly, 16 mice/sex/group). Death following 14 days-repeated exposure did not occur. Meanwhile, chronic pathological lesions were observed in the lung tissues of mice exposed to BKC for 28 days. The total number of bronchial alveolar lavage cells increased, and pulmonary homeostasis of immunologic messenger molecules was disturbed. Following, we investigated BKC-induced cellular responses using human bronchial epithelial cells. The cytotoxicity increased rapidly with concentration. Lysosomal volume, NO production, and lipid peroxidation increased in BKC-treated cells, whereas intracellular ROS level decreased accompanying structural and functional damage of mitochondria. We also found that BKC affected the expression level of immune response, DNA damage, and amino acid biosynthesis-related molecules. More interestingly, lamellar body- and autophagosome-like structures were notably observed in cells exposed to BKC, and necrotic and apoptotic cell death were identified accompanying cell accumulation in the G2/M phase. Therefore, we suggest that repeated respiratory exposure of BKC causes pulmonary inflammation and lung tissue damage and that dead and damaged cells may contribute to the inflammatory response. In addition, the formation process of lamellar body-like structures may function as a key toxicity mechanism.
Collapse
Affiliation(s)
- Eun-Jung Park
- Department of Biochemistry and Molecular Biology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; Human Health and Environmental Toxins Research Center, Kyung Hee University, 02447, Republic of Korea; Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Republic of Korea.
| | - Seung-Woo Jin
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Republic of Korea
| | - Min-Sung Kang
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Republic of Korea; Jeonbuk Branch Institute, Korea Institute of Toxicology, 56212, Republic of Korea
| | - Mi-Jin Yang
- Jeonbuk Branch Institute, Korea Institute of Toxicology, 56212, Republic of Korea
| | - Sung-Hwan Kim
- Jeonbuk Branch Institute, Korea Institute of Toxicology, 56212, Republic of Korea
| | - Hyoung-Yun Han
- Korea Institute of Toxicology, 141 Gajeong-ro, Yuseong-gu, Daejeon 34114, Republic of Korea
| | - Jeong Won Kang
- Department of Chemical and Biological Engineering, Korea University, 0284, Republic of Korea; Graduate School of Energy and Environment, Korea University, 0284, Republic of Korea
| |
Collapse
|
30
|
Nuciferine protects against high-fat diet-induced hepatic steatosis and insulin resistance via activating TFEB-mediated autophagy–lysosomal pathway. Acta Pharm Sin B 2021; 12:2869-2886. [PMID: 35755273 PMCID: PMC9214335 DOI: 10.1016/j.apsb.2021.12.012] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/10/2021] [Accepted: 11/17/2021] [Indexed: 12/30/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is characterized by hepatic steatosis and insulin resistance and there are currently no approved drugs for its treatment. Hyperactivation of mTOR complex 1 (mTORC1) and subsequent impairment of the transcription factor EB (TFEB)-mediated autophagy–lysosomal pathway (ALP) are implicated in the development of NAFLD. Accordingly, agents that augment hepatic TFEB transcriptional activity may have therapeutic potential against NAFLD. The objective of this study was to investigate the effects of nuciferine, a major active component from lotus leaf, on NAFLD and its underlying mechanism of action. Here we show that nuciferine activated ALP and alleviated steatosis, insulin resistance in the livers of NAFLD mice and palmitic acid-challenged hepatocytes in a TFEB-dependent manner. Mechanistic investigation revealed that nuciferine interacts with the Ragulator subunit hepatitis B X-interacting protein and impairs the interaction of the Ragulator complex with Rag GTPases, thereby suppressing lysosomal localization and activity of mTORC1, which activates TFEB-mediated ALP and further ameliorates hepatic steatosis and insulin resistance. Our present results indicate that nuciferine may be a potential agent for treating NAFLD and that regulation of the mTORC1–TFEB–ALP axis could represent a novel pharmacological strategy to combat NAFLD.
Collapse
|
31
|
Ogino N, Miyagawa K, Nagaoka K, Sumida K, Kusanaga M, Oe S, Honma Y, Shibata M, Harada M, Suganuma N, Ogino K. Airborne fine particulate matter in Japan induces lipid synthesis and inhibits autophagy in HepG2 cells. Int J Biochem Cell Biol 2021; 141:106099. [PMID: 34673217 DOI: 10.1016/j.biocel.2021.106099] [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: 06/22/2021] [Revised: 10/06/2021] [Accepted: 10/13/2021] [Indexed: 11/15/2022]
Abstract
Inhalation of particulate matter with a diameter less than 2.5 µm has been reported to exacerbates fatty liver disease. However, the components and mechanisms of particulate matter involved in hepatic lipid metabolism and autophagy have not been fully elucidated. We found that atmospheric particulate matter in Japan stimulated lipogenesis in hepatocytes even when its lipid component was removed. Furthermore, we demonstrated that particulate matter did not promote autophagosome formation but inhibited autophagic degradation in hepatocytes. In previous toxicity experiments, particulate matter collected from atmosphere often contained contaminants originating from filters. In this study, we exposed the powdery particulate matter with less contaminants collected using a cyclone and impactor system to HepG2 cells, human hepatocyte. This particulate matter induced lipogenesis and endoplasmic reticulum stress in HepG2 cells as well as previous reports of particulate matter in the USA and China. On the other hand, when autophagic flux were examined in detail, the particulate matter did not promote autophagosome formation, but inhibited autophagic degradation. Since these effects were similar to those of palmitate, a fatty acid, we prepared particulate matter in which lipid component was removed by acetone and compared the effects on HepG2 cells with those of untreated one. The particulate matter without lipid component induced lipid droplets as well as did the untreated one although it induced less endoplasmic reticulum stress. These results suggest that hepatic lipid synthesis is stimulated not only by the uptake of lipid but also by other components in the particulate matter.
Collapse
Affiliation(s)
- Noriyoshi Ogino
- Department of Environmental Medicine, Kochi Medical School, Kohasu, Oko-cho, Nankoku City, Kochi 783-8505, Japan; Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Iseigaoka 1-1, Yahatanishi-ku, Kitakyushu 807-8555, Japan.
| | - Koichiro Miyagawa
- Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Iseigaoka 1-1, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| | - Kenjiro Nagaoka
- Laboratory of Hygienic Chemistry, College of Pharmaceutical Sciences, Matsuyama University, Matsuyama, Ehime 790-8578, Japan.
| | - Kazuhiro Sumida
- Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Iseigaoka 1-1, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| | - Masashi Kusanaga
- Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Iseigaoka 1-1, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| | - Shinji Oe
- Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Iseigaoka 1-1, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| | - Yuichi Honma
- Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Iseigaoka 1-1, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| | - Michihiko Shibata
- Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Iseigaoka 1-1, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| | - Masaru Harada
- Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Iseigaoka 1-1, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| | - Narufumi Suganuma
- Department of Environmental Medicine, Kochi Medical School, Kohasu, Oko-cho, Nankoku City, Kochi 783-8505, Japan
| | - Keiki Ogino
- Department of Environmental Medicine, Kochi Medical School, Kohasu, Oko-cho, Nankoku City, Kochi 783-8505, Japan.
