1
|
Wang L, Jia J, Yu X, Luo M, Li J, Khan GJ, He C, Duan H, Zhai K. Molecular mechanism of rapamycin-induced autophagy activation to attenuate smoking-induced COPD. Biochem Biophys Res Commun 2025; 764:151819. [PMID: 40253907 DOI: 10.1016/j.bbrc.2025.151819] [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: 01/22/2025] [Revised: 03/25/2025] [Accepted: 04/14/2025] [Indexed: 04/22/2025]
Abstract
Chronic obstructive pulmonary disease (COPD) is one of the severe lung and respiratory airway disorders, with high prevalence rate in China. In this paper, we employed network pharmacology predictions to identify autophagy as a signaling pathway associated with COPD. To explore the protective effect of autophagy against COPD and its specific mechanism, we established a mouse model of COPD and administered 3-methyladenine (3-MA) and rapamycin (RAPA) to intervene in autophagy. The lung function of the mice was assessed using an animal pulmonary function analysis system, and lung tissue structure was evaluated through hematoxylin and eosin (HE) staining. The TUNEL staining method was employed to determine the level of apoptosis in lung tissue. Western blot analysis was conducted to measure the expression of autophagy and apoptosis-related proteins, while RT-qPCR was used to assess the expression of apoptosis-related mRNA. The results showed that RAPA effectively improved lung function, attenuated pathological lung injury and increased autophagy level in COPD mice. Apoptosis analysis showed that the apoptosis rate was elevated in COPD and 3- MA mice, whereas it was significantly reduced in RAPA mice. Our findings suggest that stimulation of autophagy may be a potential therapy for the future treatment of COPD.
Collapse
Affiliation(s)
- Li Wang
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830011, China
| | - Jianhu Jia
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830011, China
| | - Xinyan Yu
- School of Biological and Food Engineering, Engineering Research Center for Development and High Value Utilization of Genuine Medicinal Materials in North Anhui Province, Suzhou University, Suzhou, Anhui, 234000, China
| | - Mengmeng Luo
- School of Biological and Food Engineering, Engineering Research Center for Development and High Value Utilization of Genuine Medicinal Materials in North Anhui Province, Suzhou University, Suzhou, Anhui, 234000, China; College of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui, 241000, China
| | - Jie Li
- School of Biological and Food Engineering, Engineering Research Center for Development and High Value Utilization of Genuine Medicinal Materials in North Anhui Province, Suzhou University, Suzhou, Anhui, 234000, China; School of Pharmacy, Wannan Medical College, Wuhu, Anhui, 241002, China
| | - Ghulam Jilany Khan
- Department of Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, University of Central Punjab, Lahore, 54000, Pakistan
| | - Chenghui He
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830011, China; Xinjiang Institute of Materia Medica, Key Laboratory of Xinjiang Uygur Medicine, Urumqi, 830004, China.
| | - Hong Duan
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830011, China; School of Biological and Food Engineering, Engineering Research Center for Development and High Value Utilization of Genuine Medicinal Materials in North Anhui Province, Suzhou University, Suzhou, Anhui, 234000, China.
| | - Kefeng Zhai
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830011, China; School of Biological and Food Engineering, Engineering Research Center for Development and High Value Utilization of Genuine Medicinal Materials in North Anhui Province, Suzhou University, Suzhou, Anhui, 234000, China.
| |
Collapse
|
2
|
Yan H, Lv S, Pi H, Yu H, Yin W, Wang Y, Lan Y, Liu W. Inflammation mediates the relationship between cardiometabolic index and vulnerable plaque in patients with acute coronary syndrome. Lipids Health Dis 2025; 24:194. [PMID: 40437601 PMCID: PMC12121154 DOI: 10.1186/s12944-025-02608-4] [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: 03/25/2025] [Accepted: 05/14/2025] [Indexed: 06/01/2025] Open
Abstract
BACKGROUND As a novel indicator reflecting metabolic status and visceral adiposity distribution, the cardiometabolic index (CMI) has gained attention in cardiovascular risk stratification. This investigation employed optical coherence tomography (OCT) to examine potential associations between CMI and vulnerable plaque, as well as the role of inflammation. METHODS This study conducted a cross-sectional analysis of 270 acute coronary syndrome (ACS) patients who had OCT imaging evaluation. Patients were categorized based on CMI tertiles, with CMI calculated using the formula [waist (cm)/height (cm)]×[triglycerides (mmol/L)/HDL-C (mmol/L)]. OCT was used to assess plaque events in culprit lesions and plaque components in non-culprit lesions, and inflammatory markers were measured. A mediation analysis framework was implemented to investigate inflammatory pathways in CMI-vulnerable plaque relationships. RESULTS CMI tertiles were linked to vulnerable plaque traits: thin-cap fibroatheromas (TCFA), macrophages (Tertiles1 vs. Tertiles2 vs. Tertiles3, TCFA: 10.0% vs. 20.0% vs. 26.7%, P = 0.016; macrophages: 17.8% vs. 28.9% vs. 36.7%, P = 0.019). Multivariate regression demonstrated CMI elevation independently predicted a higher prevalence of TCFA (OR:1.40, 95%CI: 1.25-2.89, P = 0.003), more macrophage infiltration (OR:1.61, 95% CI:1.09-2.37, P = 0.017), reduced FCT (β:-30.65, 95% CI:-50.72-10.57, P = 0.003), and enlarged maximum lipid arc (β:20.78, 95% CI:6.55-35.01, P = 0.004). Moreover, CMI was positively related to hsCRP, WBC, and neutrophils. Mediation analysis revealed that hsCRP mediated about 17.0% of the association between CMI and minimum FCT [Indirect effect=-5.21, 95% CI=(-12.70, -1.27), P = 0.016]. CONCLUSIONS CMI is a key forecaster of vulnerable plaque in patients with ACS. Systemic inflammation is associated with the relationship between CMI and vulnerable plaque features, suggesting a potential mechanistic link.
