1
|
Michie KL, Kunz HE, Dasari S, Lanza IR. The Influence of Aging on the Unfolded Protein Response in Human Skeletal Muscle at Rest and after Acute Exercise. Med Sci Sports Exerc 2024; 56:2135-2145. [PMID: 38934509 DOI: 10.1249/mss.0000000000003508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Abstract
BACKGROUND The unfolded protein response (UPR) is a proteostatic process that is activated in response to endoplasmic reticulum stress. It is currently unclear how aging influences the chronic and adaptive UPR in human skeletal muscle. Here we determined the effect of aging on UPR activation at rest, in response to exercise, and the associations with muscle function. METHODS Thirty young (20-35 yr) and 50 older (65-85 yr) individuals were enrolled. Vastus lateralis biopsies were performed at rest and 3 and 48 h after a single bout of resistance exercise. The abundance of UPR-related transcripts and proteins was measured by RNA sequencing and Western blotting, respectively. Fractional synthetic rates of muscle protein were determined by mass spectrometry after intravenous infusion of 13 C 6 phenylalanine. RESULTS Older adults demonstrated elevated transcriptional and proteomic markers of UPR activation in resting muscle. Resting UPR gene expression was negatively associated with muscle strength and power in older adults. The UPR is similarly activated by acute resistance exercise in young and older adults and positively associated with muscle function but not the anabolic response to exercise. CONCLUSIONS Skeletal muscle from older adults exhibits chronically activated UPR, which accompanies functional decline. The adaptive UPR is a proteostatic mechanism that is upregulated in response to exercise in young and older adults and positively associated with muscle function.
Collapse
Affiliation(s)
- Kelly L Michie
- Endocrine Research Unit, Division of Endocrinology, Mayo Clinic, Rochester, MN
| | - Hawley E Kunz
- Endocrine Research Unit, Division of Endocrinology, Mayo Clinic, Rochester, MN
| | - Surendra Dasari
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - Ian R Lanza
- Endocrine Research Unit, Division of Endocrinology, Mayo Clinic, Rochester, MN
| |
Collapse
|
2
|
Białek W, Hryniewicz-Jankowska A, Czechowicz P, Sławski J, Collawn JF, Czogalla A, Bartoszewski R. The lipid side of unfolded protein response. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159515. [PMID: 38844203 DOI: 10.1016/j.bbalip.2024.159515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/16/2024] [Accepted: 05/31/2024] [Indexed: 06/12/2024]
Abstract
Although our current knowledge of the molecular crosstalk between the ER stress, the unfolded protein response (UPR), and lipid homeostasis remains limited, there is increasing evidence that dysregulation of either protein or lipid homeostasis profoundly affects the other. Most research regarding UPR signaling in human diseases has focused on the causes and consequences of disrupted protein folding. The UPR itself consists of very complex pathways that function to not only maintain protein homeostasis, but just as importantly, modulate lipid biogenesis to allow the ER to adjust and promote cell survival. Lipid dysregulation is known to activate many aspects of the UPR, but the complexity of this crosstalk remains a major research barrier. ER lipid disequilibrium and lipotoxicity are known to be important contributors to numerous human pathologies, including insulin resistance, liver disease, cardiovascular diseases, neurodegenerative diseases, and cancer. Despite their medical significance and continuous research, however, the molecular mechanisms that modulate lipid synthesis during ER stress conditions, and their impact on cell fate decisions, remain poorly understood. Here we summarize the current view on crosstalk and connections between altered lipid metabolism, ER stress, and the UPR.
Collapse
Affiliation(s)
- Wojciech Białek
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | | | - Paulina Czechowicz
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Jakub Sławski
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - James F Collawn
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, USA
| | - Aleksander Czogalla
- Department of Cytobiochemistry, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Rafał Bartoszewski
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland.
| |
Collapse
|
3
|
Li M, Zhao B, Wang J, Zhang H, Yang Y, Song S, Psifidi A, Wu W, Loor JJ, Xu C. Caveolin 1 in bovine liver is associated with fatty acid-induced lipid accumulation and the ER unfolded protein response: role in fatty liver development. J Dairy Sci 2024:S0022-0302(24)01178-0. [PMID: 39343220 DOI: 10.3168/jds.2024-25349] [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: 06/27/2024] [Accepted: 08/29/2024] [Indexed: 10/01/2024]
Abstract
Disruption of endoplasmic reticulum (ER) homeostasis, i.e., ER stress, is intrinsically linked with lipid metabolism disorders in dairy cows. Caveolin 1 (CAV1) is a ubiquitously-expressed membrane-associated scaffolding protein involved in regulating the secretory pathway within the ER. Whether inhibiting the activity of CAV1 affects the ER and its potential role in hepatic lipid deposition in dairy cows is unknown. Biopsies of liver tissue from Holstein cows (days in milk: median = 13 d, range = 5 to 21) diagnosed as healthy (n = 6, hepatic TAG levels < 1%, milk production: median = 38.9 kg/day, Interquartile range = 38.0 and 40.8) or suffering from fatty liver (n = 6, hepatic TAG levels > 5%, milk production: median = 36.6 kg/day, Interquartile range = 35.7 and 38.1) revealed that fatty liver was associated with lower abundance of CAV1 gene and protein, higher phosphorylation (p) levels of PERK and IRE1α, and increased abundance of ATF6, GRP78, CHOP protein, and several unfolded protein response (UPR) genes (ATF4, sXBP1, and GRP78). Proteins related to de novo fatty acid synthesis, including ACC1, SREBP-1c, PPARγ, and downstream targets genes of SREBP1 (ACACA and FASN) also had greater abundance. This in vivo analysis highlighted a mechanistic link between CAV1 protein abundance, ER stress, and lipid metabolism during fatty liver. A mechanistic study was then performed in vitro with primary hepatocytes isolated from 5 healthy calves (weight, 40-45 kg; 1 d old). Initially, hepatocytes were treated with FFA (1.2 mM) for 1, 3, 6, or 12 h. FFA treatment reduced CAV1 protein abundance linearly while it reduced abundance of ER stress-related proteins, p-IRE1α, p-PERK, GRP78, ATF6, and CHOP. Proteins related to de novo fatty acid synthesis (ACC1, SREBP-1c, PPARγ) also increased linearly, and lipid droplets accumulated progressively over time following FFA treatment. Subsequently, to assess the role of CAV1 in FFA-induced ER stress and de novo fatty acid synthesis, hepatocytes were transfected with pCMV-CAV1 (cattle)-3 × FLAG-Neo (pc-CAV1) plasmid to overexpress CAV1 or with siRNA to silence CAV1 (siCAV1) transcription. Overexpression of CAV1 alleviated ER stress by reducing levels of p-PERK and p-IRE1α, as well as the protein abundance of ATF6, GRP78, CHOP, and several UPR genes (GRP78, ATF4, and sXBP1). Similarly, CAV1 overexpression decreased protein abundance of ACC1, SREBP-1c, PPARγ, and downstream targets genes of SREBP1 (ACACA and FASN). Conversely, silencing CAV1 exacerbated FFA-induced ER stress and de novo fatty acid synthesis. Considering the negative role of FFA-induced ER stress on lipid accumulation in hepatocytes, a second in vitro experiment involved hepatocytes treated with 0.5 μg/mL tunicamycin (TM, a typical ER stress inducer) for 24 h with or without overexpressing CAV1 (pc-CAV1). Overexpressing caveolin 1 reversed TM-induced increases in mRNA and protein associated with ER stress and de novo fatty acid synthesis. Furthermore, use of hepatocytes transfected with pc-CAV1 for 48 h and subjected to co-immunoprecipitation revealed that CAV1 interacts with IRE1α and ATF6. Overall, the data suggest that CAV1 may help reduce hepatic ER stress and mitigate fatty acid synthesis by binding to and inhibiting IRE1α and ATF6 signaling.
Collapse
Affiliation(s)
- Ming Li
- College of Veterinary Medicine, China Agricultural University, 100193, Beijing, China
| | - Bichen Zhao
- College of Veterinary Medicine, China Agricultural University, 100193, Beijing, China
| | - Jingyi Wang
- College of Veterinary Medicine, China Agricultural University, 100193, Beijing, China
| | - Huijing Zhang
- College of Veterinary Medicine, China Agricultural University, 100193, Beijing, China
| | - Yue Yang
- College of Veterinary Medicine, China Agricultural University, 100193, Beijing, China
| | - Shihao Song
- College of Veterinary Medicine, China Agricultural University, 100193, Beijing, China
| | - Androniki Psifidi
- Department of Clinical Science and Services, Royal Veterinary College, North Mymms, Hertfordshire, AL9 7TA, United Kingdom
| | - Wenda Wu
- School of Food and Biological Engineering, University of Technology, Hefei 230009, China
| | - Juan J Loor
- Mammalian NutriPhysio Genomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - Chuang Xu
- College of Veterinary Medicine, China Agricultural University, 100193, Beijing, China..
| |
Collapse
|
4
|
Harada N, Yoshikatsu A, Yamamoto H, Nakaya Y. 2-Deoxy-D-Glucose Downregulates Fatty Acid Synthase Gene Expression Via an Endoplasmic Reticulum Stress-Dependent Pathway in HeLa Cells. Cell Biochem Biophys 2024; 82:2285-2296. [PMID: 38824236 DOI: 10.1007/s12013-024-01339-0] [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] [Accepted: 05/24/2024] [Indexed: 06/03/2024]
Abstract
Fatty acid synthase (FASN) catalyzes the rate-limiting step of cellular lipogenesis. FASN expression is upregulated in various types of cancer cells, implying that FASN is a potential target for cancer therapy. 2-Deoxy-D-glucose (2-DG) specifically targets cancer cells by inhibiting glycolysis and glucose metabolism, resulting in multiple anticancer effects. However, whether the effects of 2-DG involve lipogenic metabolism remains to be elucidated. We investigated the effect of 2-DG administration on FASN expression in HeLa human cervical cancer cells. 2-DG treatment for 24 h decreased FASN mRNA and protein levels and suppressed the activity of an exogenous rat Fasn promoter. The use of a chemical activator or inhibitors or of a mammalian expression plasmid showed that neither AMPK nor the Sp1 transcription factor is responsible for the inhibitory effect of 2-DG on FASN expression. Administration of thapsigargin, an endoplasmic reticulum (ER) stress inducer, or 4-(2-aminoethyl) benzenesulfonyl fluoride (AEBSF), a site 1 protease inhibitor, mimicked the inhibitory effect of 2-DG on FASN expression. 2-DG did not further decrease FASN expression in the presence of thapsigargin or AEBSF. Site 1 protease mediates activation of ATF6, an ER stress mediator, as well as sterol regulatory element-binding protein 1 (SREBP1), a robust transcription factor for FASN. Administration of 2-DG or thapsigargin for 24 h suppressed activation of ATF6 and SREBP1, as did AEBSF. We speculated that these effects of 2-DG or thapsigargin are due to feedback inhibition via increased GRP78 expression following ER stress. Supporting this, exogenous overexpression of GRP78 in HeLa cells suppressed SREBP1 activation and Fasn promoter activity. These results suggest that 2-DG suppresses FASN expression via an ER stress-dependent pathway, providing new insight into the molecular basis of FASN regulation in cancer.
Collapse
Affiliation(s)
- Nagakatsu Harada
- Department of Health and Nutrition, Faculty of Nursing and Nutrition, The University of Shimane, 151 Nishihayashigi, Izumo city, 693-8550, Shimane, Japan.
| | - Aya Yoshikatsu
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15, Kuramoto-cho, Tokushima city, 770-8503, Tokushima, Japan
| | - Hironori Yamamoto
- Department of Health and Nutrition, Faculty of Human Life, Jin-ai University, 3-1-1 Ohde-cho, Echizen city, 915-8568, Fukui, Japan
| | - Yutaka Nakaya
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15, Kuramoto-cho, Tokushima city, 770-8503, Tokushima, Japan
| |
Collapse
|
5
|
Yang Y, Zhang X, Zhao Q, Zhang J, Lou X. Compromised COPII vesicle trafficking leads to glycogenic hepatopathy. Dis Model Mech 2024; 17:dmm050748. [PMID: 39139065 PMCID: PMC11463966 DOI: 10.1242/dmm.050748] [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: 02/06/2024] [Accepted: 08/06/2024] [Indexed: 08/15/2024] Open
Abstract
Being a vital cellular process, coat protein complex II (COPII) vesicle trafficking has been found to play a crucial role in liver metabolism. However, its functions and the underlying mechanisms in systemic metabolic homeostasis have not been fully understood. Here, with a newly identified gene trap zebrafish line (sec31anju221), we show that compromised COPII vesicle trafficking leads to biphasic abnormal hepatic metabolism. During the larval stage, deficiency of COPII-mediated trafficking leads to activation of the unfolded protein response and the development of hepatic steatosis. By using epistasis analysis, we found that the eIF2α-ATF4 pathway serves as the primary effector for liver steatosis. In adult sec31anju221 fish, the hepatosteatosis was reversed and the phenotype switched to glycogenic hepatopathy. Proteomic profiling and biochemical assays indicate that sec31anju221 fish are in a state of hypothyroidism. Moreover, our study shows that thyroid hormone treatment alleviates the metabolic defects. This study provides insights into processes of liver diseases associated with vesicle trafficking impairments and expands our understanding of the pathological interplay between thyroid and liver.
Collapse
Affiliation(s)
- Yuxi Yang
- Medical School, Nanjing University, Nanjing 210093, China
| | - Xue Zhang
- Research Center for Life Sciences Computing, Zhejiang Laboratory, Hangzhou 311100, China
| | - Qingshun Zhao
- Medical School, Nanjing University, Nanjing 210093, China
| | - Jingzi Zhang
- Medical School, Nanjing University, Nanjing 210093, China
| | - Xin Lou
- Research Center for Life Sciences Computing, Zhejiang Laboratory, Hangzhou 311100, China
| |
Collapse
|
6
|
Sabinari I, Horakova O, Cajka T, Kleinova V, Wieckowski MR, Rossmeisl M. Influence of Lipid Class Used for Omega-3 Fatty Acid Supplementation on Liver Fat Accumulation in MASLD. Physiol Res 2024; 73:S295-S320. [PMID: 39016154 PMCID: PMC11412347 DOI: 10.33549/physiolres.935396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) occurs in subjects with obesity and metabolic syndrome. MASLD may progress from simple steatosis (i.e., hepatic steatosis) to steatohepatitis, characterized by inflammatory changes and liver cell damage, substantially increasing mortality. Lifestyle measures associated with weight loss and/or appropriate diet help reduce liver fat accumulation, thereby potentially limiting progression to steatohepatitis. As for diet, both total energy and macronutrient composition significantly influence the liver's fat content. For example, the type of dietary fatty acids can affect the metabolism of lipids and hence their tissue accumulation, with saturated fatty acids having a greater ability to promote fat storage in the liver than polyunsaturated ones. In particular, polyunsaturated fatty acids of n-3 series (omega-3), such as docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA), have been intensively studied for their antisteatotic effects, both in preclinical animal models of obesity and hepatic steatosis and in overweight/obese patients. Their effects may depend not only on the dose and duration of administration of omega-3, or DHA/EPA ratio, but also on the lipid class used for their supplementation. This review summarizes the available evidence from recent comparative studies using omega-3 supplementation via different lipid classes. Albeit the evidence is mainly limited to preclinical studies, it suggests that phospholipids and possibly wax esters could provide greater efficacy against MASLD compared to traditional chemical forms of omega-3 supplementation (i.e., triacylglycerols, ethyl esters). This cannot be attributed solely to improved EPA and/or DHA bioavailability, but other mechanisms may be involved. Keywords: MASLD • Metabolic dysfunction-associated steatotic liver disease • NAFLD • Non-alcoholic fatty liver disease • n-3 polyunsaturated fatty acids.
