1
|
White RB, Wild AR, O'Leary TP, Thompson AJ, Flibotte S, Peng A, Rogalski JC, Mair M, Derhami N, Bamji SX. The X-Linked Intellectual Disability Gene, ZDHHC9 , Is Important for Oligodendrocyte Subtype Determination and Myelination. Glia 2025; 73:1452-1466. [PMID: 40105030 PMCID: PMC12121472 DOI: 10.1002/glia.70016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 03/20/2025]
Abstract
Two percent of patients with X-linked intellectual disability (XLID) exhibit loss-of-function mutations in the enzyme, ZDHHC9. One of the main anatomical deficits observed in these patients is a decrease in corpus callosum volume and a concurrent disruption in white matter integrity. In this study, we demonstrate that deletion of Zdhhc9 in mice disrupts the balance of mature oligodendrocyte subtypes within the corpus callosum. While overall mature oligodendrocyte numbers are unchanged, there is a marked increase in MOL5/6 cells that are enriched in genes associated with cell adhesion and synapses, and a concomitant decrease in MOL2/3 cells that are enriched in genes associated with myelination. In line with this, we observed a decrease in the density of myelinated axons and disruptions in myelin compaction in the corpus callosum of Zdhhc9 knockout mice. RNA sequencing and proteomic analysis further revealed a reduction in genes and proteins essential for lipid metabolism, cholesterol synthesis, gene expression, and myelin compaction, offering insights into the underlying mechanisms of the pathology. These findings reveal a previously underappreciated and fundamental role for ZDHHC9 and protein palmitoylation in regulating oligodendrocyte subtype determination and myelinogenesis, offering mechanistic insights into the deficits observed in white matter volume in patients with mutations in ZDHHC9.
Collapse
Affiliation(s)
- Rocio B. White
- Department of Cellular and Physiological Sciences, Life Sciences Institute and Djavad Mowafaghian Centre for Brain HealthUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Angela R. Wild
- Department of Cellular and Physiological Sciences, Life Sciences Institute and Djavad Mowafaghian Centre for Brain HealthUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Timothy P. O'Leary
- Department of Cellular and Physiological Sciences, Life Sciences Institute and Djavad Mowafaghian Centre for Brain HealthUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Andrew J. Thompson
- Department of Cellular and Physiological Sciences, Life Sciences Institute and Djavad Mowafaghian Centre for Brain HealthUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Stephane Flibotte
- Life Sciences Institute Bioinformatics FacilityUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Angie Peng
- Department of Cellular and Physiological Sciences, Life Sciences Institute and Djavad Mowafaghian Centre for Brain HealthUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Jason C. Rogalski
- Department of Biochemistry and Molecular BiologyUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Mila Mair
- Department of Cellular and Physiological Sciences, Life Sciences Institute and Djavad Mowafaghian Centre for Brain HealthUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Neeki Derhami
- Department of Cellular and Physiological Sciences, Life Sciences Institute and Djavad Mowafaghian Centre for Brain HealthUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Shernaz X. Bamji
- Department of Cellular and Physiological Sciences, Life Sciences Institute and Djavad Mowafaghian Centre for Brain HealthUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| |
Collapse
|
2
|
Alluri A, Saxena P, Mishra A, Gutti RK. Association of long non-coding RNA in lipid metabolism: Implications in leukemia. Int J Biochem Cell Biol 2025; 184:106785. [PMID: 40246061 DOI: 10.1016/j.biocel.2025.106785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 04/09/2025] [Accepted: 04/15/2025] [Indexed: 04/19/2025]
Abstract
Cancer has high mortality rate and occupies second position among major diseases. Despite extensive research and therapies, in every nook and corner of the world, death rate is increasing exponentially. Hallmarks of cancer are benchmarks of cancer cells describing the fundamental principle and capabilities of the cells transforming from normal to malignant tumour. One of the major ones among them is the deregulation of cellular metabolism or metabolic reprogramming, involving alterations in glucose and lipid metabolism. Progressive research in this area has visualized the vital role of lncRNAs in lipid metabolism with respect to AML. lncRNAs involve in various cellular processes and also contribute for significant functions of the cell like chromatin remodelling, transcriptional activation and repression, gene regulation, immune response, cell differentiation, and cell cycle regulation, in addition to oncogenic processes such as proliferation, angiogenesis, migration, and apoptosis. Structural similarities are observed among mRNAs and lncRNAs in terms of poly A-tail and 5' cap however protein-coding regions are lacking. A large body of evidence has shown that lncRNAs directly or indirectly mediate lipid metabolism by activating downstream genes. Considering their potential involvement in leukemia, these lncRNAs can be explored and considered as biomarkers for therapeutics, prognosis, and diagnosis. The present review is planned to summarize the functional classification of lncRNAs, the role of lipid metabolism in cancer, different lncRNAs involved in leukemia, and different cancer types related to lipid metabolism.
Collapse
Affiliation(s)
- Anjani Alluri
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, (PO) Gachibowli, Hyderabad, TS 500046, India
| | - Pallavi Saxena
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, (PO) Gachibowli, Hyderabad, TS 500046, India
| | - Amit Mishra
- Department of Bioscience & Bioengineering, Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, RJ 342037, India
| | - Ravi Kumar Gutti
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, (PO) Gachibowli, Hyderabad, TS 500046, India.
| |
Collapse
|
3
|
Ning B, Wang SA, Young MJ, Chen YC, Hung Y, Huong TT, Chang WC, Wang YC, Yu ML, Hsu KC, Hung JJ. USP24 upregulation stabilizes PKA-Cα to promote lipogenesis, inflammation, and fibrosis during MASH progression. J Biomed Sci 2025; 32:54. [PMID: 40448065 DOI: 10.1186/s12929-025-01148-4] [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: 02/20/2025] [Accepted: 05/20/2025] [Indexed: 06/02/2025] Open
Abstract
BACKGROUND Ubiquitin-specific peptidase 24 (USP24), a deubiquitinating enzyme, regulates protein stability by removing ubiquitin. This study investigates the role of UPS24 in lipid metabolism, inflammation, and fibrosis. It also explores the effect of targeting USP24 on metabolic disorders, focusing on high-fat diet (HFD)-induced obesity and liver diseases. METHODS This study utilized CRISPR/Cas9 to create functional knockout mice (USP24C1695A) and treated HFD-fed mice with USP24 inhibitor (USP24-i-101). The effects of USP24 inhibition or knockout on 3T3-L1 derived adipocytes, primary hepatocytes, hepatic stellate cells, and murine hepatocyte cell line AML12 (alpha mouse liver 12) cells were assessed with RNA-sequencing. Molecular mechanisms and the interaction between USP24 and PKA-Cα were studied with co-immunoprecipitation. Downstream signaling pathways involving CREB, SREBP1, PPARγ, and C/EBPβ, as well as USP24 role in liver inflammation and fibrosis, were studied using western blot and real-time PCR. Clinical and animal tissue samples were examined with immunohistochemistry to identify the correlations between USP24 and metabolic-associated liver diseases. RESULTS Knockout or inhibition of USP24 reduced body weight, lipid accumulation, inflammation, and fibrosis in HFD-fed mice. The expression of genes related to lipogenesis, inflammation, and fibrosis was downregulated in USP24C1695A mice and those treated with USP24 inhibitor (USP24-i-101). USP24 inhibition decreased lipid droplet accumulation in adipocytes and hepatocytes, suppressed inflammation in hepatocytes and AML12 cells, and reduced fibrosis in hepatic stellate cells. Mechanistically, USP24 expression was upregulated by PKA activation during adipocyte differentiation, leading to increased PKA-Cα stability and CREB phosphorylation, which promoted lipogenic gene expression. Free fatty acids (FFA) increased USP24 expression, activating NF-κB and TGFβ pathways to induce inflammation (Cox2) and fibrosis (α-SMA). USP24 was highly expressed in patients with metabolic dysfunction-associated steatohepatitis (MASH) and correlated with Cox2 and α-SMA levels.
Collapse
Affiliation(s)
- Beh Ning
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, 701, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shao-An Wang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ming-Jer Young
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, 701, Taiwan
| | - Yung-Ching Chen
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, 701, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yun Hung
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tran Thu Huong
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, 701, Taiwan
| | - Wen-Chang Chang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yi-Ching Wang
- Institute of Pharmacology, National Cheng Kung University, Tainan, Taiwan
| | - Ming-Lung Yu
- Center of Excellence for Metabolic Associated Fatty Liver Disease, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Kai-Cheng Hsu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Jan-Jong Hung
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, 701, Taiwan.
| |
Collapse
|
4
|
Xia M, Li J, Martinez Aguilar LM, Wang J, Trillos Almanza MC, Li Y, Buist-Homan M, Moshage H. Arctigenin Attenuates Hepatic Stellate Cell Activation via Endoplasmic Reticulum-Associated Degradation (ERAD)-Mediated Restoration of Lipid Homeostasis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025. [PMID: 40415275 DOI: 10.1021/acs.jafc.5c01366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2025]
Abstract
Arctigenin, a natural lignan from Arctium lappa L., exhibits potent antifibrotic activity, yet its molecular mechanisms remain unclear. Endoplasmic reticulum (ER) stress is known to promote hepatic stellate cell (HSC) activation and liver fibrosis. This study investigates the therapeutic potential of arctigenin in HSC activation through ER stress modulation. Primary rat HSCs were activated (3-7 days) and treated with tunicamycin (ER stress inducer) or 4-PBA (ER stress inhibitor). Arctigenin attenuated ER stress markers (e.g., GRP78) and suppressed the expression of fibrotic marker α-SMA in ER stress-challenged activating (day 3) and activated (day 7) HSCs. Arctigenin restored lipid homeostasis by modulation of both lipogenesis (via Dgat2 and Ppar-γ upregulation) and lipolysis (suppression via ATGL inhibition). ER stress activated ER-associated degradation (ERAD), triggering the formation of small lipid droplets (LD). Arctigenin normalized the ERAD activity, thereby rescuing LD integrity and suppressing HSC activation. Our findings demonstrate that arctigenin mitigates HSC activation by suppressing ER stress and restoring lipid homeostasis via modulating ERAD-mediated lipid dysregulation. As a dietary and medicinal compound, arctigenin emerges as a promising therapeutic candidate for liver fibrosis.
Collapse
Affiliation(s)
- Mengmeng Xia
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen 9713 GZ, The Netherlands
| | - Jia Li
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen 9713 GZ, The Netherlands
| | - Lizbeth Magnolia Martinez Aguilar
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen 9713 GZ, The Netherlands
| | - Junyu Wang
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen 9713 GZ, The Netherlands
| | - Maria Camila Trillos Almanza
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen 9713 GZ, The Netherlands
| | - Yakun Li
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen 9713 GZ, The Netherlands
| | - Manon Buist-Homan
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen 9713 GZ, The Netherlands
| | - Han Moshage
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen 9713 GZ, The Netherlands
| |
Collapse
|
5
|
Tang H, Zhang Y, Zhao D, Guo M, Yuan X, Wang X. Unlocking the lipid code: SREBPs as key drivers in gastrointestinal tumour metabolism. Lipids Health Dis 2025; 24:190. [PMID: 40413517 DOI: 10.1186/s12944-025-02612-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 05/15/2025] [Indexed: 05/27/2025] Open
Abstract
In recent years, metabolic reprogramming has emerged as a significant breakthrough in elucidating the onset and progression of gastrointestinal (GI) malignancies. As central regulatory hubs for lipid metabolism, sterol regulatory element binding proteins (SREBPs) integrate dietary metabolic signals and carcinogenic stimuli through subtype-specific mechanisms, thereby promoting malignant tumour phenotypes. In this review, we first present the molecular background, structural characteristics, and posttranscriptional regulatory networks associated with SREBPs. We subsequently describe a systematic analysis of the distinct activation patterns of SREBPs in liver, gastric, colorectal, and other gastrointestinal cancers. Furthermore, we explore targeted intervention strategies for different SREBP subtypes, including small molecule inhibitors (such as fatostatin, which inhibits SREBP cleavage), natural compounds (such as berberine, which modulates the AMPK/mTOR pathway), and statin-mediated inhibition of the mevalonic acid pathway. These strategies may enhance tumour cell sensitivity to chemotherapeutic agents (such as 5-FU, gezil, and tabine) and improve the response to synergistic chemoradiotherapy by reversing adaptive metabolic resistance driven by the tumour microenvironment. Through this review, we hope to provide new insights into precise interventions targeting various subtypes of the SREBP molecule.
Collapse
Affiliation(s)
- Haowen Tang
- Department of Thoracic Oncology, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yuting Zhang
- Department of Thoracic Oncology, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Danni Zhao
- Department of Thoracic Oncology, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Minjie Guo
- Department of Thoracic Oncology, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xiao Yuan
- Cancer Institute of Jiangsu University, Zhenjiang, China.
| | - Xu Wang
- Department of Thoracic Oncology, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, China.
| |
Collapse
|
6
|
Hajirahimkhan A, Brown KA, Clare SE, Khan SA. SREBP1-Dependent Metabolism as a Potential Target for Breast Cancer Risk Reduction. Cancers (Basel) 2025; 17:1664. [PMID: 40427160 PMCID: PMC12110029 DOI: 10.3390/cancers17101664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2025] [Revised: 05/05/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
There are an estimated 10 million U [...].
Collapse
Affiliation(s)
- Atieh Hajirahimkhan
- Division of Breast Surgery, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, 303 E Superior, 4-220, Chicago, IL 60611, USA; (S.E.C.); (S.A.K.)
| | - Kristy A. Brown
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA;
- Cancer Prevention and Control Program, University of Kansas Cancer Center, Kansas City, KS 66160, USA
| | - Susan E. Clare
- Division of Breast Surgery, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, 303 E Superior, 4-220, Chicago, IL 60611, USA; (S.E.C.); (S.A.K.)
| | - Seema Ahsan Khan
- Division of Breast Surgery, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, 303 E Superior, 4-220, Chicago, IL 60611, USA; (S.E.C.); (S.A.K.)
| |
Collapse
|
7
|
Serio S, Papait R. Histone Methyltransferase SETD2: A Key Player in Cardiometabolic HFpEF. Circ Res 2025; 136:1096-1098. [PMID: 40339050 DOI: 10.1161/circresaha.125.326463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Affiliation(s)
- Simone Serio
- Institute of Genetic and Biomedical Research (IRGB), Milan Unit, National Research Council of Italy (S.S.)
- Department of Cardiovascular Medicine, IRCCS Humanitas Research Hospital, Rozzano, Italy (S.S., R.P.)
| | - Roberto Papait
- Department of Cardiovascular Medicine, IRCCS Humanitas Research Hospital, Rozzano, Italy (S.S., R.P.)
