1
|
Thakur MR, Tupe RS. l-Arginine: A multifaceted regulator of diabetic cardiomyopathy. Biochem Biophys Res Commun 2025; 761:151720. [PMID: 40186920 DOI: 10.1016/j.bbrc.2025.151720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 04/07/2025]
Abstract
In diabetes mellitus, dysregulated glucose and lipid metabolism lead to diabetic cardiomyopathy (DCM) by imparting pathological myocardial remodeling and cellular injury. Accelerated glycation, oxidative stress, and activated inflammatory pathways culminate in cardiac fibrosis and hypertrophy in DCM. The regulatory effects of l-Arginine (L-Arg) have been elucidated in the pathological changes of DCM, including myocardial fibrosis, hypertrophy, and apoptosis, by inhibiting glycation and oxidative stress-induced inflammation. Disturbed L-Arg metabolism and decreased intracellular L-Arg pool are correlated with the progression of DCM; therefore, L-Arg supplementation has been prescribed for various cardiovascular dysfunctions. This review expands the therapeutic potential of L-Arg supplementation in DCM by elucidating its molecular mechanism of action and exploring potential clinical outcomes.
Collapse
Affiliation(s)
- Muskan R Thakur
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Lavale, Pune, 412115, Maharashtra, India
| | - Rashmi S Tupe
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Lavale, Pune, 412115, Maharashtra, India.
| |
Collapse
|
2
|
Luse MA, Schug WJ, Dunaway LS, Nyshadham S, Loeb SA, Carvalho A, Tessema R, Pavelic C, Keller TCS, Shu X, Ruddiman CA, Kosmach A, Sveeggen TM, Mitchell R, Bagher P, Minshal RD, Leitnger N, Columbus L, Levental KR, Levental I, Cortese-Krott M, Isakson BE. Nitrosation of CD36 Regulates Endothelial Function and Serum Lipids. Arterioscler Thromb Vasc Biol 2025. [PMID: 40242868 DOI: 10.1161/atvbaha.124.321964] [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: 10/07/2024] [Accepted: 03/24/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND During obesity, endothelial cells (ECs) become lipid laden, leading to endothelial dysfunction. We tested posttranslational modification on CD36 that may regulate EC lipid accumulation. METHODS We used an EC-specific Cav1 (caveolin-1) knockout mouse, nitrosation and palmitoylation assays, and whole animal Nγ-nitro-l-arginine methyl ester administration to examine blood lipids. RESULTS EC-specific Cav1 knockout male mice are hyperlipidemic regardless of diet but retain endothelial cell function. We found these mice have significantly increased NO in response to the lack of Cav1, and the presence or absence of NO toggled inversely EC lipid content and plasma lipid in mice. The NO nitrosated the fatty acid translocase CD36 at the same cysteines that are palmitoylated on CD36. The nitrosation of CD36 prevented its trafficking to the plasma membrane and decreased lipid accumulation. The physiological effect of this mechanism was a reliance on NO for endothelial function and not dilation. CONCLUSIONS This work suggests that CD36 nitrosation occurs as a protective mechanism to prevent EC lipotoxicity.
Collapse
Affiliation(s)
- Melissa A Luse
- Robert M. Berne Cardiovascular Research Center (M.A.L., W.J.S., L.S.D., S.N., S.A.L., A.C., R.T., C.P., T.C.S.K., C.A.R., N.L., B.E.I.)
- Department of Molecular Physiology and Biophysics (M.A.L., W.J.S., S.A.L., T.C.S.K., K.R.L., I.L., B.E.I.)
| | - Wyatt J Schug
- Robert M. Berne Cardiovascular Research Center (M.A.L., W.J.S., L.S.D., S.N., S.A.L., A.C., R.T., C.P., T.C.S.K., C.A.R., N.L., B.E.I.)
- Department of Molecular Physiology and Biophysics (M.A.L., W.J.S., S.A.L., T.C.S.K., K.R.L., I.L., B.E.I.)
| | - Luke S Dunaway
- Robert M. Berne Cardiovascular Research Center (M.A.L., W.J.S., L.S.D., S.N., S.A.L., A.C., R.T., C.P., T.C.S.K., C.A.R., N.L., B.E.I.)
| | - Shruthi Nyshadham
- Robert M. Berne Cardiovascular Research Center (M.A.L., W.J.S., L.S.D., S.N., S.A.L., A.C., R.T., C.P., T.C.S.K., C.A.R., N.L., B.E.I.)
| | - Skylar A Loeb
- Robert M. Berne Cardiovascular Research Center (M.A.L., W.J.S., L.S.D., S.N., S.A.L., A.C., R.T., C.P., T.C.S.K., C.A.R., N.L., B.E.I.)
- Department of Molecular Physiology and Biophysics (M.A.L., W.J.S., S.A.L., T.C.S.K., K.R.L., I.L., B.E.I.)
| | - Alicia Carvalho
- Robert M. Berne Cardiovascular Research Center (M.A.L., W.J.S., L.S.D., S.N., S.A.L., A.C., R.T., C.P., T.C.S.K., C.A.R., N.L., B.E.I.)
| | - Rachel Tessema
- Robert M. Berne Cardiovascular Research Center (M.A.L., W.J.S., L.S.D., S.N., S.A.L., A.C., R.T., C.P., T.C.S.K., C.A.R., N.L., B.E.I.)
| | - Caitlin Pavelic
- Robert M. Berne Cardiovascular Research Center (M.A.L., W.J.S., L.S.D., S.N., S.A.L., A.C., R.T., C.P., T.C.S.K., C.A.R., N.L., B.E.I.)
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville (C.P., C.A.R., N.L.)
| | - T C Stevenson Keller
- Robert M. Berne Cardiovascular Research Center (M.A.L., W.J.S., L.S.D., S.N., S.A.L., A.C., R.T., C.P., T.C.S.K., C.A.R., N.L., B.E.I.)
- Department of Molecular Physiology and Biophysics (M.A.L., W.J.S., S.A.L., T.C.S.K., K.R.L., I.L., B.E.I.)
| | - Xiaohong Shu
- College of Pharmacy, Dalian Medical University, China (X.S.)
| | - Claire A Ruddiman
- Robert M. Berne Cardiovascular Research Center (M.A.L., W.J.S., L.S.D., S.N., S.A.L., A.C., R.T., C.P., T.C.S.K., C.A.R., N.L., B.E.I.)
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville (C.P., C.A.R., N.L.)
| | - Anna Kosmach
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (A.K., T.M.S., R.M., P.B.)
| | - Timothy M Sveeggen
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (A.K., T.M.S., R.M., P.B.)
| | - Ray Mitchell
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (A.K., T.M.S., R.M., P.B.)
| | - Pooneh Bagher
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (A.K., T.M.S., R.M., P.B.)
| | | | - Norbert Leitnger
- Robert M. Berne Cardiovascular Research Center (M.A.L., W.J.S., L.S.D., S.N., S.A.L., A.C., R.T., C.P., T.C.S.K., C.A.R., N.L., B.E.I.)
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville (C.P., C.A.R., N.L.)
| | - Linda Columbus
- Department of Chemistry, University of Virginia, Charlottesville (L.C.)
| | - Kandice R Levental
- Department of Molecular Physiology and Biophysics (M.A.L., W.J.S., S.A.L., T.C.S.K., K.R.L., I.L., B.E.I.)
| | - Ilya Levental
- Department of Molecular Physiology and Biophysics (M.A.L., W.J.S., S.A.L., T.C.S.K., K.R.L., I.L., B.E.I.)
| | | | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center (M.A.L., W.J.S., L.S.D., S.N., S.A.L., A.C., R.T., C.P., T.C.S.K., C.A.R., N.L., B.E.I.)
- Department of Molecular Physiology and Biophysics (M.A.L., W.J.S., S.A.L., T.C.S.K., K.R.L., I.L., B.E.I.)
| |
Collapse
|
3
|
Mihalikova D, Stamm P, Kvandova M, Pednekar C, Strohm L, Ubbens H, Oelze M, Kuntic M, Witzler C, Bayo Jimenez MT, Rajlic S, Frenis K, Tang Q, Ruan Y, Karbach S, Kleinert H, Hahad O, von Kriegsheim A, Xia N, Grune T, Li H, Kröller-Schön S, Gericke A, Ruf W, Wild PS, Lurz P, Münzel T, Daiber A, Jansen T. Exposure to aircraft noise exacerbates cardiovascular and oxidative damage in three mouse models of diabetes. Eur J Prev Cardiol 2025; 32:301-314. [PMID: 39351780 DOI: 10.1093/eurjpc/zwae320] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/12/2024] [Accepted: 09/27/2024] [Indexed: 03/06/2025]
Abstract
AIMS Epidemiology links noise to increased risk of metabolic diseases like diabetes and obesity. Translational studies in humans and experimental animals showed that noise causes reactive oxygen species (ROS)-mediated cardiovascular damage. The interaction between noise and diabetes, specifically potential additive adverse effects, remains to be determined. METHODS AND RESULTS C57BL/6 mice were treated with streptozotocin (i.p. injections, 50 mg/kg/day for 5 days) to induce type 1 diabetes mellitus, with S961 (subcutaneous osmotic mini-pumps, 0.57 mg/kg/day for 7 days) or fed a high-fat diet (HFD, 20 weeks) to induce type 2 diabetes mellitus. Control and diabetic mice were exposed to aircraft noise to an average sound pressure level of 72 dB(A) for 4 days. While body weight was unaffected, noise reduced insulin production in all diabetes models. The oral glucose tolerance test showed only an additive aggravation by noise in the HFD model. Noise increased blood pressure and aggravated diabetes-induced aortic, mesenteric, and cerebral arterioles' endothelial dysfunction. ROS formation in cerebral arterioles, the aorta, the heart, and isolated mitochondria was consistently increased by noise in all models of diabetes. Mitochondrial respiration was impaired by diabetes and noise, however without additive effects. Noise increased ROS and caused inflammation in adipose tissue in the HFD model. RNA-sequencing data and alteration of gene pathway clusters also supported additive damage by noise in the setting of diabetes. CONCLUSION In all three models of diabetes, aircraft noise exacerbates oxidative stress, inflammation, and endothelial dysfunction in mice with pre-existing diabetes. Thus, noise may potentiate the already increased cardiovascular risk in diabetic patients.
Collapse
MESH Headings
- Animals
- Oxidative Stress
- Mice, Inbred C57BL
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/physiopathology
- Diabetes Mellitus, Experimental/blood
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/genetics
- Male
- Reactive Oxygen Species/metabolism
- Noise, Transportation/adverse effects
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/physiopathology
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/genetics
- Aircraft
- Diet, High-Fat
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/blood
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 1/physiopathology
- Diabetes Mellitus, Type 1/chemically induced
- Diabetes Mellitus, Type 1/genetics
- Blood Glucose/metabolism
- Mice
- Disease Models, Animal
- Insulin/blood
Collapse
Affiliation(s)
- Dominika Mihalikova
- Department for Cardiology, Cardiology 1, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Paul Stamm
- Department for Cardiology, Cardiology 1, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Miroslava Kvandova
- Department for Cardiology, Cardiology 1, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
- Institute of Normal and Pathological Physiology, Centre of Experimental Medicine, Slovak Academy ofSciences, Bratislava, Slovakia
| | | | - Lea Strohm
- Department for Cardiology, Cardiology 1, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Henning Ubbens
- Department for Cardiology, Cardiology 1, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Matthias Oelze
- Department for Cardiology, Cardiology 1, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Marin Kuntic
- Department for Cardiology, Cardiology 1, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Claudius Witzler
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Maria Teresa Bayo Jimenez
- Department for Cardiology, Cardiology 1, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
- Laboratory of RNA Archaeology, Instituto de Parasitología y Biomedicina 'López-Neyra' (CSIC), Granada, Spain
| | - Sanela Rajlic
- Department for Cardiology, Cardiology 1, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Katie Frenis
- Department for Cardiology, Cardiology 1, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
- Hematology/Oncology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
- Stem Cell Program, Boston Children's Hospital, Boston, MA, USA
| | - Qi Tang
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Yue Ruan
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Susanne Karbach
- Department for Cardiology, Cardiology 1, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Langenbeckstr. 1, 55131 Mainz, Germany
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Hartmut Kleinert
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Omar Hahad
- Department for Cardiology, Cardiology 1, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Langenbeckstr. 1, 55131 Mainz, Germany
| | | | - Ning Xia
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Tilman Grune
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Huige Li
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Langenbeckstr. 1, 55131 Mainz, Germany
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Swenja Kröller-Schön
- Department for Cardiology, Cardiology 1, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Adrian Gericke
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Wolfram Ruf
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Langenbeckstr. 1, 55131 Mainz, Germany
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Philipp S Wild
- Department for Cardiology, Cardiology 1, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Langenbeckstr. 1, 55131 Mainz, Germany
- Preventive Cardiology and Preventive Medicine, Department of Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Institute of Molecular Biology (IMB), Johannes Gutenberg University, Mainz, Germany
| | - Philipp Lurz
- Department for Cardiology, Cardiology 1, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Thomas Münzel
- Department for Cardiology, Cardiology 1, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Langenbeckstr. 1, 55131 Mainz, Germany
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Andreas Daiber
- Department for Cardiology, Cardiology 1, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Langenbeckstr. 1, 55131 Mainz, Germany
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Thomas Jansen
- Department for Cardiology, Cardiology 1, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
- Department of Cardiology, KVB Hospital, Königstein, Germany
| |
Collapse
|
4
|
Signorello MG, Leoncini G. The Cortisol Effect on the NO/cGMP Pathway. Int J Mol Sci 2025; 26:1421. [PMID: 40003888 PMCID: PMC11855650 DOI: 10.3390/ijms26041421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/30/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Previously, it has been shown that cortisol induces oxidative stress in human platelets, stimulating reactive oxygen species production, superoxide anion formation, lipid peroxidation, and depleting antioxidant defenses. In this study, the cortisol effect on platelet function has been described. Results demonstrate that cortisol stimulates platelet activation and aggregation, leading to CD62P surface exposure and intracellular calcium elevation. Cortisol potentiates its aggregating effect, reducing the level of the powerful anti-aggregating agent nitric oxide (NO). Likewise, cortisol reduces cGMP levels. Moreover, specific inhibitors of the Src/Syk/PI3K/AKT pathways reverse the inhibiting effect of cortisol, partially restoring NO and cGMP levels. Unexpectedly, cortisol stimulates endothelial nitric oxide synthase (eNOS) activity, measured in platelet lysates prepared by whole cells treated with the hormone. The phosphorylation of the Ser1177 eNOS activating-residue is increased by cortisol. The Src/Syk/PI3K/AKT pathways appear to be involved in the phosphorylation of this residue. Moreover, cortisol induces the formation of nitrotyrosine, that can be considered a biomarker for reactive nitrogen species, including peroxynitrite. In conclusion, through these mechanisms, cortisol potentiates its capacity to induce oxidative stress in human platelets.
Collapse
|
5
|
Fraga CG, Cremonini E, Galleano M, Oteiza PI. Natural Products and Diabetes: (-)-Epicatechin and Mechanisms Involved in the Regulation of Insulin Sensitivity. Handb Exp Pharmacol 2025; 287:159-173. [PMID: 38421444 DOI: 10.1007/164_2024_707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
Type 2 diabetes (T2D) is a disease that occurs when cells do not respond normally to insulin, a condition called insulin resistance, which leads to high blood glucose levels. Although it can be treated pharmacologically, dietary habits beyond carbohydrate restriction can be highly relevant in the management of T2D. Emerging evidence supports the possibility that natural products (NPs) could contribute to managing blood glucose or counteract the undesirable effects of hyperglycemia and insulin resistance. This chapter summarizes the relevant preclinical evidence involving the flavonoid (-)-epicatechin (EC) in the optimization of glucose homeostasis, reducing insulin resistance and/or diabetes-associated disorders. Major effects of EC are observed on (i) intestinal functions, including digestive enzymes, glucose transporters, microbiota, and intestinal permeability, and (ii) redox homeostasis, including oxidative stress and inflammation. There is still a need for further clinical studies to confirm the in vitro and rodent data, allowing recommendations for EC, particularly in prediabetic and T2D patients. The collection of similar data and the lack of clinical evidence for EC is also applicable to other NPs.