Collapse
Affiliation(s)
- I Sabinari
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic.
| | | | | | | | | | | |
Collapse
|
7
|
García-Juárez M, García-Rodríguez A, Cruz-Carrillo G, Flores-Maldonado O, Becerril-Garcia M, Garza-Ocañas L, Torre-Villalvazo I, Camacho-Morales A. Intermittent Fasting Improves Social Interaction and Decreases Inflammatory Markers in Cortex and Hippocampus. Mol Neurobiol 2024:10.1007/s12035-024-04340-z. [PMID: 39002056 DOI: 10.1007/s12035-024-04340-z] [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: 12/15/2023] [Accepted: 06/28/2024] [Indexed: 07/15/2024]
Abstract
Autism spectrum disorder (ASD) is a psychiatric condition characterized by reduced social interaction, anxiety, and stereotypic behaviors related to neuroinflammation and microglia activation. We demonstrated that maternal exposure to Western diet (cafeteria diet or CAF) induced microglia activation, systemic proinflammatory profile, and ASD-like behavior in the offspring. Here, we aimed to identify the effect of alternate day fasting (ADF) as a non-pharmacologic strategy to modulate neuroinflammation and ASD-like behavior in the offspring prenatally exposed to CAF diet. We found that ADF increased plasma beta-hydroxybutyrate (BHB) levels in the offspring exposed to control and CAF diets but not in the cortex (Cx) and hippocampus (Hpp). We observed that ADF increased the CD45 + cells in Cx of both groups; In control individuals, ADF promoted accumulation of CD206 + microglia cells in choroid plexus (CP) and increased in CD45 + macrophages cells and lymphocytes in the Cx. Gestational exposure to CAF diet promoted defective sociability in the offspring; ADF improved social interaction and increased microglia CD206 + in the Hpp and microglia complexity in the dentate gyrus. Additionally, ADF led to attenuation of the ER stress markers (Bip/ATF6/p-JNK) in the Cx and Hpp. Finally, biological modeling showed that fasting promotes higher microglia complexity in Cx, which is related to improvement in social interaction, whereas in dentate gyrus sociability is correlated with less microglia complexity. These data suggest a contribution of intermittent fasting as a physiological stimulus capable of modulating microglia phenotype and complexity in the brain, and social interaction in male mice.
Collapse
Affiliation(s)
- Martín García-Juárez
- Facultad de Medicina, Departamento de Bioquímica, Universidad Autónoma de Nuevo León, Madero y Dr. Aguirre Pequeño. Col. Mitras Centro, C.P. 64460, Monterrey, Nuevo León, Mexico
- Centro de Investigación y Desarrollo en Ciencias de La Salud, Universidad Autónoma de Nuevo León, Unidad de Neurometabolismo, Monterrey, Nuevo León, Mexico
| | - Adamary García-Rodríguez
- Facultad de Medicina, Departamento de Bioquímica, Universidad Autónoma de Nuevo León, Madero y Dr. Aguirre Pequeño. Col. Mitras Centro, C.P. 64460, Monterrey, Nuevo León, Mexico
- Centro de Investigación y Desarrollo en Ciencias de La Salud, Universidad Autónoma de Nuevo León, Unidad de Neurometabolismo, Monterrey, Nuevo León, Mexico
| | - Gabriela Cruz-Carrillo
- Facultad de Medicina, Departamento de Bioquímica, Universidad Autónoma de Nuevo León, Madero y Dr. Aguirre Pequeño. Col. Mitras Centro, C.P. 64460, Monterrey, Nuevo León, Mexico
- Centro de Investigación y Desarrollo en Ciencias de La Salud, Universidad Autónoma de Nuevo León, Unidad de Neurometabolismo, Monterrey, Nuevo León, Mexico
| | - Orlando Flores-Maldonado
- Facultad de Medicina, Departamento de Microbiología, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, Mexico
| | - Miguel Becerril-Garcia
- Facultad de Medicina, Departamento de Microbiología, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, Mexico
| | - Lourdes Garza-Ocañas
- Department of Pharmacology and Toxicology, College of Medicine, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, México
| | - Ivan Torre-Villalvazo
- Departamento de Fisiología de La Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), 14080, Mexico City, Mexico
| | - Alberto Camacho-Morales
- Facultad de Medicina, Departamento de Bioquímica, Universidad Autónoma de Nuevo León, Madero y Dr. Aguirre Pequeño. Col. Mitras Centro, C.P. 64460, Monterrey, Nuevo León, Mexico.
- Centro de Investigación y Desarrollo en Ciencias de La Salud, Universidad Autónoma de Nuevo León, Unidad de Neurometabolismo, Monterrey, Nuevo León, Mexico.
- College of Medicine, Universidad Autónoma de Nuevo Leon, San Nicolás de los Garza, NL, Mexico.
| |
Collapse
|
8
|
Qian Q, Li M, Zhang Z, Davis SW, Rahmouni K, Norris AW, Cao H, Ding WX, Hotamisligil GS, Yang L. Obesity disrupts the pituitary-hepatic UPR communication leading to NAFLD progression. Cell Metab 2024; 36:1550-1565.e9. [PMID: 38718793 PMCID: PMC11222033 DOI: 10.1016/j.cmet.2024.04.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 03/05/2024] [Accepted: 04/17/2024] [Indexed: 07/05/2024]
Abstract
Obesity alters levels of pituitary hormones that govern hepatic immune-metabolic homeostasis, dysregulation of which leads to nonalcoholic fatty liver disease (NAFLD). However, the impact of obesity on intra-pituitary homeostasis is largely unknown. Here, we uncovered a blunted unfolded protein response (UPR) but elevated inflammatory signatures in pituitary glands of obese mice and humans. Furthermore, we found that obesity inflames the pituitary gland, leading to impaired pituitary inositol-requiring enzyme 1α (IRE1α)-X-box-binding protein 1 (XBP1) UPR branch, which is essential for protecting against pituitary endocrine defects and NAFLD progression. Intriguingly, pituitary IRE1-deletion resulted in hypothyroidism and suppressed the thyroid hormone receptor B (THRB)-mediated activation of Xbp1 in the liver. Conversely, activation of the hepatic THRB-XBP1 axis improved NAFLD in mice with pituitary UPR defect. Our study provides the first evidence and mechanism of obesity-induced intra-pituitary cellular defects and the pathophysiological role of pituitary-liver UPR communication in NAFLD progression.
Collapse
Affiliation(s)
- Qingwen Qian
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Mark Li
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Zeyuan Zhang
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Shannon W Davis
- Department of Biological Sciences, College of Arts and Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Kamal Rahmouni
- Department of Neuroscience and Pharmacology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Andrew W Norris
- Division of Endocrinology and Diabetes, Department of Pediatrics, Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Huojun Cao
- Iowa Institute for Oral Health Research, Division of Biostatistics and Computational Biology, Department of Endodontics, University of Iowa College of Dentistry, Iowa City, IA 52242, USA
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Gökhan S Hotamisligil
- Sabri Ülker Center for Metabolic Research, Department of Molecular Metabolism, Harvard T.H. School of Public Health, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02141, USA
| | - Ling Yang
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA.
| |
Collapse
|
9
|
Bidooki SH, Barranquero C, Sánchez-Marco J, Martínez-Beamonte R, Rodríguez-Yoldi MJ, Navarro MA, Fernandes SCM, Osada J. TXNDC5 Plays a Crucial Role in Regulating Endoplasmic Reticulum Activity through Different ER Stress Signaling Pathways in Hepatic Cells. Int J Mol Sci 2024; 25:7128. [PMID: 39000233 PMCID: PMC11241358 DOI: 10.3390/ijms25137128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
The pathogenesis of non-alcoholic fatty liver disease (NAFLD) is influenced by a number of variables, including endoplasmic reticulum stress (ER). Thioredoxin domain-containing 5 (TXNDC5) is a member of the protein disulfide isomerase family and acts as an endoplasmic reticulum (ER) chaperone. Nevertheless, the function of TXNDC5 in hepatocytes under ER stress remains largely uncharacterized. In order to identify the role of TXNDC5 in hepatic wild-type (WT) and TXNDC5-deficient (KO) AML12 cell lines, tunicamycin, palmitic acid, and thapsigargin were employed as stressors. Cell viability, mRNA, protein levels, and mRNA splicing were then assayed. The protein expression results of prominent ER stress markers indicated that the ERN1 and EIF2AK3 proteins were downregulated, while the HSPA5 protein was upregulated. Furthermore, the ATF6 protein demonstrated no significant alterations in the absence of TXNDC5 at the protein level. The knockout of TXNDC5 has been demonstrated to increase cellular ROS production and its activity is required to maintain normal mitochondrial function during tunicamycin-induced ER stress. Tunicamycin has been observed to disrupt the protein levels of HSPA5, ERN1, and EIF2AK3 in TXNDC5-deficient cells. However, palmitic acid has been observed to disrupt the protein levels of ATF6, HSPA5, and EIF2AK3. In conclusion, TXNDC5 can selectively activate distinct ER stress pathways via HSPA5, contingent on the origin of ER stress. Conversely, the absence of TXNDC5 can disrupt the EIF2AK3 cascade.
Collapse
Affiliation(s)
- Seyed Hesamoddin Bidooki
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, E-50013 Zaragoza, Spain; (S.H.B.); (J.S.-M.); (R.M.-B.); (M.A.N.)
- Instituto Agroalimentario de Aragón, CITA, Universidad de Zaragoza, E-50013 Zaragoza, Spain; (C.B.); (M.J.R.-Y.)
- Institute of Analytical Sciences and Physico-Chemistry for Environment and Materials (IPREM), Universite de Pau et des Pays de l’Adour, E2S UPPA, CNRS, 64 000 Pau, France;
- MANTA—Marine Materials Research Group, Universite de Pau et des Pays de l’Adour, E2S UPPA, 64 600 Anglet, France
| | - Cristina Barranquero
- Instituto Agroalimentario de Aragón, CITA, Universidad de Zaragoza, E-50013 Zaragoza, Spain; (C.B.); (M.J.R.-Y.)
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Javier Sánchez-Marco
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, E-50013 Zaragoza, Spain; (S.H.B.); (J.S.-M.); (R.M.-B.); (M.A.N.)
| | - Roberto Martínez-Beamonte
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, E-50013 Zaragoza, Spain; (S.H.B.); (J.S.-M.); (R.M.-B.); (M.A.N.)
- Instituto Agroalimentario de Aragón, CITA, Universidad de Zaragoza, E-50013 Zaragoza, Spain; (C.B.); (M.J.R.-Y.)
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - María J. Rodríguez-Yoldi
- Instituto Agroalimentario de Aragón, CITA, Universidad de Zaragoza, E-50013 Zaragoza, Spain; (C.B.); (M.J.R.-Y.)
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
- Departamento de Farmacología, Fisiología, Medicina Legal y Forense, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, E-50013 Zaragoza, Spain
| | - María A. Navarro
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, E-50013 Zaragoza, Spain; (S.H.B.); (J.S.-M.); (R.M.-B.); (M.A.N.)
- Instituto Agroalimentario de Aragón, CITA, Universidad de Zaragoza, E-50013 Zaragoza, Spain; (C.B.); (M.J.R.-Y.)
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Susana C. M. Fernandes
- Institute of Analytical Sciences and Physico-Chemistry for Environment and Materials (IPREM), Universite de Pau et des Pays de l’Adour, E2S UPPA, CNRS, 64 000 Pau, France;
- MANTA—Marine Materials Research Group, Universite de Pau et des Pays de l’Adour, E2S UPPA, 64 600 Anglet, France
| | - Jesús Osada
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, E-50013 Zaragoza, Spain; (S.H.B.); (J.S.-M.); (R.M.-B.); (M.A.N.)
- Instituto Agroalimentario de Aragón, CITA, Universidad de Zaragoza, E-50013 Zaragoza, Spain; (C.B.); (M.J.R.-Y.)
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| |
Collapse
|
10
|
Souza-Tavares H, Santana-Oliveira DA, Vasques-Monteiro IML, Silva-Veiga FM, Mandarim-de-Lacerda CA, Souza-Mello V. Exercise enhances hepatic mitochondrial structure and function while preventing endoplasmic reticulum stress and metabolic dysfunction-associated steatotic liver disease in mice fed a high-fat diet. Nutr Res 2024; 126:180-192. [PMID: 38759501 DOI: 10.1016/j.nutres.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/15/2024] [Accepted: 04/15/2024] [Indexed: 05/19/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) has attracted increasing attention from the scientific community because of its severe but silent progression and the lack of specific treatment. Glucolipotoxicity triggers endoplasmic reticulum (ER) stress with decreased beta-oxidation and enhanced lipogenesis, promoting the onset of MASLD, whereas regular physical exercise can prevent MASLD by preserving ER and mitochondrial function. Thus, the hypothesis of this study was that high-intensity interval training (HIIT) could prevent the development of MASLD in high-fat (HF)-fed C57BL/6J mice by maintaining insulin sensitivity, preventing ER stress, and promoting beta-oxidation. Forty male C57BL/6J mice (3 months old) comprised 4 experimental groups: the control (C) diet group, the C diet + HIIT (C-HIIT) group, the HF diet group, and the HF diet + HIIT (HF-HIIT) group. HIIT sessions lasted 12 minutes and were performed 3 times weekly by trained mice. The diet and exercise protocols lasted for 10 weeks. The HIIT protocol prevented weight gain and maintained insulin sensitivity in the HF-HIIT group. A chronic HF diet increased ER stress-related gene and protein expression, but HIIT helped to maintain ER homeostasis, preserve mitochondrial ultrastructure, and maximize beta-oxidation. The increased sirtuin-1/peroxisome proliferator-activated receptor-gamma coactivator 1-alpha expression implies that HIIT enhanced mitochondrial biogenesis and yielded adequate mitochondrial dynamics. High hepatic fibronectin type III domain containing 5/irisin agreed with the antilipogenic and anti-inflammatory effects observed in the HF-HIIT group, reinforcing the antisteatotic effects of HIIT. Thus, we confirmed that practicing HIIT 3 times per week maintained insulin sensitivity, prevented ER stress, and enhanced hepatic beta-oxidation, impeding MASLD development in this mouse model even when consuming high energy intake from saturated fatty acids.
Collapse
Affiliation(s)
- Henrique Souza-Tavares
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology. Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Daiana Araujo Santana-Oliveira
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology. Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Isabela Macedo Lopes Vasques-Monteiro
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology. Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Flavia Maria Silva-Veiga
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology. Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Carlos Alberto Mandarim-de-Lacerda
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology. Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Vanessa Souza-Mello
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology. Rio de Janeiro State University, Rio de Janeiro, Brazil.
| |
Collapse
|
11
|
Derler M, Teubenbacher T, Carapuig A, Nieswandt B, Fessler J, Kolb D, Mussbacher M. Platelets induce endoplasmic reticulum stress in macrophages in vitro. J Thromb Haemost 2024; 22:1475-1488. [PMID: 38278417 DOI: 10.1016/j.jtha.2024.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 12/23/2023] [Accepted: 01/15/2024] [Indexed: 01/28/2024]
Abstract
BACKGROUND Endoplasmic reticulum (ER) stress is a key feature of lipid-laden macrophages and contributes to the development of atherosclerotic plaques. Blood platelets are known to interact with macrophages and fine-tune effector functions such as inflammasome activation and phagocytosis. However, the effect of platelets on ER stress induction is unknown. OBJECTIVES The objective of this study is to elucidate the potential of platelets in regulating ER stress in macrophages in vitro. METHODS Bone marrow-derived macrophages and RAW 264.7 cells were incubated with isolated murine platelets, and ER stress and inflammation markers were determined by reverse transcription-quantitative polymerase chain reaction, Western blotting, and enzyme-linked immunosorbent assay. ER morphology was investigated by electron microscopy. Cell viability, lipid accumulation, and activation were measured by flow cytometry. To gain mechanistic insights, coincubation experiments were performed with platelet decoys/releasates as well as lipopolysaccharide, blocking antibodies, and TLR4 inhibitors. RESULTS Coincubation of platelets and macrophages led to elevated levels of ER stress markers (BIP, IRE1α, CHOP, and XBP1 splicing) in murine and human macrophages, which led to a pronounced enlargement of the ER. Macrophage ER stress was accompanied by increased release of proinflammatory cytokines and intracellular lipid accumulation, but not cell death. Platelet decoys, but not platelet releasates or lysate from other cells, phenocopied the effect of platelets. Blocking TLR4 inhibited inflammatory activation of macrophages but did not affect ER stress induction by platelet coincubation. CONCLUSION To our knowledge, this study is the first to demonstrate that platelets induce ER stress and unfolded protein response in macrophages by heat-sensitive membrane proteins, independent of inflammatory activation of macrophages.