| |
Collapse
|
32
|
NEFA Promotes Autophagosome Formation through Modulating PERK Signaling Pathway in Bovine Hepatocytes. Animals (Basel) 2021; 11:ani11123400. [PMID: 34944177 PMCID: PMC8697899 DOI: 10.3390/ani11123400] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 11/21/2021] [Accepted: 11/25/2021] [Indexed: 12/11/2022] Open
Abstract
During the perinatal period, the abnormally high plasma non-esterified fatty acids (NEFA) concentration caused by the negative energy balance (NEB) can impose a significant metabolic stress on the liver of dairy cows. Endoplasmic reticulum (ER) stress is an important adaptive response that can serve to maintain cell homeostasis in the event of stress. The protein kinase R-like endoplasmic reticulum kinase (PERK) pathway is the most rapidly activated cascade when ER stress occurs in cells and has an important impact on the regulation of hepatic lipid metabolism and autophagy modulation. However, it is unknown whether NEFA can affect autophagy through modulating the PERK pathway, under NEB conditions. In this study, we provide evidence that NEFA treatment markedly increased lipid accumulation, the phosphorylation level of PERK and eukaryotic initiation factor 2α (eIF2α), and the expression of glucose-regulated protein 78 (Grp78), activating transcription factor 4 (ATF4), and C/EBP homologous protein (CHOP). More importantly, NEFA treatment can cause a substantial increase in the protein levels of autophagy-related gene 7 (ATG7), Beclin-1 (BECN1), sequestosome-1 (p62), and microtubule-associated protein 1 light chain 3 (LC3)-II, and in the number of autophagosomes in primary bovine hepatocytes. The addition of GSK2656157 (PERK phosphorylation inhibitor) can significantly inhibit the effect of NEFA on autophagy and can further increase lipid accumulation. Overall, our results indicate that NEFA could promote autophagy via the PERK pathway in bovine hepatocytes. These findings provide novel evidence about the potential role of the PERK signaling pathway in maintaining bovine hepatocyte homeostasis.
Collapse
|
33
|
Lopresti MW, Cui W, Abernathy B, Fredrickson G, Barrow F, Desai AS, Revelo XS, Mashek D. Hepatic Lysosomal Acid Lipase Overexpression Worsens Hepatic Inflammation in Mice Fed a Western Diet. J Lipid Res 2021; 62:100133. [PMID: 34624333 PMCID: PMC8556525 DOI: 10.1016/j.jlr.2021.100133] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 09/16/2021] [Accepted: 09/23/2021] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is characterized by the accumulation of lipid droplets in hepatocytes. NAFLD development and progression is associated with an increase in hepatic cholesterol levels and decreased autophagy and lipophagy flux. Previous studies have shown that the expression of lysosomal acid lipase (LAL), encoded by the gene LIPA, which can hydrolyze both triglyceride and cholesteryl esters, is inversely correlated with the severity of NAFLD. In addition, ablation of LAL activity results in profound NAFLD. Based on this, we predicted that overexpressing LIPA in the livers of mice fed a Western diet would prevent the development of NAFLD. As expected, mice fed the Western diet exhibited numerous markers of NAFLD, including hepatomegaly, lipid accumulation, and inflammation. Unexpectedly, LAL overexpression did not attenuate steatosis and had only minor effects on neutral lipid composition. However, LAL overexpression exacerbated inflammatory gene expression and infiltration of immune cells in mice fed the Western diet. LAL overexpression also resulted in abnormal phagosome accumulation and lysosomal lipid accumulation depending upon the dietary treatment. Overall, we found that hepatic overexpression of LAL drove immune cell infiltration and inflammation and did not attenuate the development of NAFLD, suggesting that targeting LAL expression may not be a viable route to treat NAFLD in humans.
Collapse
Affiliation(s)
- Michael W Lopresti
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis MN
| | - Wenqi Cui
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis MN
| | - Breann Abernathy
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis MN
| | - Gavin Fredrickson
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis MN
| | - Fanta Barrow
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis MN
| | - Arnav S Desai
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis MN
| | - Xavier S Revelo
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis MN
| | - Douglas Mashek
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis MN; Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, University of Minnesota, Minneapolis MN.
| |
Collapse
|
34
|
Fang C, Weng T, Hu S, Yuan Z, Xiong H, Huang B, Cai Y, Li L, Fu X. IFN-γ-induced ER stress impairs autophagy and triggers apoptosis in lung cancer cells. Oncoimmunology 2021; 10:1962591. [PMID: 34408924 PMCID: PMC8366549 DOI: 10.1080/2162402x.2021.1962591] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Interferon-gamma (IFN-γ) is a major effector molecule of immunity and a common feature of tumors responding to immunotherapy. Active IFN-γ signaling can directly trigger apoptosis and cell cycle arrest in human cancer cells. However, the mechanisms underlying these actions remain unclear. Here, we report that IFN-γ rapidly increases protein synthesis and causes the unfolded protein response (UPR), as evidenced by the increased expression of glucose-regulated protein 78, activating transcription factor-4, and c/EBP homologous protein (CHOP) in cells treated with IFN-γ. The JAK1/2-STAT1 and AKT-mTOR signaling pathways are required for IFN-γ-induced UPR. Endoplasmic reticulum (ER) stress promotes autophagy and restores homeostasis. Surprisingly, in IFN-γ-treated cells, autophagy was impaired at the step of autophagosome-lysosomal fusion and caused by a significant decline in the expression of lysosomal membrane protein-1 and −2 (LAMP-1/LAMP-2). The ER stress inhibitor 4-PBA restored LAMP expression in IFN-γ-treated cells. IFN-γ stimulation activated the protein kinase-like ER kinase (PERK)-eukaryotic initiation factor 2a subunit (eIF2α) axis and caused a reduction in global protein synthesis. The PERK inhibitor, GSK2606414, partially restored global protein synthesis and LAMP expression in cells treated with IFN-γ. We further investigated the functional consequences of IFN-γ-induced ER stress. We show that inhibition of ER stress significantly prevents IFN-γ-triggered apoptosis. CHOP knockdown abrogated IFN-γ-mediated apoptosis. Inhibition of ER stress also restored cyclin D1 expression in IFN-γ-treated cells. Thus, ER stress and the UPR caused by IFN-γ represent novel mechanisms underlying IFN-γ-mediated anticancer effects. This study expands our understanding of IFN-γ-mediated signaling and its cellular actions in tumor cells.