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy (R.P.)
| |
Collapse
|
8
|
Venturi S, Rendine M, Marino M, Klimis-Zacas D, Riso P, Del Bo' C. Differential Effects of Wild Blueberry (Poly)Phenol Metabolites in Modulating Lipid Metabolism and Oxidative Stress in 3T3-L1 Adipocytes. Mol Nutr Food Res 2025:e70101. [PMID: 40345987 DOI: 10.1002/mnfr.70101] [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: 12/12/2024] [Revised: 04/01/2025] [Accepted: 04/28/2025] [Indexed: 05/11/2025]
Abstract
Adipocyte hypertrophy, driven by lipid accumulation, is crucial in the development of obesity. Wild blueberry (WB; Vaccinium angustifolium) (poly)phenols (PPs) metabolites may modulate adipogenesis and the development of obesity. This study examines WB PP metabolites' effects on lipid accumulation, lipid metabolism, and oxidative stress in mature 3T3-L1 adipocytes. Differentiated 3T3-L1 adipocytes were treated for 48 h with free fatty acids (FFAs; oleic/palmitic acid 750 µM, 2:1 ratio) and WB-derived PPs, including ferulic acid (FA), isoferulic acid (IA), vanillic acid (VA), and syringic acid (SA) at physiological and supra-physiological concentrations. Assessments included lipid accumulation, glycerol release, and markers of lipid metabolism (sterol regulatory element-binding protein 1c [SREBP-1], fatty acid synthase [FASN], FAB4) and oxidative stress (DNA damage, 8-hydroxy 2-deoxyguanosine [8OHdG], nuclear erythroid factor 2-related factors 2 (NRF2), heme oxygenase 1 [HO-1]). FFAs significantly increased lipid accumulation, glycerol release, and FASN levels, while reducing HO-1 levels, without affecting other markers. WB PP metabolites did not reduce lipid accumulation, but IA and VA reduced FASN levels (-25% and -26%; p < 0.05), and SA improved HO-1 levels (+150%; p < 0.05). Despite the different effects observed, the findings obtained under our experimental conditions seem to suggest that IA, VA, and SA may modulate lipid metabolism and oxidative stress markers. However, further studies are fundamental to corroborate the findings obtained and support the contribution of these BB PPs metabolites and other compounds in the prevention and management of obesity.
Collapse
Affiliation(s)
- Samuele Venturi
- Division of Human Nutrition, Department of Food, Environmental and Nutritional Sciences (DeFENS), Università degli Studi di Milano, Milan, Italy
| | - Marco Rendine
- Division of Human Nutrition, Department of Food, Environmental and Nutritional Sciences (DeFENS), Università degli Studi di Milano, Milan, Italy
| | - Mirko Marino
- Division of Human Nutrition, Department of Food, Environmental and Nutritional Sciences (DeFENS), Università degli Studi di Milano, Milan, Italy
| | | | - Patrizia Riso
- Division of Human Nutrition, Department of Food, Environmental and Nutritional Sciences (DeFENS), Università degli Studi di Milano, Milan, Italy
| | - Cristian Del Bo'
- Division of Human Nutrition, Department of Food, Environmental and Nutritional Sciences (DeFENS), Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
9
|
Bucar F, Raimann L, Schoiswohl G, Kollau A, Mayer B, Schrammel A. Identification of 5-Geranyloxy-7-Methoxy-Coumarin as Antiadipogenic Constituent of Peels From Citrus limon. Phytother Res 2025; 39:1879-1882. [PMID: 39932107 DOI: 10.1002/ptr.8391] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 07/12/2024] [Indexed: 05/21/2025]
Affiliation(s)
- Franz Bucar
- Institute of Pharmaceutical Sciences, Pharmacognosy, University of Graz, Graz, Austria
| | - Lisa Raimann
- Institute of Pharmaceutical Sciences, Pharmacognosy, University of Graz, Graz, Austria
| | - Gabriele Schoiswohl
- Institute of Pharmaceutical Sciences, Pharmacology and Toxicology, University of Graz, Graz, Austria
- Department of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Alexander Kollau
- Institute of Pharmaceutical Sciences, Pharmacology and Toxicology, University of Graz, Graz, Austria
| | - Bernd Mayer
- Institute of Pharmaceutical Sciences, Pharmacology and Toxicology, University of Graz, Graz, Austria
| | - Astrid Schrammel
- Institute of Pharmaceutical Sciences, Pharmacology and Toxicology, University of Graz, Graz, Austria
| |
Collapse
|
10
|
Asgari H, Jalali SMA, Faghani M, Hematzadeh A. Effects of Dietary Supplementation of Coenzyme Q10 on Growth Performance, Biochemical and Physiological Attributes of Rainbow Trout (Oncorhynchus mykiss). Vet Med Sci 2025; 11:e70324. [PMID: 40192499 PMCID: PMC11974040 DOI: 10.1002/vms3.70324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/17/2024] [Accepted: 03/21/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Coenzyme Q10 (CoQ10) is a natural antioxidant and plays a vital role in the energy production of animal cells; however, its physiological and biochemical properties in fish are unclear. OBJECTIVES The current experiment was investigated to explore the effects of dietary CoQ10 on growth performance and biochemical and physiological attributes of rainbow trout (Oncorhynchus mykiss). METHODS A 56-day feeding trial was conducted with 5 experimental diets supplemented with CoQ10 concentrations at 0, 25, 50, 100 and 200 mg kg-1 of diet and fed to 400 rainbow trout (10 ± 0.1 g, initial body weight). RESULTS Dietary supplementation with CoQ10, especially at the highest dietary level, significantly improved the feed conversion ratio, final body weight and lipid and protein efficiency ratio of fish (p < 0.05). Moreover, the fish carcass protein and lipid content significantly increased with supplementation of 200 mg CoQ10 kg-1 diet (p < 0.05). The blood serum levels of triglycerides (TGs), cholesterol (CHO) and uric acid significantly reduced, whereas albumin, total protein, high-density lipoprotein and lymphocyte count increased with supplementation of 200 mg CoQ10 kg-1 diet (p < 0.05). Administration of 100 mg CoQ10 kg-1 diet significantly reduced the liver gene expression of sterol regulatory element-binding protein1 (SREBP1), whereas supplementation with 200 mg CoQ10 kg-1 diet increased muscle gene expression of SREBP1 (p < 0.05). However, there were negative associations and correlations between blood TGs and CHO levels with liver and muscle SREBP1 gene expression. CONCLUSIONS Overall, dietary supplementation of CoQ10, particularly at the levels of 100-200 mg kg-1 of diet, can improve growth performance and health in rainbow trout by modifying blood metabolites and SREBP1 gene expression.
Collapse
Affiliation(s)
- Hajar Asgari
- Department of Animal SciencesSh K.C., Islamic Azad UniversityShahr‐e KordIran
| | - Sayed Mohammad Ali Jalali
- Research Center of Nutrition and Organic Products (RCNOP), Shahrekord BranchIslamic Azad UniversityShahrekordIran
| | - Mostafa Faghani
- Department of Animal SciencesSh K.C., Islamic Azad UniversityShahr‐e KordIran
| | - Azar Hematzadeh
- Department of Animal SciencesSh K.C., Islamic Azad UniversityShahr‐e KordIran
| |
Collapse
|
11
|
Thakur MR, Tupe RS. l-Arginine: A multifaceted regulator of diabetic cardiomyopathy. Biochem Biophys Res Commun 2025; 761:151720. [PMID: 40186920 DOI: 10.1016/j.bbrc.2025.151720] [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: 02/14/2025] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 04/07/2025]
Abstract
In diabetes mellitus, dysregulated glucose and lipid metabolism lead to diabetic cardiomyopathy (DCM) by imparting pathological myocardial remodeling and cellular injury. Accelerated glycation, oxidative stress, and activated inflammatory pathways culminate in cardiac fibrosis and hypertrophy in DCM. The regulatory effects of l-Arginine (L-Arg) have been elucidated in the pathological changes of DCM, including myocardial fibrosis, hypertrophy, and apoptosis, by inhibiting glycation and oxidative stress-induced inflammation. Disturbed L-Arg metabolism and decreased intracellular L-Arg pool are correlated with the progression of DCM; therefore, L-Arg supplementation has been prescribed for various cardiovascular dysfunctions. This review expands the therapeutic potential of L-Arg supplementation in DCM by elucidating its molecular mechanism of action and exploring potential clinical outcomes.
Collapse
Affiliation(s)
- Muskan R Thakur
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Lavale, Pune, 412115, Maharashtra, India
| | - Rashmi S Tupe
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Lavale, Pune, 412115, Maharashtra, India.
| |
Collapse
|
12
|
Zhixiang Y, Yongxing X, Juan L, Qing Y, Yangyang L, Zhangrui Z, Yuehua G. The effects of soy protein and soy isoflavones intake on chronic kidney disease: a systematic review and meta-analysis. Int Urol Nephrol 2025; 57:1533-1553. [PMID: 39611889 DOI: 10.1007/s11255-024-04301-4] [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: 03/19/2024] [Accepted: 11/20/2024] [Indexed: 11/30/2024]
Abstract
PURPOSE Existing literatures on the potential impact of soy protein consumption on kidney function present conflicting findings. In this study, a meta-analysis has been conducted to assess the impact of soy protein consumption in comparison to animal protein consumption among individuals with chronic kidney disease (CKD). METHODS A structured electronic search was conducted on Medline, EMBASE, and Cochrane Library for randomized controlled trials published up to March 2024. The outcome measures were serum creatinine (SCR), triglyceride (TG), total cholesterol (TC), calcium (Ca), C-reactive protein, proteinuria, high-density lipoprotein (HDL), low-density lipoprotein (LDL), uric acid (UA) and phosphorus concentrations. Mean differences were calculated for net changes using random-effects models. RESULTS Eighteen trials with a total of 522 participants were included in this systematic review. The results showed that consumption of soy protein led to a significant decrease in total cholesterol, LDL, and proteinuria levels. The average reduction was - 20.55 mg/dL (95% CI - 38.25, - 2.85 mg/dL) for total cholesterol (P = 0.02), - 8.26 mg/dL (95% CI - 13.35, - 3.17 mg/dL; P = 0.001) for LDL and - 140.53 (95% CI - 205.83, - 75.23 mg/day) for proteinuria. No statistically significant impact was observed on serum creatinine, triglycerides, calcium, C-reactive protein, HDL, uric acid, or phosphorus levels. CONCLUSION The findings of the meta-analysis showed a potential protective impact of soy protein intake on hyperlipidemia and proteinuria in CKD patients. It is important to note that the evidence presented may be of limited accuracy due to relatively small number of trials and participants.
Collapse
Affiliation(s)
- Yu Zhixiang
- Department of Nephrology, The Ninth Medical Center of Chinese PLA General Hospital (Former Name: The Chinese PLA Strategic Support Force Medical Center), Beijing, 100101, China
| | - Xu Yongxing
- Department of Nephrology, The Ninth Medical Center of Chinese PLA General Hospital (Former Name: The Chinese PLA Strategic Support Force Medical Center), Beijing, 100101, China
- Chinese PLA Medical School, Beijing, 100853, China
- Department of Nephrology, The First Medical Center of Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing, 100853, China
| | - Liu Juan
- Beijing Chaoyang 11th Retired Cadre Rest Center, Beijing, 100101, China
| | - Yang Qing
- Department of Nephrology, The Ninth Medical Center of Chinese PLA General Hospital (Former Name: The Chinese PLA Strategic Support Force Medical Center), Beijing, 100101, China
| | - Lu Yangyang
- Department of Nephrology, The Ninth Medical Center of Chinese PLA General Hospital (Former Name: The Chinese PLA Strategic Support Force Medical Center), Beijing, 100101, China
| | - Zhen Zhangrui
- Department of Nephrology, The Ninth Medical Center of Chinese PLA General Hospital (Former Name: The Chinese PLA Strategic Support Force Medical Center), Beijing, 100101, China
| | - Gao Yuehua
- Department of Nephrology, The Ninth Medical Center of Chinese PLA General Hospital (Former Name: The Chinese PLA Strategic Support Force Medical Center), Beijing, 100101, China.
| |
Collapse
|
13
|
Gong Y, Xu R, Gao G, Li S, Liu Y. The role of fatty acid metabolism on B cells and B cell-related autoimmune diseases. Inflamm Res 2025; 74:75. [PMID: 40299047 DOI: 10.1007/s00011-025-02042-3] [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: 02/08/2025] [Revised: 04/08/2025] [Accepted: 04/15/2025] [Indexed: 04/30/2025] Open
Abstract
Fatty acid metabolism plays a critical role in regulating immune cell function, including B cells, which are central to humoral immunity and the pathogenesis of autoimmune diseases. Emerging evidence suggests that fatty acid metabolism influences B cell development, activation, differentiation, and antibody production, thereby impacting B cell-related autoimmune diseases such as systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), and multiple sclerosis (MS). In this review, we discuss the mechanisms by which fatty acid metabolism modulates B cell biology, including energy provision, membrane composition, and signaling pathways. We highlight how alterations in fatty acid synthesis, oxidation, and uptake affect B cell function and contribute to autoimmune pathogenesis. Additionally, we explore the therapeutic potential of targeting fatty acid metabolism in B cells to treat autoimmune diseases. Understanding the interplay between fatty acid metabolism and B cell immunity may provide novel insights into the development of precision therapies for B cell-mediated autoimmune disorders.
Collapse
Affiliation(s)
- Yanmei Gong
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Neuroimmunology, Jinan, Shandong, China
| | - Ruiqi Xu
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Neuroimmunology, Jinan, Shandong, China
| | - Guohui Gao
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Neuroimmunology, Jinan, Shandong, China
| | - Simiao Li
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Neuroimmunology, Jinan, Shandong, China
| | - Ying Liu
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Neuroimmunology, Jinan, Shandong, China.
- Shandong Institute of Neuroimmunology, Jinan, 250014, People's Republic of China, China.