Collapse
Affiliation(s)
- Martina Derler
- Department of Pharmacology and Toxicology, Institute of Pharmaceutical Sciences, University of Graz, Graz, Austria
| | - Theresa Teubenbacher
- Department of Pharmacology and Toxicology, Institute of Pharmaceutical Sciences, University of Graz, Graz, Austria
| | - Anna Carapuig
- Department of Pharmacology and Toxicology, Institute of Pharmaceutical Sciences, University of Graz, Graz, Austria
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine, Chair of Experimental Biomedicine I, University Hospital Würzburg, Würzburg, Germany; Rudolf Virchow Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Johannes Fessler
- Division of Immunology and Pathophysiology, Medical University of Graz, Graz, Austria
| | - Dagmar Kolb
- Center for Medical Research, Core Facility Ultrastructure Analysis, Medical University of Graz, Graz, Austria; Division of Cell Biology, Histology, and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria
| | - Marion Mussbacher
- Department of Pharmacology and Toxicology, Institute of Pharmaceutical Sciences, University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria.
| |
Collapse
|
12
|
Miao K, Zhao Y, Xue N. Gkongensin A, an HSP90β inhibitor, improves hyperlipidemia, hepatic steatosis, and insulin resistance. Heliyon 2024; 10:e29367. [PMID: 38655315 PMCID: PMC11036013 DOI: 10.1016/j.heliyon.2024.e29367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 03/29/2024] [Accepted: 04/07/2024] [Indexed: 04/26/2024] Open
Abstract
The prevalence of obesity and its primary associated comorbidities, such as type 2 diabetes and fatty liver disease, has reached epidemic proportions, with no successful treatment available at present. Heat shock protein 90 (HSP90), a crucial chaperone, plays a key role in de novo lipogenesis (DNL) by stabilizing and maintaining sterol regulatory element binding protein (SREBP) activity. Kongensin A (KA), derived from Croton kongensis, inhibits RIP3-mediated necrosis, showing promise as an anti-necrotic and anti-inflammatory agent. It is not yet clear if KA, acting as an HSP90 inhibitor, can enhance hyperlipidemia, hepatic steatosis, and insulin resistance in obese individuals by controlling lipid metabolism. In this study, we first found that KA can potentially decrease lipid content at the cellular level. C57BL/6J mice were given a high-fat diet (HFD) and received KA and lovastatin through oral administration for 7 weeks. KA improved hyperlipidemia, fatty liver, and insulin resistance, as well as reduced body weight in diet-induced obese (DIO) mice, with no significant alteration in food intake. In vitro, KA suppressed DNL and reduced the amounts of mSREBPs. KA promoted mSREBP degradation via the FBW7-mediated ubiquitin-proteasome pathway. KA decreased the level of p-Akt Ser308, and p-GSK3β Ser9 by inhibiting the interaction between HSP90β and Akt. Overall, KA enhanced hyperlipidemia, hepatic steatosis, and insulin resistance by blocking SREBP activity, thereby impacting the FBW7-controlled ubiquitin-proteasome pathway.
Collapse
Affiliation(s)
- Kun Miao
- Department of Hand Surgery, Fuzhou Second General Hospital, 350007, Fuzhou, Fujian, China
| | - Yawei Zhao
- Department of Pharmacy, Jurong Hospital Affiliated to Jiangsu University, Jurong, 212400, Jiangsu, China
| | - Ning Xue
- Department of Acupuncture, Jurong Hospital Affiliated to Jiangsu University, Jurong, 212400, Jiangsu, China
| |
Collapse
|
13
|
Kleiboeker B, He A, Tan M, Lu D, Hu D, Liu X, Goodarzi P, Hsu FF, Razani B, Semenkovich CF, Lodhi IJ. Adipose tissue peroxisomal lipid synthesis orchestrates obesity and insulin resistance through LXR-dependent lipogenesis. Mol Metab 2024; 82:101913. [PMID: 38458567 PMCID: PMC10950804 DOI: 10.1016/j.molmet.2024.101913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 02/29/2024] [Accepted: 03/04/2024] [Indexed: 03/10/2024] Open
Abstract
OBJECTIVE Adipose tissue mass is maintained by a balance between lipolysis and lipid storage. The contribution of adipose tissue lipogenesis to fat mass, especially in the setting of high-fat feeding, is considered minor. Here we investigated the effect of adipose-specific inactivation of the peroxisomal lipid synthetic protein PexRAP on fatty acid synthase (FASN)-mediated lipogenesis and its impact on adiposity and metabolic homeostasis. METHODS To explore the role of PexRAP in adipose tissue, we metabolically phenotyped mice with adipose-specific knockout of PexRAP. Bulk RNA sequencing was used to determine transcriptomic responses to PexRAP deletion and 14C-malonyl CoA allowed us to measure de novo lipogenic activity in adipose tissue of these mice. In vitro cell culture models were used to elucidate the mechanism of cellular responses to PexRAP deletion. RESULTS Adipose-specific PexRAP deletion promoted diet-induced obesity and insulin resistance through activation of de novo lipogenesis. Mechanistically, PexRAP inactivation inhibited the flux of carbons to ethanolamine plasmalogens. This increased the nuclear PC/PE ratio and promoted cholesterol mislocalization, resulting in activation of liver X receptor (LXR), a nuclear receptor known to be activated by increased intracellular cholesterol. LXR activation led to increased expression of the phospholipid remodeling enzyme LPCAT3 and induced FASN-mediated lipogenesis, which promoted diet-induced obesity and insulin resistance. CONCLUSIONS These studies reveal an unexpected role for peroxisome-derived lipids in regulating LXR-dependent lipogenesis and suggest that activation of lipogenesis, combined with dietary lipid overload, exacerbates obesity and metabolic dysregulation.
Collapse
Affiliation(s)
- Brian Kleiboeker
- Division of Endocrinology, Metabolism & Lipid Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Anyuan He
- Division of Endocrinology, Metabolism & Lipid Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Min Tan
- Division of Endocrinology, Metabolism & Lipid Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Dongliang Lu
- Division of Endocrinology, Metabolism & Lipid Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Donghua Hu
- Division of Endocrinology, Metabolism & Lipid Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Xuejing Liu
- Division of Endocrinology, Metabolism & Lipid Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Parniyan Goodarzi
- Division of Endocrinology, Metabolism & Lipid Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Fong-Fu Hsu
- Division of Endocrinology, Metabolism & Lipid Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Babak Razani
- Cardiovascular Division, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Clay F Semenkovich
- Division of Endocrinology, Metabolism & Lipid Research, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Irfan J Lodhi
- Division of Endocrinology, Metabolism & Lipid Research, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
14
|
Mohammadpour-Asl S, Roshan-Milani B, Roshan-Milani S, Saboory E, Ghobadian B, Chodari L. Endoplasmic reticulum stress PERK-ATF4-CHOP pathway is involved in non-alcoholic fatty liver disease in type 1 diabetic rats: The rescue effect of treatment exercise and insulin-like growth factor I. Heliyon 2024; 10:e27225. [PMID: 38468961 PMCID: PMC10926145 DOI: 10.1016/j.heliyon.2024.e27225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 02/23/2024] [Accepted: 02/26/2024] [Indexed: 03/13/2024] Open
Abstract
Endoplasmic Reticulum Stress (ERS) is a key factor in the development of Non-Alcoholic Fatty Liver Disease (NAFLD) in diabetes. The current study aimed to examine the effects of exercise and IGF-I on ERS markers in liver tissue. Rats were divided into five groups (n = 8 per group), including control (CON), diabetes (DIA), diabetes + exercise (DIA + EX), diabetes + IGF-I (DIA + IGF-I), and diabetes + exercise + IGF-I (DIA + EX + IGF-I). Type 1 diabetes was induced by an I.P. injection of streptozotocin (60 mg/kg). After 30 days of treatment with exercise or IGF-I alone or in combination, liver tissue was assessed for caspase 12, 8, and CHOP protein levels, and expression of ERS markers (ATF-6, PERK, IRE-1A) and lipid metabolism-involved genes (FAS, FXR, SREBP-1c) by western immunoblotting. In addition, for the evaluation of histopathological changes in the liver, Hematoxylin - Eosin and Masson's Trichrome staining were done. Compared to the control group, diabetes significantly caused liver fibrosis, induced ERS, increased caspase 12 and 8 levels in the liver, and changed expression levels of genes associated with lipid metabolism, including FAS, FXR, and SREBP-1c. Treatment with either exercise or IGF-I reduced fibrosis levels suppressed ER stress markers and apoptosis, and improved expression of genes associated with lipid metabolism. In addition, simultaneous treatment with exercise and IGF-I showed a synergistic effect compared to DIA + E and DIA + IGF-I. The results suggest that IGF-1 and exercise reduced liver fibrosis possibly by reducing ERS, creating adaptive ER stress status, and improving protein folding.
Collapse
Affiliation(s)
- Shadi Mohammadpour-Asl
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
- Department of Physiology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | | | - Shiva Roshan-Milani
- Department of Physiology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
- Neurophysiology Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Ehsan Saboory
- Department of Addiction Studies, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Bijan Ghobadian
- Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Leila Chodari
- Department of Physiology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
- Neurophysiology Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
15
|
Zhao T, Tian Y, Zhao J, Sun D, Ma Y, Wang W, Yan W, Jiao P, Ma J. Loss of mitogen-activated protein kinase phosphate-5 aggravates islet dysfunction in mice with type 1 and type 2 diabetes. FASEB J 2024; 38:e23437. [PMID: 38305849 DOI: 10.1096/fj.202301479r] [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: 07/20/2023] [Revised: 12/14/2023] [Accepted: 01/08/2024] [Indexed: 02/03/2024]
Abstract
Impaired functionality and loss of islet β-cells are the primary abnormalities underlying the pathogenesis of both type 1 and 2 diabetes (T1DM and T2DM). However, specific therapeutic and preventive mechanisms underlying these conditions remain unclear. Mitogen-activated protein kinase phosphatase-5 (MKP-5) has been implicated in carcinogenesis, lipid metabolism regulation, and immune cell activation. In a previous study, we demonstrated the involvement of exogenous MKP-5 in the regulation of obesity-induced T2DM. However, the role of endogenous MKP-5 in the T1DM and T2DM processes is unclear. Thus, mice with MKP-5 knockout (KO) were generated and used to establish mouse models of both T1DM and T2DM. Our results showed that MKP-5 KO exacerbated diabetes-related symptoms in mice with both T1DM and T2DM. Given that most phenotypic studies on islet dysfunction have focused on mice with T2DM rather than T1DM, we specifically aimed to investigate the role of endoplasmic reticulum stress (ERS) and autophagy in T2DM KO islets. To accomplish this, we performed RNA sequence analysis to gain comprehensive insight into the molecular mechanisms associated with ERS and autophagy in T2DM KO islets. The results showed that the islets from mice with MKP-5 KO triggered 5' adenosine monophosphate-activated protein kinase (AMPK)-mediated autophagy inhibition and glucose-regulated protein 78 (GRP-78)-dominated ERS. Hence, we concluded that the autophagy impairment, resulting in islet dysfunction in mice with MKP-5 KO, is mediated through GRP-78 involvement. These findings provide valuable insights into the molecular pathogenesis of diabetes and highlight the significant role of MKP-5. Moreover, this knowledge holds promise for novel therapeutic strategies targeting MKP-5 for diabetes management.
Collapse
Affiliation(s)
- Tongjian Zhao
- School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
| | - Yafei Tian
- School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
| | - Jianan Zhao
- School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
| | - Dandan Sun
- School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
| | - Yongjun Ma
- School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
| | - Wei Wang
- School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
| | - Weiqun Yan
- School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
| | - Ping Jiao
- School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
| | - Jie Ma
- School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
| |
Collapse
|
16
|
Ding X, He X, Tang B, Lan T. Integrated traditional Chinese and Western medicine in the prevention and treatment of non-alcoholic fatty liver disease: future directions and strategies. Chin Med 2024; 19:21. [PMID: 38310315 PMCID: PMC10838467 DOI: 10.1186/s13020-024-00894-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 01/23/2024] [Indexed: 02/05/2024] Open
Abstract
Traditional Chinese medicine (TCM) has been widely used for several centuries for metabolic diseases, including non-alcoholic fatty liver disease (NAFLD). At present, NAFLD has become the most prevalent form of chronic liver disease worldwide and can progress to non-alcoholic steatohepatitis (NASH), cirrhosis, and even hepatocellular carcinoma. However, there is still a lack of effective treatment strategies in Western medicine. The development of NAFLD is driven by multiple mechanisms, including genetic factors, insulin resistance, lipotoxicity, mitochondrial dysfunction, endoplasmic reticulum stress, inflammation, gut microbiota dysbiosis, and adipose tissue dysfunction. Currently, certain drugs, including insulin sensitizers, statins, vitamin E, ursodeoxycholic acid and betaine, are proven to be beneficial for the clinical treatment of NAFLD. Due to its complex pathogenesis, personalized medicine that integrates various mechanisms may provide better benefits to patients with NAFLD. The holistic view and syndrome differentiation of TCM have advantages in treating NAFLD, which are similar to the principles of personalized medicine. In TCM, NAFLD is primarily classified into five types based on clinical experience. It is located in the liver and is closely related to spleen and kidney functions. However, due to the multi-component characteristics of traditional Chinese medicine, its application in the treatment of NAFLD has been considerably limited. In this review, we summarize the advances in the pathogenesis and treatment of NAFLD, drawn from both the Western medicine and TCM perspectives. We highlight that Chinese and Western medicine have complementary advantages and should receive increased attention in the prevention and treatment of NAFLD.
Collapse
Affiliation(s)
- Xin Ding
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, 280 Wai Huan Dong Road, Guangzhou, 510006, China
| | - Xu He
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, 280 Wai Huan Dong Road, Guangzhou, 510006, China
| | - Bulang Tang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, 280 Wai Huan Dong Road, Guangzhou, 510006, China
| | - Tian Lan
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, 280 Wai Huan Dong Road, Guangzhou, 510006, China.
- School of Pharmacy, Harbin Medical University, Harbin, 150086, China.
| |
Collapse
|
17
|
Li J, Wu YJ. Tri-ortho-cresyl phosphate induces hepatic steatosis by mTOR activation and ER stress induction. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 271:116010. [PMID: 38280340 DOI: 10.1016/j.ecoenv.2024.116010] [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: 06/04/2023] [Revised: 12/17/2023] [Accepted: 01/21/2024] [Indexed: 01/29/2024]
Abstract
Tri-ortho-cresyl phosphate (TOCP), an organophosphorus compound (OP), which is widely used as plasticizer, flame retardant and other industrial products, has been reported to cause multiple toxicities including neurotoxicity and reproductive toxicity. However, it remains to be elusive whether TOCP induces hepatotoxicity. The purpose of this study was to investigate the effect of TOCP on hepatocytes and the lipid metabolism in particular. The adult mice were given a single dose of TOCP (800 mg/kg, p.o.) and the histological changes in liver tissue and lipid content in serum were determined. The results showed that more vacuoles and lipid droplets were observed in the liver of the mice exposed to TOCP. And triglyceride concentrations in serum and liver tissue significantly increased. However, the histopathological changes of the liver and the elevated triglyceride levels in the exposed mice can be reversed by endoplasmic reticulum (ER) stress inhibitor 4-phenylbutyric acid and mTOR signal inhibitor rapamycin. It was also found that the changes of expression levels of the biomarkers of ER stress and mTOR signaling pathway, such as GRP78, CHOP, and p-mTOR, in the exposed mice were consistent with those observed in the cultured primary hepatocytes treated with the same chemicals. These results showed that TOCP activated mTOR signal and ER stress to induce de novo lipid synthesis, which led to the hepatic steatosis in mouse.
Collapse
Affiliation(s)
- Jing Li
- Laboratory of Molecular Toxicology, State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yi-Jun Wu
- Laboratory of Molecular Toxicology, State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
18
|
Chandrasekaran P, Weiskirchen R. The Role of SCAP/SREBP as Central Regulators of Lipid Metabolism in Hepatic Steatosis. Int J Mol Sci 2024; 25:1109. [PMID: 38256181 PMCID: PMC10815951 DOI: 10.3390/ijms25021109] [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: 12/14/2023] [Revised: 01/09/2024] [Accepted: 01/14/2024] [Indexed: 01/24/2024] Open
Abstract
The prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) is rapidly increasing worldwide at an alarming pace, due to an increase in obesity, sedentary and unhealthy lifestyles, and unbalanced dietary habits. MASLD is a unique, multi-factorial condition with several phases of progression including steatosis, steatohepatitis, fibrosis, cirrhosis, and hepatocellular carcinoma. Sterol element binding protein 1c (SREBP1c) is the main transcription factor involved in regulating hepatic de novo lipogenesis. This transcription factor is synthesized as an inactive precursor, and its proteolytic maturation is initiated in the membrane of the endoplasmic reticulum upon stimulation by insulin. SREBP cleavage activating protein (SCAP) is required as a chaperon protein to escort SREBP from the endoplasmic reticulum and to facilitate the proteolytic release of the N-terminal domain of SREBP into the Golgi. SCAP inhibition prevents activation of SREBP and inhibits the expression of genes involved in triglyceride and fatty acid synthesis, resulting in the inhibition of de novo lipogenesis. In line, previous studies have shown that SCAP inhibition can resolve hepatic steatosis in animal models and intensive research is going on to understand the effects of SCAP in the pathogenesis of human disease. This review focuses on the versatile roles of SCAP/SREBP regulation in de novo lipogenesis and the structure and molecular features of SCAP/SREBP in the progression of hepatic steatosis. In addition, recent studies that attempt to target the SCAP/SREBP axis as a therapeutic option to interfere with MASLD are discussed.