Collapse
Affiliation(s)
- Can Fang
- Thoracic Surgery Laboratory, Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Weng
- Thoracic Surgery Laboratory, Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shaojie Hu
- Thoracic Surgery Laboratory, Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiwei Yuan
- Thoracic Surgery Laboratory, Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Xiong
- Thoracic Surgery Laboratory, Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bing Huang
- Thoracic Surgery Laboratory, Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yixin Cai
- Thoracic Surgery Laboratory, Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lequn Li
- Thoracic Surgery Laboratory, Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangning Fu
- Thoracic Surgery Laboratory, Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
35
|
Lou J, Yang X, Shan W, Jin Z, Ding J, Hu Y, Liao Q, Du Q, Xie R, Xu J. Effects of calcium‑permeable ion channels on various digestive diseases in the regulation of autophagy (Review). Mol Med Rep 2021; 24:680. [PMID: 34318907 DOI: 10.3892/mmr.2021.12319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 05/07/2021] [Indexed: 12/09/2022] Open
Abstract
Autophagy is a process of degradation and catabolism in cells. By removing damaged or dysfunctional organelles, autophagy interacts with the ubiquitin‑proteasome degradation system to jointly regulate cell function and energy homeostasis. Since autophagy plays a key role in physiology, disorders of the autophagy mechanism are associated with various diseases. Therefore, thorough understanding of the autophagy regulatory mechanism are crucially important in the diagnosis and treatment of diseases. To date, ion channels may affect the development and treatment of diseases by regulating autophagy, especially calcium‑permeable ion channels, in the process of digestive system diseases. However, the mechanism by which calcium ions and their channels regulate autophagy is still poorly understood, thus emphasizing the need for further research in this field. The present review intends to discuss the association, mechanism and application of calcium ions, their channels and autophagy in the occurrence and development of digestive system diseases.
Collapse
Affiliation(s)
- Jun Lou
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Xiaoxu Yang
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Weixi Shan
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Zhe Jin
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Jianhong Ding
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Yanxia Hu
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Qiushi Liao
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Qian Du
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Rui Xie
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Jingyu Xu
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| |
Collapse
|
36
|
Guo W, Zhong W, Hao L, Dong H, Sun X, Yue R, Li T, Zhou Z. Fatty Acids Inhibit LAMP2-Mediated Autophagy Flux via Activating ER Stress Pathway in Alcohol-Related Liver Disease. Cell Mol Gastroenterol Hepatol 2021; 12:1599-1615. [PMID: 34284164 PMCID: PMC8536789 DOI: 10.1016/j.jcmgh.2021.07.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/30/2021] [Accepted: 07/01/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Alcohol-related liver disease (ALD) is characterized by accumulation of hepatic free fatty acids (FFAs) and triglyceride (TG)-enriched lipid droplets and cell death. The present study aimed to investigate how FFA or TG induces hepatocyte injury, thereby contributing to the development of ALD. METHODS Hepatocyte-specific DGAT1 knockout (DGAT1Δhep) mice and lysosome-associated membrane protein 2 (LAMP2) overexpression mice were generated and subjected to chronic alcohol feeding. Cell studies were conducted to define the causal role and underlying mechanism of FFA-induced hepatocellular injury. RESULTS Hepatocyte-specific DGAT1 deletion exacerbated alcohol-induced liver injury by increasing lipid accumulation and endoplasmic reticulum (ER) stress, reducing LAMP2 protein levels, and impairing autophagy function. Cell studies revealed that FFAs, rather than TG, induced ER stress via ATF4 activation, which, in turn, down-regulated LAMP2, thereby impairing autophagy flux. LAMP2 overexpression in the liver restored autophagy function and ameliorated alcohol-induced liver injury in mice. Reducing hepatic FFAs by peroxisome proliferator-activated receptor α activation attenuated ER stress, restored LAMP2 protein levels, and improved autophagy flux. In addition, suppression of LAMP2 and autophagy function was also detected in the liver of patients with severe alcoholic hepatitis. CONCLUSIONS This study demonstrates that accumulation of hepatic FFAs, rather than TG, plays a crucial role in the pathogenesis of ALD by suppressing LAMP2-autophagy flux pathway through ER stress signaling, which represents an important mechanism of FFA-induced hepatocellular injury in ALD.
Collapse
Affiliation(s)
- Wei Guo
- Center for Translational Biomedical Research, Kannapolis, North Carolina
| | - Wei Zhong
- Center for Translational Biomedical Research, Kannapolis, North Carolina,Department of Nutrition, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, North Carolina
| | - Liuyi Hao
- Center for Translational Biomedical Research, Kannapolis, North Carolina
| | - Haibo Dong
- Center for Translational Biomedical Research, Kannapolis, North Carolina
| | - Xinguo Sun
- Center for Translational Biomedical Research, Kannapolis, North Carolina
| | - Ruichao Yue
- Center for Translational Biomedical Research, Kannapolis, North Carolina
| | - Tianjiao Li
- Department of Nutrition, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, North Carolina
| | - Zhanxiang Zhou
- Center for Translational Biomedical Research, Kannapolis, North Carolina,Department of Nutrition, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, North Carolina,Correspondence Address correspondence to: Zhanxiang Zhou, PhD, Center for Translational Biomedical Research, University of North Carolina at Greensboro, 600 Laureate Way, Suite 2203, Kannapolis, North Carolina 28081.fax: (704) 250-5809.
| |
Collapse
|
37
|
Álvarez-Mercado AI, Rojano-Alfonso C, Micó-Carnero M, Caballeria-Casals A, Peralta C, Casillas-Ramírez A. New Insights Into the Role of Autophagy in Liver Surgery in the Setting of Metabolic Syndrome and Related Diseases. Front Cell Dev Biol 2021; 9:670273. [PMID: 34141709 PMCID: PMC8204012 DOI: 10.3389/fcell.2021.670273] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 04/23/2021] [Indexed: 01/18/2023] Open
Abstract
Visceral obesity is an important component of metabolic syndrome, a cluster of diseases that also includes diabetes and insulin resistance. A combination of these metabolic disorders damages liver function, which manifests as non-alcoholic fatty liver disease (NAFLD). NAFLD is a common cause of abnormal liver function, and numerous studies have established the enormously deleterious role of hepatic steatosis in ischemia-reperfusion (I/R) injury that inevitably occurs in both liver resection and transplantation. Thus, steatotic livers exhibit a higher frequency of post-surgical complications after hepatectomy, and using liver grafts from donors with NAFLD is associated with an increased risk of post-surgical morbidity and mortality in the recipient. Diabetes, another MetS-related metabolic disorder, also worsens hepatic I/R injury, and similar to NAFLD, diabetes is associated with a poor prognosis after liver surgery. Due to the large increase in the prevalence of MetS, NAFLD, and diabetes, their association is frequent in the population and therefore, in patients requiring liver resection and in potential liver graft donors. This scenario requires advancement in therapies to improve postoperative results in patients suffering from metabolic diseases and undergoing liver surgery; and in this sense, the bases for designing therapeutic strategies are in-depth knowledge about the molecular signaling pathways underlying the effects of MetS-related diseases and I/R injury on liver tissue. A common denominator in all these diseases is autophagy. In fact, in the context of obesity, autophagy is profoundly diminished in hepatocytes and alters mitochondrial functions in the liver. In insulin resistance conditions, there is a suppression of autophagy in the liver, which is associated with the accumulation of lipids, being this is a risk factor for NAFLD. Also, oxidative stress occurring in hepatic I/R injury promotes autophagy. The present review aims to shed some light on the role of autophagy in livers undergoing surgery and also suffering from metabolic diseases, which may lead to the discovery of effective therapeutic targets that could be translated from laboratory to clinical practice, to improve postoperative results of liver surgeries when performed in the presence of one or more metabolic diseases.
