1
|
Ashraf MS, Tuli K, Moiz S, Sharma SK, Sharma D, Adnan M. AMP kinase: A promising therapeutic drug target for post-COVID-19 complications. Life Sci 2024; 359:123202. [PMID: 39489398 DOI: 10.1016/j.lfs.2024.123202] [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: 06/14/2024] [Revised: 10/27/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024]
Abstract
The COVID-19 pandemic, caused by SARS-CoV-2, has resulted in severe respiratory issues and persistent complications, particularly affecting glucose metabolism. Patients with or without pre-existing diabetes often experience worsened symptoms, highlighting the need for innovative therapeutic approaches. AMPK, a crucial regulator of cellular energy balance, plays a pivotal role in glucose metabolism, insulin sensitivity, and inflammatory responses. AMPK activation, through allosteric or kinase-dependent mechanisms, impacts cellular processes like glucose uptake, fatty acid oxidation, and autophagy. The tissue-specific distribution of AMPK emphasizes its role in maintaining metabolic homeostasis throughout the body. Intriguingly, SARS-CoV-2 infection inhibits AMPK, contributing to metabolic dysregulation and post-COVID-19 complications. AMPK activators like capsaicinoids, curcumin, phytoestrogens, cilostazol, and momordicosides have demonstrated the potential to regulate AMPK activity. Compounds from various sources improve fatty acid oxidation and insulin sensitivity, with metformin showing opposing effects on AMPK activation compared to the virus, suggesting potential therapeutic options. The diverse effects of AMPK activation extend to its role in countering viral infections, further highlighting its significance in COVID-19. This review explores AMPK activation mechanisms, its role in metabolic disorders, and the potential use of natural compounds to target AMPK for post-COVID-19 complications. Also, it aims to review the possible methods of activating AMPK to prevent post-COVID-19 diabetes and cardiovascular complications. It also explores the use of natural compounds for their therapeutic effects in targeting the AMPK pathways. Targeting AMPK activation emerges as a promising avenue to mitigate the long-term effects of COVID-19, offering hope for improved patient outcomes and a better quality of life.
Collapse
Affiliation(s)
- Mohammad Saquib Ashraf
- Department of Medical Laboratory Science College of Pharmacy, Nursing and Medical Science Riyadh ELM University, Riyadh, P.O. Box 12734, Saudi Arabia.
| | - Kanika Tuli
- Guru Nanak Institute of Pharmacy, Dalewal, Hoshiarpur 144208, Punjab, India
| | - Shadman Moiz
- Department of Biotechnology, Lalit Narayan Mithila University, Darbhanga 846004, Bihar, India
| | - Satish Kumar Sharma
- Department of Pharmacology, Glocal School of Pharmacy, The Glocal University, Saharanpur, India
| | - Deepa Sharma
- UMM Matrix Innovations Private Limited, Delhi 110044, India
| | - Mohd Adnan
- Department of Biology, College of Science, University of Ha'il, Ha'il, P.O. Box 2440, Saudi Arabia; Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 602105, India
| |
Collapse
|
2
|
Park JE, Han JS. Ferulic acid improves palmitate-induced insulin resistance by regulating IRS-1/Akt and AMPK pathways in L6 skeletal muscle cells. Toxicol Res (Camb) 2024; 13:tfae197. [PMID: 39664501 PMCID: PMC11630505 DOI: 10.1093/toxres/tfae197] [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: 07/03/2024] [Revised: 11/04/2024] [Accepted: 11/17/2024] [Indexed: 12/13/2024] Open
Abstract
Objective Increased plasma-free fatty acid (FFA) induced by obesity can trigger insulin resistance and it is a significantly dangerous constituent in the progression of diabetes. Although ferulic acid has various physiological functions, no studies have examined ferulic acid's effects on insulin-resistant muscle cells. This study investigated the effect of ferulic acid on improving palmitic acid-induced insulin resistance in L6 skeletal muscle cells. Methods Palmitic acid induces insulin resistance by inhibiting the phosphorylation of IRS-1tyr and stimulating the phosphorylation of IRS-1ser in diabetes. Thus, palmitic acid (0.75 mM) was used as an insulin resistance inducer and ferulic acid was treated at various concentrations (2, 5, 10, and 20 uM) in L6 skeletal muscle cells. Results Palmitic acid significantly reduced the cell viability of L6 skeletal muscle cells, whereas ferulic acid treatment significantly increased cell viability in a concentration-dependent manner. Palmitic acid significantly reduced glucose uptake due to insulin resistance in the muscle cells; however, ferulic acid treatment remarkably increased glucose uptake. Ferulic acid promoted the phosphorylation of IRS-1tyr that palmitic acid inhibited, while also suppressing the palmitic acid-induced phosphorylation of IRS-1ser. Ferulic acid activated PI3K and then stimulated the phosphorylation of Akt, which increased PM-GLUT4 expression, thereby stimulating glucose uptake into insulin-resistant muscle cells. Ferulic acid also increased glycogen synthesis by phosphorylating GSK3β via the Akt pathway. Additionally, ferulic acid significantly promoted phosphorylation of AMPK, enhancing PM-GLUT4 levels and glucose uptake. Conclusions These results suggest that ferulic acid may improve palmitate-induced insulin resistance by regulating IRS-1/ Akt and the AMPK pathway in L6 skeletal muscle cells.
Collapse
Affiliation(s)
- Jae Eun Park
- Department of Food Science and Nutrition, Pusan National University, 2, Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan, 46241, Republic of Korea
| | - Ji Sook Han
- Department of Food Science and Nutrition, Pusan National University, 2, Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan, 46241, Republic of Korea
| |
Collapse
|
3
|
Mendez DA, Soñanez-Organis JG, Yang X, Vazquez-Anaya G, Nishiyama A, Ortiz RM. Exogenous thyroxine increases cardiac GLUT4 translocation in insulin resistant OLETF rats. Mol Cell Endocrinol 2024; 590:112254. [PMID: 38677465 DOI: 10.1016/j.mce.2024.112254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 04/16/2024] [Accepted: 04/23/2024] [Indexed: 04/29/2024]
Abstract
During insulin resistance, the heart undergoes a metabolic shift in which fatty acids (FA) account for roughly about 99% of the ATP production. This metabolic shift is indicative of impaired glucose metabolism. A shift in FA metabolism with impaired glucose tolerance can increase reactive oxygen species (ROS), lipotoxicity, and mitochondrial dysfunction, ultimately leading to cardiomyopathy. Thyroid hormones (TH) may improve the glucose intolerance by increasing glucose reabsorption and metabolism in peripheral tissues, but little is known on its effects on cardiac tissue during insulin resistance. In the present study, insulin resistant Otsuka Long Evans Tokushima Fatty (OLETF) rats were used to assess the effects of exogenous thyroxine (T4) on glucose metabolism in cardiac tissue. Rats were assigned to four groups: (1) lean, Long Evans Tokushima Otsuka (LETO; n=6), (2) LETO + T4 (8 μg/100 g BM/d × 5 wks; n = 7), (3) untreated OLETF (n = 6), and (4) OLETF + T4 (8 μg/100 g BM/d × 5 wks; n = 7). T4 increased GLUT4 gene expression by 85% in OLETF and increased GLUT4 protein translocation to the membrane by 294%. Additionally, T4 increased p-AS160 by 285%, phosphofructokinase-1 (PFK-1) mRNA, the rate limiting step in glycolysis, by 98% and hexokinase II by 64% in OLETF. T4 decreased both CPT2 mRNA and protein expression in OLETF. The results suggest that exogenous T4 has the potential to increase glucose uptake and metabolism while simultaneously reducing fatty acid transport in the heart of insulin resistant rats. Thus, L-thyroxine may have therapeutic value to help correct the impaired substrate metabolism associated with diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Dora A Mendez
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, CA, USA.
| | - José G Soñanez-Organis
- Division of Science and Engineering, Department of Chemical Biological and Agropecuary Sciences, University of Sonora, Navojoa, SON, Mexico
| | - Xue Yang
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, CA, USA
| | - Guillermo Vazquez-Anaya
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, CA, USA
| | - Akira Nishiyama
- Department of Pharmacology, Kagawa University Medical School, Kagawa, Japan
| | - Rudy M Ortiz
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, CA, USA
| |
Collapse
|
4
|
Margolis LM, Wilson MA, Drummer DJ, Carrigan CT, Murphy NE, Allen JT, Dawson MA, Mantzoros CS, Young AJ, Pasiakos SM. Pioglitazone does not enhance exogenous glucose oxidation or metabolic clearance rate during aerobic exercise in men under acute high-altitude exposure. Am J Physiol Regul Integr Comp Physiol 2024; 327:R25-R34. [PMID: 38682243 PMCID: PMC11381008 DOI: 10.1152/ajpregu.00064.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 04/09/2024] [Accepted: 04/23/2024] [Indexed: 05/01/2024]
Abstract
Insulin insensitivity decreases exogenous glucose oxidation and metabolic clearance rate (MCR) during aerobic exercise in unacclimatized lowlanders at high altitude (HA). Whether use of an oral insulin sensitizer before acute HA exposure enhances exogenous glucose oxidation is unclear. This study investigated the impact of pioglitazone (PIO) on exogenous glucose oxidation and glucose turnover compared with placebo (PLA) during aerobic exercise at HA. With the use of a randomized crossover design, native lowlanders (n = 7 males, means ± SD, age: 23 ± 6 yr, body mass: 84 ± 11 kg) consumed 145 g (1.8 g/min) of glucose while performing 80 min of steady-state (1.43 ± 0.16 V̇o2 L/min) treadmill exercise at HA (460 mmHg; [Formula: see text] 96.6 mmHg) following short-term (5 days) use of PIO (15 mg oral dose per day) or PLA (microcrystalline cellulose pill). Substrate oxidation and glucose turnover were determined using indirect calorimetry and stable isotopes ([13C]glucose and 6,6-[2H2]glucose). Exogenous glucose oxidation was not different between PIO (0.31 ± 0.03 g/min) and PLA (0.32 ± 0.09 g/min). Total carbohydrate oxidation (PIO: 1.65 ± 0.22 g/min, PLA: 1.68 ± 0.32 g/min) or fat oxidation (PIO: 0.10 ± 0.0.08 g/min, PLA: 0.09 ± 0.07 g/min) was not different between treatments. There was no treatment effect on glucose rate of appearance (PIO: 2.46 ± 0.27, PLA: 2.43 ± 0.27 mg/kg/min), disappearance (PIO: 2.19 ± 0.17, PLA: 2.20 ± 0.22 mg/kg/min), or MCR (PIO: 1.63 ± 0.37, PLA: 1.73 ± 0.40 mL/kg/min). Results from this study indicate that PIO is not an effective intervention to enhance exogenous glucose oxidation or MCR during acute HA exposure. Lack of effect with PIO suggests that the etiology of glucose metabolism dysregulation during acute HA exposure may not result from insulin resistance in peripheral tissues.NEW & NOTEWORTHY Short-term (5 days) use of the oral insulin sensitizer pioglitazone does not alter circulating glucose or insulin responses to enhance exogenous glucose oxidation during steady-state aerobic exercise in young healthy men under simulated acute (8 h) high-altitude (460 mmHg) conditions. These results indicate that dysregulations in glucose metabolism in native lowlanders sojourning at high altitude may not be due to insulin resistance at peripheral tissue.
Collapse
Affiliation(s)
- Lee M Margolis
- United States Army Research Institute of Environmental Medicine, Natick, Massachusetts, United States
| | - Marques A Wilson
- United States Army Research Institute of Environmental Medicine, Natick, Massachusetts, United States
| | - Devin J Drummer
- United States Army Research Institute of Environmental Medicine, Natick, Massachusetts, United States
- Oak Ridge Institute for Science and Education, Belcamp, Maryland, United States
| | - Christopher T Carrigan
- United States Army Research Institute of Environmental Medicine, Natick, Massachusetts, United States
| | - Nancy E Murphy
- United States Army Research Institute of Environmental Medicine, Natick, Massachusetts, United States
| | - Jillian T Allen
- United States Army Research Institute of Environmental Medicine, Natick, Massachusetts, United States
| | - M Alan Dawson
- United States Army Research Institute of Environmental Medicine, Natick, Massachusetts, United States
| | - Christos S Mantzoros
- Department of Medicine, Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States
- Department of Medicine, Veterans Affairs Boston Healthcare System, Boston, Massachusetts, United States
| | - Andrew J Young
- United States Army Research Institute of Environmental Medicine, Natick, Massachusetts, United States
- Oak Ridge Institute for Science and Education, Belcamp, Maryland, United States
| | - Stefan M Pasiakos
- Office of Dietary Supplements, National Institutes of Health, Bethesda, Maryland, United States
| |
Collapse
|
5
|
Rochowski MT, Jayathilake K, Balcerak JM, Tamil Selvan M, Gunasekara S, Rudd J, Miller C, Lacombe VA. Alterations of whole body glucose metabolism in a feline SARS-CoV-2 infection model. Am J Physiol Regul Integr Comp Physiol 2024; 326:R499-R506. [PMID: 38574344 PMCID: PMC11381005 DOI: 10.1152/ajpregu.00228.2023] [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: 10/04/2023] [Revised: 03/06/2024] [Accepted: 03/30/2024] [Indexed: 04/06/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been especially devastating to patients with comorbidities, including metabolic and cardiovascular diseases. Elevated blood glucose during SARS-CoV-2 infection increased mortality of patients with COVID-19, although the mechanisms are not well understood. It has been previously demonstrated that glucose transport and utilization is a crucial pathway for other highly infectious RNA viruses. Thus, we hypothesized that SARS-CoV-2 infection could lead to alterations in cellular and whole body glucose metabolism. Specific pathogen-free domestic cats were intratracheally inoculated with USA-WA1/2020 (wild-type) SARS-CoV-2 or vehicle-inoculated, then euthanized at 4- and 8-days postinoculation (dpi). Blood glucose and cortisol concentrations were elevated at 4 and 8 dpi. Blood ketones, insulin, and angiotensin II concentrations remained unchanged throughout the experimental timeline. SARS-CoV-2 RNA was detected in the lung and heart, without changes in angiotensin-converting enzyme 2 (ACE2) RNA expression. In the lung, SARS-CoV-2 infection increased glucose transporter 1 (GLUT1) protein levels at 4 and 8 dpi, whereas GLUT4 level was only upregulated at 8 dpi. In the heart, GLUT-1 and -4 protein levels remained unchanged. Furthermore, GLUT1 level was upregulated in the skeletal muscle at 8 dpi, and AMPK was activated in the hearts of infected cats. SARS-CoV-2 infection increased blood glucose concentration and pulmonary GLUT protein levels. These findings suggest that SARS-CoV-2 infection induces metabolic reprogramming primarily in the lung to support viral replication. Furthermore, this translational feline model mimicked human COVID-19 and could be used to explore novel therapeutic targets to treat metabolic disease during SARS-CoV-2 infection.NEW & NOTEWORTHY Our study on a feline model of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, mirroring human COVID-19, revealed alterations in whole body and cellular glucose metabolism. Infected cats developed mild hyperglycemia, increased protein levels of glucose transporters in the lung, and AMPK activation in the heart. These findings suggest that SARS-CoV-2 infection induces metabolic reprogramming in the cardiorespiratory system to support viral replication. Understanding these mechanisms could lead to novel antiviral therapeutic strategies.