- Shandong Provincial Medicine and Health Key Laboratory of Neuroimmunology, Jinan, Shandong, China.
| |
Collapse
|
14
|
Barsky ST, Monks DA. The role of androgens and global and tissue-specific androgen receptor expression on body composition, exercise adaptation, and performance. Biol Sex Differ 2025; 16:28. [PMID: 40269952 PMCID: PMC12016402 DOI: 10.1186/s13293-025-00707-6] [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: 11/25/2024] [Accepted: 03/23/2025] [Indexed: 04/25/2025] Open
Abstract
Gonadal testosterone stimulates skeletal muscle anabolism and contributes to sexually differentiated adipose distribution through incompletely understood mechanisms. Observations in humans and animal models have indicated a major role for androgen receptor (AR) in mediating sex differences in body composition throughout the lifespan. Traditional surgical, genetic and pharmacological studies have tested systemic actions of circulating androgens, and more recent transgenic approaches have allowed for tests of AR gene function in specific androgen responsive niches contributing to body composition, including: skeletal muscle and surrounding interstitial cells, white and brown adipose, as well as trabecular and cortical bone. Less well understood is how these functions of gonadal androgens interact with exercise. Here, we summarize the understood mechanisms of action of AR and its interactions with exercise, specifically on outcomes of body composition and muscle function, and the global- and tissue-specific role of AR in regulating skeletal muscle, adipose, and bone morphology. Additionally, we describe the known effects of androgen and AR manipulation on female body composition, muscle morphology, and sport performance, while highlighting a need for greater inclusion of female subjects in human and animal muscle physiology and endocrinology research.
Collapse
Affiliation(s)
- Sabrina Tzivia Barsky
- Department of Cell & Systems Biology, Faculty of Arts & Science, University of Toronto, Toronto, ON, Canada
| | - Douglas Ashley Monks
- Department of Cell & Systems Biology, Faculty of Arts & Science, University of Toronto, Toronto, ON, Canada.
- Department of Psychology, Faculty of Arts & Science, University of Toronto Mississauga, 3359 Mississauga Road North, Deerfield Hall DH4098, Mississauga, ON, L5L 1C6, Canada.
| |
Collapse
|
15
|
Lee AK, Welander PV. A geranylgeranyl reductase homolog required for cholesterol production in Myxococcota. J Bacteriol 2025; 207:e0049524. [PMID: 40067012 PMCID: PMC12004948 DOI: 10.1128/jb.00495-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 02/14/2025] [Indexed: 04/18/2025] Open
Abstract
Myxococcota is a phylum of sterol-producing bacteria. They exhibit a clade depth for sterol biosynthesis unparalleled in the bacterial domain and produce sterols of a biosynthetic complexity that rivals eukaryotes. Additionally, the sterol biosynthesis pathways found in this phylum have been proposed as a potential source for sterol biosynthesis in the last eukaryotic common ancestor, lending evolutionary importance to our understanding of this pathway in Myxococcota. However, sterol production has only been characterized in a few species, and outstanding questions about the evolutionary history of this pathway remain. Here, we identify two myxobacteria, Minicystis rosea and Sandaracinus amylolyticus, capable of cholesterol biosynthesis. These two myxobacteria possess a cholesterol biosynthesis pathway that differs in both the ordering and enzymes involved in biosynthesis compared with Enhygromyxa salina, a myxobacterium previously demonstrated to produce cholesterol, as well as the canonical pathways found in eukaryotes. We characterize an alternative bacterial reductase responsible for performing C-24 reduction, further delineating bacterial cholesterol production from eukaryotes. Finally, we examine the distribution and phylogenetic relationships of sterol biosynthesis proteins across both cultured and uncultured Myxococcota species, providing evidence for multiple acquisition events and instances of both horizontal and vertical transfer at the family level. Altogether, this work further demonstrates the capacity of myxobacteria to synthesize eukaryotic sterols but with an underlying diversity in the biochemical reactions that govern sterol synthesis, suggesting a complex evolutionary history and refining our understanding of how myxobacterial cholesterol production relates to their eukaryotic counterparts. IMPORTANCE Sterols are essential and ubiquitous lipids in eukaryotes, but their significance in bacteria is less understood. Sterol production in Myxococcota, a phylum of developmentally complex predatory bacteria, has provided insight into novel sterol biochemistry and prompted discussion regarding the evolution of this pathway within both the eukaryotic and bacterial domains. Here, we characterize cholesterol biosynthesis in two myxobacteria, providing evidence for distinct pathway organization and identifying a unique protein responsible for C-24 reduction. We couple these results with the phylogenomic analysis of sterol biosynthesis within Myxococcota, revealing a complicated evolutionary history marked by vertical and horizontal transfer. This suggests a mosaic acquisition of this pathway in Myxococcota and highlights the complex role myxobacteria may have had in sterol transfer to eukaryotes.
Collapse
Affiliation(s)
- Alysha K. Lee
- Department of Earth Systems Science, Stanford University, Stanford, California, USA
| | - Paula V. Welander
- Department of Earth Systems Science, Stanford University, Stanford, California, USA
| |
Collapse
|
16
|
Wu Y, Ge H, Zhao H, Zou K, Wang P, Wang Y, Zhang Y. The active ingredient β-sitosterol in the anti-inflammatory agents alleviates perianal inflammation in rats by inhibiting the expression of Srebf2, activating the PPAR signaling pathway, and altering the composition of gut microbiota. Int Immunopharmacol 2025; 152:114470. [PMID: 40086059 DOI: 10.1016/j.intimp.2025.114470] [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: 10/22/2024] [Revised: 02/24/2025] [Accepted: 03/09/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND Anti-inflammatory herbal formulations are common in traditional Chinese medicine for clearing heat and detoxifying; however, the specific active components and their mechanisms remain unclear. OBJECTIVE This study investigates the role of Sitosterol in alleviating perianal inflammation and its underlying mechanisms. METHODS Sitosterol was identified as a key active ingredient through the TCMSP database. Its structure was analyzed using PubChem, target genes were explored with STITCH, and KEGG pathways related to Srebf2 were revealed by STRING. An animal model of perianal inflammation was induced with 75 % acetic acid and treated with Sitosterol, water, normal saline, or antibiotics. The effects on gut microbiota were assessed using 16S rRNA sequencing, and inflammation was evaluated through HE stains, IHC, and TUNEL assays. In vitro, LPS-treated Caco-2 cells were used to measure proliferation, apoptosis, and cytokine levels, with PPAR pathway involvement examined using GW6471. RESULTS Sitosterol emerged as the primary active ingredient targeting Srebf2, with KEGG analysis highlighting the PPAR signaling pathway. In rats, Sitosterol reduced weight loss, inflammatory cell infiltration, edema, and vasodilation in perianal tissue. Additionally, it decreased PCNA levels, increased apoptosis, and elevated serum levels of IL-1β, IL-6, and TNF-α, particularly at high doses compared to antibiotics. Sitosterol also restored gut microbiota. Srebf2 knockdown improved tissue conditions and modulated cytokine levels, effects that were countered by GW6471. In LPS-treated Caco-2 cells, Sitosterol reversed reductions in cell viability and proliferation and modulated the expression of proteins and cytokines. CONCLUSION Sitosterol restores gut microbiota composition and further alleviates perianal inflammation in rats by inhibiting Srebf2 expression and activating the PPAR signaling pathway.
Collapse
Affiliation(s)
- Yanlan Wu
- Colon and Rectal Surgery, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Hao Ge
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Haoran Zhao
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Kaiping Zou
- Department of Pharmacy, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Pei Wang
- Jiangsu Clinical Innovation Center For Anorectal Diseases of T.C.M, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Yi Wang
- Colon and Rectal Surgery, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Yang Zhang
- Colon and Rectal Surgery, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Clinical Innovation Center For Anorectal Diseases of T.C.M, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
17
|
Wang JY, Chen MP, Jiang JX, Wan YK, Li X, Zhang YW, Fang Y, Huang HH, Qin ZY, Hou J. Lipin1-dependent transcriptional inactivation of SREBPs contributes to selinexor sensitivity in multiple myeloma. Acta Pharmacol Sin 2025:10.1038/s41401-025-01553-3. [PMID: 40229499 DOI: 10.1038/s41401-025-01553-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 03/23/2025] [Indexed: 04/16/2025]
Abstract
Selective nuclear export inhibitor selinexor (SEL) represents a promising therapeutic strategy for relapsed/refractory multiple myeloma (RRMM). But its mechanisms of action as well as factors that influence therapeutic responses have not been fully characterized yet. In this study we employed catTFRE proteomics technique to profile changes in nuclear abundance of activated transcription factors (TFs)/co-factors (TCs) in myeloma cells following SEL treatment. We found that pharmacological inhibition of exportin-1 (XPO1) by SEL leads to a significant nuclear accumulation of Lipin1 in NCI-H929 cells. Nuclear-localized Lipin1 acted as a transcriptional cofactor that suppressed the transcriptional activity of SREBPs. By performing subcellular localization analysis, molecular docking, co-immunoprecipitation and other assays, we demonstrated that Lipin1 was subjected to XPO1-dependent nuclear export. We demonstrated that SEL downregulated the expression of key lipogenesis-related genes regulated by SREBPs including FASN, SCD, DHCR24 and FDPS, leading to reduced fatty acid and cholesterol synthesis in MM cell lines and primary CD138+ cells. Using shRNA-mediated knockdown assays, we elucidated the critical role of Lipin1 in mediating the inhibitory effects of SEL on the SREBPs pathway and its contribution to SEL sensitivity both in vitro and in murine xenograft models. In conclusion, we reveal a novel mechanism by which SEL downregulates cellular lipid biosynthesis, thereby inhibiting the proliferation of myeloma cells. This study highlights the critical role of Lipin1 in the anti-myeloma effects of SEL, suggesting its potential as a biomarker for identifying patients who are most likely to benefit from SEL-based therapies.
Collapse
Affiliation(s)
- Jun-Ying Wang
- Department of Hematology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Meng-Ping Chen
- Department of Hematology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Jin-Xing Jiang
- Department of Hematology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Yi-Ke Wan
- Department of Hematology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Xin Li
- Department of Hematology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Yi-Wei Zhang
- Department of Hematology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Yi Fang
- Department of Hematology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Hong-Hui Huang
- Department of Hematology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Zhao-Yu Qin
- Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai, 200433, China.
| | - Jian Hou
- Department of Hematology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| |
Collapse
|
18
|
Zhang K, Wang L, Gao W, Guo R. Retinol-binding protein 4 in skeletal and cardiac muscle: molecular mechanisms, clinical implications, and future perspectives. Front Cell Dev Biol 2025; 13:1587165. [PMID: 40276651 PMCID: PMC12018443 DOI: 10.3389/fcell.2025.1587165] [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: 03/04/2025] [Accepted: 03/24/2025] [Indexed: 04/26/2025] Open
Abstract
Retinol-binding protein 4 (RBP4) has emerged as a critical adipokine involved in the pathophysiology of metabolic and cardiovascular diseases. Beyond its classical role in retinol transport, RBP4 influences insulin resistance, inflammation, lipid metabolism, mitochondrial function, and cellular apoptosis in both skeletal and cardiac muscles. Elevated levels of RBP4 are associated with obesity, type 2 mellitus diabetes, and cardiovascular diseases, making it a potential biomarker and therapeutic target. This comprehensive review elucidates the molecular mechanisms by which RBP4 affects skeletal and cardiac muscle physiology. We discuss its clinical implications as a biomarker for disease risk and progression, explore therapeutic strategies targeting RBP4, and highlight future research directions. Understanding the multifaceted roles of RBP4 could pave the way for novel interventions against metabolic and cardiovascular disorders.
Collapse
Affiliation(s)
- Kangzhen Zhang
- Department of Geriatrics, Nanjing Central Hospital, Nanjing, China
| | - Lijuan Wang
- Department of General Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Wei Gao
- Department of Geriatrics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Rong Guo
- Department of Cardiology, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
19
|
Anas MA, Aprianto MA, Sapan Y, Almira FN, Aldis RE, Atapattu NSBM, Kidd MT, Akit H, Montha N. Black soldier fly larvae oil downregulated gene expression related to fat metabolism of broilers fed low protein diet. Poult Sci 2025; 104:104831. [PMID: 40101509 PMCID: PMC11964662 DOI: 10.1016/j.psj.2025.104831] [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: 09/22/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 03/20/2025] Open
Abstract
Feeding low crude protein (LCP) diets supplemented with crystalline amino acids improves environmental and welfare parameters of broilers. However, increased body fat contents in broilers fed LCP diets have become a concern. Black soldier fly larvae oil (BSFLO), rich in lauric acid, has been reported to inhibit lipogenesis and reduce body fat. A 3 × 2 factorial experiment was conducted to evaluate the effect of BSFLO on performance, blood biochemistry, carcass quality, fat metabolism gene expression, and litter quality in broilers fed protein-reduced diets. A total of 288 broilers were divided into 6 treatments: three CP levels (200, 185, or 170 g/kg; high [HCP], medium [MCP], or low [LCP]) and two oil sources (BSFLO and Crude Palm Oil [CPO]), with 6 replicate pens of 8 birds each. Results showed a 15 g/kg CP reduction had no effect on body weight and feed intake (P > 0.05) but increased FCR (P = 0.001). A 30 g/kg CP significantly reduced the body weight and feed intake with inferior FCR (P < 0.05). However, negative effect of low CP diets on FCR was mitigated by BSFLO (P = 0.008). Reducing CP by 30 g/kg increased fat pads (P = 0.033), whereas BSFLO reduced fat pads (P = 0.049) at all three CP levels. Protein-reduced diets increased blood cholesterol (P = 0.002), HDL (P < 0.001), and LDL (P = 0.002). BSFLO decreased blood triglyceride (P = 0.026) and cholesterol (P < 0.001). Reducing 30 g/kg CP increased meat cooking loss (P = 0.035), while BSFLO decreased cooking loss (P < 0.001). BSFLO increased meat protein (P < 0.001) and decreased cholesterol (P = 0.003). The inclusion of BSFLO in protein-reduced diet down-regulated the gene expression of FAS, ACC, SREBP-1, and HMGR in broilers (P < 0.001). Reducing CP levels decreased litter pH (P = 0.011), nitrogen (P < 0.001), ammonia (P < 0.001) and moisture (P = 0.018). The study concludes that BSFLO reduced body fat by down-regulating the lipogenesis gene expression. In addition, BSFLO enhanced feed efficiency in broilers fed protein-reduced diet.