Collapse
Affiliation(s)
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), Rheinisch-Westfälische Technische Hochschule (RWTH) University Hospital Aachen, D-52074 Aachen, Germany
| |
Collapse
|
19
|
Wang X, Chen Y, Meng H, Meng F. SREBPs as the potential target for solving the polypharmacy dilemma. Front Physiol 2024; 14:1272540. [PMID: 38269061 PMCID: PMC10806128 DOI: 10.3389/fphys.2023.1272540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 12/26/2023] [Indexed: 01/26/2024] Open
Abstract
The phenomenon of polypharmacy is a common occurrence among older people with multiple health conditions due to the rapid increase in population aging and the popularization of clinical guidelines. The prevalence of metabolic syndrome is growing quickly, representing a serious threat to both the public and the worldwide healthcare systems. In addition, it enhances the risk of cardiovascular disease as well as mortality and morbidity. Sterol regulatory element binding proteins (SREBPs) are basic helix-loop-helix leucine zipper transcription factors that transcriptionally modulate genes that regulate lipid biosynthesis and uptake, thereby serving an essential role in biological systems regulation. In this article, we have described the structure of SREBPs and explored their activation and regulation of signals. We also reveal that SREBPs are intricately involved in the modulation of metabolic diseases and thus have tremendous potential as the novel target for single-drug therapy for multiple diseases.
Collapse
Affiliation(s)
| | | | | | - Fanbo Meng
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis (Jilin Provincial Engineering Laboratory for Endothelial Function and Genetic Diagnosis of Cardiovascular Disease, Jilin Provincial Molecular Biology Research Center for Precision Medicine of Major Cardiovascular Disease, Jilin Provincial Cardiovascular Research Institute), Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
20
|
Yilmaz E. Endoplasmic Reticulum Stress and Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:373-390. [PMID: 39287859 DOI: 10.1007/978-3-031-63657-8_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
In recent years, the world has seen an alarming increase in obesity and is closely associated with insulin resistance, which is a state of low-grade inflammation, the latter characterized by elevated levels of proinflammatory cytokines in blood and tissues. A shift in energy balance alters systemic metabolic regulation and the important role that chronic inflammation, endoplasmic reticulum (ER) dysfunction, and activation of the unfolded protein response (UPR) plays in this process.Why obesity is so closely associated with insulin resistance and inflammation is not understood well. This suggests that there are probably many causes for obesity-related insulin resistance and inflammation. One of the faulty mechanisms is protein homeostasis, protein quality control system included protein folding, chaperone activity, and ER-associated degradation leading to endoplasmic reticulum (ER) stress.The ER is a vast membranous network responsible for the trafficking of a wide range of proteins and plays a central role in integrating multiple metabolic signals critical in cellular homeostasis. Conditions that may trigger unfolded protein response activation include increased protein synthesis, the presence of mutant or misfolded proteins, inhibition of protein glycosylation, imbalance of ER calcium levels, glucose and energy deprivation, hypoxia, pathogens, or pathogen-associated components and toxins. Thus, characterizing the mechanisms contributing to obesity and identifying potential targets for its prevention and treatment will have a great impact on the control of associated conditions, particularly T2D.
Collapse
Affiliation(s)
- Erkan Yilmaz
- Biotechnology Institute, Ankara University, Kecioren, Ankara, Turkey.
| |
Collapse
|
21
|
Geiger M, Gorica E, Mohammed SA, Mongelli A, Mengozi A, Delfine V, Ruschitzka F, Costantino S, Paneni F. Epigenetic Network in Immunometabolic Disease. Adv Biol (Weinh) 2024; 8:e2300211. [PMID: 37794610 DOI: 10.1002/adbi.202300211] [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: 06/08/2023] [Revised: 09/08/2023] [Indexed: 10/06/2023]
Abstract
Although a large amount of data consistently shows that genes affect immunometabolic characteristics and outcomes, epigenetic mechanisms are also heavily implicated. Epigenetic changes, including DNA methylation, histone modification, and noncoding RNA, determine gene activity by altering the accessibility of chromatin to transcription factors. Various factors influence these alterations, including genetics, lifestyle, and environmental cues. Moreover, acquired epigenetic signals can be transmitted across generations, thus contributing to early disease traits in the offspring. A closer investigation is critical in this aspect as it can help to understand the underlying molecular mechanisms further and gain insights into potential therapeutic targets for preventing and treating diseases arising from immuno-metabolic dysregulation. In this review, the role of chromatin alterations in the transcriptional modulation of genes involved in insulin resistance, systemic inflammation, macrophage polarization, endothelial dysfunction, metabolic cardiomyopathy, and nonalcoholic fatty liver disease (NAFLD), is discussed. An overview of emerging chromatin-modifying drugs and the importance of the individual epigenetic profile for personalized therapeutic approaches in patients with immuno-metabolic disorders is also presented.
Collapse
Affiliation(s)
- Martin Geiger
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Era Gorica
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Shafeeq Ahmed Mohammed
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Alessia Mongelli
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Alessandro Mengozi
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Valentina Delfine
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Frank Ruschitzka
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Sarah Costantino
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
- University Heart Center, University Hospital Zurich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Francesco Paneni
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
- University Heart Center, University Hospital Zurich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
- Department of Research and Education, University Hospital Zurich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| |
Collapse
|
22
|
Kim HR, Young CN. Circumventricular organ-hypothalamic circuit endoplasmic reticulum stress drives hepatic steatosis during obesity. Obesity (Silver Spring) 2024; 32:59-69. [PMID: 37794528 DOI: 10.1002/oby.23895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 08/08/2023] [Accepted: 08/08/2023] [Indexed: 10/06/2023]
Abstract
OBJECTIVE Nonalcoholic fatty liver disease (NAFLD), characterized by excess liver triglyceride accumulation (hepatic steatosis), leads to an increased risk for cardiometabolic diseases and obesity-related mortality. Emerging evidence points to endoplasmic reticulum (ER) stress in the central nervous system as critical in NAFLD pathogenesis. Here, we tested the contribution of ER stress in a circumventricular organ-hypothalamic circuit in NAFLD development during obesity. METHODS C57BL/6J male mice were fed a high-fat diet (HFD) or normal chow. A combination of histological, viral tracing, intersectional viral targeting, and in vivo integrative physiological approaches were used to examine the role of ER stress in subfornical organ to hypothalamic paraventricular nucleus projecting neurons (SFO➔PVN) in NAFLD during diet-induced obesity. RESULTS Immunohistochemical analysis revealed marked unfolded protein response activation in the SFO, particularly in excitatory SFO➔PVN neurons of HFD-fed animals. Moreover, intersectional viral inhibition of ER stress in SFO➔PVN neurons resulted in a reduction in hepatomegaly, hepatic steatosis, and a blunted increase in body weight gain during diet-induced obesity, independent of changes in food intake, substrate partitioning, energy expenditure, and ambulatory activity. CONCLUSIONS These results indicate that ER stress in an SFO➔PVN neural circuit contributes to hepatic steatosis during obesity.
Collapse
Affiliation(s)
- Han Rae Kim
- Department of Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Colin N Young
- Department of Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| |
Collapse
|
23
|
Ohguro H, Umetsu A, Sato T, Furuhashi M, Watanabe M. Lipid Metabolism Regulators Are the Possible Determinant for Characteristics of Myopic Human Scleral Stroma Fibroblasts (HSSFs). Int J Mol Sci 2023; 25:501. [PMID: 38203671 PMCID: PMC10778967 DOI: 10.3390/ijms25010501] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/25/2023] [Accepted: 12/26/2023] [Indexed: 01/12/2024] Open
Abstract
The purpose of the current investigation was to elucidate what kinds of responsible mechanisms induce elongation of the sclera in myopic eyes. To do this, two-dimensional (2D) cultures of human scleral stromal fibroblasts (HSSFs) obtained from eyes with two different axial length (AL) groups, <26 mm (low AL group, n = 2) and >27 mm (high AL group, n = 3), were subjected to (1) measurements of Seahorse mitochondrial and glycolytic indices to evaluate biological aspects and (2) analysis by RNA sequencing. Extracellular flux analysis revealed that metabolic indices related to mitochondrial and glycolytic functions were higher in the low AL group than in the high AL group, suggesting that metabolic activities of HSSF cells are different depending the degree of AL. Based upon RNA sequencing of these low and high AL groups, the bioinformatic analyses using gene ontology (GO) enrichment analysis and ingenuity pathway analysis (IPA) of differentially expressed genes (DEGs) identified that sterol regulatory element-binding transcription factor 2 (SREBF2) is both a possible upstream regulator and a causal network regulator. Furthermore, SREBF1, insulin-induced gene 1 (INSIG1), and insulin-like growth factor 1 (IGF1) were detected as upstream regulators, and protein tyrosine phosphatase receptor type O (PTPRO) was detected as a causal network regulator. Since those possible regulators were all pivotally involved in lipid metabolisms including fatty acid (FA), triglyceride (TG) and cholesterol (Chol) biosynthesis, the findings reported here indicate that FA, TG and Chol biosynthesis regulation may be responsible mechanisms inducing AL elongation via HSSF.
Collapse
Affiliation(s)
- Hiroshi Ohguro
- Department of Ophthalmology, School of Medicine, Sapporo Medical University, Sapporo 060-8556, Hokkaido, Japan; (H.O.); (A.U.)
| | - Araya Umetsu
- Department of Ophthalmology, School of Medicine, Sapporo Medical University, Sapporo 060-8556, Hokkaido, Japan; (H.O.); (A.U.)
| | - Tatsuya Sato
- Department of Cardiovascular, Renal and Metabolic Medicine, School of Medicine, Sapporo Medical University, Sapporo 060-8556, Hokkaido, Japan
- Department of Cellular Physiology and Signal Transduction, School of Medicine, Sapporo Medical University, Sapporo 060-8556, Hokkaido, Japan
| | - Masato Furuhashi
- Department of Cardiovascular, Renal and Metabolic Medicine, School of Medicine, Sapporo Medical University, Sapporo 060-8556, Hokkaido, Japan
| | - Megumi Watanabe
- Department of Ophthalmology, School of Medicine, Sapporo Medical University, Sapporo 060-8556, Hokkaido, Japan; (H.O.); (A.U.)
| |
Collapse
|
24
|
Li N, Li X, Ding Y, Liu X, Diggle K, Kisseleva T, Brenner DA. SREBP Regulation of Lipid Metabolism in Liver Disease, and Therapeutic Strategies. Biomedicines 2023; 11:3280. [PMID: 38137501 PMCID: PMC10740981 DOI: 10.3390/biomedicines11123280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/26/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Sterol regulatory element-binding proteins (SREBPs) are master transcription factors that play a crucial role in regulating genes involved in the biogenesis of cholesterol, fatty acids, and triglycerides. As such, they are implicated in several serious liver diseases, including non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), fibrosis, and hepatocellular carcinoma (HCC). SREBPs are subject to regulation by multiple cofactors and critical signaling pathways, making them an important target for therapeutic interventions. In this review, we first introduce the structure and activation of SREBPs, before focusing on their function in liver disease. We examine the mechanisms by which SREBPs regulate lipogenesis, explore how alterations in these processes are associated with liver disease, and evaluate potential therapeutic strategies using small molecules, natural products, or herb extracts that target these pathways. Through this analysis, we provide new insights into the versatility and multitargets of SREBPs as factors in the modulation of different physiological stages of liver disease, highlighting their potential targets for therapeutic treatment.
Collapse
Affiliation(s)
- Na Li
- College of Medical Technology, Shanghai University of Medicine & Health Sciences, Shanghai 201318, China
- Graduate School of Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiaodan Li
- College of Medical Technology, Shanghai University of Medicine & Health Sciences, Shanghai 201318, China
- Graduate School of Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yifu Ding
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Shanghai 200031, China;
| | - Xiao Liu
- Department of Surgery, University of California San Diego, La Jolla, CA 92093, USA (T.K.)
| | - Karin Diggle
- Department of Surgery, University of California San Diego, La Jolla, CA 92093, USA (T.K.)
| | - Tatiana Kisseleva
- Department of Surgery, University of California San Diego, La Jolla, CA 92093, USA (T.K.)
| | - David A. Brenner
- Department of Surgery, University of California San Diego, La Jolla, CA 92093, USA (T.K.)
- Sanford Burnham Prebys, La Jolla, CA 92037, USA
| |
Collapse
|
25
|
Abasubong KP, Jiang GZ, Guo HX, Wang X, Li XF, Yan-Zou D, Liu WB, Desouky HE. High-fat diet alters intestinal microbiota and induces endoplasmic reticulum stress via the activation of apoptosis and inflammation in blunt snout bream. FISH PHYSIOLOGY AND BIOCHEMISTRY 2023; 49:1079-1095. [PMID: 37831370 DOI: 10.1007/s10695-023-01240-2] [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: 04/26/2023] [Accepted: 09/16/2023] [Indexed: 10/14/2023]
Abstract
The primary organ for absorbing dietary fat is the gut. High dietary lipid intake negatively affects health and absorption by causing fat deposition in the intestine. This research explores the effect of a high-fat diet (HFD) on intestinal microbiota and its connections with endoplasmic reticulum stress and inflammation. 60 fish (average weight: 45.84 ± 0.07 g) were randomly fed a control diet (6% fat) and a high-fat diet (12 % fat) in four replicates for 12 weeks. From the result, hepatosomatic index (HSI), Visceralsomatic index (VSI), abdominal fat (ADF), Intestosomatic index (ISI), mesenteric fat (MFI), Triglycerides (TG), total cholesterol (TC), non-esterified fatty acid (NEFA) content were substantially greater on HFD compared to the control diet. Moreover, fish provided the HFD significantly obtained lower superoxide dismutase (SOD) and glutathione peroxidase (GPX) activities. In contrast, an opposite result was seen in malondialdehyde (MDA) content in comparison to the control. HFD significantly altered intestinal microbiota in blunt snout bream, characterized by an increased abundance of Aeromonas, Plesiomonas proteobacteria, and firmicutes with a reduced abundance of Cetobacterium and ZOR0006. The transcriptional levels of glucose-regulated protein 78 (grp78), inositol requiring enzyme 1 (ire1), spliced X box-binding protein 1 (xbp1), DnaJ heat shock protein family (Hsp40) member B9 (dnajb9), tumor necrosis factor alpha (tnf-α), nuclear factor-kappa B (nf-κb), monocyte chemoattractant protein-1 (mcp-1), and interleukin-6 (il-6) in the intestine were markedly upregulated in fish fed HFD than the control group. Also, the outcome was similar in bax, caspases-3, and caspases-9, ZO-1, Occludin-1, and Occludin-2 expressions. In conclusion, HFD could alter microbiota and facilitate chronic inflammatory signals via activating endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Kenneth Prudence Abasubong
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, People's Republic of China
- National Laboratory of Animal Science, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, People's Republic of China
| | - Guang-Zhen Jiang
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, People's Republic of China
- National Laboratory of Animal Science, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, People's Republic of China
| | - Hui-Xing Guo
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, People's Republic of China
- National Laboratory of Animal Science, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, People's Republic of China
| | - Xi Wang
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, People's Republic of China
- National Laboratory of Animal Science, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, People's Republic of China
| | - Xiang-Fei Li
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, People's Republic of China
- National Laboratory of Animal Science, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, People's Republic of China
| | - Dong Yan-Zou
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, People's Republic of China
- National Laboratory of Animal Science, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, People's Republic of China
| | - Wen-Bin Liu
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, People's Republic of China.