Collapse
Affiliation(s)
- Matthew T Rochowski
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, United States
- Harold Hamm Diabetes Center, Oklahoma City, Oklahoma, United States
| | - Kaushalya Jayathilake
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, United States
| | - John-Michael Balcerak
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, United States
| | - Miruthula Tamil Selvan
- Department of Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, United States
| | - Sachithra Gunasekara
- Department of Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, United States
| | - Jennifer Rudd
- Department of Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, United States
| | - Craig Miller
- Department of Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, United States
| | - Véronique A Lacombe
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, United States
- Harold Hamm Diabetes Center, Oklahoma City, Oklahoma, United States
| |
Collapse
|
6
|
Popova D, Sun J, Chow HM, Hart RP. A critical review of ethanol effects on neuronal firing: A metabolic perspective. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2024; 48:450-458. [PMID: 38217065 DOI: 10.1111/acer.15266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 12/22/2023] [Accepted: 01/02/2024] [Indexed: 01/14/2024]
Abstract
Ethanol metabolism is relatively understudied in neurons, even though changes in neuronal metabolism are known to affect their activity. Recent work demonstrates that ethanol is preferentially metabolized over glucose as a source of carbon and energy, and it reprograms neurons to a state of reduced energy potential and diminished capacity to utilize glucose once ethanol is exhausted. Ethanol intake has been associated with changes in neuronal firing and specific brain activity (EEG) patterns have been linked with risk for alcohol use disorder (AUD). Furthermore, a haplotype of the inwardly rectifying potassium channel subunit, GIRK2, which plays a critical role in regulating excitability of neurons, has been linked with AUD and shown to be directly regulated by ethanol. At the same time, overexpression of GIRK2 prevents ethanol-induced metabolic changes. Based on the available evidence, we conclude that the mechanisms underlying the effects of ethanol on neuronal metabolism are a novel target for developing therapies for AUD.
Collapse
Affiliation(s)
- Dina Popova
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey, USA
| | - Jacquelyne Sun
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Hei-Man Chow
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong, Hong Kong
- Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Ronald P Hart
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey, USA
| |
Collapse
|
7
|
Pan Q, Ai W, Guo S. TGF-β1 Signaling Impairs Metformin Action on Glycemic Control. Int J Mol Sci 2024; 25:2424. [PMID: 38397103 PMCID: PMC10889280 DOI: 10.3390/ijms25042424] [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: 01/07/2024] [Revised: 02/02/2024] [Accepted: 02/16/2024] [Indexed: 02/25/2024] Open
Abstract
Hyperglycemia is a hallmark of type 2 diabetes (T2D). Metformin, the first-line drug used to treat T2D, maintains blood glucose within a normal range by suppressing hepatic glucose production (HGP). However, resistance to metformin treatment is developed in most T2D patients over time. Transforming growth factor beta 1 (TGF-β1) levels are elevated both in the liver and serum of T2D humans and mice. Here, we found that TGF-β1 treatment impairs metformin action on suppressing HGP via inhibiting AMPK phosphorylation at Threonine 172 (T172). Hepatic TGF-β1 deficiency improves metformin action on glycemic control in high fat diet (HFD)-induced obese mice. In our hepatic insulin resistant mouse model (hepatic insulin receptor substrate 1 (IRS1) and IRS2 double knockout (DKO)), metformin action on glycemic control was impaired, which is largely improved by further deletion of hepatic TGF-β1 (TKObeta1) or hepatic Foxo1 (TKOfoxo1). Moreover, blockade of TGF-β1 signaling by chemical inhibitor of TGF-β1 type I receptor LY2157299 improves to metformin sensitivity in mice. Taken together, our current study suggests that hepatic TGF-β1 signaling impairs metformin action on glycemic control, and suppression of TGF-β1 signaling could serve as part of combination therapy with metformin for T2D treatment.
Collapse
Affiliation(s)
| | | | - Shaodong Guo
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX 77843, USA; (Q.P.); (W.A.)
| |
Collapse
|
8
|
Derosa G, D’Angelo A, Maffioli P, Cucinella L, Nappi RE. The Use of Nigella sativa in Cardiometabolic Diseases. Biomedicines 2024; 12:405. [PMID: 38398007 PMCID: PMC10886913 DOI: 10.3390/biomedicines12020405] [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: 10/01/2023] [Revised: 11/05/2023] [Accepted: 11/09/2023] [Indexed: 02/25/2024] Open
Abstract
Nigella sativa L. is an herb that is commonly used in cooking and in traditional medicine, particularly in Arab countries, the Indian subcontinent, and some areas of eastern Europe. Nigella sativa is also called "black cumin" or "black seeds", as the seeds are the most-used part of the plant. They contain the main bioactive component thymoquinone (TQ), which is responsible for the pleiotropic pharmacological properties of the seeds, including anti-oxidant, anti-inflammatory, anti-hypertensive, anti-hepatotoxic, hypoglycemic, and lipid-lowering properties. In this narrative review, both the potential mechanisms of action of Nigella sativa and the fundamental role played by pharmaceutical technology in optimizing preparations based on this herb in terms of yield, quality, and effectiveness have been outlined. Moreover, an analysis of the market of products containing Nigella sativa was carried out based on the current literature with an international perspective, along with a specific focus on Italy.
Collapse
Affiliation(s)
- Giuseppe Derosa
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy;
- Centre of Diabetes, Metabolic Diseases, and Dyslipidemias, University of Pavia, 27100 Pavia, Italy;
- Regional Centre for Prevention, Surveillance, Diagnosis and Treatment of Dyslipidemias and Atherosclerosis, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
- Italian Nutraceutical Society (SINut), 40100 Bologna, Italy
- Laboratory of Molecular Medicine, Fondazione IRCCS Policlinico San Matteo, University of Pavia, 27100 Pavia, Italy
| | - Angela D’Angelo
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy;
- Laboratory of Molecular Medicine, Fondazione IRCCS Policlinico San Matteo, University of Pavia, 27100 Pavia, Italy
| | - Pamela Maffioli
- Centre of Diabetes, Metabolic Diseases, and Dyslipidemias, University of Pavia, 27100 Pavia, Italy;
- Regional Centre for Prevention, Surveillance, Diagnosis and Treatment of Dyslipidemias and Atherosclerosis, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
- Italian Nutraceutical Society (SINut), 40100 Bologna, Italy
| | - Laura Cucinella
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.E.N.)
- Research Center for Reproductive Medicine and Gynecological Endocrinology, Menopause Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Rossella Elena Nappi
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.E.N.)
- Research Center for Reproductive Medicine and Gynecological Endocrinology, Menopause Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| |
Collapse
|
9
|
Wang H, Shen M, Shu X, Guo B, Jia T, Feng J, Lu Z, Chen Y, Lin J, Liu Y, Zhang J, Zhang X, Sun D. Cardiac Metabolism, Reprogramming, and Diseases. J Cardiovasc Transl Res 2024; 17:71-84. [PMID: 37668897 DOI: 10.1007/s12265-023-10432-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 08/22/2023] [Indexed: 09/06/2023]
Abstract
Cardiovascular diseases (CVD) account for the largest bulk of deaths worldwide, posing a massive burden on societies and the global healthcare system. Besides, the incidence and prevalence of these diseases are on the rise, demanding imminent action to revert this trend. Cardiovascular pathogenesis harbors a variety of molecular and cellular mechanisms among which dysregulated metabolism is of significant importance and may even proceed other mechanisms. The healthy heart metabolism primarily relies on fatty acids for the ultimate production of energy through oxidative phosphorylation in mitochondria. Other metabolites such as glucose, amino acids, and ketone bodies come next. Under pathological conditions, there is a shift in metabolic pathways and the preference of metabolites, termed metabolic remodeling or reprogramming. In this review, we aim to summarize cardiovascular metabolism and remodeling in different subsets of CVD to come up with a new paradigm for understanding and treatment of these diseases.
Collapse
Affiliation(s)
- Haichang Wang
- Heart Hospital, Xi'an International Medical Center, Xi'an, China
| | - Min Shen
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Xiaofei Shu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Baolin Guo
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Tengfei Jia
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Jiaxu Feng
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Zuocheng Lu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Yanyan Chen
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Jie Lin
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Yue Liu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Jiye Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Xuan Zhang
- Institute for Hospital Management Research, Chinese PLA General Hospital, Beijing, China.
| | - Dongdong Sun
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
10
|
Rotllan N, Julve J, Escolà-Gil JC. Type 2 Diabetes and HDL Dysfunction: A Key Contributor to Glycemic Control. Curr Med Chem 2024; 31:280-285. [PMID: 36722477 DOI: 10.2174/0929867330666230201124125] [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: 07/08/2022] [Revised: 10/31/2022] [Accepted: 12/08/2022] [Indexed: 02/02/2023]
Abstract
High-density lipoproteins (HDL) have been shown to exert multiple cardioprotective and antidiabetic functions, such as their ability to promote cellular cholesterol efflux and their antioxidant, anti-inflammatory, and antiapoptotic properties. Type 2 diabetes (T2D) is usually associated with low high-density lipoprotein cholesterol (HDL-C) levels as well as with significant alterations in the HDL composition, thereby impairing its main functions. HDL dysfunction also negatively impacts both pancreatic β-cell function and skeletal muscle insulin sensitivity, perpetuating this adverse self-feeding cycle. The impairment of these pathways is partly dependent on cellular ATP-binding cassette transporter (ABC) A1-mediated efflux to lipid-poor apolipoprotein (apo) A-I in the extracellular space. In line with these findings, experimental interventions aimed at improving HDL functions, such as infusions of synthetic HDL or lipid-poor apoA-I, significantly improved glycemic control in T2D patients and experimental models of the disease. Cholesteryl ester transfer protein (CETP) inhibitors are specific drugs designed to increase HDLC and HDL functions. Posthoc analyses of large clinical trials with CETP inhibitors have demonstrated their potential anti-diabetic properties. Research on HDL functionality and HDL-based therapies could be a crucial step toward improved glycemic control in T2D subjects.
Collapse
Affiliation(s)
- Noemi Rotllan
- Institut de recerca de l'Hospital de la Santa Creu i Sant Pau, Institut d'Investigacions Biomèdiques (IIB) Sant Pau, Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
| | - Josep Julve
- Institut de recerca de l'Hospital de la Santa Creu i Sant Pau, Institut d'Investigacions Biomèdiques (IIB) Sant Pau, Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
| | - Joan Carles Escolà-Gil
- Institut de recerca de l'Hospital de la Santa Creu i Sant Pau, Institut d'Investigacions Biomèdiques (IIB) Sant Pau, Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
11
|
Yuan W, Fang W, Zhang R, Lyu H, Xiao S, Guo D, Ali DW, Michalak M, Chen XZ, Zhou C, Tang J. Therapeutic strategies targeting AMPK-dependent autophagy in cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119537. [PMID: 37463638 DOI: 10.1016/j.bbamcr.2023.119537] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 07/04/2023] [Accepted: 07/07/2023] [Indexed: 07/20/2023]
Abstract
Macroautophagy is a health-modifying process of engulfing misfolded or aggregated proteins or damaged organelles, coating these proteins or organelles into vesicles, fusion of vesicles with lysosomes to form autophagic lysosomes, and degradation of the encapsulated contents. It is also a self-rescue strategy in response to harsh environments and plays an essential role in cancer cells. AMP-activated protein kinase (AMPK) is the central pathway that regulates autophagy initiation and autophagosome formation by phosphorylating targets such as mTORC1 and unc-51 like activating kinase 1 (ULK1). AMPK is an evolutionarily conserved serine/threonine protein kinase that acts as an energy sensor in cells and regulates various metabolic processes, including those involved in cancer. The regulatory network of AMPK is complicated and can be regulated by multiple upstream factors, such as LKB1, AKT, PPAR, SIRT1, or noncoding RNAs. Currently, AMPK is being investigated as a novel target for anticancer therapies based on its role in macroautophagy regulation. Herein, we review the effects of AMPK-dependent autophagy on tumor cell survival and treatment strategies targeting AMPK.
Collapse
Affiliation(s)
- Wenbin Yuan
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Wanyi Fang
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Rui Zhang
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Hao Lyu
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Shuai Xiao
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Dong Guo
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Declan William Ali
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Marek Michalak
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Xing-Zhen Chen
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Cefan Zhou
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China.
| | - Jingfeng Tang
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China.
| |
Collapse
|
12
|
Peña-Jorquera H, Cid-Jofré V, Landaeta-Díaz L, Petermann-Rocha F, Martorell M, Zbinden-Foncea H, Ferrari G, Jorquera-Aguilera C, Cristi-Montero C. Plant-Based Nutrition: Exploring Health Benefits for Atherosclerosis, Chronic Diseases, and Metabolic Syndrome-A Comprehensive Review. Nutrients 2023; 15:3244. [PMID: 37513660 PMCID: PMC10386413 DOI: 10.3390/nu15143244] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/14/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
Atherosclerosis, chronic non-communicable diseases, and metabolic syndrome are highly interconnected and collectively contribute to global health concerns that reduce life expectancy and quality of life. These conditions arise from multiple risk factors, including inflammation, insulin resistance, impaired blood lipid profile, endothelial dysfunction, and increased cardiovascular risk. Adopting a plant-based diet has gained popularity as a viable alternative to promote health and mitigate the incidence of, and risk factors associated with, these three health conditions. Understanding the potential benefits of a plant-based diet for human health is crucial, particularly in the face of the rising prevalence of chronic diseases like diabetes, hypertension, dyslipidemia, atherosclerosis, and cancer. Thus, this review focused on the plausible advantages of consuming a type of food pattern for the prevention and/or treatment of chronic diseases, emphasizing the dietary aspects that contribute to these conditions and the evidence supporting the benefits of a plant-based diet for human health. To facilitate a more in-depth analysis, we present separate evidence for each of these three concepts, acknowledging their intrinsic connection while providing a specific focus on each one. This review underscores the potential of a plant-based diet to target the underlying causes of these chronic diseases and enhance health outcomes for individuals and populations.