Collapse
Affiliation(s)
- Muhsin Al Anas
- Department of Animal Nutrition and Feed Science, Faculty of Animal Science, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia.
| | - Muhammad Anang Aprianto
- Department of Animal Nutrition and Feed Science, Faculty of Animal Science, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | - Yizrel Sapan
- Department of Animal Nutrition and Feed Science, Faculty of Animal Science, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | - Fadella Nur Almira
- Department of Animal Science, Faculty of Agriculture and Food Sciences and Environmental Management, University of Debrecen, Debrecen 4032, Hungary; Doctoral School of Animal Science, University of Debrecen, Debrecen 4032, Hungary
| | - Rinanti Eka Aldis
- Department of Animal Nutrition and Feed Science, Faculty of Animal Science, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | - N S B M Atapattu
- Department of Animal Science, Faculty of Agriculture, University of Ruhuna, Mapalana, Kamburupitiya 81100, Sri Lanka
| | - Michael T Kidd
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR 72701, United States
| | - Henny Akit
- Department of Animal Science, Faculty of Agriculture, Universiti Putra Malaysia, Serdang, 43400, Malaysia
| | - Napatsorn Montha
- Department of Animal Science and Aquatic, Faculty of Agriculture, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
20
|
Bhushan S, Sohal A, Noureddin M, Kowdley KV. Resmetirom: the first approved therapy for treating metabolic dysfunction associated steatohepatitis. Expert Opin Pharmacother 2025; 26:663-675. [PMID: 40100944 DOI: 10.1080/14656566.2025.2478917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 03/10/2025] [Indexed: 03/20/2025]
Abstract
INTRODUCTION Metabolic dysfunction associated steatohepatitis (MASH), previously referred to as nonalcoholic steatohepatitis (NASH), has emerged as one of the leading indications for liver transplantation in the United States. The disease is associated with increased cardiovascular mortality in patients with early-stage liver fibrosis and a heightened risk of hepatic complications in those with advanced fibrosis. Despite its growing prevalence and significant healthcare burden, there were no approved drugs to treat this chronic disease. In March 2024, Resmetirom, a selective thyroid hormone receptor-beta agonist, became the first drug to receive FDA approval for the treatment of patients with MASH and fibrosis stages F2/F3. This accelerated approval was granted based on significantly higher rates of MASH resolution and fibrosis. AREAS COVERED This review summarizes the current literature on the mechanism of action, preclinical data, pharmacokinetics, clinical efficacy, indications, and contraindications of resmetirom in the management of patients with MASH. EXPERT OPINION The approval of resmetirom for patients with MASH and moderate to advanced hepatic fibrosis is a major advance in the management of MASH. The recent positive results of the ESSENCE trial of semaglutide, if associated with conditional approval, may offer clinicians two options to treat MASH in patients with moderate to advanced fibrosis.
Collapse
Affiliation(s)
| | | | | | - Kris V Kowdley
- Liver Institute Northwest, Seattle, USA
- Elson S. Floyd College of medicine, Washington State University, Spokane, USA
| |
Collapse
|
21
|
Tam FF, Dumlao JM, Lee AH, Choy JC. Endogenous production of nitric oxide by iNOS in human cells restricts inflammatory activation and cholesterol/fatty acid biosynthesis. Free Radic Biol Med 2025; 231:1-10. [PMID: 39978607 DOI: 10.1016/j.freeradbiomed.2025.02.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 01/31/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025]
Abstract
Nitric oxide (NO) is a bioactive gas that is known to control many physiological processes. In human parenchymal cells, the function of iNOS-derived NO is incompletely understood. Here, we used RNA-seq to examine the role of iNOS-derived NO in the control of gene expression in a human lung epithelial cell line treated with inflammatory cytokines. iNOS-derived NO restricted the expression of genes involved in immune signaling, including the immune-related genes CXCL9 and E-selectin that were not previously known to be inhibited by iNOS. We also determined that iNOS-derived NO inhibits the expression of genes needed for cholesterol/fatty acid biosynthesis in response to cytokine stimulation, a process not previously known to be affected by NO. These findings establish the regulation of immune activation and cholesterol/fatty acid biosynthesis as main functions of iNOS in human parenchymal cells.
Collapse
Affiliation(s)
- Franklin F Tam
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Jenice M Dumlao
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Amy Hy Lee
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Jonathan C Choy
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada.
| |
Collapse
|
22
|
Phengpol N, Promsan S, Pengrattanachot N, Jaruan O, Sutthasupha P, Lungkaphin A. Maternal obesity promotes impaired renal autophagic process and kidney injury in male offspring. Int J Obes (Lond) 2025:10.1038/s41366-025-01751-3. [PMID: 40133698 DOI: 10.1038/s41366-025-01751-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 02/12/2025] [Accepted: 03/14/2025] [Indexed: 03/27/2025]
Abstract
BACKGROUND Obesity during pregnancy increases the risk of obesity, insulin resistance, diabetes, and the development and progression of chronic kidney disease (CKD) in later life in offspring. Impaired renal autophagic process is linked to kidney dysfunction in the setting of increased renal lipid accumulation. The aim of this study was to elucidate the effect of maternal obesity on kidney injury related to impaired renal autophagic process in the offspring. METHODS Maternal obesity model was conducted using female C57BL/6 mice fed with high-fat diet (HFD) for 8 weeks before mating. HFD was consecutively maintained throughout gestation and lactation. Male offspring were selected for investigation after weaning. Metabolic parameters and kidney morphology were performed. Renal insulin signaling, lipid metabolism, lipid accumulation, fibrosis and autophagy were determined. RESULTS Male offspring of HFD fed mothers developed obesity with insulin resistance, hyperglycemia, hyperlipidemia and consequently promoted kidney injury. Maternal obesity increased CD36, FAS, SREBP1c and Perilipin-2 while suppressed PPARα and CPT1A. The reduction of AMPK, SIRT1, Beclin-1, LC3B, and LAMP2 and the elevation of mTOR and SQSTM1/P62 were observed. These findings indicated the impairment of autophagy and renal lipid metabolism exaggerating renal lipid accumulation in the offspring of maternal obesity. CONCLUSIONS This study demonstrated that long-term HFD consumption in mothers promoted obesity with insulin resistance related kidney injury through the impairment of autophagic process and renal lipid metabolism in the offspring. These circumstances accelerated kidney injury and contributed to an increased susceptibility to CKD in male offspring of maternal obesity.
Collapse
Affiliation(s)
- Nichakorn Phengpol
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Sasivimon Promsan
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | | | - Onanong Jaruan
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Prempree Sutthasupha
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Anusorn Lungkaphin
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
- Functional Foods for Health and Disease, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
- Functional Food Research Center for Well-being, Multidisciplinary Research Institute Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
23
|
Xiao H, Du X, Hou H, Guo W, Tao Z, Bao S, Wen Z, Jing N, Gao WQ, Dong B, Fang YX. ATOH8 confers the vulnerability of tumor cells to ferroptosis by repressing SCD expression. Cell Death Differ 2025:10.1038/s41418-025-01482-y. [PMID: 40133667 DOI: 10.1038/s41418-025-01482-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 03/02/2025] [Accepted: 03/14/2025] [Indexed: 03/27/2025] Open
Abstract
Emerging evidence indicates that transcriptional regulation plays pivotal roles in modulating cellular vulnerability to ferroptosis. However, the intricate mechanisms governing these processes remain poorly understood. In this study, we identify ATOH8, a basic helix-loop-helix (bHLH) transcription factor, as a key player in ferroptosis regulation. ATOH8 is significantly upregulated in tumor cells following treatment with a ferroptosis inducer. Overexpression of ATOH8 increases the susceptibility of tumor cells to ferroptosis, while deletion of ATOH8 promotes ferroptosis evasion. Mechanistically, ATOH8 confers the sensitivity of tumor cells to ferroptosis by suppressing the transcription of stearoyl-CoA desaturase (SCD). Additionally, another bHLH family member, TCF3, is found to functions as a co-factor with ATOH8 by forming a TCF3-ATOH8 transcriptional repressive complex that suppresses SCD transcription. Furthermore, searching for upstream element reveals that EZH2 epigenetically suppresses ATOH8 expression by promoting DNA methylation in the ATOH8 promoter region and increasing the level of H3K27 me3. Importantly, pharmacological inhibition of EZH2 in a combined with a ferroptosis inducer markedly impedes tumor growth both in vitro and in vivo. Collectively, our study elucidates a molecular link between ferroptosis and epigenetic and transcriptional regulation, highlighting the potential of EZH2 and ATOH8 as therapeutic targets for cancer treatment.
Collapse
Affiliation(s)
- Huixiang Xiao
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Xinxing Du
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Huan Hou
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, #1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Wenyun Guo
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Zhenkeke Tao
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Shijia Bao
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Zhenzhen Wen
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Nan Jing
- School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Wei-Qiang Gao
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
- School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, China.
| | - Baijun Dong
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Yu-Xiang Fang
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| |
Collapse
|
24
|
Soultsioti M, de Jong AWM, Blomberg N, Tas A, Giera M, Snijder EJ, Bárcena M. Perturbation of de novo lipogenesis hinders MERS-CoV assembly and release, but not the biogenesis of viral replication organelles. J Virol 2025; 99:e0228224. [PMID: 39976449 PMCID: PMC11915874 DOI: 10.1128/jvi.02282-24] [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: 01/10/2025] [Accepted: 01/20/2025] [Indexed: 02/21/2025] Open
Abstract
Coronaviruses hijack host cell metabolic pathways and resources to support their replication. They induce extensive host endomembrane remodeling to generate viral replication organelles and exploit host membranes for assembly and budding of their enveloped progeny virions. Because of the overall significance of host membranes, we sought to gain insight into the role of host factors involved in lipid metabolism in cells infected with Middle East respiratory syndrome coronavirus (MERS-CoV). We employed a single-cycle infection approach in combination with pharmacological inhibitors, biochemical assays, lipidomics, and light and electron microscopy. Pharmacological inhibition of acetyl-CoA carboxylase (ACC) and fatty acid synthase (FASN), key host factors in de novo fatty acid biosynthesis, led to pronounced inhibition of MERS-CoV particle release. Inhibition of ACC led to a profound metabolic switch in Huh7 cells, altering their lipidomic profile and inducing lipolysis. However, despite the extensive changes induced by the ACC inhibitor, the biogenesis of viral replication organelles remained unaffected. Instead, ACC inhibition appeared to affect the trafficking and post-translational modifications of the MERS-CoV envelope proteins. Electron microscopy revealed an accumulation of nucleocapsids in early budding stages, indicating that MERS-CoV assembly is adversely impacted by ACC inhibition. Notably, inhibition of palmitoylation resulted in similar effects, while supplementation of exogenous palmitic acid reversed the compound's inhibitory effects, possibly reflecting a crucial need for palmitoylation of the MERS-CoV spike and envelope proteins for their role in virus particle assembly.IMPORTANCEMiddle East respiratory syndrome coronavirus (MERS-CoV) is the etiological agent of a zoonotic respiratory disease of limited transmissibility between humans. However, MERS-CoV is still considered a high-priority pathogen and is closely monitored by WHO due to its high lethality rate of around 35% of laboratory-confirmed infections. Like other positive-strand RNA viruses, MERS-CoV relies on the host cell's endomembranes to support various stages of its replication cycle. However, in spite of this general reliance of MERS-CoV replication on host cell lipid metabolism, mechanistic insights are still very limited. In our study, we show that pharmacological inhibition of acetyl-CoA carboxylase (ACC), a key enzyme in the host cell's fatty acid biosynthesis pathway, significantly disrupts MERS-CoV particle assembly without exerting a negative effect on the biogenesis of viral replication organelles. Furthermore, our study highlights the potential of ACC as a target for the development of host-directed antiviral therapeutics against coronaviruses.
Collapse
Affiliation(s)
- M. Soultsioti
- Molecular Virology Laboratory, Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, the Netherlands
| | - A. W. M. de Jong
- Section Electron Microscopy, Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - N. Blomberg
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - A. Tas
- Molecular Virology Laboratory, Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, the Netherlands
| | - M. Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - E. J. Snijder
- Molecular Virology Laboratory, Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, the Netherlands
| | - M. Bárcena
- Section Electron Microscopy, Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
25
|
Hornisch M, Piazza I. Regulation of gene expression through protein-metabolite interactions. NPJ METABOLIC HEALTH AND DISEASE 2025; 3:7. [PMID: 40052108 PMCID: PMC11879850 DOI: 10.1038/s44324-024-00047-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 12/20/2024] [Indexed: 03/09/2025]
Abstract
Organisms have to adapt to changes in their environment. Cellular adaptation requires sensing, signalling and ultimately the activation of cellular programs. Metabolites are environmental signals that are sensed by proteins, such as metabolic enzymes, protein kinases and nuclear receptors. Recent studies have discovered novel metabolite sensors that function as gene regulatory proteins such as chromatin associated factors or RNA binding proteins. Due to their function in regulating gene expression, metabolite-induced allosteric control of these proteins facilitates a crosstalk between metabolism and gene expression. Here we discuss the direct control of gene regulatory processes by metabolites and recent progresses that expand our abilities to systematically characterize metabolite-protein interaction networks. Obtaining a profound map of such networks is of great interest for aiding metabolic disease treatment and drug target identification.
Collapse
Affiliation(s)
- Maximilian Hornisch
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Str. 10, Berlin, 13092 Germany
| | - Ilaria Piazza
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Str. 10, Berlin, 13092 Germany
- SciLifeLab, Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solna, 171 65 Sweden
| |
Collapse
|
26
|
Li Y, Li X, Cournoyer P, Choudhuri S, Guo L, Chen S. Comparing the cannabidiol-induced transcriptomic profiles in human and mouse Sertoli cells. Toxicology 2025; 512:154068. [PMID: 39894194 DOI: 10.1016/j.tox.2025.154068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/15/2025] [Accepted: 01/25/2025] [Indexed: 02/04/2025]
Abstract
Cannabidiol (CBD), a major cannabinoid found in Cannabis sativa L., has been used in the treatment of seizures associated with Lennox-Gastaut syndrome, Dravet syndrome, and tuberous sclerosis complex. Recently, concerns have been raised regarding the male reproductive toxicity of CBD in animal models, such as monkeys, rats, and mice. In our previous studies, we reported that CBD inhibited cell proliferation in both primary human Sertoli cells and mouse Sertoli TM4 cells. Transcriptomic analysis revealed that in primary human Sertoli cells CBD disrupted DNA replication, cell cycle, and DNA repair, ultimately causing cellular senescence. In this study, we further investigated the molecular changes induced by CBD in mouse Sertoli TM4 cells using RNA-sequencing analyses and compared the transcriptomic profile with that of primary human Sertoli cells. Our findings demonstrated that, unlike in primary human Sertoli cells, CBD did not induce cellular senescence but caused apoptosis in mouse Sertoli TM4 cells. Through transcriptomic data analysis in mouse Sertoli TM4 cells, immune and cellular stress responses were identified. Moreover, transcriptomic comparisons revealed major differences in molecular changes induced by CBD between mouse Sertoli TM4 and primary human Sertoli cells. This suggests that primary human Sertoli cells and mouse Sertoli cells may respond differently to CBD.