- National Laboratory of Animal Science, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, People's Republic of China.
| | - Hesham Eed Desouky
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, People's Republic of China
- National Laboratory of Animal Science, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, People's Republic of China
- Department of Animal and Poultry Production, Faculty of Agriculture, Damanhour University, Damanhour, Beheria, 22713, Egypt
| |
Collapse
|
26
|
Sun J, Yan Q, Zhang Z, Xu T, Gong Y, Li W, Mai K, Ai Q. Exploring the role of SWI/SNF complex subunit BAF60c in lipid metabolism and inflammation in fish. iScience 2023; 26:108207. [PMID: 37942006 PMCID: PMC10628743 DOI: 10.1016/j.isci.2023.108207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/26/2023] [Accepted: 10/11/2023] [Indexed: 11/10/2023] Open
Abstract
Chromatin remodeling plays an important role in regulating gene transcription, in which chromatin remodeling complex is a crucial aspect. Brg1/Brm-associated factor 60c (BAF60c) subunit forms a bridge between chromatin remodeling complexes and transcription factors in mammals; hence, it has received extensive attention. However, the roles of BAF60c in fish remain largely unexplored. In this study, we identified BAF60c-interacting proteins by using HIS-pull-down and LC-MS/MS analysis in fish. Subsequently, the RNA-seq analysis was performed to identify the overall effects of BAF60c. Then, the function of BAF60c was verified through BAF60c knockdown and overexpression experiments. We demonstrated for the first time that BAF60c interacts with glucose-regulated protein 78 (GRP78) and regulates lipid metabolism, endoplasmic reticulum (ER) stress, and inflammation. Knockdown of BAF60c reduces fatty acid biosynthesis, ER stress, and inflammation. In conclusion, the results enriched BAF60c-interacting protein network and explored the function of BAF60c in lipid metabolism and inflammation in fish.
Collapse
Affiliation(s)
- Jie Sun
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) & Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, Qingdao, Shandong 266003, People’s Republic of China
| | - Qiuxin Yan
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) & Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, Qingdao, Shandong 266003, People’s Republic of China
| | - Zhihao Zhang
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) & Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, Qingdao, Shandong 266003, People’s Republic of China
| | - Ting Xu
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) & Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, Qingdao, Shandong 266003, People’s Republic of China
| | - Ye Gong
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) & Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, Qingdao, Shandong 266003, People’s Republic of China
| | - Weijia Li
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) & Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, Qingdao, Shandong 266003, People’s Republic of China
| | - Kangsen Mai
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) & Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, Qingdao, Shandong 266003, People’s Republic of China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao, Shandong 266237, People’s Republic of China
| | - Qinghui Ai
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) & Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, Qingdao, Shandong 266003, People’s Republic of China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao, Shandong 266237, People’s Republic of China
| |
Collapse
|
27
|
Na M, Yang X, Deng Y, Yin Z, Li M. Endoplasmic reticulum stress in the pathogenesis of alcoholic liver disease. PeerJ 2023; 11:e16398. [PMID: 38025713 PMCID: PMC10655704 DOI: 10.7717/peerj.16398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/12/2023] [Indexed: 12/01/2023] Open
Abstract
The endoplasmic reticulum (ER) plays a pivotal role in protein synthesis, folding, and modification. Under stress conditions such as oxidative stress and inflammation, the ER can become overwhelmed, leading to an accumulation of misfolded proteins and ensuing ER stress. This triggers the unfolded protein response (UPR) designed to restore ER homeostasis. Alcoholic liver disease (ALD), a spectrum disorder resulting from chronic alcohol consumption, encompasses conditions from fatty liver and alcoholic hepatitis to cirrhosis. Metabolites of alcohol can incite oxidative stress and inflammation in hepatic cells, instigating ER stress. Prolonged alcohol exposure further disrupts protein homeostasis, exacerbating ER stress which can lead to irreversible hepatocellular damage and ALD progression. Elucidating the contribution of ER stress to ALD pathogenesis may pave the way for innovative therapeutic interventions. This review delves into ER stress, its basic signaling pathways, and its role in the alcoholic liver injury.
Collapse
Affiliation(s)
- Man Na
- Department of Pharmacy, The 926th Hospital of Joint Logistics Support Force of Chinese People’s Liberation Army, Kaiyuan, Yunan, China
| | - Xingbiao Yang
- Department of Pharmacy, The 926th Hospital of Joint Logistics Support Force of Chinese People’s Liberation Army, Kaiyuan, Yunan, China
| | - Yongkun Deng
- Department of Pharmacy, The 926th Hospital of Joint Logistics Support Force of Chinese People’s Liberation Army, Kaiyuan, Yunan, China
| | - Zhaoheng Yin
- Department of Pharmacy, The 926th Hospital of Joint Logistics Support Force of Chinese People’s Liberation Army, Kaiyuan, Yunan, China
| | - Mingwei Li
- Department of Pharmacy, The 926th Hospital of Joint Logistics Support Force of Chinese People’s Liberation Army, Kaiyuan, Yunan, China
| |
Collapse
|
28
|
Esmaeilzadeh A, Mohammadi V, Elahi R, Rezakhani N. The role of heat shock proteins (HSPs) in type 2 diabetes mellitus pathophysiology. J Diabetes Complications 2023; 37:108564. [PMID: 37852076 DOI: 10.1016/j.jdiacomp.2023.108564] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 07/05/2023] [Accepted: 07/21/2023] [Indexed: 10/20/2023]
Abstract
Type 2 diabetes mellitus (T2DM) is a metabolic disorder characterized by sustained hyperglycemia caused by impaired insulin signaling and secretion. Metabolic stress, caused by an inappropriate diet, is one of the major hallmarks provoking inflammation, endoplasmic reticulum (ER) stress, and mitochondrial dysfunction. Heat shock proteins (HSPs) are a group of highly conserved proteins that have a crucial role in chaperoning damaged and misfolded proteins to avoid disruption of cellular homeostasis under stress conditions. To do this, HSPs interact with diverse intra-and extracellular pathways among which are the insulin signaling, insulin secretion, and apoptosis pathways. Therefore, HSP dysfunction, e.g. HSP70, may lead to disruption of the pathways responsible for insulin secretion and uptake. Consistently, the altered expression of other HSPs and genetic polymorphisms in HSP-producing genes in diabetic subjects has made HSPs hot research in T2DM. This paper provides a comprehensive overview of the role of different HSPs in T2DM pathogenesis, affected cellular pathways, and the potential therapeutic strategies targeting HSPs in T2DM.
Collapse
Affiliation(s)
- Abdolreza Esmaeilzadeh
- Department of Immunology, Zanjan University of Medical Sciences, Zanjan, Iran; Cancer Gene Therapy Research Center (CGRC), Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Vahid Mohammadi
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Reza Elahi
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Negin Rezakhani
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
29
|
Baier D, Mendrina T, Schoenhacker‐Alte B, Pirker C, Mohr T, Rusz M, Regner B, Schaier M, Sgarioto N, Raynal NJ, Nowikovsky K, Schmidt WM, Heffeter P, Meier‐Menches SM, Koellensperger G, Keppler BK, Berger W. The Lipid Metabolism as Target and Modulator of BOLD-100 Anticancer Activity: Crosstalk with Histone Acetylation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301939. [PMID: 37752764 PMCID: PMC10646284 DOI: 10.1002/advs.202301939] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 08/18/2023] [Indexed: 09/28/2023]
Abstract
The leading first-in-class ruthenium-complex BOLD-100 currently undergoes clinical phase-II anticancer evaluation. Recently, BOLD-100 is identified as anti-Warburg compound. The present study shows that also deregulated lipid metabolism parameters characterize acquired BOLD-100-resistant colon and pancreatic carcinoma cells. Acute BOLD-100 treatment reduces lipid droplet contents of BOLD-100-sensitive but not -resistant cells. Despite enhanced glycolysis fueling lipid accumulation, BOLD-100-resistant cells reveal diminished lactate secretion based on monocarboxylate transporter 1 (MCT1) loss mediated by a frame-shift mutation in the MCT1 chaperone basigin. Glycolysis and lipid catabolism converge in the production of protein/histone acetylation substrate acetyl-coenzymeA (CoA). Mass spectrometric and nuclear magnetic resonance analyses uncover spontaneous cell-free BOLD-100-CoA adduct formation suggesting acetyl-CoA depletion as mechanism bridging BOLD-100-induced lipid metabolism alterations and histone acetylation-mediated gene expression deregulation. Indeed, BOLD-100 treatment decreases histone acetylation selectively in sensitive cells. Pharmacological targeting confirms histone de-acetylation as central mode-of-action of BOLD-100 and metabolic programs stabilizing histone acetylation as relevant Achilles' heel of acquired BOLD-100-resistant cell and xenograft models. Accordingly, histone gene expression changes also predict intrinsic BOLD-100 responsiveness. Summarizing, BOLD-100 is identified as epigenetically active substance acting via targeting several onco-metabolic pathways. Identification of the lipid metabolism as driver of acquired BOLD-100 resistance opens novel strategies to tackle therapy failure.
Collapse
Affiliation(s)
- Dina Baier
- Center for Cancer Research and Comprehensive Cancer CenterMedical University ViennaBorschkegasse 8aVienna1090Austria
- Institute of Inorganic ChemistryUniversity of ViennaWaehringer Str. 42Vienna1090Austria
- Research Cluster “Translational Cancer Therapy Research”Vienna1090Austria
| | - Theresa Mendrina
- Center for Cancer Research and Comprehensive Cancer CenterMedical University ViennaBorschkegasse 8aVienna1090Austria
- Institute of Inorganic ChemistryUniversity of ViennaWaehringer Str. 42Vienna1090Austria
- Research Cluster “Translational Cancer Therapy Research”Vienna1090Austria
| | - Beatrix Schoenhacker‐Alte
- Center for Cancer Research and Comprehensive Cancer CenterMedical University ViennaBorschkegasse 8aVienna1090Austria
- Institute of Inorganic ChemistryUniversity of ViennaWaehringer Str. 42Vienna1090Austria
- Research Cluster “Translational Cancer Therapy Research”Vienna1090Austria
| | - Christine Pirker
- Center for Cancer Research and Comprehensive Cancer CenterMedical University ViennaBorschkegasse 8aVienna1090Austria
- Research Cluster “Translational Cancer Therapy Research”Vienna1090Austria
| | - Thomas Mohr
- Center for Cancer Research and Comprehensive Cancer CenterMedical University ViennaBorschkegasse 8aVienna1090Austria
- Joint Metabolome FacilityUniversity of Vienna and Medical University of ViennaWaehringer Str. 38Vienna1090Austria
- ScienceConsultGuntramsdorf2351Austria
| | - Mate Rusz
- Institute of Inorganic ChemistryUniversity of ViennaWaehringer Str. 42Vienna1090Austria
- Research Cluster “Translational Cancer Therapy Research”Vienna1090Austria
- Institute of Analytical ChemistryFaculty of ChemistryUniversity of ViennaWaehringer Str. 38Vienna1090Austria
| | - Benedict Regner
- Anna Spiegel Center of Translational ResearchDepartment of Medicine IMedical University ViennaLazarettgasse 14Vienna1090Austria
| | - Martin Schaier
- Institute of Analytical ChemistryFaculty of ChemistryUniversity of ViennaWaehringer Str. 38Vienna1090Austria
| | - Nicolas Sgarioto
- Départment de pharmacologie et physiologieFaculté de médecineCentre de recherché de l hôpitalUniversité de MontréalSaint‐Justine (7.17.020), 3175 Chemin de la Côte Ste‐CatherineQuebecH3T1C5Canada
| | - Noël J.‐M. Raynal
- Départment de pharmacologie et physiologieFaculté de médecineCentre de recherché de l hôpitalUniversité de MontréalSaint‐Justine (7.17.020), 3175 Chemin de la Côte Ste‐CatherineQuebecH3T1C5Canada
| | - Karin Nowikovsky
- Unit of Physiology and BiophysicsDepartment of Biomedical SciencesUniversity of Veterinary Medicine ViennaVeterinaerplatz 1Vienna1210Austria
| | - Wolfgang M. Schmidt
- Neuromuscular Research DepartmentCenter for Anatomy and Cell BiologyMedical University of ViennaWähringer Str. 13Vienna1090Austria
| | - Petra Heffeter
- Center for Cancer Research and Comprehensive Cancer CenterMedical University ViennaBorschkegasse 8aVienna1090Austria
- Research Cluster “Translational Cancer Therapy Research”Vienna1090Austria
| | - Samuel M. Meier‐Menches
- Institute of Inorganic ChemistryUniversity of ViennaWaehringer Str. 42Vienna1090Austria
- Joint Metabolome FacilityUniversity of Vienna and Medical University of ViennaWaehringer Str. 38Vienna1090Austria
- Institute of Analytical ChemistryFaculty of ChemistryUniversity of ViennaWaehringer Str. 38Vienna1090Austria
| | - Gunda Koellensperger
- Institute of Analytical ChemistryFaculty of ChemistryUniversity of ViennaWaehringer Str. 38Vienna1090Austria
| | - Bernhard K. Keppler
- Institute of Inorganic ChemistryUniversity of ViennaWaehringer Str. 42Vienna1090Austria
- Research Cluster “Translational Cancer Therapy Research”Vienna1090Austria
| | - Walter Berger
- Center for Cancer Research and Comprehensive Cancer CenterMedical University ViennaBorschkegasse 8aVienna1090Austria
- Research Cluster “Translational Cancer Therapy Research”Vienna1090Austria
| |
Collapse
|
30
|
Buyl K, Vrints M, Fernando R, Desmae T, Van Eeckhoutte T, Jans M, Van Der Schueren J, Boeckmans J, Rodrigues RM, De Boe V, Rogiers V, De Kock J, Beirinckx F, Vanhaecke T. Human skin stem cell-derived hepatic cells as in vitro drug discovery model for insulin-driven de novo lipogenesis. Eur J Pharmacol 2023; 957:175989. [PMID: 37572939 DOI: 10.1016/j.ejphar.2023.175989] [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: 03/29/2023] [Revised: 08/10/2023] [Accepted: 08/10/2023] [Indexed: 08/14/2023]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), formerly known as non-alcoholic fatty liver disease (NAFLD), is characterized by intrahepatic triglyceride accumulation and can progress to metabolic dysfunction-associated steatohepatitis (MASH) and liver fibrosis. Hepatic de novo lipogenesis (DNL), activated by glucose and insulin, is a central pathway contributing to early-stage development of MASLD. The emerging global prevalence of MASLD highlights the urgent need for pharmaceutical intervention to combat this health threat. However, the identification of novel drugs that could inhibit hepatic DNL is hampered by a lack of reliable, insulin-sensitive, human, in vitro, hepatic models. Here, we report human skin stem cell-derived hepatic cells (hSKP-HPC) as a unique in vitro model to study insulin-driven DNL (iDNL), evidenced by both gene expression and lipid accumulation readouts. Insulin-sensitive hSKP-HPC showed increased sterol regulatory element-binding protein 1c (SREBP-1c) expression, a key transcription factor for DNL. Furthermore, this physiologically relevant in vitro human steatosis model allowed both inhibition and activation of the iDNL pathway using reference inhibitors and activators, respectively. Optimisation of the lipid accumulation assay to a high-throughput, 384-well format enabled the screening of a library of annotated compounds, delivering new insights on key players in the iDNL pathway and MASLD pathophysiology. Together, these results establish the value of the hSKP-HPC model in preclinical development of antisteatotic drugs to combat MASLD.