Collapse
Affiliation(s)
- Ana Isabel Álvarez-Mercado
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, Granada, Spain
- Institute of Nutrition and Food Technology “José Mataix”, Biomedical Research Center, Parque Tecnológico Ciencias de la Salud, Granada, Spain
- Instituto de Investigación Biosanitaria ibs. GRANADA, Complejo Hospitalario Universitario de Granada, Granada, Spain
| | - Carlos Rojano-Alfonso
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Marc Micó-Carnero
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | | | - Carmen Peralta
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Araní Casillas-Ramírez
- Hospital Regional de Alta Especialidad de Ciudad Victoria “Bicentenario 2010”, Ciudad Victoria, Mexico
- Facultad de Medicina e Ingeniería en Sistemas Computacionales de Matamoros, Universidad Autónoma de Tamaulipas, Matamoros, Mexico
| |
Collapse
|
38
|
Shao M, Shi R, Gao ZX, Gao SS, Li JF, Li H, Cui SZ, Hu WM, Chen TY, Wu GR, Zhang J, Xu J, Sy MS, Li C. Crizotinib and Doxorubicin Cooperatively Reduces Drug Resistance by Mitigating MDR1 to Increase Hepatocellular Carcinoma Cells Death. Front Oncol 2021; 11:650052. [PMID: 34094940 PMCID: PMC8170002 DOI: 10.3389/fonc.2021.650052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 04/16/2021] [Indexed: 11/24/2022] Open
Abstract
As the sixth most lethal cancers worldwide, hepatocellular carcinoma (HCC) has been treated with doxorubicin (Dox) for decades. However, chemotherapy resistance, especially for Dox is an even more prominent problem due to its high cardiotoxicity. To find a regimen to reduce Dox resistance, and identify the mechanisms behind it, we tried to identify combination of drugs that can overcome drug resistance by screening tyrosine kinase inhibitor(s) with Dox with various HCC cell lines in vitro and in vivo. We report here that combination of Crizo and Dox has a synergistic effect on inducing HCC cell death. Accordingly, Crizo plus Dox increases Dox accumulation in nucleus 3-16 times compared to Dox only; HCC cell death enhanced at least 50% in vitro and tumor weights reduced ranging from 35 to 65%. Combining these two drugs reduces multiple drug resistance 1 (MDR1) protein as a result of activation of protein kinase RNA-like endoplasmic reticulum kinase (PERK), which phosphorylates eIF2α, leading to protein translational repression. Additionally, PERK stimulation activates C-Jun terminal kinase (JNK), resulting in accumulation of unfused autophagosome to enhance autophagic cell death via Poly-ADP-ribosyltransferase (PARP-1) cleavage. When the activity of PERK or JNK is blocked, unfused autophagosome is diminished, cleaved PARP-1 is reduced, and cell death is abated. Therefore, Crizo plus Dox sensitize HCC drug resistance by engaging PERK-p- eIF2α-MDR1, and kill HCC cells by engaging PERK-JNK- autophagic cell death pathways. These newly discovered mechanisms of Crizo plus Dox not only provide a potential treatment for HCC but also point to an approach to overcome MDR1 related drug resistance in other cancers.
Collapse
Affiliation(s)
- Ming Shao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Run Shi
- State Key Laboratory of Respiratory Disease, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Zhen-Xing Gao
- State Key Laboratory of Respiratory Disease, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Shan-Shan Gao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Jing-Feng Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Huan Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Shu-Zhong Cui
- State Key Laboratory of Respiratory Disease, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China.,Abdominal Surgery, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Wei-Min Hu
- State Key Laboratory of Respiratory Disease, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Tian-Yun Chen
- Department of Stomatology, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Gui-Ru Wu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Jie Zhang
- Department of Stomatology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Jiang Xu
- Department of Stomatology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Man-Sun Sy
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Chaoyang Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,State Key Laboratory of Respiratory Disease, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
39
|
Gao W, Fang Z, Lei L, Ju L, Jin B, Loor JJ, Liang Y, Shi Z, Shen T, Yu H, Chen M, Ouyang H, Song Y, Wang Z, Liu G, Li X, Du X. Propionate alleviates palmitic acid-induced endoplasmic reticulum stress by enhancing autophagy in calf hepatic cells. J Dairy Sci 2021; 104:9316-9326. [PMID: 34001357 DOI: 10.3168/jds.2020-19969] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 04/10/2021] [Indexed: 12/30/2022]
Abstract
Negative energy balance-induced high blood concentrations of free fatty acids during the early postpartum period in dairy cows is a major cause of liver injury. Cows in severe negative energy balance often have suboptimal intakes of feed, which contributes to shortfalls in production of ruminal propionate and circulating glucose. Although increasing propionate production by the rumen through feed additives such as propylene glycol is effective in helping cows alleviate the shortfall in dietary energy supply, mechanisms whereby propionate affects liver function beyond gluconeogenesis are unknown. Therefore, the objective of this study was to investigate whether propionate could protect calf hepatic cells from palmitic acid (PA)-induced lipotoxicity and the underlying mechanisms. Calf hepatic cells were isolated from 5 healthy calves (1 d old, female, 30-40 kg, fasting) and treated with various concentrations of PA (0, 100, 200, or 400 μM) and propionate (0, 1, 2, or 4 mM) after being administered with or without autophagic inhibitor. Propionate enhanced autophagic activity in calf hepatic cells, as indicated by elevated expression of autophagy markers LC3-II (microtubule-associated protein 1 light chain 3-II, encoded by MAP1LC3) and decreased expression of SQSTM1 (sequestosome-1, also called p62). Conversely, PA suppressed autophagic activity and decreased cell viability, which was improved by propionate in calf hepatic cells. In addition, propionate decreased the phosphorylation of proteins EIF2AK3 (kinase R/PKR like ER kinase) and ERN1 (inositol-requiring enzyme 1α) and cleaved ATF6 (activating transcription factor 6) in PA-treated calf hepatic cells, indicating the suppression effect of propionate on endoplasmic reticulum (ER) stress. However, inhibition of autophagic activity by chloroquine or bafilomycin A1 impede the beneficial effects of propionate on ER stress and cell viability. These results demonstrated that propionate alleviates ER stress and elevates cell viability in PA-treated calf hepatic cells by enhancing autophagy, which implies that autophagy may be a promising target in improving liver injury of dairy cows during transition period.