Collapse
Affiliation(s)
- Humberto Peña-Jorquera
- IRyS Group, Physical Education School, Pontificia Universidad Católica de Valparaíso, Viña del Mar 2530388, Chile
| | - Valeska Cid-Jofré
- Centro de Investigación Biomédica y Aplicada (CIBAP), Escuela de Medicina, Facultad de Ciencias Médicas, Universidad de Santiago de Chile (USACH), Santiago 9160019, Chile
| | - Leslie Landaeta-Díaz
- Facultad de Salud y Ciencias Sociales, Universidad de las Américas, Santiago 7500975, Chile
- Núcleo en Ciencias Ambientales y Alimentarias, Universidad de las Américas, Santiago 7500975, Chile
| | - Fanny Petermann-Rocha
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad Diego Portales, Santiago 8370068, Chile
- BHF Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK
| | - Miquel Martorell
- Department of Nutrition and Dietetics, Faculty of Pharmacy, Centre for Healthy Living, University of Concepción, Concepción 4070386, Chile
| | - Hermann Zbinden-Foncea
- Laboratorio de Fisiología del Ejercicio y Metabolismo, Escuela de Kinesiología, Facultad de Medicina, Universidad Finis Terrae, Santiago 7500000, Chile
- Facultad de Ciencias de la Salud, Universidad Francisco de Vitoria, 28223 Madrid, Spain
| | - Gerson Ferrari
- Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Av. Pedro de Valdivia 425, Providencia 7500912, Chile
- Escuela de Ciencias de la Actividad Física, el Deporte y la Salud, Universidad de Santiago de Chile (USACH), Santiago 9170022, Chile
| | - Carlos Jorquera-Aguilera
- Escuela de Nutrición y Dietética, Facultad de Ciencias, Universidad Mayor, Santiago 8580745, Chile
| | - Carlos Cristi-Montero
- IRyS Group, Physical Education School, Pontificia Universidad Católica de Valparaíso, Viña del Mar 2530388, Chile
| |
Collapse
|
13
|
Izbicka E, Streeper RT. Mitigation of Insulin Resistance by Natural Products from a New Class of Molecules, Membrane-Active Immunomodulators. Pharmaceuticals (Basel) 2023; 16:913. [PMID: 37513825 PMCID: PMC10386479 DOI: 10.3390/ph16070913] [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: 04/10/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 07/30/2023] Open
Abstract
Insulin resistance (IR), accompanied by an impaired cellular glucose uptake, characterizes diverse pathologies that include, but are not limited to, metabolic disease, prediabetes and type 2 diabetes. Chronic inflammation associated with deranged cellular signaling is thought to contribute to IR. The key molecular players in IR are plasma membrane proteins, including the insulin receptor and glucose transporter 4. Certain natural products, such as lipids, phenols, terpenes, antibiotics and alkaloids have beneficial effects on IR, yet their mode of action remains obscured. We hypothesized that these products belong to a novel class of bioactive molecules that we have named membrane-active immunomodulators (MAIMs). A representative MAIM, the naturally occurring medium chain fatty acid ester diethyl azelate (DEA), has been shown to increase the fluidity of cell plasma membranes with subsequent downstream effects on cellular signaling. DEA has also been shown to improve markers of IR, including blood glucose, insulin and lipid levels, in humans. The literature supports the notion that DEA and other natural MAIMs share similar mechanisms of action in improving IR. These findings shed a new light on the mechanism of IR mitigation using natural products, and may facilitate the discovery of other compounds with similar activities.
Collapse
|
14
|
Covert JD, Grice BA, Thornburg MG, Kaur M, Ryan AP, Tackett L, Bhamidipati T, Stull ND, Kim T, Habegger KM, McClain DA, Brozinick JT, Elmendorf JS. An early, reversible cholesterolgenic etiology of diet-induced insulin resistance. Mol Metab 2023; 72:101715. [PMID: 37019209 PMCID: PMC10114231 DOI: 10.1016/j.molmet.2023.101715] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/27/2023] [Accepted: 03/21/2023] [Indexed: 04/07/2023] Open
Abstract
OBJECTIVE A buildup of skeletal muscle plasma membrane (PM) cholesterol content in mice occurs within 1 week of a Western-style high-fat diet and causes insulin resistance. The mechanism driving this cholesterol accumulation and insulin resistance is not known. Promising cell data implicate that the hexosamine biosynthesis pathway (HBP) triggers a cholesterolgenic response via increasing the transcriptional activity of Sp1. In this study we aimed to determine whether increased HBP/Sp1 activity represented a preventable cause of insulin resistance. METHODS C57BL/6NJ mice were fed either a low-fat (LF, 10% kcal) or high-fat (HF, 45% kcal) diet for 1 week. During this 1-week diet the mice were treated daily with either saline or mithramycin-A (MTM), a specific Sp1/DNA-binding inhibitor. A series of metabolic and tissue analyses were then performed on these mice, as well as on mice with targeted skeletal muscle overexpression of the rate-limiting HBP enzyme glutamine-fructose-6-phosphate-amidotransferase (GFAT) that were maintained on a regular chow diet. RESULTS Saline-treated mice fed this HF diet for 1 week did not have an increase in adiposity, lean mass, or body mass while displaying early insulin resistance. Consistent with an HBP/Sp1 cholesterolgenic response, Sp1 displayed increased O-GlcNAcylation and binding to the HMGCR promoter that increased HMGCR expression in skeletal muscle from saline-treated HF-fed mice. Skeletal muscle from these saline-treated HF-fed mice also showed a resultant elevation of PM cholesterol with an accompanying loss of cortical filamentous actin (F-actin) that is essential for insulin-stimulated glucose transport. Treating these mice daily with MTM during the 1-week HF diet fully prevented the diet-induced Sp1 cholesterolgenic response, loss of cortical F-actin, and development of insulin resistance. Similarly, increases in HMGCR expression and cholesterol were measured in muscle from GFAT transgenic mice compared to age- and weight-match wildtype littermate control mice. In the GFAT Tg mice we found that these increases were alleviated by MTM. CONCLUSIONS These data identify increased HBP/Sp1 activity as an early mechanism of diet-induced insulin resistance. Therapies targeting this mechanism may decelerate T2D development.
Collapse
Affiliation(s)
- Jacob D Covert
- Department of Anatomy, Cell Biology and Physiology, Indianapolis, IN, United States; Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Brian A Grice
- Department of Anatomy, Cell Biology and Physiology, Indianapolis, IN, United States; Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Matthew G Thornburg
- Department of Anatomy, Cell Biology and Physiology, Indianapolis, IN, United States; Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Manpreet Kaur
- Department of Anatomy, Cell Biology and Physiology, Indianapolis, IN, United States; Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Andrew P Ryan
- Department of Anatomy, Cell Biology and Physiology, Indianapolis, IN, United States; Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, United States; Eli Lilly and Company, Indianapolis, IN, United States
| | - Lixuan Tackett
- Department of Anatomy, Cell Biology and Physiology, Indianapolis, IN, United States; Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Theja Bhamidipati
- Department of Anatomy, Cell Biology and Physiology, Indianapolis, IN, United States; Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Natalie D Stull
- Indiana Biosciences Research Institute Indianapolis, IN, United States
| | - Teayoun Kim
- Comprehensive Diabetes Center and Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Kirk M Habegger
- Comprehensive Diabetes Center and Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Donald A McClain
- Section of Endocrinology and Metabolism, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Joseph T Brozinick
- Department of Anatomy, Cell Biology and Physiology, Indianapolis, IN, United States; Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, United States; Comprehensive Diabetes Center and Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jeffrey S Elmendorf
- Department of Anatomy, Cell Biology and Physiology, Indianapolis, IN, United States; Department of Biochemistry and Molecular Biology, Indianapolis, IN, United States; Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, United States.
| |
Collapse
|
15
|
AlTamimi JZ, AlFaris NA, Alshammari GM, Alagal RI, Aljabryn DH, Yahya MA. The Protective Effect of 11-Keto-β-Boswellic Acid against Diabetic Cardiomyopathy in Rats Entails Activation of AMPK. Nutrients 2023; 15:nu15071660. [PMID: 37049501 PMCID: PMC10097356 DOI: 10.3390/nu15071660] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 03/25/2023] [Accepted: 03/25/2023] [Indexed: 04/03/2023] Open
Abstract
This study examined the protective effect of 11-keto-β-boswellic acid (AKBA) against streptozotocin (STZ)-induced diabetic cardiomyopathy (DC) in rats and examined the possible mechanisms of action. Male rats were divided into 5 groups (n = 8/each): (1) control, AKBA (10 mg/kg, orally), STZ (65 mg/kg, i.p.), STZ + AKBA (10 mg/kg, orally), and STZ + AKBA + compound C (CC/an AMPK inhibitor, 0.2 mg/kg, i.p.). AKBA improved the structure and the systolic and diastolic functions of the left ventricles (LVs) of STZ rats. It also attenuated the increase in plasma glucose, plasma insulin, and serum and hepatic levels of triglycerides (TGs), cholesterol (CHOL), and free fatty acids (FFAs) in these diabetic rats. AKBA stimulated the ventricular activities of phosphofructokinase (PFK), pyruvate dehydrogenase (PDH), and acetyl CoA carboxylase (ACC); increased levels of malonyl CoA; and reduced levels of carnitine palmitoyltransferase I (CPT1), indicating improvement in glucose and FA oxidation. It also reduced levels of malondialdehyde (MDA); increased mitochondria efficiency and ATP production; stimulated mRNA, total, and nuclear levels of Nrf2; increased levels of glutathione (GSH), heme oxygenase (HO-1), superoxide dismutase (SOD), and catalase (CAT); but reduced the expression and nuclear translocation of NF-κB and levels of tumor-necrosis factor-α (TNF-α) and interleukin-6 (IL-6). These effects were concomitant with increased activities of AMPK in the LVs of the control and STZ-diabetic rats. Treatment with CC abolished all these protective effects of AKBA. In conclusion, AKBA protects against DC in rats, mainly by activating the AMPK-dependent control of insulin release, cardiac metabolism, and antioxidant and anti-inflammatory effects.
Collapse
|
16
|
Felemban AH, Alshammari GM, Yagoub AEA, Al-Harbi LN, Alhussain MH, Yahya MA. Activation of AMPK Entails the Protective Effect of Royal Jelly against High-Fat-Diet-Induced Hyperglycemia, Hyperlipidemia, and Non-Alcoholic Fatty Liver Disease in Rats. Nutrients 2023; 15:nu15061471. [PMID: 36986201 PMCID: PMC10056733 DOI: 10.3390/nu15061471] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/14/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
This study examined the mechanism underlying the protective effect of royal jelly (RJ) against high-fat-diet (HFD)-mediated non-alcoholic liver disease (NAFLD) in rats. Adult male rats were divided into five groups (n = 8 each): control fed a standard diet, control + RJ (300 mg/kg), HFD, HFD + RJ (300 mg/kg), and HFD + RJ + CC (0.2 mg/kg). The treatment with RJ reduced weight gain, increased fat pads, and attenuated fasting hyperglycemia, hyperinsulinemia, and glucose tolerance in the HFD-fed rats. It also reduced the serum levels of liver function enzymes, interleukin 6 (IL-6), tumor necrosis factor-α (TNF-α), and leptin but significantly increased the serum levels of adiponectin. In addition, and with no effect on lipid excretion in stool, RJ significantly decreased the hepatic mRNA expression of SREBP1, serum, hepatic cholesterol, and triglycerides but increased hepatic mRNA levels of PPARα. Furthermore, RJ reduced the hepatic levels of TNF-α, IL-6, and malondialdehyde (MDA) in the livers of these rats. Of note, with no effect on the mRNA levels of AMPK, RJ stimulated the phosphorylation of AMPK and increased the levels of superoxide dismutase (SOD) and total glutathione (GSH) in the livers of the control and HFD-fed rats. In conclusion, RJ attenuates NAFLD via its antioxidant potential and adiponectin-independent activation of liver AMPK.
Collapse
Affiliation(s)
- Alaa Hasanain Felemban
- Department of Food Science and Nutrition, College of Food and Agricultural Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ghedeir M Alshammari
- Department of Food Science and Nutrition, College of Food and Agricultural Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abu ElGasim Ahmed Yagoub
- Department of Food Science and Nutrition, College of Food and Agricultural Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Laila Naif Al-Harbi
- Department of Food Science and Nutrition, College of Food and Agricultural Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Maha H Alhussain
- Department of Food Science and Nutrition, College of Food and Agricultural Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed Abdo Yahya
- Department of Food Science and Nutrition, College of Food and Agricultural Science, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
17
|
Wu D, Yuan X, Zhou R, Chen W, Li W, Li Z, Li X, Zhu R, Wang H, Yang Y. Aqueous extract of Sanghuangporus baumii induces autophagy to inhibit cervical carcinoma growth. Food Funct 2023; 14:2374-2384. [PMID: 36779533 DOI: 10.1039/d2fo02887e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Sanghuangporus baumii, an edible fungus rich in heteropolysaccharides, has been found to have some anti-cervical cancer effects. In the current study, the effects of an aqueous extract of S. baumii on cervical cancer were investigated in a U14 cervical carcinoma cell implanted female Kunming mouse model. An aqueous extract of S. baumii (SHWE) was administered to tumor-bearing mice by gavage for 21 days. SHWE treatment significantly inhibited tumor growth by 67.4% at a dose of 400 mg per kg bodyweight. Transcriptomic results showed that the expression of key genes GABARAP, VMP1, VAMP8 and STX17 which are involved in the autophagy pathway was regulated after SHWE treatment, suggesting that SHWE may induce autophagy in tumors. The results were further confirmed by measuring the LC3II/LC3I ratio using western blotting. Moreover, some differentially expressed genes were involved in the insulin signaling pathway, implying that SHWE induced autophagy by disturbing glucose uptake and utilization in tumors. The analysis of the gut microbiota indicated that SHWE treatment stimulated the proliferation of Akkermansia, a well-known probiotic that presented benefits in metabolic regulation and cancer therapy. In conclusion, SHWE administration modified the gut microbiota, disturbed the glucose metabolism and induced autophagy in tumors, and then inhibited the development of cervical carcinoma in vivo.
Collapse
Affiliation(s)
- Di Wu
- Institute of Edible Fungi, Shanghai Academy of Agriculture Sciences, National Engineering Research Center of Edible Fungi, Key Laboratory of Edible Fungi Resources and Utilization (South), Ministry of Agriculture, Shanghai 201403, China.
| | - Xuemei Yuan
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, China.
| | - Ruijie Zhou
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, China.
| | - Wanchao Chen
- Institute of Edible Fungi, Shanghai Academy of Agriculture Sciences, National Engineering Research Center of Edible Fungi, Key Laboratory of Edible Fungi Resources and Utilization (South), Ministry of Agriculture, Shanghai 201403, China.
| | - Wen Li
- Institute of Edible Fungi, Shanghai Academy of Agriculture Sciences, National Engineering Research Center of Edible Fungi, Key Laboratory of Edible Fungi Resources and Utilization (South), Ministry of Agriculture, Shanghai 201403, China.
| | - Zhengpeng Li
- Institute of Edible Fungi, Shanghai Academy of Agriculture Sciences, National Engineering Research Center of Edible Fungi, Key Laboratory of Edible Fungi Resources and Utilization (South), Ministry of Agriculture, Shanghai 201403, China.
| | - Xueyin Li
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, China.
| | - Rui Zhu
- Department of Integrated Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hualin Wang
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, China.
| | - Yan Yang
- Institute of Edible Fungi, Shanghai Academy of Agriculture Sciences, National Engineering Research Center of Edible Fungi, Key Laboratory of Edible Fungi Resources and Utilization (South), Ministry of Agriculture, Shanghai 201403, China.
| |
Collapse
|
18
|
Kim K, Yoon H. Gamma-Aminobutyric Acid Signaling in Damage Response, Metabolism, and Disease. Int J Mol Sci 2023; 24:ijms24054584. [PMID: 36902014 PMCID: PMC10003236 DOI: 10.3390/ijms24054584] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 03/03/2023] Open
Abstract
Gamma-aminobutyric acid (GABA) plays a crucial role in signal transduction and can function as a neurotransmitter. Although many studies have been conducted on GABA in brain biology, the cellular function and physiological relevance of GABA in other metabolic organs remain unclear. Here, we will discuss recent advances in understanding GABA metabolism with a focus on its biosynthesis and cellular functions in other organs. The mechanisms of GABA in liver biology and disease have revealed new ways to link the biosynthesis of GABA to its cellular function. By reviewing what is known about the distinct effects of GABA and GABA-mediated metabolites in physiological pathways, we provide a framework for understanding newly identified targets regulating the damage response, with implications for ameliorating metabolic diseases. With this review, we suggest that further research is necessary to develop GABA's beneficial and toxic effects on metabolic disease progression.