Collapse
Affiliation(s)
- Cesar G Fraga
- Fisicoquímica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.
- Instituto de Bioquímica y Medicina Molecular (IBIMOL), UBA-CONICET, Buenos Aires, Argentina.
- Department of Nutrition, University of California, Davis, CA, USA.
| | | | - Monica Galleano
- Fisicoquímica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Bioquímica y Medicina Molecular (IBIMOL), UBA-CONICET, Buenos Aires, Argentina
| | - Patricia I Oteiza
- Department of Nutrition, University of California, Davis, CA, USA
- Department of Environmental Toxicology, University of California, Davis, CA, USA
| |
Collapse
|
6
|
Sarlak Z, Naderi N, Amidi B, Ghorbanzadeh V. Sodium Butyrate, A Gut Microbiota Derived Metabolite in Type 2 Diabetes Mellitus and Cardiovascular Disease: A Review. Cardiovasc Hematol Agents Med Chem 2025; 23:1-10. [PMID: 39206487 DOI: 10.2174/0118715257307380240820052940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 06/13/2024] [Accepted: 07/03/2024] [Indexed: 09/04/2024]
Abstract
Type 2 diabetes is characterized by elevated blood glucose levels, leading to an increased risk of cardiovascular diseases. Sodium butyrate, the sodium salt of the short-chain fatty acid butyric acid produced by gut microbiota fermentation, has shown promising effects on metabolic diseases, including type 2 diabetes and cardiovascular diseases. Sodium butyrate demonstrates anti-inflammatory, anti-oxidative, and lipid-lowering properties and can improve insulin sensitivity and reduce hepatic steatosis. In this review, we investigate how sodium butyrate influences cardiovascular complications of type 2 diabetes, including atherosclerosis (AS), heart failure (HF), hypertension, and angiogenesis. Moreover, we explore the pathophysiology of cardiovascular disease in type 2 diabetes, focusing on hyperglycemia, oxidative stress, inflammation, and genetic factors playing crucial roles. The review suggests that sodium butyrate can be a potential preventive and therapeutic agent for cardiovascular complications in individuals with type 2 diabetes.
Collapse
Affiliation(s)
- Zeynab Sarlak
- Department of Biology, Khorramabad branch, Islamic Azad University, Khorramabad, Iran
| | - Narges Naderi
- Cardiovascular Research Center, Shahid Rahimi Hospital, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Bardia Amidi
- Cardiovascular Research Center, Shahid Rahimi Hospital, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Vajihe Ghorbanzadeh
- Cardiovascular Research Center, Shahid Rahimi Hospital, Lorestan University of Medical Sciences, Khorramabad, Iran
| |
Collapse
|
7
|
Miyazaki S, Takino JI, Nagamine K, Takeuchi M, Hori T. RasGRP2 Attenuates TAGE Modification of eNOS in Vascular Endothelial Cells. Biol Pharm Bull 2025; 48:262-266. [PMID: 40101981 DOI: 10.1248/bpb.b24-00730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Toxic advanced glycation end-products (TAGEs) are glyceraldehyde (GA)-derived AGEs with strong cytotoxic effects. TAGEs are also involved in lifestyle-related diseases. Notably, modification of TAGEs by GA causes protein dysfunction. As endothelial nitric oxide synthase (eNOS) is constitutively expressed in vascular endothelial cells and is a source of nitric oxide (NO), we focused on it as a TAGE modification-targeting protein. Our laboratory has reported that Ras guanyl nucleotide-releasing protein 2 (RasGRP2) activates Rap1 and R-Ras, among other small GTPases, and suppresses apoptosis and TAGE-induced vascular hyperpermeability in vascular endothelial cells. Therefore, in this study, we investigated the effects of RasGRP2 on cell death, TAGE formation, and TAGE modification of eNOS in vascular endothelial cells following GA treatment using RasGRP2-overexpressing (R) cells and mock (M) immortalized human umbilical vein endothelial cells. GA treatment decreases the viability of both cell types in a concentration-dependent manner. In M cells, GA treatment increased the formation of TAGEs and TAGE modification of eNOS in a concentration-dependent manner, but this increase was suppressed in R cells. Additionally, co-treatment with aminoguanidine, an inhibitor of AGEs formation, suppressed cell death and TAGE modification of eNOS induced by GA. These results indicate that GA induces cell death, the formation of TAGEs, and TAGE modification of eNOS in vascular endothelial cells. Additionally, RasGRP2 is a protective factor that suppresses TAGE formation.
Collapse
Affiliation(s)
- Shouhei Miyazaki
- Faculty of Pharmaceutical Sciences, Hiroshima International University, 5-1-1 Hirokoshingai, Kure, Hiroshima 737-0112, Japan
| | - Jun-Ichi Takino
- Faculty of Pharmaceutical Sciences, Hiroshima International University, 5-1-1 Hirokoshingai, Kure, Hiroshima 737-0112, Japan
| | - Kentaro Nagamine
- Faculty of Health Sciences, Hiroshima International University, 5-1-1 Hirokoshingai, Kure, Hiroshima 737-0112, Japan
| | - Masayoshi Takeuchi
- Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, 1-1 Daigaku, Uchinada-machi, Kahoku, Ishikawa 920-0293, Japan
| | - Takamitsu Hori
- Faculty of Pharmaceutical Sciences, Hiroshima International University, 5-1-1 Hirokoshingai, Kure, Hiroshima 737-0112, Japan
| |
Collapse
|
8
|
Mashaba GR, Phoswa WN, Mokgalaboni K. The Effect of Statins on Carotid Intima-Media Thickness and C-Reactive Protein in Type 2 Diabetes Mellitus: A Meta-Analysis. J Cardiovasc Dev Dis 2024; 11:276. [PMID: 39330333 PMCID: PMC11432595 DOI: 10.3390/jcdd11090276] [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: 07/25/2024] [Revised: 08/21/2024] [Accepted: 09/02/2024] [Indexed: 09/28/2024] Open
Abstract
BACKGROUND The effect of statins on CIMT progression and C-reactive protein (CRP) in T2DM patients is widely reported. However, some studies demonstrated no effect of statins on CIMT and CRP in T2DM patients, while others reported otherwise. Thus, the current study comprehensively and quantitatively analyzes data from previous studies to evaluate the overall effect of statins on CIMT and CRP in T2DM to rule out any inconsistencies observed in previous clinical evidence. Therefore, the aim of this meta-oanalysis was to evaluate the effect of statins on CIMT progression and CRP in T2DM. METHODS A comprehensive search for studies was performed using PubMed, Scopus, Web of Sciences, and the Cochrane Library, for publications from their inception to 16 July 2024. The meta-analysis was conducted using Jamovi (version 4.2.8) and Review Manager (version 5.4), with the overall effect sizes reported as standardized mean differences (SMD) and 95% confidence intervals (CI). RESULTS Evidence from eleven studies (fifteen statin dosages) that met the inclusion criteria with a sample size of 983 T2DM patients on statin treatment was analyzed. The overall effect size from the random effect model meta-analysis showed a reduction in the CIMT status amongst T2DM patients post-statin treatment compared to at baseline [SMD = -0.47, 95%CI (-0.76, -0.18), p = 0.001]. Furthermore, there was a reduction in the level of CRP in T2DM patients post-treatment [SMD = -1.80, 95% CI (-2.76, -0.84), p < 0.001]. CONCLUSIONS Evidence gathered in this study suggests that statin therapy effectively reduces CIMT and CRP levels among patients living with T2DM. Interestingly, this evidence suggests that 20 mg of atorvastatin is more effective in reducing CIMT and CRP. Therefore, we recommend conducting further trials with larger sample sizes and proper methodology for T2DM.
Collapse
Affiliation(s)
- Given Reneilwe Mashaba
- Department of Life and Consumer Science, College of Agriculture and Environmental Sciences, University of South Africa, Florida Campus, Roodepoort 1710, South Africa;
- DIMAMO Population Health Research Centre, University of Limpopo, Sovenga, Private Bag X1106, Polokwane 0727, South Africa
| | - Wendy Nokhwezi Phoswa
- Department of Life and Consumer Science, College of Agriculture and Environmental Sciences, University of South Africa, Florida Campus, Roodepoort 1710, South Africa;
| | - Kabelo Mokgalaboni
- Department of Life and Consumer Science, College of Agriculture and Environmental Sciences, University of South Africa, Florida Campus, Roodepoort 1710, South Africa;
| |
Collapse
|
9
|
Qiu X, Lan X, Li L, Chen H, Zhang N, Zheng X, Xie X. The role of perirenal adipose tissue deposition in chronic kidney disease progression: Mechanisms and therapeutic implications. Life Sci 2024; 352:122866. [PMID: 38936605 DOI: 10.1016/j.lfs.2024.122866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/11/2024] [Accepted: 06/20/2024] [Indexed: 06/29/2024]
Abstract
Chronic kidney disease (CKD) represents a significant and escalating global health challenge, with morbidity and mortality rates rising steadily. Evidence increasingly implicates perirenal adipose tissue (PRAT) deposition as a contributing factor in the pathogenesis of CKD. This review explores how PRAT deposition may exert deleterious effects on renal structure and function. The anatomical proximity of PRAT to the kidneys not only potentially causes mechanical compression but also leads to the dysregulated secretion of adipokines and inflammatory mediators, such as adiponectin, leptin, visfatin, tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and exosomes. Additionally, PRAT deposition may contribute to renal lipotoxicity through elevated levels of free fatty acids (FFA), triglycerides (TAG), diacylglycerol (DAG), and ceramides (Cer). PRAT deposition is also linked to the hyperactivation of the renin-angiotensin-aldosterone system (RAAS), which further exacerbates CKD progression. Recognizing PRAT deposition as an independent risk factor for CKD underscores the potential of targeting PRAT as a novel strategy for the prevention and management of CKD. This review further discusses interventions that could include measuring PRAT thickness to establish a baseline, managing metabolic risk factors that promote its deposition, and inhibiting key PRAT-induced signaling pathways.
Collapse
Affiliation(s)
- Xiang Qiu
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Xin Lan
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Langhui Li
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Huan Chen
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China; Nucleic Acid Medicine of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Ningjuan Zhang
- The School of Clinical Medical Sciences, Southwest Medical University, Luzhou, China
| | - Xiaoli Zheng
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China.
| | - Xiang Xie
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China.
| |
Collapse
|
10
|
Guo Z. The role of glucagon-like peptide-1/GLP-1R and autophagy in diabetic cardiovascular disease. Pharmacol Rep 2024; 76:754-779. [PMID: 38890260 DOI: 10.1007/s43440-024-00609-1] [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: 11/30/2023] [Revised: 05/25/2024] [Accepted: 06/03/2024] [Indexed: 06/20/2024]
Abstract
Diabetes leads to a significantly accelerated incidence of various related macrovascular complications, including peripheral vascular disease and cardiovascular disease (the most common cause of mortality in diabetes), as well as microvascular complications such as kidney disease and retinopathy. Endothelial dysfunction is the main pathogenic event of diabetes-related vascular disease at the earliest stage of vascular injury. Understanding the molecular processes involved in the development of diabetes and its debilitating vascular complications might bring up more effective and specific clinical therapies. Long-acting glucagon-like peptide (GLP)-1 analogs are currently available in treating diabetes with widely established safety and extensively evaluated efficacy. In recent years, autophagy, as a critical lysosome-dependent self-degradative process to maintain homeostasis, has been shown to be involved in the vascular endothelium damage in diabetes. In this review, the GLP-1/GLP-1R system implicated in diabetic endothelial dysfunction and related autophagy mechanism underlying the pathogenesis of diabetic vascular complications are briefly presented. This review also highlights a possible crosstalk between autophagy and the GLP-1/GLP-1R axis in the treatment of diabetic angiopathy.
Collapse
Affiliation(s)
- Zi Guo
- Section of Nephrology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06510, USA.
| |
Collapse
|
11
|
Kuntic M, Hahad O, Al-Kindi S, Oelze M, Lelieveld J, Daiber A, Münzel T. Pathomechanistic Synergy Between Particulate Matter and Traffic Noise-Induced Cardiovascular Damage and the Classical Risk Factor Hypertension. Antioxid Redox Signal 2024. [PMID: 38874533 DOI: 10.1089/ars.2024.0659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Affiliation(s)
- Marin Kuntic
- Department of Cardiology 1, Medical Center of the Johannes Gutenberg University, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Mainz, Germany
| | - Omar Hahad
- Department of Cardiology 1, Medical Center of the Johannes Gutenberg University, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Mainz, Germany
| | - Sadeer Al-Kindi
- Cardiovascular Prevention & Wellness and Center for CV Computational & Precision Health, Houston Methodist DeBakey Heart & Vascular Center, Houston, Texas, USA
| | - Matthias Oelze
- Department of Cardiology 1, Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Jos Lelieveld
- Max Planck Institute for Chemistry, Atmospheric Chemistry, Mainz, Germany
| | - Andreas Daiber
- Department of Cardiology 1, Medical Center of the Johannes Gutenberg University, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Mainz, Germany
| | - Thomas Münzel
- Department of Cardiology 1, Medical Center of the Johannes Gutenberg University, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Mainz, Germany
| |
Collapse
|
12
|
Dou B, Zhu Y, Sun M, Wang L, Tang Y, Tian S, Wang F. Mechanisms of Flavonoids and Their Derivatives in Endothelial Dysfunction Induced by Oxidative Stress in Diabetes. Molecules 2024; 29:3265. [PMID: 39064844 PMCID: PMC11279171 DOI: 10.3390/molecules29143265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/02/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
Diabetic complications pose a significant threat to life and have a negative impact on quality of life in individuals with diabetes. Among the various factors contributing to the development of these complications, endothelial dysfunction plays a key role. The main mechanism underlying endothelial dysfunction in diabetes is oxidative stress, which adversely affects the production and availability of nitric oxide (NO). Flavonoids, a group of phenolic compounds found in vegetables, fruits, and fungi, exhibit strong antioxidant and anti-inflammatory properties. Several studies have provided evidence to suggest that flavonoids have a protective effect on diabetic complications. This review focuses on the imbalance between reactive oxygen species and the antioxidant system, as well as the changes in endothelial factors in diabetes. Furthermore, we summarize the protective mechanisms of flavonoids and their derivatives on endothelial dysfunction in diabetes by alleviating oxidative stress and modulating other signaling pathways. Although several studies underline the positive influence of flavonoids and their derivatives on endothelial dysfunction induced by oxidative stress in diabetes, numerous aspects still require clarification, such as optimal consumption levels, bioavailability, and side effects. Consequently, further investigations are necessary to enhance our understanding of the therapeutic potential of flavonoids and their derivatives in the treatment of diabetic complications.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Furong Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250300, China
| |
Collapse
|
13
|
Kiskin FN, Yang Y, Yang H, Zhang JZ. Cracking the code of the cardiovascular enigma: hPSC-derived endothelial cells unveil the secrets of endothelial dysfunction. J Mol Cell Cardiol 2024; 192:65-78. [PMID: 38761989 DOI: 10.1016/j.yjmcc.2024.05.005] [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: 03/13/2024] [Revised: 05/08/2024] [Accepted: 05/10/2024] [Indexed: 05/20/2024]
Abstract
Endothelial dysfunction is a central contributor to the development of most cardiovascular diseases and is characterised by the reduced synthesis or bioavailability of the vasodilator nitric oxide together with other abnormalities such as inflammation, senescence, and oxidative stress. The use of patient-specific and genome-edited human pluripotent stem cell-derived endothelial cells (hPSC-ECs) has shed novel insights into the role of endothelial dysfunction in cardiovascular diseases with strong genetic components such as genetic cardiomyopathies and pulmonary arterial hypertension. However, their utility in studying complex multifactorial diseases such as atherosclerosis, metabolic syndrome and heart failure poses notable challenges. In this review, we provide an overview of the different methods used to generate and characterise hPSC-ECs before comprehensively assessing their effectiveness in cardiovascular disease modelling and high-throughput drug screening. Furthermore, we explore current obstacles that will need to be overcome to unleash the full potential of hPSC-ECs in facilitating patient-specific precision medicine. Addressing these challenges holds great promise in advancing our understanding of intricate cardiovascular diseases and in tailoring personalised therapeutic strategies.