Collapse
Affiliation(s)
- Karolien Buyl
- Department of in Vitro Toxicology and Dermato-Cosmetology (IVTD), Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, B-1090, Brussels, Belgium.
| | - Martine Vrints
- Galapagos NV, Industriepark Mechelen Noord, Generaal De Wittelaan L11 A3, B-2880, Mechelen, Belgium
| | - Ruani Fernando
- Department of in Vitro Toxicology and Dermato-Cosmetology (IVTD), Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, B-1090, Brussels, Belgium
| | - Terry Desmae
- Department of in Vitro Toxicology and Dermato-Cosmetology (IVTD), Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, B-1090, Brussels, Belgium
| | - Thomas Van Eeckhoutte
- Galapagos NV, Industriepark Mechelen Noord, Generaal De Wittelaan L11 A3, B-2880, Mechelen, Belgium
| | - Mia Jans
- Galapagos NV, Industriepark Mechelen Noord, Generaal De Wittelaan L11 A3, B-2880, Mechelen, Belgium
| | - Jan Van Der Schueren
- Galapagos NV, Industriepark Mechelen Noord, Generaal De Wittelaan L11 A3, B-2880, Mechelen, Belgium
| | - Joost Boeckmans
- Department of in Vitro Toxicology and Dermato-Cosmetology (IVTD), Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, B-1090, Brussels, Belgium
| | - Robim M Rodrigues
- Department of in Vitro Toxicology and Dermato-Cosmetology (IVTD), Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, B-1090, Brussels, Belgium
| | - Veerle De Boe
- Department of Urology, Universitair Ziekenhuis Brussel (UZ-Brussel), Laarbeeklaan 101, B-1090, Brussels, Belgium
| | - Vera Rogiers
- Department of in Vitro Toxicology and Dermato-Cosmetology (IVTD), Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, B-1090, Brussels, Belgium
| | - Joery De Kock
- Department of in Vitro Toxicology and Dermato-Cosmetology (IVTD), Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, B-1090, Brussels, Belgium
| | - Filip Beirinckx
- Galapagos NV, Industriepark Mechelen Noord, Generaal De Wittelaan L11 A3, B-2880, Mechelen, Belgium
| | - Tamara Vanhaecke
- Department of in Vitro Toxicology and Dermato-Cosmetology (IVTD), Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, B-1090, Brussels, Belgium
| |
Collapse
|
31
|
Hayashi H, Sawada K, Tanaka H, Muro K, Hasebe T, Nakajima S, Okumura T, Fujiya M. The effect of heat-killed Lactobacillus brevis SBL88 on improving selective hepatic insulin resistance in non-alcoholic fatty liver disease mice without altering the gut microbiota. J Gastroenterol Hepatol 2023; 38:1847-1854. [PMID: 37646384 DOI: 10.1111/jgh.16337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 09/01/2023]
Abstract
BACKGROUND AND AIM There have been several reports that some probiotics improve non-alcoholic fatty liver disease (NAFLD); however, many studies have involved cocktail therapies. We evaluated whether heat-killed Lactobacillus brevis SBL88 (L. brevis SBL88) monotherapy improves the clinical features of NAFLD. METHODS The NAFLD model was induced in mice fed a high-fat diet (HFD) (HFD mice) or HFD + 1% heat-killed L. brevis SBL88 (SBL mice) for 16 weeks. Histopathological liver findings were analyzed. To evaluate the gut microbiota, a modified terminal restriction fragment length polymorphism analysis of the feces was performed. RNA sequencing in the liver was performed with Ion Proton™. To investigate the direct effects of heat-killed L. brevis SBL88, an in vitro study was performed. RESULTS Histopathological findings revealed that fat droplets in the liver were significantly reduced in SBL mice; however, terminal restriction fragment length polymorphism did not show alterations in the gut microbiota between HFD mice and SBL mice. RNA sequencing and pathway analysis revealed that the regulation of lipid and insulin metabolism was affected. The mRNA expression of insulin receptor substrate 2 (IRS-2) was significantly higher in SBL mice, whereas the expression of IRS-1 was not significantly different. Phospho-IRS-2 expression was also significantly increased in SBL mice. In addition, an in vitro study revealed significant alterations in IRS-2 and forkhead box protein O1 expression levels. CONCLUSION SBL mice exhibited partially improved selective hepatic insulin resistance. Our data suggest that heat-killed L. brevis SBL88 could attenuate the clinical features of NAFLD that are not mediated by alterations in the gut microbiota.
Collapse
Affiliation(s)
- Hidemi Hayashi
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Koji Sawada
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Hiroki Tanaka
- Division of Tumor Pathology, Department of Pathology, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Kazuki Muro
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Takumu Hasebe
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Shunsuke Nakajima
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Toshikatsu Okumura
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Mikihiro Fujiya
- Gastroenterology and Endoscopy, Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| |
Collapse
|
32
|
Shreya S, Grosset CF, Jain BP. Unfolded Protein Response Signaling in Liver Disorders: A 2023 Updated Review. Int J Mol Sci 2023; 24:14066. [PMID: 37762367 PMCID: PMC10531763 DOI: 10.3390/ijms241814066] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/04/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Endoplasmic reticulum (ER) is the site for synthesis and folding of secreted and transmembrane proteins. Disturbance in the functioning of ER leads to the accumulation of unfolded and misfolded proteins, which finally activate the unfolded protein response (UPR) signaling. The three branches of UPR-IRE1 (Inositol requiring enzyme 1), PERK (Protein kinase RNA-activated (PKR)-like ER kinase), and ATF6 (Activating transcription factor 6)-modulate the gene expression pattern through increased expression of chaperones and restore ER homeostasis by enhancing ER protein folding capacity. The liver is a central organ which performs a variety of functions which help in maintaining the overall well-being of our body. The liver plays many roles in cellular physiology, blood homeostasis, and detoxification, and is the main site at which protein synthesis occurs. Disturbance in ER homeostasis is triggered by calcium level imbalance, change in redox status, viral infection, and so on. ER dysfunction and subsequent UPR signaling participate in various hepatic disorders like metabolic (dysfunction) associated fatty liver disease, liver cancer, viral hepatitis, and cholestasis. The exact role of ER stress and UPR signaling in various liver diseases is not fully understood and needs further investigation. Targeting UPR signaling with drugs is the subject of intensive research for therapeutic use in liver diseases. The present review summarizes the role of UPR signaling in liver disorders and describes why UPR regulators are promising therapeutic targets.
Collapse
Affiliation(s)
- Smriti Shreya
- Gene Expression and Signaling Lab, Department of Zoology, Mahatma Gandhi Central University, Motihari 845401, Bihar, India;
| | - Christophe F. Grosset
- MIRCADE Team, U1312, Bordeaux Institute in Oncology, BRIC, Université de Bordeaux, 146 Rue Léo Saignat, F-33000 Bordeaux, France
| | - Buddhi Prakash Jain
- Gene Expression and Signaling Lab, Department of Zoology, Mahatma Gandhi Central University, Motihari 845401, Bihar, India;
| |
Collapse
|
33
|
Pioltine EM, Costa CB, Franchi FF, dos Santos PH, Nogueira MFG. Tauroursodeoxycholic Acid Supplementation in In Vitro Culture of Indicine Bovine Embryos: Molecular and Cellular Effects on the In Vitro Cryotolerance. Int J Mol Sci 2023; 24:14060. [PMID: 37762363 PMCID: PMC10531190 DOI: 10.3390/ijms241814060] [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: 07/21/2023] [Revised: 08/30/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
During embryo development, the endoplasmic reticulum (ER) acts as an important site for protein biosynthesis; however, in vitro culture (IVC) can negatively affect ER homeostasis. Therefore, the aim of our study was to evaluate the effects of the supplementation of tauroursodeoxycholic acid (TUDCA), an ER stress inhibitor, in the IVC of bovine embryos. Two experiments were carried out: Exp. 1: an evaluation of blastocyst rate, hatching kinetics, and gene expression of hatched embryos after being treated with different concentrations of TUDCA (50, 200, or 1000 μM) in the IVC; Exp. 2: an evaluation of the re-expansion, hatching, and gene expression of hatched embryos previously treated with 200 µM of TUDCA at IVC and submitted to vitrification. There was no increase in the blastocyst and hatched blastocyst rates treated with TUDCA in the IVC. However, embryos submitted to vitrification after treatment with 200 µM of TUDCA underwent an increased hatching rate post-warming together with a down-regulation in the expression of ER stress-related genes and the accumulation of lipids. In conclusion, this work showed that the addition of TUDCA during in vitro culture can improve the cryotolerance of the bovine blastocyst through the putative modulation of ER and oxidative stress.
Collapse
Affiliation(s)
- Elisa Mariano Pioltine
- Multi-User Laboratory of Phytomedicines Pharmacology, and Biotechnology (PhitoPharmaTec), Department of Pharmacology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu 18618-000, Brazil
| | - Camila Bortoliero Costa
- Multi-User Laboratory of Phytomedicines Pharmacology, and Biotechnology (PhitoPharmaTec), Department of Pharmacology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu 18618-000, Brazil
- Laboratory of Embryonic Micromanipulation, Department of Biological Sciences, School of Sciences and Languages, São Paulo State University (UNESP), Assis 19806-900, Brazil
| | - Fernanda Fagali Franchi
- Multi-User Laboratory of Phytomedicines Pharmacology, and Biotechnology (PhitoPharmaTec), Department of Pharmacology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu 18618-000, Brazil
| | - Priscila Helena dos Santos
- Multi-User Laboratory of Phytomedicines Pharmacology, and Biotechnology (PhitoPharmaTec), Department of Pharmacology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu 18618-000, Brazil
| | - Marcelo Fábio Gouveia Nogueira
- Multi-User Laboratory of Phytomedicines Pharmacology, and Biotechnology (PhitoPharmaTec), Department of Pharmacology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu 18618-000, Brazil
- Laboratory of Embryonic Micromanipulation, Department of Biological Sciences, School of Sciences and Languages, São Paulo State University (UNESP), Assis 19806-900, Brazil
| |
Collapse
|
34
|
Liu H, Xing Y, Nie Q, Li Z, Meng C, Ma H. Association Between Sensitivity to Thyroid Hormones and Metabolic Dysfunction-Associated Fatty Liver Disease in Euthyroid Subjects: A Cross-Sectional Study. Diabetes Metab Syndr Obes 2023; 16:2153-2163. [PMID: 37492438 PMCID: PMC10363669 DOI: 10.2147/dmso.s420872] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 07/03/2023] [Indexed: 07/27/2023] Open
Abstract
Objective Thyroid hormones (THs) exert instrumental effects in regulating lipids metabolism. Whereas, research investigating the relationship between sensitivity indices to THs and metabolic dysfunction-associated fatty liver disease (MAFLD) have contradicted this. This study was designed to approach the correlation between sensitivity indices to THs and MAFLD in euthyroid subjects. Methods An overall sample of 6356 euthyroid participants were enrolled in a Chinese hospital. Free triiodothyronine to free thyroxine ratio (FT3/FT4), thyrotropin triiodothyronine resistance index (TT3RI), thyrotropin thyroxine resistance index (TT4RI), thyroid stimulating hormone index (TSHI) and thyroid feedback quantile-based indices (TFQIFT3 and TFQIFT4) were collected as sensitivity indicators to THs. Participants were split into two groups based on whether they suffered with MAFLD or not. And participants were categorized into quartiles based on sensitivity indicators to THs. The effects of sensitivity indices to THs on MAFLD were analyzed using regression analysis. Bootstrap was performed to assess the mediation effect of triglyceride glucose (TyG) index on the relationship between sensitivity parameters to THs and MAFLD. Results The incidence of MAFLD in euthyroid subjects was 34.47%. As FT3/FT4, TT3RI and TFQIFT3 levels rose, so did the MAFLD prevalence. After adjustment for confounders, logistic regression analyses indicated that the high-level FT3/FT4 and TFQIFT3 still remained risk factors for MAFLD. The relevance of FT3/FT4 and MAFLD was stronger among those whose age ≤ 40 years and had non-visceral obesity. And the interrelation between TFQIFT3 and MAFLD was stronger in subjects whose age ≤ 40 years. Mediation analyses suggested that TyG index had a noteworthy indirect impact on the relationship between FT3/FT4, TFQIFT3 and MAFLD. Conclusion Increased FT3/FT4 and TFQIFT3 were significantly related to MAFLD prevalence in populations with normal thyroid function. TyG index partly mediated the relevance between FT3/FT4, TFQIFT3 and MAFLD.
Collapse
Affiliation(s)
- Huanxin Liu
- Health Examination Center, Hebei General Hospital, Shijiazhuang, Hebei, 050051, People’s Republic of China
| | - Yuling Xing
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei, 050051, People’s Republic of China
| | - Qian Nie
- Health Examination Center, Hebei General Hospital, Shijiazhuang, Hebei, 050051, People’s Republic of China
| | - Zhong Li
- Department of General Surgery, Shijiazhuang People’s Hospital, Shijiazhuang, 050011, People’s Republic of China
| | - Cuiqiao Meng
- Health Examination Center, Hebei General Hospital, Shijiazhuang, Hebei, 050051, People’s Republic of China
| | - Huijuan Ma
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei, 050051, People’s Republic of China
- Key Laboratory of Metabolic Disease in Hebei Province, Hebei General Hospital, Shijiazhuang, Hebei, 050051, People’s Republic of China
| |
Collapse
|
35
|
Basha A, May SC, Anderson RM, Samala N, Mirmira RG. Non-Alcoholic Fatty Liver Disease: Translating Disease Mechanisms into Therapeutics Using Animal Models. Int J Mol Sci 2023; 24:9996. [PMID: 37373143 PMCID: PMC10298283 DOI: 10.3390/ijms24129996] [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: 05/17/2023] [Revised: 06/06/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a range of pathologies arising from fat accumulation in the liver in the absence of excess alcohol use or other causes of liver disease. Its complications include cirrhosis and liver failure, hepatocellular carcinoma, and eventual death. NAFLD is the most common cause of liver disease globally and is estimated to affect nearly one-third of individuals in the United States. Despite knowledge that the incidence and prevalence of NAFLD are increasing, the pathophysiology of the disease and its progression to cirrhosis remain insufficiently understood. The molecular pathogenesis of NAFLD involves insulin resistance, inflammation, oxidative stress, and endoplasmic reticulum stress. Better insight into these molecular pathways would allow for therapies that target specific stages of NAFLD. Preclinical animal models have aided in defining these mechanisms and have served as platforms for screening and testing of potential therapeutic approaches. In this review, we will discuss the cellular and molecular mechanisms thought to contribute to NAFLD, with a focus on the role of animal models in elucidating these mechanisms and in developing therapies.
Collapse
Affiliation(s)
- Amina Basha
- Kovler Diabetes Center, Section of Adult and Pediatric Endocrinology, Diabetes and Metabolism, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Sarah C. May
- Kovler Diabetes Center, Section of Adult and Pediatric Endocrinology, Diabetes and Metabolism, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Ryan M. Anderson
- Kovler Diabetes Center, Section of Adult and Pediatric Endocrinology, Diabetes and Metabolism, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Niharika Samala
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Raghavendra G. Mirmira
- Kovler Diabetes Center, Section of Adult and Pediatric Endocrinology, Diabetes and Metabolism, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
36
|
Lee HK, Nam YH, Shin SW, Kim MC, An JI, Kim NW, Shim JH, Srinath S, Hong BN, Kwak JH, Kang TH. Erigeron annuus Extract Alleviates Insulin Resistance via Regulating the Expression of Mitochondrial Damage and Endoplasmic Reticulum Stress-Related Genes. Nutrients 2023; 15:2685. [PMID: 37375588 DOI: 10.3390/nu15122685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/06/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Diabetes is a prevalent and debilitating metabolic disorder affecting a large population worldwide. The condition is characterized by insulin resistance and impaired function of pancreatic β-cells, leading to elevated blood glucose levels. In this study, the antidiabetic effects of Erigeron annuus extract (EAE) on zebrafish with damaged pancreatic islets caused by insulin resistance were investigated. The study utilized the zebrafish model to monitor live pancreatic islets. RNA sequencing was also conducted to determine the mechanism by which EAE exerts its antidiabetic effect. The results showed that EAE was effective in recovering reduced islets in excess insulin-induced zebrafish. The effective concentration at 50% (EC50) of EAE was determined to be 0.54 μg/mL, while the lethal concentration at 50% (LC50) was calculated as 202.5 μg/mL. RNA sequencing indicated that the mode of action of EAE is related to its ability to induce mitochondrial damage and suppress endoplasmic reticulum stress. The findings of this study demonstrate the efficacy and therapeutic potential of EAE in treating insulin resistance in zebrafish. The results suggest that EAE may offer a promising approach for the management of diabetes by reducing mitochondrial damage and suppressing endoplasmic reticulum stress. Further research is required to establish the clinical application of EAE in diabetic patients.