Collapse
|
19
|
Lee SR, Jeong SH, Mukae M, Jeong KJ, Kwun HJ, Hong EJ. GLUT4 degradation by GLUTFOURINH® in mice resembles moderate-obese diabetes of human with hyperglycemia and low lipid accumulation. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166668. [PMID: 36822448 DOI: 10.1016/j.bbadis.2023.166668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 01/28/2023] [Accepted: 02/14/2023] [Indexed: 02/23/2023]
Abstract
BACKGROUNDS AND AIMS Type 2 diabetes mellitus (T2D) is a chronic disease characterized by insulin resistance and hyperglycemia. To investigate T2D, genetic and chemical induced hyper-obese rodent models have been experimentally developed. However, establishment of moderate-obese diabetes model will confer diverse opportunities for translational studies. In this study, we found the chemical, GLUTFOURINH® (GFI), induces post-translational degradation of glucose transporter 4 (GLUT4). We aimed to establish novel diabetic model by using GFI. METHODS AND RESULTS Low plasma membrane GLUT4 (pmGLUT4) levels by GFI resulted in reduction of intracellular glucose uptake and TG, and increase of intracellular FFA in A204 cells. Likewise, GFI treatment decreased intracellular TG and increased intracellular FFA levels in Hep3B and 3T3-L1 cells. Mice were administered with GFI (16 mg/kg) for short-term (3-day) and long-term (28- and 31-day) to compared with vehicle injection, HFD model, and T2D model, respectively. Short-term and long-term GFI treatments induced hyperglycemia and hyperinsulinemia with low pmGLUT4 levels. Compared to HFD model, long-term GFI with HFD reduced adipose weight and intracellular TG accumulation, but increased plasma FFA. GFI treatment resulted in insulin resistance by showing low QUICKI and high HOMA-IR values, and low insulin response during insulin tolerance test. Additionally, low pmGLUT4 by GFI heightened hyperglycemia, hyperinsulinemia, and insulin resistance compared to T2D model. CONCLUSIONS In summary, we report GLUT4 degradation by novel chemical (GFI) induces moderate-obese diabetes representing hyperglycemia, insulin resistance and low intracellular lipid accumulation. The GLUT4 degradation by GFI has translational value for studying diseases related to moderate-obese diabetes.
Collapse
Affiliation(s)
- Sang R Lee
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Su Hee Jeong
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Moeka Mukae
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Kang Joo Jeong
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Hyo-Jung Kwun
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Eui-Ju Hong
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea.
| |
Collapse
|
20
|
Ryu B, Park EJ, Doan TP, Cho HM, An JP, Pham TLG, Pham HTT, Oh WK. Heliciopsides A-E, Unusual Macrocyclic and Phenolic Glycosides from the Leaves of Heliciopsis terminalis and Their Stimulation of Glucose Uptake. Pharmaceuticals (Basel) 2022; 15:1315. [PMID: 36355487 PMCID: PMC9695999 DOI: 10.3390/ph15111315] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/10/2022] [Accepted: 10/21/2022] [Indexed: 11/26/2023] Open
Abstract
Ten phenolic constituents, including three new macrocyclic glycosides (1-3), a new phenolic glycoside (5), a new biphenyl glycoside (6), and five known compounds (4, 7-10), were isolated from a 70% MeOH extract of the leaves of Heliciopsis terminalis by liquid chromatography-tandem mass spectrometry (LC-MS/MS)-guided molecular networking. The chemical structures of new compounds 1-3, 5 and 6 were established based on comprehensive spectroscopic data analysis, including 1D and 2D NMR and HRESIMS techniques. All isolated compounds (1-10) were evaluated for their stimulation of glucose uptake in differentiated 3T3-L1 adipocytes using 2-deoxy-2-[(7-nitro-2,1,3-benzoxadiazol-4-yl)amino]-d-glucose (2-NBDG) as a fluorescent glucose analog. Compounds 3, 6 and 8 showed stimulatory effects on the uptake of 2-NBDG in 3T3-L1 adipocyte cells. Among them, compounds 3 and 6 activated the AMPK signaling pathway in differentiated C2C12 myoblasts.
Collapse
Affiliation(s)
- Byeol Ryu
- Korea Bioactive Natural Material Bank, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Korea
| | - Eun-Jin Park
- Korea Bioactive Natural Material Bank, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Korea
| | - Thi-Phuong Doan
- Korea Bioactive Natural Material Bank, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Korea
| | - Hyo-Moon Cho
- Korea Bioactive Natural Material Bank, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Korea
| | - Jin-Pyo An
- Korea Bioactive Natural Material Bank, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Korea
| | | | - Ha-Thanh-Tung Pham
- Department of Botany, Hanoi University of Pharmacy, Hanoi 000084, Vietnam
| | - Won-Keun Oh
- Korea Bioactive Natural Material Bank, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
21
|
Thymus serpyllum Exhibits Anti-Diabetic Potential in Streptozotocin-Induced Diabetes Mellitus Type 2 Mice: A Combined Biochemical and In Vivo Study. Nutrients 2022; 14:nu14173561. [PMID: 36079819 PMCID: PMC9460602 DOI: 10.3390/nu14173561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/24/2022] [Accepted: 08/26/2022] [Indexed: 12/02/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a complex metabolic disorder that is characterized by hyperglycemia, insulin resistance, and lack of insulin production. It has been previously reported that Thymus serpyllum has therapeutic potential against many diseases. To investigate the antidiabetic action of Thymus serpyllum, this study aimed to analyze its restorative impact in diabetic mice, in which it was administered in diet. Diabetes was induced in BALB/c mice fed with a high-fat diet and two intraperitoneal injections of streptozotocin. With the onset of diabetes, the mice were administered daily with aqueous extract of Thymus serpyllum (500 mg/kg/d and 800 mg/kg/d) for 4 weeks. Body weight and fasting blood glucose levels were measured after every 1 week of the treatment. Subsequently, intraperitoneal glucose tolerance and insulin tolerance tests were conducted. In addition, liver tissue was isolated for assessment in terms of levels of gene expression of the AMPK, IRS1, and GLUT2 gene. Treatment with the aqueous extract of Thymus serpyllum was found to be significantly effective in controlling hyperglycemia and improving glucose and insulin tolerance. Predictable with these impacts, the extract of Thymus serpyllum upregulated the AMPK expression at the mRNA level, as well as upregulating the expression of IRS1 and GLUT2 gene. Histopathological examination of the liver, kidney, and pancreas also revealed the restorative impact in terms of cellular morphology. The results hence demonstrated that oral administration of aqueous extract of Thymus serpyllum can potentially attenuate hyperglycemia in the liver muscle of streptozotocin (STZ)-induced diabetic mice via AMPK and IRS1 upregulation.
Collapse
|
22
|
N-Octyl Caffeamide, a Caffeic Acid Amide Derivative, Prevents Progression of Diabetes and Hepatic Steatosis in High-Fat Diet Induced Obese Mice. Int J Mol Sci 2022; 23:ijms23168948. [PMID: 36012215 PMCID: PMC9409300 DOI: 10.3390/ijms23168948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/08/2022] [Accepted: 08/09/2022] [Indexed: 11/30/2022] Open
Abstract
The underlying pathological mechanisms of diabetes are complicated and varied in diabetic patients, which may lead to the current medications often failing to maintain glycemic control in the long term. Thus, the discovery of diverse new compounds for developing medicines to treat diabetes and its complications are urgently needed. Polyphenols are metabolites of plants and have been employed in the prevention and treatment of a variety of diseases. Caffeic acid phenethyl ester (CAPE) is a category of compounds structurally similar to polyphenols. In this study, we aimed to investigate the antidiabetic activity and potential molecular mechanisms of a novel synthetic CAPE derivative N-octyl caffeamide (36M) using high-fat (HF) diet induced obese mouse models. Our results demonstrate that 36M prevented the progression of diabetes in the HF diet fed obese mice via increasing phosphorylation of adenosine monophosphate-activated protein kinase (AMPK) and inhibiting expression of protein tyrosine phosphatase 1B (PTP1B). We also found that 36M could prevent hepatic lipid storage in the HF diet fed mice via inhibition of fatty acid synthase and lipid droplet proteins, including perilipins and Fsp27. In conclusion, 36M is a potential candidate compound that can be developed as AMPK inhibitor and PTP1B inhibitor for treating diabetes and hepatic steatosis.
Collapse
|
23
|
Ansari P, Akther S, Hannan JMA, Seidel V, Nujat NJ, Abdel-Wahab YHA. Pharmacologically Active Phytomolecules Isolated from Traditional Antidiabetic Plants and Their Therapeutic Role for the Management of Diabetes Mellitus. Molecules 2022; 27:molecules27134278. [PMID: 35807526 PMCID: PMC9268530 DOI: 10.3390/molecules27134278] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/29/2022] [Accepted: 07/01/2022] [Indexed: 01/09/2023] Open
Abstract
Diabetes mellitus is a chronic complication that affects people of all ages. The increased prevalence of diabetes worldwide has led to the development of several synthetic drugs to tackle this health problem. Such drugs, although effective as antihyperglycemic agents, are accompanied by various side effects, costly, and inaccessible to the majority of people living in underdeveloped countries. Medicinal plants have been used traditionally throughout the ages to treat various ailments due to their availability and safe nature. Medicinal plants are a rich source of phytochemicals that possess several health benefits. As diabetes continues to become prevalent, health care practitioners are considering plant-based medicines as a potential source of antidiabetic drugs due to their high potency and fewer side effects. To better understand the mechanism of action of medicinal plants, their active phytoconstituents are being isolated and investigated thoroughly. In this review article, we have focused on pharmacologically active phytomolecules isolated from medicinal plants presenting antidiabetic activity and the role they play in the treatment and management of diabetes. These natural compounds may represent as good candidates for a novel therapeutic approach and/or effective and alternative therapies for diabetes.
Collapse
Affiliation(s)
- Prawej Ansari
- Department of Pharmacy, Independent University, Dhaka 1229, Bangladesh; (S.A.); (J.M.A.H.); (N.J.N.)
- School of Biomedical Sciences, Ulster University, Coleraine BT52 1SA, UK;
- Correspondence: ; Tel.: +880-1323-879720
| | - Samia Akther
- Department of Pharmacy, Independent University, Dhaka 1229, Bangladesh; (S.A.); (J.M.A.H.); (N.J.N.)
| | - J. M. A. Hannan
- Department of Pharmacy, Independent University, Dhaka 1229, Bangladesh; (S.A.); (J.M.A.H.); (N.J.N.)
| | - Veronique Seidel
- Natural Products Research Laboratory, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK;
| | - Nusrat Jahan Nujat
- Department of Pharmacy, Independent University, Dhaka 1229, Bangladesh; (S.A.); (J.M.A.H.); (N.J.N.)
| | | |
Collapse
|
24
|
Lawal AO, Folorunso IM, Iwaloye O. Morin hydrate protects type-2-diabetic wistar rats exposed to diesel exhaust particles from inflammation and oxidative stress. J Diabetes Metab Disord 2022; 21:805-816. [PMID: 35673443 PMCID: PMC9167336 DOI: 10.1007/s40200-022-01057-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 05/10/2022] [Indexed: 11/24/2022]
Abstract
Background Studies have demonstrated that exposure to diesel exhaust particle (DEP) aggravates diabetes condition by inducing oxidative and pro-inflammatory effects. Morin hydrate (MH), a flavonol found in common guava, among others has been demonstrated to possess a variety of biological activities. The present study was designed to investigate the effects of morin hydrate (MH) on the pancreas of type-2 diabetic (T2D) wistar rats exposed to DEP. Methods Rats were induced with type 2 diabetes by oral fructose therapy for 14 days followed by injection of streptozotocin (45 mg/kg). These rats were pre-treated with DEP (0.4 mg/kg and 0.5 mg/kg) through nasal instillation prior to receiving oral MH (30 mg/kg).This study determined oxidative stress parameters using biochemical assay, and some pancreatic genes involved in oxidative stress, inflammation and glucose uptake were quantified using RT-polymerase chain reaction (PCR). Results The results indicate that MH reverses oxidative stress in T2D rats exposed to DEP via substantial increase in superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx) activity and reduced glutathione (GSH) levels, but a decrease in malondialdehyde (MDA) and conjugated diene (CD) levels. Moreover, PCR assay showed that MH mitigate inflammation and oxidative stress but promote glucose uptake by increasing the mRNA expression of IL-10, HO-1, and GLUT 4; decreasing mRNA expression of IL-1 and modulating AKT/PI3K/GLUT4 and AMPK/GLUT4 signaling. Histopathological examination revealed that MH reverses DEP induced pancreatic fibrosis and necrosis. Conclusion The results suggest that MH alleviate inflammation and oxidative stress and promote glucose uptake in the pancreas of type-2 diabetic rats, either in the presence or absence of DEP.
