1
|
Patel S, Naik L, Das M, Nayak DK, Dandsena PK, Mishra A, Kumar A, Dirisala VR, Mishra A, Das S, Singh R, Behura A, Dhiman R. Furamidine-induced autophagy exerts an anti-mycobacterial effect in a SIRT1-pAMPK-FOXO3a-dependent manner by elevation of intracellular Ca 2+ level expression. Microbiol Res 2025; 290:127976. [PMID: 39591744 DOI: 10.1016/j.micres.2024.127976] [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: 08/02/2024] [Revised: 11/02/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024]
Abstract
Mycobacterium tuberculosis (M. tb), the etiological agent of tuberculosis (TB), continues to be a major contributor to global mortality rates. To effectively combat this pandemic, TB control has to be enhanced in several areas, including point-of-care diagnostics, shorter and safer drug regimens, and preventative vaccination. The latest findings have highlighted autophagy as a host-defense mechanism that eradicates many invading bacteria, including M. tb. Thus, novel approaches like the stimulation of autophagy using various pharmaceutical drugs can be undertaken to deal with this noxious pathogen. The present study has been formulated to evaluate the anti-mycobacterial potential of Furamidine, a DNA minor groove binder (MGB). Initially, a non-cytotoxic concentration of Furamidine (10 µM) was used to assess its impact on the intracellular persistence of mycobacteria in differentiated THP-1 (dTHP-1) cells. Furamidine treatment compromised intracellular mycobacterial growth compared to control cells. Autophagy, a well-known host-defensive strategy, was investigated as a possible contributor to revealing the mechanism of action. Multiparametric approaches such as LC3-I to II conversion, protein level expression of different autophagic markers, and MDC staining were employed to study autophagic response that conclusively suggested the autophagy induction potential of Furamidine in dTHP-1 cells. Further, elevated LC3-II expression and increased autophagic vacuole accumulation under Baf-A1 treatment demonstrated the positive regulation of autophagic flux upon Furamidine treatment. Mechanistic investigations showed increased intracellular calcium (Ca2+) level expression, SIRT1, pAMPK, and FOXO3a activation upon its treatment. Inhibition of Ca2+ level expression suppressed Ca2+-mediated-FOXO3a level in Furamidine-treated cells. Furthermore, administering various inhibitors hampered the Furamidine-induced autophagy that impacted intracellular mycobacteria clearance. These results conclude that Furamidine triggered the Ca2+/pAMPK/SIRT1/FOXO3a pathway, causing less mycobacterial load in dTHP-1 cells.
Collapse
Affiliation(s)
- Salina Patel
- Laboratory of Mycobacterial Immunology, Department of Life Science, National Institute of Technology, Rourkela, Odisha 769008, India
| | - Lincoln Naik
- Laboratory of Mycobacterial Immunology, Department of Life Science, National Institute of Technology, Rourkela, Odisha 769008, India
| | - Mousumi Das
- Laboratory of Mycobacterial Immunology, Department of Life Science, National Institute of Technology, Rourkela, Odisha 769008, India
| | - Dev Kiran Nayak
- Laboratory of Mycobacterial Immunology, Department of Life Science, National Institute of Technology, Rourkela, Odisha 769008, India
| | - Pramathesh Kumar Dandsena
- Laboratory of Mycobacterial Immunology, Department of Life Science, National Institute of Technology, Rourkela, Odisha 769008, India
| | - Abtar Mishra
- Laboratory of Mycobacterial Immunology, Department of Life Science, National Institute of Technology, Rourkela, Odisha 769008, India
| | - Ashish Kumar
- Laboratory of Mycobacterial Immunology, Department of Life Science, National Institute of Technology, Rourkela, Odisha 769008, India
| | - Vijaya R Dirisala
- Department of Biotechnology, Vignan's Foundation for Science, Technology and Research, Vadlamudi, Guntur District, AP-522213, India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Rajasthan 342011, India
| | - Surajit Das
- Laboratory of Environmental Microbiology and Ecology, Department of Life Science, National Institute of Technology, Rourkela, Odisha 769008, India
| | - Ramandeep Singh
- Tuberculosis Research Laboratory, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurugram Expressway, PO Box # 4, Faridabad, Haryana 121001, India
| | - Assirbad Behura
- Laboratory of Mycobacterial Immunology, Department of Life Science, National Institute of Technology, Rourkela, Odisha 769008, India.
| | - Rohan Dhiman
- Laboratory of Mycobacterial Immunology, Department of Life Science, National Institute of Technology, Rourkela, Odisha 769008, India.
| |
Collapse
|
2
|
Park SG, Kim JW, Song JH, Kwon SH, Oh SH, Piao X, Wang Z, Ryu JH, Kim N, Kim OS, Koh JT. The microbial metabolite imidazole propionate dysregulates bone homeostasis by inhibiting AMP-activated protein kinase (AMPK) signaling. Commun Biol 2024; 7:1644. [PMID: 39695168 DOI: 10.1038/s42003-024-07316-w] [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: 09/30/2023] [Accepted: 11/25/2024] [Indexed: 12/20/2024] Open
Abstract
Microbial metabolites provide numerous benefits to the human body but can also contribute to diseases such as obesity, diabetes, cancer, and bone disorders. However, the role of imidazole propionate (ImP), a histidine-derived metabolite produced by the intestinal microbiome, in bone metabolism and the development of osteoporosis is still poorly understood. In this study, we investigated the role of ImP and its underlying mechanisms in regulating bone homeostasis. When ImP was administered to 8-week-old mice for 4 weeks, bone loss was observed, along with a decrease in alkaline phosphatase-positive osteoblast cells. Additionally, bone marrow stromal cells (BMSCs) isolated from ImP-treated mice exhibited reduced osteogenic potential. In BMSCs from control mice, ImP treatment inhibited BMP2-induced osteoblast differentiation while promoting adipocyte differentiation. However, ImP had no effect on RANKL-induced osteoclast differentiation or activity in bone marrow macrophages. Mechanistically, ImP treatment increased p38γ phosphorylation and decreased AMPK (T172) phosphorylation in BMSCs. Suppression of p38γ expression using si-p38γ reversed the inhibitory effects of ImP on osteoblast differentiation, with a concurrent increase in AMPK (T172) phosphorylation. Conversely, ImP stimulated adipocyte differentiation by decreasing AMPK (T172) phosphorylation. Treatment with the AMPK agonist metformin significantly reversed the inhibitory effects of ImP on osteoblast differentiation and the promotion of adipocyte differentiation, along with enhanced AMPK (T172) phosphorylation. These findings suggest that the microbial metabolite ImP may disrupt bone homeostasis by stimulating p38γ phosphorylation and inhibiting the AMPK pathway, presenting a potential therapeutic target for managing metabolic bone diseases.
Collapse
Affiliation(s)
- Suk-Gyun Park
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Jung-Woo Kim
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Ju Han Song
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Seung-Hee Kwon
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Sin-Hye Oh
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Xianyu Piao
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Zhao Wang
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Je-Hwang Ryu
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Nacksung Kim
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Ok-Su Kim
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
- Department of Periodontology, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Jeong-Tae Koh
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea.
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea.
| |
Collapse
|
3
|
Mariño Dal Magro B, Dos Santos BG, August PM, Menegotto MZ, Driemeier D, Matté C. Maternal swimming with overload allied to postnatal high-fat, high-sugar diet induce subtle impairment on rat offspring's ovarian redox homeostasis. Reprod Fertil Dev 2024; 37:RD24132. [PMID: 39699999 DOI: 10.1071/rd24132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 11/27/2024] [Indexed: 12/21/2024] Open
Abstract
Context The Developmental Origins of Health and Disease (DOHaD) concept suggests that early-life interventions significantly influence the long-term health outcomes of offspring. Emerging evidence supports that maternal physical exercise and balanced nutrition can positively impact the health of the next generation. Aims This study investigated the effects of maternal swimming combined with postnatal high-fat, high-sugar (HFHS) diet on the ovarian health of adult female Wistar rat offspring. Methods Adult female Wistar rats performed swimming exercise in a controlled temperature environment (32°C, 2% bodyweight overload adjusted daily) during 4weeks, starting 1week prior mating. The female offspring received a control or HFHS diet from postnatal day (PND) 21 to PND 90. We analyzed offspring's body weight, ovarian histomorphology, redox status, and associated molecular pathways 5' adenosine monophosphate-activated protein kinase (AMPKα), forkhead box O3 (FoxO3), and mitofusin 1 (Mfn-1). Key results Our findings reveal that maternal swimming exerted an effect on offspring body weight gain, delaying it. Individually, maternal exercise reduced superoxide dismutase (SOD) activity and mitofusin-1 levels, while the postnatal HFHS diet alone decreased both SOD and glutathione peroxidase (GPx) activities and increased the pFOXO3a/FOXO3a ratio in the ovaries. Conclusions We showed that combination of maternal swimming with a 2% overload and a postnatal HFHS diet can negatively affect the ovarian redox balance in offspring. Implications Prenatal and postnatal lifestyle might affect reproductive function in females.
Collapse
Affiliation(s)
- Bárbara Mariño Dal Magro
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Bernardo Gindri Dos Santos
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Pauline Maciel August
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Manuela Zeferino Menegotto
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - David Driemeier
- Setor de Anatomia Patológica Veterinária, Departamento de Patologia Clínica Veterinária, Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Cristiane Matté
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; and Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
4
|
Alarfaj SJ, Bahaa MM, Elmasry TA, Elberri EI, El-Khateeb E, Hamouda AO, Salahuddin MM, Kamal M, Gadallah ANAA, Eltantawy N, Yasser M, Negm WA, Hamouda MA, Alsegiani AS, Alrubia S, Eldesoqui M, Abdallah MS. Fenofibrate as an Adjunct Therapy for Ulcerative Colitis: Targeting Inflammation via SIRT1, NLRP3, and AMPK Pathways: A Randomized Controlled Pilot Study. Drug Des Devel Ther 2024; 18:5239-5253. [PMID: 39575188 PMCID: PMC11578921 DOI: 10.2147/dddt.s490772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 11/08/2024] [Indexed: 11/24/2024] Open
Abstract
Background Ulcerative colitis (UC) is an idiopathic chronic inflammation of colonic and rectal mucosa. The peroxisome proliferator-activated receptor α (PPARα) has been identified as having protective effects in UC. Aim The study aimed to investigate the efficacy of fenofibrate, a PPARα agonist, in UC. Methods A total of 70 patients with mild to moderate UC were allocated randomly and assigned to two groups (n = 35 each) from Gastroenterology Department, Faculty of Medicine, Menoufia University. The mesalamine group received a placebo along with 1 g of mesalamine three times daily, while the fenofibrate group received 1 g of mesalamine three times and fenofibrate 160 mg once daily. The study duration was for six months. A gastroenterologist assessed patients by non-invasive Partial Mayo Score (PMS) and the Inflammatory Bowel Disease Questionnaire (IBDQ) to evaluate clinical response and remission. The serum levels of silent information regulator 1 (SIRT1), NOD-like receptor protein 3 (NLRP3), and adenosine monophosphate activated protein kinase (AMPK), as well as fecal calprotectin levels were examined to determine the biological effect of fenofibrate. Results After treatment, the fenofibrate group showed statistically significant reductions in PMS (p = 0.044) and improved digestive domain of IBDQ (p = 0.023). Additionally, there were significant decreases in serum NLRP3 (p = 0.041) and fecal calprotectin (p = 0.035), along with significant increases in SIRT1 (p = 0.002) and AMPK (p = 0.0003). The fenofibrate group also had higher response and remission rates compared to the mesalamine group. Conclusion Fenofibrate may be a promising adjunct for improving clinical outcomes, quality of life, and modulating inflammation in mild to moderate patients with UC. Trial Registration Identifier NCT05781698.
Collapse
Affiliation(s)
- Sumaiah J Alarfaj
- Department of Pharmacy Practice, College of Pharmacy, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Mostafa M Bahaa
- Pharmacy Practice Department, Faculty of Pharmacy, Horus University, New Damietta, Egypt
| | - Thanaa A Elmasry
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Eman I Elberri
- Clinical Pharmacy Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Eman El-Khateeb
- Clinical Pharmacy Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Amir O Hamouda
- Department of Biochemistry and Pharmacology, Faculty of Pharmacy, Horus University, New Damietta, Egypt
| | - Muhammed M Salahuddin
- Department of Biochemistry and Pharmacology, Faculty of Pharmacy, Horus University, New Damietta, Egypt
| | - Marwa Kamal
- Department of Clinical Pharmacy, Faculty of Pharmacy, Fayoum University, Fayoum, Egypt
| | | | - Nashwa Eltantawy
- Department of Pharmacy Practice, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University, Cairo, Egypt
| | - Mohamed Yasser
- Department of Pharmaceutics, Faculty of Pharmacy, Port Said University, Port Said, Egypt
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Horus University, New Damietta, Egypt
- Department of Pharmaceutics, Faculty of Pharmacy, East Port Said National University, Port Said, Egypt
| | - Walaa A Negm
- Pharmacognosy Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Manal A Hamouda
- Department of Clinical Pharmacy, Faculty of Pharmacy, Menofia University, Menofia, Egypt
| | - Amsha S Alsegiani
- Pharmaceutical Chemistry Department, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Sarah Alrubia
- Pharmaceutical Chemistry Department, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mamdouh Eldesoqui
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh, Saudi Arabia
| | - Mahmoud S Abdallah
- Department of Clinical Pharmacy, Faculty of Pharmacy, University of Sadat City (USC), Sadat City, Menoufia, Egypt
- Department of PharmD, Faculty of Pharmacy, Jadara University, Irbid, Jordan
| |
Collapse
|
5
|
Huang S, Liang H, Chen Y, Liu C, Luo P, Wang H, Du Q. Hypoxanthine ameliorates diet-induced insulin resistance by improving hepatic lipid metabolism and gluconeogenesis via AMPK/mTOR/PPARα pathway. Life Sci 2024; 357:123096. [PMID: 39369847 DOI: 10.1016/j.lfs.2024.123096] [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: 06/11/2024] [Revised: 09/09/2024] [Accepted: 09/28/2024] [Indexed: 10/08/2024]
Abstract
AIM Insulin resistance (IR) is a pivotal metabolic disorder associated with type 2 diabetes and metabolic syndrome. This study investigated the potential of hypoxanthine (Hx), a purine metabolite and uric acid precursor, in ameliorating IR and regulating hepatic glucose and lipid metabolism. METHODS We utilized both in vitro IR-HepG2 cells and in vivo diet-induced IR mice to investigate the impact of Hx. The HepG2 cells were treated with Hx to evaluate its effects on glucose production and lipid deposition. Activity-based protein profiling (ABPP) was applied to identify Hx-target proteins and the underlying pathways. In vivo studies involved administration of Hx to IR mice, followed by assessments of IR-associated indices, with explores on the potential regulating mechanisms on hepatic glucose and lipid metabolism. KEY FINDINGS Hx intervention significantly reduced glucose production and lipid deposition in a dose-dependent manner without affecting cell viability in IR-HepG2 cells. ABPP identified key Hx-target proteins engaged in fatty acid and pyruvate metabolism. In vivo, Hx treatment reduced IR severities, as evidenced by decreased HOMA-IR, fasting blood glucose, and serum lipid profiles. Histological assessments confirmed reduced liver lipid deposition. Mechanistic insights revealed that Hx suppresses hepatic gluconeogenesis and fatty acid synthesis, and promotes fatty acid oxidation via the AMPK/mTOR/PPARα pathway. SIGNIFICANCE This study delineates a novel role of Hx in regulating hepatic metabolism, offering a potential therapeutic strategy for IR and associated metabolic disorders. The findings provide a foundation for further investigation into the role of purine metabolites in metabolic regulation and their clinical implications.