Collapse
Affiliation(s)
- Fedir N Kiskin
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| | - Yuan Yang
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| | - Hao Yang
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| | - Joe Z Zhang
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| |
Collapse
|
14
|
Wilkerson JL, Tatum SM, Holland WL, Summers SA. Ceramides are fuel gauges on the drive to cardiometabolic disease. Physiol Rev 2024; 104:1061-1119. [PMID: 38300524 PMCID: PMC11381030 DOI: 10.1152/physrev.00008.2023] [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: 02/14/2023] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 02/02/2024] Open
Abstract
Ceramides are signals of fatty acid excess that accumulate when a cell's energetic needs have been met and its nutrient storage has reached capacity. As these sphingolipids accrue, they alter the metabolism and survival of cells throughout the body including in the heart, liver, blood vessels, skeletal muscle, brain, and kidney. These ceramide actions elicit the tissue dysfunction that underlies cardiometabolic diseases such as diabetes, coronary artery disease, metabolic-associated steatohepatitis, and heart failure. Here, we review the biosynthesis and degradation pathways that maintain ceramide levels in normal physiology and discuss how the loss of ceramide homeostasis drives cardiometabolic pathologies. We highlight signaling nodes that sense small changes in ceramides and in turn reprogram cellular metabolism and stimulate apoptosis. Finally, we evaluate the emerging therapeutic utility of these unique lipids as biomarkers that forecast disease risk and as targets of ceramide-lowering interventions that ameliorate disease.
Collapse
Affiliation(s)
- Joseph L Wilkerson
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, United States
| | - Sean M Tatum
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, United States
| | - William L Holland
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, United States
| | - Scott A Summers
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, United States
| |
Collapse
|
15
|
Tan M, Liu Y, Xu Y, Yan G, Zhou N, Chen H, Jiang Z, Peng L. Plant-Derived Exosomes as Novel Nanotherapeutics Contrive Glycolysis Reprogramming-Mediated Angiogenesis for Diabetic Ulcer Healing. Biomater Res 2024; 28:0035. [PMID: 38840655 PMCID: PMC11151174 DOI: 10.34133/bmr.0035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 04/26/2024] [Indexed: 06/07/2024] Open
Abstract
Reversal of endothelial cell (EC) dysfunction under high-glucose (HG) conditions to achieve angiogenesis has remained a big challenge in diabetic ulcers. Herein, exosomes derived from medicinal plant ginseng (GExos) were shown as excellent nanotherapeutics with biomimetic cell membrane-like structures to be able to efficiently transfer the encapsulated active substances to ECs, resulting in a marked reprogramming of glycolysis by up-regulating anaerobic glycolysis and down-regulating oxidative stress, which further restore the proliferation, migration, and tubule formation abilities of ECs under HG conditions. In vivo, GExos enhance the angiogenesis and nascent vessel network reconstruction in full-thickness diabetic complicated skin ulcer wounds in mice with high biosafety. GExos were shown as promising nanotherapeutics in stimulating glycolysis reprogramming-mediated angiogenesis in diabetic ulcers, possessing wide application potential for reversing hyperglycemic dysangiogenesis and stimulating vascular regeneration.
Collapse
Affiliation(s)
- Minhong Tan
- College of Pharmaceutical Sciences,
Zhejiang University, Hangzhou 310058, PR China
- School of Materials Science and Engineering,
Zhejiang University, Hangzhou 310058, PR China
| | - Yuda Liu
- College of Pharmaceutical Sciences,
Zhejiang University, Hangzhou 310058, PR China
| | - Yang Xu
- College of Pharmaceutical Sciences,
Zhejiang University, Hangzhou 310058, PR China
| | - Ge Yan
- College of Pharmaceutical Sciences,
Zhejiang University, Hangzhou 310058, PR China
| | - Nan Zhou
- College of Pharmaceutical Sciences,
Zhejiang University, Hangzhou 310058, PR China
| | - Haoran Chen
- College of Pharmaceutical Sciences,
Zhejiang University, Hangzhou 310058, PR China
| | - Zhihong Jiang
- State Key Laboratory of Quality Research in Chinese Medicine,
Macau University of Science and Technology, Macau, PR China
| | - Lihua Peng
- College of Pharmaceutical Sciences,
Zhejiang University, Hangzhou 310058, PR China
- State Key Laboratory of Quality Research in Chinese Medicine,
Macau University of Science and Technology, Macau, PR China
| |
Collapse
|
16
|
Radulescu D, Mihai FD, Trasca MET, Caluianu EI, Calafeteanu CDM, Radulescu PM, Mercut R, Ciupeanu-Calugaru ED, Marinescu GA, Siloşi CA, Nistor CCE, Danoiu S. Oxidative Stress in Military Missions-Impact and Management Strategies: A Narrative Analysis. Life (Basel) 2024; 14:567. [PMID: 38792589 PMCID: PMC11121804 DOI: 10.3390/life14050567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024] Open
Abstract
This narrative review comprehensively examines the impact of oxidative stress on military personnel, highlighting the crucial role of physical exercise and tailored diets, particularly the ketogenic diet, in minimizing this stress. Through a meticulous analysis of the recent literature, the study emphasizes how regular physical exercise not only enhances cardiovascular, cognitive, and musculoskeletal health but is also essential in neutralizing the effects of oxidative stress, thereby improving endurance and performance during long-term missions. Furthermore, the implementation of the ketogenic diet provides an efficient and consistent energy source through ketone bodies, tailored to the specific energy requirements of military activities, and significantly contributes to the reduction in reactive oxygen species production, thus protecting against cellular deterioration under extreme stress. The study also underlines the importance of integrating advanced technologies, such as wearable devices and smart sensors that allow for the precise and real-time monitoring of oxidative stress and physiological responses, thus facilitating the customization of training and nutritional regimes. Observations from this review emphasize significant variability among individuals in responses to oxidative stress, highlighting the need for a personalized approach in formulating intervention strategies. It is crucial to develop and implement well-monitored, personalized supplementation protocols to ensure that each member of the military personnel receives a regimen tailored to their specific needs, thereby maximizing the effectiveness of measures to combat oxidative stress. This analysis makes a valuable contribution to the specialized literature, proposing a detailed framework for addressing oxidative stress in the armed forces and opening new directions for future research with the aim of optimizing clinical practices and improving the health and performance of military personnel under stress and specific challenges of the military field.
Collapse
Affiliation(s)
- Dumitru Radulescu
- Department of Surgery, The Military Emergency Clinical Hospital ‘Dr. Stefan Odobleja’ Craiova, 200749 Craiova, Romania; (D.R.); (E.-I.C.); (P.-M.R.); (G.-A.M.)
| | - Florina-Diana Mihai
- Doctoral School, University of Medicine and Pharmacy of Craiova, 2 Petru Rares Street, 200349 Craiova, Romania;
| | - Major Emil-Tiberius Trasca
- Department of Surgery, The Military Emergency Clinical Hospital ‘Dr. Stefan Odobleja’ Craiova, 200749 Craiova, Romania; (D.R.); (E.-I.C.); (P.-M.R.); (G.-A.M.)
| | - Elena-Irina Caluianu
- Department of Surgery, The Military Emergency Clinical Hospital ‘Dr. Stefan Odobleja’ Craiova, 200749 Craiova, Romania; (D.R.); (E.-I.C.); (P.-M.R.); (G.-A.M.)
| | - Captain Dan Marian Calafeteanu
- Department of Ortopedics, The Military Emergency Clinical Hospital ‘Dr. Stefan Odobleja’ Craiova, 200749 Craiova, Romania;
| | - Patricia-Mihaela Radulescu
- Department of Surgery, The Military Emergency Clinical Hospital ‘Dr. Stefan Odobleja’ Craiova, 200749 Craiova, Romania; (D.R.); (E.-I.C.); (P.-M.R.); (G.-A.M.)
| | - Razvan Mercut
- Department of Plastic and Reconstructive Surgery, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | | | - Georgiana-Andreea Marinescu
- Department of Surgery, The Military Emergency Clinical Hospital ‘Dr. Stefan Odobleja’ Craiova, 200749 Craiova, Romania; (D.R.); (E.-I.C.); (P.-M.R.); (G.-A.M.)
| | - Cristian-Adrian Siloşi
- Doctoral School, University of Medicine and Pharmacy of Craiova, 2 Petru Rares Street, 200349 Craiova, Romania;
| | | | - Suzana Danoiu
- Department of Pathophysiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| |
Collapse
|
17
|
Münzel T, Molitor M, Kuntic M, Hahad O, Röösli M, Engelmann N, Basner M, Daiber A, Sørensen M. Transportation Noise Pollution and Cardiovascular Health. Circ Res 2024; 134:1113-1135. [PMID: 38662856 DOI: 10.1161/circresaha.123.323584] [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] [Indexed: 05/29/2024]
Abstract
Epidemiological studies have found that transportation noise increases the risk for cardiovascular morbidity and mortality, with solid evidence for ischemic heart disease, heart failure, and stroke. According to the World Health Organization, at least 1.6 million healthy life years are lost annually from traffic-related noise in Western Europe. Traffic noise at night causes fragmentation and shortening of sleep, elevation of stress hormone levels, and increased oxidative stress in the vasculature and the brain. These factors can promote vascular (endothelial) dysfunction, inflammation, and arterial hypertension, thus elevating cardiovascular risk. The present review focusses on the indirect, nonauditory cardiovascular health effects of noise. We provide an updated overview of epidemiological research on the effects of transportation noise on cardiovascular risk factors and disease, and mechanistic insights based on the latest clinical and experimental studies and propose new risk markers to address noise-induced cardiovascular effects in the general population. We will discuss the potential effects of noise on vascular dysfunction, oxidative stress, and inflammation in humans and animals. We will elaborately explain the underlying pathomechanisms by alterations of gene networks, epigenetic pathways, circadian rhythm, signal transduction along the neuronal-cardiovascular axis, and metabolism. We will describe current and future noise mitigation strategies. Finally, we will conduct an overall evaluation of the status of the current evidence of noise as a significant cardiovascular risk factor.
Collapse
Affiliation(s)
- Thomas Münzel
- Department of Cardiology, University Medical Center Mainz, Germany (T.M., M.M., M.K., O.H., A.D.)
- German Centre for Cardiovascular Research (DZHK), Rhine-Main, Germany (T.M., M.M., O.H., A.D.)
| | - Michael Molitor
- Department of Cardiology, University Medical Center Mainz, Germany (T.M., M.M., M.K., O.H., A.D.)
- German Centre for Cardiovascular Research (DZHK), Rhine-Main, Germany (T.M., M.M., O.H., A.D.)
| | - Marin Kuntic
- Department of Cardiology, University Medical Center Mainz, Germany (T.M., M.M., M.K., O.H., A.D.)
| | - Omar Hahad
- Department of Cardiology, University Medical Center Mainz, Germany (T.M., M.M., M.K., O.H., A.D.)
- German Centre for Cardiovascular Research (DZHK), Rhine-Main, Germany (T.M., M.M., O.H., A.D.)
| | - Martin Röösli
- Swiss Tropical and Public Health Institute, Department Epidemiology and Public Health, University of Basel, Switzerland (M.R., N.E.)
| | - Nicole Engelmann
- Swiss Tropical and Public Health Institute, Department Epidemiology and Public Health, University of Basel, Switzerland (M.R., N.E.)
| | - Mathias Basner
- Unit for Experimental Psychiatry, Division of Sleep and Chronobiology, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA (M.B.)
| | - Andreas Daiber
- Department of Cardiology, University Medical Center Mainz, Germany (T.M., M.M., M.K., O.H., A.D.)
- German Centre for Cardiovascular Research (DZHK), Rhine-Main, Germany (T.M., M.M., O.H., A.D.)
| | - Mette Sørensen
- Danish Cancer Institute, Danish Cancer Society, Copenhagen, Denmark (M.S.)
- Department of Natural Science and Environment, Roskilde University, Denmark (M.S.)
| |
Collapse
|
18
|
van der Velden AIM, IJpelaar DHT, Chandie Shaw PK, Pijl H, Vink H, van der Vlag J, Rabelink TJ, van den Berg BM. Role of dietary interventions on microvascular health in South-Asian Surinamese people with type 2 diabetes in the Netherlands: A randomized controlled trial. Nutr Diabetes 2024; 14:17. [PMID: 38600065 PMCID: PMC11006941 DOI: 10.1038/s41387-024-00275-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 03/25/2024] [Accepted: 04/03/2024] [Indexed: 04/12/2024] Open
Abstract
BACKGROUND/OBJECTIVES We investigated whether dietary interventions, i.e. a fasting mimicking diet (FMD, Prolon®) or glycocalyx mimetic supplementation (EndocalyxTM) could stabilize microvascular function in Surinamese South-Asian patients with type 2 diabetes (SA-T2DM) in the Netherlands, a patient population more prone to develop vascular complications. SUBJECTS/METHODS A randomized, placebo controlled, 3-arm intervention study was conducted in 56 SA-T2DM patients between 18 and 75 years old, for 3 consecutive months, with one additional follow up measurement 3 months after the last intervention. Sublingual microcirculation was assessed with SDF-imaging coupled to the GlycoCheckTM software, detecting red blood cell velocity, capillary density, static and dynamic perfused boundary region (PBR), and the overall microvascular health score (MVHS). Linear mixed models and interaction analysis were used to investigate the effects the interventions had on microvascular function. RESULTS Despite a temporal improvement in BMI and HbA1c after FMD the major treatment effect on microvascular health was worsening for RBC-velocity independent PBRdynamic, especially at follow-up. Glycocalyx supplementation, however, reduced urinary MCP-1 presence and improved both PBRdynamic and MVHSdynamic, which persisted at follow-up. CONCLUSIONS We showed that despite temporal beneficial changes in BMI and HbA1c after FMD, this intervention is not able to preserve microvascular endothelial health in Dutch South-Asian patients with T2DM. In contrast, glycocalyx mimetics preserves the microvascular endothelial health and reduces the inflammatory cytokine MCP-1. CLINICAL STUDY REGISTRATION NCT03889236.
Collapse
Affiliation(s)
- Anouk I M van der Velden
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory of Vascular and Regenerative Medicine, LUMC, Leiden, The Netherlands
| | - Daphne H T IJpelaar
- Department of Internal Medicine and Nephrology, Green Heart Hospital, Gouda, The Netherlands
| | - Prataap K Chandie Shaw
- Department of Internal Medicine and Nephrology, Haaglanden Medical Center, The Hague, The Netherlands
| | - Hanno Pijl
- Einthoven Laboratory of Vascular and Regenerative Medicine, LUMC, Leiden, The Netherlands
- Department of Internal Medicine (Endocrinology), LUMC, Leiden, The Netherlands
| | - Hans Vink
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht, The Netherlands
- MicroVascular Health Solutions LLC, Alpine, Utah, USA
| | - Johan van der Vlag
- Department of Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ton J Rabelink
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory of Vascular and Regenerative Medicine, LUMC, Leiden, The Netherlands
| | - Bernard M van den Berg
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, The Netherlands.