Collapse
Affiliation(s)
- Haihao Yan
- Department of Cardiology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Sai Lv
- Department of Cardiology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Haiyao Pi
- Department of Cardiology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Haixu Yu
- Department of Cardiology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Weijun Yin
- Department of Cardiology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Yaran Wang
- Department of Cardiology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Yonghao Lan
- Department of Cardiology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Wei Liu
- Department of Cardiology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China.
| |
Collapse
|
3
|
Alzarea EA, Al-Kuraishy HM, Al-Gareeb AI, Alexiou A, Papadakis M, Beshay ON, Batiha GES. The Conceivable Role of Metabolic Syndrome in the Pathogenesis of Alzheimer's Disease: Cellular and Subcellular Alterations in Underpinning a Tale of Two. Neuromolecular Med 2025; 27:35. [PMID: 40379890 DOI: 10.1007/s12017-025-08832-6] [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: 10/31/2024] [Accepted: 01/09/2025] [Indexed: 05/19/2025]
Abstract
Alzheimer's disease (AD)is an age-related neurodegenerative disease characterized by memory decline and cognitive impairment .AD is common in people aged > 65 years, though most of AD cases are sporadic, which accounts for 95%, and 1-5% of AD is caused by familial causes . The causes of AD are aging, environmental toxins, and cardiometabolic factors that induce the degeneration of cholinergic neurons. It has been shown that the metabolic syndrome which is a clustering of dissimilar constituents including insulin resistance (IR), glucose intolerance, visceral obesity, hypertension, and dyslipidemia is implicated in the pathogenesis of AD. Metabolic syndrome disapprovingly affects cognitive function and the development in AD by inducing the development of oxidative stress, neuroinflammation, and brain IR. These changes, together with brain IR, impair cerebrovascular reactivity causing cognitive impairment and dementia. Nevertheless, the fundamental mechanism by which metabolic syndrome persuades AD risk is not entirely explicated. Accordingly, this review aims to discuss the connotation between metabolic syndrome and AD. In conclusion, metabolic syndrome is regarded as a possible risk factor for the initiation of AD neuropathology by diverse signaling pathways such as brain IR, activation of inflammatory signaling pathways, neuroinflammation, defective proteostasis, and dysregulation of lipid mediators.
Collapse
Affiliation(s)
- Ekremah A Alzarea
- Hematopathology, Department of Pathology, College of Medicine, Jouf University, Sakaka, Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Jabir Ibn Hayyan Medical University, Al-Ameer Qu./Najaf-Iraq, PO.Box13, Kufa, Iraq
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University, Chandigarh-Ludhiana Highway, Mohali, Punjab, Australia
- Department of Research & Development, Funogen, Athens, Greece
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany.
| | - Olivia N Beshay
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, AlBeheira, Egypt
| |
Collapse
|
4
|
Zapata JK, Gómez-Ambrosi J, Frühbeck G. Childhood obesity: The threatening apprentice of the adiposity empire. Rev Endocr Metab Disord 2025:10.1007/s11154-025-09959-4. [PMID: 40195232 DOI: 10.1007/s11154-025-09959-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/18/2025] [Indexed: 04/09/2025]
Abstract
Childhood obesity is a global health problem, with its prevalence having tripled since 1975. The increase in its prevalence has been predominantly in developing countries, but also in those with high economic status. Nowadays, there are multiple obesity definitions, however, one of the most accurate is the one which defines obesity as the accumulation of excessive body adiposity and not as an body weight excess. Nevertheless, the body mass index (BMI) is the most frequently used tool for its classification, according to the cut-off points established by the Center for Disease Control and World Health Organization tables. In children and adolescents an adiposity excess is related to the appearance of cardiovascular disease in adulthood and with many comorbidities such as metabolic syndrome, insulin resistance, type 2 diabetes, hypertension and metabolic dysfunction-associated steatotic liver disease, among others. Currently, there is still controversy about which is the ideal indicator for measuring overweight and obesity. BMI is still used as a standardized measure but may miss cases in which body composition is pathological despite a BMI within the normal-weight category. An adequate knowledge of the impact on health of dysfunctional adiposity as well as its accurate diagnosis will allow health professionals to address this condition in a more precise and comprehensive manner, and substantially improve the associated cardiometabolic risk and prognosis.
Collapse
Affiliation(s)
- J Karina Zapata
- Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Javier Gómez-Ambrosi
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain.
- Centro de Investigación Biomédica en Red-Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain.
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain.
| | - Gema Frühbeck
- Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain.
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain.
- Centro de Investigación Biomédica en Red-Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain.
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain.
| |
Collapse
|
5
|
Li S, Zhou X, Duan Q, Niu S, Li P, Feng Y, Zhang Y, Xu X, Gong SP, Cao H. Autophagy and Its Association with Macrophages in Clonal Hematopoiesis Leading to Atherosclerosis. Int J Mol Sci 2025; 26:3252. [PMID: 40244103 PMCID: PMC11989900 DOI: 10.3390/ijms26073252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/04/2025] [Accepted: 03/17/2025] [Indexed: 04/18/2025] Open
Abstract
Atherosclerosis, a chronic inflammatory disease characterized by lipid accumulation and immune cell infiltration, is linked to plaque formation and cardiovascular events. While traditionally associated with lipid metabolism and endothelial dysfunction, recent research highlights the roles of autophagy and clonal hematopoiesis (CH) in its pathogenesis. Autophagy, a cellular process crucial for degrading damaged components, regulates macrophage homeostasis and inflammation, both of which are pivotal in atherosclerosis. In macrophages, autophagy influences lipid metabolism, cytokine regulation, and oxidative stress, helping to prevent plaque instability. Defective autophagy exacerbates inflammation, impairs cholesterol efflux, and accelerates disease progression. Additionally, autophagic processes in endothelial cells and smooth muscle cells further contribute to atherosclerotic pathology. Recent studies also emphasize the interplay between autophagy and CH, wherein somatic mutations in genes like TET2, JAK2, and DNMT3A drive immune cell expansion and enhance inflammatory responses in atherosclerotic plaques. These mutations modify macrophage function, intensifying the inflammatory environment and accelerating atherosclerosis. Chaperone-mediated autophagy (CMA), a selective form of autophagy, also plays a critical role in regulating macrophage inflammation by degrading pro-inflammatory cytokines and oxidized low-density lipoprotein (ox-LDL). Impaired CMA activity leads to the accumulation of these substrates, activating the NLRP3 inflammasome and worsening inflammation. Preclinical studies suggest that pharmacologically activating CMA may mitigate atherosclerosis progression. In animal models, reduced CMA activity accelerates plaque instability and increases inflammation. This review highlights the importance of autophagic regulation in macrophages, focusing on its role in inflammation, plaque formation, and the contributions of CH. Building upon current advances, we propose a hypothesis in which autophagy, programmed cell death, and clonal hematopoiesis form a critical intrinsic axis that modulates the fundamental functions of macrophages, playing a complex role in the development of atherosclerosis. Understanding these mechanisms offers potential therapeutic strategies targeting autophagy and inflammation to reduce the burden of atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Shuanhu Li
- Key Laboratory of Pharmacodynamics and Material Basis of Chinese Medicine of Shaanxi Administration of Traditional Chinese Medicine, Engineering Research Center of Brain Health Industry of Chinese Medicine, Pharmacology of Chinese Medicine, Shaanxi University of Chinese Medicine, University Government Committee of Shaanxi Province, Xianyang 712046, China;
| | - Xin Zhou
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi’an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi’an Medical University, Xi’an 710021, China; (S.N.); (P.L.); (Y.F.); (Y.Z.); (S.-P.G.)