Collapse
Affiliation(s)
- Sizhe Huang
- Centre of General Practice, the Seventh Affiliated Hospital, Southern Medical University, Foshan 528000, Guangdong, PR China
| | - Hengmiao Liang
- Centre of General Practice, the Seventh Affiliated Hospital, Southern Medical University, Foshan 528000, Guangdong, PR China
| | - Yuting Chen
- Department of Laboratory Medicine, the Seventh Affiliated Hospital, Southern Medical University, Foshan 528000, Guangdong, PR China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Ce Liu
- Department of Laboratory Medicine, the Seventh Affiliated Hospital, Southern Medical University, Foshan 528000, Guangdong, PR China
| | - Piao Luo
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China.
| | - Huijun Wang
- Centre of General Practice, the Seventh Affiliated Hospital, Southern Medical University, Foshan 528000, Guangdong, PR China; Department of Laboratory Medicine, the Seventh Affiliated Hospital, Southern Medical University, Foshan 528000, Guangdong, PR China.
| | - Qingfeng Du
- Centre of General Practice, the Seventh Affiliated Hospital, Southern Medical University, Foshan 528000, Guangdong, PR China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China.
| |
Collapse
|
6
|
Yanagi T, Kikuchi H, Takeuchi K, Susa K, Mori T, Chiga M, Yamamoto K, Furukawa A, Kanazawa T, Kato Y, Takahashi N, Suzuki T, Mori Y, Carter BC, Mori M, Nakano Y, Fujiki T, Hara Y, Suzuki S, Ando F, Mandai S, Honda S, Torii S, Shimizu S, Tanaka H, Fujii Y, Rai T, Uchida S, Sohara E. ULK1-regulated AMP sensing by AMPK and its application for the treatment of chronic kidney disease. Kidney Int 2024; 106:887-906. [PMID: 39428173 DOI: 10.1016/j.kint.2024.08.024] [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: 08/17/2023] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 10/22/2024]
Abstract
Adenosine monophosphate (AMP)-activated protein kinase (AMPK) is a central kinase involved in energy homeostasis. Increased intracellular AMP levels result in AMPK activation through the binding of AMP to the γ-subunit of AMPK. Recently, we reported that AMP-induced AMPK activation is impaired in the kidneys in chronic kidney disease (CKD) despite an increase in the AMP/ATP ratio. However, the mechanisms by which AMP sensing is disrupted in CKD are unclear. Here, we identified mechanisms of energy homeostasis in which Unc-51-like kinase 1 (ULK1)-dependent phosphorylation of AMPKγ1 at Ser260/Thr262 promoting AMP sensitivity of AMPK. AMPK activation by AMP was impaired in Ulk1 knockout mice despite an increased AMP/ATP ratio. ULK1 expression is markedly downregulated in CKD kidneys, leading to AMP sensing failure. Additionally, MK8722, an allosteric AMPK activator, stimulated AMPK in the kidneys of a CKD mouse model (5/6th nephrectomy) via a pathway that is independent of AMP sensing. Thus, our study shows that MK8722 treatment significantly attenuates the deterioration of kidney function in CKD and may be a potential therapeutic option in CKD therapeutics.
Collapse
Affiliation(s)
- Tomoki Yanagi
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Hiroaki Kikuchi
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.
| | - Koh Takeuchi
- Graduate School of Pharmaceutical Sciences, the University of Tokyo, Tokyo, Japan
| | - Koichiro Susa
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Takayasu Mori
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Motoko Chiga
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Kouhei Yamamoto
- Department of Human Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Asuka Furukawa
- Department of Human Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Takumi Kanazawa
- Department of Human Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yuki Kato
- Department of Human Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Naohiro Takahashi
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Takefumi Suzuki
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yutaro Mori
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Benjamin C Carter
- Department of Biochemistry, Purdue University, West Lafayette, Indiana, USA
| | - Makiko Mori
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yuta Nakano
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Tamami Fujiki
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yu Hara
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Soichiro Suzuki
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Fumiaki Ando
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Shintaro Mandai
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Shinya Honda
- Department of Pathological Cell Biology, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Satoru Torii
- Department of Pathological Cell Biology, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Shigeomi Shimizu
- Department of Pathological Cell Biology, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Hajime Tanaka
- Department of Urology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yasuhisa Fujii
- Department of Urology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Tatemitsu Rai
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan; Department of Nephrology and Hypertension, Dokkyo Medical University, Tochigi, Japan
| | - Shinichi Uchida
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Eisei Sohara
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.
| |
Collapse
|
7
|
Ma N, Xu C, Wang Y, Cui K, Kuang H. Telomerase reverse transcriptase protects against diabetic kidney disease by promoting AMPK/PGC-1a-regulated mitochondrial energy homeostasis. Chem Biol Interact 2024; 403:111238. [PMID: 39265716 DOI: 10.1016/j.cbi.2024.111238] [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: 06/16/2024] [Revised: 09/04/2024] [Accepted: 09/09/2024] [Indexed: 09/14/2024]
Abstract
Disordered glucose and lipid metabolism, coupled with disturbed mitochondrial bioenergetics, are pivotal in the initiation and development of diabetic kidney disease (DKD). While the essential role of telomerase reverse transcriptase (TERT) in regulating mitochondrial function in the cardiovascular system has been recognized, its specific function in maintaining mitochondrial homeostasis in DKD remains unclear. This study aimed to explore how TERT regulates mitochondrial function and the underlying mechanisms. In vitro, human renal proximal tubular HK-2 cells exposed to high glucose/high fat (HG/HF) presented significant downregulation of TERT and AMPK dephosphorylation. This led to decreased ATP production, altered NAD+/NADH ratios, reduced mitochondrial complex activities, increased mitochondrial dysfunction, lipid accumulation, and reactive oxygen species (ROS) production. Knockdown of TERT (si-TERT) further exacerbated mitochondrial dysfunction, decreased mitochondrial membrane potential, and lowered levels of cellular oxidative phosphorylation and glycolysis, as determined via a Seahorse X24 flux analyzer. Conversely, mitochondrial dysfunction was significantly alleviated after pcDNA-TERT plasmid transfection and adeno-associated virus (AAV) 9-TERT gene therapy in vivo. Notably, treatment with an AMPK inhibitor, activator, and si-PGC-1a (peroxisome proliferator-activated receptor γ coactivator-1α), resulted in mitochondrial dysfunction and decreased expression of genes related to energy metabolism and mitochondrial biogenesis. Our findings reveal that TERT protects mitochondrial function and homeostasis by partially activating the AMPK/PGC-1a signaling pathway. These results establish a crucial foundation for understanding TERT's critical role inmitochondrial regulation and its protective effect on DKD.
Collapse
Affiliation(s)
- Nan Ma
- Department of Endocrinology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chengye Xu
- Department of Endocrinology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yan Wang
- Department of Endocrinology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Kexin Cui
- Department of Endocrinology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hongyu Kuang
- Department of Endocrinology, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
8
|
Wang YX, Pi JC, Yao YF, Peng XP, Li WJ, Xie MY. Hypoglycemic effects of white hyacinth bean polysaccharide on type 2 diabetes mellitus rats involvement with entero-insular axis and GLP-1 via metabolomics study. Int J Biol Macromol 2024; 281:136489. [PMID: 39393741 DOI: 10.1016/j.ijbiomac.2024.136489] [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: 05/06/2024] [Revised: 09/02/2024] [Accepted: 10/08/2024] [Indexed: 10/13/2024]
Abstract
The present study aimed to investigate the potential effects of white hyacinth bean polysaccharide (WHBP) against type 2 diabetic mellitus (T2DM) which was established by high-glucose/high-fat for 8 weeks, combined with a low-dose streptozotocin (STZ) injection. Our results showed that WHBP behaved the hypoglycemic effect by attenuating fasting blood glucose in vivo. WHBP-mediated anti-diabetic effects associated with the attenuation of insulin resistance and pancreatic impairment, as evidenced by the mitigation of pathological changes, inflammatory response and oxidative stress in the pancreas of T2DM rats. Meanwhile, gut protection was also shown during WHBP-mediated anti-diabetic effects, and glucagon-like peptide-1 (GLP-1), a mediator of the entero-insular axis, was observed to be elevated in both gut and pancreas of WHBP groups when compared to DM group, suggesting that hypoglycemic effects of WHBP were implicated in gut-pancreas interaction. Subsequently, untargeted metabolomics analysis performed by UPLC-QTOF/MS and showed that WHBP administration significantly adjusted the levels of 40 metabolites when compared to DM group. Further data concerning pathway analysis showed that WHBP administration significantly regulated the phenylalanine metabolism, tryptophan metabolism, arginine and proline, isoleucine metabolism, and glycerophospholipid metabolism in T2DM rats. Together, our results suggested that WHBP performed hypoglycemic effects and pancreatic protection linked to entero-insular axis involvement with GLP-1 and reversed metabolic disturbances in T2DM rats.
Collapse
Affiliation(s)
- Yi-Xuan Wang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Jin-Chan Pi
- College of Future Technology, Nanchang University, Nanchang 330031, China
| | - Yu-Fei Yao
- Department of Critical Care Medicine, The Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang 330006, China
| | - Xiao-Ping Peng
- Department of Cardiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Wen-Juan Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China.
| | - Ming-Yong Xie
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| |
Collapse
|
9
|
Torres-Torres J, Monroy-Muñoz IE, Perez-Duran J, Solis-Paredes JM, Camacho-Martinez ZA, Baca D, Espino-Y-Sosa S, Martinez-Portilla R, Rojas-Zepeda L, Borboa-Olivares H, Reyes-Muñoz E. Cellular and Molecular Pathophysiology of Gestational Diabetes. Int J Mol Sci 2024; 25:11641. [PMID: 39519193 PMCID: PMC11546748 DOI: 10.3390/ijms252111641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/25/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Gestational diabetes (GD) is a metabolic disorder characterized by glucose intolerance during pregnancy, significantly impacting maternal and fetal health. Its global prevalence is approximately 14%, with risk factors including obesity, family history of diabetes, advanced maternal age, and ethnicity, which are linked to cellular and molecular disruptions in glucose regulation and insulin resistance. GD is associated with short- and long-term complications for both the mother and the newborn. For mothers, GD increases the risk of developing type 2 diabetes, cardiovascular diseases, and metabolic syndrome. In the offspring, exposure to GD in utero predisposes them to obesity, glucose intolerance, and metabolic disorders later in life. This review aims to elucidate the complex cellular and molecular mechanisms underlying GD to inform the development of effective therapeutic strategies. A systematic review was conducted using medical subject headings (MeSH) terms related to GD's cellular and molecular pathophysiology. Inclusion criteria encompassed original studies, systematic reviews, and meta-analyses focusing on GD's impact on maternal and fetal health, adhering to PRISMA guidelines. Data extraction captured study characteristics, maternal and fetal outcomes, key findings, and conclusions. GD disrupts insulin signaling pathways, leading to impaired glucose uptake and insulin resistance. Mitochondrial dysfunction reduces ATP production and increases reactive oxygen species, exacerbating oxidative stress. Hormonal influences, chronic inflammation, and dysregulation of the mammalian target of rapamycin (mTOR) pathway further impair insulin signaling. Gut microbiota alterations, gene expression, and epigenetic modifications play significant roles in GD. Ferroptosis and placental dysfunction primarily contribute to intrauterine growth restriction. Conversely, fetal macrosomia arises from maternal hyperglycemia and subsequent fetal hyperinsulinemia, resulting in excessive fetal growth. The chronic inflammatory state and oxidative stress associated with GD exacerbate these complications, creating a hostile intrauterine environment. GD's complex pathophysiology involves multiple disruptions in insulin signaling, mitochondrial function, inflammation, and oxidative stress. Effective management requires early detection, preventive strategies, and international collaboration to standardize care and improve outcomes for mothers and babies.
Collapse
Affiliation(s)
- Johnatan Torres-Torres
- Department of Reproductive and Perinatal Health Research, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
- Obstetric and Gynecology Department, Hospital General de México Dr. Eduardo Liceaga, Mexico City 06720, Mexico
| | - Irma Eloisa Monroy-Muñoz
- Department of Reproductive and Perinatal Health Research, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
| | - Javier Perez-Duran
- Department of Reproductive and Perinatal Health Research, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
| | - Juan Mario Solis-Paredes
- Department of Reproductive and Perinatal Health Research, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
| | | | - Deyanira Baca
- Obstetric and Gynecology Department, Hospital General de México Dr. Eduardo Liceaga, Mexico City 06720, Mexico
| | - Salvador Espino-Y-Sosa
- Department of Reproductive and Perinatal Health Research, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
- Centro de Investigacion en Ciencias de la Salud, Universidad Anahuac Mexico, Campus Norte, Huixquilucan 52786, Mexico
| | - Raigam Martinez-Portilla
- Department of Reproductive and Perinatal Health Research, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
| | - Lourdes Rojas-Zepeda
- Maternal-Fetal Department, Instituto Materno Infantil del Estado de Mexico, Toluca 50170, Mexico
| | - Hector Borboa-Olivares
- Community Interventions Research Branch, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
| | - Enrique Reyes-Muñoz
- Research Division, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
| |
Collapse
|
10
|
He L, Su Z, Wang S. The anti-obesity effects of polyphenols: a comprehensive review of molecular mechanisms and signal pathways in regulating adipocytes. Front Nutr 2024; 11:1393575. [PMID: 39539361 PMCID: PMC11557335 DOI: 10.3389/fnut.2024.1393575] [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: 03/01/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024] Open
Abstract
Excess weight gain is a growing concern worldwide, fueled by increased consumption of calorie-dense foods and more sedentary lifestyles. Obesity in China is also becoming increasingly problematic, developing into a major public health concern. Obesity not only increases the risk of associated disease but also imposes a burden on health care systems, and it is thus imperative that an effective intervention approach be identified. Recent studies have demonstrated that the polyphenol-rich Mediterranean diet has considerable potential in this regard. Polyphenols can inhibit the production of adipocytes and reduce adverse reactions, such as inflammation, insulin resistance, and gut microflora imbalance. In this review, we examine four polyphenols (curcumin, ellagic acid, ferulic acid, and quercetin) in terms of their potential as interventions targeting obesity. The mechanisms that help promote adipocyte browning, increase thermogenic factors, increase thermogenesis, and regulate adipocyte differentiation are summarized, and key signaling pathways, including PPARγ, C/EBP-, and others, are reviewed.
Collapse
Affiliation(s)
- Lan He
- Department of Cardiology, The First People’s Hospital of Wenling, Taizhou University Affiliated Wenling Hospital, Zhejiang, China
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan, China
| | - Zhan Su
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan, China
| | - Shuangshuang Wang
- Department of Cardiology, The First People’s Hospital of Wenling, Taizhou University Affiliated Wenling Hospital, Zhejiang, China
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Affiliated First Hospital of Ningbo University, Ningbo, China
| |
Collapse
|
11
|
Santana CVN, Magno LAV, Ramos AV, Rios MA, Sandrim VC, De Marco LA, de Miranda DM, Romano-Silva MA. Genetic Variations in AMPK, FOXO3A, and POMC Increase the Risk of Extreme Obesity. J Obes 2024; 2024:3813621. [PMID: 39484290 PMCID: PMC11527528 DOI: 10.1155/2024/3813621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 09/11/2024] [Indexed: 11/03/2024] Open
Abstract
Objective: Genetic variability significantly impacts metabolism, weight gain, and feeding behaviors, predisposing individuals to obesity. This study explored how variations in key genes related to obesity-FOXO3A (forkhead box O3), AMPK (protein kinase AMP-activated), and POMC (proopiomelanocortin)-are associated with extreme obesity (EOB). Methods: We conducted a case-control study with 251 EOB patients and 212 healthy controls with a body mass index (BMI) of less than 25 kg/m2. We genotyped 10 single nucleotide variants (SNVs) using TaqMan-based assays. Results: Four SNVs-rs1536057 in FOXO3A, rs103685 in AMPK, rs934778, and rs6545975 in POMC-were associated with an increased risk of EOB. The strongest association was observed with rs934778 (POMC), which had a maximum odds ratio (OR) of 5.26 (95% CI: 2.86-9.09). While these genetic variations are closely linked to EOB, they do not affect serum glucose, triglycerides, HDL, LDL, BMI, or waist circumference. Conclusions: These findings indicate that factors beyond traditional metabolic pathways, potentially related to feeding behavior or hormonal regulation, may also link these genetic variations to obesity. Further research in a larger sample is essential to validate these findings and explore their potential to guide clinical interventions and public health strategies.