- Einthoven Laboratory of Vascular and Regenerative Medicine, LUMC, Leiden, The Netherlands.
| |
Collapse
|
19
|
Ma S, Xie X, Yuan R, Xin Q, Miao Y, Leng SX, Chen K, Cong W. Vascular Aging and Atherosclerosis: A Perspective on Aging. Aging Dis 2024; 16:AD.2024.0201-1. [PMID: 38502584 PMCID: PMC11745439 DOI: 10.14336/ad.2024.0201-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 02/01/2024] [Indexed: 03/21/2024] Open
Abstract
Vascular aging (VA) is recognized as a pivotal factor in the development and progression of atherosclerosis (AS). Although various epidemiological and clinical research has demonstrated an intimate connection between aging and AS, the candidate mechanisms still require thorough examination. This review adopts an aging-centric perspective to deepen the comprehension of the intricate relationship between biological aging, vascular cell senescence, and AS. Various aging-related physiological factors influence the physical system's reactions, including oxygen radicals, inflammation, lipids, angiotensin II, mechanical forces, glucose levels, and insulin resistance. These factors cause endothelial dysfunction, barrier damage, sclerosis, and inflammation for VA and promote AS via distinct or shared pathways. Furthermore, the increase of senescent cells inside the vascular tissues, caused by genetic damage, dysregulation, secretome changes, and epigenetic modifications, might be the primary cause of VA.
Collapse
Affiliation(s)
- Shudong Ma
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China.
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Xuena Xie
- School of Pharmacy, Macau University of Science and Technology, Macau, China.
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Rong Yuan
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Qiqi Xin
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Yu Miao
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Sean Xiao Leng
- Division of Geriatric Medicine and Gerontology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA.
| | - Keji Chen
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China.
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Weihong Cong
- School of Pharmacy, Macau University of Science and Technology, Macau, China.
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
20
|
Tang S, An X, Sun W, Zhang Y, Yang C, Kang X, Sun Y, Jiang L, Zhao X, Gao Q, Ji H, Lian F. Parallelism and non-parallelism in diabetic nephropathy and diabetic retinopathy. Front Endocrinol (Lausanne) 2024; 15:1336123. [PMID: 38419958 PMCID: PMC10899692 DOI: 10.3389/fendo.2024.1336123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 01/23/2024] [Indexed: 03/02/2024] Open
Abstract
Diabetic nephropathy (DN) and diabetic retinopathy (DR), as microvascular complications of diabetes mellitus, are currently the leading causes of end-stage renal disease (ESRD) and blindness, respectively, in the adult working population, and they are major public health problems with social and economic burdens. The parallelism between the two in the process of occurrence and development manifests in the high overlap of disease-causing risk factors and pathogenesis, high rates of comorbidity, mutually predictive effects, and partial concordance in the clinical use of medications. However, since the two organs, the eye and the kidney, have their unique internal environment and physiological processes, each with specific influencing molecules, and the target organs have non-parallelism due to different pathological changes and responses to various influencing factors, this article provides an overview of the parallelism and non-parallelism between DN and DR to further recognize the commonalities and differences between the two diseases and provide references for early diagnosis, clinical guidance on the use of medication, and the development of new drugs.
Collapse
Affiliation(s)
- Shanshan Tang
- College of Traditional Chinese Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Xuedong An
- Guang’an Men Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Wenjie Sun
- Guang’an Men Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuehong Zhang
- Fangshan Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Cunqing Yang
- Guang’an Men Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaomin Kang
- Guang’an Men Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuting Sun
- Guang’an Men Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Linlin Jiang
- Guang’an Men Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Xuefei Zhao
- Guang’an Men Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Qing Gao
- Guang’an Men Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Hangyu Ji
- Guang’an Men Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Fengmei Lian
- Guang’an Men Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
21
|
Yang C, Zhu Q, Chen Y, Ji K, Li S, Wu Q, Pan Q, Li J. Review of the Protective Mechanism of Curcumin on Cardiovascular Disease. Drug Des Devel Ther 2024; 18:165-192. [PMID: 38312990 PMCID: PMC10838105 DOI: 10.2147/dddt.s445555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 01/16/2024] [Indexed: 02/06/2024] Open
Abstract
Cardiovascular diseases (CVDs) are the most common cause of death worldwide and has been the focus of research in the medical community. Curcumin is a polyphenolic compound extracted from the root of turmeric. Curcumin has been shown to have a variety of pharmacological properties over the past decades. Curcumin can significantly protect cardiomyocyte injury after ischemia and hypoxia, inhibit myocardial hypertrophy and fibrosis, improve ventricular remodeling, reduce drug-induced myocardial injury, improve diabetic cardiomyopathy(DCM), alleviate vascular endothelial dysfunction, inhibit foam cell formation, and reduce vascular smooth muscle cells(VSMCs) proliferation. Clinical studies have shown that curcumin has a protective effect on blood vessels. Toxicological studies have shown that curcumin is safe. But high doses of curcumin also have some side effects, such as liver damage and defects in embryonic heart development. This article reviews the mechanism of curcumin intervention on CVDs in recent years, in order to provide reference for the development of new drugs in the future.
Collapse
Affiliation(s)
- Chunkun Yang
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Qinwei Zhu
- Department of Emergency, Weifang Hospital of Traditional Chinese Medicine, Weifang, Shandong, People's Republic of China
| | - Yanbo Chen
- Department of Arrhythmia, Weifang People's Hospital, Weifang, Shandong, People's Republic of China
| | - Kui Ji
- Department of Emergency, Weifang Hospital of Traditional Chinese Medicine, Weifang, Shandong, People's Republic of China
| | - Shuanghong Li
- Department of Emergency, Weifang Hospital of Traditional Chinese Medicine, Weifang, Shandong, People's Republic of China
| | - Qian Wu
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Qingquan Pan
- Department of Emergency, Weifang Hospital of Traditional Chinese Medicine, Weifang, Shandong, People's Republic of China
| | - Jun Li
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| |
Collapse
|
22
|
Buonfiglio F, Wasielica-Poslednik J, Pfeiffer N, Gericke A. Diabetic Keratopathy: Redox Signaling Pathways and Therapeutic Prospects. Antioxidants (Basel) 2024; 13:120. [PMID: 38247544 PMCID: PMC10812573 DOI: 10.3390/antiox13010120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/10/2024] [Accepted: 01/16/2024] [Indexed: 01/23/2024] Open
Abstract
Diabetes mellitus, the most prevalent endocrine disorder, not only impacts the retina but also significantly involves the ocular surface. Diabetes contributes to the development of dry eye disease and induces morphological and functional corneal alterations, particularly affecting nerves and epithelial cells. These changes manifest as epithelial defects, reduced sensitivity, and delayed wound healing, collectively encapsulated in the context of diabetic keratopathy. In advanced stages of this condition, the progression to corneal ulcers and scarring further unfolds, eventually leading to corneal opacities. This critical complication hampers vision and carries the potential for irreversible visual loss. The primary objective of this review article is to offer a comprehensive overview of the pathomechanisms underlying diabetic keratopathy. Emphasis is placed on exploring the redox molecular pathways responsible for the aberrant structural changes observed in the cornea and tear film during diabetes. Additionally, we provide insights into the latest experimental findings concerning potential treatments targeting oxidative stress. This endeavor aims to enhance our understanding of the intricate interplay between diabetes and ocular complications, offering valuable perspectives for future therapeutic interventions.
Collapse
Affiliation(s)
- Francesco Buonfiglio
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (J.W.-P.); (N.P.)
| | | | | | - Adrian Gericke
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (J.W.-P.); (N.P.)
| |
Collapse
|
23
|
Rabbani N, Thornalley PJ. Hexokinase-linked glycolytic overload and unscheduled glycolysis in hyperglycemia-induced pathogenesis of insulin resistance, beta-cell glucotoxicity, and diabetic vascular complications. Front Endocrinol (Lausanne) 2024; 14:1268308. [PMID: 38292764 PMCID: PMC10824962 DOI: 10.3389/fendo.2023.1268308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 12/12/2023] [Indexed: 02/01/2024] Open
Abstract
Hyperglycemia is a risk factor for the development of insulin resistance, beta-cell glucotoxicity, and vascular complications of diabetes. We propose the hypothesis, hexokinase-linked glycolytic overload and unscheduled glycolysis, in explanation. Hexokinases (HKs) catalyze the first step of glucose metabolism. Increased flux of glucose metabolism through glycolysis gated by HKs, when occurring without concomitant increased activity of glycolytic enzymes-unscheduled glycolysis-produces increased levels of glycolytic intermediates with overspill into effector pathways of cell dysfunction and pathogenesis. HK1 is saturated with glucose in euglycemia and, where it is the major HK, provides for basal glycolytic flux without glycolytic overload. HK2 has similar saturation characteristics, except that, in persistent hyperglycemia, it is stabilized to proteolysis by high intracellular glucose concentration, increasing HK activity and initiating glycolytic overload and unscheduled glycolysis. This drives the development of vascular complications of diabetes. Similar HK2-linked unscheduled glycolysis in skeletal muscle and adipose tissue in impaired fasting glucose drives the development of peripheral insulin resistance. Glucokinase (GCK or HK4)-linked glycolytic overload and unscheduled glycolysis occurs in persistent hyperglycemia in hepatocytes and beta-cells, contributing to hepatic insulin resistance and beta-cell glucotoxicity, leading to the development of type 2 diabetes. Downstream effector pathways of HK-linked unscheduled glycolysis are mitochondrial dysfunction and increased reactive oxygen species (ROS) formation; activation of hexosamine, protein kinase c, and dicarbonyl stress pathways; and increased Mlx/Mondo A signaling. Mitochondrial dysfunction and increased ROS was proposed as the initiator of metabolic dysfunction in hyperglycemia, but it is rather one of the multiple downstream effector pathways. Correction of HK2 dysregulation is proposed as a novel therapeutic target. Pharmacotherapy addressing it corrected insulin resistance in overweight and obese subjects in clinical trial. Overall, the damaging effects of hyperglycemia are a consequence of HK-gated increased flux of glucose metabolism without increased glycolytic enzyme activities to accommodate it.
Collapse
Affiliation(s)
| | - Paul J. Thornalley
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| |
Collapse
|
24
|
Di Pietro P, Abate AC, Prete V, Damato A, Venturini E, Rusciano MR, Izzo C, Visco V, Ciccarelli M, Vecchione C, Carrizzo A. C2CD4B Evokes Oxidative Stress and Vascular Dysfunction via a PI3K/Akt/PKCα-Signaling Pathway. Antioxidants (Basel) 2024; 13:101. [PMID: 38247525 PMCID: PMC10812653 DOI: 10.3390/antiox13010101] [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: 11/29/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 01/23/2024] Open
Abstract
High glucose-induced endothelial dysfunction is an important pathological feature of diabetic vasculopathy. While genome-wide studies have identified an association between type 2 diabetes mellitus (T2DM) and increased expression of a C2 calcium-dependent domain containing 4B (C2CD4B), no study has yet explored the possible direct effect of C2CD4B on vascular function. Vascular reactivity studies were conducted using a pressure myograph, and nitric oxide and oxidative stress were assessed through difluorofluorescein diacetate and dihydroethidium, respectively. We demonstrate that high glucose upregulated both mRNA and protein expression of C2CD4B in mice mesenteric arteries in a time-dependent manner. Notably, the inhibition of C2CD4B expression by genetic knockdown efficiently prevented hyperglycemia-induced oxidative stress, endothelial dysfunction, and loss of nitric oxide (NO) bioavailability. Recombinant C2CD4B evoked endothelial dysfunction of mice mesenteric arteries, an effect associated with increased reactive oxygen species (ROS) and decreased NO production. In isolated human umbilical vein endothelial cells (HUVECs), C2CD4B increased phosphorylation of endothelial nitric oxide synthase (eNOS) at the inhibitory site Thr495 and reduced eNOS dimerization. Pharmacological inhibitors of phosphoinositide 3-kinase (PI3K), Akt, and PKCα effectively attenuated oxidative stress, NO reduction, impairment of endothelial function, and eNOS uncoupling induced by C2CD4B. These data demonstrate, for the first time, that C2CD4B exerts a direct effect on vascular endothelium via a phosphoinositide 3-kinase (PI3K)/Akt/PKCα-signaling pathway, providing a new perspective on C2CD4B as a promising therapeutic target for the prevention of oxidative stress in diabetes-induced endothelial dysfunction.
Collapse
Affiliation(s)
- Paola Di Pietro
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (P.D.P.); (A.C.A.); (V.P.); (M.R.R.); (C.I.); (V.V.); (M.C.); (C.V.)
| | - Angela Carmelita Abate
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (P.D.P.); (A.C.A.); (V.P.); (M.R.R.); (C.I.); (V.V.); (M.C.); (C.V.)
| | - Valeria Prete
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (P.D.P.); (A.C.A.); (V.P.); (M.R.R.); (C.I.); (V.V.); (M.C.); (C.V.)
| | - Antonio Damato
- Vascular Physiopathology Unit, IRCCS Neuromed, 86077 Pozzilli, Italy; (A.D.); (E.V.)
| | - Eleonora Venturini
- Vascular Physiopathology Unit, IRCCS Neuromed, 86077 Pozzilli, Italy; (A.D.); (E.V.)
| | - Maria Rosaria Rusciano
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (P.D.P.); (A.C.A.); (V.P.); (M.R.R.); (C.I.); (V.V.); (M.C.); (C.V.)
| | - Carmine Izzo
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (P.D.P.); (A.C.A.); (V.P.); (M.R.R.); (C.I.); (V.V.); (M.C.); (C.V.)
| | - Valeria Visco
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (P.D.P.); (A.C.A.); (V.P.); (M.R.R.); (C.I.); (V.V.); (M.C.); (C.V.)
| | - Michele Ciccarelli
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (P.D.P.); (A.C.A.); (V.P.); (M.R.R.); (C.I.); (V.V.); (M.C.); (C.V.)
| | - Carmine Vecchione
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (P.D.P.); (A.C.A.); (V.P.); (M.R.R.); (C.I.); (V.V.); (M.C.); (C.V.)
- Vascular Physiopathology Unit, IRCCS Neuromed, 86077 Pozzilli, Italy; (A.D.); (E.V.)
| | - Albino Carrizzo
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (P.D.P.); (A.C.A.); (V.P.); (M.R.R.); (C.I.); (V.V.); (M.C.); (C.V.)