| | - Qinchun Duan
- Laboratory of Cell Biology, Genetics and Developmental Biology, College of Life Sciences, Shaanxi Normal University, Xi’an 710062, China; (Q.D.); or (X.X.)
| | - Shukun Niu
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi’an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi’an Medical University, Xi’an 710021, China; (S.N.); (P.L.); (Y.F.); (Y.Z.); (S.-P.G.)
| | - Pengquan Li
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi’an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi’an Medical University, Xi’an 710021, China; (S.N.); (P.L.); (Y.F.); (Y.Z.); (S.-P.G.)
| | - Yihan Feng
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi’an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi’an Medical University, Xi’an 710021, China; (S.N.); (P.L.); (Y.F.); (Y.Z.); (S.-P.G.)
| | - Ye Zhang
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi’an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi’an Medical University, Xi’an 710021, China; (S.N.); (P.L.); (Y.F.); (Y.Z.); (S.-P.G.)
| | - Xuehong Xu
- Laboratory of Cell Biology, Genetics and Developmental Biology, College of Life Sciences, Shaanxi Normal University, Xi’an 710062, China; (Q.D.); or (X.X.)
| | - Shou-Ping Gong
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi’an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi’an Medical University, Xi’an 710021, China; (S.N.); (P.L.); (Y.F.); (Y.Z.); (S.-P.G.)
| | - Huiling Cao
- Key Laboratory of Pharmacodynamics and Material Basis of Chinese Medicine of Shaanxi Administration of Traditional Chinese Medicine, Engineering Research Center of Brain Health Industry of Chinese Medicine, Pharmacology of Chinese Medicine, Shaanxi University of Chinese Medicine, University Government Committee of Shaanxi Province, Xianyang 712046, China;
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi’an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi’an Medical University, Xi’an 710021, China; (S.N.); (P.L.); (Y.F.); (Y.Z.); (S.-P.G.)
| |
Collapse
|
6
|
Shi L, Sun T, Huo D, Geng L, Zhao C, Xia W. ETV5-Mediated Transcriptional Repression of DDIT4 Blocks Macrophage Pro-Inflammatory Activation in Diabetic Atherosclerosis. Cardiovasc Toxicol 2025; 25:379-394. [PMID: 39864045 DOI: 10.1007/s12012-024-09956-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 12/28/2024] [Indexed: 01/27/2025]
Abstract
Atherosclerosis risk is elevated in diabetic patients, but the underlying mechanism such as the involvement of macrophages remains unclear. Here, we investigated the underlying mechanism related to the pro-inflammatory activation of macrophages in the development of diabetic atherosclerosis. Bioinformatics tools were used to analyze the macrophage-related transcriptome differences in patients with atherosclerosis and diabetic mice. LDLR-/- mice with DDIT4 depletion were generated and fed a Western diet to induce atherosclerosis. DDIT4 expression was elevated in diabetic mice and patients with atherosclerosis. Macrophage proinflammatory factors F4/80, Il-6, and TNFα were reduced in DDIT4-/-LDLR-/- mice and necrotic areas were decreased in the aortic root. Atherosclerotic plaque stability was increased in DDIT4-/-LDLR-/- mice, as evidenced by increased collagen and smooth muscle cell content. DDIT4, regulated by ETV5, acted on macrophages, affecting lipid accumulation, migration capacity, and pro-inflammatory responses. Knockdown of ETV5 increased expression of DDIT4 and pro-inflammatory factors in macrophages, increased necrotic core area in the aortic root, and decreased stability of atherosclerotic plaques in mice, which was abated by DDIT4 knockdown. The findings provide new insight into how diabetes promotes atherosclerosis and support a model wherein loss of ETV5 sustains transcription of DDIT4 and the pro-inflammatory activation of macrophages.
Collapse
MESH Headings
- Animals
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Atherosclerosis/prevention & control
- Plaque, Atherosclerotic
- Macrophages/metabolism
- Macrophages/pathology
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Transcription Factors/deficiency
- Humans
- Mice, Knockout
- Disease Models, Animal
- Inflammation Mediators/metabolism
- Male
- Mice, Inbred C57BL
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Macrophage Activation
- Diabetic Angiopathies/genetics
- Diabetic Angiopathies/pathology
- Diabetic Angiopathies/metabolism
- Diabetic Angiopathies/prevention & control
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Receptors, LDL/deficiency
- Receptors, LDL/genetics
- Aortic Diseases/pathology
- Aortic Diseases/genetics
- Aortic Diseases/metabolism
- Aortic Diseases/prevention & control
- Signal Transduction
- Aorta/pathology
- Aorta/metabolism
- Transcription, Genetic
- Cytokines/metabolism
- Female
- Mice
Collapse
Affiliation(s)
- Lili Shi
- Department of Cadre Ward, The First Affiliated Hospital of Harbin Medical University, No. 23, Postal Street, Harbin, 150001, Heilongjiang, PR China
| | - Tingting Sun
- Department of Cadre Ward, The First Affiliated Hospital of Harbin Medical University, No. 23, Postal Street, Harbin, 150001, Heilongjiang, PR China
| | - Di Huo
- Department of Cadre Ward, The First Affiliated Hospital of Harbin Medical University, No. 23, Postal Street, Harbin, 150001, Heilongjiang, PR China
| | - Lin Geng
- Department of Cadre Ward, The First Affiliated Hospital of Harbin Medical University, No. 23, Postal Street, Harbin, 150001, Heilongjiang, PR China
| | - Chao Zhao
- Department of Cadre Ward, The First Affiliated Hospital of Harbin Medical University, No. 23, Postal Street, Harbin, 150001, Heilongjiang, PR China
| | - Wenbo Xia
- Department of Cadre Ward, The First Affiliated Hospital of Harbin Medical University, No. 23, Postal Street, Harbin, 150001, Heilongjiang, PR China.