Collapse
Affiliation(s)
- Cinthia Vila Nova Santana
- Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
- Escola Bahiana de Medicina e Saúde Pública, Salvador, Brazil
| | - Luiz Alexandre Viana Magno
- Programa de Pós-Graduação em Ciências da Saúde (PPGCS), Faculdade Ciências Médicas de Minas Gerais (FCMMG), Belo Horizonte, Brazil
- INCT em Neurotecnologia Responsável (INCT-NeurotecR), Belo Horizonte, Brazil
| | | | - Maria Angélica Rios
- Programa de Pós-Graduação em Ciências da Saúde (PPGCS), Faculdade Ciências Médicas de Minas Gerais (FCMMG), Belo Horizonte, Brazil
| | - Valéria Cristina Sandrim
- Instituto de Biociências Botucatu, Universidade Estadual Paulista Júlio de Mesquita Filho (UNESP), Botucatu, Brazil
| | - Luiz Armando De Marco
- Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
- INCT em Neurotecnologia Responsável (INCT-NeurotecR), Belo Horizonte, Brazil
| | - Débora Marques de Miranda
- Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
- INCT em Neurotecnologia Responsável (INCT-NeurotecR), Belo Horizonte, Brazil
| | - Marco Aurélio Romano-Silva
- Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
- INCT em Neurotecnologia Responsável (INCT-NeurotecR), Belo Horizonte, Brazil
| |
Collapse
|
12
|
Li Y, Yi S, Jiang W, Gong M. Exploring the Relationship Between Different Obesity Metabolism Indices and Hyperuricemia in Patients with Hypertension and Coronary Heart Disease. Diabetes Metab Syndr Obes 2024; 17:3817-3832. [PMID: 39440026 PMCID: PMC11495196 DOI: 10.2147/dmso.s491255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024] Open
Abstract
Background Previous studies have established a strong association between obesity, high metabolism, and the development of hyperuricemia. However, the relationship between obesity metabolism indices and hyperuricemia in high-risk patients with hypertension and coronary heart disease (CHD) remains unclear. The purpose of this study was to investigate this relationship in patients with both hypertension and CHD, and to identify the obesity metabolism index with the best diagnostic value. Methods A two-center study encompassed 6344 participants with hypertension and CHD. Multiple logistic regression was utilized to examine the correlation between six obesity metabolism indices and hyperuricemia, with restricted cubic spline (RCS) analysis to identify non-linear relationships. Diagnostic value was assessed via receiver operating characteristic (ROC) curves and decision curve analysis (DCA). Results Multivariable logistic regression revealed a significant correlation between increased obesity metabolism indices and hyperuricemia. Furthermore, RCS analysis revealed a nonlinear dose-response relationship (P for nonlinear < 0.001). Moreover, ROC and DCA results showed that METS-VF index, which combined visceral obesity and metabolic parameters, became the most reliable diagnostic tool. Conclusion The study underscores a strong association between elevated obesity metabolism indices and hyperuricemia in patients with hypertension and CHD. The METS-VF index, amalgamating visceral obesity and metabolic parameters, emerged as the most reliable diagnostic tool.
Collapse
Affiliation(s)
- Yuanyuan Li
- Cardiovascular Center of the Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, People’s Republic of China
| | - Shanting Yi
- Cardiovascular Center of the Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, People’s Republic of China
| | - Wencai Jiang
- Department of Cardiology, Suining Central Hospital, Suining, 629000, People’s Republic of China
| | - Meihui Gong
- Cardiovascular Center of the Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, People’s Republic of China
| |
Collapse
|
13
|
Zhu XF, Mo YT, Hu YQ, Feng YX, Liu EH. Association between single-point insulin sensitivity estimator and heart failure in older adults: A cross-sectional study. Exp Gerontol 2024; 196:112578. [PMID: 39245081 DOI: 10.1016/j.exger.2024.112578] [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: 06/17/2024] [Revised: 08/22/2024] [Accepted: 09/05/2024] [Indexed: 09/10/2024]
Abstract
BACKGROUND Heart failure (HF) is a condition caused by a malfunction of the heart's pumping function. The single-point insulin sensitivity estimator (SPISE) index is a novel indicator for assessing insulin resistance in humans. However, the connection between the SPISE index and the risk of HF in the elderly is unknown. Therefore, our study aims to evaluate the connection between the SPISE index and HF in older adults. METHODS The study was based on data collected from the 1999-2020 National Health and Nutrition Examination Survey database and included 6165 participants aged ≥60 years. The multivariable linear regression model and the smooth fitting curve model were applied to investigate the connection between the SPISE index and HF in the elderly. Furthermore, the subgroup analysis was performed to investigate the interactive factors. RESULTS In this study, the mean age of the population was 69.38 years. After adjusting for all covariates, we observed that the SPISE index was inversely related to the prevalence of HF (OR = 0.87, 95 % CI = 0.80-0.94, P < 0.001) in older adults. The interaction analysis showed that the association might be affected by diabetes mellitus and smoking status. Additionally, an inflection point between the SPISE index and HF was found among older women. CONCLUSIONS An inverse correlation was detected between the SPISE index and HF in the elderly. This could provide new insight into the prevention and management of HF in the elderly population.
Collapse
Affiliation(s)
- Xiao-Feng Zhu
- Department of Clinical Medicine, The Nanshan School of Guangzhou Medical University, Guangzhou, 511436, China.
| | - Ye-Tong Mo
- Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, 511436, China
| | - Yu-Qi Hu
- Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, 511436, China
| | - Yu-Xue Feng
- Department of Clinical Medicine, The First Clinical School of Guangzhou Medical University, Guangzhou, 511436, China
| | - En-Hui Liu
- Department of Pediatrics, Pediatrics School, Guangzhou Medical University, Guangzhou, 511436, China
| |
Collapse
|
14
|
O’Brien J, Niehaus P, Chang K, Remark J, Barrett J, Dasgupta A, Adenegan M, Salimian M, Kevas Y, Chandrasekaran K, Kristian T, Chellappan R, Rubin S, Kiemen A, Lu CPJ, Russell JW, Ho CY. Skin keratinocyte-derived SIRT1 and BDNF modulate mechanical allodynia in mouse models of diabetic neuropathy. Brain 2024; 147:3471-3486. [PMID: 38554393 PMCID: PMC11449144 DOI: 10.1093/brain/awae100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 03/06/2024] [Accepted: 03/08/2024] [Indexed: 04/01/2024] Open
Abstract
Diabetic neuropathy is a debilitating disorder characterized by spontaneous and mechanical allodynia. The role of skin mechanoreceptors in the development of mechanical allodynia is unclear. We discovered that mice with diabetic neuropathy had decreased sirtuin 1 (SIRT1) deacetylase activity in foot skin, leading to reduced expression of brain-derived neurotrophic factor (BDNF) and subsequent loss of innervation in Meissner corpuscles, a mechanoreceptor expressing the BDNF receptor TrkB. When SIRT1 was depleted from skin, the mechanical allodynia worsened in diabetic neuropathy mice, likely due to retrograde degeneration of the Meissner-corpuscle innervating Aβ axons and aberrant formation of Meissner corpuscles which may have increased the mechanosensitivity. The same phenomenon was also noted in skin-keratinocyte specific BDNF knockout mice. Furthermore, overexpression of SIRT1 in skin induced Meissner corpuscle reinnervation and regeneration, resulting in significant improvement of diabetic mechanical allodynia. Overall, the findings suggested that skin-derived SIRT1 and BDNF function in the same pathway in skin sensory apparatus regeneration and highlighted the potential of developing topical SIRT1-activating compounds as a novel treatment for diabetic mechanical allodynia.
Collapse
Affiliation(s)
- Jennifer O’Brien
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Peter Niehaus
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Koping Chang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Pathology, National Taiwan University, Taipei, 100, Taiwan
| | - Juliana Remark
- Hansjörg Wyss Department of Plastic Surgery, Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | - Joy Barrett
- Hansjörg Wyss Department of Plastic Surgery, Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | - Abhishikta Dasgupta
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Morayo Adenegan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Mohammad Salimian
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Yanni Kevas
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Krish Chandrasekaran
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, USA
| | - Tibor Kristian
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD 21021, USA
| | - Rajeshwari Chellappan
- Department of Pathology, University of Alabama Birmingham, Birmingham, AL 35233, USA
| | - Samuel Rubin
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Chemistry, College of William and Mary, Williamsburg, VA 23187, USA
| | - Ashley Kiemen
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Catherine Pei-Ju Lu
- Hansjörg Wyss Department of Plastic Surgery, Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | - James W Russell
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, USA
| | - Cheng-Ying Ho
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
15
|
Vogt J, Wolf L, Hoelzle LE, Feger M, Föller M. AMP-dependent kinase stimulates the expression of αKlotho. FEBS Open Bio 2024; 14:1691-1700. [PMID: 39090792 PMCID: PMC11452301 DOI: 10.1002/2211-5463.13872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 06/14/2024] [Accepted: 07/17/2024] [Indexed: 08/04/2024] Open
Abstract
Renal αKlotho along with fibroblast growth factor 23 regulates phosphate and vitamin D metabolism. Its cleavage yields soluble Klotho controlling intracellular processes. αKlotho has anti-inflammatory and antioxidant effects and is nephro- and cardioprotective. AMP-dependent kinase (AMPK) is a nephro- and cardioprotective energy sensor. Given that both αKlotho and AMPK have beneficial effects in similar organs, we studied whether AMPK regulates αKlotho gene expression in Madin-Darby canine kidney, normal rat kidney 52E, and human kidney 2 cells. Using quantitative real-time PCR and western blotting, we measured αKlotho expression upon pharmacological manipulation or siRNA-mediated knockdown of AMPKα. AMPK activator 5-aminoimidazole-4-carboxamide ribonucleoside (AICAR) enhanced αKlotho expression, an effect reduced in the presence of AMPK inhibitor compound C or siRNA targeting AMPK catalytic α-subunits (α1 and α2). Similarly, AMPK activators metformin and phenformin upregulated αKlotho transcripts. Taken together, our results suggest that AMPK is a powerful inducer of αKlotho and could thereby contribute to the development of future therapeutic interventions.
Collapse
Affiliation(s)
- Julia Vogt
- Department of PhysiologyUniversity of HohenheimStuttgartGermany
| | - Lisa Wolf
- Department of PhysiologyUniversity of HohenheimStuttgartGermany
| | - Ludwig E. Hoelzle
- Institute of Animal Science, University of HohenheimStuttgartGermany
| | - Martina Feger
- Department of PhysiologyUniversity of HohenheimStuttgartGermany
| | - Michael Föller
- Department of PhysiologyUniversity of HohenheimStuttgartGermany
| |
Collapse
|
16
|
Seok JK, Yang G, Jee JI, Kang HC, Cho YY, Lee HS, Lee JY. Hepatocyte-specific RIG-I loss attenuates metabolic dysfunction-associated steatotic liver disease in mice via changes in mitochondrial respiration and metabolite profiles. Toxicol Res 2024; 40:683-695. [PMID: 39345739 PMCID: PMC11436585 DOI: 10.1007/s43188-024-00264-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 10/01/2024] Open
Abstract
Pattern recognition receptor (PRR)-mediated inflammation is an important determinant of the initiation and progression of metabolic diseases such as metabolic dysfunction-associated steatotic liver disease (MASLD). In this study, we investigated whether RIG-I is involved in hepatic metabolic reprogramming in a high-fat diet (HFD)-induced MASLD model in hepatocyte-specific RIG-I-KO (RIG-I∆hep) mice. Our study revealed that hepatic deficiency of RIG-I improved HFD-induced metabolic imbalances, including glucose impairment and insulin resistance. Hepatic steatosis and liver triglyceride levels were reduced in RIG-I-deficient hepatocytes in HFD-induced MASLD mice, and this was accompanied by the reduced expression of lipogenesis genes, such as PPARγ, Dga2, and Pck1. Hepatic RIG-I deficiency alters whole-body metabolic rates in the HFD-induced MASLD model; there is higher energy consumption in RIG-I∆hep mice. Deletion of RIG-I activated glycolysis and tricarboxylic acid (TCA) cycle-related metabolites in hepatocytes from both HFD-induced MASLD mice and methionine-choline-deficient diet (MCD)-fed mice. RIG-I deficiency enhanced AMPK activation and mitochondrial function in hepatocytes from HFD-induced MASLD mice. These findings indicate that deletion of RIG-I can activate cellular metabolism in hepatocytes by switching on both glycolysis and mitochondrial respiration, resulting in metabolic changes induced by a HFD and stimulation of mitochondrial activity. In summary, RIG-I may be a key regulator of cellular metabolism that influences the development of metabolic diseases such as MASLD. Supplementary Information The online version contains supplementary material available at 10.1007/s43188-024-00264-x.
Collapse
Affiliation(s)
- Jin Kyung Seok
- College of Pharmacy, The Catholic University of Korea, Bucheon, 14662 Republic of Korea
| | - Gabsik Yang
- Department of Pharmacology, College of Korean Medicine, Woosuk University, Jeonbuk, 55338 Republic of Korea
| | - Jung In Jee
- College of Pharmacy, The Catholic University of Korea, Bucheon, 14662 Republic of Korea
| | - Han Chang Kang
- College of Pharmacy, The Catholic University of Korea, Bucheon, 14662 Republic of Korea
| | - Yong-Yeon Cho
- College of Pharmacy, The Catholic University of Korea, Bucheon, 14662 Republic of Korea
| | - Hye Suk Lee
- College of Pharmacy, The Catholic University of Korea, Bucheon, 14662 Republic of Korea
| | - Joo Young Lee
- College of Pharmacy, The Catholic University of Korea, Bucheon, 14662 Republic of Korea
| |
Collapse
|
17
|
Sun M, Lu Z, Chen WM, Lv S, Fu N, Yang Y, Wang Y, Miao M, Wu SY, Zhang J. Metformin monotherapy versus predominantly older non-metformin antidiabetic medications for cerebrovascular risk in early type 2 diabetes management. Diabetes Obes Metab 2024; 26:3914-3925. [PMID: 38952343 DOI: 10.1111/dom.15739] [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: 05/18/2024] [Revised: 06/06/2024] [Accepted: 06/07/2024] [Indexed: 07/03/2024]
Abstract
AIM Choosing the initial treatment for type 2 diabetes (T2D) is pivotal, requiring consideration of solid clinical evidence and patient characteristics. Despite metformin's historical preference, its efficacy in preventing cerebrovascular events lacked empirical validation. This study aimed to evaluate the associations between first-line monotherapy (metformin or non-metformin antidiabetic medications) and cerebrovascular complications in patients with T2D without diabetic complications. METHODS We analysed 9090 patients with T2D without complications who were prescribed either metformin or non-metformin medications as initial therapy. Propensity score matching ensured group comparability. Cox regression analyses, stratified by initial metformin use, assessed cerebrovascular disease risk, adjusting for multiple covariates and using competing risk analysis. Metformin exposure was measured using cumulative defined daily doses. RESULTS Metformin users had a significantly lower crude incidence of cerebrovascular diseases compared with non-users (p < .0001). Adjusted hazard ratios (aHRs) consistently showed an association between metformin use and a lower risk of overall cerebrovascular diseases (aHRs: 0.67-0.69) and severe events (aHRs: 0.67-0.69). The association with reduced risk of mild cerebrovascular diseases was significant across all models (aHRs: 0.73-0.74). Higher cumulative defined daily doses of metformin correlated with reduced cerebrovascular risk (incidence rate ratio: 0.62-0.94, p < .0001), indicating a dose-dependent effect. CONCLUSION Metformin monotherapy is associated with a reduced risk of cerebrovascular diseases in early-stage T2D, highlighting its dose-dependent efficacy. However, the observed benefits might also be influenced by baseline differences and the increased risks associated with other medications, such as sulphonylureas. These findings emphasize the need for personalized diabetes management, particularly in mitigating cerebrovascular risk in early T2D stages.
Collapse
Affiliation(s)
- Mingyang Sun
- Department of Anaesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, China
| | - Zhongyuan Lu
- Department of Anaesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Wan-Ming Chen
- Graduate Institute of Business Administration, College of Management, Fu Jen Catholic University, Taipei, Taiwan
- Artificial Intelligence Development Centre, Fu Jen Catholic University, Taipei, Taiwan
| | - Shuang Lv
- Department of Anaesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, China
| | - Ningning Fu
- Department of Anaesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, China
| | - Yitian Yang
- Department of Anaesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, China
| | - Yangyang Wang
- Department of Anaesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, China
| | - Mengrong Miao
- Department of Anaesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, China
| | - Szu-Yuan Wu
- Graduate Institute of Business Administration, College of Management, Fu Jen Catholic University, Taipei, Taiwan
- Artificial Intelligence Development Centre, Fu Jen Catholic University, Taipei, Taiwan
- Department of Food Nutrition and Health Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan
- Big Data Centre, Lo-Hsu Medical Foundation, Lotung Poh-Ai Hospital, Yilan, Taiwan
- Division of Radiation Oncology, Lo-Hsu Medical Foundation, Lotung Poh-Ai Hospital, Yilan, Taiwan
- Department of Healthcare Administration, College of Medical and Health Science, Asia University, Taichung, Taiwan
- Cancer Centre, Lo-Hsu Medical Foundation, Lotung Poh-Ai Hospital, Yilan, Taiwan
- Centres for Regional Anaesthesia and Pain Medicine, Taipei Municipal Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Management, College of Management, Fo Guang University, Yilan, Taiwan
| | - Jiaqiang Zhang
- Department of Anaesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, China
| |
Collapse
|
18
|
Wei Y, Li X, Cui R, Liu J, Wang G. Associations between sensitivity to thyroid hormones and insulin resistance in euthyroid adults with obesity. Front Endocrinol (Lausanne) 2024; 15:1366830. [PMID: 39175570 PMCID: PMC11338882 DOI: 10.3389/fendo.2024.1366830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 07/10/2024] [Indexed: 08/24/2024] Open
Abstract
Background Impaired sensitivity to thyroid hormones (TH) was associated with metabolic syndrome. The study aimed to explore the association between central TH sensitivity indices and insulin resistance (IR) in euthyroid adults with obesity. Methods This cross-sectional study enrolled 293 euthyroid outpatients with obesity in Beijing Chao-Yang Hospital. We used the thyroid feedback quantile-based index (TFQI), thyroid stimulating hormone index (TSHI), and thyrotrophic T4 resistance index (TT4RI) to indicate central TH sensitivity. IR was assessed by homeostasis model assessment of insulin resistance (HOMA-IR), hepatic insulin resistance index (hepatic-IR), the Matsuda index, and the adipose tissue insulin resistance index (Adipo-IR). Participants were categorized according to tertiles of TH sensitivity indices. We used multiple linear regressions to explore the associations. Results There was a significant stepwise increase in HOMA-IR and Adipo-IR from the lowest to the highest tertiles of TH sensitivity indices (all P<0.05). After adjustment for age, sex, body mass index, hypertension, hyperlipidemia, and diabetes, only Adipo-IR was significantly associated with TH sensitivity indices. On average, each unit increased in TFQI, TSHI, and TT4RI was associated with 1.19 (P=0.053), 1.16 (P=0.04), and 1.01 (P=0.03) units increased in Adipo-IR, respectively. There was no significant association between TH sensitivity indices and HOMA-IR, hepatic-IR, and the Matsuda index after adjustment for other risk factors. Conclusions Reduced central TH sensitivity was associated with increased adipose tissue insulin resistance in euthyroid adults with obesity. The results further confirmed the importance of TH sensitivity on metabolic diseases.