Collapse
Affiliation(s)
- Yuxi Li
- Division of Biochemical Toxicology, National Center for Toxicological Research, U S. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR 72079, USA
| | - Xilin Li
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U S. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR 72079, USA
| | - Patrick Cournoyer
- Office of the Commissioner, U S. Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Supratim Choudhuri
- Division of Food Ingredients, Office of Premarket Additive Safety, Office of Food Chemical Safety, Dietary Supplements, and Innovation, Human Foods Program, U S. Food and Drug Administration, College Park, MD 20740, USA
| | - Lei Guo
- Division of Biochemical Toxicology, National Center for Toxicological Research, U S. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR 72079, USA
| | - Si Chen
- Division of Biochemical Toxicology, National Center for Toxicological Research, U S. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR 72079, USA.
| |
Collapse
|
27
|
Naz T, Zhao XY, Li S, Saeed T, Ullah S, Nazir Y, Liu Q, Mohamed H, Song Y. The interplay of transcriptional regulator SREBP1 with AMPK promotes lipid biosynthesis in Mucor circinelloides WJ11. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159592. [PMID: 39733936 DOI: 10.1016/j.bbalip.2024.159592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 12/18/2024] [Accepted: 12/24/2024] [Indexed: 12/31/2024]
Abstract
SREBP1 is a transcription factor that influences lipogenesis by regulating key genes associated with lipid biosynthesis, while AMPK, modulates lipid metabolism by regulating acetyl-CoA carboxylase. The exact role of these metabolic regulators in oleaginous microbes remains unclear. This study identified and manipulated the genes encoding SREBP1 (sre1) and α1 subunit of AMPK (ampk-α1) in Mucor circinelloides WJ11. Individual overexpression of sre1 yielded 32.5 % lipids and 21 g/L biomass, while ampk-α1 deletion combined with sre1 overexpression yielded 42.5 % lipids and 25 g/L biomass in mutant strains. This increase correlated with upregulated expression of key lipogenic genes and enzyme activity, enhancing lipid production and biomass. These surges were correlated with the increased mRNA levels of key genes (acl, acc1, acc2, cme1, fas1, g6pdh1, g6pdh2 and 6pgdh2). Enzyme activity analysis further showed that upregulation of ACL, ACC, ME, FAS, G6PDH and 6PGDH might provide more precursors and NADPH for lipid biosynthesis in sre1 overexpressing strains. Conversely, the activities of these genes and enzymes were markedly downregulated in sre1 deleted mutants consistent with lower lipid production and biomass than the control. These findings open new avenues for research by exploring the coordinated role of sre1 and ampk-α1 in lipid metabolism in M. circinelloides.
Collapse
Affiliation(s)
- Tahira Naz
- Colin Ratledge Center for Microbial Lipids, School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo 255000, China; State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Xiang Yu Zhao
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, China.
| | - Shaoqi Li
- Colin Ratledge Center for Microbial Lipids, School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo 255000, China
| | - Tariq Saeed
- Colin Ratledge Center for Microbial Lipids, School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo 255000, China; Department of Diet and Nutritional Sciences, Ibadat International University, Islamabad 45750, Pakistan.
| | - Samee Ullah
- Colin Ratledge Center for Microbial Lipids, School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo 255000, China; University Institute of Diet and Nutritional Sciences, Faculty of Allied Health Sciences, The University of Lahore, Lahore 54000, Pakistan
| | - Yusuf Nazir
- Colin Ratledge Center for Microbial Lipids, School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo 255000, China; Department of Food Sciences, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Bangi 43600, Malaysia
| | - Qing Liu
- Colin Ratledge Center for Microbial Lipids, School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo 255000, China.
| | - Hassan Mohamed
- Colin Ratledge Center for Microbial Lipids, School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo 255000, China; Department of Botany and Microbiology, Faculty of Science, Al-Azhar University, Assiut 71524, Egypt.
| | - Yuanda Song
- Colin Ratledge Center for Microbial Lipids, School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo 255000, China.
| |
Collapse
|
28
|
Dao W, Chen H, Ouyang Y, Huang L, Fan X, Miao Y. Molecular Characteristics and Role of Buffalo SREBF2 in Triglyceride and Cholesterol Biosynthesis in Mammary Epithelial Cells. Genes (Basel) 2025; 16:237. [PMID: 40004566 PMCID: PMC11855135 DOI: 10.3390/genes16020237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/15/2025] [Accepted: 02/16/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Sterol regulatory element-binding transcription factor 2 (SREBF2) is a key transcription factor involved in regulating cholesterol homeostasis. However, its role in buffalo mammary gland lipid metabolism remains unclear. Methods: To address this, we isolated and characterized the SREBF2 gene from buffalo mammary glands and performed an in-depth analysis of its molecular characteristics, tissue-specific expression, and functional roles in buffalo mammary epithelial cells (BuMECs). Additionally, we investigated the single nucleotide polymorphisms (SNPs) of SREBF2 in both river and swamp buffalo. Results: The coding sequence (CDS) of buffalo SREBF2 is 3327 bp long and encodes a protein of 1108 amino acid residues. Bioinformatics analysis revealed that the molecular characteristics of buffalo SREBF2 were highly similar across Bovidae species, with collinearity being observed among them. An expression profile analysis revealed that SREBF2 is expressed in all 11 tested tissues of buffalo, with its expression level in the mammary gland being higher during lactation than in the dry period. The knockdown of SREBF2 in BuMECs during lactation led to a significant reduction in the expression of genes involved in triglyceride (TAG) and cholesterol synthesis, including PI3K, AKT, mTOR, SREBF1, PPARG, INSIG1, ACACA, SCD, DGAT1, LPL, CD36, HMGCR, and SQLE. This knockdown led to a 23.53% and 94.56% reduction in TAG and cholesterol levels in BuMECs, respectively. In addition, a total of 22 SNPs were identified in both buffalo types, of which four non-synonymous substitutions (c.301G>C, c.304A>T, c.1240G>A, and c.2944G>A) were found exclusively in the SREBF2 CDS of swamp buffalo, and the assessment revealed that these substitutions had no impact on SREBF2 function. Conclusions: These findings emphasize the critical role of SREBF2 in regulating both triglyceride and cholesterol biosynthesis, providing valuable insights into its functions in buffalo mammary glands.
Collapse
Affiliation(s)
- Wenbin Dao
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (W.D.); (L.H.); (X.F.)
| | - Hongyan Chen
- Faculty of Animal Husbandry and Veterinary Medicine, Yunnan Vocational College of Agriculture, Kunming 650212, China;
| | - Yina Ouyang
- Yunnan Institute of Animal Science and Veterinary, Kunming 650224, China;
| | - Lige Huang
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (W.D.); (L.H.); (X.F.)
| | - Xinyang Fan
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (W.D.); (L.H.); (X.F.)
| | - Yongwang Miao
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (W.D.); (L.H.); (X.F.)
| |
Collapse
|
29
|
Li S, Yuan H, Li L, Li Q, Lin P, Li K. Oxidative Stress and Reprogramming of Lipid Metabolism in Cancers. Antioxidants (Basel) 2025; 14:201. [PMID: 40002387 PMCID: PMC11851681 DOI: 10.3390/antiox14020201] [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: 12/10/2024] [Revised: 01/30/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Oxidative stress is a common event involved in cancer pathophysiology, frequently accompanied by unique lipid metabolic reprogramming phenomena. Oxidative stress is caused mainly by an imbalance between the production of reactive oxygen species (ROS) and the antioxidant system in cancer cells. Emerging evidence has reported that oxidative stress regulates the expression and activity of lipid metabolism-related enzymes, leading to the alteration of cellular lipid metabolism; this involves a significant increase in fatty acid synthesis and a shift in the way in which lipids are taken up and utilized. The dysregulation of lipid metabolism provides abundant intermediates to synthesize biological macromolecules for the rapid proliferation of cancer cells; moreover, it contributes to the maintenance of intracellular redox homeostasis by producing a variety of reducing agents. Moreover, lipid derivatives and metabolites play critical roles in signal transduction within cancer cells and in the tumor microenvironment that evades immune destruction and facilitates tumor invasion and metastasis. These findings suggest a close relationship between oxidative stress and lipid metabolism during the malignant progression of cancers. This review focuses on the crosstalk between the redox system and lipid metabolic reprogramming, which provides an in-depth insight into the modulation of ROS on lipid metabolic reprogramming in cancers and discusses potential strategies for targeting lipid metabolism for cancer therapy.
Collapse
Affiliation(s)
| | | | | | | | - Ping Lin
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center and Lab of Experimental Oncology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; (S.L.); (H.Y.); (L.L.); (Q.L.)
| | - Kai Li
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center and Lab of Experimental Oncology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; (S.L.); (H.Y.); (L.L.); (Q.L.)
| |
Collapse
|
30
|
Parrotta ME, Colangeli L, Scipione V, Vitale C, Sbraccia P, Guglielmi V. Time Restricted Eating: A Valuable Alternative to Calorie Restriction for Addressing Obesity? Curr Obes Rep 2025; 14:17. [PMID: 39899119 PMCID: PMC11790783 DOI: 10.1007/s13679-025-00609-z] [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] [Accepted: 01/20/2025] [Indexed: 02/04/2025]
Abstract
PURPOSE OF REVIEW In this review, we summarize the molecular effects of time-restricted eating (TRE) and its possible role in appetite regulation. We also discuss the potential clinical benefits of TRE in obesity. RECENT FINDINGS TRE is an emerging dietary approach consisting in limiting food intake to a specific window of time each day. The rationale behind this strategy is to restore the circadian misalignment, commonly seen in obesity. Preclinical studies have shown that restricting food intake only during the active phase of the day can positively influence several cellular functions including senescence, mitochondrial activity, inflammation, autophagy and nutrients' sensing pathways. Furthermore, TRE may play a role by modulating appetite and satiety hormones, though further research is needed to clarify its exact mechanisms. Clinical trials involving patients with obesity or type 2 diabetes suggest that TRE can be effective for weight loss, but its broader effects on improving other clinical outcomes, such as cardiovascular risk factors, remain less certain. The epidemic proportions of obesity cause urgency to find dietary, pharmacological and surgical interventions that can be effective in the medium and long term. According to its molecular effects, TRE can be an interesting alternative to caloric restriction in the treatment of obesity, but the considerable variability across clinical trials regarding population, intervention, and follow-up duration makes it difficult to reach definitive conclusions.
Collapse
Affiliation(s)
| | - Luca Colangeli
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Internal Medicine Unit - Obesity Center, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Valeria Scipione
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Carolina Vitale
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Paolo Sbraccia
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Internal Medicine Unit - Obesity Center, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Valeria Guglielmi
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.
- Internal Medicine Unit - Obesity Center, University Hospital Policlinico Tor Vergata, Rome, Italy.
| |
Collapse
|
31
|
Zhao Z, Wu W, Zhang Q, Xing T, Bai Y, Li S, Zhang D, Che H, Guo X. Mechanism and therapeutic potential of hippo signaling pathway in type 2 diabetes and its complications. Biomed Pharmacother 2025; 183:117817. [PMID: 39842269 DOI: 10.1016/j.biopha.2025.117817] [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: 10/15/2024] [Revised: 12/22/2024] [Accepted: 01/09/2025] [Indexed: 01/24/2025] Open
Abstract
Loss of pancreatic islet cell mass and function is one of the most important factors in the development of type 2 diabetes mellitus, and hyperglycemia-induced lesions in other organs are also associated with apoptosis or hyperproliferation of the corresponding tissue cells. The Hippo signaling pathway is a key signal in the regulation of cell growth, proliferation and apoptosis, which has been shown to play an important role in the regulation of diabetes mellitus and its complications. Excessive activation of the Hippo signaling pathway under high glucose conditions triggered apoptosis and decreased insulin secretion in pancreatic islet cells, while dysregulation of the Hippo signaling pathway in the cells of other organ tissues led to proliferation or apoptosis and promoted tissue fibrosis, which aggravated the progression of diabetes mellitus and its complications. This article reviews the mechanisms of Hippo signaling, its individual and reciprocal regulation in diabetic pancreatic pathology, and its emerging role in the pathophysiology of diabetic complications. Potential therapeutics for diabetes mellitus that have been shown to target the Hippo signaling pathway are also summarized to provide information for the clinical management of type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Ziqi Zhao
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Weijie Wu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Qianyi Zhang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Tiancheng Xing
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Yiling Bai
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Shuoqi Li
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Dandan Zhang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Huilian Che
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| | - Xiaohui Guo
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| |
Collapse
|
32
|
Wang H, Luo S, Yin Y, Liu Y, Sun X, Qiu L, Wu X. DLAT is involved in ovarian cancer progression by modulating lipid metabolism through the JAK2/STAT5A/SREBP1 signaling pathway. Cancer Cell Int 2025; 25:25. [PMID: 39871246 PMCID: PMC11773875 DOI: 10.1186/s12935-025-03656-7] [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: 10/28/2024] [Accepted: 01/17/2025] [Indexed: 01/29/2025] Open
Abstract
BACKGROUND Ovarian cancer (OC) remains a lethal gynecological malignancy with an alarming mortality rate, primarily attributed to delayed diagnosis and a lack of effective treatment modalities. Accumulated evidence highlights the pivotal role of reprogrammed lipid metabolism in fueling OC progression, however, the intricate underlying molecular mechanisms are not fully elucidated. METHODS DLAT expression was assessed in OC tissues and cell lines by immunohistochemistry, western blot and qRT-PCR analysis. The effects of DLAT silencing on changes in lipid metabolism, cell viability, migration, and invasion were examined in SKOV3 and OVCAR3 cells using CCK-8, colony formation, Transwell migration and invasion, and wound-healing assays. GSEA analysis was used to examine the relationship between DLAT and lipid metabolism-related enzymes. Rescue experiments in which SREBP1 was overexpressed in DLAT-silenced cells were carried out. Western blot analysis was performed to determine whether the JAK2/STAT5 signaling pathway was involved in DLAT-regulated SREBP1 expression. Commercially available triglyceride and cholesterol detection kits, as well as Nile Red and Oil red O staining were used to measure lipid metabolism. A subcutaneous tumor model was established in BALB/c mice to confirm the role of the DLAT/SREBP1 axis in OC growth and metastasis in vivo. RESULTS DLAT expression was significantly upregulated in OC patient tissue and associated with poor prognosis. Silencing DLAT reduced lipid content and impaired OC cell proliferation, migration, and invasion. DLAT upregulated SREBP1 expression via the JAK2/STAT5 signaling pathway, enhancing expression of fatty acid synthesis enzymes and altering lipid metabolism. SREBP1 was essential for DLAT-dependent OC cell growth and metastasis both in vitro and in vivo. CONCLUSION This study uncovers a novel DLAT/JAK2/STAT5/SREBP1 axis that reprograms lipid metabolism in OC, providing insights into metabolic vulnerabilities and potential therapeutic targets for OC treatment.