| |
Collapse
|
7
|
Tang KT, Chen YH, Wang JH, Hsieh TY, Chao YH, Chen DY, Chen HH, Lin CC. Autophagic Flux Might Be Blocked in Patients With Primary Antiphospholipid Antibody Syndrome. Int J Rheum Dis 2025; 28:e70063. [PMID: 40103288 DOI: 10.1111/1756-185x.70063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/18/2024] [Accepted: 12/31/2024] [Indexed: 03/20/2025]
Affiliation(s)
- Kuo-Tung Tang
- Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, Taichung, Taiwan
- School of Medicine, National Yang-Ming Chiao-Tung University, Taipei, Taiwan
- Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Yi-Hua Chen
- Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Food and Nutrition, Providence University, Taichung, Taiwan
| | - Jou-Hsuan Wang
- Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung-Hsing University, Taichung, Taiwan
| | - Tsu-Yi Hsieh
- Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Ya-Hsuan Chao
- Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung-Hsing University, Taichung, Taiwan
- Division of Chest Medicine, Department of Internal Medicine, Changhua Christian Hospital, Changhua, Taiwan
| | - Der-Yuan Chen
- Rheumatology and Immunology Center, China Medical University Hospital, Taichung, Taiwan
- College of Medicine, China Medical University, Taichung, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Hsin-Hua Chen
- Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
- Division of General Medicine, Department of Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- Program in Translational Medicine and Rong Hsing Research Center for Translational Medi-Cine, National Chung Hsing University, Taichung, Taiwan
- Big Data Center, National Chung Hsing University, Taichung, Taiwan
- Department of Industrial Engineering and Enterprise Information, Tunghai University, Taichung, Taiwan
| | - Chi-Chen Lin
- Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung, Taiwan
- Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung-Hsing University, Taichung, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
- Department of Pharmacology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
8
|
Wang P, Li JX, Kong YY, Zheng SL, Miao CY. Nicotinamide Mononucleotide and Nicotinamide Riboside Improve Dyslipidemia and Fatty Liver but Promote Atherosclerosis in Apolipoprotein E Knockout Mice. Pharmaceuticals (Basel) 2025; 18:281. [PMID: 40143060 PMCID: PMC11944796 DOI: 10.3390/ph18030281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 01/27/2025] [Accepted: 02/10/2025] [Indexed: 03/28/2025] Open
Abstract
Background: Nicotinamide mononucleotide (NMN) and nicotinamide riboside (NR) are intermediary products in NAD+ metabolism. NMN and NR supplementation can elevate NAD+ levels in tissues, addressing health issues associated with aging and obesity. However, the impact of NMN and NR on atherosclerosis remains incompletely elucidated. Methods: C57BL/6J and Apolipoprotein E knockout (ApoE-/-) mice were used to explore the impact of NMN and NR supplementation on serum lipids, fatty liver, and atherosclerosis. Additionally, various suppliers, administration protocols, and doses on ApoE-/- mice were investigated. Results: The intragastric administration of NMN (300 mg/kg) and NR (230 mg/kg) reduced body weight, serum lipids, and fatty liver but aggravated atherosclerosis in ApoE-/- mice after 4 months of administration with different suppliers. Atherosclerosis also deteriorated after 2 months of different NMN administration protocols (intragastric and water administration) in ApoE-/- mice with existing plaques. The effects of NMN were dose-dependent, and doses around 100 mg/kg had little harmful effects on atherosclerosis. Conclusions: NMN and NR improve dyslipidemia and fatty liver but promote atherosclerosis in ApoE-/- mice. These findings emphasize the safe dosage for the clinical trials of NMN.
Collapse
Affiliation(s)
| | | | | | | | - Chao-Yu Miao
- Department of Pharmacology, Second Military Medical University/Naval Medical University, Shanghai 200433, China; (P.W.); (J.-X.L.); (Y.-Y.K.); (S.-L.Z.)
| |
Collapse
|
9
|
Kausar MA, Anwar S, Khan YS, Saleh AA, Ahmed MAA, Kaur S, Iqbal N, Siddiqui WA, Najm MZ. Autophagy and Cancer: Insights into Molecular Mechanisms and Therapeutic Approaches for Chronic Myeloid Leukemia. Biomolecules 2025; 15:215. [PMID: 40001518 PMCID: PMC11853340 DOI: 10.3390/biom15020215] [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: 12/29/2024] [Revised: 01/20/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
Autophagy is a critical cellular process that maintains homeostasis by recycling damaged or aberrant components. This process is orchestrated by a network of proteins that form autophagosomes, which engulf and degrade intracellular material. In cancer, autophagy plays a dual role: it suppresses tumor initiation in the early stages but supports tumor growth and survival in advanced stages. Chronic myeloid leukemia (CML), a hematological malignancy, is characterized by the Philadelphia chromosome, a chromosomal abnormality resulting from a translocation between chromosomes 9 and 22. Autophagy has emerged as a key factor in CML pathogenesis, promoting cancer cell survival and contributing to resistance against tyrosine kinase inhibitors (TKIs), the primary treatment for CML. Targeting autophagic pathways is being actively explored as a therapeutic approach to overcome drug resistance and enhance cancer cell death. Recent research highlights the intricate interplay between autophagy and CML progression, underscoring its role in disease biology and treatment outcomes. This review aims to provide a comprehensive analysis of the molecular and cellular mechanisms underlying CML, with a focus on the therapeutic potential of targeting autophagy.