Collapse
Affiliation(s)
- Akeem O. Lawal
- Bioinformatics and Molecular Biology Unit, Department of Biochemistry, School of Sciences, Federal University of Technology, P.M.B. 704, Akure, Ondo-State Nigeria
| | - Ibukun M. Folorunso
- Bioinformatics and Molecular Biology Unit, Department of Biochemistry, School of Sciences, Federal University of Technology, P.M.B. 704, Akure, Ondo-State Nigeria
| | - Opeyemi Iwaloye
- Bioinformatics and Molecular Biology Unit, Department of Biochemistry, School of Sciences, Federal University of Technology, P.M.B. 704, Akure, Ondo-State Nigeria
| |
Collapse
|
25
|
Kim J, Son J, Ahn D, Nam G, Zhao X, Park H, Jeong W, Chung SJ. Structure-Activity Relationship of Synthetic Ginkgolic Acid Analogs for Treating Type 2 Diabetes by PTPN9 Inhibition. Int J Mol Sci 2022; 23:ijms23073927. [PMID: 35409287 PMCID: PMC8999917 DOI: 10.3390/ijms23073927] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 02/01/2023] Open
Abstract
Ginkgolic acid (C13:0) (GA), isolated from Ginkgo biloba, is a potential therapeutic agent for type 2 diabetes. A series of GA analogs were designed and synthesized for the evaluation of their structure–activity relationship with respect to their antidiabetic effects. Unlike GA, the synthetic analog 1e exhibited improved inhibitory activity against PTPN9 and significantly stimulated glucose uptake via AMPK phosphorylation in differentiated 3T3-L1 adipocytes and C2C12 myotubes; it also induced insulin-dependent AKT activation in C2C12 myotubes in a concentration-dependent manner. Docking simulation results showed that 1e had a better binding affinity through a unique hydrophobic interaction with a PTPN9 hydrophobic groove. Moreover, 1e ameliorated palmitate-induced insulin resistance in C2C12 cells. This study showed that 1e increases glucose uptake and suppresses palmitate-induced insulin resistance in C2C12 myotubes via PTPN9 inhibition; thus, it is a promising therapeutic candidate for treating type 2 diabetes.
Collapse
Affiliation(s)
- Jinsoo Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea; (J.K.); (D.A.); (G.N.); (X.Z.)
| | - Jinyoung Son
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Korea; (J.S.); (H.P.); (W.J.)
| | - Dohee Ahn
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea; (J.K.); (D.A.); (G.N.); (X.Z.)
| | - Gibeom Nam
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea; (J.K.); (D.A.); (G.N.); (X.Z.)
| | - Xiaodi Zhao
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea; (J.K.); (D.A.); (G.N.); (X.Z.)
| | - Hyuna Park
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Korea; (J.S.); (H.P.); (W.J.)
| | - Woojoo Jeong
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Korea; (J.S.); (H.P.); (W.J.)
| | - Sang J. Chung
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea; (J.K.); (D.A.); (G.N.); (X.Z.)
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Korea; (J.S.); (H.P.); (W.J.)
- Correspondence:
| |
Collapse
|
26
|
de Sousa Neto IV, da Cunha Nascimento D, Prestes J, da Fonseca EF, Celes RS, Rolnick N, de Sousa Barbalho YG, Silva ADO, Stival MM, de Lima LR, Funghetto SS. Initial Muscle Quality Affects Individual Responsiveness of Interleukin-6 and Creatine Kinase following Acute Eccentric Exercise in Sedentary Obese Older Women. BIOLOGY 2022; 11:biology11040537. [PMID: 35453736 PMCID: PMC9026080 DOI: 10.3390/biology11040537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/09/2022] [Accepted: 03/11/2022] [Indexed: 11/18/2022]
Abstract
This study aimed to evaluate the time course and responsiveness of plasma interleukin-6 (IL-6) and creatine kinase (CK) levels following acute eccentric resistance exercise in sedentary obese older women with a different muscle quality index (MQI). Eighty-eight participants (69.4 ± 6.06 years) completed an acute eccentric resistance exercise (7 sets of 10 repetitions at 110% of 10-repetition maximum with 3 min rest interval). Participants were divided into two groups: high or low MQI according to 50th percentile cut-off. The responsiveness was based on minimal clinical important difference. There were no differences between groups and time on IL-6 and CK levels (p > 0.05). However, the high MQI group displayed a lower proportion of low responders (1 for laboratory and 2 for field-based vs. 5 and 4) and a higher proportion of high responders for IL-6 (7 for laboratory and 6 for field-based vs. 4 and 5) compared to low MQI group. In addition, the high MQI group showed a higher proportion of high responders for CK (11 for laboratory and 9 for field-based vs. 6 and 6) compared to low MQI. A prior MQI screening can provide feedback to understand the magnitude response. Individual responsiveness should be taken into consideration for maximizing eccentric exercise prescription.
Collapse
Affiliation(s)
- Ivo Vieira de Sousa Neto
- Laboratory of Molecular Analysis, Faculty of Ceilândia, Universidade de Brasília, Brasilia 70910-900, Brazil
- Graduate Program of Sciences and Technology of Health, Faculty of Ceilândia, Universidade de Brasília, Brasilia 70910-900, Brazil; (Y.G.d.S.B.); (M.M.S.); (S.S.F.)
- Correspondence:
| | - Dahan da Cunha Nascimento
- Graduate Program of Physical Education, Catholic University of Brasilia, Brasilia 70910-900, Brazil; (D.d.C.N.); (J.P.); (E.F.d.F.); (R.S.C.)
| | - Jonato Prestes
- Graduate Program of Physical Education, Catholic University of Brasilia, Brasilia 70910-900, Brazil; (D.d.C.N.); (J.P.); (E.F.d.F.); (R.S.C.)
| | - Eduardo Fernandes da Fonseca
- Graduate Program of Physical Education, Catholic University of Brasilia, Brasilia 70910-900, Brazil; (D.d.C.N.); (J.P.); (E.F.d.F.); (R.S.C.)
| | - Rodrigo Souza Celes
- Graduate Program of Physical Education, Catholic University of Brasilia, Brasilia 70910-900, Brazil; (D.d.C.N.); (J.P.); (E.F.d.F.); (R.S.C.)
| | - Nicholas Rolnick
- The Human Performance Mechanic, Lehman College, New York, NY 10468, USA;
| | - Yuri Gustavo de Sousa Barbalho
- Graduate Program of Sciences and Technology of Health, Faculty of Ceilândia, Universidade de Brasília, Brasilia 70910-900, Brazil; (Y.G.d.S.B.); (M.M.S.); (S.S.F.)
| | | | - Marina Morato Stival
- Graduate Program of Sciences and Technology of Health, Faculty of Ceilândia, Universidade de Brasília, Brasilia 70910-900, Brazil; (Y.G.d.S.B.); (M.M.S.); (S.S.F.)
- Department of Nursing, Faculty of Ceilândia, Universidade de Brasília, Brasilia 70910-900, Brazil;
| | - Luciano Ramos de Lima
- Department of Nursing, Faculty of Ceilândia, Universidade de Brasília, Brasilia 70910-900, Brazil;
| | - Silvana Schwerz Funghetto
- Graduate Program of Sciences and Technology of Health, Faculty of Ceilândia, Universidade de Brasília, Brasilia 70910-900, Brazil; (Y.G.d.S.B.); (M.M.S.); (S.S.F.)
- Department of Nursing, Faculty of Ceilândia, Universidade de Brasília, Brasilia 70910-900, Brazil;
| |
Collapse
|
27
|
de la Cruz-Ojeda P, Flores-Campos R, Navarro-Villarán E, Muntané J. The Role of Non-Coding RNAs in Autophagy During Carcinogenesis. Front Cell Dev Biol 2022; 10:799392. [PMID: 35309939 PMCID: PMC8926078 DOI: 10.3389/fcell.2022.799392] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 01/18/2022] [Indexed: 12/12/2022] Open
Abstract
Macroautophagy (autophagy herein) is a cellular stress response and a survival pathway involved in self-renewal and quality control processes to maintain cellular homeostasis. The alteration of autophagy has been implicated in numerous diseases such as cancer where it plays a dual role. Autophagy serves as a tumor suppressor in the early phases of cancer formation with the restoration of homeostasis and eliminating cellular altered constituents, yet in later phases, autophagy may support and/or facilitate tumor growth, metastasis and may contribute to treatment resistance. Key components of autophagy interact with either pro- and anti-apoptotic factors regulating the proximity of tumor cells to apoptotic cliff promoting cell survival. Autophagy is regulated by key cell signaling pathways such as Akt (protein kinase B, PKB), mammalian target of rapamycin (mTOR) and AMP-activated protein kinase (AMPK) involved in cell survival and metabolism. The expression of critical members of upstream cell signaling, as well as those directly involved in the autophagic and apoptotic machineries are regulated by microRNAs (miRNAs) and long non-coding RNAs (lncRNAs). Consequently, non-coding RNAs play a relevant role in carcinogenesis and treatment response in cancer. The review is an update of the current knowledge in the regulation by miRNA and lncRNA of the autophagic components and their functional impact to provide an integrated and comprehensive regulatory network of autophagy in cancer.
Collapse
Affiliation(s)
- Patricia de la Cruz-Ojeda
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain.,Department of Medical Physiology and Biophysics, University of Seville, Seville, Spain.,Networked Biomedical Research Center Hepatic and Digestive Diseases (CIBEREHD o Ciberehd), Institute of Health Carlos III, Madrid, Spain
| | - Rocío Flores-Campos
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
| | - Elena Navarro-Villarán
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain.,Department of Medical Physiology and Biophysics, University of Seville, Seville, Spain.,Networked Biomedical Research Center Hepatic and Digestive Diseases (CIBEREHD o Ciberehd), Institute of Health Carlos III, Madrid, Spain
| | - Jordi Muntané
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain.,Department of Medical Physiology and Biophysics, University of Seville, Seville, Spain.,Networked Biomedical Research Center Hepatic and Digestive Diseases (CIBEREHD o Ciberehd), Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
28
|
The WWOX/HIF1A Axis Downregulation Alters Glucose Metabolism and Predispose to Metabolic Disorders. Int J Mol Sci 2022; 23:ijms23063326. [PMID: 35328751 PMCID: PMC8955937 DOI: 10.3390/ijms23063326] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/10/2022] [Accepted: 03/17/2022] [Indexed: 02/01/2023] Open
Abstract
Recent reports indicate that the hypoxia-induced factor (HIF1α) and the Warburg effect play an initiating role in glucotoxicity, which underlies disorders in metabolic diseases. WWOX has been identified as a HIF1α regulator. WWOX downregulation leads to an increased expression of HIF1α target genes encoding glucose transporters and glycolysis’ enzymes. It has been proven in the normoglycemic mice cells and in gestational diabetes patients. The aim of the study was to determine WWOX’s role in glucose metabolism regulation in hyperglycemia and hypoxia to confirm its importance in the development of metabolic disorders. For this purpose, the WWOX gene was silenced in human normal fibroblasts, and then cells were cultured under different sugar and oxygen levels. Thereafter, it was investigated how WWOX silencing alters the genes and proteins expression profile of glucose transporters and glycolysis pathway enzymes, and their activity. In normoxia normoglycemia, higher glycolysis genes expression, their activity, and the lactate concentration were observed in WWOX KO fibroblasts in comparison to control cells. In normoxia hyperglycemia, it was observed a decrease of insulin-dependent glucose uptake and a further increase of lactate. It likely intensifies hyperglycemia condition, which deepen the glucose toxic effect. Then, in hypoxia hyperglycemia, WWOX KO caused weaker glucose uptake and elevated lactate production. In conclusion, the WWOX/HIF1A axis downregulation alters glucose metabolism and probably predispose to metabolic disorders.
Collapse
|
29
|
Grunwald SA, Haafke S, Grieben U, Kassner U, Steinhagen-Thiessen E, Spuler S. Statins Aggravate the Risk of Insulin Resistance in Human Muscle. Int J Mol Sci 2022; 23:2398. [PMID: 35216514 PMCID: PMC8876152 DOI: 10.3390/ijms23042398] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 02/13/2022] [Accepted: 02/17/2022] [Indexed: 01/23/2023] Open
Abstract
Beside their beneficial effects on cardiovascular events, statins are thought to contribute to insulin resistance and type-2 diabetes. It is not known whether these effects are long-term events from statin-treatment or already triggered with the first statin-intake. Skeletal muscle is considered the main site for insulin-stimulated glucose uptake and therefore, a primary target for insulin resistance in the human body. We analyzed localization and expression of proteins related to GLUT4 mediated glucose uptake via AMPKα or AKT in human skeletal muscle tissue from patients with statin-intake >6 months and in primary human myotubes after 96 h statin treatment. The ratio for AMPKα activity significantly increased in human skeletal muscle cells treated with statins for long- and short-term. Furthermore, the insulin-stimulated counterpart, AKT, significantly decreased in activity and protein level, while GSK3ß and mTOR protein expression reduced in statin-treated primary human myotubes, only. However, GLUT4 was normally distributed whereas CAV3 was internalized from plasma membrane around the nucleus in statin-treated primary human myotubes. Statin-treatment activates AMPKα-dependent glucose uptake and remains active after long-term statin treatment. Permanent blocking of its insulin-dependent counterpart AKT activation may lead to metabolic inflexibility and insulin resistance in the long run and may be a direct consequence of statin-treatment.
Collapse
Affiliation(s)
- Stefanie A. Grunwald
- Muscle Research Unit, Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13125 Berlin, Germany; (S.H.); (U.G.)
| | - Stefanie Haafke
- Muscle Research Unit, Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13125 Berlin, Germany; (S.H.); (U.G.)
| | - Ulrike Grieben
- Muscle Research Unit, Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13125 Berlin, Germany; (S.H.); (U.G.)
| | - Ursula Kassner
- Interdisciplinary Lipid Metabolic Center, Charité Universitätsmedizin Berlin, 13353 Berlin, Germany; (U.K.); (E.S.-T.)
| | - Elisabeth Steinhagen-Thiessen
- Interdisciplinary Lipid Metabolic Center, Charité Universitätsmedizin Berlin, 13353 Berlin, Germany; (U.K.); (E.S.-T.)
| | - Simone Spuler
- Muscle Research Unit, Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13125 Berlin, Germany; (S.H.); (U.G.)
| |
Collapse
|
30
|
Knudsen JR, Madsen AB, Li Z, Andersen NR, Schjerling P, Jensen TE. Gene deletion of γ-actin impairs insulin-stimulated skeletal muscle glucose uptake in growing mice but not in mature adult mice. Physiol Rep 2022; 10:e15183. [PMID: 35224890 PMCID: PMC8882697 DOI: 10.14814/phy2.15183] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 04/14/2023] Open
Abstract
The cortical cytoskeleton, consisting of the cytoplasmic actin isoforms β and/or γ-actin, has been implicated in insulin-stimulated GLUT4 translocation and glucose uptake in muscle and adipose cell culture. Furthermore, transgenic inhibition of multiple actin-regulating proteins in muscle inhibits insulin-stimulated muscle glucose uptake. The current study tested if γ-actin was required for insulin-stimulated glucose uptake in mouse skeletal muscle. Based on our previously reported age-dependent phenotype in muscle-specific β-actin gene deletion (-/- ) mice, we included cohorts of growing 8-14 weeks old and mature 18-32 weeks old muscle-specific γ-actin-/- mice or wild-type littermates. In growing mice, insulin significantly increased the glucose uptake in slow-twitch oxidative soleus and fast-twitch glycolytic EDL muscles from wild-type mice, but not γ-actin-/- . In relative values, the maximal insulin-stimulated glucose uptake was reduced by ~50% in soleus and by ~70% in EDL muscles from growing γ-actin-/- mice compared to growing wild-type mice. In contrast, the insulin-stimulated glucose uptake responses in mature adult γ-actin-/- soleus and EDL muscles were indistinguishable from the responses in wild-type muscles. Mature adult insulin-stimulated phosphorylations on Akt, p70S6K, and ULK1 were not significantly affected by genotype. Hence, insulin-stimulated muscle glucose uptake shows an age-dependent impairment in young growing but not in fully grown γ-actin-/- mice, bearing phenotypic resemblance to β-actin-/- mice. Overall, γ-actin does not appear required for insulin-stimulated muscle glucose uptake in adulthood. Furthermore, our data emphasize the need to consider the rapid growth of young mice as a potential confounder in transgenic mouse phenotyping studies.