Collapse
Affiliation(s)
- Wenwen Gao
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun 130062, Jilin, China
| | - Zhiyuan Fang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun 130062, Jilin, China
| | - Lin Lei
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun 130062, Jilin, China
| | - Lingxue Ju
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun 130062, Jilin, China
| | - Bo Jin
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun 130062, Jilin, China
| | - Juan J Loor
- Mammalian NutriPhysioGenomics Laboratory, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - Yusheng Liang
- Mammalian NutriPhysioGenomics Laboratory, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - Zhen Shi
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun 130062, Jilin, China
| | - Taiyu Shen
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun 130062, Jilin, China
| | - Hao Yu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun 130062, Jilin, China
| | - Meng Chen
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun 130062, Jilin, China
| | - Hongsheng Ouyang
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University, 5333 Xi'an Road, Changchun 130062, China
| | - Yuxiang Song
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun 130062, Jilin, China
| | - Zhe Wang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun 130062, Jilin, China
| | - Guowen Liu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun 130062, Jilin, China
| | - Xinwei Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun 130062, Jilin, China
| | - Xiliang Du
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun 130062, Jilin, China.
| |
Collapse
|
40
|
Ramos VDM, Kowaltowski AJ, Kakimoto PA. Autophagy in Hepatic Steatosis: A Structured Review. Front Cell Dev Biol 2021; 9:657389. [PMID: 33937257 PMCID: PMC8081956 DOI: 10.3389/fcell.2021.657389] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 03/16/2021] [Indexed: 01/18/2023] Open
Abstract
Steatosis is the accumulation of neutral lipids in the cytoplasm. In the liver, it is associated with overeating and a sedentary lifestyle, but may also be a result of xenobiotic toxicity and genetics. Non-alcoholic fatty liver disease (NAFLD) defines an array of liver conditions varying from simple steatosis to inflammation and fibrosis. Over the last years, autophagic processes have been shown to be directly associated with the development and progression of these conditions. However, the precise role of autophagy in steatosis development is still unclear. Specifically, autophagy is necessary for the regulation of basic metabolism in hepatocytes, such as glycogenolysis and gluconeogenesis, response to insulin and glucagon signaling, and cellular responses to free amino acid contents. Also, genetic knockout models for autophagy-related proteins suggest a critical relationship between autophagy and hepatic lipid metabolism, but some results are still ambiguous. While autophagy may seem necessary to support lipid oxidation in some contexts, other evidence suggests that autophagic activity can lead to lipid accumulation instead. This structured literature review aims to critically discuss, compare, and organize results over the last 10 years regarding rodent steatosis models that measured several autophagy markers, with genetic and pharmacological interventions that may help elucidate the molecular mechanisms involved.
Collapse
Affiliation(s)
| | | | - Pamela A. Kakimoto
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
41
|
Role of HO-1 against Saturated Fatty Acid-Induced Oxidative Stress in Hepatocytes. Nutrients 2021; 13:nu13030993. [PMID: 33808635 PMCID: PMC8003531 DOI: 10.3390/nu13030993] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/11/2021] [Accepted: 03/16/2021] [Indexed: 01/24/2023] Open
Abstract
Increased circulating levels of free fatty acids, especially saturated ones, are involved in disease progression in the non-alcoholic fatty liver. Although the mechanism of saturated fatty acid-induced toxicity in the liver is not fully understood, oxidative stress may be deeply involved. We examined the effect of increased palmitic acid, the most common saturated fatty acid in the blood, on the liver of BALB/c mice via tail vein injection with palmitate. After 24 h, among several anti-oxidative stress response genes, only heme oxygenase-1 (HO-1) was significantly upregulated in palmitate-injected mice compared with that in vehicle-injected mice. Elevation of HO-1 mRNA was also observed in the fatty liver of high-fat-diet-fed mice. To further investigate the role of HO-1 on palmitic acid-induced oxidative stress, in vitro experiments were performed to expose palmitate to HepG2 cells. SiRNA-mediated knockdown of HO-1 significantly increased the oxidative stress induced by palmitate, whereas pre-treatment with SnCl2, a well-known HO-1 inducer, significantly decreased it. Moreover, SB203580, a selective p38 inhibitor, reduced HO-1 mRNA expression and increased palmitate-induced oxidative stress in HepG2 cells. These results suggest that the HO-1-mediated anti-oxidative stress compensatory reaction plays an essential role against saturated fatty acid-induced lipotoxicity in the liver.
Collapse
|
42
|
Luo Y, Fu Y, Huang Z, Li M. Transition metals and metal complexes in autophagy and diseases. J Cell Physiol 2021; 236:7144-7158. [PMID: 33694161 DOI: 10.1002/jcp.30359] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 02/19/2021] [Accepted: 02/27/2021] [Indexed: 12/19/2022]
Abstract
Transition metals refer to the elements in the d and ds blocks of the periodic table. Since the success of cisplatin and auranofin, transition metal-based compounds have become a prospective source for drug development, particularly in cancer treatment. In recent years, extensive studies have shown that numerous transition metal-based compounds could modulate autophagy, promising a new therapeutic strategy for metal-related diseases and the design of metal-based agents. Copper, zinc, and manganese, which are common components in physiological pathways, play important roles in the progression of cancer, neurodegenerative diseases, and cardiovascular diseases. Furthermore, enrichment of copper, zinc, or manganese can regulate autophagy. Thus, we summarized the current advances in elucidating the mechanisms of some metals/metal-based compounds and their functions in autophagy regulation, which is conducive to explore the intricate roles of autophagy and exploit novel therapeutic drugs for human diseases.
Collapse
Affiliation(s)
- Yuping Luo
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yuanyuan Fu
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Zhiying Huang
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Min Li
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
43
|
How does hepatic lipid accumulation lead to lipotoxicity in non-alcoholic fatty liver disease? Hepatol Int 2021; 15:21-35. [PMID: 33548031 PMCID: PMC7886759 DOI: 10.1007/s12072-020-10121-2] [Citation(s) in RCA: 221] [Impact Index Per Article: 55.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 12/10/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD), characterized as excess lipid accumulation in the liver which is not due to alcohol use, has emerged as one of the major health problems around the world. The dysregulated lipid metabolism creates a lipotoxic environment which promotes the development of NAFLD, especially the progression from simple steatosis (NAFL) to non-alcoholic steatohepatitis (NASH). PURPOSEAND AIM This review focuses on the mechanisms of lipid accumulation in the liver, with an emphasis on the metabolic fate of free fatty acids (FFAs) in NAFLD and presents an update on the relevant cellular processes/mechanisms that are involved in lipotoxicity. The changes in the levels of various lipid species that result from the imbalance between lipolysis/lipid uptake/lipogenesis and lipid oxidation/secretion can cause organellar dysfunction, e.g. ER stress, mitochondrial dysfunction, lysosomal dysfunction, JNK activation, secretion of extracellular vesicles (EVs) and aggravate (or be exacerbated by) hypoxia which ultimately lead to cell death. The aim of this review is to provide an overview of how abnormal lipid metabolism leads to lipotoxicity and the cellular mechanisms of lipotoxicity in the context of NAFLD.