Collapse
MESH Headings
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Autophagy/drug effects
- Protein Kinase Inhibitors/therapeutic use
- Protein Kinase Inhibitors/pharmacology
- Animals
- Drug Resistance, Neoplasm/drug effects
- Antineoplastic Agents/therapeutic use
- Antineoplastic Agents/pharmacology
Collapse
Affiliation(s)
- Mohd Adnan Kausar
- Department of Biochemistry, College of Medicine, University of Ha’il, Hail 55476, Saudi Arabia;
| | - Sadaf Anwar
- Department of Biochemistry, College of Medicine, University of Ha’il, Hail 55476, Saudi Arabia;
| | - Yusuf Saleem Khan
- Department of Anatomy, College of Medicine, University of Ha’il, Hail 55476, Saudi Arabia;
| | - Ayman A. Saleh
- Department of Pathology, College of Medicine, University of Ha’il, Hail 55476, Saudi Arabia;
| | | | - Simran Kaur
- School of Biosciences, Apeejay Stya University, Sohna, Gurugram 122103, Haryana, India;
| | - Naveed Iqbal
- Department of Obstetrics and Gynecology, College of Medicine, University of Ha’il, Ha’il 55476, Saudi Arabia;
| | - Waseem Ahmad Siddiqui
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202001, Uttar Pradesh, India;
| | - Mohammad Zeeshan Najm
- School of Biosciences, Apeejay Stya University, Sohna, Gurugram 122103, Haryana, India;
| |
Collapse
|
10
|
Buttari B, Tramutola A, Rojo AI, Chondrogianni N, Saha S, Berry A, Giona L, Miranda JP, Profumo E, Davinelli S, Daiber A, Cuadrado A, Di Domenico F. Proteostasis Decline and Redox Imbalance in Age-Related Diseases: The Therapeutic Potential of NRF2. Biomolecules 2025; 15:113. [PMID: 39858508 PMCID: PMC11764413 DOI: 10.3390/biom15010113] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 12/20/2024] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (NRF2) is a master regulator of cellular homeostasis, overseeing the expression of a wide array of genes involved in cytoprotective processes such as antioxidant and proteostasis control, mitochondrial function, inflammation, and the metabolism of lipids and glucose. The accumulation of misfolded proteins triggers the release, stabilization, and nuclear translocation of NRF2, which in turn enhances the expression of critical components of both the proteasomal and lysosomal degradation pathways. This process facilitates the clearance of toxic protein aggregates, thereby actively maintaining cellular proteostasis. As we age, the efficiency of the NRF2 pathway declines due to several factors including increased activity of its repressors, impaired NRF2-mediated antioxidant and cytoprotective gene expression, and potential epigenetic changes, though the precise mechanisms remain unclear. This leads to diminished antioxidant defenses, increased oxidative damage, and exacerbated metabolic dysregulation and inflammation-key contributors to age-related diseases. Given NRF2's role in mitigating proteotoxic stress, the pharmacological modulation of NRF2 has emerged as a promising therapeutic strategy, even in aged preclinical models. By inducing NRF2, it is possible to mitigate the damaging effects of oxidative stress, metabolic dysfunction, and inflammation, thus reducing protein misfolding. The review highlights NRF2's therapeutic implications for neurodegenerative diseases and cardiovascular conditions, emphasizing its role in improving proteostasis and redox homeostasis Additionally, it summarizes current research into NRF2 as a therapeutic target, offering hope for innovative treatments to counteract the effects of aging and associated diseases.
Collapse
Affiliation(s)
- Brigitta Buttari
- Department of Cardiovascular and Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, 00161 Rome, Italy; (B.B.); (E.P.)
| | - Antonella Tramutola
- Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza University, 00185 Rome, Italy;
| | - Ana I. Rojo
- Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), National Institute of Health Carlos III (ISCIII), Instituto de Investigación Sanitaria La Paz (IdiPaz), 28049 Madrid, Spain; (A.I.R.); (A.C.)
| | - Niki Chondrogianni
- Institute of Chemical Biology, National Hellenic Research Foundation, 116 35 Athens, Greece;
| | - Sarmistha Saha
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Mathura 00185, Uttar Pradesh, India;
| | - Alessandra Berry
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.B.); (L.G.)
| | - Letizia Giona
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.B.); (L.G.)
- PhD Program in Science of Nutrition, Metabolism, Aging and Gender-Related Diseases, Faculty of Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Joana P. Miranda
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisboa, Portugal;
| | - Elisabetta Profumo
- Department of Cardiovascular and Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, 00161 Rome, Italy; (B.B.); (E.P.)
| | - Sergio Davinelli
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy;
| | - Andreas Daiber
- Department for Cardiology 1, University Medical Center Mainz, Molecular Cardiology, Johannes Gutenberg University, 55131 Mainz, Germany;
| | - Antonio Cuadrado
- Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), National Institute of Health Carlos III (ISCIII), Instituto de Investigación Sanitaria La Paz (IdiPaz), 28049 Madrid, Spain; (A.I.R.); (A.C.)