Collapse
Affiliation(s)
- Jonas R. Knudsen
- Section for Molecular PhysiologyDepartment of Nutrition, Exercise and SportsUniversity of CopenhagenCopenhagenDenmark
| | - Agnete B. Madsen
- Section for Molecular PhysiologyDepartment of Nutrition, Exercise and SportsUniversity of CopenhagenCopenhagenDenmark
| | - Zhencheng Li
- Section for Molecular PhysiologyDepartment of Nutrition, Exercise and SportsUniversity of CopenhagenCopenhagenDenmark
| | - Nicoline R. Andersen
- Section for Molecular PhysiologyDepartment of Nutrition, Exercise and SportsUniversity of CopenhagenCopenhagenDenmark
| | - Peter Schjerling
- Department of Orthopedic Surgery MInstitute of Sports Medicine CopenhagenBispebjerg HospitalCopenhagenDenmark
| | - Thomas E. Jensen
- Section for Molecular PhysiologyDepartment of Nutrition, Exercise and SportsUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
31
|
Wu JW, Hu H, Hua JS, Ma LK. ATPase inhibitory factor 1 protects the heart from acute myocardial ischemia/reperfusion injury through activating AMPK signaling pathway. Int J Biol Sci 2022; 18:731-741. [PMID: 35002521 PMCID: PMC8741848 DOI: 10.7150/ijbs.64956] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 11/24/2021] [Indexed: 11/23/2022] Open
Abstract
Rationale: Myocardial ischemia/reperfusion (I/R) injury is a common clinic scenario that occurs in the context of reperfusion therapy for acute myocardial infarction (AMI). The mitochondrial F1Fo-ATPase inhibitory factor 1 (IF1) blocks the reversal of the F1Fo-ATP synthase to prevent detrimental consumption of cellular ATP and associated demise. In the present study, we study the role and mechanism of IF1 in myocardial I/R injury. Methods: Mice were ligated the left anterior descending coronary artery to build the I/R model in vivo. Rat hearts were isolated and perfused with constant pressure according to Langendorff. Also, neonatal cardiomyocytes hypoxia-reoxygenation (H/R) model was also used. Myocardial infarction area, cardiac function, cellular function, and cell viability was conducted and compared. Results: Our data revealed that IF1 is upregulated in hearts after I/R and cardiomyocytes with hypoxia/re-oxygenation (H/R). IF1 delivered with adenovirus and adeno-associated virus serotype 9 (AAV9) ameliorated cardiac dysfunction and pathological development induced by I/R ex vivo and in vivo. Mechanistically, IF1 stimulates glucose uptake and glycolysis activity and stimulates AMPK activation during in vivo basal and I/R and in vitro OGD/R conditions, and activation of AMPK by IF1 is responsible for its cardioprotective effects against H/R-induced injury. Conclusions: These results suggest that increased IF1 in the I/R heart confer cardioprotective effects via activating AMPK signaling. Therefore, IF1 can be used as a potential therapeutic target for the treatment of pathological ischemic injury and heart failure.
Collapse
Affiliation(s)
- Jia-Wei Wu
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Hao Hu
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Jin-Sheng Hua
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Li-Kun Ma
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, 230001, China
| |
Collapse
|
32
|
Wang H, Zheng A, Arias EB, Cartee GD. Prior AICAR induces elevated glucose uptake concomitant with greater γ3-AMPK activation and reduced membrane cholesterol in skeletal muscle from 26-month-old rats. Facets (Ott) 2022; 7:774-791. [PMID: 36381195 PMCID: PMC9648397 DOI: 10.1139/facets-2021-0166] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2024] Open
Abstract
Attenuated skeletal muscle glucose uptake (GU) has been observed with advancing age. It is important to elucidate the mechanisms linked to interventions that oppose this detrimental outcome. Earlier research using young rodents and (or) cultured myocytes reported that treatment with 5-aminoimidazole-4-carboxamide-1-β-d-ribofuranoside (AICAR; an AMP-activated protein kinase (AMPK) activator) can increase γ3-AMPK activity and reduce membrane cholesterol content, each of which has been proposed to elevate GU. However, the effect of AICAR treatment on γ3-AMPK activity and membrane cholesterol in skeletal muscle of aged animals has not been reported. Our purpose was to evaluate the effects of AICAR treatment on these potential mechanisms for enhanced glucose uptake in the skeletal muscle of aged animals. Epitrochlearis muscles from 26-27-month-old male rats were isolated and incubated ± AICAR, followed by 3 h incubation without AICAR, and then incubation with 3-O-methyl-[3 H] glucose (to assess GU ± insulin). Muscles were also analyzed for γ3-AMPK activity and membrane cholesterol content. Prior AICAR treatment led to increased γ3-AMPK activity, reduced membrane cholesterol content, and enhanced glucose uptake in skeletal muscle from aged rats. These observations revealed that two potential mechanisms for greater GU previously observed in younger animals and (or) cell models are also potentially relevant for enhanced GU by muscles from older animals.
Collapse
Affiliation(s)
- Haiyan Wang
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Amy Zheng
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Edward B. Arias
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Gregory D. Cartee
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
- Institute of Gerontology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
33
|
Shukal DK, Malaviya PB, Sharma T. Role of the AMPK signalling pathway in the aetiopathogenesis of ocular diseases. Hum Exp Toxicol 2022; 41:9603271211063165. [PMID: 35196887 DOI: 10.1177/09603271211063165] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND AMP-activated protein kinase (AMPK) plays a precise role as a master regulator of cellular energy homeostasis. AMPK is activated in response to the signalling cues that exhaust cellular ATP levels such as hypoxia, ischaemia, glucose depletion and heat shock. As a central regulator of both lipid and glucose metabolism, AMPK is considered to be a potential therapeutic target for the treatment of various diseases, including eye disorders. OBJECTIVE To review all the shreds of evidence concerning the role of the AMPK signalling pathway in the pathogenesis of ocular diseases. METHOD Scientific data search and review of available information evaluating the influence of AMPK signalling on ocular diseases. RESULTS Review highlights the significance of AMPK signalling in the aetiopathogenesis of ocular diseases, including cataract, glaucoma, diabetic retinopathy, retinoblastoma, age-related macular degeneration, corneal diseases, etc. The review also provides the information on the AMPK-associated pathways with reference to ocular disease, which includes mitochondrial biogenesis, autophagy and regulation of inflammatory response. CONCLUSION The study concludes the role of AMPK in ocular diseases. There is growing interest in the therapeutic utilization of the AMPK pathway for ocular disease treatment. Furthermore, inhibition of AMPK signalling might represent more pertinent strategy than AMPK activation for ocular disease treatment. Such information will guide the development of more effective AMPK modulators for ocular diseases.[Formula: see text].
Collapse
Affiliation(s)
- Dhaval K Shukal
- 534329Iladevi Cataract and IOL Research Centre, Memnagar, Ahmedabad, Gujarat, India.,76793Manipal Academy of Higher Education, Mangalore, Karnataka, India
| | - Pooja B Malaviya
- 534329Iladevi Cataract and IOL Research Centre, Memnagar, Ahmedabad, Gujarat, India.,76793Manipal Academy of Higher Education, Mangalore, Karnataka, India
| | - Tusha Sharma
- 534329Iladevi Cataract and IOL Research Centre, Memnagar, Ahmedabad, Gujarat, India
| |
Collapse
|
34
|
Garlapati C, Joshi S, Turaga RC, Mishra M, Reid MD, Kapoor S, Artinian L, Rehder V, Aneja R. Monoethanolamine-induced glucose deprivation promotes apoptosis through metabolic rewiring in prostate cancer. Am J Cancer Res 2021; 11:9089-9106. [PMID: 34522228 PMCID: PMC8419048 DOI: 10.7150/thno.62724] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 08/13/2021] [Indexed: 11/16/2022] Open
Abstract
Rationale: Cancer cells rely on glucose metabolism for fulfilling their high energy demands. We previously reported that monoethanolamine (Etn), an orally deliverable lipid formulation, reduced intracellular glucose and glutamine levels in prostate cancer (PCa). Glucose deprivation upon Etn treatment exacerbated metabolic stress in PCa, thereby enhancing cell death. Moreover, Etn was potent in inhibiting tumor growth in a PCa xenograft model. However, the precise mechanisms underlying Etn-induced metabolic stress in PCa remain elusive. The purpose of the present study was to elucidate the mechanisms contributing to Etn-mediated metabolic rewiring in PCa. Methods: Glucose transporters (GLUTs) facilitate glucose transport across the plasma membrane. Thus, we assessed the expression of GLUTs and the internalization of GLUT1 in PCa. We also evaluated the effects of Etn on membrane dynamics, mitochondrial structure and function, lipid droplet density, autophagy, and apoptosis in PCa cells. Results: Compared to other GLUTs, GLUT1 was highly upregulated in PCa. We observed enhanced GLUT1 internalization, altered membrane dynamics, and perturbed mitochondrial structure and function upon Etn treatment. Etn-induced bioenergetic stress enhanced lipolysis, decreased lipid droplet density, promoted accumulation of autophagosomes, and increased apoptosis. Conclusion: We provide the first evidence that Etn alters GLUT1 trafficking leading to metabolic stress in PCa. By upregulating phosphatidylethanolamine (PE), Etn modulates membrane fluidity and affects mitochondrial structure and function. Etn also induces autophagy in PCa cells, thereby promoting apoptosis. These data strongly suggest that Etn rewires cellular bioenergetics and could serve as a promising anticancer agent for PCa.
Collapse
|
35
|
Papain Ameliorates Lipid Accumulation and Inflammation in High-Fat Diet-Induced Obesity Mice and 3T3-L1 Adipocytes via AMPK Activation. Int J Mol Sci 2021; 22:ijms22189885. [PMID: 34576066 PMCID: PMC8468764 DOI: 10.3390/ijms22189885] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/08/2021] [Accepted: 09/10/2021] [Indexed: 12/19/2022] Open
Abstract
Papain is a proteolytic enzyme present in the leaves, fruits, roots, and latex of the Carica papaya (papaya) plant. Although it exhibits a wide range of activities, there are no reports on the anti-obesity effects of papain. This study examined the anti-obesity effect and obesity-involved anti-inflammatory mechanism of papain in in vivo and in vitro models using high-fat diet (HFD)-induced obese mice and 3T3-L1 preadipocytes. Oral administration of papain reduced HFD-induced weight of the body, liver, and adipose tissues of mice. Papain also reduced hepatic lipid accumulation and adipocyte size. Moreover, serum total cholesterol and triglyceride levels were markedly reduced in papain-treated mice. In addition, papain inhibited the differentiation of preadipocytes and oil accumulation in 3T3-L1 preadipocytes and rat primary preadipocytes. Mechanistically, papain significantly downregulated the protein levels of key adipogenesis regulators and reversed the expression of pro-inflammatory cytokines and adipokines in HFD-induced obese mice and 3T3-L1 preadipocytes. Papain also markedly enhanced activation of the AMP-activated protein kinase pathway in both models. Collectively, these results suggest that papain exerts anti-obesity effects in HFD-induced mice and 3T3-L1 preadipocytes by regulating levels of adipogenic factors involved in lipid metabolism and inflammation; thus, it could be useful in the prevention and treatment of obesity.
Collapse
|
36
|
Chen Y, Xu W, Zhang Q, Zhang Y, Mu R. Intraperitoneal injection of genistein affects the distribution and metabolism of cholesterol in female yellow catfish Tachysurus fulvidraco. FISH PHYSIOLOGY AND BIOCHEMISTRY 2021; 47:1299-1311. [PMID: 34241762 DOI: 10.1007/s10695-021-00985-y] [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: 02/15/2021] [Accepted: 07/01/2021] [Indexed: 06/13/2023]
Abstract
Genistein is an abundant phytoestrogen in soybean. This study aimed to determine the effects of genistein on cholesterol distribution and metabolism in female yellow catfish. Three hundred fish (49.2 ± 1.4 g) were randomly divided into five treatments and received intraperitoneal injections as follows: (1) blank, no injection; (2) control, vehicle only; (3) E2, 17β-estradiol at 10 μg·g-1 body weight; (4) low genistein doses, genistein at 10 μg·g-1 body weight; (5) high genistein doses, genistein at 100 μg·g-1 body weight. Both high and low genistein doses significantly reduced (p < 0.05) serum TC and LDL-C 24 h after injection. Moreover, the high genistein doses significantly reduced (p < 0.05) serum HDL-C. Both high and low doses of genistein significantly increased (p < 0.05) hepatic TC. Only high genistein doses significantly increased (p < 0.05) ovary TC. In the liver, both high and low genistein doses significantly increased (p < 0.05) protein and mRNA expression of ldlr. Meanwhile, high genistein doses significantly decreased (p < 0.05) mRNA expression of hmgcr. In ovary tissue, high genistein doses significantly decreased (p < 0.05) mRNA expression of cyp11a1. These results suggested that genistein affected the cholesterol distribution in female yellow catfish. Both high and low doses of genistein reduced cholesterol content in blood and increased its content in the liver by increasing the uptake of blood cholesterol. Meanwhile, high genistein doses may inhibit hepatic cholesterol synthesis. Additionally, high genistein doses could increase cholesterol transfer from serum into the ovary and disturb cholesterol conversion to pregnenolone.
Collapse
Affiliation(s)
- Yushi Chen
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
- School of Marine Sciences, Ningbo University, Ningbo, 315211, China
| | - Wenbin Xu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China.