- Vascular Physiopathology Unit, IRCCS Neuromed, 86077 Pozzilli, Italy; (A.D.); (E.V.)
| |
Collapse
|
25
|
Vedika R, Sharma P, Reddy A. Signature precursor and mature microRNAs in cervical ripening during gestational diabetes mellitus lead to pre-term labor and other impediments in future. J Diabetes Metab Disord 2023; 22:945-965. [PMID: 37975145 PMCID: PMC10638342 DOI: 10.1007/s40200-023-01232-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 04/29/2023] [Indexed: 11/19/2023]
Abstract
Gestational diabetes mellitus (GDM) is a pathological condition in which the placenta releases a hormone called human placental lactogen that prevents maternal insulin uptake. GDM is characterised by varying degrees of carbohydrate intolerance and is first identified during pregnancy. Around 5-17% of pregnancies are GDM pregnancies. Older or obese women have a higher risk of developing GDM during gestation. Hyperglycemia is a classic manifestation of GDM and leads to alterations in eNOS and iNOS expression and subsequently causes ROS and RNS overproduction. ROS and RNS play an important role in maintaining normal physiology, when present in low concentrations. Increased concentrations of ROS is harmful and can cause cellular and tissue damage. Oxidative stress is defined as an imbalance between pro-oxidant and antioxidant molecules that manifests due to hyperglycemia. miRNAs are short, non-coding RNAs that play a critical role in regulating gene expression. Studies have shown that the placenta expresses more than 500 miRNAs, which play a crucial role in trophoblast division, movement, and apoptosis. Latest research has revealed that hyperglycemic conditions and increased oxidative stress, characteristic of GDM, can lead to the dysregulation of miRNAs. The placenta also releases miRNAs into the maternal circulation. The secreted miRNAs are encapsulated in exosomes or vesicles. These exosomes interact with tissues and organs at distant sites, releasing their cargo intracellularly. This crosstalk between hyperglycemia, ROS and miRNA expression in GDM has detrimental effects on both foetal and maternal health. One of the complications of GDM is preterm labour. GDM induced iNOS expression has been implicated in cervical ripening, which in turn causes preterm birth. This article focuses on the speculations of oxidative and nitrative stress markers that lead to detrimental effects in GDM. We have also envisaged the role of non-coding miRNA interactions in regulating gene expression for oxidative damage. Graphical Abstract Holistic view of miRNA in GDM. I)(A) Placenta as a metabolic organ that provides the foetus with nutrients, oxygen and hormones to maintain pregnancy. Human placental lactogen (hPL) is one such hormone that is released into maternal circulation. hPL is known to induce insulin resistance. (B) ß-cell dysfunction leads to reduced glucose sensing and insulin production. Insulin resistance, a characteristic of GDM, exacerbates insulin ß cell dysfunction leading to maternal hyperglycemia. Hyperglycemia leads to increased ROS and RNS production through several mechanisms. Consequently, GDM is characterised by increased oxidative and nitrative stress.II)Exposure to maternal hyperglycemia causes increased ROS and RNS production in trophoblast cells. Oxidative stress caused by hyperglycemia may lead to eNOS uncoupling, causing eNOS to behave as a superoxide producing enzyme. iNOS expression in trophoblast cells leads to increased NO production. iNOS-derived NO reacts with ROS to produce RNS, thereby increasing nitrosative stress. Expression of antioxidant defences are reduced. Hyperglycemia and oxidative stress may alter the expression of some miRNAs. Some miRNAs are upregulated while others are downregulated. Some miRNAs are secreted into maternal circulation in the form of exosomes. Oxidative stress markers, nitrative stress markers and circulating miRNAs are found to be increased in maternal circulation.
Collapse
Affiliation(s)
- R. Vedika
- Animal cell culture laboratory, Department of Biotechnology, SRMIST, Kattankulathur, Tamil Nadu India
| | - Priyanshy Sharma
- Animal cell culture laboratory, Department of Biotechnology, SRMIST, Kattankulathur, Tamil Nadu India
| | - Amala Reddy
- Animal cell culture laboratory, Department of Biotechnology, SRMIST, Kattankulathur, Tamil Nadu India
- Department of Biotechnology, SRMIST, Kattankulathur, Kancheepuram 603203 India
| |
Collapse
|
26
|
Haba D, Qin Q, Takizawa C, Tomida S, Minematsu T, Sanada H, Nakagami G. Local low-frequency vibration accelerates healing of full-thickness wounds in a hyperglycemic rat model. J Diabetes Investig 2023; 14:1356-1367. [PMID: 37688317 PMCID: PMC10688122 DOI: 10.1111/jdi.14072] [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: 04/07/2023] [Revised: 07/06/2023] [Accepted: 08/07/2023] [Indexed: 09/10/2023] Open
Abstract
AIMS/INTRODUCTION Local low-frequency vibration (LLFV) promotes vasodilation and blood flow, enhancing wound healing in diabetic foot ulcers with angiopathy. However, vibration-induced vasodilation does not occur, owing to chronic hyperglycemia and inflammation. We hypothesized that LLFV improves glycometabolism and inflammation, leading to vasodilation and angiogenesis in diabetic wounds. Therefore, this study investigated the effect of LLFV on wound healing in hyperglycemic rats, primarily focusing on glycometabolism, inflammation, vasodilation, and angiogenesis. MATERIALS AND METHODS Streptozotocin-induced hyperglycemic Sprague-Dawley rats were used in this study. We applied LLFV to experimentally-induced wounds at 50 Hz and 0, 600, 1,000 or 1,500 mVpp for 40 min/day from post-wounding days (PWD) 1-14. RESULTS The relative wound areas in the 600 and 1,000 mVpp groups on PWD 5-7 were significantly smaller than those at 0 mVpp. The expression of Glo-1 (1,500 mVpp) and Slc2A4 (1,000 and 1,500 mVpp) was upregulated on PWD 4 and 14, respectively. However, there was no difference in methylglyoxal expression levels in any group until PWD 14. At 1,000 mVpp, the expression of Tnfa on PWD 4, and that of Ptx3 and Ccl2 on PWD 14 was downregulated. Furthermore, the M1/M2 macrophage ratio was considerably decreased on both days. The expression of Nos3, Vegfa and vascular endothelial growth factor A was upregulated on PWD 4. In addition, vasodilation and angiogenesis were more obvious on PWD 14 with 1,000 mVpp. CONCLUSIONS The results suggest that LLFV promotes wound healing, improves glycometabolism and inflammation, and enhances vasodilation and angiogenesis in hyperglycemic wounds.
Collapse
Affiliation(s)
- Daijiro Haba
- Global Nursing Research Center, Graduate School of MedicineThe University of TokyoTokyoJapan
- Department of Gerontological Nursing/Wound Care Management, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Qi Qin
- Department of Gerontological Nursing/Wound Care Management, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Chihiro Takizawa
- Department of Gerontological Nursing/Wound Care Management, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Sanai Tomida
- Department of Gerontological Nursing/Wound Care Management, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Takeo Minematsu
- Global Nursing Research Center, Graduate School of MedicineThe University of TokyoTokyoJapan
- Department of Skincare Science, Graduate School of MedicineThe University of TokyoTokyoJapan
- Ishikawa Prefectural Nursing UniversityIshikawaJapan
| | - Hiromi Sanada
- Global Nursing Research Center, Graduate School of MedicineThe University of TokyoTokyoJapan
- Department of Gerontological Nursing/Wound Care Management, Graduate School of MedicineThe University of TokyoTokyoJapan
- Ishikawa Prefectural Nursing UniversityIshikawaJapan
| | - Gojiro Nakagami
- Global Nursing Research Center, Graduate School of MedicineThe University of TokyoTokyoJapan
- Department of Gerontological Nursing/Wound Care Management, Graduate School of MedicineThe University of TokyoTokyoJapan
| |
Collapse
|
27
|
Wang LF, Li Q, Wen K, Zhao QH, Zhang YT, Zhao JL, Ding Q, Guan XH, Xiao YF, Deng KY, Xin HB. CD38 Deficiency Alleviates Diabetic Cardiomyopathy by Coordinately Inhibiting Pyroptosis and Apoptosis. Int J Mol Sci 2023; 24:16008. [PMID: 37958991 PMCID: PMC10650707 DOI: 10.3390/ijms242116008] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/29/2023] [Accepted: 11/03/2023] [Indexed: 11/15/2023] Open
Abstract
Diabetic cardiomyopathy is one of the diabetes mellitus-induced cardiovascular complications that can result in heart failure in severe cases, which is characterized by cardiomyocyte apoptosis, local inflammation, oxidative stress, and myocardial fibrosis. CD38, a main hydrolase of NAD+ in mammals, plays an important role in various cardiovascular diseases, according to our previous studies. However, the role of CD38 in diabetes-induced cardiomyopathy is still unknown. Here, we report that global deletion of the CD38 gene significantly prevented diabetic cardiomyopathy induced by high-fat diet plus streptozotocin (STZ) injection in CD38 knockout (CD38-KO) mice. We observed that CD38 expression was up-regulated, whereas the expression of Sirt3 was down-regulated in the hearts of diabetic mice. CD38 deficiency significantly promoted glucose metabolism and improved cardiac functions, exemplified by increased left ventricular ejection fraction and fractional shortening. In addition, we observed that CD38 deficiency markedly decreased diabetes or high glucose and palmitic acid (HG + PA)-induced pyroptosis and apoptosis in CD38 knockout hearts or cardiomyocytes, respectively. Furthermore, we found that the expression levels of Sirt3, mainly located in mitochondria, and its target gene FOXO3a were increased in CD38-deficient hearts and cardiomyocytes with CD38 knockdown under diabetic induction conditions. In conclusion, we demonstrated that CD38 deficiency protected mice from diabetes-induced diabetic cardiomyopathy by reducing pyroptosis and apoptosis via activating NAD+/Sirt3/FOXO3a signaling pathways.
Collapse
Affiliation(s)
- Ling-Fang Wang
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China; (Q.L.); (K.W.); (Q.-H.Z.); (Y.-T.Z.); (J.-L.Z.); (Q.D.); (X.-H.G.); (Y.-F.X.); (K.-Y.D.)
| | | | | | | | | | | | | | | | | | | | - Hong-Bo Xin
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China; (Q.L.); (K.W.); (Q.-H.Z.); (Y.-T.Z.); (J.-L.Z.); (Q.D.); (X.-H.G.); (Y.-F.X.); (K.-Y.D.)
| |
Collapse
|
28
|
Lisi V, Senesi G, Balbi C. Converging protective pathways: Exploring the linkage between physical exercise, extracellular vesicles and oxidative stress. Free Radic Biol Med 2023; 208:718-727. [PMID: 37739138 DOI: 10.1016/j.freeradbiomed.2023.09.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/27/2023] [Accepted: 09/18/2023] [Indexed: 09/24/2023]
Abstract
Physical Exercise (EXR) has been shown to have numerous beneficial effects on various systems in the human body. It leads to a decrease in the risk of mortality from chronic diseases, such as cardiovascular disease, cancer, metabolic and central nervous system disorders. EXR results in improving cardiovascular fitness, cognitive function, immune activity, endocrine action, and musculoskeletal health. These positive effects make EXR a valuable intervention for promoting overall health and well-being in individuals of all ages. These beneficial effects are partially mediated by the role of the regular EXR in the adaptation to redox homeostasis counteracting the sudden increase of ROS, the hallmark of many chronic diseases. EXR can trigger the release of numerous humoral factors, e.g. protein, microRNA (miRs), and DNA, that can be shuttled as cargo of Extracellular vesicles (EVs). EVs show different cargo modification after oxidative stress stimuli as well as after EXR. In this review, we aim to highlight the main studies on the role of EVs released during EXR and oxidative stress conditions in enhancing the antioxidant enzymes pathway and in the decrease of oxidative stress environment mediated by their cargo.
Collapse
Affiliation(s)
- Veronica Lisi
- Unit of Biology and Genetics of Movement, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis 15, 00135 Rome, Italy.
| | - Giorgia Senesi
- Cellular and Molecular Cardiology, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Carolina Balbi
- Cellular and Molecular Cardiology, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland; Center for Molecular Cardiology, Zurich, Switzerland
| |
Collapse
|
29
|
Li H, Gao Y, Lin Y. Progress in molecular mechanisms of coronary microvascular dysfunction. Microcirculation 2023; 30:e12827. [PMID: 37608689 DOI: 10.1111/micc.12827] [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: 06/23/2023] [Accepted: 08/03/2023] [Indexed: 08/24/2023]
Abstract
Coronary microvascular dysfunction is a high-risk factor for many cardiovascular events. However, because of multiple risk factors and limited understanding about its underlying pathophysiological mechanisms, it was easily misdiagnosed. Therefore, its clinical diagnosis and treatment were greatly restricted. Coronary microcirculation refers to microvessels that play an important role in the physiological regulation of myocardial perfusion and regulating blood flow distribution, fulfilling myocardial metabolic needs and moderating peripheral vascular resistance. In coronary microvascular dysfunction, vascular endothelial celldamage is a critical link. The main feature of early coronary microvascular dysfunction is the impairment of endothelial cell proliferation, adhesion, migration, apoptosis, and secretion. Moreover, coronary microvascular dysfunction risk factors include hyperglycemia, lipid metabolism disorders, ischemia-reperfusion injury, aging, and hypertension, similar to coronary atherosclerosis. There are various mechanisms by which these risk factors harm endothelial function and cause microcirculatory disturbances. Therefore, we reviewed coronary microvascular dysfunction's risk factors and pathogenesis in this article.
Collapse
Affiliation(s)
- Hao Li
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Yuping Gao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Yuanyuan Lin
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
30
|
Xiao Q, Wang D, Li D, Huang J, Ma F, Zhang H, Sheng Y, Zhang C, Ha X. Protein kinase C: A potential therapeutic target for endothelial dysfunction in diabetes. J Diabetes Complications 2023; 37:108565. [PMID: 37540984 DOI: 10.1016/j.jdiacomp.2023.108565] [Citation(s) in RCA: 2] [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: 04/25/2023] [Revised: 07/13/2023] [Accepted: 07/22/2023] [Indexed: 08/06/2023]
Abstract
Protein kinase C (PKC) is a family of serine/threonine protein kinases that play an important role in many organs and systems and whose activation contributes significantly to endothelial dysfunction in diabetes. The increase in diacylglycerol (DAG) under high glucose conditions mediates PKC activation and synthesis, which stimulates oxidative stress and inflammation, resulting in impaired endothelial cell function. This article reviews the contribution of PKC to the development of diabetes-related endothelial dysfunction and summarizes the drugs that inhibit PKC activation, with the aim of exploring therapeutic modalities that may alleviate endothelial dysfunction in diabetic patients.
Collapse
Affiliation(s)
- Qian Xiao
- Department of Laboratory, Ninth Forty Hospital of the Chinese People's Liberation Army Joint Security Force, Lanzhou 730050, Gansu, China; School of Public Health, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, Gansu, China
| | - Dan Wang
- Department of Laboratory, Ninth Forty Hospital of the Chinese People's Liberation Army Joint Security Force, Lanzhou 730050, Gansu, China; School of Public Health, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, Gansu, China
| | - Danyang Li
- School of Public Health, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, Gansu, China
| | - Jing Huang
- Department of Laboratory, Ninth Forty Hospital of the Chinese People's Liberation Army Joint Security Force, Lanzhou 730050, Gansu, China; School of Public Health, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, Gansu, China
| | - Feifei Ma
- Department of Laboratory, Ninth Forty Hospital of the Chinese People's Liberation Army Joint Security Force, Lanzhou 730050, Gansu, China; College of Veterinary Medicine, Gansu Agriculture University, Lanzhou 730070, Gansu, China
| | - Haocheng Zhang
- Department of Laboratory, Ninth Forty Hospital of the Chinese People's Liberation Army Joint Security Force, Lanzhou 730050, Gansu, China; The Second School of Clinical Medicine, Lanzhou University, Lanzhou, 730030, Gansu, China
| | - Yingda Sheng
- Department of Laboratory, Ninth Forty Hospital of the Chinese People's Liberation Army Joint Security Force, Lanzhou 730050, Gansu, China; School of Public Health, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, Gansu, China
| | - Caimei Zhang
- Department of Laboratory, Ninth Forty Hospital of the Chinese People's Liberation Army Joint Security Force, Lanzhou 730050, Gansu, China; School of Public Health, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, Gansu, China
| | - Xiaoqin Ha
- Department of Laboratory, Ninth Forty Hospital of the Chinese People's Liberation Army Joint Security Force, Lanzhou 730050, Gansu, China; School of Public Health, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, Gansu, China.
| |
Collapse
|
31
|
Ghotbi Z, Estakhr M, Hosseini M, Shahripour RB. Cerebral Vasomotor Reactivity in COVID-19: A Narrative Review. Life (Basel) 2023; 13:1614. [PMID: 37511989 PMCID: PMC10381148 DOI: 10.3390/life13071614] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/09/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) primarily affects the respiratory system but can also lead to neurological complications. Among COVID-19 patients, the endothelium is considered the Achilles heel. A variety of endothelial dysfunctions may result from SARS-CoV-2 infection and subsequent endotheliitis, such as altered vascular tone, oxidative stress, and cytokine storms. The cerebral hemodynamic impairment that is caused is associated with a higher probability of severe disease and poor outcomes in patients with COVID-19. This review summarizes the most relevant literature on the role of vasomotor reactivity (VMR) in COVID-19 patients. An overview of the research articles is presented. Most of the studies have supported the hypothesis that endothelial dysfunction and cerebral VMR impairment occur in COVID-19 patients. Researchers believe these alterations may be due to direct viral invasion of the brain or indirect effects, such as inflammation and cytokines. Recently, researchers have concluded that viruses such as the Human Herpes Virus 8 and the Hantavirus predominantly affect endothelial cells and, therefore, affect cerebral hemodynamics. Especially in COVID-19 patients, impaired VMR is associated with a higher risk of severe disease and poor outcomes. Using VMR, one can gain valuable insight into a patient's disease progression and make more informed decisions regarding appropriate treatment options. A new pandemic may develop with the COVID-19 virus or other viruses, making it essential that healthcare providers and researchers remain focused on developing new strategies for improving survival in such patients, particularly those with cerebrovascular risk factors.