Collapse
Affiliation(s)
| | | | | | - Jia Liu
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Guang Wang
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
19
|
Lee JH, Jung IR, Tu-Sekine B, Jin S, Anokye-Danso F, Ahima RS, Kim SF. Genetic Deletion of Skeletal Muscle Inositol Polyphosphate Multikinase Disrupts Glucose Homeostasis and Impairs Exercise Tolerance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.28.605526. [PMID: 39131310 PMCID: PMC11312436 DOI: 10.1101/2024.07.28.605526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Inositol phosphates are critical signaling messengers involved in a wide range of biological pathways in which inositol polyphosphate multikinase (IPMK) functions as a rate-limiting enzyme for inositol polyphosphate metabolism. IPMK has been implicated in cellular metabolism, but its function at the systemic level is still poorly understood. Since skeletal muscle is a major contributor to energy homeostasis, we have developed a mouse model in which skeletal muscle IPMK is specifically deleted and examined how a loss of IPMK affects whole-body metabolism. Here, we report that mice in which IPMK knockout is deleted, specifically in the skeletal muscle, displayed an increased body weight, disrupted glucose tolerance, and reduced exercise tolerance under the normal diet. Moreover, these changes were associated with an increased accumulation of triglyceride in skeletal muscle. Furthermore, we have confirmed that a loss of IPMK led to reduced beta-oxidation, increased triglyceride accumulation, and impaired insulin response in IPMK-deficient muscle cells. Thus, our results suggest that IPMK mediates the whole-body metabolism via regulating muscle metabolism and may be potentially targeted for the treatment of metabolic syndromes.
Collapse
Affiliation(s)
- Ji-Hyun Lee
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Johns Hopkins University, Baltimore, Maryland, USA. 21224
| | - Ik-Rak Jung
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Johns Hopkins University, Baltimore, Maryland, USA. 21224
| | - Becky Tu-Sekine
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Johns Hopkins University, Baltimore, Maryland, USA. 21224
| | - Sunghee Jin
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Johns Hopkins University, Baltimore, Maryland, USA. 21224
| | - Frederick Anokye-Danso
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Johns Hopkins University, Baltimore, Maryland, USA. 21224
| | - Rexford S Ahima
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Johns Hopkins University, Baltimore, Maryland, USA. 21224
| | - Sangwon F Kim
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Johns Hopkins University, Baltimore, Maryland, USA. 21224
| |
Collapse
|
20
|
Kim HJ, Kwon O. Nutrition and exercise: Cornerstones of health with emphasis on obesity and type 2 diabetes management-A narrative review. Obes Rev 2024; 25:e13762. [PMID: 38715378 DOI: 10.1111/obr.13762] [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: 09/05/2023] [Revised: 02/22/2024] [Accepted: 04/06/2024] [Indexed: 07/11/2024]
Abstract
While a broad consensus exists that integrated nutrition and regular exercise are foundational for health maintenance and serve as a robust non-pharmacological strategy against cardiometabolic diseases, the nuanced interplay between these elements remains incompletely understood. Through multifaceted interactions, these factors profoundly influence primary metabolic organs, notably the skeletal muscle and adipose tissue. Despite the critical nature of this interactivity, a holistic understanding of the combined effects of physical activity and dietary practices is still emerging. This narrative review aims to elucidate the intertwined roles of nutrition and exercise. It provides a comprehensive overview of their synergistic dynamics and emphasizes the importance of a dual-focus approach in mitigating and managing cardiometabolic disorders, predominantly obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Hye Jin Kim
- Laboratory of Developmental Biology and Genomics, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- Korea Mouse Phenotyping Center (KMPC), Seoul, Republic of Korea
- Logme Inc., Seoul, Republic of Korea
| | - Oran Kwon
- Logme Inc., Seoul, Republic of Korea
- Department of Nutritional Science and Food Management, Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul, Republic of Korea
| |
Collapse
|
21
|
Vo N, Zhang Q, Sung HK. From fasting to fat reshaping: exploring the molecular pathways of intermittent fasting-induced adipose tissue remodeling. JOURNAL OF PHARMACY & PHARMACEUTICAL SCIENCES : A PUBLICATION OF THE CANADIAN SOCIETY FOR PHARMACEUTICAL SCIENCES, SOCIETE CANADIENNE DES SCIENCES PHARMACEUTIQUES 2024; 27:13062. [PMID: 39104461 PMCID: PMC11298356 DOI: 10.3389/jpps.2024.13062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 07/05/2024] [Indexed: 08/07/2024]
Abstract
Obesity, characterised by excessive fat accumulation, is a complex chronic condition that results from dysfunctional adipose tissue expansion due to prolonged calorie surplus. This leads to rapid adipocyte enlargement that exceeds the support capacity of the surrounding neurovascular network, resulting in increased hypoxia, inflammation, and insulin resistance. Intermittent fasting (IF), a dietary regimen that cycles between periods of fasting and eating, has emerged as an effective strategy to combat obesity and improve metabolic homeostasis by promoting healthy adipose tissue remodeling. However, the precise molecular and cellular mechanisms behind the metabolic improvements and remodeling of white adipose tissue (WAT) driven by IF remain elusive. This review aims to summarise and discuss the relationship between IF and adipose tissue remodeling and explore the potential mechanisms through which IF induces alterations in WAT. This includes several key structural changes, including angiogenesis and sympathetic innervation of WAT. We will also discuss the involvement of key signalling pathways, such as PI3K, SIRT, mTOR, and AMPK, which potentially play a crucial role in IF-mediated metabolic adaptations.
Collapse
Affiliation(s)
- Nathaniel Vo
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Qiwei Zhang
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Hoon-Ki Sung
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
22
|
Kim B, Lee Y, Lee C, Jung ES, Kang H, Holzapfel WH. Comprehensive Amelioration of Metabolic Dysfunction through Administration of Lactiplantibacillus plantarum APsulloc 331261 (GTB1™) in High-Fat-Diet-Fed Mice. Foods 2024; 13:2227. [PMID: 39063311 PMCID: PMC11276112 DOI: 10.3390/foods13142227] [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: 06/10/2024] [Revised: 07/10/2024] [Accepted: 07/13/2024] [Indexed: 07/28/2024] Open
Abstract
The beneficial effects of probiotics for the improvement of metabolic disorders have been studied intensively; however, these effects are evident in a probiotic strain-specific and disease-specific manner. Thus, it is still essential to evaluate the efficacy of each strain against a target disease. Here, we present an anti-obese and anti-diabetic probiotic strain, Lactiplantibacillus plantarum APsulloc331261 (GTB1™), which was isolated from green tea and tested for safety previously. In high-fat-diet-induced obese mice, GTB1™ exerted multiple beneficial effects, including significant reductions in adiposity, glucose intolerance, and dyslipidemia, which were further supported by improvements in levels of circulating hormones and adipokines. Lipid metabolism in adipose tissues was restored through the activation of PPAR/PGC1α signaling by GTB1™ treatment, which was facilitated by intestinal microbiota composition changes and short-chain fatty acid production. Our findings provide evidence to suggest that GTB1™ is a potential candidate for probiotic supplementation for comprehensive improvement in metabolic disorders.
Collapse
Affiliation(s)
- Bobae Kim
- Basic Research Center, HEM Pharma Inc., Pohang 37554, Republic of Korea; (B.K.); (Y.L.); (C.L.)
- Department of Advanced Convergence, Handong Global University, Pohang 37554, Republic of Korea
| | - Yuri Lee
- Basic Research Center, HEM Pharma Inc., Pohang 37554, Republic of Korea; (B.K.); (Y.L.); (C.L.)
- Department of Advanced Convergence, Handong Global University, Pohang 37554, Republic of Korea
| | - Chungho Lee
- Basic Research Center, HEM Pharma Inc., Pohang 37554, Republic of Korea; (B.K.); (Y.L.); (C.L.)
| | - Eun Sung Jung
- Multi-Omics Center, HEM Pharma Inc., Suwon 16229, Republic of Korea;
| | - Hyeji Kang
- Basic Research Center, HEM Pharma Inc., Pohang 37554, Republic of Korea; (B.K.); (Y.L.); (C.L.)
- Global Green Research Institute, Handong Global University, Pohang 37554, Republic of Korea
| | - Wilhelm H. Holzapfel
- Basic Research Center, HEM Pharma Inc., Pohang 37554, Republic of Korea; (B.K.); (Y.L.); (C.L.)
- Department of Advanced Convergence, Handong Global University, Pohang 37554, Republic of Korea
| |
Collapse
|
23
|
Biao Y, Li D, Zhang Y, Gao J, Xiao Y, Yu Z, Li L. Wulingsan Alleviates MAFLD by Activating Autophagy via Regulating the AMPK/mTOR/ULK1 Signaling Pathway. Can J Gastroenterol Hepatol 2024; 2024:9777866. [PMID: 39035827 PMCID: PMC11260214 DOI: 10.1155/2024/9777866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 04/19/2024] [Accepted: 06/24/2024] [Indexed: 07/23/2024] Open
Abstract
Here, we presented the study of the molecular mechanisms underlying the action of Wulingsan (WLS) in rats with metabolic-associated fatty liver disease (MAFLD) induced by a high-fat diet (HFD). High-performance liquid chromatography was employed to identify the chemical components of WLS. After 2 weeks of HFD induction, MAFLD rats were treated with WLS in three different doses for 6 weeks, a positive control treatment or with a vehicle. Lipid metabolism, liver function, oxidative stress, and inflammatory factors as well as pathomorphological changes in liver parenchyma were assessed in all groups. Finally, the expressions of autophagy-related markers, adenosine monophosphate-activated protein kinase (AMPK)/mechanistic target of rapamycin (mTOR)/unc-51-like kinase-1 (ULK1) signaling pathway-related genes, and proteins in liver were detected. The results revealed that WLS significantly ameliorated liver injury, the dysfunction of the lipid metabolism, the oxidative stress, and overall inflammatory status. Furthermore, WLS increased the expressions of LC3B-II, Beclin1, p-AMPK, and ULK1, along with decreased p62, p-mTOR, and sterol regulatory element-binding protein-1c levels. In conclusion, we showed that WLS is capable of alleviating HFD-induced MAFLD by improving lipid accumulation, suppressing oxidative stress and inflammation, and promoting autophagy.
Collapse
Affiliation(s)
- Yaning Biao
- School of Basic MedicineHebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Dantong Li
- School of PharmacyHebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Yixin Zhang
- School of PharmacyHebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Jingmiao Gao
- School of PharmacyHebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Yi Xiao
- School of PharmacyHebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Zehe Yu
- School of PharmacyHebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Li Li
- School of PharmacyHebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
24
|
Akamine MAV, Ferreira Soares BMA, Telles JPM, Cicupira Rodrigues de Assis A, Rodriguez GNV, Soares PR, Chalela WA, Scudeler TL. Role of Dapagliflozin in Ischemic Preconditioning in Patients with Symptomatic Coronary Artery Disease-DAPA-IP Study Protocol. Pharmaceuticals (Basel) 2024; 17:920. [PMID: 39065769 PMCID: PMC11280174 DOI: 10.3390/ph17070920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/05/2024] [Accepted: 07/07/2024] [Indexed: 07/28/2024] Open
Abstract
Background: Ischemic preconditioning (IP) is a powerful cellular protection mechanism. The cellular pathways underlying IP are extremely complex and involve the participation of cell triggers, intracellular signaling pathways, and end-effectors. Experimental studies have shown that sodium-glucose transport protein 2 (SGLT2) inhibitors promote activation of 5'-adenosine monophosphate (AMP)-activated protein kinase (AMPK), the main regulator of adenosine 5'-triphosphate homeostasis and energy metabolism in the body. Despite its cardioprotective profile demonstrated by numerous clinical trials, the results of studies on the action of SGLT2 inhibitors in IP are scarce. This study will investigate the effects of dapagliflozin on IP in patients with coronary artery disease (CAD). Methods: The study will include 50 patients with multivessel CAD, ischemia documented by stress testing, and preserved left ventricular ejection fraction (LVEF). Patients will undergo four exercise tests, the first two with a time interval of 30 min between them after washout of cardiovascular or hypoglycemic medications and the last two after 7 days of dapagliflozin 10 mg once a day, also with a time interval of 30 min between them. Discussion: The role of SGLT2 inhibitors on IP is not clearly established. Several clinical trials have shown that SGLT2 inhibitors reduce the occurrence cardiovascular events, notably heart failure. However, such studies have not shown beneficial metabolic effects of SGLT2 inhibitors, such as reducing myocardial infarction or stroke. On the other hand, experimental studies with animal models have shown the beneficial effects of SGLT2 inhibitors on IP, a mechanism that confers cardiac and vascular protection from subsequent ischemia-reperfusion (IR) injury. This is the first clinical study to evaluate the effects of SGLT2 inhibitors on IP, which could result in an important advance in the treatment of patients with stable CAD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Thiago Luis Scudeler
- Instituto do Coração (InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Enéas de Carvalho Aguiar, 44, Cerqueira César, São Paulo 05403-000, Brazil; (M.A.V.A.); (B.M.A.F.S.); (J.P.M.T.); (A.C.R.d.A.); (G.N.V.R.); (P.R.S.); (W.A.C.)
| |
Collapse
|
25
|
MacMillan S, Burns DP, O'Halloran KD, Evans AM. SubSol-HIe is an AMPK-dependent hypoxia-responsive subnucleus of the nucleus tractus solitarius that coordinates the hypoxic ventilatory response and protects against apnoea in mice. Pflugers Arch 2024; 476:1087-1107. [PMID: 38635058 PMCID: PMC11166843 DOI: 10.1007/s00424-024-02957-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/24/2024] [Accepted: 03/31/2024] [Indexed: 04/19/2024]
Abstract
Functional magnetic resonance imaging (fMRI) suggests that the hypoxic ventilatory response is facilitated by the AMP-activated protein kinase (AMPK), not at the carotid bodies, but within a subnucleus (Bregma -7.5 to -7.1 mm) of the nucleus tractus solitarius that exhibits right-sided bilateral asymmetry. Here, we map this subnucleus using cFos expression as a surrogate for neuronal activation and mice in which the genes encoding the AMPK-α1 (Prkaa1) and AMPK-α2 (Prkaa2) catalytic subunits were deleted in catecholaminergic cells by Cre expression via the tyrosine hydroxylase promoter. Comparative analysis of brainstem sections, relative to controls, revealed that AMPK-α1/α2 deletion inhibited, with right-sided bilateral asymmetry, cFos expression in and thus activation of a neuronal cluster that partially spanned three interconnected anatomical nuclei adjacent to the area postrema: SolDL (Bregma -7.44 mm to -7.48 mm), SolDM (Bregma -7.44 mm to -7.48 mm) and SubP (Bregma -7.48 mm to -7.56 mm). This approximates the volume identified by fMRI. Moreover, these nuclei are known to be in receipt of carotid body afferent inputs, and catecholaminergic neurons of SubP and SolDL innervate aspects of the ventrolateral medulla responsible for respiratory rhythmogenesis. Accordingly, AMPK-α1/α2 deletion attenuated hypoxia-evoked increases in minute ventilation (normalised to metabolism), reductions in expiration time, and increases sigh frequency, but increased apnoea frequency during hypoxia. The metabolic response to hypoxia in AMPK-α1/α2 knockout mice and the brainstem and spinal cord catecholamine levels were equivalent to controls. We conclude that within the brainstem an AMPK-dependent, hypoxia-responsive subnucleus partially spans SubP, SolDM and SolDL, namely SubSol-HIe, and is critical to coordination of active expiration, the hypoxic ventilatory response and defence against apnoea.