Collapse
|
44
|
Honma Y, Miyagawa K, Hara Y, Hayashi T, Kusanaga M, Ogino N, Minami S, Oe S, Ikeda M, Hino K, Harada M. Correlation of hepatitis C virus-mediated endoplasmic reticulum stress with autophagic flux impairment and hepatocarcinogenesis. Med Mol Morphol 2021; 54:108-121. [PMID: 33386512 DOI: 10.1007/s00795-020-00271-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/13/2020] [Indexed: 12/29/2022]
Abstract
Hepatitis C virus (HCV) infection has been known to use autophagy for its replication. However, the mechanisms by which HCV modulates autophagy remain controversial. We used HCV-Japanese fulminant hepatitis-1-infected Huh7 cells. HCV infection induced the accumulation of autophagosomes. Morphological analyses of monomeric red fluorescent protein (mRFP)-green fluorescent protein (GFP) tandem fluorescent-tagged LC3 transfection showed HCV infection impaired autophagic flux. Autophagosome-lysosome fusion assessed by transfection of mRFP- or GFP-LC3 and immunostaining of lysosomal-associated membrane protein 1 was inhibited by HCV infection. Decrease of HCV-induced endoplasmic reticulum (ER) stress by 4-phenylbutyric acid, a chemical chaperone, improved the HCV-mediated autophagic flux impairment. HCV infection-induced oxidative stress and subsequently DNA damage, but not apoptosis. Furthermore, HCV induced cytoprotective effects against the cellular stress by facilitating the formation of cytoplasmic inclusion bodies as shown by p62 expression and by modulating keratin protein expression and activated nuclear factor erythroid 2-related factor 2. HCV eradication by direct-acting antivirals improved autophagic flux, but DNA damage persisted. In conclusion, HCV-induced ER stress correlates with autophagic flux impairment. Decrease of ER stress is considered to be a promising therapeutic strategy for HCV-related chronic liver diseases. However, we should be aware that the risk of hepatocarcinogenesis remains even after HCV eradication.
Collapse
Affiliation(s)
- Yuichi Honma
- Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan.
| | - Koichiro Miyagawa
- Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan
| | - Yuichi Hara
- Department of Hepatology and Pancreatology, Kawasaki Medical School, Kurashiki, Japan
| | - Tsuguru Hayashi
- Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan
| | - Masashi Kusanaga
- Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan
| | - Noriyoshi Ogino
- Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan
| | - Sota Minami
- Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan
| | - Shinji Oe
- Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan
| | - Masanori Ikeda
- Department of Persistent and Oncogenic Viruses, Center for Chronic Viral Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Keisuke Hino
- Department of Hepatology and Pancreatology, Kawasaki Medical School, Kurashiki, Japan
| | - Masaru Harada
- Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan.
| |
Collapse
|
45
|
Li J, Li X, Liu D, Zhang S, Tan N, Yokota H, Zhang P. Phosphorylation of eIF2α signaling pathway attenuates obesity-induced non-alcoholic fatty liver disease in an ER stress and autophagy-dependent manner. Cell Death Dis 2020; 11:1069. [PMID: 33318479 PMCID: PMC7736876 DOI: 10.1038/s41419-020-03264-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 11/15/2020] [Accepted: 11/16/2020] [Indexed: 12/12/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common liver disorder and frequently exacerbates in postmenopausal women. In NAFLD, the endoplasmic reticulum (ER) plays an important role in lipid metabolism, in which salubrinal is a selective inhibitor of eIF2α de-phosphorylation in response to ER stress. To determine the potential mechanism of obesity-induced NAFLD, we employed salubrinal and evaluated the effect of ER stress and autophagy on lipid metabolism. Ninety-five female C57BL/6 mice were randomly divided into five groups: standard chow diet, high-fat (HF) diet, HF with salubrinal, HF with ovariectomy, and HF with ovariectomy and salubrinal. All mice except for SC were given HF diet. After the 8-week obesity induction, salubrinal was subcutaneously injected for the next 8 weeks. The expression of ER stress and autophagy markers was evaluated in vivo and in vitro. Compared to the normal mice, the serum lipid level and adipose tissue were increased in obese mice, while salubrinal attenuated obesity by blocking lipid disorder. Also, the histological severity of hepatic steatosis and fibrosis in the liver and lipidosis was suppressed in response to salubrinal. Furthermore, salubrinal inhibited ER stress by increasing the expression of p-eIF2α and ATF4 with a decrease in the level of CHOP. It promoted autophagy by increasing LC3II/I and inhibiting p62. Correlation analysis indicated that lipogenesis in the development of NAFLD was associated with ER stress. Collectively, we demonstrated that eIF2α played a key role in obesity-induced NAFLD, and salubrinal alleviated hepatic steatosis and lipid metabolism by altering ER stress and autophagy through eIF2α signaling.
Collapse
Affiliation(s)
- Jie Li
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin, 300070, China
| | - Xinle Li
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin, 300070, China
| | - Daquan Liu
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin, 300070, China
| | - Shiqi Zhang
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Nian Tan
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Hiroki Yokota
- Department of Biomedical Engineering, Indiana University-Purdue University, Indianapolis, IN, 46202, USA
| | - Ping Zhang
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China.
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin, 300070, China.
- Department of Biomedical Engineering, Indiana University-Purdue University, Indianapolis, IN, 46202, USA.