Collapse
Affiliation(s)
- Zahra Ghotbi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz P.O. Box 71348-14336, Iran
| | - Mehrdad Estakhr
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz P.O. Box 71348-14336, Iran
| | - Melika Hosseini
- Comprehensive Stroke Center, Department of Neurosciences, Loma Linda University, Loma Linda, CA 92354, USA
| | - Reza Bavarsad Shahripour
- Comprehensive Stroke Center, Department of Neurosciences, Loma Linda University, Loma Linda, CA 92354, USA
- UCSD Comprehensive Stroke Center, Department of Neurosciences, University of California, San Diego, CA 92093, USA
| |
Collapse
|
32
|
Bahadoran Z, Mirmiran P, Kashfi K, Ghasemi A. Vascular nitric oxide resistance in type 2 diabetes. Cell Death Dis 2023; 14:410. [PMID: 37433795 PMCID: PMC10336063 DOI: 10.1038/s41419-023-05935-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 06/18/2023] [Accepted: 06/28/2023] [Indexed: 07/13/2023]
Abstract
Vascular nitric oxide (NO•) resistance, manifested by an impaired vasodilator function of NO• in both the macro- and microvessels, is a common state in type 2 diabetes (T2D) associated with developing cardiovascular events and death. Here, we summarize experimental and human evidence of vascular NO• resistance in T2D and discuss its underlying mechanisms. Human studies indicate a ~ 13-94% decrease in the endothelium (ET)-dependent vascular smooth muscle (VSM) relaxation and a 6-42% reduced response to NO• donors, i.e., sodium nitroprusside (SNP) and glyceryl trinitrate (GTN), in patients with T2D. A decreased vascular NO• production, NO• inactivation, and impaired responsiveness of VSM to NO• [occurred due to quenching NO• activity, desensitization of its receptor soluble guanylate cyclase (sGC), and/or impairment of its downstream pathway, cyclic guanosine monophosphate (cGMP)-protein kinase G (PKG)] are the known mechanisms underlying the vascular NO• resistance in T2D. Hyperglycemia-induced overproduction of reactive oxygen species (ROS) and vascular insulin resistance are key players in this state. Therefore, upregulating vascular NO• availability, re-sensitizing or bypassing the non-responsive pathways to NO•, and targeting key vascular sources of ROS production may be clinically relevant pharmacological approaches to circumvent T2D-induced vascular NO• resistance.
Collapse
Affiliation(s)
- Zahra Bahadoran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parvin Mirmiran
- Department of Clinical Nutrition, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Khosrow Kashfi
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, 10031, USA
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
33
|
Chuaiphichai S, Dickinson Y, Whiteman CAR, Au-Yeung D, McNeill E, Channon KM, Douglas G. Endothelial cell vasodilator dysfunction mediates progressive pregnancy-induced hypertension in endothelial cell tetrahydrobiopterin deficient mice. Vascul Pharmacol 2023; 150:107168. [PMID: 36966985 DOI: 10.1016/j.vph.2023.107168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 03/05/2023] [Accepted: 03/19/2023] [Indexed: 04/10/2023]
Abstract
BACKGROUND AND PURPOSE Pregnancy-associated vascular remodelling is essential for both maternal and fetal health. We have previously shown that maternal endothelial cell tetrahydrobiopterin (BH4) deficiency causes poor pregnancy outcomes. Here, we investigated the role and mechanisms of endothelial cell-mediated vasorelaxation function in these outcomes. EXPERIMENTAL APPROACH The vascular reactivity of mouse aortas and uterine arteries from non-pregnant and pregnant endothelial cell-specific BH4 deficient mice (Gch1fl/flTie2cre mice) was assessed by wire myography. Systolic blood pressure was assessed by tail cuff plethysmography. KEY RESULTS In late pregnancy, systolic blood pressure was significantly higher (∼24 mmHg) in Gch1fl/flTie2cre mice compared with wild-type littermates. This was accompanied by enhanced vasoconstriction and reduced endothelial-dependent vasodilation in both aorta and uterine arteries from pregnant Gch1fl/flTie2cre mice. In uterine arteries loss of eNOS-derived vasodilators was partially compensated by upregulation of intermediate and large-conductance Ca2+-activated K+ channels. In rescue experiments, oral BH4 supplementation alone did not rescue vascular dysfunction and pregnancy-induced hypertension in Gch1fl/flTie2cre mice. However, combination with the fully reduced folate, 5-methyltetrahydrofolate (5-MTHF), restored endothelial cell vasodilator function and blood pressure. CONCLUSIONS AND IMPLICATIONS We identify a critical requirement for maternal endothelial cell Gch1/BH4 biosynthesis in endothelial cell vasodilator function in pregnancy. Targeting vascular Gch1 and BH4 biosynthesis with reduced folates may provide a novel therapeutic target for the prevention and treatment of pregnancy-related hypertension.
Collapse
Affiliation(s)
- Surawee Chuaiphichai
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK.
| | - Yasmin Dickinson
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Christopher A R Whiteman
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Desson Au-Yeung
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Eileen McNeill
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Keith M Channon
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Gillian Douglas
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| |
Collapse
|
34
|
Lisi V, Senesi G, Bertola N, Pecoraro M, Bolis S, Gualerzi A, Picciolini S, Raimondi A, Fantini C, Moretti E, Parisi A, Sgrò P, Di Luigi L, Geiger R, Ravera S, Vassalli G, Caporossi D, Balbi C. Plasma-derived extracellular vesicles released after endurance exercise exert cardioprotective activity through the activation of antioxidant pathways. Redox Biol 2023; 63:102737. [PMID: 37236143 DOI: 10.1016/j.redox.2023.102737] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/08/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Cardiovascular diseases (CVD) can cause various conditions, including an increase in reactive oxygen species (ROS) levels that can decrease nitric oxide (NO) availability and promote vasoconstriction, leading to arterial hypertension. Physical exercise (PE) has been found to be protective against CVD by helping to maintain redox homeostasis through a decrease in ROS levels, achieved by increased expression of antioxidant enzymes (AOEs) and modulation of heat shock proteins (HSPs). Extracellular vesicles (EVs) circulating in the body are a major source of regulatory signals, including proteins and nucleic acids. Interestingly, the cardioprotective role of EVs released after PE has not been fully described. The aim of this study was to investigate the role of circulating EVs, obtained through Size Exclusion Chromatography (SEC) of plasma samples from healthy young males (age: 26.95 ± 3.07; estimated maximum oxygen consumption rate (VO2max): 51.22 ± 4.85 (mL/kg/min)) at basal level (Pre_EVs) and immediately after a single bout of endurance exercise (30' treadmill, 70% heart rate (HR) -Post_EVs). Gene ontology (GO) analysis of proteomic data from isolated EVs, revealed enrichment in proteins endowed with catalytic activity in Post_EVs, compare to Pre_EVs, with MAP2K1 being the most significantly upregulated protein. Enzymatic assays on EVs derived from Pre and Post samples showed increment in Glutathione Reductase (GR) and Catalase (CAT) activity in Post_EVs. At functional level, Post_EVs, but not Pre_EVs, enhanced the activity of antioxidant enzymes (AOEs) and reduced oxidative damage accumulation in treated human iPS-derived cardiomyocytes (hCM) at basal level and under stress conditions (Hydrogen Peroxide (H2O2) treatment), resulting in a global cardioprotective effect. In conclusion, our data demonstrated, for the first time, that a single 30-min endurance exercise is able to alter the cargo of circulating EVs, resulting in cardioprotective effect through antioxidant activity.
Collapse
Affiliation(s)
- Veronica Lisi
- Unit of Biology and Genetics of Movement, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis 15, 00135, Rome, Italy
| | - Giorgia Senesi
- Cellular and Molecular Cardiology, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Nadia Bertola
- Department of Experimental Medicine, University of Genoa, 16132, Genova, Italy
| | - Matteo Pecoraro
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Sara Bolis
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Alice Gualerzi
- Laboratory of Nanomedicine and Clinical Biophotonics (LABION), IRCCS Fondazione Don Carlo Gnocchi, Milan, Italy
| | - Silvia Picciolini
- Laboratory of Nanomedicine and Clinical Biophotonics (LABION), IRCCS Fondazione Don Carlo Gnocchi, Milan, Italy
| | - Andrea Raimondi
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland; Centro Imaging Sperimentale, IRCCS Istituto Scientifico San Raffaele, Via Olgettina 52, 20132, Milan, Italy
| | - Cristina Fantini
- Unit of Biology and Genetics of Movement, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis 15, 00135, Rome, Italy
| | - Elisa Moretti
- Laboratory of Physical Exercise and Sport Science, Department of Exercise, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis 15, 00135, Rome, Italy
| | - Attilio Parisi
- Laboratory of Physical Exercise and Sport Science, Department of Exercise, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis 15, 00135, Rome, Italy
| | - Paolo Sgrò
- Laboratory of Physical Exercise and Sport Science, Department of Exercise, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis 15, 00135, Rome, Italy
| | - Luigi Di Luigi
- Endocrinology Unit, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis 15, 00135, Rome, Italy
| | - Roger Geiger
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Silvia Ravera
- Department of Experimental Medicine, University of Genoa, 16132, Genova, Italy
| | - Giuseppe Vassalli
- Cellular and Molecular Cardiology, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland; Center for Molecular Cardiology, Zurich, Switzerland
| | - Daniela Caporossi
- Unit of Biology and Genetics of Movement, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis 15, 00135, Rome, Italy
| | - Carolina Balbi
- Cellular and Molecular Cardiology, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland; Center for Molecular Cardiology, Zurich, Switzerland.
| |
Collapse
|
35
|
Lorentzen KA, Hernanz R, Pinilla E, Nyengaard JR, Wogensen L, Simonsen U. Sex-Dependent Impairment of Endothelium-Dependent Relaxation in Aorta of Mice with Overexpression of Hyaluronan in Tunica Media. Int J Mol Sci 2023; 24:8436. [PMID: 37176139 PMCID: PMC10179165 DOI: 10.3390/ijms24098436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 05/01/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023] Open
Abstract
Diabetic macroangiopathy is characterized by increased extracellular matrix deposition, including excessive hyaluronan accumulation, vessel thickening and stiffness, and endothelial dysfunction in large arteries. We hypothesized that the overexpression of hyaluronan in the tunica media also led to endothelial cell (EC) dysfunction. To address this hypothesis, we investigated the following in the aortas of mice with excessive hyaluronan accumulation in the tunica media (HAS-2) and wild-type mice: EC dysfunction via myograph studies, nitric oxide (NO) bioavailability via diaminofluorescence, superoxide formation via dihydroethidium fluorescence, and the distances between ECs via stereological methods. EC dysfunction, characterized by blunted relaxations in response to acetylcholine and decreased NO bioavailability, was found in the aortas of male HAS-2 mice, while it was unaltered in the aortas of female HAS-2 mice. Superoxide levels increased and extracellular superoxide dismutase (ecSOD) expression decreased in the aortas of male and female HAS-2 mice. The EC-EC distances and LDL receptor expression were markedly increased in the HAS-2 aortas of male mice. Our findings suggest hyaluronan increases oxidative stress in the vascular wall and that together with increased EC distance, it is associated with a sex-specific decrease in NO levels and endothelial dysfunction in the aorta of male HAS-2 transgenic mice.
Collapse
Affiliation(s)
- Karen Axelgaard Lorentzen
- Research Laboratory for Biochemical Pathology, Department of Clinical Medicine, Aarhus University Hospital, Aarhus University, 8000 Aarhus, Denmark
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, 8000 Aarhus, Denmark
| | - Raquel Hernanz
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, 8000 Aarhus, Denmark
- Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos, 28933 Alcorcón, Spain
| | - Estéfano Pinilla
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, 8000 Aarhus, Denmark
| | - Jens Randel Nyengaard
- Core Center for Molecular Morphology, Section for Stereology and Microscopy, Center for Stochastic Geometry and Advanced Bioimaging, The Department of Clinical Medicine—Stereology, Aarhus University, 8000 Aarhus, Denmark
| | - Lise Wogensen
- Research Laboratory for Biochemical Pathology, Department of Clinical Medicine, Aarhus University Hospital, Aarhus University, 8000 Aarhus, Denmark
| | - Ulf Simonsen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, 8000 Aarhus, Denmark
| |
Collapse
|
36
|
Osaki A, Kagami K, Ishinoda Y, Sato A, Kimura T, Horii S, Ito K, Toya T, Ido Y, Namba T, Masaki N, Nagatomo Y, Adachi T. Reactive Oxygen Species in the Aorta and Perivascular Adipose Tissue Precedes Endothelial Dysfunction in the Aorta of Mice with a High-Fat High-Sucrose Diet and Additional Factors. Int J Mol Sci 2023; 24:ijms24076486. [PMID: 37047458 PMCID: PMC10095299 DOI: 10.3390/ijms24076486] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/19/2023] [Accepted: 03/24/2023] [Indexed: 03/31/2023] Open
Abstract
Metabolic syndrome (Mets) is the major contributor to the onset of metabolic complications, such as hypertension, type 2 diabetes mellitus (DM), dyslipidemia, and non-alcoholic fatty liver disease, resulting in cardiovascular diseases. C57BL/6 mice on a high-fat and high-sucrose diet (HFHSD) are a well-established model of Mets but have minor endothelial dysfunction in isolated aortas without perivascular adipose tissue (PVAT). The purpose of this study was to evaluate the effects of additional factors such as DM, dyslipidemia, and steatohepatitis on endothelial dysfunction in aortas without PVAT. Here, we employed eight-week-old male C57BL/6 mice fed with a normal diet (ND), HFHSD, steatohepatitis choline-deficient HFHSD (HFHSD-SH), and HFHSD containing 1% cholesterol and 0.1% deoxycholic acid (HFHSD-Chol) for 16 weeks. At week 20, some HFHSD-fed mice were treated with streptozocin to develop diabetes (HFHSD-DM). In PVAT-free aortas, the endothelial-dependent relaxation (EDR) did not differ between ND and HFHSD (p = 0.25), but in aortas with PVAT, the EDR of HFHSD-fed mice was impaired compared with ND-fed mice (p = 0.005). HFHSD-DM, HFHSD-SH, and HFHSD-Chol impaired the EDR in aortas without PVAT (p < 0.001, p = 0.019, and p = 0.009 vs. ND, respectively). Furthermore, tempol rescued the EDR in those models. In the Mets model, the EDR is compromised by PVAT, but with the addition of DM, dyslipidemia, and SH, the vessels themselves may result in impaired EDR.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Yuji Nagatomo
- Correspondence: (Y.N.); (T.A.); Tel.: +81-4-2995-1597 (T.A.); Fax: +81-4-2996-5200 (T.A.)