Collapse
Affiliation(s)
- Hyo Kyu Lee
- Department of Oriental Medicine Biotechnology, College of Life Sciences and Graduate School of Biotechnology, Kyung Hee University, Global Campus, Yongin 17104, Gyeonggi-do, Republic of Korea
| | - Youn Hee Nam
- Department of Oriental Medicine Biotechnology, College of Life Sciences and Graduate School of Biotechnology, Kyung Hee University, Global Campus, Yongin 17104, Gyeonggi-do, Republic of Korea
| | - Sung Woo Shin
- Department of Oriental Medicine Biotechnology, College of Life Sciences and Graduate School of Biotechnology, Kyung Hee University, Global Campus, Yongin 17104, Gyeonggi-do, Republic of Korea
| | - Min Cheol Kim
- Department of Oriental Medicine Biotechnology, College of Life Sciences and Graduate School of Biotechnology, Kyung Hee University, Global Campus, Yongin 17104, Gyeonggi-do, Republic of Korea
| | - Jung In An
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Gyeonggi-do, Republic of Korea
| | - Na Woo Kim
- Department of Oriental Medicine Biotechnology, College of Life Sciences and Graduate School of Biotechnology, Kyung Hee University, Global Campus, Yongin 17104, Gyeonggi-do, Republic of Korea
| | - Ji Heon Shim
- Department of Oriental Medicine Biotechnology, College of Life Sciences and Graduate School of Biotechnology, Kyung Hee University, Global Campus, Yongin 17104, Gyeonggi-do, Republic of Korea
| | - Sunitha Srinath
- Department of Oriental Medicine Biotechnology, College of Life Sciences and Graduate School of Biotechnology, Kyung Hee University, Global Campus, Yongin 17104, Gyeonggi-do, Republic of Korea
| | - Bin Na Hong
- Department of Oriental Medicine Biotechnology, College of Life Sciences and Graduate School of Biotechnology, Kyung Hee University, Global Campus, Yongin 17104, Gyeonggi-do, Republic of Korea
| | - Jong Hwan Kwak
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Gyeonggi-do, Republic of Korea
| | - Tong Ho Kang
- Department of Oriental Medicine Biotechnology, College of Life Sciences and Graduate School of Biotechnology, Kyung Hee University, Global Campus, Yongin 17104, Gyeonggi-do, Republic of Korea
| |
Collapse
|
37
|
Chalhoub G, Jamnik A, Pajed L, Kolleritsch S, Hois V, Bagaric A, Prem D, Tilp A, Kolb D, Wolinski H, Taschler U, Züllig T, Rechberger GN, Fuchs C, Trauner M, Schoiswohl G, Haemmerle G. Carboxylesterase 2a deletion provokes hepatic steatosis and insulin resistance in mice involving impaired diacylglycerol and lysophosphatidylcholine catabolism. Mol Metab 2023; 72:101725. [PMID: 37059417 PMCID: PMC10148186 DOI: 10.1016/j.molmet.2023.101725] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/04/2023] [Accepted: 04/08/2023] [Indexed: 04/16/2023] Open
Abstract
OBJECTIVE Hepatic triacylglycerol accumulation and insulin resistance are key features of NAFLD. However, NAFLD development and progression are rather triggered by the aberrant generation of lipid metabolites and signaling molecules including diacylglycerol (DAG) and lysophosphatidylcholine (lysoPC). Recent studies showed decreased expression of carboxylesterase 2 (CES2) in the liver of NASH patients and hepatic DAG accumulation was linked to low CES2 activity in obese individuals. The mouse genome encodes several Ces2 genes with Ces2a showing highest expression in the liver. Herein we investigated the role of mouse Ces2a and human CES2 in lipid metabolism in vivo and in vitro. METHODS Lipid metabolism and insulin signaling were investigated in mice lacking Ces2a and in a human liver cell line upon pharmacological CES2 inhibition. Lipid hydrolytic activities were determined in vivo and from recombinant proteins. RESULTS Ces2a deficient mice (Ces2a-ko) are obese and feeding a high-fat diet (HFD) provokes severe hepatic steatosis and insulin resistance together with elevated inflammatory and fibrotic gene expression. Lipidomic analysis revealed a marked rise in DAG and lysoPC levels in the liver of Ces2a-ko mice fed HFD. Hepatic lipid accumulation in Ces2a deficiency is linked to lower DAG and lysoPC hydrolytic activities in liver microsomal preparations. Moreover, Ces2a deficiency significantly increases hepatic expression and activity of MGAT1, a PPAR gamma target gene, suggesting aberrant lipid signaling upon Ces2a deficiency. Mechanistically, we found that recombinant Ces2a and CES2 show significant hydrolytic activity towards lysoPC (and DAG) and pharmacological inhibition of CES2 in human HepG2 cells largely phenocopies the lipid metabolic changes present in Ces2a-ko mice including reduced lysoPC and DAG hydrolysis, DAG accumulation and impaired insulin signaling. CONCLUSIONS Ces2a and CES2 are critical players in hepatic lipid signaling likely via the hydrolysis of DAG and lysoPC at the ER.
Collapse
Affiliation(s)
- Gabriel Chalhoub
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Alina Jamnik
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Laura Pajed
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | | | - Victoria Hois
- Division of Endocrinology and Diabetology, Medical University of Graz, Austria
| | - Antonia Bagaric
- Department of Pharmacology and Toxicology, University of Graz, Graz, Austria
| | - Dominik Prem
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Anna Tilp
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Dagmar Kolb
- Core Facility Ultrastructure Analysis, Medical University of Graz, Graz, Austria
| | - Heimo Wolinski
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Ulrike Taschler
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Thomas Züllig
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | | | - Claudia Fuchs
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Michael Trauner
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Gabriele Schoiswohl
- Department of Pharmacology and Toxicology, University of Graz, Graz, Austria.
| | - Guenter Haemmerle
- Institute of Molecular Biosciences, University of Graz, Graz, Austria.
| |
Collapse
|
38
|
Varghese DS, Oommen D, John A, Ali BR. GRP78/BiP alleviates oxLDL-induced hepatotoxicity in familial hypercholesterolemia caused by missense variants of LDLR in a HepG2 cellular model. Lipids Health Dis 2023; 22:69. [PMID: 37248472 DOI: 10.1186/s12944-023-01835-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 05/13/2023] [Indexed: 05/31/2023] Open
Abstract
BACKGROUND AND AIMS The accumulation of misfolded proteins, encoded by genetic variants of functional genes leads to Endoplasmic Reticulum (ER) stress, which is a critical consequence in human disorders such as familial hypercholesterolemia, cardiovascular and hepatic diseases. In addition to the identification of ER stress as a contributing factor to pathogenicity, extensive studies on the role of oxidized Low-Density Lipoprotein (oxLDL) and its ill effects in expediting cardiovascular diseases and other metabolic comorbidities are well documented. However, the current understanding of its role in hepatic insults needs to be revised. This study elucidates the molecular mechanisms underlying the progression of oxLDL and ER stress-induced cytotoxicity in HepG2. METHODS HepG2 cells stably expressing wild-type Low-Density lipoprotein receptor (WT-LDLR) and missense variants of LDLR that are pathogenically associated with familial hypercholesterolemia were used as the in vitro models. The relative mRNA expression and protein profiles of ER stress sensors, inflammatory and apoptotic markers, together with cytotoxic assays and measurement of mitochondrial membrane potential, were carried out in HepG2 cells treated with 100 µg per ml oxLDL for 24 to 48 h. 1-way or 2-way ANOVA was used for statistical analyses of datasets. RESULTS ER stress responses are elicited along all three arms of the unfolded protein response (UPR), with adverse cytotoxic and inflammatory responses in oxLDL-treated conditions. Interestingly, oxLDL-treated ER-stressed HepG2 cells manifested intriguingly low expression of BiP- the master regulator of ER stress, as observed earlier by various researchers in liver biopsies of Non-Alcoholic Steatohepatitis (NASH) patients. This study shows that overexpression of BiP rescues hepatic cells from cytotoxic and inflammatory mechanisms instigated by ER stress in combination with oxLDL, along the ER and mitochondrial membrane and restores cellular homeostasis. CONCLUSION The data provide interesting leads that identify patients with familial hypercholesterolemia conditions and potentially other Endoplasmic Reticulum Associated Degradation (ERAD) diseases as highly susceptible to developing hepatic insults with molecular signatures like those manifested in Non-Alcoholic Fatty Liver Disease (NAFLD) and NASH. LIMITATIONS AND FUTURE PERSPECTIVES Although the use of HepG2 cells as the model is a major caveat of the study, the findings of this research may be used as the pilot study to expand further investigations in primary hepatocytes or iPSC- derived cellular models.
Collapse
Affiliation(s)
- Divya Saro Varghese
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Deepu Oommen
- Present Address: Indian Institute of Science, C V Raman Road, 560012, Bangalore, India
| | - Anne John
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Bassam R Ali
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates.
- Zayed Centre for Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates.
| |
Collapse
|
39
|
Rehati A, Abuduaini B, Liang Z, Chen D, He F. Identification of heat shock protein family A member 5 (HSPA5) targets involved in nonalcoholic fatty liver disease. Genes Immun 2023:10.1038/s41435-023-00205-y. [PMID: 37156995 DOI: 10.1038/s41435-023-00205-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 02/27/2023] [Accepted: 04/17/2023] [Indexed: 05/10/2023]
Abstract
Heat shock protein family A (Hsp70) member 5 (HSPA5) is an endoplasmic reticulum chaperone, which regulates cell metabolism, particularly lipid metabolism. While HSPA5's role in regulating cell function is well described, HSPA5 binding to RNA and its biological function in nonalcoholic fatty liver disease (NAFLD) is still lacking. In the present study, the ability of HSPA5 to modulate alternative splicing (AS) of cellular genes was assessed using Real-Time PCR on 89 NAFLD-associated genes. RNA immunoprecipitation coupled to RNA sequencing (RIP-Seq) assays were also performed to identify cellular mRNAs bound by HSPA5. We obtained the HSPA5-bound RNA profile in HeLa cells and peak calling analysis revealed that HSPA5 binds to coding genes and lncRNAs. Moreover, RIP-Seq assays demonstrated that HSPA5 immunoprecipitates specific cellular mRNAs such as EGFR, NEAT1, LRP1 and TGFß1, which are important in the pathology of NAFLD. Finally, HSPA5 binding sites may be associated with splicing sites. We used the HOMER algorithm to search for motifs enriched in coding sequence (CDs) peaks, which identified over-representation of the AGAG motif in both sets of immunoprecipitated peaks. HSPA5 regulated genes at the 5'UTR alternative splicing and introns and in an AG-rich sequence-dependent manner. We propose that the HSPA5-AGAG interaction might play an important role in regulating alternative splicing of NAFLD-related genes. This report is the first to demonstrate that HSPA5 regulated pre-RNA alternative splicing, stability, or translation and affected target protein(s) via binding to lncRNA and mRNA linked to NAFLD.
Collapse
Affiliation(s)
- Aliya Rehati
- Department of Gastroenterology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China
| | - Buzukela Abuduaini
- Department of Intensive Care Unit, The First Affiliated Hospital of Xinjiang Medical University, 393 South Li Yu Shan Road, Urumqi, 830054, Xinjiang, China.
| | - Zhao Liang
- Department of General Surgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China
| | - Dong Chen
- ABLife BioBigData Institute, Wuhan, 430075, Hubei, China
| | - Fangping He
- Department of Gastroenterology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China.
| |
Collapse
|
40
|
Jeon YG, Kim YY, Lee G, Kim JB. Physiological and pathological roles of lipogenesis. Nat Metab 2023; 5:735-759. [PMID: 37142787 DOI: 10.1038/s42255-023-00786-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 03/15/2023] [Indexed: 05/06/2023]
Abstract
Lipids are essential metabolites, which function as energy sources, structural components and signalling mediators. Most cells are able to convert carbohydrates into fatty acids, which are often converted into neutral lipids for storage in the form of lipid droplets. Accumulating evidence suggests that lipogenesis plays a crucial role not only in metabolic tissues for systemic energy homoeostasis but also in immune and nervous systems for their proliferation, differentiation and even pathophysiological roles. Thus, excessive or insufficient lipogenesis is closely associated with aberrations in lipid homoeostasis, potentially leading to pathological consequences, such as dyslipidaemia, diabetes, fatty liver, autoimmune diseases, neurodegenerative diseases and cancers. For systemic energy homoeostasis, multiple enzymes involved in lipogenesis are tightly controlled by transcriptional and post-translational modifications. In this Review, we discuss recent findings regarding the regulatory mechanisms, physiological roles and pathological importance of lipogenesis in multiple tissues such as adipose tissue and the liver, as well as the immune and nervous systems. Furthermore, we briefly introduce the therapeutic implications of lipogenesis modulation.
Collapse
Affiliation(s)
- Yong Geun Jeon
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Ye Young Kim
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Gung Lee
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Jae Bum Kim
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, South Korea.
| |
Collapse
|
41
|
Kim HJ, Kim M. Diallyl disulfide alleviates hypercholesterolemia induced by a western diet by suppressing endoplasmic reticulum stress in apolipoprotein E-deficient mice. BMC Complement Med Ther 2023; 23:141. [PMID: 37138269 PMCID: PMC10155326 DOI: 10.1186/s12906-023-03920-1] [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: 06/29/2022] [Accepted: 03/14/2023] [Indexed: 05/05/2023] Open
Abstract
BACKGROUND The endoplasmic reticulum (ER) plays a pivotal role in maintaining cellular metabolic homeostasis. ER stress refers to the accumulation of misfolded proteins, which can trigger an unfolded protein response for survival or death in the cells. Diallyl disulfide (DADS), a major active compound in garlic, has many health benefits for patients with metabolic diseases, especially cardiovascular or fatty liver diseases. However, its role in attenuating hypercholesterolemia by suppressing ER stress remains unknown. Therefore, in this study, we determined whether DADS supplementation could reduce ER stress in apolipoprotein E-deficient (ApoE-/-) mice fed a Western-type diet (WD). METHODS ApoE-/- mice were fed either a WD alone or a WD supplemented with 0.1% DADS for 12 weeks (n = 10). Levels of plasma total cholesterol, triglyceride, leptin, and insulin were determined. Western blotting was performed to measure protein levels involved in ER stress markers. Histology and Immunostaining were performed on aortic root sections to confirm the effect of DADS on histology and expression of ER chaperone protein GRP78. RESULTS The metabolic parameters showed that increases in fat weight, leptin resistance, and hypercholesterolemia were reversed in DADS-supplemented mice (p < 0.05). In addition, DADS ameliorated not only the protein of ER stress markers, phospho-eukaryotic initiation factor 2 subunit alpha and C/EBP homologous protein in the liver (p < 0.05) but also glucose-related protein 78 localization in the aorta. CONCLUSIONS This indicates that DADS inhibits diet-induced hypercholesterolemia, at least in parts by regulating ER stress markers. DADS may be a good candidate for treating individuals with diet-induced hypercholesterolemia.
Collapse
Affiliation(s)
- Hyun Ju Kim
- Kimchi Functionality Research Group, World Institute of Kimchi, Nam-Gu, Gwangju, 61755, Republic of Korea.
| | - Mijeong Kim
- Department of Food Science and Nutrition and Kimchi Research Institute, Pusan National University, 2, Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan, 46241, Republic of Korea
| |
Collapse
|
42
|
Chen S, Su Y, Zhang M, Zhang Y, Xiu P, Luo W, Zhang Q, Zhang X, Liang H, Lee APW, Shao L, Xiu J. Insights into the toxicological effects of nanomaterials on atherosclerosis: mechanisms involved and influence factors. J Nanobiotechnology 2023; 21:140. [PMID: 37118804 PMCID: PMC10148422 DOI: 10.1186/s12951-023-01899-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 04/16/2023] [Indexed: 04/30/2023] Open
Abstract
Atherosclerosis is one of the most common types of cardiovascular disease and is driven by lipid accumulation and chronic inflammation in the arteries, which leads to stenosis and thrombosis. Researchers have been working to design multifunctional nanomedicines with the ability to target, diagnose, and treat atherosclerosis, but recent studies have also identified that nanomaterials can cause atherosclerosis. Therefore, this review aims to outline the molecular mechanisms and physicochemical properties of nanomaterials that promote atherosclerosis. By analyzing the toxicological effects of nanomaterials on cells involved in the pathogenesis of atherosclerosis such as vascular endothelial cells, vascular smooth muscle cells and immune cells, we aim to provide new perspectives for the prevention and treatment of atherosclerosis, and raise awareness of nanotoxicology to advance the clinical translation and sustainable development of nanomaterials.