- Tianjin Key Laboratory of Spine and Spinal Cord, Tianjin Medical University, Tianjin, 300052, China.
| |
Collapse
|
46
|
Baiges-Gaya G, Fernández-Arroyo S, Luciano-Mateo F, Cabré N, Rodríguez-Tomàs E, Hernández-Aguilera A, Castañé H, Romeu M, Nogués MR, Camps J, Joven J. Hepatic metabolic adaptation and adipose tissue expansion are altered in mice with steatohepatitis induced by high-fat high sucrose diet. J Nutr Biochem 2020; 89:108559. [PMID: 33264665 DOI: 10.1016/j.jnutbio.2020.108559] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 10/05/2020] [Accepted: 11/21/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Obesity is a chronic progressive disease with several metabolic alterations. Nonalcoholic fatty liver disease (NAFLD) is an important comorbidity of obesity that can progress to nonalcoholic steatohepatitis (NASH), cirrhosis or hepatocarcinoma. This study aimed at clarifying the molecular mechanisms underlying the metabolic alterations in hepatic and adipose tissue during high-fat high-sucrose diet-induced NAFLD development in mice. METHODS Twenty-four male mice (C57BL/6J) were randomly allocated into 3 groups (n = 8 mice per group) to receive a chow diet, a high-fat diet (HFD), or a high-fat high-sucrose diet (HF-HSD) for 20 weeks. At sacrifice, liver and adipose tissue were obtained for histopathological, metabolomic, and protein expression analyses. RESULTS HF-HSD (but not HFD) was associated with NASH and increased oxidative stress. These animals presented an inhibition of hepatic autophagy and alterations in AMP-activated protein kinase/mammalian target of rapamycin activity. We also observed that the ability of metabolic adaptation was adversely affected by the increase of damaged mitochondria. NASH development was associated with changes in adipose tissue dynamics and increased amounts of saturated fatty acids, monounsaturated fatty acids and polyunsaturated fatty acids in visceral adipose tissue. CONCLUSION HF-HSD led to a metabolic blockage and impaired hepatic mitochondria turnover. In addition, the continuous accumulation of fatty acids produced adipose tissue dysfunction and hepatic fat accumulation that favored the progression to NASH.
Collapse
Affiliation(s)
- Gerard Baiges-Gaya
- Universitat Rovira i Virgili, Departament de Medicina i Cirurgia, Facultat de Medicina, Reus, Spain; Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d'investigació Sanitària Pere Virgili, Reus, Spain
| | - Salvador Fernández-Arroyo
- Universitat Rovira i Virgili, Departament de Medicina i Cirurgia, Facultat de Medicina, Reus, Spain; Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d'investigació Sanitària Pere Virgili, Reus, Spain
| | - Fedra Luciano-Mateo
- Universitat Rovira i Virgili, Departament de Medicina i Cirurgia, Facultat de Medicina, Reus, Spain; Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d'investigació Sanitària Pere Virgili, Reus, Spain
| | - Noemí Cabré
- Universitat Rovira i Virgili, Departament de Medicina i Cirurgia, Facultat de Medicina, Reus, Spain; Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d'investigació Sanitària Pere Virgili, Reus, Spain
| | - Elisabet Rodríguez-Tomàs
- Universitat Rovira i Virgili, Departament de Medicina i Cirurgia, Facultat de Medicina, Reus, Spain; Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d'investigació Sanitària Pere Virgili, Reus, Spain
| | - Anna Hernández-Aguilera
- Universitat Rovira i Virgili, Departament de Medicina i Cirurgia, Facultat de Medicina, Reus, Spain; Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d'investigació Sanitària Pere Virgili, Reus, Spain
| | - Helena Castañé
- Universitat Rovira i Virgili, Departament de Medicina i Cirurgia, Facultat de Medicina, Reus, Spain; Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d'investigació Sanitària Pere Virgili, Reus, Spain
| | - Marta Romeu
- Universitat Rovira i Virgili, Departament de Ciències Mèdiques Bàsiques, Facultat de Medicina, Unitat de Farmacologia, Reus, Spain
| | - Maria-Rosa Nogués
- Universitat Rovira i Virgili, Departament de Ciències Mèdiques Bàsiques, Facultat de Medicina, Unitat de Farmacologia, Reus, Spain
| | - Jordi Camps
- Universitat Rovira i Virgili, Departament de Medicina i Cirurgia, Facultat de Medicina, Reus, Spain; Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d'investigació Sanitària Pere Virgili, Reus, Spain.
| | - Jorge Joven
- Universitat Rovira i Virgili, Departament de Medicina i Cirurgia, Facultat de Medicina, Reus, Spain; Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d'investigació Sanitària Pere Virgili, Reus, Spain; Campus of International Excellence Southern Catalonia, Tarragona, Spain.
| |
Collapse
|
47
|
Fang Y, Ji L, Zhu C, Xiao Y, Zhang J, Lu J, Yin J, Wei L. Liraglutide Alleviates Hepatic Steatosis by Activating the TFEB-Regulated Autophagy-Lysosomal Pathway. Front Cell Dev Biol 2020; 8:602574. [PMID: 33330497 PMCID: PMC7729067 DOI: 10.3389/fcell.2020.602574] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 11/10/2020] [Indexed: 12/14/2022] Open
Abstract
Liraglutide, a glucagon-like peptide-1 receptor agonist (GLP-1RA), has been demonstrated to alleviate non-alcoholic fatty liver disease (NAFLD). However, the underlying mechanism has not been fully elucidated. Increasing evidence suggests that autophagy is involved in the pathogenesis of hepatic steatosis. In this study, we examined whether liraglutide could alleviate hepatic steatosis through autophagy-dependent lipid degradation and investigated the underlying mechanisms. Herein, the effects of liraglutide on NAFLD were evaluated in a high-fat diet (HFD)-induced mouse model of NAFLD as well as in mouse primary and HepG2 hepatocytes exposed to palmitic acid (PA). The expression of the GLP-1 receptor (GLP-1R) was measured in vivo and in vitro. Oil red O staining was performed to detect lipid accumulation in hepatocytes. Electron microscopy was used to observe the morphology of autophagic vesicles and autolysosomes. Autophagic flux activity was measured by infecting HepG2 cells with mRFP-GFP-LC3 adenovirus. The roles of GLP-1R and transcription factor EB (TFEB) in autophagy-lysosomal activation were explored using small interfering RNA. Liraglutide treatment alleviated hepatic steatosis in vivo and in vitro. In models of hepatic steatosis, microtubule-associated protein 1B light chain-3-II (LC3-II) and SQSTM1/P62 levels were elevated in parallel to blockade of autophagic flux. Liraglutide treatment restored autophagic activity by improving lysosomal function. Furthermore, treatment with autophagy inhibitor chloroquine weakened liraglutide-induced autophagy activation and lipid degradation. TFEB has been identified as a key regulator of lysosome biogenesis and autophagy. The protein levels of nuclear TFEB and its downstream targets CTSB and LAMP1 were decreased in hepatocytes treated with PA, and these decreases were reversed by liraglutide treatment. Knockdown of TFEB expression compromised the effects of liraglutide on lysosome biogenesis and hepatic lipid accumulation. Mechanistically, GLP-1R expression was decreased in HFD mouse livers as well as PA-stimulated hepatocytes, and liraglutide treatment reversed the downregulation of GLP-1R expression in vivo and in vitro. Moreover, GLP-1R inhibition could mimic the effect of the TFEB downregulation-mediated decrease in lysosome biogenesis. Thus, our findings suggest that liraglutide attenuated hepatic steatosis via restoring autophagic flux, specifically the GLP-1R-TFEB-mediated autophagy-lysosomal pathway.