| | - Fabio Di Domenico
- Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza University, 00185 Rome, Italy;
| |
Collapse
|
11
|
Lin Y, Yang M, Liu Q, Cai Y, Zhang Z, Xu C, Luo M. Apolipoprotein E Gene ε4 Allele is Associated with Atherosclerosis in Multiple Vascular Beds. Int J Gen Med 2024; 17:5039-5048. [PMID: 39512258 PMCID: PMC11542474 DOI: 10.2147/ijgm.s475771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 10/26/2024] [Indexed: 11/15/2024] Open
Abstract
Background Atherosclerosis is a systemic disease that can involve multiple vascular beds. The risk factors for atherosclerosis in multiple vascular beds remain unclear. Apolipoprotein E (APOE) is involved in inflammation and lipid deposition in the process of atherosclerosis. The objective of this study was to investigate whether APOE polymorphisms are associated with atherosclerosis in multiple vascular beds. Methods A total of 416 patients with atherosclerosis in single vascular bed and 658 patients with atherosclerosis in multiple vascular beds were included. APOE genotypes were detected and the differences of APOE genotypes between the groups were compared. Logistic regression analysis was performed to analyze the relationship between APOE genotypes and atherosclerosis in multiple vascular beds. Results APOE E3/E4 genotype frequency was lower in the patients with atherosclerosis in multiple vascular beds than that of patients with atherosclerosis in single vascular bed (11.4% vs 17.8%, P=0.004). There was no significant difference in age and gender distribution, proportion of history of smoking, alcohol consumption, hypertension, and diabetes mellitus between the two groups (all P>0.05), and among patients with different APOE alleles (all P>0.05). Logistic regression analysis indicated that APOE E3/E4 genotype (E3/E4 vs E3/E3: odds ratio (OR) 0.598, 95% confidence interval (CI): 0.419-0.854, P=0.005), and APOE ε4 allele (ε4 vs ε3: OR 0.630, 95% CI: 0.444-0.895, P=0.010) associated with atherosclerosis in multiple vascular beds. Conclusion APOE ε4 allele is associated with atherosclerosis in multiple vascular beds.
Collapse
Affiliation(s)
- Youni Lin
- Center for Cardiovascular Diseases, Meizhou People’s Hospital, Meizhou, People’s Republic of China
| | - Min Yang
- Center for Cardiovascular Diseases, Meizhou People’s Hospital, Meizhou, People’s Republic of China
| | - Qifeng Liu
- Center for Cardiovascular Diseases, Meizhou People’s Hospital, Meizhou, People’s Republic of China
| | - Yufu Cai
- Center for Cardiovascular Diseases, Meizhou People’s Hospital, Meizhou, People’s Republic of China
| | - Zhouhua Zhang
- Center for Cardiovascular Diseases, Meizhou People’s Hospital, Meizhou, People’s Republic of China
| | - Chongfei Xu
- Center for Cardiovascular Diseases, Meizhou People’s Hospital, Meizhou, People’s Republic of China
| | - Ming Luo
- Center for Cardiovascular Diseases, Meizhou People’s Hospital, Meizhou, People’s Republic of China
| |
Collapse
|
12
|
Zhu L, Liao Y, Jiang B. Role of ROS and autophagy in the pathological process of atherosclerosis. J Physiol Biochem 2024; 80:743-756. [PMID: 39110405 DOI: 10.1007/s13105-024-01039-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 07/25/2024] [Indexed: 12/29/2024]
Abstract
Activation of autophagy and production of reactive oxygen species occur at various stages of atherosclerosis. To clarify the role and mechanism of autophagy and reactive oxygen species in atherosclerosis is of great significance to the prevention and treatment of atherosclerosis. Recent studies have shown that basal autophagy plays an important role in protecting cells from oxidative stress, reducing apoptosis and enhancing atherosclerotic plaque stability. Autophagy deficiency and excessive accumulation of reactive oxygen species can impair the function of endothelial cells, macrophages and smooth muscle cells, trigger autophagic cell death, and lead to instability and even rupture of plaques. However, the main signaling pathways regulating autophagy, the molecular mechanisms of autophagy and reactive oxygen species interaction, how they are initiated and distributed in plaques, and how they affect atherosclerosis progression, remain to be clarified. At present, there is no autophagy inducer used to treat atherosclerosis clinically. Therefore, it is urgent to clarify the mechanism of autophagy and find new targets for autophagy. Antioxidant agents generally have defects such as low reactive oxygen species scavenging efficiency and high cytotoxicity. Highly potent autophagy inducers and reactive oxygen species scavengers still need to be further developed and validated to provide more possibilities for innovative treatments for atherosclerosis.
Collapse
Affiliation(s)
- Liyuan Zhu
- Center of Clinical Pharmacology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yingnan Liao
- Department of Laboratory Medicine, Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical Genetics, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Research Unit for Blindness Prevention, Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Bo Jiang
- Center of Clinical Pharmacology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
13
|
Liu X, Zhou Q, Meng J, Zuo H, Li R, Zhang R, Lu H, Zhang Z, Li H, Zeng S, Tian M, Wang H, Hu K, Li N, Mao L, Hou S. Autophagy-mediated activation of the AIM2 inflammasome enhances M1 polarization of microglia and exacerbates retinal neovascularization. MedComm (Beijing) 2024; 5:e668. [PMID: 39081514 PMCID: PMC11286542 DOI: 10.1002/mco2.668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Retinopathy of prematurity (ROP) is a retinal neovascularization (RNV) disease that is characterized by abnormal blood vessel development in the retina. Importantly, the etiology of ROP remains understudied. We re-analyzed previously published single-cell data and discovered a strong correlation between microglia and RNV diseases, particularly ROP. Subsequently, we found that reactive oxygen species reduced autophagy-dependent protein degradation of absent in melanoma 2 (AIM2) in hypoxic BV2 cells, leading to increased AIM2 protein accumulation. Furthermore, we engineered AIM2 knockout mice and observed that the RNV was significantly reduced compared to wild-type mice. In vitro vascular function assays also demonstrated diminished angiogenic capabilities following AIM2 knockdown in hypoxic BV2 cells. Mechanistically, AIM2 enhanced the M1-type polarization of microglia via the ASC/CASP1/IL-1β pathway, resulting in RNV. Notably, the administration of recombinant protein IL-1β exacerbated angiogenesis, while its inhibition ameliorated the condition. Taken together, our study provides a novel therapeutic target for ROP and offers insight into the interaction between pyroptosis and autophagy.