Collapse
Affiliation(s)
- Hui Wang
- Department of Gynecology, Obstetrics & Gynecology Hospital of Fudan University, Shanghai, China
| | - Shen Luo
- Department of Gynecology, Obstetrics & Gynecology Hospital of Fudan University, Shanghai, China
| | - Yue Yin
- Department of Gynecology, Obstetrics & Gynecology Hospital of Fudan University, Shanghai, China
| | - Yang Liu
- Department of Gynecology, Obstetrics & Gynecology Hospital of Fudan University, Shanghai, China
| | - Xiaomei Sun
- Department of Gynecology, Obstetrics & Gynecology Hospital of Fudan University, Shanghai, China
| | - Ling Qiu
- Department of Gynecology, Obstetrics & Gynecology Hospital of Fudan University, Shanghai, China.
| | - Xin Wu
- Department of Gynecology, Obstetrics & Gynecology Hospital of Fudan University, Shanghai, China.
| |
Collapse
|
33
|
Zhang R, Wang J, Wu C, Wang L, Liu P, Li P. Lipidomics-based natural products for chronic kidney disease treatment. Heliyon 2025; 11:e41620. [PMID: 39866478 PMCID: PMC11758422 DOI: 10.1016/j.heliyon.2024.e41620] [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: 08/01/2024] [Revised: 12/17/2024] [Accepted: 12/31/2024] [Indexed: 01/28/2025] Open
Abstract
Chronic kidney disease (CKD) is by far the most prevalent disease in the world and is now a major global public health problem because of the increase in diabetes, hypertension and obesity. Traditional biomarkers of kidney function lack sensitivity and specificity for early detection and monitoring of CKD progression, necessitating more sensitive biomarkers for early diagnostic intervention. Dyslipidemia is a hallmark of CKD. Advancements in mass spectrometry (MS)-based lipidomics platforms have facilitated comprehensive analysis of lipids in biological samples and have revealed changes in the lipidome that are associated with metabolic disorders, which can be used as new biomarkers for kidney diseases. It is also critical for the discovery of new therapeutic targets and drugs. In this article, we focus on lipids in CKD, lipidomics methodologies and their applications in CKD. Additionally, we introduce novel biomarkers identified through lipidomics approaches and natural products derived from lipidomics for the treatment of CKD. We believe that our study makes a significant contribution to literature by demonstrating that natural products can improve CKD from a lipidomic perspective.
Collapse
Affiliation(s)
- Rui Zhang
- Renal Division, Department of Medicine, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| | - Jingjing Wang
- Renal Division, Department of Medicine, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| | - Chenguang Wu
- Renal Division, Department of Medicine, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| | - Lifan Wang
- Renal Division, Department of Medicine, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| | - Peng Liu
- Shunyi Hospital, Beijing Hospital of Traditional Chinese Medicine, Beijing, China
| | - Ping Li
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
34
|
Li J, Huang Y, Deng J, Yu M, Zouboulis CC, Wang G, Wang J. Tea (Camellia sinensis) Seed Saponins Act as Sebosuppression Agents via the AMPK/mTOR Pathway. J Cosmet Dermatol 2025; 24:e16793. [PMID: 39836532 PMCID: PMC11750075 DOI: 10.1111/jocd.16793] [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: 08/18/2024] [Revised: 12/12/2024] [Accepted: 01/02/2025] [Indexed: 01/23/2025]
Abstract
BACKGROUND Excessive lipogenesis of the skin triggers some dermatological concerns, such as enlarged pores, acne, and blackheads. Although topical drug treatments can offer temporary relief, their prolonged usage may lead to side effects of dryness, irritation, or allergic reactions. Consequently, the development of safer and efficacious ingredients in cosmetics for managing sebum overproduction represents a significant yet challenging endeavor. AIM Saponins were extracted from tea (Camellia sinensis) seed meal and purified by macroporous resin in order to investigate the impact of tea seed saponins (TSS) on lipid production in human immortalized sebaceous cells. Moreover, we attempted to reveal the underlying mechanism of TSS on the sebosuppression effect in SZ95 sebocytes stimulated by linoleic acid (LA). METHODS The compositions and chemical structures of TSS were determined using UV-vis absorption spectrum, Fourier transform-infrared (FTIR) spectrum, and ultra-high-performance liquid chromatography-mass spectrometry analysis. An in vitro model of cellular lipid accumulation induced by LA was established. Total lipid synthesis in intracellular SZ95 sebocytes was assessed through Nile Red staining, while triglyceride, cholesterol, and fatty acids were quantified by commercially assay kits. Western blot and quantitative real-time polymerase chain reaction were employed to analyze the protein expression levels involved in the AMP-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR) pathway as well as the downstream protein and mRNA expressions of sterol regulatory element-binding protein-1 (SREBP-1), peroxisome proliferator-activated receptor γ (PPARγ), and fatty acid synthase (FAS). The localizations of SREBP-1 within the cytoplasm or nucleus were characterized using immunofluorescence staining. RESULTS Five saponins were identified in the extracted TSS, all of which were oleanic acid-type pentacyclic triterpenes. TSS treatment significantly alleviated LA-induced lipid accumulation in SZ95 sebocytes. In addition, TSS activated the AMPK/mTOR pathway and downregulated the downstream protein and mRNA expression of transcription factors and enzymes, including SREBP-1, PPARγ, and FAS. Moreover, the TSS blocked the nuclear transfer of SREBP-1 from cytoplasm to nucleus. CONCLUSION In human sebocytes, TSS exhibited sebosuppressive effect as revealed by the inhibited production of total lipids as well as triglyceride, cholesterol, and fatty acids. Moreover, the anti-lipogenesis mechanism by TSS involved the activation of the AMPK/mTOR pathway and downregulated downstream transcription factors and enzymes of SREBP-1, PPARγ, and FAS. Additionally, TSS blocked the SREBP-1 nuclear translocation. These results may justify the potent of TSS as a new candidate for modulating lipogenesis in human SZ95 sebocytes.
Collapse
Affiliation(s)
- Jian Li
- Cosmetic Research Center, School of Chemical and Material EngineeringJiangnan UniversityWuxiChina
| | - Yuan‐cheng Huang
- Guangzhou Huashi Cosmetic Technology Co. Ltd.GuangzhouGuangdongChina
| | - Jian‐ming Deng
- Guangzhou Huashi Cosmetic Technology Co. Ltd.GuangzhouGuangdongChina
| | - Min Yu
- Guangzhou Huashi Cosmetic Technology Co. Ltd.GuangzhouGuangdongChina
| | - Christos C. Zouboulis
- Department of Dermatology, Venereology, Allergology and Immunology, Staedtisches Klinikum DessauBrandenburg Medical School Theodore Fontane and Faculty of Health SciencesDessauGermany
| | - Guang‐Li Wang
- Cosmetic Research Center, School of Chemical and Material EngineeringJiangnan UniversityWuxiChina
| | - Jing Wang
- Cosmetic Research Center, School of Chemical and Material EngineeringJiangnan UniversityWuxiChina
| |
Collapse
|
35
|
Jia X, Liu J, Jiang W, Chang L, Shen X, Jiang G, Li X, Chi C, Liu W, Zhang D. Binding site redundancy is critical for the regulation of fas by miR-30c in blunt snout bream (Megalobrama amblycephala). Comp Biochem Physiol A Mol Integr Physiol 2025; 299:111763. [PMID: 39395751 DOI: 10.1016/j.cbpa.2024.111763] [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: 09/11/2024] [Revised: 10/09/2024] [Accepted: 10/09/2024] [Indexed: 10/14/2024]
Abstract
MiR-30c and fatty acid synthase (fas) both play important roles in physiological processes such as lipid synthesis and fat metabolism. Predictive analysis revealed that fas is a target gene of miR-30c with multiple seed sites. Seed sites are useful to predict miRNA targeting relationships; however, detailed analyses of seed sites in fish genomes remain poorly studied. In this study, the regulatory relationship between miR-30c and fas, number and effect of seed regions, and mechanism by which miR-30c regulates lipid metabolism were evaluated in blunt snout bream (Megalobrama amblycephala). Four miR-30c target sites for fas were identified using various prediction tools. miR-30c mimics were transfected into 293 T cells, and dual-luciferase reporter assays were used to evaluate the roles of different fas target sites. When a single target site was mutated, relative luciferase activity was higher than that in the control group, with different activity levels depending on the mutation site. When multiple target sites were mutated, relative luciferase activity increased significantly as the number of mutation sites increased and was the highest when the four sites were mutated simultaneously. The miR-30c agomir was injected into the abdominal cavity of M. amblycephala at various concentrations for analyses of physiological and biochemical parameters in the liver and blood and the expression of genes related to lipid metabolism in the liver. Total cholesterol, free fatty acid, triglyceride, and low density lipoprotein levels were significantly lower after miR-30c agomir injection comparing to the control (P < 0.05). Additionally, the expression levels of genes related to lipid metabolism were significantly lower after miR-30c agomir injection than in the control (P < 0.05). In summary, this study identified four specific miR-30c target sites in the 3' UTR of fas mRNA; the effects of these sites are cumulative, and the redundancy ensures the accurate regulation of fas during evolution. In addition, miR-30c has a negative regulatory effect on fas and regulates lipid metabolism via various genes related to this process. Therefore, the regulation of miR-30c can effectively ameliorate the side effects of a high-fat diet on liver function in M. amblycephala.
Collapse
Affiliation(s)
- Xiaoyan Jia
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Jie Liu
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Weibo Jiang
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Le Chang
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiaoxue Shen
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Guangzhen Jiang
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiangfei Li
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Cheng Chi
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Wenbin Liu
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Dingdong Zhang
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
36
|
Wang Z, Fu H, Zhang N. Study on the mechanism of SAR1B in sodium acetate promoting milk fat synthesis. In Vitro Cell Dev Biol Anim 2025; 61:24-35. [PMID: 39316237 DOI: 10.1007/s11626-024-00974-4] [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: 03/26/2024] [Accepted: 08/31/2024] [Indexed: 09/25/2024]
Abstract
Acetate can promote milk fat synthesis in dairy cow mammary epithelial cells (BMECs). In this study, gene function analysis was used to explore the role of Ras family secretion-related GTP binding protein 1B (SAR1B) in milk fat synthesis of BMECs and its role and molecular mechanism in acetate-promoted milk fat synthesis. We found that the synthesis of lipid droplets and triglycerides was inhibited, and the expression levels of key genes and proteins in milk fat synthesis such as FASN and ACC were decreased in SAR1B knockout, which was reversed by overexpression of SAR1B. Addition of sodium acetate in BMECs can promote the expression of SAR1B, and SAR1B plays an important role in the synthesis of milk fat promoted by sodium acetate. We further investigated the underlying mechanism of SAR1B upregulation by sodium acetate, and found that sodium acetate could affect SAR1B expression through the positive regulation of SAR1B gene promoter activity by C/EBPβ and PPARγ. In conclusion, the results suggest that SAR1B can promote milk fat synthesis in BMECs, while C/EBPβ and PPARγ play important roles in sodium acetate to promote the expression of SAR1B.
Collapse
Affiliation(s)
- Zhixia Wang
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, 150030, Heilongjiang, China
| | - Haixin Fu
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, 150030, Heilongjiang, China
| | - Na Zhang
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, 150030, Heilongjiang, China.
| |
Collapse
|
37
|
Merret PE, Sparfel L, Lavau C, Lagadic-Gossmann D, Martin-Chouly C. Extracellular vesicles as a potential source of biomarkers for endocrine disruptors in MASLD: A short review on the case of DEHP. Biochimie 2025; 228:127-137. [PMID: 39307409 DOI: 10.1016/j.biochi.2024.09.009] [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: 02/14/2024] [Revised: 08/28/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024]
Abstract
Metabolic dysfunction-Associated Steatotic Liver Disease (MASLD) is a chronic disease with increasing prevalence and for which non-invasive biomarkers are needed. Environmental endocrine disruptors (EDs) are known to be involved in the onset and progression of MASLD and assays to monitor their impact on the liver are being developed. Extracellular vesicles (EVs) mediate cell communication and their content reflects the pathophysiological state of the cells from which they are released. They can thus serve as biomarkers of the pathological state of the liver and of exposure to EDs. In this review, we present the relationships between DEHP (Di(2-ethylhexyl) phthalate) and MASLD and highlight the potential of EVs as biomarkers of DEHP exposure and the resulting progression of MASLD.
Collapse
Affiliation(s)
- Pierre-Etienne Merret
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, F-35000, Rennes, France
| | - Lydie Sparfel
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, F-35000, Rennes, France
| | - Catherine Lavau
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, F-35000, Rennes, France
| | - Dominique Lagadic-Gossmann
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, F-35000, Rennes, France.
| | - Corinne Martin-Chouly
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, F-35000, Rennes, France.
| |
Collapse
|
38
|
Kumari S, Gupta S, Jamil A, Tabatabaei D, Karakashev S. Exploring Metabolic Approaches for Epithelial Ovarian Cancer Therapy. J Cell Physiol 2025; 240:e31495. [PMID: 39676338 DOI: 10.1002/jcp.31495] [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] [Received: 04/29/2024] [Revised: 10/21/2024] [Accepted: 11/18/2024] [Indexed: 12/17/2024]
Abstract
Epithelial ovarian cancer (EOC) has the highest mortality rate among malignant tumors of the female reproductive system and the lowest survival rate. This poor prognosis is due to the aggressive nature of EOC, its late-stage diagnosis, and the tumor's ability to adapt to stressors through metabolic reprogramming. EOC cells sustain their rapid proliferation by altering the uptake, utilization, and regulation of carbohydrates, lipids, and amino acids. These metabolic changes support tumor growth and contribute to metastasis, chemotherapy resistance, and immune evasion. Targeting these metabolic vulnerabilities has shown promise in preclinical studies, with some therapies advancing to clinical trials. However, challenges remain due to tumor heterogeneity, adaptive resistance mechanisms, and the influence of the tumor microenvironment. This review provides a comprehensive summary of metabolic targets for EOC treatment and offers an overview of the current landscape of clinical trials focusing on ovarian cancer metabolism. Future efforts should prioritize combination therapies that integrate metabolic inhibitors with immunotherapies or chemotherapy. Advances in precision medicine and multi-omics approaches will be crucial for identifying patient-specific metabolic dependencies and improving outcomes. By addressing these challenges, metabolism-based therapies can significantly transform the treatment of this devastating disease.