Collapse
Affiliation(s)
- Siyu Chen
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yuan Su
- Stomatology Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, 528300, China
| | - Manjin Zhang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Yulin Zhang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Peiming Xiu
- Guangdong Medical University, Dongguan, 523808, China
| | - Wei Luo
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Qiuxia Zhang
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xinlu Zhang
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Hongbin Liang
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Alex Pui-Wai Lee
- Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Longquan Shao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China.
| | - Jiancheng Xiu
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
43
|
Mohan S, Nair A, Poornima MS, Raghu KG. Vanillic acid mitigates hyperinsulinemia induced ER stress mediated altered calcium homeostasis, MAMs distortion and surplus lipogenesis in HepG2 cells. Chem Biol Interact 2023; 375:110365. [PMID: 36764371 DOI: 10.1016/j.cbi.2023.110365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 10/11/2022] [Accepted: 01/23/2023] [Indexed: 02/10/2023]
Abstract
Hyperinsulinemia (HI) induced insulin resistance (IR) and associated pathologies are the burning and unsolvable issues in diabetes treatment. The cellular, molecular and biochemical events associated with HI are not yet elucidated. Similarly, no focused research on designing therapeutic strategies with natural products for attenuation of HI are seen in literature. Keeping this in mind we planned the present study to evaluate the alterations occurring at ER/Ca2+ homeostasis/mitochondria associated endoplasmic reticulum membranes (MAMs) in HepG2 cells during HI and to evaluate the possible beneficial effect of vanillic acid (VA) to mitigate the complications. An in vitro model of HI was established by treating HepG2 cells with human insulin (1 μM) for 24 h. Then, ER stress, Ca2+ homeostasis, MAMs, IR and hepatic lipogenesis were studied at protein level. Various proteins critical to ER, Ca2+ homeostasis and MAMs such as p-IRE-1α, ATF6, p-PERK, p-eIF2α, CHOP, XBP1, p-CAMKII, InsP3R, SERCA, JNK, GRP78, VDAC, Cyp D, GRP75, MFN2, PTEN and mTORC were studied and found altered significantly causing ER stress, defect in Ca2+ movements and distortion of MAMs. The decreased expression of IRS2 and an unaltered expression of IRS1 confirmed the development of selective insulin resistance in hepatocytes during HI and this was the crucial factor for the progression of the hepatic lipid accumulation. We found simultaneous treatment of VA is beneficial up to a certain extent to protect HepG2 cells from the adverse effect of HI via its antioxidant, antilipogenic, mitochondrial and ER protection properties.
Collapse
Affiliation(s)
- Sreelekshmi Mohan
- Biochemistry and Molecular Mechanism Laboratory, Agro-processing and Technology Division, Council of Scientific and Industrial Research (CSIR) - National Institute for Interdisciplinary Science and Technology (NIIST), Thiruvananthapuram, 695019, Kerala, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Anupama Nair
- Biochemistry and Molecular Mechanism Laboratory, Agro-processing and Technology Division, Council of Scientific and Industrial Research (CSIR) - National Institute for Interdisciplinary Science and Technology (NIIST), Thiruvananthapuram, 695019, Kerala, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - M S Poornima
- Biochemistry and Molecular Mechanism Laboratory, Agro-processing and Technology Division, Council of Scientific and Industrial Research (CSIR) - National Institute for Interdisciplinary Science and Technology (NIIST), Thiruvananthapuram, 695019, Kerala, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - K G Raghu
- Biochemistry and Molecular Mechanism Laboratory, Agro-processing and Technology Division, Council of Scientific and Industrial Research (CSIR) - National Institute for Interdisciplinary Science and Technology (NIIST), Thiruvananthapuram, 695019, Kerala, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
44
|
Ramatchandirin B, Pearah A, He L. Regulation of Liver Glucose and Lipid Metabolism by Transcriptional Factors and Coactivators. Life (Basel) 2023; 13:life13020515. [PMID: 36836874 PMCID: PMC9962321 DOI: 10.3390/life13020515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/08/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023] Open
Abstract
The prevalence of nonalcoholic fatty liver disease (NAFLD) worldwide is on the rise and NAFLD is becoming the most common cause of chronic liver disease. In the USA, NAFLD affects over 30% of the population, with similar occurrence rates reported from Europe and Asia. This is due to the global increase in obesity and type 2 diabetes mellitus (T2DM) because patients with obesity and T2DM commonly have NAFLD, and patients with NAFLD are often obese and have T2DM with insulin resistance and dyslipidemia as well as hypertriglyceridemia. Excessive accumulation of triglycerides is a hallmark of NAFLD and NAFLD is now recognized as the liver disease component of metabolic syndrome. Liver glucose and lipid metabolisms are intertwined and carbon flux can be used to generate glucose or lipids; therefore, in this review we discuss the important transcription factors and coactivators that regulate glucose and lipid metabolism.
Collapse
Affiliation(s)
| | - Alexia Pearah
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Ling He
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, 600 N. Wolfe St, Baltimore, MD 21287, USA
- Correspondence: ; Tel.: +1-410-502-5765; Fax: +1-410-502-5779
| |
Collapse
|
45
|
Ajoolabady A, Kaplowitz N, Lebeaupin C, Kroemer G, Kaufman RJ, Malhi H, Ren J. Endoplasmic reticulum stress in liver diseases. Hepatology 2023; 77:619-639. [PMID: 35524448 PMCID: PMC9637239 DOI: 10.1002/hep.32562] [Citation(s) in RCA: 79] [Impact Index Per Article: 79.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 03/05/2022] [Accepted: 03/08/2022] [Indexed: 02/02/2023]
Abstract
The endoplasmic reticulum (ER) is an intracellular organelle that fosters the correct folding of linear polypeptides and proteins, a process tightly governed by the ER-resident enzymes and chaperones. Failure to shape the proper 3-dimensional architecture of proteins culminates in the accumulation of misfolded or unfolded proteins within the ER, disturbs ER homeostasis, and leads to canonically defined ER stress. Recent studies have elucidated that cellular perturbations, such as lipotoxicity, can also lead to ER stress. In response to ER stress, the unfolded protein response (UPR) is activated to reestablish ER homeostasis ("adaptive UPR"), or, conversely, to provoke cell death when ER stress is overwhelmed and sustained ("maladaptive UPR"). It is well documented that ER stress contributes to the onset and progression of multiple hepatic pathologies including NAFLD, alcohol-associated liver disease, viral hepatitis, liver ischemia, drug toxicity, and liver cancers. Here, we review key studies dealing with the emerging role of ER stress and the UPR in the pathophysiology of liver diseases from cellular, murine, and human models. Specifically, we will summarize current available knowledge on pharmacological and non-pharmacological interventions that may be used to target maladaptive UPR for the treatment of nonmalignant liver diseases.
Collapse
Affiliation(s)
- Amir Ajoolabady
- Department of Cardiology, Shanghai Institute for Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, China
| | - Neil Kaplowitz
- Division of Gastrointestinal and Liver Disease, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- USC Research Center for Liver Disease, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Cynthia Lebeaupin
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Randal J. Kaufman
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Harmeet Malhi
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Jun Ren
- Department of Cardiology, Shanghai Institute for Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, China
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
46
|
Arruda AP, Parlakgül G. Endoplasmic Reticulum Architecture and Inter-Organelle Communication in Metabolic Health and Disease. Cold Spring Harb Perspect Biol 2023; 15:cshperspect.a041261. [PMID: 35940911 PMCID: PMC9899651 DOI: 10.1101/cshperspect.a041261] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The endoplasmic reticulum (ER) is a key organelle involved in the regulation of lipid and glucose metabolism, proteostasis, Ca2+ signaling, and detoxification. The structural organization of the ER is very dynamic and complex, with distinct subdomains such as the nuclear envelope and the peripheral ER organized into ER sheets and tubules. ER also forms physical contact sites with all other cellular organelles and with the plasma membrane. Both form and function of the ER are highly adaptive, with a potent capacity to respond to transient changes in environmental cues such as nutritional fluctuations. However, under obesity-induced chronic stress, the ER fails to adapt, leading to ER dysfunction and the development of metabolic pathologies such as insulin resistance and fatty liver disease. Here, we discuss how the remodeling of ER structure and contact sites with other organelles results in diversification of metabolic function and how perturbations to this structural flexibility by chronic overnutrition contribute to ER dysfunction and metabolic pathologies in obesity.
Collapse
Affiliation(s)
- Ana Paula Arruda
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, California 94720, USA.,Chan Zuckerberg Biohub, San Francisco, California 94158, USA
| | - Güneş Parlakgül
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, California 94720, USA.,Sabri Ülker Center for Metabolic Research and Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| |
Collapse
|
47
|
Rhinacanthin C Ameliorates Insulin Resistance and Lipid Accumulation in NAFLD Mice via the AMPK/SIRT1 and SREBP-1c/FAS/ACC Signaling Pathways. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2023; 2023:6603522. [PMID: 36660274 PMCID: PMC9845057 DOI: 10.1155/2023/6603522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 12/09/2022] [Accepted: 12/22/2022] [Indexed: 01/12/2023]
Abstract
Rhinacanthin C (RC) is a naphthoquinone ester with an anti-inflammatory activity extracted from Rhinacanthus nasutus (L.) Kurz (Rn). It has been proven to improve hyperglycemia and hyperlipidemia, but the prevention and mechanism of RC in nonalcoholic fatty liver disease (NAFLD) are not clear. In the current study, we first extracted RC from Rn using ethyl acetate and identified it by HPLC, MS, and NMR. At the same time, molecular docking analysis of RC with AMPK and SREBP-1c was performed using AutoDock software. In addition, the mouse model of NAFLD was induced by a high-fat diet in vivo, and low, medium, and high concentrations of RC were used for intervention. The results showed that RC significantly reduced the body mass and liver body coefficient of NAFLD mice, inhibited liver inflammation and fat accumulation, and improved insulin resistance. Further studies showed that RC significantly reduced the levels of serum leptin and resistin, upregulated the expression levels of adiponectin and adiponectin receptor in the liver, and inhibited the expression levels of MCP-1, TNF-α, and IL-6. In terms of mechanism, RC upregulates the expression of p-AMPK and SIRT1 and downregulates the expression of p-p65, SREBP-1c, Fas, Acc-α, PPAR-γ, and SCD1. These studies suggest that RC improves insulin resistance and lipid accumulation in NAFLD by activating the AMPK/SIRT1 and SREBP-1c/Fas/ACC pathways, respectively.
Collapse
|
48
|
Chen G, Wei T, Ju F, Li H. Protein quality control and aggregation in the endoplasmic reticulum: From basic to bedside. Front Cell Dev Biol 2023; 11:1156152. [PMID: 37152279 PMCID: PMC10154544 DOI: 10.3389/fcell.2023.1156152] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 04/10/2023] [Indexed: 05/09/2023] Open
Abstract
Endoplasmic reticulum (ER) is the largest membrane-bound compartment in all cells and functions as a key regulator in protein biosynthesis, lipid metabolism, and calcium balance. Mammalian endoplasmic reticulum has evolved with an orchestrated protein quality control system to handle defective proteins and ensure endoplasmic reticulum homeostasis. Nevertheless, the accumulation and aggregation of misfolded proteins in the endoplasmic reticulum may occur during pathological conditions. The inability of endoplasmic reticulum quality control system to clear faulty proteins and aggregates from the endoplasmic reticulum results in the development of many human disorders. The efforts to comprehensively understand endoplasmic reticulum quality control network and protein aggregation will benefit the diagnostics and therapeutics of endoplasmic reticulum storage diseases. Herein, we overview recent advances in mammalian endoplasmic reticulum protein quality control system, describe protein phase transition model, and summarize the approaches to monitor protein aggregation. Moreover, we discuss the therapeutic applications of enhancing endoplasmic reticulum protein quality control pathways in endoplasmic reticulum storage diseases.
Collapse
Affiliation(s)
- Guofang Chen
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Tingyi Wei
- Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Precision Medicine, Shanghai, China
| | - Furong Ju
- Ming Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Sha Tin, Hong kong SAR, China
| | - Haisen Li
- School of Life Sciences, Fudan University, Shanghai, China
- AoBio Medical, Shanghai, China
- *Correspondence: Haisen Li,
| |
Collapse
|
49
|
Celik C, Lee SYT, Yap WS, Thibault G. Endoplasmic reticulum stress and lipids in health and diseases. Prog Lipid Res 2023; 89:101198. [PMID: 36379317 DOI: 10.1016/j.plipres.2022.101198] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 11/03/2022] [Accepted: 11/09/2022] [Indexed: 11/14/2022]
Abstract
The endoplasmic reticulum (ER) is a complex and dynamic organelle that regulates many cellular pathways, including protein synthesis, protein quality control, and lipid synthesis. When one or multiple ER roles are dysregulated and saturated, the ER enters a stress state, which, in turn, activates the highly conserved unfolded protein response (UPR). By sensing the accumulation of unfolded proteins or lipid bilayer stress (LBS) at the ER, the UPR triggers pathways to restore ER homeostasis and eventually induces apoptosis if the stress remains unresolved. In recent years, it has emerged that the UPR works intimately with other cellular pathways to maintain lipid homeostasis at the ER, and so does at cellular levels. Lipid distribution, along with lipid anabolism and catabolism, are tightly regulated, in part, by the ER. Dysfunctional and overwhelmed lipid-related pathways, independently or in combination with ER stress, can have reciprocal effects on other cellular functions, contributing to the development of diseases. In this review, we summarize the current understanding of the UPR in response to proteotoxic stress and LBS and the breadth of the functions mitigated by the UPR in different tissues and in the context of diseases.
Collapse
Affiliation(s)
- Cenk Celik
- School of Biological Sciences, Nanyang Technological University, Singapore
| | | | - Wei Sheng Yap
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Guillaume Thibault
- School of Biological Sciences, Nanyang Technological University, Singapore; Mechanobiology Institute, National University of Singapore, Singapore; Institute of Molecular and Cell Biology, A*STAR, Singapore.
| |
Collapse
|
50
|
Chen W, Yin H, Xiao J, Liu W, Qu Q, Gong F, He X. The effect of aging on glucose metabolism improvement after Roux-en-Y gastric bypass in type 2 diabetes rats. Nutr Diabetes 2022; 12:51. [PMID: 36564376 PMCID: PMC9789110 DOI: 10.1038/s41387-022-00229-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 12/01/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND This study aimed to investigate the effect of aging on glucose metabolism improvement after Roux-en-Y gastric bypass (RYGB) in rat models with type 2 diabetes mellitus (T2DM). METHODS Twenty aged Goto-Kakizaki rats were randomly assigned into RYGB-A group and sham RYGB (SR-A) group, and 10 adult Goto-Kakizaki rats also accept RYGB procedures (RYGB-Y). Glucose metabolism, resting energy expenditure (REE), glucagon-like peptide-1 (GLP-1) and total bile acid level were measured. RESULTS RYGB could significantly improve glucose metabolism in aged diabetic rats. The fasting blood glucose level in the RYGB-A group decreased from 15.8 ± 1.1 mmol/l before surgery to 12.3 ± 1.5 mmol/l 16 weeks after surgery (P < 0.01), and the AUCOGTT value decreased from 2603.9 ± 155.4 (mmol/l) min to 2299.9 ± 252.8 (mmol/l) min (P = 0.08). The decrease range of fasting blood glucose in the RYGB-A group was less than that in the RYGB-Y group (20.5% ± 6.5% vs. 40.6% ± 10.6%, P < 0.01), so is the decrease range of AUCOGTT value (11.6% ± 14.8% vs. 38.5% ± 8.3%, P < 0.01). Moreover, at the 16th postoperative week, the increase range of REE of the RYGB-A group was lower than that of the RYGB-Y group (15.3% ± 11.1% vs. 29.1% ± 12.1%, P = 0.04). The increased range of bile acid of the RYGB-A group was less than that of the RYGB-Y group (80.2 ± 59.3 % vs.212.3 ± 139.0 %, P < 0.01). The GLP-1 level of the RYGB-A group was less than that of the RYGB-Y group (12.8 ± 3.9 pmol/L vs. 18.7 ± 5.6 pmol/L, P = 0.02). There was no significant difference between the RYGB-A group and the RYGB-Y group in the level of the triiodothyronine level. CONCLUSIONS RYGB could induce a glucose metabolism improvement in aged diabetic rats, and aging might moderate the effect of RYGB.
Collapse
Affiliation(s)
- Weijie Chen
- grid.413106.10000 0000 9889 6335Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Shuaifuyuan 1#, Beijing, 100730 PR China
| | - Haixin Yin
- grid.413106.10000 0000 9889 6335Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Shuaifuyuan 1#, Beijing, 100730 PR China
| | - Jianchun Xiao
- grid.413106.10000 0000 9889 6335Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Shuaifuyuan 1#, Beijing, 100730 PR China
| | - Wei Liu
- grid.413106.10000 0000 9889 6335Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Shuaifuyuan 1#, Beijing, 100730 PR China
| | - Qiang Qu
- grid.413106.10000 0000 9889 6335Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Shuaifuyuan 1#, Beijing, 100730 PR China
| | - Fengying Gong
- grid.413106.10000 0000 9889 6335Department of Endocrinology, Key Laboratory of Endocrinology of the Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Shuaifuyuan 1#, Beijing, 100730 PR China
| | - Xiaodong He
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Shuaifuyuan 1#, Beijing, 100730, PR China.
| |
Collapse
|