Collapse
Affiliation(s)
- Sandy MacMillan
- Centre for Discovery Brain Sciences, College of Medicine and Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - David P Burns
- Department of Physiology, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland
| | - Ken D O'Halloran
- Department of Physiology, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland
| | - A Mark Evans
- Centre for Discovery Brain Sciences, College of Medicine and Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh, EH8 9XD, UK.
| |
Collapse
|
26
|
Dalle S. Targeting Protein Kinases to Protect Beta-Cell Function and Survival in Diabetes. Int J Mol Sci 2024; 25:6425. [PMID: 38928130 PMCID: PMC11203834 DOI: 10.3390/ijms25126425] [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: 04/30/2024] [Revised: 05/30/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024] Open
Abstract
The prevalence of diabetes is increasing worldwide. Massive death of pancreatic beta-cells causes type 1 diabetes. Progressive loss of beta-cell function and mass characterizes type 2 diabetes. To date, none of the available antidiabetic drugs promotes the maintenance of a functional mass of endogenous beta-cells, revealing an unmet medical need. Dysfunction and apoptotic death of beta-cells occur, in particular, through the activation of intracellular protein kinases. In recent years, protein kinases have become highly studied targets of the pharmaceutical industry for drug development. A number of drugs that inhibit protein kinases have been approved for the treatment of cancers. The question of whether safe drugs that inhibit protein kinase activity can be developed and used to protect the function and survival of beta-cells in diabetes is still unresolved. This review presents arguments suggesting that several protein kinases in beta-cells may represent targets of interest for the development of drugs to treat diabetes.
Collapse
Affiliation(s)
- Stéphane Dalle
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), 34094 Montpellier, France
| |
Collapse
|
27
|
Song L, Li Y, Xu M. Exogenous Nucleotides Ameliorate Insulin Resistance Induced by Palmitic Acid in HepG2 Cells through the IRS-1/AKT/FOXO1 Pathways. Nutrients 2024; 16:1801. [PMID: 38931156 PMCID: PMC11206901 DOI: 10.3390/nu16121801] [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: 04/10/2024] [Revised: 06/01/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Nucleotides (NTs) act as pivotal regulatory factors in numerous biological processes, playing indispensable roles in growth, development, and metabolism across organisms. This study delves into the effects of exogenous NTs on hepatic insulin resistance using palmitic-acid-induced HepG2 cells, administering interventions at three distinct dosage levels of exogenous NTs. The findings underscore that exogenous NT intervention augments glucose consumption in HepG2 cells, modulates the expression of glycogen-synthesis-related enzymes (glycogen synthase kinase 3β and glycogen synthase), and influences glycogen content. Additionally, it governs the expression levels of hepatic enzymes (hexokinase, phosphoenolpyruvate carboxykinase, and glucose-6-phosphatase). Moreover, exogenous NT intervention orchestrates insulin signaling pathway (insulin receptor substrate-1, protein kinase B, and forkhead box protein O1) and AMP-activated protein kinase (AMPK) activity in HepG2 cells. Furthermore, exogenous NT intervention fine-tunes the expression levels of oxidative stress-related markers (malondialdehyde, glutathione peroxidase, and NADPH oxidase 4) and the expression of inflammation-related nuclear transcription factor (NF-κB). Lastly, exogenous NT intervention regulates the expression levels of glucose transporter proteins (GLUTs). Consequently, exogenous NTs ameliorate insulin resistance in HepG2 cells by modulating the IRS-1/AKT/FOXO1 pathways and regulate glucose consumption, glycogen content, insulin signaling pathways, AMPK activity, oxidative stress, and inflammatory status.
Collapse
Affiliation(s)
- Lixia Song
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China; (L.S.); (Y.L.)
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| | - Yong Li
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China; (L.S.); (Y.L.)
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| | - Meihong Xu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China; (L.S.); (Y.L.)
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
- Institute of Medical Technology, Peking University Health Science Center, Beijing 100019, China
| |
Collapse
|
28
|
Aldossary KM, Ali LS, Abdallah MS, Bahaa MM, Elmasry TA, Elberri EI, Kotkata FA, El Sabaa RM, Elmorsi YM, Kamel MM, Negm WA, Elberri AI, Hamouda AO, AlRasheed HA, Salahuddin MM, Yasser M, Hamouda MA. Effect of a high dose atorvastatin as added-on therapy on symptoms and serum AMPK/NLRP3 inflammasome and IL-6/STAT3 axes in patients with major depressive disorder: randomized controlled clinical study. Front Pharmacol 2024; 15:1381523. [PMID: 38855751 PMCID: PMC11157054 DOI: 10.3389/fphar.2024.1381523] [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: 02/03/2024] [Accepted: 04/08/2024] [Indexed: 06/11/2024] Open
Abstract
Background Neuroinflammation pathways have been associated with the development of major depressive disorders (MDD). The anti-inflammatory characteristics of statins have been demonstrated to have significance in the pathophysiology of depression. Aim To investigate the mechanistic pathways of high dose atorvastatin in MDD. Patients and methods This trial included 60 patients with MDD who met the eligibility requirements. Two groups of patients (n = 30) were recruited by selecting patients from the Psychiatry Department. Group 1 received 20 mg of fluoxetine plus a placebo once daily. Group 2 received fluoxetine and atorvastatin (80 mg) once daily. All patients were assessed by a psychiatrist using the Hamilton Depression Rating Scale (HDRS). A HDRS score of ≤7 indicates remission or partial remission [HDRS<17 and>7]. Response was defined as ≥ 50% drop in the HDRS score. The serum concentrations of nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 (NLRP-3), interleukin-6 (IL-6), adenosine monophosphate activated protein kinase (AMPK), and signal transducer and activator of transcription factor-3 (STAT-3) were measured. Results The atorvastatin group showed a significant reduction in the levels of all measured markers along with a statistical increase in the levels of AMPK when compared to the fluoxetine group. The atorvastatin group displayed a significant decrease in HDRS when compared to its baseline and the fluoxetine group. The response rate and partial remission were higher in the atorvastatin group than fluoxetine (p = 0.03, and p = 0.005), respectively. Conclusion These results imply that atorvastatin at high doses may be a promising adjuvant therapy for MDD patients by altering the signaling pathways for AMPK/NLRP3 and IL-6/STAT-3. Clinical Trial Registration clinicaltrials.gov, identifier NCT05792540.
Collapse
Affiliation(s)
- Khlood Mohammad Aldossary
- Department of Pharmacy Practice, College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Lashin Saad Ali
- Department of Basic Medical Science, Faculty of Dentistry, Al-Ahliyya Amman University, Amman, Jordan
- Physiology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Mahmoud S. Abdallah
- Department of Clinical Pharmacy, Faculty of Pharmacy, University of Sadat City (USC), Sadat City, Menoufia, Egypt
- Department of PharmD, Faculty of Pharmacy, Jadara University, Irbid, Jordan
| | - Mostafa M. Bahaa
- Pharmacy Practice Department, Faculty of Pharmacy, Horus University, New Damietta, Egypt
| | - Thanaa A. Elmasry
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Tanta University, Tanta, Al-Gharbia, Egypt
| | - Eman I. Elberri
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tanta University, Tanta, Al-Gharbia, Egypt
| | - Fedaa A. Kotkata
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tanta University, Tanta, Al-Gharbia, Egypt
| | - Ramy M. El Sabaa
- Department of Clinical Pharmacy, Faculty of Pharmacy, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| | - Yasmine M. Elmorsi
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tanta University, Tanta, Al-Gharbia, Egypt
| | - Mostafa M. Kamel
- Psychiatry Department, Faculty of Medicine, Tanta University, Egypt
| | - Walaa A. Negm
- Pharmacognosy Department, Faculty of Pharmacy, Tanta University, Tanta, Al-Gharbia, Egypt
| | - Aya Ibrahim Elberri
- Genetic Engineering and Molecular Biology Division, Department of Zoology, Faculty of Science, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| | - Amir O. Hamouda
- Department of Biochemistry and Pharmacology, Faculty of Pharmacy, Horus University, New Damietta, Egypt
| | - Hayam Ali AlRasheed
- Department of Pharmacy Practice, College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Muhammed M. Salahuddin
- Department of Biochemistry and Pharmacology, Faculty of Pharmacy, Horus University, New Damietta, Egypt
| | - Mohamed Yasser
- Department of Pharmaceutics, Faculty of Pharmacy, Port Said University, Port Said, Egypt
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Horus University, New Damietta, Egypt
| | - Manal A. Hamouda
- Department of Clinical Pharmacy, Faculty of Pharmacy, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| |
Collapse
|
29
|
Jia W, Zhong L, Ren Q, Teng D, Gong L, Dong H, Li J, Wang C, He YX, Yang J. Microcystin-RR promote lipid accumulation through CD36 mediated signal pathway and fatty acid uptake in HepG2 cells. ENVIRONMENTAL RESEARCH 2024; 249:118402. [PMID: 38309560 DOI: 10.1016/j.envres.2024.118402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/19/2024] [Accepted: 02/01/2024] [Indexed: 02/05/2024]
Abstract
Microcystins (MC)-RR is a significant analogue of MC-LR, which has been identified as a hepatotoxin capable of influencing lipid metabolism and promoting the progression of liver-related metabolic diseases. However, the toxicity and biological function of MC-RR are still not well understood. In this study, the toxic effects and its role in lipid metabolism of MC-RR were investigated in hepatoblastoma cells (HepG2cells). The results demonstrated that MC-RR dose-dependently reduced cell viability and induced apoptosis. Additionally, even at low concentrations, MC-RR promoted lipid accumulation through up-regulating levels of triglyceride, total cholesterol, phosphatidylcholines and phosphatidylethaolamine in HepG2 cells, with no impact on cell viability. Proteomics and transcriptomics analysis further revealed significant alterations in the protein and gene expression profiles in HepG2 cells treated with MC-RR. Bioinformatic analysis, along with subsequent validation, indicated the upregulation of CD36 and activation of the AMPK and PI3K/AKT/mTOR in response to MC-RR exposure. Finally, knockdown of CD36 markedly ameliorated MC-RR-induced lipid accumulation in HepG2 cells. These findings collectively suggest that MC-RR promotes lipid accumulation in HepG2 cells through CD36-mediated signal pathway and fatty acid uptake. Our findings provide new insights into the hepatotoxic mechanism of MC-RR.
Collapse
Affiliation(s)
- Wenjuan Jia
- School of Basic Medical Sciences, Qingdao University, Qingdao, 266071, China; Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, China.
| | - Lin Zhong
- School of Basic Medical Sciences, Qingdao University, Qingdao, 266071, China; Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, China
| | - Qingmiao Ren
- The Precision Medicine Laboratory, The First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Da Teng
- School of Basic Medical Sciences, Qingdao University, Qingdao, 266071, China; Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, China
| | - Lei Gong
- Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, China
| | - Haibin Dong
- Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, China
| | - Jun Li
- School of Basic Medical Sciences, Qingdao University, Qingdao, 266071, China; Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, China
| | - Chunxiao Wang
- Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, China
| | - Yong-Xing He
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, 730000, China.
| | - Jun Yang
- Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, China.
| |
Collapse
|
30
|
Kim TH. Ginsenosides for the treatment of insulin resistance and diabetes: Therapeutic perspectives and mechanistic insights. J Ginseng Res 2024; 48:276-285. [PMID: 38707641 PMCID: PMC11068994 DOI: 10.1016/j.jgr.2024.03.002] [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: 12/15/2023] [Revised: 01/26/2024] [Accepted: 03/04/2024] [Indexed: 05/07/2024] Open
Abstract
Diabetes mellitus (DM) is a systemic disorder of energy metabolism characterized by a sustained elevation of blood glucose in conjunction with impaired insulin action in multiple peripheral tissues (i.e., insulin resistance). Although extensive research has been conducted to identify therapeutic targets for the treatment of DM, its global prevalence and associated mortailty rates are still increasing, possibly because of challenges related to long-term adherence, limited efficacy, and undesirable side effects of currently available medications, implying an urgent need to develop effective and safe pharmacotherapies for DM. Phytochemicals have recently drawn attention as novel pharmacotherapies for DM based on their clinical relevance, therapeutic efficacy, and safety. Ginsenosides, pharmacologically active ingredients primarily found in ginseng, have long been used as adjuvants to traditional medications in Asian countries and have been reported to exert promising therapeutic efficacy in various metabolic diseases, including hyperglycemia and diabetes. This review summarizes the current pharmacological effects of ginsenosides and their mechanistic insights for the treatment of insulin resistance and DM, providing comprehensive perspectives for the development of novel strategies to treat DM and related metabolic complications.
Collapse
Affiliation(s)
- Tae Hyun Kim
- Drug Information Research Institute, Muscle Physiome Research Center, College of Pharmacy, Sookmyung Women's University, Seoul, Republic of Korea
| |
Collapse
|
31
|
Yuan Q, Zhang X, Yang X, Zhang Q, Wei X, Ding Z, Chen J, Hua H, Huang D, Xu Y, Wang X, Gao C, Liu S, Zhang H. Knockdown of ACOT4 alleviates gluconeogenesis and lipid accumulation in hepatocytes. Heliyon 2024; 10:e27618. [PMID: 38495177 PMCID: PMC10940928 DOI: 10.1016/j.heliyon.2024.e27618] [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: 08/08/2023] [Revised: 02/28/2024] [Accepted: 03/04/2024] [Indexed: 03/19/2024] Open
Abstract
Acyl-CoA thioesterase 4 (ACOT4) has been reported to be related to acetyl-CoA carboxylase activity regulation; However, its exact functions in liver lipid and glucose metabolism are still unclear. Here, we discovered explored the regulatory roles of ACOT4 in hepatic lipid and glucose metabolism in vitro. We found that the expression level of ACOT4 was significantly increased in the hepatic of db/db and ob/ob mice as well as obese mice fed a high fat diet. Adenovirus-mediated overexpression of ACOT4 promoted gluconeogenesis and high-glucose/high-insulin-induced lipid accumulation and impaired insulin sensitivity in primary mouse hepatocytes, whereas ACOT4 knockdown notably suppressed gluconeogenesis and decreased the triglycerides accumulation in hepatocytes. Furthermore, ACOT4 knockdown increased insulin-induced phosphorylation of AKT and GSK-3β in primary mouse hepatocytes. Mechanistically, we found that upregulation of ACOT4 expression inhibited AMP-activated protein kinase (AMPK) activity, and its knockdown had the opposite effect. However, activator A769662 and inhibitor compound C of AMPK suppressed the impact of the change in ACOT4 expression on AMPK activity. Our data indicated that ACOT4 is related to hepatic glucose and lipid metabolism, primarily via the regulation of AMPK activity. In conclusion, ACOT4 is a potential target for the therapy of non-alcoholic fatty liver (NAFLD) and type 2 diabetes.