- School of Marine Sciences, Ningbo University, Ningbo, 315211, China.
| | - Qingji Zhang
- School of Marine Sciences, Ningbo University, Ningbo, 315211, China
| | - Yilin Zhang
- School of Marine Sciences, Ningbo University, Ningbo, 315211, China
| | - Ren Mu
- College of Biological Science and Agriculture, Qiannan Normal University for Nationalities, Longshan Avenue, Duyun, 558000, Guizhou Province, China.
| |
Collapse
|
37
|
Masson SWC, Woodhead JST, D'Souza RF, Broome SC, MacRae C, Cho HC, Atiola RD, Futi T, Dent JR, Shepherd PR, Merry TL. β-Catenin is required for optimal exercise- and contraction-stimulated skeletal muscle glucose uptake. J Physiol 2021; 599:3897-3912. [PMID: 34180063 DOI: 10.1113/jp281352] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 06/22/2021] [Indexed: 01/14/2023] Open
Abstract
KEY POINTS Loss of β-catenin impairs in vivo and isolated muscle exercise/contraction-stimulated glucose uptake. β-Catenin is required for exercise-induced skeletal muscle actin cytoskeleton remodelling. β-Catenin675 phosphorylation during exercise may be intensity dependent. ABSTRACT The conserved structural protein β-catenin is an emerging regulator of vesicle trafficking in multiple tissues and supports insulin-stimulated glucose transporter 4 (GLUT4) translocation in skeletal muscle by facilitating cortical actin remodelling. Actin remodelling may be a convergence point between insulin and exercise/contraction-stimulated glucose uptake. Here we investigated whether β-catenin is involved in regulating exercise/contraction-stimulated glucose uptake. We report that the muscle-specific deletion of β-catenin induced in adult mice (BCAT-mKO) impairs both exercise- and contraction (isolated muscle)-induced glucose uptake without affecting running performance or canonical exercise signalling pathways. Furthermore, high intensity exercise in mice and contraction of myotubes and isolated muscles led to the phosphorylation of β-cateninS675 , and this was impaired by Rac1 inhibition. Moderate intensity exercise in control and Rac1 muscle-specific knockout mice did not induce muscle β-cateninS675 phosphorylation, suggesting exercise intensity-dependent regulation of β-cateninS675 . Introduction of a non-phosphorylatable S675A mutant of β-catenin into myoblasts impaired GLUT4 translocation and actin remodelling stimulated by carbachol, a Rac1 and RhoA activator. Exercise-induced increases in cross-sectional phalloidin staining (F-actin marker) of gastrocnemius muscle was impaired in muscle from BCAT-mKO mice. Collectively our findings suggest that β-catenin is required for optimal glucose transport in muscle during exercise/contraction, potentially via facilitating actin cytoskeleton remodelling.
Collapse
Affiliation(s)
- Stewart W C Masson
- Discipline of Nutrition, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Jonathan S T Woodhead
- Discipline of Nutrition, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Randall F D'Souza
- Discipline of Nutrition, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Sophie C Broome
- Discipline of Nutrition, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Caitlin MacRae
- Discipline of Nutrition, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Hyun C Cho
- Discipline of Nutrition, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Robert D Atiola
- Discipline of Nutrition, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Tumanu Futi
- Discipline of Nutrition, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Jessica R Dent
- Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Peter R Shepherd
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand.,Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Troy L Merry
- Discipline of Nutrition, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| |
Collapse
|
38
|
Hadi V, Pahlavani N, Malekahmadi M, Nattagh-Eshtivani E, Navashenaq JG, Hadi S, Ferns GA, Ghayour-Mobarhan M, Askari G, Norouzy A. Nigella sativa in controlling Type 2 diabetes, cardiovascular, and rheumatoid arthritis diseases: Molecular aspects. JOURNAL OF RESEARCH IN MEDICAL SCIENCES 2021; 26:20. [PMID: 34221050 PMCID: PMC8240544 DOI: 10.4103/jrms.jrms_236_20] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/08/2020] [Accepted: 08/27/2020] [Indexed: 11/30/2022]
Abstract
Oxidative stress is an important factor in the etiology of several chronic diseases that include cardiovascular disease (CVD), Type 2 diabetes (T2D), and rheumatoid arthritis (RA). Oxidative stress can lead to inflammation, and this can contribute to these chronic diseases. Reducing inflammation and oxidative stress may, therefore, be useful in the prevention and treatment of these conditions. One of the treatment options for chronic diseases is the use of traditional medicine and herbs, such as Nigella sativa. This is one of the herbs that have recently been assessed for its ability to reduce inflammation and oxidative stress. We have reviewed the reported effects of N. sativa on risk factors of chronic diseases (CVD, DM, and RA) with emphasis on molecular and cellular mechanisms in controlling inflammation and oxidative stress. Various mechanisms have been proposed to contribute to the beneficial properties of N. sativa, including a reduction of lipid peroxidation via its antioxidant properties; agonist of peroxisome proliferator-activated receptor gamma in adipose tissue; activation of AMP-activated protein kinase, increased antioxidants, inhibition of nuclear factor-kappa B pathway; increased in interleukin-10 expression, CD4+ T-cell percentage, T regulatory cell percentage (CD4+ CD25+ T-cell) in peripheral blood, and CD4+/CD8+ ratio, but to prove this claim, it is necessary to conduct experimental and well-designed clinical trial studies with a larger sample size on the effects of N. sativa on these chronic diseases.
Collapse
Affiliation(s)
- Vahid Hadi
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Health, AJA University of Medical Sciences, Tehran, Iran
| | - Naseh Pahlavani
- Social Development and Health Promotion Research Center, Gonabad University of Medical Sciences, Gonabad, Iran.,Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Mahsa Malekahmadi
- Research Center for Gastroenterology and Liver Disease, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elyas Nattagh-Eshtivani
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Jamshid Gholizadeh Navashenaq
- Noncommunicable Diseases Research Center, Bam University of Medical Sciences, Bam, Iran.,Department of Immunology, School of Medicine, Bam University of Medical Sciences, Bam, Iran
| | - Saeid Hadi
- Department of Health, AJA University of Medical Sciences, Tehran, Iran.,Department of Community Nutrition, School of Nutrition and Food Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Gordon A Ferns
- Division of Medical Education, Brighton and Sussex Medical School, Brighton, Sussex, England
| | - Majid Ghayour-Mobarhan
- Cardiovascular Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Nutrition, International UNESCO Center for Health Related Basic Sciences and Human Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gholamreza Askari
- Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Abdolreza Norouzy
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
39
|
Wang H, Arias EB, Cartee GD. Reduced membrane cholesterol content in skeletal muscle is not essential for greater insulin-stimulated glucose uptake after acute exercise by rats. Appl Physiol Nutr Metab 2021; 46:685-689. [PMID: 33765397 DOI: 10.1139/apnm-2021-0130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
One exercise session can elevate insulin-stimulated glucose uptake (GU) by skeletal muscle, but it is uncertain if this effect is accompanied by altered membrane cholesterol content, which is reportedly inversely related to insulin-stimulated GU. Muscles from sedentary (SED) or exercised 3 h post-exercise (3hPEX) rats were evaluated for GU, membrane cholesterol, and phosphorylation of cholesterol regulatory proteins (pHMCGRSer872 and pABCA1Ser2054). Insulin-stimulated GU for 3hPEX exceeded SED. Membrane cholesterol, pHMCGRSer872 and pABCA1Ser2054 did not differ between groups. Novelty: Alterations in membrane cholesterol and phosphorylation of proteins that regulate muscle cholesterol are not essential for elevated insulin-stimulated GU in skeletal muscle after acute exercise.
Collapse
Affiliation(s)
- Haiyan Wang
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI 48109-1048, USA.,Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI 48109-1048, USA
| | - Edward B Arias
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI 48109-1048, USA.,Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI 48109-1048, USA
| | - Gregory D Cartee
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI 48109-1048, USA.,Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI 48109-1048, USA
| |
Collapse
|
40
|
Human Cytomegalovirus Induces the Expression of the AMPKa2 Subunit to Drive Glycolytic Activation and Support Productive Viral Infection. J Virol 2021; 95:JVI.01321-20. [PMID: 33268515 PMCID: PMC8092818 DOI: 10.1128/jvi.01321-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Human Cytomegalovirus (HCMV) infection modulates cellular metabolism to support viral replication. Calcium/calmodulin-dependent kinase kinase (CaMKK) and AMP-activated protein kinase (AMPK) regulate metabolic activation and have been found to be important for successful HCMV infection. Here, we explored the contributions that specific CaMKK isoforms and AMPK subunit isoforms make toward HCMV infection. Our results indicate that various CaMKK and AMPK isoforms contribute to infection in unique ways. For example, CaMKK1 is important for HCMV infection at a low multiplicity of infection, but is dispensable for AMPK activation at the earliest times of infection, which our data suggest is more reliant on CaMKK2. Our results also indicate that HCMV specifically induces the expression of the non-ubiquitous AMPKa2 catalytic subunit, found to be important for both HCMV-mediated glycolytic activation and high titer infection. Further, we find that AMPK-mediated glycolytic activation is important for infection, as overexpression of GLUT4, the high capacity glucose transporter, partially rescues viral replication in the face of AMPK inhibition. Collectively, our data indicate that HCMV infection selectively induces the expression of specific metabolic regulatory kinases, relying on their activity to support glycolytic activation and productive infection.IMPORTANCE Viruses are obligate parasites that depend on the host cell to provide the energy and molecular building blocks to mass produce infectious viral progeny. The processes that govern viral modulation of cellular resources have emerged as critical for successful infection. Here, we find that HCMV depends on two kinase isoforms to support infection, CaMKK1 and AMPKa2. We find that HCMV specifically induces expression of the AMPKa2 subunit to induce metabolic activation and drive robust viral replication. These results suggest that HCMV has evolved mechanisms to target specific metabolic regulatory kinase subunits to support productive infection, thereby providing insight into how HCMV hijacks cellular metabolism for its replication, and sheds light on potential viral therapeutic vulnerabilities.
Collapse
|
41
|
The Cardiometabolic Health Benefits of Sauna Exposure in Individuals with High-Stress Occupations. A Mechanistic Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18031105. [PMID: 33513711 PMCID: PMC7908414 DOI: 10.3390/ijerph18031105] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/22/2021] [Accepted: 01/24/2021] [Indexed: 01/11/2023]
Abstract
Components of the metabolic syndrome (i.e., hypertension, insulin resistance, obesity, atherosclerosis) are a leading cause of death in the United States and result in low-grade chronic inflammation, excessive oxidative stress, and the eventual development of cardiometabolic diseases (CMD). High-stress occupations (HSO: firefighters, police, military personnel, first responders, etc.) increase the risk of developing CMD because they expose individuals to chronic and multiple stressors (i.e., sleep deprivation, poor nutrition habits, lack of physical activity, psychological stress). Interestingly, heat exposure and, more specifically, sauna bathing have been shown to improve multiple markers of CMD, potentially acting as hormetic stressors, at the cellular level and in the whole organism. Therefore, sauna bathing might be a practical and alternative intervention for disease prevention for individuals with HSO. The purpose of this review is to detail the mechanisms and pathways involved in the response to both acute and chronic sauna bathing and collectively present sauna bathing as a potential treatment, in addition to current standard of care, for mitigating CMD to both clinicians and individuals serving in HSO.
Collapse
|
42
|
Kuramoto K, Sawada Y, Yamada T, Nagashima T, Ohnuki K, Shin T. Novel Indirect AMP-Activated Protein Kinase Activators: Identification of a Second-Generation Clinical Candidate with Improved Physicochemical Properties and Reduced hERG Inhibitory Activity. Chem Pharm Bull (Tokyo) 2021; 68:452-465. [PMID: 32378543 DOI: 10.1248/cpb.c20-00015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This study reports the synthesis and evaluation of novel indirect AMP-activated protein kinase (AMPK) activators. The series of compounds selectively inhibited cell growth in several human breast cancer cell lines by activating AMPK. We performed back-up medicinal chemistry synthetic research on ASP4132, a previously reported as a compound for clinical development that acts as an indirect AMPK activator. This led to the successful identification of 4-({4-[5-({1-[(5-ethoxypyrazin-2-yl)methyl]-4-fluoropiperidin-4-yl}methoxy)-3-methylpyridine-2-carbonyl]piperazin-1-yl}methyl)benzonitrile succinate (27b), a potent, highly aqueous soluble and metabolically stable compound in human hepatocytes. Compound 27b also showed weaker human Ether-a-go-go Related Gene (hERG) inhibitory activity than that of compound 13 and ASP4132. Therefore, 27b was a promising AMPK activator and a second-generation clinical candidate for treatment for human cancer.
Collapse
Affiliation(s)
| | - Yuki Sawada
- Drug Discovery Research, Astellas Pharma Inc
| | | | | | - Kei Ohnuki
- Drug Discovery Research, Astellas Pharma Inc
| | | |
Collapse
|
43
|
Son JS, Chae SA, Wang H, Chen Y, Bravo Iniguez A, de Avila JM, Jiang Z, Zhu MJ, Du M. Maternal Inactivity Programs Skeletal Muscle Dysfunction in Offspring Mice by Attenuating Apelin Signaling and Mitochondrial Biogenesis. Cell Rep 2020; 33:108461. [PMID: 33264618 PMCID: PMC8137280 DOI: 10.1016/j.celrep.2020.108461] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 10/14/2020] [Accepted: 11/10/2020] [Indexed: 12/25/2022] Open
Abstract
Although maternal exercise (ME) becomes increasingly uncommon, the effects of ME on offspring muscle metabolic health remain largely undefined. Maternal mice are subject to daily exercise during pregnancy, which enhances mitochondrial biogenesis during fetal muscle development; this is correlated with higher mitochondrial content and oxidative muscle fibers in offspring muscle and improved endurance capacity. Apelin, an exerkine, is elevated due to ME, and maternal apelin administration mirrors the effect of ME on mitochondrial biogenesis in fetal muscle. Importantly, both ME and apelin induce DNA demethylation of the peroxisome proliferator-activated receptor γ coactivator-1α (Ppargc1a) promoter and enhance its expression and mitochondrial biogenesis in fetal muscle. Such changes in DNA methylation were maintained in offspring, with ME offspring muscle expressing higher levels of PGC-1α1/4 isoforms, explaining improved muscle function. In summary, ME enhances DNA demethylation of the Ppargc1a promoter in fetal muscle, which has positive programming effects on the exercise endurance capacity and protects offspring muscle against metabolic dysfunction. Son et al. demonstrate that maternal exercise facilitates fetal muscle development, which improves muscle function and exercise endurance in offspring. Maternal administration of apelin, an exerkine, mirrors the beneficial effects of maternal exercise on mitochondrial biogenesis and fetal muscle development. These findings suggest apelin and its receptor as potential drug targets for improving fetal muscle development of sedentary mothers.