Collapse
Affiliation(s)
- Yunyun Fang
- Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Linlin Ji
- Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Chaoyu Zhu
- Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yuanyuan Xiao
- Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Jingjing Zhang
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
| | - Junxi Lu
- Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Jun Yin
- Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Department of Endocrinology and Metabolism, Shanghai Eighth People's Hospital, Shanghai, China
| | - Li Wei
- Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
48
|
Prolonged Lipid Accumulation in Cultured Primary Human Hepatocytes Rather Leads to ER Stress than Oxidative Stress. Int J Mol Sci 2020; 21:ijms21197097. [PMID: 32993055 PMCID: PMC7582586 DOI: 10.3390/ijms21197097] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 09/18/2020] [Accepted: 09/23/2020] [Indexed: 12/12/2022] Open
Abstract
Overweight has become a major health care problem in Western societies and is accompanied by an increasing incidence and prevalence of non-alcoholic fatty liver disease (NAFLD). The progression from NAFLD to non-alcoholic steatohepatitis (NASH) marks a crucial tipping point in the progression of severe and irreversible liver diseases. This study aims to gain further insight into the molecular processes leading to the evolution from steatosis to steatohepatitis. Steatosis was induced in cultures of primary human hepatocytes by continuous five-day exposure to free fatty acids (FFAs). The kinetics of lipid accumulation, lipotoxicity, and oxidative stress were measured. Additionally, ER stress was evaluated by analyzing the protein expression profiles of its key players: PERK, IRE1a, and ATF6a. Our data revealed that hepatocytes are capable of storing enormous amounts of lipids without showing signs of lipotoxicity. Prolonged lipid accumulation did not create an imbalance in hepatocyte redox homeostasis or a reduction in antioxidative capacity. However, we observed an FFA-dependent increase in ER stress, revealing thresholds for triggering the activation of pathways associated with lipid stress, inhibition of protein translation, and apoptosis. Our study clearly showed that even severe lipid accumulation can be attenuated by cellular defenses, but regenerative capacities may be reduced.
Collapse
|
49
|
Liu D, Liu NY, Chen LT, Shao Y, Shi XM, Zhu DY. Perfluorooctane sulfonate induced toxicity in embryonic stem cell-derived cardiomyocytes via inhibiting autophagy-lysosome pathway. Toxicol In Vitro 2020; 69:104988. [PMID: 32861759 DOI: 10.1016/j.tiv.2020.104988] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/22/2020] [Accepted: 08/24/2020] [Indexed: 01/07/2023]
Abstract
Perfluorooctane sulfonate (PFOS), a classic environmental pollutant, is reported to cause cardiotoxicity in animals and humans. It has been demonstrated that PFOS exposure down-regulates expression of cardiac-development related genes and proteins. However, the related mechanism of PFOS has not been fully elucidated. In the present study, the embryonic stem (ES) cells-derived cardiomyocytes (ESC-CMs) was employed to investigate PFOS-mediated mechanism in developmental toxicity of cardiomyocytes. Our previous study shows that PFOS induces cardiomyocyte toxicity via causing mitochondrial damage. Nevertheless, the underlying mechanism by which PFOS affects the autophagy-related mitochondrial toxicity in ESC-CMs remains unclear. Here, we found that PFOS induced the swelling of mitochondria and the autophagosome accumulation in ESC-CMs at 40 μM concentration. PFOS increased the levels of LC3-II, p62, and ubiquitinated proteins. PFOS also induced an increase of LC3 and p62 localization into mitochondria, indicating that mitophagy degradation was impaired. The results of autophagic flux using chloroquine and RFP-GFP-LC3 analysis showed that the accumulation of autophagosome was not caused by the formation but by the impaired degradation. PFOS was capable of blocking the fusion between autophagosome and lysosome. PFOS caused dysfunction of lysosomes because it down-regulated Lamp2a and cathepsin D, but it did not induced lysosome membrane permeabilization. Meanwhile, PFOS-mediated lysosomal function and the inhibitory effect of autophagic flux could be reversed by PP242 at 40 nM concentration, an mTOR inhibitor. Furthermore, PP242 restored PFOS-induced ATP depletion and mitochondrial membrane potential. In conclusion, PFOS induced mitochondrial dysfunction via blocking autophagy-lysosome degradation, leading to cardiomyocyte toxicity from ES cells.
Collapse
Affiliation(s)
- Dan Liu
- Institute of Pharmacology and Toxicology, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Nuo-Ya Liu
- Institute of Pharmacology and Toxicology, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Li-Ting Chen
- Institute of Pharmacology and Toxicology, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Ying Shao
- Institute of Pharmacology and Toxicology, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Xiao-Meng Shi
- Undergraduate Students in Research Training Project at Zhejiang University, Hangzhou 310058, China
| | - Dan-Yan Zhu
- Institute of Pharmacology and Toxicology, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China.
| |
Collapse
|
50
|
Zhou X, Fouda S, Li D, Zhang K, Ye JM. Involvement of the Autophagy-ER Stress Axis in High Fat/Carbohydrate Diet-Induced Nonalcoholic Fatty Liver Disease. Nutrients 2020; 12:nu12092626. [PMID: 32872238 PMCID: PMC7551457 DOI: 10.3390/nu12092626] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 08/07/2020] [Accepted: 08/26/2020] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease that can progress from simple hepatic steatosis to nonalcoholic steatohepatitis (NASH), and even further to liver cirrhosis or liver cancer. Overconsumption of high fat and/or carbohydrate are among the most common lifestyle factors that drive the development and progression of NAFLD. This review evaluates recent reports on the involvement of autophagy and endoplasmic reticulum (ER) stress in the pathogenesis of NAFLD. Here, we reveal a mechanism of an intrinsically linked axis of impaired autophagy and unresolved ER stress that mediates the development and progression of NAFLD resulting from the overconsumption of high fat and/or carbohydrate.
Collapse
Affiliation(s)
- Xiu Zhou
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China; (X.Z.); (D.L.); (K.Z.)
- International Healthcare Innovation Institute, Jiangmen 529040, China
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou 510006, China
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC 3083, Australia;
| | - Sherouk Fouda
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC 3083, Australia;
| | - Dongli Li
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China; (X.Z.); (D.L.); (K.Z.)
- International Healthcare Innovation Institute, Jiangmen 529040, China
| | - Kun Zhang
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China; (X.Z.); (D.L.); (K.Z.)
- International Healthcare Innovation Institute, Jiangmen 529040, China
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou 510006, China
| | - Ji-Ming Ye
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China; (X.Z.); (D.L.); (K.Z.)
- International Healthcare Innovation Institute, Jiangmen 529040, China
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC 3083, Australia;
- Correspondence: ; Tel.: +61-3-9925-7419; Fax: +61-3-9925-7178
| |
Collapse
|