Collapse
Affiliation(s)
- Xianyang Liu
- The First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of OphthalmologyChongqingChina
| | - Qian Zhou
- The First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of OphthalmologyChongqingChina
| | - Jiayu Meng
- Sichuan Provincial Key Laboratory for Human Disease Gene StudySichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Hangjia Zuo
- The First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of OphthalmologyChongqingChina
| | - Ruonan Li
- The First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of OphthalmologyChongqingChina
| | - Rui Zhang
- Department of OphthalmologyQilu HospitalCheeloo College of MedicineShandong UniversityJinanChina
| | - Huiping Lu
- The First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of OphthalmologyChongqingChina
| | - Zhi Zhang
- The First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of OphthalmologyChongqingChina
| | - Hongshun Li
- The First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of OphthalmologyChongqingChina
| | - Shuhao Zeng
- The First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of OphthalmologyChongqingChina
| | - Meng Tian
- Beijing Institute of OphthalmologyBeijing Tongren Eye CenterBeijing Ophthalmology & Visual Sciences Key LaboratoryBeijing Tongren HospitalCapital Medical UniversityBeijingChina
| | - Hong Wang
- Beijing Institute of OphthalmologyBeijing Tongren Eye CenterBeijing Ophthalmology & Visual Sciences Key LaboratoryBeijing Tongren HospitalCapital Medical UniversityBeijingChina
| | - Ke Hu
- The First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of OphthalmologyChongqingChina
| | - Na Li
- Department of Laboratory Medicine, Beijing Tongren HospitalCapital Medical UniversityBeijingChina
| | - Liming Mao
- Department of ImmunologySchool of MedicineNantong UniversityNantongChina
| | - Shengping Hou
- The First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Beijing Institute of OphthalmologyBeijing Tongren Eye CenterBeijing Ophthalmology & Visual Sciences Key LaboratoryBeijing Tongren HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
14
|
Sharma H, Mossman K, Austin RC. Fatal attractions that trigger inflammation and drive atherosclerotic disease. Eur J Clin Invest 2024; 54:e14169. [PMID: 38287209 DOI: 10.1111/eci.14169] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 12/14/2023] [Accepted: 01/09/2024] [Indexed: 01/31/2024]
Abstract
BACKGROUND Atherosclerosis is the salient, underlying cause of cardiovascular diseases, such as arrhythmia, coronary artery disease, cardiomyopathy, pulmonary embolism and myocardial infarction. In recent years, atherosclerosis pathophysiology has evolved from a lipid-based to an inflammation-centric ideology. METHODS This narrative review is comprised of review and original articles that were found through the PubMed search engine. The following search terms or amalgamation of terms were used: "cardiovascular disease," "atherosclerosis," "inflammation," "GRP78," "Hsp60," "oxidative low-density lipoproteins," "aldehyde dehydrogenase," "β2-glycoprotein," "lipoprotein lipase A," "human cytomegalovirus." "SARS-CoV-2," "chlamydia pneumonia," "autophagy," "thrombosis" and "therapeutics." RESULTS Emerging evidence supports the concept that atherosclerosis is associated with the interaction between cell surface expression of stress response chaperones, including GRP78 and Hsp60, and their respective autoantibodies. Moreover, various other autoantigens and their autoantibodies have displayed a compelling connection with the development of atherosclerosis, including oxidative low-density lipoproteins, aldehyde dehydrogenase, β2-glycoprotein and lipoprotein lipase A. Atherosclerosis progression is also concurrent with viral and bacterial activators of various diseases. This narrative review will focus on the contributions of human cytomegalovirus as well as SARS-CoV-2 and chlamydia pneumonia in atherosclerosis development. Notably, the interaction of an autoantigen with their respective autoantibodies or the presence of a foreign antigen can enhance inflammation development, which leads to atherosclerotic lesion progression. CONCLUSION We will highlight and discuss the complex role of the interaction between autoantigens and autoantibodies, and the presence of foreign antigens in the development of atherosclerotic lesions in relationship to pro-inflammatory responses.
Collapse
Affiliation(s)
- Hitesh Sharma
- Division of Nephrology, Department of Medicine, McMaster University, The Research Institute of St. Joe's Hamilton and the Hamilton Centre for Kidney Research, Hamilton, Ontario, Canada
| | - Karen Mossman
- Department of Medicine, Michael DeGroote Institute for Infectious Disease Research and the McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Richard C Austin
- Division of Nephrology, Department of Medicine, McMaster University, The Research Institute of St. Joe's Hamilton and the Hamilton Centre for Kidney Research, Hamilton, Ontario, Canada
| |
Collapse
|
15
|
Cui X, Wang B, Han D, Cheng M, Yuan P, Du P, Hou Y, Su C, Tang J, Zhang J. Exacerbation of atherosclerosis by STX17 knockdown: Unravelling the role of autophagy and inflammation. J Cell Mol Med 2024; 28:e18402. [PMID: 39008328 PMCID: PMC11133389 DOI: 10.1111/jcmm.18402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 04/13/2024] [Accepted: 04/27/2024] [Indexed: 07/16/2024] Open
Abstract
Syntaxin 17 (STX17) has been identified as a crucial factor in mediating the fusion of autophagosomes and lysosomes. However, its specific involvement in the context of atherosclerosis (AS) remains unclear. This study sought to elucidate the role and mechanistic contributions of STX17 in the initiation and progression of AS. Utilizing both in vivo and in vitro AS model systems, we employed ApoE knockout (KO) mice subjected to a high-fat diet and human umbilical vein endothelial cells (HUVECs) treated with oxidized low-density lipoprotein (ox-LDL) to assess STX17 expression. To investigate underlying mechanisms, we employed shRNA-STX17 lentivirus to knock down STX17 expression, followed by evaluating autophagy and inflammation in HUVECs. In both in vivo and in vitro AS models, STX17 expression was significantly upregulated. Knockdown of STX17 exacerbated HUVEC damage, both with and without ox-LDL treatment. Additionally, we observed that STX17 knockdown impaired autophagosome degradation, impeded autophagy flux and also resulted in the accumulation of dysfunctional lysosomes in HUVECs. Moreover, STX17 knockdown intensified the inflammatory response following ox-LDL treatment in HUVECs. Further mechanistic exploration revealed an association between STX17 and STING; reducing STX17 expression increased STING levels. Further knockdown of STING enhanced autophagy flux. In summary, our findings suggest that STX17 knockdown worsens AS by impeding autophagy flux and amplifying the inflammatory response. Additionally, the interaction between STX17 and STING may play a crucial role in STX17-mediated autophagy.