Collapse
Affiliation(s)
- Qianqian Yuan
- Department of Biochemistry and Molecular Biology, Metabolic Disease Research Center, School of Basic Medicine, Anhui Medical University, Hefei, 230032, China
| | - Xiaomin Zhang
- Department of Biochemistry and Molecular Biology, Metabolic Disease Research Center, School of Basic Medicine, Anhui Medical University, Hefei, 230032, China
| | - Xiaonan Yang
- Department of Otorhinolaryngology Head and Neck Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Qing Zhang
- Department of Biochemistry and Molecular Biology, Metabolic Disease Research Center, School of Basic Medicine, Anhui Medical University, Hefei, 230032, China
| | - Xiang Wei
- Department of Biochemistry and Molecular Biology, Metabolic Disease Research Center, School of Basic Medicine, Anhui Medical University, Hefei, 230032, China
- Department of Hyperbaric Oxygen, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, 230011, China
| | - Zhimin Ding
- Department of Otorhinolaryngology Head and Neck Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Jiajie Chen
- Department of Dermatology, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Hongting Hua
- Department of Otorhinolaryngology Head and Neck Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Dake Huang
- Synthetic Laboratory of School of Basic Medicine Sciences, Anhui Medical University, Hefei, 230032, China
| | - Yongxia Xu
- Department of Endocrinology, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Xiuyun Wang
- Department of Biochemistry and Molecular Biology, Metabolic Disease Research Center, School of Basic Medicine, Anhui Medical University, Hefei, 230032, China
| | - Chaobing Gao
- Department of Otorhinolaryngology Head and Neck Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Shengxiu Liu
- Department of Dermatology, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Huabing Zhang
- Department of Biochemistry and Molecular Biology, Metabolic Disease Research Center, School of Basic Medicine, Anhui Medical University, Hefei, 230032, China
- Anhui Provincial Institute of Translational Medicine, Hefei, 230022, Anhui, China
| |
Collapse
|
32
|
Bustraan S, Bennett J, Whilding C, Pennycook BR, Smith D, Barr AR, Read J, Carling D, Pollard A. AMP-activated protein kinase activation suppresses leptin expression independently of adipogenesis in primary murine adipocytes. Biochem J 2024; 481:345-362. [PMID: 38314646 PMCID: PMC11088909 DOI: 10.1042/bcj20240003] [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: 01/06/2024] [Revised: 02/01/2024] [Accepted: 02/04/2024] [Indexed: 02/06/2024]
Abstract
Adipogenesis, defined as the development of mature adipocytes from stem cell precursors, is vital for the expansion, turnover and health of adipose tissue. Loss of adipogenic potential in adipose stem cells, or impairment of adipogenesis is now recognised as an underlying cause of adipose tissue dysfunction and is associated with metabolic disease. In this study, we sought to determine the role of AMP-activated protein kinase (AMPK), an evolutionarily conserved master regulator of energy homeostasis, in adipogenesis. Primary murine adipose-derived stem cells were treated with a small molecule AMPK activator (BI-9774) during key phases of adipogenesis, to determine the effect of AMPK activation on adipocyte commitment, maturation and function. To determine the contribution of the repression of lipogenesis by AMPK in these processes, we compared the effect of pharmacological inhibition of acetyl-CoA carboxylase (ACC). We show that AMPK activation inhibits adipogenesis in a time- and concentration-dependent manner. Transient AMPK activation during adipogenic commitment leads to a significant, ACC-independent, repression of adipogenic transcription factor expression. Furthermore, we identify a striking, previously unexplored inhibition of leptin gene expression in response to both short-term and chronic AMPK activation irrespective of adipogenesis. These findings reveal that in addition to its effect on adipogenesis, AMPK activation switches off leptin gene expression in primary mouse adipocytes independently of adipogenesis. Our results identify leptin expression as a novel target of AMPK through mechanisms yet to be identified.
Collapse
Affiliation(s)
- Sophia Bustraan
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, U.K
- Medical Research Council Laboratory of Medical Sciences, London, U.K
| | - Jane Bennett
- Medical Research Council Laboratory of Medical Sciences, London, U.K
| | - Chad Whilding
- Medical Research Council Laboratory of Medical Sciences, London, U.K
| | | | - David Smith
- Emerging Innovations Unit, Discovery Sciences, R&D, AstraZeneca, Cambridge, U.K
| | - Alexis R. Barr
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, U.K
- Medical Research Council Laboratory of Medical Sciences, London, U.K
| | - Jon Read
- Mechanistic and Structural Biology, Biopharmaceuticals R&D, AstraZeneca, Cambridge, U.K
| | - David Carling
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, U.K
- Medical Research Council Laboratory of Medical Sciences, London, U.K
| | - Alice Pollard
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, U.K
| |
Collapse
|
33
|
Żołnierkiewicz O, Rogacka D. Hyperglycemia - A culprit of podocyte pathology in the context of glycogen metabolism. Arch Biochem Biophys 2024; 753:109927. [PMID: 38350532 DOI: 10.1016/j.abb.2024.109927] [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: 10/26/2023] [Revised: 01/31/2024] [Accepted: 02/10/2024] [Indexed: 02/15/2024]
Abstract
Prolonged disruption in the balance of glucose can result in metabolic disorders. The kidneys play a significant role in regulating blood glucose levels. However, when exposed to chronic hyperglycemia, the kidneys' ability to handle glucose metabolism may be impaired, leading to an accumulation of glycogen. Earlier studies have shown that there can be a significant increase in glucose storage in the form of glycogen in the kidneys in diabetes. Podocytes play a crucial role in maintaining the integrity of filtration barrier. In diabetes, exposure to elevated glucose levels can lead to significant metabolic and structural changes in podocytes, contributing to kidney damage and the development of diabetic kidney disease. The accumulation of glycogen in podocytes is not a well-established phenomenon. However, a recent study has demonstrated the presence of glycogen granules in podocytes. This review delves into the intricate connections between hyperglycemia and glycogen metabolism within the context of the kidney, with special emphasis on podocytes. The aberrant storage of glycogen has the potential to detrimentally impact podocyte functionality and perturb their structural integrity. This review provides a comprehensive analysis of the alterations in cellular signaling pathways that may potentially lead to glycogen overproduction in podocytes.
Collapse
Affiliation(s)
- Olga Żołnierkiewicz
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Wita Stwosza 63, 80-308, Gdansk, Poland
| | - Dorota Rogacka
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Wita Stwosza 63, 80-308, Gdansk, Poland; University of Gdansk, Faculty of Chemistry, Department of Molecular Biotechnology, Wita Stwosza 63, 80-308, Gdansk, Poland.
| |
Collapse
|
34
|
Chen K, Liu S, Wei Y. Sub-nanosized vanadate hybrid clusters maintain glucose homeostasis and restore treatment response in inflammatory disease in obese mice. NANO RESEARCH 2024; 17:1818-1826. [DOI: 10.1007/s12274-023-6366-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 09/11/2024]
|
35
|
Swargiary D, Kashyap B, Sarma P, Ahmed SA, Gurumayum S, Barge SR, Basumatary D, Borah JC. Free radical scavenging polyphenols isolated from Phyllanthus niruri L. ameliorates hyperglycemia via SIRT1 induction and GLUT4 translocation in in vitro and in vivo models. Fitoterapia 2024; 173:105803. [PMID: 38171388 DOI: 10.1016/j.fitote.2023.105803] [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: 09/29/2023] [Revised: 12/12/2023] [Accepted: 12/28/2023] [Indexed: 01/05/2024]
Abstract
Type 2 diabetes milletus (T2DM) is a complex multifaceted disorder characterized by insulin resistance in skeletal muscle. Phyllanthus niruri L. is well reported sub-tropical therapeutically beneficial ayurvedic medicinal plant from Euphorbiaceae family used in various body ailments such as metabolic disorder including diabetes. The present study emphasizes on the therapeutic potential of Phyllanthus niruri L. and its phytochemical(s) against insulin resistance conditions and impaired antioxidant activity thereby aiding as an anti-hyperglycemic agent in targeting T2DM. Three compounds were isolated from the most active ethyl acetate fraction namely compound 1 as 1-O-galloyl-6-O-luteoyl-β-D-glucoside, compound 2 as brevifolincarboxylic acid and compound 3 as ricinoleic acid. Compounds 1 and 2, the two polyphenols enhanced the uptake of glucose and inhibited ROS levels in palmitate induced C2C12 myotubes. PNEAF showed the potent enhancement of glucose uptake in palmitate-induced insulin resistance condition in C2C12 myotubes and significant ROS inhibition was observed in skeletal muscle cell line. PNEAF treated IR C2C12 myotubes and STZ induced Wistar rats elevated SIRT1, PGC1-α signaling cascade through phosphorylation of AMPK and GLUT4 translocation resulting in insulin sensitization. Our study revealed an insight into the efficacy of marker compounds isolated from P. niruri and its enriched ethyl acetate fraction as ROS scavenging agent and helps in attenuating insulin resistance condition in C2C12 myotubes as well as in STZ induced Wistar rat by restoring glucose metabolism. Overall, this study can provide prospects for the marker-assisted development of P. niruri as a phytopharmaceutical drug for the insulin resistance related diabetic complications.
Collapse
Affiliation(s)
- Deepsikha Swargiary
- Chemical Biology Lab-I, Institute of Advanced Study in Science and Technology (IASST), Guwahati, Assam, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P, India
| | - Bhaswati Kashyap
- Chemical Biology Lab-I, Institute of Advanced Study in Science and Technology (IASST), Guwahati, Assam, India
| | - Pranamika Sarma
- Chemical Biology Lab-I, Institute of Advanced Study in Science and Technology (IASST), Guwahati, Assam, India
| | - Semim Akhtar Ahmed
- Chemical Biology Lab-I, Institute of Advanced Study in Science and Technology (IASST), Guwahati, Assam, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P, India
| | - Shalini Gurumayum
- Chemical Biology Lab-I, Institute of Advanced Study in Science and Technology (IASST), Guwahati, Assam, India
| | - Sagar Ramrao Barge
- Chemical Biology Lab-I, Institute of Advanced Study in Science and Technology (IASST), Guwahati, Assam, India
| | - Devi Basumatary
- Chemical Biology Lab-I, Institute of Advanced Study in Science and Technology (IASST), Guwahati, Assam, India
| | - Jagat C Borah
- Chemical Biology Lab-I, Institute of Advanced Study in Science and Technology (IASST), Guwahati, Assam, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P, India.
| |
Collapse
|
36
|
Khapchaev AY, Vorotnikov AV, Antonova OA, Samsonov MV, Shestakova EA, Sklyanik IA, Tomilova AO, Shestakova MV, Shirinsky VP. Shear Stress and the AMP-Activated Protein Kinase Independently Protect the Vascular Endothelium from Palmitate Lipotoxicity. Biomedicines 2024; 12:339. [PMID: 38397940 PMCID: PMC10886486 DOI: 10.3390/biomedicines12020339] [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: 12/14/2023] [Revised: 01/21/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
Saturated free fatty acids are thought to play a critical role in metabolic disorders associated with obesity, insulin resistance, type 2 diabetes (T2D), and their vascular complications via effects on the vascular endothelium. The most abundant saturated free fatty acid, palmitate, exerts lipotoxic effects on the vascular endothelium, eventually leading to cell death. Shear stress activates the endothelial AMP-activated protein kinase (AMPK), a cellular energy sensor, and protects endothelial cells from lipotoxicity, however their relationship is uncertain. Here, we used isoform-specific shRNA-mediated silencing of AMPK to explore its involvement in the long-term protection of macrovascular human umbilical vein endothelial cells (HUVECs) against palmitate lipotoxicity and to relate it to the effects of shear stress. We demonstrated that it is the α1 catalytic subunit of AMPK that is critical for HUVEC protection under static conditions, whereas AMPK-α2 autocompensated a substantial loss of AMPK-α1, but failed to protect the cells from palmitate. Shear stress equally protected the wild type HUVECs and those lacking either α1, or α2, or both AMPK-α isoforms; however, the protective effect of AMPK reappeared after returning to static conditions. Moreover, in human adipose microvascular endothelial cells isolated from obese diabetic individuals, shear stress was a strong protector from palmitate lipotoxicity, thus highlighting the importance of circulation that is often obstructed in obesity/T2D. Altogether, these results indicate that AMPK is important for vascular endothelial cell protection against lipotoxicity in the static environment, however it may be dispensable for persistent and more effective protection exerted by shear stress.
Collapse
Affiliation(s)
- Asker Y. Khapchaev
- Institute of Experimental Cardiology Named after Academician V.N. Smirnov, National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Moscow 121552, Russia; (O.A.A.); (M.V.S.); (V.P.S.)
| | - Alexander V. Vorotnikov
- Institute of Experimental Cardiology Named after Academician V.N. Smirnov, National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Moscow 121552, Russia; (O.A.A.); (M.V.S.); (V.P.S.)
| | - Olga A. Antonova
- Institute of Experimental Cardiology Named after Academician V.N. Smirnov, National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Moscow 121552, Russia; (O.A.A.); (M.V.S.); (V.P.S.)
| | - Mikhail V. Samsonov
- Institute of Experimental Cardiology Named after Academician V.N. Smirnov, National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Moscow 121552, Russia; (O.A.A.); (M.V.S.); (V.P.S.)
| | - Ekaterina A. Shestakova
- Diabetes Institute, Endocrinology Research Center, Moscow 117036, Russia; (E.A.S.); (I.A.S.); (A.O.T.); (M.V.S.)
| | - Igor A. Sklyanik
- Diabetes Institute, Endocrinology Research Center, Moscow 117036, Russia; (E.A.S.); (I.A.S.); (A.O.T.); (M.V.S.)
| | - Alina O. Tomilova
- Diabetes Institute, Endocrinology Research Center, Moscow 117036, Russia; (E.A.S.); (I.A.S.); (A.O.T.); (M.V.S.)
| | - Marina V. Shestakova
- Diabetes Institute, Endocrinology Research Center, Moscow 117036, Russia; (E.A.S.); (I.A.S.); (A.O.T.); (M.V.S.)
| | - Vladimir P. Shirinsky
- Institute of Experimental Cardiology Named after Academician V.N. Smirnov, National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Moscow 121552, Russia; (O.A.A.); (M.V.S.); (V.P.S.)
| |
Collapse
|
37
|
Henin G, Loumaye A, Leclercq IA, Lanthier N. Myosteatosis: Diagnosis, pathophysiology and consequences in metabolic dysfunction-associated steatotic liver disease. JHEP Rep 2024; 6:100963. [PMID: 38322420 PMCID: PMC10844870 DOI: 10.1016/j.jhepr.2023.100963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 10/27/2023] [Accepted: 10/27/2023] [Indexed: 02/08/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is associated with an increased risk of multisystemic complications, including muscle changes such as sarcopenia and myosteatosis that can reciprocally affect liver function. We conducted a systematic review to highlight innovative assessment tools, pathophysiological mechanisms and metabolic consequences related to myosteatosis in MASLD, based on original articles screened from PUBMED, EMBASE and COCHRANE databases. Forty-six original manuscripts (14 pre-clinical and 32 clinical studies) were included. Microscopy (8/14) and tissue lipid extraction (8/14) are the two main assessment techniques used to measure muscle lipid content in pre-clinical studies. In clinical studies, imaging is the most used assessment tool and included CT (14/32), MRI (12/32) and ultrasound (4/32). Assessed muscles varied across studies but mainly included paravertebral (4/14 in pre-clinical; 13/32 in clinical studies) and lower limb muscles (10/14 in preclinical; 13/32 in clinical studies). Myosteatosis is already highly prevalent in non-cirrhotic stages of MASLD and correlates with disease activity when using muscle density assessed by CT. Numerous pathophysiological mechanisms were found and included: high-fat and high-fructose diet, dysregulation in fatty acid transport and ketogenesis, endocrine disorders and impaired microRNA122 pathway signalling. In this review we also uncover several potential consequences of myosteatosis in MASLD, such as insulin resistance, MASLD progression from steatosis to metabolic steatohepatitis and loss of muscle strength. In conclusion, data on myosteatosis in MASLD are already available. Screening for myosteatosis could be highly relevant in the context of MASLD, considering its correlation with MASLD activity as well as its related consequences.
Collapse
Affiliation(s)
- Guillaume Henin
- Service d’Hépato-Gastroentérologie, Cliniques universitaires Saint-Luc, UCLouvain, Brussels, Belgium
- Laboratory of Hepatogastroenterology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Audrey Loumaye
- Service d’Endocrinologie, Diabétologie et Nutrition, Cliniques universitaires Saint-Luc, UCLouvain, Brussels, Belgium
| | - Isabelle A. Leclercq
- Laboratory of Hepatogastroenterology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Nicolas Lanthier
- Service d’Hépato-Gastroentérologie, Cliniques universitaires Saint-Luc, UCLouvain, Brussels, Belgium
- Laboratory of Hepatogastroenterology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| |
Collapse
|
38
|
Ren QN, Huang DH, Zhang XN, Wang YN, Zhou YF, Zhang MY, Wang SC, Mai SJ, Wu DH, Wang HY. Two somatic mutations in the androgen receptor N-terminal domain are oncogenic drivers in hepatocellular carcinoma. Commun Biol 2024; 7:22. [PMID: 38182647 PMCID: PMC10770045 DOI: 10.1038/s42003-023-05704-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 12/13/2023] [Indexed: 01/07/2024] Open
Abstract
The androgen receptor (AR) plays an important role in male-dominant hepatocellular carcinoma, and specific acquired somatic mutations of AR have been observed in HCC patients. Our previous research have established the role of AR wild type as one of the key oncogenes in hepatocarcinogenesis. However, the role of hepatic acquired somatic mutations of AR remains unknown. In this study, we identify two crucial acquired somatic mutations, Q62L and E81Q, situated close to the N-terminal activation function domain-1 of AR. These mutations lead to constitutive activation of AR, both independently and synergistically with androgens, making them potent driver oncogene mutations. Mechanistically, these N-terminal AR somatic mutations enhance de novo lipogenesis by activating sterol regulatory element-binding protein-1 and promote glycogen accumulation through glycogen phosphorylase, brain form, thereby disrupting the AMPK pathway and contributing to tumorigenesis. Moreover, the AR mutations show sensitivity to the AMPK activator A769662. Overall, this study establishes the role of these N- terminal hepatic mutations of AR as highly malignant oncogenic drivers in hepatocarcinogenesis and highlights their potential as therapeutic targets for patients harboring these somatic mutations.