Collapse
Affiliation(s)
- Jun Seok Son
- Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, WA 99164, USA; School of Molecular Biosciences, Washington State University, Pullman, WA 99164, USA
| | - Song Ah Chae
- Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, WA 99164, USA
| | - Hongyang Wang
- Institute of Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai, China
| | - Yanting Chen
- Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, WA 99164, USA
| | | | - Jeanene M de Avila
- Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, WA 99164, USA
| | - Zhihua Jiang
- Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, WA 99164, USA
| | - Mei-Jun Zhu
- School of Food Science, Washington State University, Pullman, WA 99164, USA
| | - Min Du
- Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, WA 99164, USA; School of Molecular Biosciences, Washington State University, Pullman, WA 99164, USA.
| |
Collapse
|
44
|
Kim HW, Park EJ, Cho HM, An JP, Chin YW, Kim J, Sung SH, Oh WK. Glucose Uptake-Stimulating Galloyl Ester Triterpenoids from Castanopsis sieboldii. JOURNAL OF NATURAL PRODUCTS 2020; 83:3093-3101. [PMID: 32965112 DOI: 10.1021/acs.jnatprod.0c00645] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Using molecular networking-guided isolation, three new galloyl ester triterpenoids (1-3), two new hexahydroxydiphenic acid-conjugated triterpenoids (6 and 7), and four known compounds (4, 5, 8, and 9) were isolated from the fruits and leaves of Castanopsis sieboldii. The chemical structures of 1-3, 6, and 7 were elucidated on the basis of interpreting their NMR, HRESIMS, and ECD spectra. All compounds (1-9) were evaluated for their glucose uptake-stimulating activities in differentiated adipocytes using 2-deoxy-2-[(7-nitro-2,1,3-benzoxadiazol-4-yl)amino]-d-glucose as a fluorescent-tagged glucose probe. Compounds 2 and 9 resulted in a 1.5-fold increase in glucose uptake. Among them, compound 2 from the fruits showed an upregulation of p-AMPK/AMPK ratio in differentiated C2C12 myoblasts to support the mechanism proposed of glucose uptake stimulation.
Collapse
Affiliation(s)
- Hyun Woo Kim
- Korea Bioactive Natural Material Bank, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Eun Jin Park
- Korea Bioactive Natural Material Bank, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyo Moon Cho
- Korea Bioactive Natural Material Bank, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Jin-Pyo An
- Korea Bioactive Natural Material Bank, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Young Won Chin
- Korea Bioactive Natural Material Bank, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Jinwoong Kim
- Korea Bioactive Natural Material Bank, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Sang Hyun Sung
- Korea Bioactive Natural Material Bank, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Won Keun Oh
- Korea Bioactive Natural Material Bank, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
45
|
Eslamizad M, Albrecht D, Kuhla B. The effect of chronic, mild heat stress on metabolic changes of nutrition and adaptations in rumen papillae of lactating dairy cows. J Dairy Sci 2020; 103:8601-8614. [PMID: 32600758 DOI: 10.3168/jds.2020-18417] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 04/18/2020] [Indexed: 12/25/2022]
Abstract
Global warming and accompanying high ambient temperatures reduce feed intake of dairy cows and shift the blood flow from the core of the body to the periphery. As a result, hypoxia may occur in the digestive tract accompanied by disruption of the intestinal barrier, local endotoxemia and inflammation, and altered nutrient absorption. However, whether the barrier of the rumen, like the intestine, is affected by ambient heat has not been studied so far. Lactating Holstein dairy cows were subjected to heat stress at 28°C (temperature-humidity index = 76; n = 5) with ad libitum feed intake or to thermoneutral conditions at 15°C (temperature-humidity index = 60; n = 5) and pair-feeding to heat-stressed animals for a total of 4 d. Gas exchange and feed intake behavior were measured in a respiration chamber, and rumen epithelia were taken after slaughter. Heat stress significantly reduced meal size and whole-body fat oxidation but increased meal frequency and carbohydrate oxidation. The mRNA expression of toll-like receptor 4 (TLR4) and tight junction proteins and the phosphorylation of TLR4 downstream targets (interleukin-1 receptor-associated kinase 4, stress-activated protein kinase, p38 mitogen-activated protein kinase, and nuclear factor k-B) in the rumen epithelium were not affected by heat. The proteomics approach revealed increased expression of rumen epithelium proteins involved in the AMP-activated protein kinase (AMPK) and insulin signaling pathways in heat-stressed cows. Also, proteins involved in chaperone-mediated folding of proteins were upregulated, whereas those involved in antioxidant defense system were downregulated. Further, we found evidence for increased carbohydrate phosphorylation accompanied with an increased flux of carbohydrates through the hexosamine biosynthetic pathway, providing substrates for protein glycosylation. In conclusion, the mild heat stress did not induce barrier dysfunction or inflammatory responses in the rumen epithelium of dairy cows, probably because of adaptations in feed intake behavior and defense mechanisms at the tissue level.
Collapse
Affiliation(s)
- Mehdi Eslamizad
- Institute of Nutritional Physiology "Oskar Kellner," Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| | - Dirk Albrecht
- Institute of Microbiology, Ernst-Moritz-Arndt-University, Felix-Hausdorff-Straße 8, 17487 Greifswald, Germany
| | - Björn Kuhla
- Institute of Nutritional Physiology "Oskar Kellner," Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany.
| |
Collapse
|
46
|
Cespedes A, Villa M, Benito-Cuesta I, Perez-Alvarez MJ, Ordoñez L, Wandosell F. Energy-Sensing Pathways in Ischemia: The Counterbalance Between AMPK and mTORC. Curr Pharm Des 2020; 25:4763-4770. [PMID: 31820693 DOI: 10.2174/1381612825666191210152156] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 12/06/2019] [Indexed: 01/02/2023]
Abstract
Stroke is an important cause of death and disability, and it is the second leading cause of death worldwide. In humans, middle cerebral artery occlusion (MCAO) is the most common cause of ischemic stroke. The damage occurs due to the lack of nutrients and oxygen contributed by the blood flow. The present review aims to analyze to what extent the lack of each of the elements of the system leads to damage and which mechanisms are unaffected by this deficiency. We believe that the specific analysis of the effect of lack of each component could lead to the emergence of new therapeutic targets for this important brain pathology.
Collapse
Affiliation(s)
- Angel Cespedes
- Centro de Biología Molecular "Severo Ochoa". CSIC-UAM. Nicolás Cabrera 1, 28049 Madrid, Spain.,Research Group of Neurodegenerative Diseases, Department of Animal Health, Faculty of Veterinary Medicine and Zootechnics - Tolima University, Santa Helena - 730006299, Ibagué, Colombia
| | - Mario Villa
- Centro de Biología Molecular "Severo Ochoa". CSIC-UAM. Nicolás Cabrera 1, 28049 Madrid, Spain.,Departamento de Biología (Fisiología Animal). Facultad de Ciencias. Universidad Autónoma de Madrid. C/Darwin 2. 28049 Madrid, Spain
| | - Irene Benito-Cuesta
- Centro de Biología Molecular "Severo Ochoa". CSIC-UAM. Nicolás Cabrera 1, 28049 Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Maria J Perez-Alvarez
- Centro de Biología Molecular "Severo Ochoa". CSIC-UAM. Nicolás Cabrera 1, 28049 Madrid, Spain.,Departamento de Biología (Fisiología Animal). Facultad de Ciencias. Universidad Autónoma de Madrid. C/Darwin 2. 28049 Madrid, Spain
| | - Lara Ordoñez
- Centro de Biología Molecular "Severo Ochoa". CSIC-UAM. Nicolás Cabrera 1, 28049 Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Francisco Wandosell
- Centro de Biología Molecular "Severo Ochoa". CSIC-UAM. Nicolás Cabrera 1, 28049 Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
47
|
Coomer CA, Carlon-Andres I, Iliopoulou M, Dustin ML, Compeer EB, Compton AA, Padilla-Parra S. Single-cell glycolytic activity regulates membrane tension and HIV-1 fusion. PLoS Pathog 2020; 16:e1008359. [PMID: 32084246 PMCID: PMC7055913 DOI: 10.1371/journal.ppat.1008359] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 03/04/2020] [Accepted: 01/27/2020] [Indexed: 12/21/2022] Open
Abstract
There has been resurgence in determining the role of host metabolism in viral infection yet deciphering how the metabolic state of single cells affects viral entry and fusion remains unknown. Here, we have developed a novel assay multiplexing genetically-encoded biosensors with single virus tracking (SVT) to evaluate the influence of global metabolic processes on the success rate of virus entry in single cells. We found that cells with a lower ATP:ADP ratio prior to virus addition were less permissive to virus fusion and infection. These results indicated a relationship between host metabolic state and the likelihood for virus-cell fusion to occur. SVT revealed that HIV-1 virions were arrested at hemifusion in glycolytically-inactive cells. Interestingly, cells acutely treated with glycolysis inhibitor 2-deoxyglucose (2-DG) become resistant to virus infection and also display less surface membrane cholesterol. Addition of cholesterol in these in glycolytically-inactive cells rescued the virus entry block at hemifusion and enabled completion of HIV-1 fusion. Further investigation with FRET-based membrane tension and membrane order reporters revealed a link between host cell glycolytic activity and host membrane order and tension. Indeed, cells treated with 2-DG possessed lower plasma membrane lipid order and higher tension values, respectively. Our novel imaging approach that combines lifetime imaging (FLIM) and SVT revealed not only changes in plasma membrane tension at the point of viral fusion, but also that HIV is less likely to enter cells at areas of higher membrane tension. We therefore have identified a connection between host cell glycolytic activity and membrane tension that influences HIV-1 fusion in real-time at the single-virus fusion level in live cells.
Collapse
Affiliation(s)
- Charles A. Coomer
- Cellular Imaging Group, Wellcome Centre Human Genetics, University of Oxford, Oxford, United Kingdom
- HIV Dynamics and Replication Program, National Cancer Institute, Frederick, Maryland, United States of America
- University of Kentucky, College of Medicine, Lexington, Kentucky, United States of America
- Division of Structural Biology, Wellcome Centre Human Genetics, University of Oxford, United Kingdom
| | - Irene Carlon-Andres
- Cellular Imaging Group, Wellcome Centre Human Genetics, University of Oxford, Oxford, United Kingdom
- Division of Structural Biology, Wellcome Centre Human Genetics, University of Oxford, United Kingdom
| | - Maro Iliopoulou
- Cellular Imaging Group, Wellcome Centre Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Michael L. Dustin
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Ewoud B. Compeer
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Alex A. Compton
- HIV Dynamics and Replication Program, National Cancer Institute, Frederick, Maryland, United States of America
| | - Sergi Padilla-Parra
- Cellular Imaging Group, Wellcome Centre Human Genetics, University of Oxford, Oxford, United Kingdom
- Division of Structural Biology, Wellcome Centre Human Genetics, University of Oxford, United Kingdom
| |
Collapse
|
48
|
Krishan S, Sahni S, Leck LYW, Jansson PJ, Richardson DR. Regulation of autophagy and apoptosis by Dp44mT-mediated activation of AMPK in pancreatic cancer cells. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165657. [PMID: 31904416 DOI: 10.1016/j.bbadis.2019.165657] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 12/16/2019] [Accepted: 12/20/2019] [Indexed: 01/14/2023]
Abstract
Upon activation, the 5'-adenosine monophosphate-activated protein kinase (AMPK) increases catabolism, while inhibiting anabolism. The anti-cancer agent, di-2-pyridylketone 4,4-dimethyl-3-thiosemicarbazone (Dp44mT), activates AMPK in multiple tumor cell-types (Biochim. Biophys Acta 2016;1863:2916-2933). This acts as an initial cell "rescue response" after iron-depletion mediated by Dp44mT. Considering Dp44mT-mediated AMPK activation, the role of AMPK on Dp44mT cytotoxicity was examined. Dp44mT increased the p-AMPK/AMPK ratio in multiple tumor cell-types over short (24 h) and longer (72 h) incubations. Notably, Dp44mT was more effective in inhibiting tumor cell proliferation after AMPK silencing, potentially due to the loss of AMPK-mediated metabolic plasticity that protects cells against Dp44mT cytotoxicity. The silencing of AMPK-increased cellular cholesterol and stabilized lysosomes against Dp44mT-mediated lysosomal membrane permeabilization. This was substantiated by studies demonstrating that the cholesterol-depleting agent, methyl-β-cyclodextrin (MβCD), restores Dp44mT-mediated lysosomal membrane permeabilization in AMPK silenced cells. The increased levels of cholesterol after AMPK silencing were independent of the ability of AMPK to inhibit the rate-limiting step of cholesterol synthesis via the inactivating phosphorylation of 3-hydroxy-3-methylglutaryl CoA reductase (HMGCR) at Ser872. In fact, Dp44mT did not increase phosphorylation of HMGCR at (Ser872), but decreased total HMGCR expression similarly in both the presence or absence of AMPK silencing. Dp44mT was demonstrated to increase autophagic initiation after AMPK silencing via an AMPK- and Beclin-1-independent mechanism. Further, there was increased cleaved caspase 3 and cleaved PARP after incubation of AMPK silenced cells with Dp44mT. Overall, AMPK silencing promotes Dp44mT anti-proliferative activity, suggesting a role for AMPK in rescuing its cytotoxicity by inhibiting autophagy and also apoptosis.
Collapse
Affiliation(s)
- S Krishan
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
| | - S Sahni
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
| | - L Y W Leck
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
| | - P J Jansson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
| | - D R Richardson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia; Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| |
Collapse
|
49
|
Manandhar B, Cochran BJ, Rye KA. Role of High-Density Lipoproteins in Cholesterol Homeostasis and Glycemic Control. J Am Heart Assoc 2019; 9:e013531. [PMID: 31888429 PMCID: PMC6988162 DOI: 10.1161/jaha.119.013531] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Bikash Manandhar
- Lipid Research Group School of Medical Sciences Faculty of Medicine University of New South Wales Sydney New South Wales Australia
| | - Blake J Cochran
- Lipid Research Group School of Medical Sciences Faculty of Medicine University of New South Wales Sydney New South Wales Australia
| | - Kerry-Anne Rye
- Lipid Research Group School of Medical Sciences Faculty of Medicine University of New South Wales Sydney New South Wales Australia
| |
Collapse
|
50
|
Effects of Propolis Extract and Propolis-Derived Compounds on Obesity and Diabetes: Knowledge from Cellular and Animal Models. Molecules 2019; 24:molecules24234394. [PMID: 31805752 PMCID: PMC6930477 DOI: 10.3390/molecules24234394] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 11/28/2019] [Accepted: 11/29/2019] [Indexed: 12/23/2022] Open
Abstract
Propolis is a natural product resulting from the mixing of bee secretions with botanical exudates. Since propolis is rich in flavonoids and cinnamic acid derivatives, the application of propolis extracts has been tried in therapies against cancer, inflammation, and metabolic diseases. As metabolic diseases develop relatively slowly in patients, the therapeutic effects of propolis in humans should be evaluated over long periods of time. Moreover, several factors such as medical history, genetic inheritance, and living environment should be taken into consideration in human studies. Animal models, especially mice and rats, have some advantages, as genetic and microbiological variables can be controlled. On the other hand, cellular models allow the investigation of detailed molecular events evoked by propolis and derivative compounds. Taking advantage of animal and cellular models, accumulating evidence suggests that propolis extracts have therapeutic effects on obesity by controlling adipogenesis, adipokine secretion, food intake, and energy expenditure. Studies in animal and cellular models have also indicated that propolis modulates oxidative stress, the accumulation of advanced glycation end products (AGEs), and adipose tissue inflammation, all of which contribute to insulin resistance or defects in insulin secretion. Consequently, propolis treatment may mitigate diabetic complications such as nephropathy, retinopathy, foot ulcers, and non-alcoholic fatty liver disease. This review describes the beneficial effects of propolis on metabolic disorders.
Collapse
|