Collapse
Affiliation(s)
- Sangeeta Kumari
- Fels Cancer Institute for Personalized Medicine and Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Shraddha Gupta
- Fels Cancer Institute for Personalized Medicine and Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Aisha Jamil
- Fels Cancer Institute for Personalized Medicine and Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Deyana Tabatabaei
- Fels Cancer Institute for Personalized Medicine and Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania, USA
| | - Sergey Karakashev
- Fels Cancer Institute for Personalized Medicine and Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
- Cancer Epigenetics Institute, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| |
Collapse
|
39
|
Wang Y, Fang X, Liu J, Lv X, Lu K, Lu Y, Jiang Y. PCSK9 in T-cell function and the immune response. Biomark Res 2024; 12:163. [PMID: 39736777 DOI: 10.1186/s40364-024-00712-8] [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: 08/16/2024] [Accepted: 12/19/2024] [Indexed: 01/01/2025] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) was first reported in 2003 and confirmed to be strongly associated with familial hypercholesterolemia. Small-molecule inhibitors targeting PCSK9 provide an effective and safe method for managing hypercholesterolemia and reducing the cardiovascular risk. In recent years, increasing evidence has indicated other important roles for PCSK9 in inflammation, tumors, and even immune regulation. PCSK9 might be an attractive regulator of T-cell activation and expansion. It might mediate inflammation and regulate other types of immune cells. In this review, we summarize the current advances in the field of PCSK9 and provide a narrative of the biological processes associated with PCSK9. The relationships between PCSK9 and different T cells were investigated in depth. Finally, the signaling pathways associated with PCSK9 and the immune response are also summarized in this review.
Collapse
Affiliation(s)
- Yuying Wang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, 250021, China
| | - Xiaosheng Fang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, 250021, China
| | - Jiarui Liu
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, 250021, China
| | - Xiao Lv
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, 250021, China
| | - Kang Lu
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, 250021, China
| | - Yingxue Lu
- Department of Nephrology, Shandong Second Provincial General Hospital, Jinan , Shandong, 250021, China
| | - Yujie Jiang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, 250021, China.
| |
Collapse
|
40
|
Mejbel HA, Siegal GP, Wei S. Palmar Nodular Fasciitis Harboring a Novel SREBF1::USP6 Fusion Gene. Int J Surg Pathol 2024; 32:1494-1499. [PMID: 38304953 DOI: 10.1177/10668969241229330] [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] [Indexed: 02/03/2024]
Abstract
The diagnosis of low-grade fibroblastic/myofibroblastic tumors of acral sites can be challenging. These tumors encompass a diverse group of neoplasms with a spectrum of biologic potential ranges from benign to overtly malignant. They often demonstrate significant clinical, radiologic, and immunophenotypic overlap, in which the molecular phenotype may play an important diagnostic role to arrive at the final diagnosis. Herein, we report a case of soft tissue mass lesion presented on the palm of an adult patient for four months. Histologically, the tumor consisted of primarily low-grade spindle cells expressing smooth muscle actin. Molecular testing revealed a novel SREBF1::USP6 fusion gene, confirming the final diagnosis of nodular fasciitis and ultimately expanding its molecular profile. This case highlights the diagnostic value of single, cost-effective, targeted molecular panel to arrive at an accurate diagnosis and provide helpful therapeutic information.
Collapse
Affiliation(s)
- Haider A Mejbel
- Departments of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Gene P Siegal
- Departments of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Shi Wei
- Departments of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Pathology & Laboratory Medicine, University of Kansas School of Medicine, Kansas City, KS, USA
| |
Collapse
|
41
|
Ouyang S, Zhuo S, Yang M, Zhu T, Yu S, Li Y, Ying H, Le Y. Glycerol Kinase Drives Hepatic de novo Lipogenesis and Triglyceride Synthesis in Nonalcoholic Fatty Liver by Activating SREBP-1c Transcription, Upregulating DGAT1/2 Expression, and Promoting Glycerol Metabolism. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401311. [PMID: 39418169 PMCID: PMC11633478 DOI: 10.1002/advs.202401311] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 08/07/2024] [Indexed: 10/19/2024]
Abstract
Glycerol kinase (GK) participates in triglyceride (TG) synthesis by catalyzing glycerol metabolism. Whether GK contributes to nonalcoholic fatty liver (NAFL) is unclear. The expression of hepatic Gk is found to be increased in diet-induced and genetic mouse models of NAFL and is positively associated with hepatic SREBP-1c expression and TG levels. Cholesterol and fatty acids stimulate GK expression in hepatocytes. In HFD-induced NAFL mice, knockdown of hepatic Gk decreases expression of SREBP-1c and its target lipogenic genes as well as DGAT1/2, increases serum glycerol levels, decreases serum TG levels, and attenuates hepatic TG accumulation. Overexpression of GK in hepatocytes in mice or in culture produces opposite results. Mechanistic studies reveal that GK stimulates SREBP-1c transcription directly by binding to its gene promoter and indirectly by binding to SREBP-1c protein, thereby increasing lipogenic gene expression and de novo lipogenesis. Studies with truncated GK and mutant GKs indicate that GK induces SREBP-1c transcription independently of its enzyme activity. GK contributes to lipid homeostasis under physiological conditions by catalyzing glycerol metabolism rather than by regulating SREBP-1c transcription. Collectively, these results demonstrate that increased hepatic GK promotes de novo lipogenesis and TG synthesis in NAFL by stimulating SREBP-1c transcription and DGAT1/2 expression and catalyzing glycerol metabolism.
Collapse
Affiliation(s)
- Shuyu Ouyang
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031China
| | - Shu Zhuo
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031China
| | - Mengmei Yang
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031China
| | - Tengfei Zhu
- School of Public HealthShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Shuting Yu
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031China
| | - Yu Li
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031China
| | - Hao Ying
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031China
| | - Yingying Le
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031China
| |
Collapse
|
42
|
Peng C, Zhang X, Zhou N, Hu T, Shen Y, Chen TJ, Liu Y, Cui H, Zhu S. Apigenin inhibits lipid metabolism of hepatocellular carcinoma cells by targeting the histone demethylase KDM1A. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156024. [PMID: 39341125 DOI: 10.1016/j.phymed.2024.156024] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/26/2024] [Accepted: 09/02/2024] [Indexed: 09/30/2024]
Abstract
BACKGROUND The development of cancer is accompanied by metabolic reprogramming, and the liver serves as a central hub for lipid transportation. Apigenin, a plant-derived flavonoid, demonstrates potent anticancer properties across various cancer types and exhibits promising potential as a therapeutic agent for cancer treatment. However, there are limited studies focusing on the downstream targets of apigenin. Moreover, there are few reports on the impact of apigenin in lipid metabolism within liver cancer cells. PURPOSE The objective is to elucidate the metabolic mechanism underlying the inhibitory effect of apigenin on liver cancer progression, search for downstream targets and provide reliable data support for the clinical trials of apigenin. METHODS Anticancer effects of apigenin were detected at cellular and molecular levels in vitro, and downstream targets of apigenin, especially metabolic pathway genes, were analyzed by transcriptome. Next, the downstream target of apigenin was verified and the biological function of the downstream target was examined. Finally, the downstream target of apigenin was further verified by restoring target gene expression. RESULTS Cellular molecular experiments showed that Apigenin inhibited the proliferation, migration, invasion and lipid metabolism of hepatocellular carcinoma (HCC) cells. Transcriptome analysis showed apigenin widely regulates histone demethylase, particularly histone H3K4 lysine demethylase 1A (KDM1A). Apigenin treatment inhibited the expression of KDM1A protein and mRNA levels in liver cancer cells, molecular docking predicted the interaction between apigenin and KDM1A. Furthermore, downregulation KDM1A inhibited the proliferation and lipid metabolism of HCC cells, in the same way, overexpressing KDM1A promoted proliferation of HCC cells. Finally, restoring KDM1A expression partially attenuated the effects of apigenin on lipid metabolism in HCC cells. CONCLUSION In conclusion, our study provides compelling evidence that apigenin inhibits liver cancer progression and elucidates its mechanism of action in regulating lipid metabolism. Specifically, we find that apigenin suppresses the progression of HCC cells by downregulating genes involved in lipid metabolism. Additionally, our results indicate that KDM1A acts as a downstream target of apigenin in the inhibition of lipid metabolism in HCC. These findings offer experimental support for the potential use of apigenin as a therapeutic agent for liver cancer, highlighting its relevance in future clinical applications.
Collapse
Affiliation(s)
- Cheng Peng
- School of Life Sciences, Southwest University, Beibei, Chongqing 400716, China
| | - Ximei Zhang
- School of Life Sciences, Southwest University, Beibei, Chongqing 400716, China; Chipscreen Biosciences Pharmaceutical Ltd, Chengdu, Sichuan 610041, China
| | - Nini Zhou
- School of Life Sciences, Southwest University, Beibei, Chongqing 400716, China
| | - Ting Hu
- School of Life Sciences, Southwest University, Beibei, Chongqing 400716, China
| | - Yang Shen
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China; Jinfeng Laboratory, Chongqing 401329, China
| | - Teng Jiang Chen
- School of Life Sciences, Southwest University, Beibei, Chongqing 400716, China
| | - Yan Liu
- School of Life Sciences, Southwest University, Beibei, Chongqing 400716, China
| | - Hongjuan Cui
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China; Jinfeng Laboratory, Chongqing 401329, China
| | - Shunqin Zhu
- School of Life Sciences, Southwest University, Beibei, Chongqing 400716, China; State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China; Jinfeng Laboratory, Chongqing 401329, China.
| |
Collapse
|
43
|
Kato C, Iizuka-Ohashi M, Honda M, Konishi E, Yokota I, Boku S, Mizuta N, Morita M, Sakaguchi K, Taguchi T, Watanabe M, Naoi Y. Additional statin treatment enhances the efficacy of HER2 blockade and improves prognosis in Rac1-high/HER2-positive breast cancer. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167458. [PMID: 39128642 DOI: 10.1016/j.bbadis.2024.167458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 07/26/2024] [Accepted: 08/07/2024] [Indexed: 08/13/2024]
Abstract
The prognosis of HER2-positive breast cancer (BC) has improved with the development of anti-HER2 therapies; however, the problem remains that there are still cases where anti-HER2 therapies do not respond well. We found that the expression of SREBF2, a master transcriptional factor in the mevalonate pathway, was correlated with ERBB2 (HER2) expression and a poor prognosis in HER2-positive BC. The target gene expressions of SREBF2 were associated with higher expression of ERBB2 in HER2-positive BC cells. Statins, anti-hypercholesterolemia drugs that inhibit the mevalonate pathway, enhanced the efficacy of HER2-targeting agents with inducing apoptosis in a geranylgeranylation-dependent manner. Mechanistically, statins specifically inhibited membrane localization of Rac1, a target protein of geranylgeranylation, and suppressed the activation of HER2 downstreams AKT and ERK pathways. Consistently, retrospective analysis showed a longer recurrence-free survival in Rac1-high/HER2-positive BC patients treated with HER2-targeting agents with statins than without statins. Our findings thus suggest that Rac1 expression could be used as a biomarker to stratify HER2-positive BC patients that could benefit from dual blockade, i.e., targeting HER2 with inhibition of geranylgeranylation of Rac1 using statins, thereby opening avenues for precision medicine in a new subset of Rac1-high/HER2-positive BC.
Collapse
Affiliation(s)
- Chikage Kato
- Department of Endocrine and Breast Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Mahiro Iizuka-Ohashi
- Department of Endocrine and Breast Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Mizuki Honda
- Department of Pathology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Eiichi Konishi
- Department of Pathology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Isao Yokota
- Department of Biostatistics, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Shogen Boku
- Cancer Treatment Center, Kansai Medical University Hospital, Osaka, Japan
| | | | - Midori Morita
- Department of Endocrine and Breast Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Koichi Sakaguchi
- Department of Endocrine and Breast Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tetsuya Taguchi
- Department of Endocrine and Breast Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Motoki Watanabe
- Department of Molecular-Targeting Prevention, Kyoto Prefectural University of Medicine, Kyoto, Japan.
| | - Yasuto Naoi
- Department of Endocrine and Breast Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
44
|
Zhuang Z, Wu L, Jia W, Li Y, Lu Y, Xu M, Bai H, Bi Y, Wang Z, Chen S, Chang G, Jiang Y. Threonine modulates the STAT3-SCD1 pathway to reduce fatty acid metabolism in duck hepatocytes. Poult Sci 2024; 103:104444. [PMID: 39476611 PMCID: PMC11564961 DOI: 10.1016/j.psj.2024.104444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 10/05/2024] [Accepted: 10/25/2024] [Indexed: 11/18/2024] Open
Abstract
Dietary threonine (Thr) is known to influence fat deposition in poultry, but the precise mechanisms behind its regulatory effects on hepatic lipid metabolism remain elusive. Prior research indicated that including supplemental Thr in the feed may influence STAT3 (Signal Transducer and Activator of Transcription 3) levels in the liver of meat ducks. Numerous studies have recorded the function of STAT3 in regulating fatty acid (FA) metabolism in mammals. The primary objective of this study was to investigate whether Thr influences FA metabolism and triglycerides (TG) deposition in duck liver by regulating STAT3 expression. Primary hepatocytes were isolated from duck embryos and treated for 36 h with different doses of Thr (0, 10, 25, 50, 200 μM) in vitro or with a constructed STAT3 overexpression plasmid to examine the content of FAs and TG. RNA-seq was used to detect changes in gene expression in hepatocytes following STAT3 overexpression. The results demonstrated that both the exogenous addition of Thr and the overexpression of STAT3 significantly suppressed the capacity of hepatocytes for FAs deposition (P < 0.05). The overexpression of STAT3 also inhibited TG accumulation under conditions in response to Thr deficiency (P < 0.01). Transcriptomic analyses indicated that the overexpression of STAT3 inhibits the activity of triglyceride metabolism and unsaturated fatty acid biosynthesis (P < 0.01). Finally, a dual-luciferase reporter test demonstrated that STAT3 may systematically target and inhibit SCD1 transcription (P < 0.01). The present study indicates that supplemental Thr (50 μM) inhibits hepatic FA deposition via the STAT3-SCD1 pathway. This work enhances our comprehension of the functional roles of Thr and STAT3 in modulating lipid metabolism within duck livers. Moreover, it provides a partial theoretical foundation for the nutritional prevention and pharmacological intervention of lipid metabolism disorders in poultry.