Collapse
Affiliation(s)
- Qian-Nan Ren
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong Province, China.
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong Province, 510060, China.
| | - Dan-Hui Huang
- Department of Respiratory and Critical Care Medicine, Chronic Airways Diseases Laboratory, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Xiao-Nan Zhang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
| | - Yue-Ning Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong Province, 510060, China
| | - Yu-Feng Zhou
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong Province, 510060, China
| | - Mei-Yin Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong Province, 510060, China
| | - Shuo-Cheng Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong Province, 510060, China
| | - Shi-Juan Mai
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong Province, 510060, China
| | - De-Hua Wu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong Province, China.
| | - Hui-Yun Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong Province, 510060, China.
| |
Collapse
|
39
|
Pinnell EF, Hostnik LD, Watts MR, Timko KJ, Thriffiley AA, Stover MR, Koenig LE, Gorman OM, Toribio RE, Burns TA. Effect of 5'-adenosine monophosphate-activated protein kinase agonists on insulin and glucose dynamics in experimentally induced insulin dysregulation in horses. J Vet Intern Med 2024; 38:102-110. [PMID: 38088223 PMCID: PMC10800176 DOI: 10.1111/jvim.16970] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 12/01/2023] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND 5'-adenosine monophosphate-activated protein kinase (AMPK) agonists, particularly resveratrol (RES), have not been extensively evaluated for their effect on insulin dysregulation (ID) in horses. OBJECTIVES Evaluate the effects of treatment with RES (10 mg/kg PO q12h), metformin (MET; 30 mg/kg PO q12h), and aspirin (ASP; 20 mg/kg PO q24h) on experimentally induced ID. ANIMALS Thirty-three healthy, adult, light-breed horses. METHODS Unblinded, placebo-controlled, experimental trial evaluating effects of AMPK agonists (RES, MET, and ASP) on experimentally induced ID. Horses were randomly assigned to a treatment group (RES, MET/ASP, RES/ASP, RES/MET/ASP, or placebo [CON]) after induction of ID with dexamethasone (0.08 mg/kg PO q24h for 7 days). Frequently sampled insulin-modified IV glucose tolerance tests (FSIGTT) and oral sugar tests (OST) were performed at baseline, 7 days after ID, and ID plus 7 days of treatment. Minimal model and OST variables were compared between (1-way ANOVA) and within (1-way ANOVA for repeated measures) groups over time to determine effects of treatment on ID. RESULTS Administration of dexamethasone for 14 days resulted in significantly altered insulin and glucose dynamics (SI, DI, basal [glucose], and [insulin]) and produced clinical signs of laminitis in 5 out of 33 (15%) of horses included in the study. Combination therapy with RES, MET, and ASP did not significantly improve insulin and glucose dynamics in horses with experimentally induced ID. CONCLUSIONS AND CLINICAL IMPORTANCE Metabolic testing before glucocorticoid administration should be considered in horses with clinical signs of metabolic syndrome.
Collapse
Affiliation(s)
- Erin F. Pinnell
- Department of Veterinary Clinical SciencesThe Ohio State University College of Veterinary MedicineColumbusOhioUSA
- Department of Veterinary Clinical SciencesWashington State University College of Veterinary MedicinePullmanWashingtonUSA
| | - Laura D. Hostnik
- Department of Veterinary Clinical SciencesThe Ohio State University College of Veterinary MedicineColumbusOhioUSA
| | - Mauria R. Watts
- Department of Veterinary Clinical SciencesThe Ohio State University College of Veterinary MedicineColumbusOhioUSA
| | - Kathryn J. Timko
- Department of Veterinary Clinical SciencesThe Ohio State University College of Veterinary MedicineColumbusOhioUSA
| | - Allison A. Thriffiley
- Department of Veterinary Clinical SciencesThe Ohio State University College of Veterinary MedicineColumbusOhioUSA
| | - Mercedes R. Stover
- Department of Veterinary Clinical SciencesThe Ohio State University College of Veterinary MedicineColumbusOhioUSA
| | - Lauren E. Koenig
- Department of Veterinary Clinical SciencesThe Ohio State University College of Veterinary MedicineColumbusOhioUSA
| | - Olivia M. Gorman
- Department of Veterinary Clinical SciencesThe Ohio State University College of Veterinary MedicineColumbusOhioUSA
| | - Ramiro E. Toribio
- Department of Veterinary Clinical SciencesThe Ohio State University College of Veterinary MedicineColumbusOhioUSA
| | - Teresa A. Burns
- Department of Veterinary Clinical SciencesThe Ohio State University College of Veterinary MedicineColumbusOhioUSA
| |
Collapse
|
40
|
Ahuja A, Zboinski E, das S, Zhu X, Ma Q, Xie Y, Tu Q, Chen J. Antidiabetic features of AdipoAI, a novel AdipoR agonist. Cell Biochem Funct 2024; 42:e3910. [PMID: 38269524 PMCID: PMC10811407 DOI: 10.1002/cbf.3910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 12/12/2023] [Accepted: 12/12/2023] [Indexed: 01/26/2024]
Abstract
Adiponectin is an antidiabetic endogenous adipokine that plays a protective role against the unfavorable metabolic sequelae of obesity. Recent evidence suggests a sinister link between hypoadiponectinemia and development of insulin resistance/type 2 diabetes (T2D). Adiponectin's insulin-sensitizing property is mediated through the specific adiponectin receptors R1 and R2, which activate the AMP-activated protein kinase (AMPK) and peroxisome proliferator-activated receptor (PPAR) α pathways. AdipoAI is a novel synthetic analogue of endogenous adiponectin with possibly similar pharmacological effects. Thus, there is a need of orally active small molecules that activate Adipoq subunits, and their downstream signaling, which could ameliorate obesity related type 2 diabetes. In the study we aim to investigate the effects of AdipoAI on obesity and T2D. Through in-vitro and in-vivo analyses, we investigated the antidiabetic potentials of AdipoAI and compared it with AdipoRON, another orally active adiponectin receptors agonist. Our results showed that in-vitro treatment of AdipoAI (0-5 µM) increased adiponectin receptor subunits AdipoR1/R2 with increase in AMPK and APPL1 protein expression in C2C12 myotubes. Similarly, in-vivo, oral administration of AdipoAI (25 mg/kg) observed similar effects as that of AdipoRON (50 mg/kg) with improved control of blood glucose and insulin sensitivity in diet-induced obesity (DIO) mice models. Further, AdipoAI significantly reduced epididymal fat content with decrease in inflammatory markers and increase in PPAR-α and AMPK levels and exhibited hepatoprotective effects in liver. Further, AdipoAI and AdipoRON also observed similar results in adipose tissue. Thus, our results suggest that low doses of orally active small molecule agonist of adiponectin AdipoAI can be a promising therapeutic target for obesity and T2D.
Collapse
Affiliation(s)
- Akash Ahuja
- Division of Oral Biology, Department of Periodontology, Tufts University School of Dental Medicine, Boston, Massachusetts, USA
| | - Elissa Zboinski
- Division of Oral Biology, Department of Periodontology, Tufts University School of Dental Medicine, Boston, Massachusetts, USA
| | - Siddhartha das
- Division of Oral Biology, Department of Periodontology, Tufts University School of Dental Medicine, Boston, Massachusetts, USA
| | - Xiaofang Zhu
- Division of Oral Biology, Department of Periodontology, Tufts University School of Dental Medicine, Boston, Massachusetts, USA
| | - Qian Ma
- Division of Oral Biology, Department of Periodontology, Tufts University School of Dental Medicine, Boston, Massachusetts, USA
- Department of General Dentistry, Affiliated Stomatological Hospital of Nanjing Medical University, Jiangsu, China
| | - Ying Xie
- Division of Oral Biology, Department of Periodontology, Tufts University School of Dental Medicine, Boston, Massachusetts, USA
- Department of Periodontology, Peking University School and Hospital of Stomatology, Beijing, China
| | - Qisheng Tu
- Division of Oral Biology, Department of Periodontology, Tufts University School of Dental Medicine, Boston, Massachusetts, USA
| | - Jake Chen
- Division of Oral Biology, Department of Periodontology, Tufts University School of Dental Medicine, Boston, Massachusetts, USA
- Dept. of Developmental, Molecular and Chemical Biology, Tufts School of Medicine; Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, USA
| |
Collapse
|
41
|
Azizian H, Farhadi Z, Bader M, Alizadeh Ghalenoei J, Ghafari MA, Mahmoodzadeh S. GPER activation attenuates cardiac dysfunction by upregulating the SIRT1/3-AMPK-UCP2 pathway in postmenopausal diabetic rats. PLoS One 2023; 18:e0293630. [PMID: 38134189 PMCID: PMC10745199 DOI: 10.1371/journal.pone.0293630] [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: 07/11/2023] [Accepted: 10/16/2023] [Indexed: 12/24/2023] Open
Abstract
Postmenopausal diabetic women are at higher risk to develop cardiovascular diseases (CVD) compared with nondiabetic women. Alterations in cardiac cellular metabolism caused by changes in sirtuins are one of the main causes of CVD in postmenopausal diabetic women. Several studies have demonstrated the beneficial actions of the G protein-coupled estrogen receptor (GPER) in postmenopausal diabetic CVD. However, the molecular mechanisms by which GPER has a cardioprotective effect are still not well understood. In this study, we used an ovariectomized (OVX) type-two diabetic (T2D) rat model induced by high-fat diet/streptozotocin to investigate the effect of G-1 (GPER-agonist) on sirtuins, and their downstream pathways involved in regulation of cardiac metabolism and function. Animals were divided into five groups: Sham-Control, T2D, OVX+T2D, OVX+T2D+Vehicle, and OVX+T2D+G-1. G-1 was administrated for six weeks. At the end, hemodynamic factors were measured, and protein levels of sirtuins, AMP-activated protein kinase (AMPK), and uncoupling protein 2 (UCP2) were determined by Western blot analysis. In addition, cardiac levels of oxidative stress biomarkers were measured. The findings showed that T2D led to left ventricular dysfunction and signs of oxidative stress in the myocardium, which were accompanied by decreased protein levels of Sirt1/2/3/6, p-AMPK, and UCP2 in the heart. Moreover, the induction of the menopausal state exacerbated these changes. In contrast, treatment with G-1 ameliorated the hemodynamic changes associated with ovariectomy by increasing Sirt1/3, p-AMPK, UCP2, and improving oxidative status. The results provide evidence of the cardioprotective effects of GPER operating through Sirt1/3, p-AMPK, and UCP2, thereby improving cardiac function. Our results suggest that increasing Sirt1/3 levels may offer new therapeutic approaches for postmenopausal diabetic CVD.
Collapse
Affiliation(s)
- Hossein Azizian
- Yazd Neuroendocrine Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Zeinab Farhadi
- Yazd Neuroendocrine Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Michael Bader
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu Berlin, Berlin, Germany
- University of Lübeck, Institute for Biology, Lübeck, Germany
| | - Jalil Alizadeh Ghalenoei
- Yazd Neuroendocrine Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Mohammad Amin Ghafari
- Yazd Neuroendocrine Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Shokoufeh Mahmoodzadeh
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| |
Collapse
|
42
|
Mishra T, Gupta S, Rai P, Khandelwal N, Chourasiya M, Kushwaha V, Singh A, Varshney S, Gaikwad AN, Narender T. Anti-adipogenic action of a novel oxazole derivative through activation of AMPK pathway. Eur J Med Chem 2023; 262:115895. [PMID: 37883898 DOI: 10.1016/j.ejmech.2023.115895] [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: 06/20/2023] [Revised: 10/09/2023] [Accepted: 10/19/2023] [Indexed: 10/28/2023]
Abstract
Obesity is a chronic disorder with multifactorial etiology, including genetic, medical, dietary and other environmental factors. Both natural and synthetic heterocyclic compounds, especially oxazoles, represent an interesting group of compounds and have gained much attention due to their remarkable biological activities. Therefore, a library of 3,3-DMAH (3,3-dimethylallylhalfordinol) inspired N-alkylated oxazole bromide salts with varied substitutions were prepared and screened using the 3T3-L1 model of adipogenesis and HFD-induced obesity model in Syrian golden hamsters. Several compounds in the synthesized series displayed remarkable anti-adipogenic potential on the differentiation of 3T3-L1 preadipocytes. Compound 19e, displayed the most potent activity of all and selected for further studies. Compound 19e inhibited mitotic clonal expansion of 3T3-L1 cells and enhanced the mitochondrial oxygen consumption rate of the cells during early phase of differentiation via AMPK activation. 19e also improved the dyslipidaemia in high calorie diet fed Syrian Golden Hamsters. Therefore, compound 19e can serve as a potential lead against adipogenesis and dyslipidaemia models and could be further investigated to affirm its significance as a drug candidate.
Collapse
Affiliation(s)
- Tripti Mishra
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Lucknow, U.P., 226031, India
| | - Sanchita Gupta
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, U.P., 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Prashant Rai
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, U.P., 226031, India
| | - Nilesh Khandelwal
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, U.P., 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Mohit Chourasiya
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Lucknow, U.P., 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Vinita Kushwaha
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, U.P., 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Astha Singh
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, U.P., 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Salil Varshney
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, U.P., 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Anil Nilkanth Gaikwad
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, U.P., 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| | - Tadigoppula Narender
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Lucknow, U.P., 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
43
|
Zhang Q, He CX, Wang LY, Qian D, Tang DD, Jiang SN, Chen WW, Wu CJ, Peng W. Hydroxy-α-sanshool from the fruits of Zanthoxylum bungeanum Maxim. promotes browning of white fat by activating TRPV1 to induce PPAR-γ deacetylation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 121:155113. [PMID: 37748388 DOI: 10.1016/j.phymed.2023.155113] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 09/05/2023] [Accepted: 09/20/2023] [Indexed: 09/27/2023]
Abstract
BACKGROUND Accumulating evidence suggested increasing energy expenditure is a feasible strategy for combating obesity, and browning of white adipose tissue (WAT) to promote thermogenesis might be one of the attractive ways. Hydroxy-α-sanshool (HAS), a natural amide alkaloid extracted from the fruits of Zanthoxylum bungeanum Maxim, possesses lots of benefits in lipid metabolism regulation. METHODS The anti-obesity effect of HAS was investigated by establishing an animal model of obesity and a 3T3-L1 differentiation cell model. Effects of HAS on the whole-body fat and liver of obese mice, and the role of HAS in inducing browning of white fat were studied by Micro CT, Metabolic cage detection, Cell mitochondrial pressure detection, transmission electron microscopy and cold exposure assays. Furthermore, the Real-time PCR (qPCR), digital PCR (dPCR), western blot, Co-immunoprecipitation (Co-IP), molecular docking, drug affinity responsive target stability (DARTS), Cellular thermal shift assay (CETSA) and other methods were used to investigate the target and mechanisms of HAS. RESULTS We found that treatment with HAS helped mice combat obesity caused by a high fat diet (HFD) and improve metabolic characteristics. In addition, our results suggested that the anti-obesity effect of HAS is related to increase energy consumption and thermogenesis via induction of browning of WAT. The further investigations uncovered that HAS can up-regulate UCP-1 expression, increase mitochondria number, and elevate the cellular oxygen consumption rates (OCRs) of white adipocytes. Importantly, the results indicated that browning effects of HAS is closely associated with SIRT1-dependent PPAR-γ deacetylation through activating the TRPV1/AMPK pathway, and TRPV1 is the potential drug target of HAS for the browning effects of WAT. CONCLUSIONS Our results suggested the HAS can promote browning of WAT via regulating AMPK/SIRT-1/PPARγ signaling, and the potential drug target of HAS is the membrane receptor of TRPV1.