| | - Takeshi Adachi
- Correspondence: (Y.N.); (T.A.); Tel.: +81-4-2995-1597 (T.A.); Fax: +81-4-2996-5200 (T.A.)
| |
Collapse
|
37
|
Tai YL, Marupudi S, Figueroa GA, Russell RD. Hemodynamics and Arterial Stiffness in Response to Oral Glucose Loading in Individuals with Type II Diabetes and Controlled Hypertension. High Blood Press Cardiovasc Prev 2023; 30:175-181. [PMID: 36913100 DOI: 10.1007/s40292-023-00569-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 02/21/2023] [Indexed: 03/14/2023] Open
Abstract
INTRODUCTION Type 2 diabetes (T2D), the fastest growing pandemic, is typically accompanied by vascular complications. A central hallmark of both T2D and vascular disease is insulin resistance which causes impaired glucose transport and vasoconstriction concomitantly. Those with cardiometabolic disease display greater variation in central hemodynamics and arterial elasticity, both potent predictors of cardiovascular morbidity and mortality, which may be exacerbated by concomitant hyperglycemia and hyperinsulinemia during glucose testing. Thus, elucidating central and arterial responses to glucose testing in those with T2D may identify acute vascular pathophysiologies triggered by oral glucose loading. AIM This study compared hemodynamics and arterial stiffness to an oral glucose challenge (OGC: 50g glucose) between individuals with and without T2D. 21 healthy (48 ± 10 years) and 20 participants with clinically diagnosed T2D and controlled hypertension (52 ± 8 years) were tested. METHODS Hemodynamics and arterial compliance were assessed at baseline, and 10, 20, 30, 40, 50, and 60 min post-OGC. RESULTS Heart rate increased between 20 and 60 post-OGC in both groups (p < 0.05). Central systolic blood pressure (SBP) decreased in the T2D group between 10 and 50 min post-OGC while central diastolic blood pressure (DBP) decreased in both groups from 20 to 60 post-OGC. Central SBP decreased in T2D between 10 and 50 min post-OGC and central DBP decreased in both groups between 20 and 60 min post-OGC. Brachial SBP decreased between 10 and 50 min in healthy participants, whereas both groups displayed decreases in brachial DBP between 20 and 60 min post-OGC. Arterial stiffness was unaffected. CONCLUSIONS An OGC alters central and peripheral blood pressure in healthy and T2D participants similarly with no changes in arterial stiffness.
Collapse
Affiliation(s)
- Yu Lun Tai
- Department of Health and Human Performance, College of Health Professions, University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - Smaran Marupudi
- Department of Health and Human Performance, College of Health Professions, University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - Gabriel A Figueroa
- Department of Health and Human Performance, College of Health Professions, University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - Ryan D Russell
- Department of Health and Human Performance, College of Health Professions, University of Texas Rio Grande Valley, Brownsville, TX, USA.
| |
Collapse
|
38
|
Janaszak-Jasiecka A, Płoska A, Wierońska JM, Dobrucki LW, Kalinowski L. Endothelial dysfunction due to eNOS uncoupling: molecular mechanisms as potential therapeutic targets. Cell Mol Biol Lett 2023; 28:21. [PMID: 36890458 PMCID: PMC9996905 DOI: 10.1186/s11658-023-00423-2] [Citation(s) in RCA: 84] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 01/19/2023] [Indexed: 03/10/2023] Open
Abstract
Nitric oxide (NO) is one of the most important molecules released by endothelial cells, and its antiatherogenic properties support cardiovascular homeostasis. Diminished NO bioavailability is a common hallmark of endothelial dysfunction underlying the pathogenesis of the cardiovascular disease. Vascular NO is synthesized by endothelial nitric oxide synthase (eNOS) from the substrate L-arginine (L-Arg), with tetrahydrobiopterin (BH4) as an essential cofactor. Cardiovascular risk factors such as diabetes, dyslipidemia, hypertension, aging, or smoking increase vascular oxidative stress that strongly affects eNOS activity and leads to eNOS uncoupling. Uncoupled eNOS produces superoxide anion (O2-) instead of NO, thus becoming a source of harmful free radicals exacerbating the oxidative stress further. eNOS uncoupling is thought to be one of the major underlying causes of endothelial dysfunction observed in the pathogenesis of vascular diseases. Here, we discuss the main mechanisms of eNOS uncoupling, including oxidative depletion of the critical eNOS cofactor BH4, deficiency of eNOS substrate L-Arg, or accumulation of its analog asymmetrical dimethylarginine (ADMA), and eNOS S-glutathionylation. Moreover, potential therapeutic approaches that prevent eNOS uncoupling by improving cofactor availability, restoration of L-Arg/ADMA ratio, or modulation of eNOS S-glutathionylation are briefly outlined.
Collapse
Affiliation(s)
- Anna Janaszak-Jasiecka
- Department of Medical Laboratory Diagnostics - Fahrenheit Biobank BBMRI.Pl, Medical University of Gdansk, 7 Debinki Street, 80-211, Gdansk, Poland
| | - Agata Płoska
- Department of Medical Laboratory Diagnostics - Fahrenheit Biobank BBMRI.Pl, Medical University of Gdansk, 7 Debinki Street, 80-211, Gdansk, Poland
| | - Joanna M Wierońska
- Department of Neurobiology, Polish Academy of Sciences, Maj Institute of Pharmacology, 12 Smętna Street, 31-343, Kraków, Poland
| | - Lawrence W Dobrucki
- Department of Medical Laboratory Diagnostics - Fahrenheit Biobank BBMRI.Pl, Medical University of Gdansk, 7 Debinki Street, 80-211, Gdansk, Poland.,Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,Beckman Institute for Advanced Science and Technology, 405 N Mathews Ave, MC-251, Urbana, IL, 61801, USA.,Department of Biomedical and Translational Sciences, Carle-Illinois College of Medicine, Urbana, IL, USA
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics - Fahrenheit Biobank BBMRI.Pl, Medical University of Gdansk, 7 Debinki Street, 80-211, Gdansk, Poland. .,BioTechMed Centre, Department of Mechanics of Materials and Structures, Gdansk University of Technology, 11/12 Gabriela Narutowicza Street, 80-233, Gdansk, Poland.
| |
Collapse
|
39
|
Haji S, Shiratsuchi M, Takamatsu A, Tsuda M, Muta H, Masuda T, Nakashima Y, Ogawa Y. Endothelial function testing before conditioning therapy is useful for predicting transplant-related complications after allogeneic hematopoietic cell transplantation. Int J Hematol 2023; 117:438-445. [PMID: 36436130 DOI: 10.1007/s12185-022-03498-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/11/2022] [Accepted: 11/13/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Hematopoietic stem cell transplantation (HSCT) is a useful tool for the treatment of hematologic malignancies. However, transplantation-related complications are the main cause of non-relapse mortality. Previous reports suggest that endothelial damage is related to early complications after HSCT. Non-invasive reactive hyperemia peripheral arterial tonometry (RH-PAT) was performed to evaluate endothelial function as a predictive marker for these complications. METHODS The reactive hyperemia index (RHI) obtained from RH-PAT was evaluated before the conditioning regimen. The relationship between the RHI and the appearance of engraftment syndrome, thrombotic microangiopathy, and acute graft-versus-host disease (aGVHD) was assessed. Receiver operating characteristic curve analysis showed that an RHI value of 1.58 was the optimal cut-off for predicting transplantation-related complications. RESULTS In total, 49 patients (22 acute myelogenous leukemia, 7 acute lymphocytic leukemia, 6 myelodysplastic syndrome, 6 adult T-cell leukemia, 6 non-Hodgkin lymphoma, and 2 others) were enrolled; 34 had a normal RHI (≥ 1.59), and 15 had an abnormally low RHI (≤ 1.58). Thrombotic microangiopathy (20.2% vs 0.0%, P = 0.025) and aGVHD (80.0% vs 41.2%, P = 0.015) were significantly more frequent in patients with a low RHI. CONCLUSION Endothelial dysfunction assessed by RH-PAT before HSCT was able to predict transplantation-related complications.
Collapse
Affiliation(s)
- Shojiro Haji
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Motoaki Shiratsuchi
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan.
- Department of Hematology, Iizuka Hospital, Iizuka, 820-8505, Japan.
| | - Akiko Takamatsu
- Department of Hematology, Iizuka Hospital, Iizuka, 820-8505, Japan
| | - Mariko Tsuda
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Hiroki Muta
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Toru Masuda
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Yasuhiro Nakashima
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Yoshihiro Ogawa
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
40
|
Rajlic S, Treede H, Münzel T, Daiber A, Duerr GD. Early Detection Is the Best Prevention-Characterization of Oxidative Stress in Diabetes Mellitus and Its Consequences on the Cardiovascular System. Cells 2023; 12:583. [PMID: 36831253 PMCID: PMC9954643 DOI: 10.3390/cells12040583] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/05/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023] Open
Abstract
Previous studies demonstrated an important role of oxidative stress in the pathogenesis of cardiovascular disease (CVD) in diabetic patients due to hyperglycemia. CVD remains the leading cause of premature death in the western world. Therefore, diabetes mellitus-associated oxidative stress and subsequent inflammation should be recognized at the earliest possible stage to start with the appropriate treatment before the onset of the cardiovascular sequelae such as arterial hypertension or coronary artery disease (CAD). The pathophysiology comprises increased reactive oxygen and nitrogen species (RONS) production by enzymatic and non-enzymatic sources, e.g., mitochondria, an uncoupled nitric oxide synthase, xanthine oxidase, and the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX). Considering that RONS originate from different cellular mechanisms in separate cellular compartments, adequate, sensitive, and compartment-specific methods for their quantification are crucial for early detection. In this review, we provide an overview of these methods with important information for early, appropriate, and effective treatment of these patients and their cardiovascular sequelae.
Collapse
Affiliation(s)
- Sanela Rajlic
- Department of Cardiothoracic and Vascular Surgery, University of Medicine Mainz, 55131 Mainz, Germany
| | - Hendrik Treede
- Department of Cardiothoracic and Vascular Surgery, University of Medicine Mainz, 55131 Mainz, Germany
| | - Thomas Münzel
- Center for Cardiology, Department of Cardiology, Molecular Cardiology, University Medical Center, 55131 Mainz, Germany
| | - Andreas Daiber
- Center for Cardiology, Department of Cardiology, Molecular Cardiology, University Medical Center, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, 55131 Mainz, Germany
| | - Georg Daniel Duerr
- Department of Cardiothoracic and Vascular Surgery, University of Medicine Mainz, 55131 Mainz, Germany
| |
Collapse
|
41
|
Sufianov A, Kostin A, Begliarzade S, Kudriashov V, Ilyasova T, Liang Y, Mukhamedzyanov A, Beylerli O. Exosomal non coding RNAs as a novel target for diabetes mellitus and its complications. Noncoding RNA Res 2023; 8:192-204. [PMID: 36818396 PMCID: PMC9929646 DOI: 10.1016/j.ncrna.2023.02.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/06/2023] [Accepted: 02/06/2023] [Indexed: 02/10/2023] Open
Abstract
Diabetes mellitus (DM) is a first-line priority among the problems facing medical science and public health in almost all countries of the world. The main problem of DM is the high incidence of damage to the cardiovascular system, which in turn leads to diseases such as myocardial infarction, stroke, gangrene of the lower extremities, blindness and chronic renal failure. As a result, the study of the molecular genetic mechanisms of the pathogenesis of DM is of critical importance for the development of new diagnostic and therapeutic strategies. Molecular genetic aspects of the etiology and pathogenesis of diabetes mellitus are intensively studied in well-known laboratories around the world. One of the strategies in this direction is to study the role of exosomes in the pathogenesis of DM. Exosomes are microscopic extracellular vesicles with a diameter of 30-100 nm, released into the intercellular space by cells of various tissues and organs. The content of exosomes depends on the cell type and includes mRNA, non-coding RNAs, DNA, and so on. Non-coding RNAs, a group of RNAs with limited transcriptional activity, have been discovered to play a significant role in regulating gene expression through epigenetic and posttranscriptional modulation, such as silencing of messenger RNA. One of the problems of usage exosomes in DM is the identification of the cellular origin of exosomes and the standardization of protocols for molecular genetic studies in clinical laboratories. In addition, the question of the target orientation of exosomes and their targeted activity requires additional study. Solving these and other problems will make it possible to use exosomes for the diagnosis and delivery of drugs directly to target cells in DM. This study presents an analysis of literature data on the role of exosomes and ncRNAs in the development and progression of DM, as well as the prospects for the use of exosomes in clinical practice in this disease.
Collapse
Affiliation(s)
- Albert Sufianov
- Educational and Scientific Institute of Neurosurgery, Рeoples’ Friendship University of Russia (RUDN University), Moscow, Russia,Department of Neurosurgery, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Andrey Kostin
- Research and Educational Resource Center for Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis Innovative Technologies, Peoples' Friendship University of Russia, Moscow, Russia
| | - Sema Begliarzade
- Republican Clinical Perinatal Center, Ufa, Republic of Bashkortostan, 450106, Russia
| | | | - Tatiana Ilyasova
- Department of Internal Diseases, Bashkir State Medical University, Ufa, Republic of Bashkortostan, 450008, Russia
| | - Yanchao Liang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | | | - Ozal Beylerli
- Educational and Scientific Institute of Neurosurgery, Рeoples’ Friendship University of Russia (RUDN University), Moscow, Russia,Corresponding author. Рeoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, Moscow, 117198, Russian Federation.
| |
Collapse
|
42
|
Shi X, Tian Y, Zhai S, Liu Y, Chu S, Xiong Z. The progress of research on the application of redox nanomaterials in disease therapy. Front Chem 2023; 11:1115440. [PMID: 36814542 PMCID: PMC9939781 DOI: 10.3389/fchem.2023.1115440] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 01/23/2023] [Indexed: 02/08/2023] Open
Abstract
Redox imbalance can trigger cell dysfunction and damage and plays a vital role in the origin and progression of many diseases. Maintaining the balance between oxidants and antioxidants in vivo is a complicated and arduous task, leading to ongoing research into the construction of redox nanomaterials. Nanodrug platforms with redox characteristics can not only reduce the adverse effects of oxidative stress on tissues by removing excess oxidants from the body but also have multienzyme-like activity, which can play a cytotoxic role in tumor tissues through the catalytic oxidation of their substrates to produce harmful reactive oxygen species such as hydroxyl radicals. In this review, various redox nanomaterials currently used in disease therapy are discussed, emphasizing the treatment methods and their applications in tumors and other human tissues. Finally, the limitations of the current clinical application of redox nanomaterials are considered.