Collapse
Affiliation(s)
- Zhong Zhuang
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Lei Wu
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Wenqian Jia
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Yongpeng Li
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Yijia Lu
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Minghong Xu
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Hao Bai
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China
| | - Yulin Bi
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Zhixiu Wang
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Shihao Chen
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Guobin Chang
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Yong Jiang
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
45
|
Xie B, Zhang A, Li C, Liu Y, Deng Y, Li R, Qin H, Wu B, He T, Lan D. Differential analysis of sorting nexin 10 and sterol regulatory element-binding protein 2 expression in inflammatory bowel disease. Immunol Res 2024; 72:1417-1423. [PMID: 39412576 DOI: 10.1007/s12026-024-09539-9] [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: 07/22/2024] [Accepted: 09/04/2024] [Indexed: 02/06/2025]
Abstract
Sorting nexin 10 (SNX10) expression induces intestinal barrier dysfunction and inflammatory responses; in contrast, its inhibition promotes intestinal mucosal healing through sterol regulatory element-binding protein 2 (SREBP2)-mediated cholesterol synthesis. However, its regulatory mechanism for the pathogenesis of inflammatory bowel disease (IBD) remains unclear. In this study, we examined SNX10 and SREBP2 expression in ulcerative colitis (UC) and Crohn's disease (CD). A total of 30 and 28 patients with UC and CD, respectively, were recruited. The expression of SNX10 and SREBP2 in the colonic mucosa was measured by immunohistochemistry (IHC). We discovered that patients with CD had significantly higher expression levels of SNX10 and SREBP2 than patients with UC and healthy controls. In addition, the expression of SREBP2 in patients with UC was significantly higher than that in healthy controls. In our study, we indicated that SNX10 and SREBP2 may serve as biomarkers for identifying patients with UC and CD, thereby providing a clinical therapeutic strategy for the treatment of IBD by inhibiting SNX10.
Collapse
Affiliation(s)
- Bicheng Xie
- Department of Gastroenterology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Yunnan Digestive Disease Clinical Medical Center, No. 157, Jingbi Road, Kunming, 650032, Yunnan, China
| | - Anxing Zhang
- Department of Gastroenterology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Yunnan Digestive Disease Clinical Medical Center, No. 157, Jingbi Road, Kunming, 650032, Yunnan, China
| | - Canmei Li
- Department of Oncology, Dali Bai Autonomous Prefecture People's Hospital, Dali, 671000, Yunnan, China
| | - Yu Liu
- Department of Gastroenterology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Yunnan Digestive Disease Clinical Medical Center, No. 157, Jingbi Road, Kunming, 650032, Yunnan, China
| | - Yao Deng
- Department of Gastroenterology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Yunnan Digestive Disease Clinical Medical Center, No. 157, Jingbi Road, Kunming, 650032, Yunnan, China
| | - Ruochang Li
- Department of Gastroenterology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Yunnan Digestive Disease Clinical Medical Center, No. 157, Jingbi Road, Kunming, 650032, Yunnan, China
| | - Haichun Qin
- Department of Gastroenterology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Yunnan Digestive Disease Clinical Medical Center, No. 157, Jingbi Road, Kunming, 650032, Yunnan, China
| | - Bian Wu
- Department of General Surgery II, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Yunnan Key Laboratory of Innovative Application of Traditional Chinese Medicine, No. 157, Jingbi Road, Kunming, 650032, Yunnan, China.
| | - Tian He
- Department of Gastroenterology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Yunnan Digestive Disease Clinical Medical Center, No. 157, Jingbi Road, Kunming, 650032, Yunnan, China.
| | - Danfeng Lan
- Department of Gastroenterology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Yunnan Digestive Disease Clinical Medical Center, No. 157, Jingbi Road, Kunming, 650032, Yunnan, China.
| |
Collapse
|
46
|
Cocci P, Angeletti M, Mosconi G, Olivotto I, Zarantoniello M, Palermo FA. Replacement of fish meal with full fat Hermetia illucens modulates hepatic FXR signaling in juvenile rainbow trout ( Oncorhynchus mykiss): Exploring a potential role of ecdysteroids. Heliyon 2024; 10:e40302. [PMID: 39584117 PMCID: PMC11585762 DOI: 10.1016/j.heliyon.2024.e40302] [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: 11/09/2023] [Revised: 11/07/2024] [Accepted: 11/07/2024] [Indexed: 11/26/2024] Open
Abstract
The present study was conducted to investigate the effects of fish meal (FM) replacement with full fat Hermetia illucens (HI) on the molecular mechanisms regulating lipid and bile salt (BA) homeostasis in rainbow trout (Oncorhynchus mykiss) juveniles. We thus explore the presence of 20-hydroxyecdysone (20E) in an insect meal-based diet and evaluate its potential involvement in regulating the molecular mechanisms/basis of FXR:RXR axis signaling. Ecdysteroids are a category of steroid hormones which bind a nuclear-receptor complex composed of ecdysone receptor (EcR) and ultraspiracle protein (USP) and regulate insect molting and metamorphosis. In all vertebrates, including fish, EcR-USP homologs are the Farnesoid X receptors (FXR) and the Retinoid X receptors (RXR), which are known to regulate crucial physiological and metabolic aspects, including BA synthesis and cholesterol homeostasis. In silico prediction indicates that 20E binds the heterodimeric complex with a binding affinity constant Kd equals to 610 ± 60 nM and affects positively the dimerization process. Results also demonstrated the coordinated increased expression of FXR and RXR, as well as their downstream target genes (i.e. short heterodimer partner 1 and 2) in rainbow trout fed diets containing HI meal. This latter finding was paralleled by a significant down-regulation of CYP7a1 and CYP8b1 gene expression together with a decrease in hepatic total cholesterol, triglyceride, and BA levels. Overall, our study suggested that FXR is a potential target for 20E content in insect meal and provided preliminary data on the potential role of ecdysteroids in regulating the metabolic status of teleost fish through modulation of FXR signaling in the enterohepatic system.
Collapse
Affiliation(s)
- Paolo Cocci
- University of Camerino, School of Biosciences and Veterinary Medicine, Camerino, 62032, Italy
| | - Mauro Angeletti
- University of Camerino, School of Biosciences and Veterinary Medicine, Camerino, 62032, Italy
| | - Gilberto Mosconi
- University of Camerino, School of Biosciences and Veterinary Medicine, Camerino, 62032, Italy
| | - Ike Olivotto
- Polytechnic University of Marche, Department of Life and Environmental Sciences, Ancona, 60131, Italy
| | - Matteo Zarantoniello
- Polytechnic University of Marche, Department of Life and Environmental Sciences, Ancona, 60131, Italy
| | | |
Collapse
|
47
|
Aksoy MO, Bilinska A, Stachowiak M, Flisikowska T, Szczerbal I. Deciphering the Role of the SREBF1 Gene in the Transcriptional Regulation of Porcine Adipogenesis Using CRISPR/Cas9 Editing. Int J Mol Sci 2024; 25:12677. [PMID: 39684387 DOI: 10.3390/ijms252312677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 11/23/2024] [Accepted: 11/24/2024] [Indexed: 12/18/2024] Open
Abstract
Sterol regulatory element-binding protein 1 (SREBP1) is an important transcription factor that controls lipid metabolism and adipogenesis. Two isoforms, SREBP1a and SREBP1c, are generated by alternative splicing of the first exon of the SREBF1 gene. The porcine SREBF1 gene has mainly been studied for its role in lipid metabolism in adipose tissues, but little is known about its involvement, and the role of its two isoforms, in adipogenesis. The aim of the present study was to introduce a deletion in the 5'-regulatory region of the SREBF1c gene, considered crucial for adipogenesis, using the Clustered Regularly Interspaced Short Palindromic Repeats/CRISPR-associated protein 9 (CRISPR/Cas9) method. This approach allows for the evaluation of how inhibiting SREBF1c transcription affects the expression of other genes essential for adipocyte differentiation, particularly PPARG, CEBPA, CEBPB, CEBPD, GATA2, and FABP4. It was observed that disrupting the SREBF1c isoform had no effect on the GATA2 gene but did result in a decrease in the expression of the CEBPA and CEBPD genes, an increase in the expression of CEBPB, and an inhibition in the expression of the PPARG and FABP4 genes. These changes in gene expression blocked adipogenesis, as could be seen by the failure of lipid droplets to accumulate. Our results provide evidence highlighting the pivotal role of the SREBP1c isoform in the regulation of porcine adipogenesis.
Collapse
Affiliation(s)
- Mehmet Onur Aksoy
- Department of Genetics and Animal Breeding, Poznan University of Life Sciences, Wolynska 33, 60-637 Poznan, Poland
| | - Adrianna Bilinska
- Department of Genetics and Animal Breeding, Poznan University of Life Sciences, Wolynska 33, 60-637 Poznan, Poland
| | - Monika Stachowiak
- Department of Genetics and Animal Breeding, Poznan University of Life Sciences, Wolynska 33, 60-637 Poznan, Poland
| | - Tatiana Flisikowska
- Chair of Livestock Biotechnology, School of Life Sciences, Technical University of Munich (TUM), D-85354 Freising, Germany
| | - Izabela Szczerbal
- Department of Genetics and Animal Breeding, Poznan University of Life Sciences, Wolynska 33, 60-637 Poznan, Poland
| |
Collapse
|
48
|
Gao YP, Hu C, Hu M, Dong WS, Li K, Ye YJ, Hu YX, Zhang X. CREB3 protein family: the promising therapeutic targets for cardiovascular and metabolic diseases. Cell Biol Toxicol 2024; 40:103. [PMID: 39581923 PMCID: PMC11586310 DOI: 10.1007/s10565-024-09939-5] [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: 05/20/2024] [Accepted: 10/17/2024] [Indexed: 11/26/2024]
Abstract
Significant advancements in cardiovascular and metabolic disease research have been made with the CREB3 protein family. Studies have revealed that members of this family are crucial in the development of these diseases, contributing to the regulation of lipid metabolism, inflammation, and vascular function. These studies provide useful information for future therapeutic strategies in cardiovascular and metabolic diseases.
Collapse
Affiliation(s)
- Yi-Peng Gao
- Department of Geriatrics, Hubei Key Laboratory of Metabolic and Chronic Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Can Hu
- Department of Ultrasound, Clinical Research Center for Medical Imaging in Hubei Province, Hubei Province Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Min Hu
- Department of Cardiology, Hubei Key Laboratory of Metabolic and Chronic Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Wen-Sheng Dong
- Department of Geriatrics, Hubei Key Laboratory of Metabolic and Chronic Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Kang Li
- Department of Geriatrics, Hubei Key Laboratory of Metabolic and Chronic Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yun-Jia Ye
- Department of Geriatrics, Hubei Key Laboratory of Metabolic and Chronic Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yu-Xin Hu
- Department of Geriatrics, Hubei Key Laboratory of Metabolic and Chronic Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xin Zhang
- Department of Geriatrics, Hubei Key Laboratory of Metabolic and Chronic Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
49
|
Brenes AJ, Griesser E, Sinclair LV, Davidson L, Prescott AR, Singh F, Hogg EKJ, Espejo-Serrano C, Jiang H, Yoshikawa H, Platani M, Swedlow JR, Findlay GM, Cantrell DA, Lamond AI. Proteomic and functional comparison between human induced and embryonic stem cells. eLife 2024; 13:RP92025. [PMID: 39540879 PMCID: PMC11563575 DOI: 10.7554/elife.92025] [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] [Indexed: 11/16/2024] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) have great potential to be used as alternatives to embryonic stem cells (hESCs) in regenerative medicine and disease modelling. In this study, we characterise the proteomes of multiple hiPSC and hESC lines derived from independent donors and find that while they express a near-identical set of proteins, they show consistent quantitative differences in the abundance of a subset of proteins. hiPSCs have increased total protein content, while maintaining a comparable cell cycle profile to hESCs, with increased abundance of cytoplasmic and mitochondrial proteins required to sustain high growth rates, including nutrient transporters and metabolic proteins. Prominent changes detected in proteins involved in mitochondrial metabolism correlated with enhanced mitochondrial potential, shown using high-resolution respirometry. hiPSCs also produced higher levels of secreted proteins, including growth factors and proteins involved in the inhibition of the immune system. The data indicate that reprogramming of fibroblasts to hiPSCs produces important differences in cytoplasmic and mitochondrial proteins compared to hESCs, with consequences affecting growth and metabolism. This study improves our understanding of the molecular differences between hiPSCs and hESCs, with implications for potential risks and benefits for their use in future disease modelling and therapeutic applications.
Collapse
Affiliation(s)
- Alejandro J Brenes
- Molecular, Cell and Developmental Biology, School of Life Sciences, University of DundeeDundeeUnited Kingdom
- Cell Signalling & Immunology, School of Life Sciences, University of DundeeDundeeUnited Kingdom
- Human Pluripotent Stem Cell Facility, School of Life Sciences, University of Dundee, Dow St, Dundee DD1 5EHDundeeUnited Kingdom
| | - Eva Griesser
- Molecular, Cell and Developmental Biology, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Linda V Sinclair
- Cell Signalling & Immunology, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Lindsay Davidson
- Human Pluripotent Stem Cell Facility, School of Life Sciences, University of Dundee, Dow St, Dundee DD1 5EHDundeeUnited Kingdom
| | - Alan R Prescott
- Dundee Imaging Facility, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Francois Singh
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Elizabeth KJ Hogg
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Carmen Espejo-Serrano
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Hao Jiang
- Molecular, Cell and Developmental Biology, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Harunori Yoshikawa
- Molecular, Cell and Developmental Biology, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Melpomeni Platani
- Molecular, Cell and Developmental Biology, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Jason R Swedlow
- Molecular, Cell and Developmental Biology, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Greg M Findlay
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Doreen A Cantrell
- Cell Signalling & Immunology, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Angus I Lamond
- Molecular, Cell and Developmental Biology, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| |
Collapse
|
50
|
Liu Y, Fan Y, Liu J, Liu X, Li X, Hu J. Application and mechanism of Chinese herb medicine in the treatment of non-alcoholic fatty liver disease. Front Pharmacol 2024; 15:1499602. [PMID: 39605910 PMCID: PMC11598537 DOI: 10.3389/fphar.2024.1499602] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a chronic liver condition closely associated with metabolic syndrome, with its incidence rate continuously rising globally. Recent studies have shown that the development of NAFLD is associated with insulin resistance, lipid metabolism disorder, oxidative stress and endoplasmic reticulum stress. Therapeutic strategies for NAFLD include lifestyle modifications, pharmacological treatments, and emerging biological therapies; however, there is currently no specific drug to treat NAFLD. However Chinese herb medicine (CHM) has shown potential in the treatment of NAFLD due to its unique therapeutic concepts and methods for centuries in China. This review aims to summarize the pathogenesis of NAFLD and some CHMs that have been shown to have therapeutic effects on NAFLD, thus enriching the scientific connotation of TCM theories and facilitating the exploration of TCM in the treatment of NAFLD.
Collapse
Affiliation(s)
- Yuqiao Liu
- College of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yue Fan
- College of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jibin Liu
- College of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiyang Liu
- College of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiuyan Li
- College of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jingqing Hu
- College of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Xin-Huangpu Joint Innovation Institute of Chinese Medicine, Guangzhou, China
| |
Collapse
|