Collapse
Affiliation(s)
- Qing Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, PR China; Innovative Institute of Chinese Medicine and Pharmacy/Academy for Interdiscipline, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Cheng-Xun He
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, PR China
| | - Ling-Yu Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, PR China
| | - Die Qian
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, PR China
| | - Dan-Dan Tang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, PR China
| | - Sheng-Nan Jiang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, PR China
| | - Wen-Wen Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, PR China
| | - Chun-Jie Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, PR China; Innovative Institute of Chinese Medicine and Pharmacy/Academy for Interdiscipline, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China.
| | - Wei Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, PR China.
| |
Collapse
|
44
|
Chen Y, Li Q, Zhao S, Sun L, Yin Z, Wang X, Li X, Iwakiri Y, Han J, Duan Y. Berberine protects mice against type 2 diabetes by promoting PPARγ-FGF21-GLUT2-regulated insulin sensitivity and glucose/lipid homeostasis. Biochem Pharmacol 2023; 218:115928. [PMID: 37979703 DOI: 10.1016/j.bcp.2023.115928] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/13/2023] [Accepted: 11/15/2023] [Indexed: 11/20/2023]
Abstract
Type 2 diabetes (T2D) is a chronic, burdensome disease that is characterized by disordered insulin sensitivity and disturbed glucose/lipid homeostasis. Berberine (BBR) has multiple therapeutic actions on T2D, including regulation of glucose and lipid metabolism, improvement of insulin sensitivity and energy expenditure. Recently, the function of BBR on fibroblast growth factor 21 (FGF21) has been identified. However, if BBR ameliorates T2D through FGF21, the underlying mechanisms remain unknown. Herein, we used T2D wild type (WT) and FGF21 global knockout (FKO) mice [mouse T2D model: established by high-fat diet (HFD) feeding plus streptozotocin (STZ) injection], and hepatocyte-specific peroxisome proliferator activated receptor γ (PPARγ) deficient (PPARγHepKO) mice, and cultured human liver carcinoma cells line, HepG2 cells, to characterize the role of BBR in glucose/lipid metabolism and insulin sensitivity. We found that BBR activated FGF21 expression by up-regulating PPARγ expression at the cellular level. Meanwhile, BBR ameliorated glucosamine hydrochloride (Glcn)-induced insulin resistance and increased glucose transporter 2 (GLUT2) expression in a PPARγ/FGF21-dependent manner. In T2D mice, BBR up-regulated the expression of PPARγ, FGF21 and GLUT2 in the liver, and GLUT2 in the pancreas. BBR also reversed T2D-induced insulin resistance, liver lipid accumulation, and damage in liver and pancreas. However, FGF21 deficiency diminished these effects of BBR on diabetic mice. Altogether, our study demonstrates that the therapeutic effects of BBR on T2D were partly accomplished by activating PPARγ-FGF21-GLUT2 signaling pathway. The discovery of this new pathway provides a deeper understanding of the mechanism of BBR for T2D treatment.
Collapse
Affiliation(s)
- Yi Chen
- Department of Cell Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, China
| | - Qi Li
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Shiwei Zhao
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Lei Sun
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Zequn Yin
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Xiaolin Wang
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Xiaoju Li
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Yasuko Iwakiri
- Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT, USA
| | - Jihong Han
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China; Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China.
| | - Yajun Duan
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China.
| |
Collapse
|
45
|
Zhang LH, Wang J, Tan BH, Yin YB, Kang YM. Roux-en-Y Gastric Bypass Improves Insulin Sensitivity in Obese Rats with Type 2 Diabetes Mellitus by Regulating the Grin3a/AMPK Signal Axis in Hypothalamic Arcuate Nucleus. Diabetes Metab Syndr Obes 2023; 16:3617-3629. [PMID: 38028990 PMCID: PMC10644885 DOI: 10.2147/dmso.s430445] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/21/2023] [Indexed: 12/01/2023] Open
Abstract
Objective The objective of this study was to explore the effects and related mechanisms of Roux-en-Y gastric bypass (RYGB) on insulin sensitivity in obese rats with type 2 diabetes mellitus (T2DM). Methods The obese T2DM rat model was constructed by feeding a high-fat diet and injecting streptozotocin (STZ), and treated with RYGB. Grin3a shRNA was injected into the bilateral hypothalamic arcuate nucleus (ARC) to knockdown the Grin3a expression on T2DM rats. Eight weeks after operation, the body weight, fasting blood glucose (FBG), fasting serum insulin (FSI), homeostatic model assessment of insulin resistance (HOMA-IR), and plasma triglyceride (TG) levels were assessed. Hematoxylin & eosin (H&E) staining was adopted to observe the white adipose tissue (WAT) of rats. Western blot and qRT-PCR were used to detect the expression of Grin3a, adenosine 5' monophosphate-activated protein kinase (AMPK) and p-AMPK in ARC of rats. Later, the plasmid over-expressing or knocking down Grin3a was transfected into differentiated 3T3-L1 adipocytes, and the TG level and the formation of lipid droplets in adipocyte were assessed by TG kit and oil red O staining. The expression of lipogenic transcription factors in cells was detected by qRT-PCR. Results RYGB reduced FBG, FSI, HOMA-IR and plasma TG levels in T2DM rats while increasing Grin3a expression and p-AMPK/AMPK ratio in ARC. Knockdown of Grin3a not only reversed the decrease of FBG, FSI, HOMA-IR and plasma TG levels in T2DM rats induced by RYGB, but also reversed the up-regulation of p-AMPK/AMPK ratio in ARC affected by RYGB. Moreover, knocking down Grin3a significantly increased the TG level, promoted the formation of lipid droplets and up-regulated the expressions of lipogenic transcription factors in adipocytes. Conclusion RYGB improved the insulin sensitivity, reduced the plasma TG level and lessens the fat accumulation in obese T2DM rats by regulating the Grin3a/AMPK signal in ARC.
Collapse
Affiliation(s)
- Li-Hai Zhang
- Department of Physiology and Pathophysiology, Xi’an Jiaotong University School of Basic Medical Sciences, Xi’an, People’s Republic of China
- Six Wards of General Surgery, The First Affiliated Hospital of Jiamusi University, Jiamusi, People’s Republic of China
| | - Jiao Wang
- Department of Physiology and Pathophysiology, Xi’an Jiaotong University School of Basic Medical Sciences, Xi’an, People’s Republic of China
- Six Wards of General Surgery, The First Affiliated Hospital of Jiamusi University, Jiamusi, People’s Republic of China
| | - Bai-Hong Tan
- Six Wards of General Surgery, The First Affiliated Hospital of Jiamusi University, Jiamusi, People’s Republic of China
| | - Yan-Bin Yin
- Six Wards of General Surgery, The First Affiliated Hospital of Jiamusi University, Jiamusi, People’s Republic of China
| | - Yu-Ming Kang
- Department of Physiology and Pathophysiology, Xi’an Jiaotong University School of Basic Medical Sciences, Xi’an, People’s Republic of China
| |
Collapse
|
46
|
Valentini A, Cardillo C, Della Morte D, Tesauro M. The Role of Perivascular Adipose Tissue in the Pathogenesis of Endothelial Dysfunction in Cardiovascular Diseases and Type 2 Diabetes Mellitus. Biomedicines 2023; 11:3006. [PMID: 38002006 PMCID: PMC10669084 DOI: 10.3390/biomedicines11113006] [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: 09/25/2023] [Revised: 10/27/2023] [Accepted: 11/05/2023] [Indexed: 11/26/2023] Open
Abstract
Cardiovascular diseases (CVDs) and type 2 diabetes mellitus (T2DM) are two of the four major chronic non-communicable diseases (NCDs) representing the leading cause of death worldwide. Several studies demonstrate that endothelial dysfunction (ED) plays a central role in the pathogenesis of these chronic diseases. Although it is well known that systemic chronic inflammation and oxidative stress are primarily involved in the development of ED, recent studies have shown that perivascular adipose tissue (PVAT) is implicated in its pathogenesis, also contributing to the progression of atherosclerosis and to insulin resistance (IR). In this review, we describe the relationship between PVAT and ED, and we also analyse the role of PVAT in the pathogenesis of CVDs and T2DM, further assessing its potential therapeutic target with the aim of restoring normal ED and reducing global cardiovascular risk.
Collapse
Affiliation(s)
- Alessia Valentini
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (A.V.); (D.D.M.)
| | - Carmine Cardillo
- Department of Aging, Policlinico A. Gemelli IRCCS, 00168 Roma, Italy;
- Department of Translational Medicine and Surgery, Catholic University, 00168 Rome, Italy
| | - David Della Morte
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (A.V.); (D.D.M.)
| | - Manfredi Tesauro
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (A.V.); (D.D.M.)
| |
Collapse
|
47
|
Kaviani F, Razavi BM, Mohsenzadeh MS, Rameshrad M, Hosseinzadeh H. Thymoquinone attenuates olanzapine-induced metabolic disorders in rats. Mol Biol Rep 2023; 50:8925-8935. [PMID: 37707771 DOI: 10.1007/s11033-023-08726-5] [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: 05/15/2023] [Accepted: 07/31/2023] [Indexed: 09/15/2023]
Abstract
BACKGROUND Olanzapine (OLZ) is an atypical antipsychotic agent for psychotic disorders. Evidence has shown that OLZ is related to metabolic side effects, including obesity, hypertension, and insulin resistance. Thymoquinone (TQ) is the principal bioactive component of Nigella sativa. Several studies have been conducted to investigate the effectiveness of TQ in alleviating metabolic abnormalities. In the current research work, the protective effects of TQ on metabolic disorders induced by OLZ and possible underlying mechanisms were investigated. METHODS AND RESULTS Wistar rats were exposed to TQ alone (10 mg/kg), OLZ (5 mg/kg), or OLZ plus TQ (2.5, 5, or 10 mg/kg) given daily by intraperitoneal injection. After the treatment, variations in body weight, food intake, systolic blood pressure, serum leptin, biochemical factors, liver malondialdehyde (MDA), and glutathione (GSH) content were evaluated. Protein expression of AMPK in the liver was also measured by a western blotting test. OLZ increased body weight, food intake, MDA levels, and blood pressure. OLZ also elevated glucose, triglyceride, low-density lipoprotein cholesterol, and leptin serum levels. It decreased GSH. In the western blot, decreased AMPK protein level was obtained. These changes were attenuated by TQ co-administration. CONCLUSIONS The present study demonstrates the effectiveness of TQ on OLZ-induced metabolic abnormalities related to its antioxidant activity and regulation of glucose homeostasis and lipid metabolism.
Collapse
Affiliation(s)
- Farima Kaviani
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bibi Marjan Razavi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Targeted Drug Delivery Research Center, Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahdieh Sadat Mohsenzadeh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Food Control Laboratory, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Rameshrad
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Hosseinzadeh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
48
|
Kim J, Lee SK, Jeong SY, You H, Han SD, Park S, Kim S, Kim TM. Multifaceted action of stem cell-derived extracellular vesicles for nonalcoholic steatohepatitis. J Control Release 2023; 364:S0168-3659(23)00706-X. [PMID: 39491172 DOI: 10.1016/j.jconrel.2023.10.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 10/24/2023] [Accepted: 10/27/2023] [Indexed: 11/05/2024]
Abstract
Nonalcoholic steatohepatitis (NASH) is a chronic liver disease associated with metabolic syndrome. Extracellular vesicles (EVs) are essential signaling mediators containing functional biomolecules. EVs are secreted from various cell types, and recent studies have shown that mesenchymal stem cell-derived EVs have therapeutic potential against immune and metabolic diseases. In this study, we investigated whether EVs from induced mesenchymal stem cells (iMSC-EVs) regulate AMPK signaling and lipid metabolism using cell-based studies and two different mouse models of NASH (methionine/choline-deficient diet-induced and ob/ob mice). Protein analysis revealed that iMSC-EVs carry cargo proteins with the potential to regulate lipid metabolism. iMSC-EVs inhibited free fatty acid release from adipose tissues by downregulating the activity of lipolytic genes in NASH. In addition, iMSC-EVs improved hepatic steatosis by modulating AMPK signaling, which plays essential role in metabolic homeostasis in the liver. Moreover, iMSC-EVs reduced CD36 expression, contributing to the blockade of free fatty acid transport to the liver of NASH mice. Finally, iMSC-EVs reduced inflammation, endoplasmic reticulum stress, and apoptosis while promoting hepatic regeneration of the NASH liver. In conclusion, iMSC-EVs can potentially serve as cell-free therapeutics for NASH owing to their multifaceted modality.
Collapse
Affiliation(s)
- Jimin Kim
- Brexogen Research Center, Brexogen Inc., Songpa-Gu, Seoul 05855, South Korea
| | - Seul Ki Lee
- Brexogen Research Center, Brexogen Inc., Songpa-Gu, Seoul 05855, South Korea
| | - Seon-Yeong Jeong
- Brexogen Research Center, Brexogen Inc., Songpa-Gu, Seoul 05855, South Korea
| | - Haedeun You
- Brexogen Research Center, Brexogen Inc., Songpa-Gu, Seoul 05855, South Korea
| | - Sang-Deok Han
- Brexogen Research Center, Brexogen Inc., Songpa-Gu, Seoul 05855, South Korea
| | - Somi Park
- Brexogen Research Center, Brexogen Inc., Songpa-Gu, Seoul 05855, South Korea
| | - Soo Kim
- Brexogen Research Center, Brexogen Inc., Songpa-Gu, Seoul 05855, South Korea
| | - Tae Min Kim
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang, Gangwon-do 25354, South Korea; Institutes of Green-Bio Science and Technology, Seoul National University, Pyeongchang, Gangwon-do 25354, South Korea.
| |
Collapse
|
49
|
Liang C, Gao S, Gao J, Xu Y, Li Q. Comparison of effects of HucMSCs, exosomes, and conditioned medium on NASH. Sci Rep 2023; 13:18431. [PMID: 37891247 PMCID: PMC10611740 DOI: 10.1038/s41598-023-45828-3] [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: 06/13/2023] [Accepted: 10/24/2023] [Indexed: 10/29/2023] Open
Abstract
To investigate the effects and potential mechanisms of human umbilical cord mesenchymal stem cells, exosomes, and their conditioned media on lipid storage in oleic acid (OA) and palmitic acid (PA) treated hepatocytes and high-fat methionine- choline deficient diet (HFMRCD) induced non-alcoholic steatohepatitis (NASH) mice. AML12 cells were stimulated with OA and PA to establish the lipid storage cell model. HucMSCs, exosomes, and culture medium were then co-cultured. At the same time, C57BL/6 mice were fed an HFMRCD for 6 or 8 weeks to establish a NASH mouse model. The effect of HucMSCs, exosomes, and culture medium on lipid droplet repair of hepatocytes or NASH mice was then assessed. The weight of hepatocytes or liver tissue, Oil Red O, hematoxylin-eosin staining, Masson staining, Western blot, and qPCR were used to detect the related IL-6, TNF-α, TGF-β1 andEI24/AMPK/mTOR pathway expression in hepatocytes and liver tissue. Compared with the model group, the effect of HucMSCs-Ex on inhibiting the accumulation of lipid droplets was more obvious at the cell level. In vivo study showed that HucMSCs-Ex reduces activity scores in NASH mice and improves liver tissue morphology by reducing vacuolar degeneration, fat deposition, and collagen deposition of liver tissue. Western blot and qPCR results showed that inflammatory factors and AMPK/mTOR or EI24-related autophagy pathways were altered before and after treatment. HucMSCs, HucMSC-Ex, and CM can promote autophagy in hepatocytes or NASH mice through the AMPK/mTOR or EI24-related autophagy pathway and alleviate injury associated with lipid deposition, collagen deposition or inflammation, reversing the progression of NASH.
Collapse
Affiliation(s)
- Chenchen Liang
- School of Public Health, Dali University, Dali, 671013, Yunnan, China
| | - Siyuan Gao
- Center of Liver Diseases, The Third People's Hospital of Kunming, Kunming, 650041, Yunnan, China
| | - Jianpeng Gao
- Department of Administration, Kunming Yan'an Hospital, Kunming, 650051, Yunnan, China.
| | - Yanwen Xu
- School of Public Health, Dali University, Dali, 671013, Yunnan, China
| | - Qilong Li
- School of Public Health, Dali University, Dali, 671013, Yunnan, China
| |
Collapse
|
50
|
Zhou X, Zhang J, Sun Y, Shen J, Sun B, Ma Q. Glutamine Ameliorates Liver Steatosis via Regulation of Glycolipid Metabolism and Gut Microbiota in High-Fat Diet-Induced Obese Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:15656-15667. [PMID: 37847053 DOI: 10.1021/acs.jafc.3c05566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2023]
Abstract
Obesity and its associated conditions, such as nonalcoholic fatty liver disease (NAFLD), are risk factors for health. The aim of this study was to explore the effects of glutamine (Gln) on liver steatosis induced by a high-fat diet (HFD) and HEPG2 cells induced by oleic acid. Gln demonstrated a positive influence on hepatic homeostasis by suppressing acetyl CoA carboxylase (ACC) and fatty acid synthase (FAS) and promoting sirtuin 1 (SIRT1) expression while improving glucose metabolism by regulating serine/threonine protein kinase (AKT)/factor forkhead box O1 (FOXO1) signals in vivo and in vitro. Obese Gln-fed mice had higher colonic short-chain fatty acid (SCFA) contents and lower inflammation factor protein levels in the liver, HEPG2 cells, and jejunum. Gln-treated obese mice had an effective decrease in Firmicutes abundance. These findings indicate that Gln serves as a nutritional tool in managing obesity and related disorders.
Collapse
Affiliation(s)
- Xinbo Zhou
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Junjie Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Yutong Sun
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Jian Shen
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Bo Sun
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Qingquan Ma
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|