Collapse
Affiliation(s)
- Xiaolu Shi
- Department of Implantology, Hospital of Stomatology, Jilin University, Changchun, China
| | - Ye Tian
- Department of Implantology, Hospital of Stomatology, Jilin University, Changchun, China
| | - Shaobo Zhai
- Department of Implantology, Hospital of Stomatology, Jilin University, Changchun, China
| | - Yang Liu
- Department of Implantology, Hospital of Stomatology, Jilin University, Changchun, China
| | - Shunli Chu
- Department of Implantology, Hospital of Stomatology, Jilin University, Changchun, China,*Correspondence: Shunli Chu, ; Zhengrong Xiong,
| | - Zhengrong Xiong
- Polymer Composites Engineering Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences (CAS), Changchun, China,Department of Applied Chemistry, University of Science and Technology of China, Hefei, China,*Correspondence: Shunli Chu, ; Zhengrong Xiong,
| |
Collapse
|
43
|
Mázala-de-Oliveira T, Jannini de Sá YAP, Carvalho VDF. Impact of gut-peripheral nervous system axis on the development of diabetic neuropathy. Mem Inst Oswaldo Cruz 2023; 118:e220197. [PMID: 36946851 PMCID: PMC10027071 DOI: 10.1590/0074-02760220197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 02/14/2023] [Indexed: 03/22/2023] Open
Abstract
Diabetes is a chronic metabolic disease caused by a reduction in the production and/or action of insulin, with consequent development of hyperglycemia. Diabetic patients, especially those who develop neuropathy, presented dysbiosis, with an increase in the proportion of pathogenic bacteria and a decrease in the butyrate-producing bacteria. Due to this dysbiosis, diabetic patients presented a weakness of the intestinal permeability barrier and high bacterial product translocation to the bloodstream, in parallel to a high circulating levels of pro-inflammatory cytokines such as TNF-α. In this context, we propose here that dysbiosis-induced increased systemic levels of bacterial products, like lipopolysaccharide (LPS), leads to an increase in the production of pro-inflammatory cytokines, including TNF-α, by Schwann cells and spinal cord of diabetics, being crucial for the development of neuropathy.
Collapse
Affiliation(s)
| | | | - Vinicius de Frias Carvalho
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Rio de Janeiro, RJ, Brasil
- Instituto Nacional de Ciência e Tecnologia em Neuroimunomodulação, Rio de Janeiro, RJ, Brasil
- + Corresponding author:
| |
Collapse
|
44
|
Heart Failure and Diabetes Mellitus: Dangerous Liaisons. INTERNATIONAL JOURNAL OF HEART FAILURE 2022; 4:163-174. [PMID: 36381018 PMCID: PMC9634025 DOI: 10.36628/ijhf.2022.0022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 10/20/2022] [Accepted: 10/21/2022] [Indexed: 01/25/2023]
Abstract
Patients with diabetes mellitus (DM) have a higher prevalence of heart failure (HF) than those without it. Approximately 40% of HF patients have DM, having poorer outcomes than those without DM. Myocardial ischemia caused by endothelial dysfunction, renal dysfunction, obesity, and impaired myocardial energetics is pathophysiology of DM-induced HF (DM-HF). Also, patients with HF show an increased risk for the onset of DM due to several mechanisms including insulin resistance. This review is focused on the epidemiology, pathogenic mechanism and treatment strategy of DM-HF.
Collapse
|
45
|
Ormazabal V, Nair S, Carrión F, Mcintyre HD, Salomon C. The link between gestational diabetes and cardiovascular diseases: potential role of extracellular vesicles. Cardiovasc Diabetol 2022; 21:174. [PMID: 36057662 PMCID: PMC9441052 DOI: 10.1186/s12933-022-01597-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 08/05/2022] [Indexed: 11/25/2022] Open
Abstract
Extracellular vesicles are critical mediators of cell communication. They encapsulate a variety of molecular cargo such as proteins, lipids, and nucleic acids including miRNAs, lncRNAs, circular RNAs, and mRNAs, and through transfer of these molecular signals can alter the metabolic phenotype in recipient cells. Emerging studies show the important role of extracellular vesicle signaling in the development and progression of cardiovascular diseases and associated risk factors such as type 2 diabetes and obesity. Gestational diabetes mellitus (GDM) is hyperglycemia that develops during pregnancy and increases the future risk of developing obesity, impaired glucose metabolism, and cardiovascular disease in both the mother and infant. Available evidence shows that changes in maternal metabolism and exposure to the hyperglycemic intrauterine environment can reprogram the fetal genome, leaving metabolic imprints that define life-long health and disease susceptibility. Understanding the factors that contribute to the increased susceptibility to metabolic disorders of children born to GDM mothers is critical for implementation of preventive strategies in GDM. In this review, we discuss the current literature on the fetal programming of cardiovascular diseases in GDM and the impact of extracellular vesicle (EV) signaling in epigenetic programming in cardiovascular disease, to determine the potential link between EV signaling in GDM and the development of cardiovascular disease in infants.
Collapse
Affiliation(s)
- Valeska Ormazabal
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, UQ Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of Medicine + Biomedical Sciences, The University of Queensland, Building 71/918, Herston, QLD, 4029, Australia.,Faculty of Biological Sciences, Pharmacology Department, University of Concepcion, Concepción, Chile
| | - Soumyalekshmi Nair
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, UQ Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of Medicine + Biomedical Sciences, The University of Queensland, Building 71/918, Herston, QLD, 4029, Australia
| | - Flavio Carrión
- Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago, Chile
| | - H David Mcintyre
- Mater Research, Faculty of Medicine, University of Queensland, Mater Health, South Brisbane, Australia
| | - Carlos Salomon
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, UQ Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of Medicine + Biomedical Sciences, The University of Queensland, Building 71/918, Herston, QLD, 4029, Australia. .,Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago, Chile.
| |
Collapse
|
46
|
Fluss G, Cranwell V, Rao A, Lee JS, Elshafey R, Wallack M, Finlay D. Covid-19 Delta variant resulting in multi system thromboembolic disease. ANNALS OF VASCULAR SURGERY - BRIEF REPORTS AND INNOVATIONS 2022; 2:100101. [PMID: 35784811 PMCID: PMC9222409 DOI: 10.1016/j.avsurg.2022.100101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 06/13/2022] [Accepted: 06/15/2022] [Indexed: 12/02/2022]
Abstract
Infection with severe acute respiratory syndrome Coronavirus 2 (SARS-CoV-2/Covid-19) has been correlated with micro and macrovascular thrombosis. Reports have discussed localized thrombosis leading to ischemia, we report a case of diffuse systemic thromboembolism resulting in limb ischemia and organ damage from Covid-19 despite prompt anticoagulation. A 60-year-old unvaccinated male with a history of asthma, diabetes mellitus and coronary artery disease, presented with shortness of breath and right lower extremity pain. The patient was found to be Covid-19 Delta variant positive. CTA and transthoracic echocardiogram showed diffuse thromboembolic disease affecting the left ventricle, right kidney and bilateral lower extremities.
Collapse
|
47
|
Rotariu D, Babes EE, Tit DM, Moisi M, Bustea C, Stoicescu M, Radu AF, Vesa CM, Behl T, Bungau AF, Bungau SG. Oxidative stress - Complex pathological issues concerning the hallmark of cardiovascular and metabolic disorders. Biomed Pharmacother 2022; 152:113238. [PMID: 35687909 DOI: 10.1016/j.biopha.2022.113238] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/30/2022] [Accepted: 06/02/2022] [Indexed: 12/07/2022] Open
Abstract
Oxidative stress is a complex biological process characterized by the excessive production of reactive oxygen species (ROS) that act as destroyers of the REDOX balance in the body and, implicitly, inducing oxidative damage. All the metabolisms are impaired in oxidative stress and even nucleic acid balance is influenced. ROS will promote structural changes of the tissues and organs due to interaction with proteins and phospholipids. The constellation of the cardiovascular risk factors (CVRFs) will usually develop in subjects with predisposition to cardiac disorders. Oxidative stress is usually related with hypertension (HTN), diabetes mellitus (DM), obesity and cardiovascular diseases (CVDs) like coronary artery disease (CAD), cardiomyopathy or heart failure (HF), that can develop in subjects with the above-mentioned diseases. Elements describing the complex relationship between CVD and oxidative stress should be properly explored and described because prevention may be the optimal approach. Our paper aims to expose in detail the complex physiopathology of oxidative stress in CVD occurrence and novelties regarding the phenomenon. Biomarkers assessing oxidative stress or therapy targeting specific pathways represent a major progress that actually change the outcome of subjects with CVD. New antioxidants therapy specific for each CVD represents a captivating and interesting future perspective with tremendous benefits on subject's outcome.
Collapse
Affiliation(s)
- Dragos Rotariu
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania.
| | - Emilia Elena Babes
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy of Oradea, University of Oradea, 410073 Oradea, Romania.
| | - Delia Mirela Tit
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania; Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania.
| | - Madalina Moisi
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy of Oradea, University of Oradea, 410073 Oradea, Romania.
| | - Cristiana Bustea
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy of Oradea, University of Oradea, 410073 Oradea, Romania.
| | - Manuela Stoicescu
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy of Oradea, University of Oradea, 410073 Oradea, Romania.
| | - Andrei-Flavius Radu
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania; Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy of Oradea, University of Oradea, 410073 Oradea, Romania.
| | - Cosmin Mihai Vesa
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy of Oradea, University of Oradea, 410073 Oradea, Romania.
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India.
| | | | - Simona Gabriela Bungau
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania; Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania.
| |
Collapse
|
48
|
Čater M, Bombek LK. Protective Role of Mitochondrial Uncoupling Proteins against Age-Related Oxidative Stress in Type 2 Diabetes Mellitus. Antioxidants (Basel) 2022; 11:antiox11081473. [PMID: 36009191 PMCID: PMC9404801 DOI: 10.3390/antiox11081473] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 07/25/2022] [Accepted: 07/26/2022] [Indexed: 02/04/2023] Open
Abstract
The accumulation of oxidative damage to DNA and other biomolecules plays an important role in the etiology of aging and age-related diseases such as type 2 diabetes mellitus (T2D), atherosclerosis, and neurodegenerative disorders. Mitochondrial DNA (mtDNA) is especially sensitive to oxidative stress. Mitochondrial dysfunction resulting from the accumulation of mtDNA damage impairs normal cellular function and leads to a bioenergetic crisis that accelerates aging and associated diseases. Age-related mitochondrial dysfunction decreases ATP production, which directly affects insulin secretion by pancreatic beta cells and triggers the gradual development of the chronic metabolic dysfunction that characterizes T2D. At the same time, decreased glucose oxidation in skeletal muscle due to mitochondrial damage leads to prolonged postprandial blood glucose rise, which further worsens glucose homeostasis. ROS are not only highly reactive by-products of mitochondrial respiration capable of oxidizing DNA, proteins, and lipids but can also function as signaling and effector molecules in cell membranes mediating signal transduction and inflammation. Mitochondrial uncoupling proteins (UCPs) located in the inner mitochondrial membrane of various tissues can be activated by ROS to protect cells from mitochondrial damage. Mitochondrial UCPs facilitate the reflux of protons from the mitochondrial intermembrane space into the matrix, thereby dissipating the proton gradient required for oxidative phosphorylation. There are five known isoforms (UCP1-UCP5) of mitochondrial UCPs. UCP1 can indirectly reduce ROS formation by increasing glutathione levels, thermogenesis, and energy expenditure. In contrast, UCP2 and UCP3 regulate fatty acid metabolism and insulin secretion by beta cells and modulate insulin sensitivity. Understanding the functions of UCPs may play a critical role in developing pharmacological strategies to combat T2D. This review summarizes the current knowledge on the protective role of various UCP homologs against age-related oxidative stress in T2D.
Collapse
Affiliation(s)
- Maša Čater
- Correspondence: (M.Č.); (L.K.B.); Tel.: +386-2-2345-847 (L.K.B.)
| | | |
Collapse
|
49
|
Tissue Engineering Approaches to Uncover Therapeutic Targets for Endothelial Dysfunction in Pathological Microenvironments. Int J Mol Sci 2022; 23:ijms23137416. [PMID: 35806421 PMCID: PMC9266895 DOI: 10.3390/ijms23137416] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 06/28/2022] [Accepted: 07/01/2022] [Indexed: 02/07/2023] Open
Abstract
Endothelial cell dysfunction plays a central role in many pathologies, rendering it crucial to understand the underlying mechanism for potential therapeutics. Tissue engineering offers opportunities for in vitro studies of endothelial dysfunction in pathological mimicry environments. Here, we begin by analyzing hydrogel biomaterials as a platform for understanding the roles of the extracellular matrix and hypoxia in vascular formation. We next examine how three-dimensional bioprinting has been applied to recapitulate healthy and diseased tissue constructs in a highly controllable and patient-specific manner. Similarly, studies have utilized organs-on-a-chip technology to understand endothelial dysfunction's contribution to pathologies in tissue-specific cellular components under well-controlled physicochemical cues. Finally, we consider studies using the in vitro construction of multicellular blood vessels, termed tissue-engineered blood vessels, and the spontaneous assembly of microvascular networks in organoids to delineate pathological endothelial dysfunction.
Collapse
|
50
|
Wong AYW, Ong BSY, Lee ARYB, Mai AS, Selvarajan S, Lakshminarasappa SR, Tay SM. Topical Biological Agents as Adjuncts to Improve Wound Healing in Chronic Diabetic Wounds: A Systematic Review of Clinical Evidence and Future Directions. Cureus 2022; 14:e27180. [PMID: 36035037 PMCID: PMC9398533 DOI: 10.7759/cureus.27180] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2022] [Indexed: 11/05/2022] Open
Abstract
Diabetes is a leading chronic illness in the modern world and 19-34% develop chronic diabetic foot ulcers (DFUs) in their lifetime, often necessitating amputation. The reduction in tissue growth factors and resulting imbalance between proteolytic enzymes and their inhibitors, along with systemic factors impairing healing appear particularly important in chronic wounds. Growth factors applied topically have thus been suggested to be a non-invasive, safe, and cost-effective adjunct to improve wound healing and prevent complications. Comprehensive database searches of MEDLINE via PubMed, EMBASE, Cochrane, and ClinicalTrials.gov were performed to identify clinical evidence and ongoing trials. The risk of bias analysis included randomized controlled trials (RCTs) was performed using the Cochrane Risk of Bias 2.0 tool. We included randomized controlled trials that compared the use of a topical biologic growth factor-containing regimen to any other regimen. Primary outcomes of interest were time to wound closure, healing rate, and time. Secondary outcomes included the incidence of adverse events such as infection. A total of 41 trials from 1992-2020 were included in this review, with a total recorded 3,112 patients. Platelet-derived growth factors (PDGF) in the form of becaplermin gel are likely to reduce the time of closure, increase the incidence of wound closure, and complete wound healing. Human umbilical cord-related treatments, dehydrated human amnion and chorion allograft (dHACA), and hypothermically stored amniotic membrane (HSAM), consistently increased the rates and incidence of complete ulcer healing while reducing ulcer size and time to complete ulcer healing. Fibroblast growth factor-1 (FGF1) showed only a slight benefit in multiple studies regarding increasing complete ulcer healing rates and incidence while reducing ulcer size and time to complete ulcer healing, with a few studies showing no statistical difference from placebo. Platelet-rich fibrin (PRF) is consistent in reducing the time to complete ulcer healing and increasing wound healing rate but may not reduce ulcer size or increase the incidence of complete ulcer healing. Targeting the wound healing pathway via the extrinsic administration of growth factors is a promising option to augment wound healing in diabetic patients. Growth factors have also shown promise in specific subgroups of patients who are at risk of significantly impaired wound healing such as those with a history of secondary infection and vasculopathy. As diabetes impairs multiple stages of wound healing, combining growth factors in diabetic wound care may prove to be an area of interest. Evidence from this systematic literature review suggests that topical adjuncts probably reduce time to wound closure, reduce healing time, and increase the healing rate in patients with chronic DFUs.
Collapse
Affiliation(s)
- Andrew Yew Wei Wong
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, SGP
| | | | | | - Aaron Shengting Mai
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, SGP
| | | | | | - Sook Muay Tay
- Surgical Intensive Care, Singapore General Hospital, Singapore, SGP
| |
Collapse
|