Collapse
Affiliation(s)
- Xinyue Cui
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Key Laboratory of Cardiac Injury and Repair of Henan ProvinceZhengzhouHenanChina
| | - Bo Wang
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Key Laboratory of Cardiac Injury and Repair of Henan ProvinceZhengzhouHenanChina
| | - Dongjian Han
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Key Laboratory of Cardiac Injury and Repair of Henan ProvinceZhengzhouHenanChina
| | - Mengdie Cheng
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Key Laboratory of Cardiac Injury and Repair of Henan ProvinceZhengzhouHenanChina
| | - Peiyu Yuan
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Key Laboratory of Cardiac Injury and Repair of Henan ProvinceZhengzhouHenanChina
| | - Pengchong Du
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Key Laboratory of Cardiac Injury and Repair of Henan ProvinceZhengzhouHenanChina
| | - Yachen Hou
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Key Laboratory of Cardiac Injury and Repair of Henan ProvinceZhengzhouHenanChina
| | - Chang Su
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Key Laboratory of Cardiac Injury and Repair of Henan ProvinceZhengzhouHenanChina
| | - Junnan Tang
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Key Laboratory of Cardiac Injury and Repair of Henan ProvinceZhengzhouHenanChina
| | - Jinying Zhang
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Key Laboratory of Cardiac Injury and Repair of Henan ProvinceZhengzhouHenanChina
| |
Collapse
|
16
|
Preda A, Montecucco F, Carbone F, Camici GG, Lüscher TF, Kraler S, Liberale L. SGLT2 inhibitors: from glucose-lowering to cardiovascular benefits. Cardiovasc Res 2024; 120:443-460. [PMID: 38456601 PMCID: PMC12001887 DOI: 10.1093/cvr/cvae047] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 01/03/2024] [Accepted: 02/05/2024] [Indexed: 03/09/2024] Open
Abstract
An increasing number of individuals are at high risk of type 2 diabetes (T2D) and its cardiovascular complications, including heart failure (HF), chronic kidney disease (CKD), and eventually premature death. The sodium-glucose co-transporter-2 (SGLT2) protein sits in the proximal tubule of human nephrons to regulate glucose reabsorption and its inhibition by gliflozins represents the cornerstone of contemporary T2D and HF management. Herein, we aim to provide an updated overview of the pleiotropy of gliflozins, provide mechanistic insights and delineate related cardiovascular (CV) benefits. By discussing contemporary evidence obtained in preclinical models and landmark randomized controlled trials, we move from bench to bedside across the broad spectrum of cardio- and cerebrovascular diseases. With landmark randomized controlled trials confirming a reduction in major adverse CV events (MACE; composite endpoint of CV death, non-fatal myocardial infarction, and non-fatal stroke), SGLT2 inhibitors strongly mitigate the risk for heart failure hospitalization in diabetics and non-diabetics alike while conferring renoprotection in specific patient populations. Along four major pathophysiological axes (i.e. at systemic, vascular, cardiac, and renal levels), we provide insights into the key mechanisms that may underlie their beneficial effects, including gliflozins' role in the modulation of inflammation, oxidative stress, cellular energy metabolism, and housekeeping mechanisms. We also discuss how this drug class controls hyperglycaemia, ketogenesis, natriuresis, and hyperuricaemia, collectively contributing to their pleiotropic effects. Finally, evolving data in the setting of cerebrovascular diseases and arrhythmias are presented and potential implications for future research and clinical practice are comprehensively reviewed.
Collapse
Affiliation(s)
- Alberto Preda
- Department of Clinical Cardiology, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa—Italian Cardiovascular Network, Genoa, Italy
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa—Italian Cardiovascular Network, Genoa, Italy
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zürich, Schlieren, Switzerland
- Department of Research and Education, University Hospital Zurich, Zurich, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zürich, Schlieren, Switzerland
- Royal Brompton and Harefield Hospitals and Imperial College and King’s College, London, United Kingdom
| | - Simon Kraler
- Center for Molecular Cardiology, University of Zürich, Schlieren, Switzerland
- Department of Internal Medicine, Cantonal Hospital Baden, Baden, Switzerland
| | - Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa—Italian Cardiovascular Network, Genoa, Italy
| |
Collapse
|
17
|
Zhou G, Liu Y, Wu H, Zhang D, Yang Q, Li Y. Research Progress on Histone Deacetylases Regulating Programmed Cell Death in Atherosclerosis. J Cardiovasc Transl Res 2024; 17:308-321. [PMID: 37821683 DOI: 10.1007/s12265-023-10444-z] [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: 05/07/2023] [Accepted: 09/28/2023] [Indexed: 10/13/2023]
Abstract
Histone deacetylases (HDACs) are epigenetic modifying enzyme that is closely related to chromatin structure and gene transcription, and numerous studies have found that HDACs play an important regulatory role in atherosclerosis disease. Apoptosis, autophagy and programmed necrosis as the three typical programmed cell death modalities that can lead to cell loss and are closely related to the developmental process of atherosclerosis. In recent years, accumulating evidence has shown that the programmed cell death mediated by HDACs is increasingly important in the pathophysiology of atherosclerosis. This paper first gives a brief overview of HDACs, the mechanism of programmed cell death, and their role in atherosclerosis, and then further elaborates on the role and mechanism of HDACs in regulating apoptosis, autophagy, and programmed necrosis in atherosclerosis, respectively, to provide new effective measures and theoretical basis for the prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Gang Zhou
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, 443003, China
- Department of Central Experimental Laboratory, Yichang Central People's Hospital, Yichang, 443003, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, 443003, China
| | - Yanfang Liu
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, 443003, China
- Department of Central Experimental Laboratory, Yichang Central People's Hospital, Yichang, 443003, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, 443003, China
| | - Hui Wu
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, 443003, China.
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, 443003, China.
- Department of Cardiology, Yichang Central People's Hospital, Yiling Road 183, Yichang, 443003, Hubei, China.
| | - Dong Zhang
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, 443003, China
- Department of Central Experimental Laboratory, Yichang Central People's Hospital, Yichang, 443003, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, 443003, China
| | - Qingzhuo Yang
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, 443003, China
- Department of Central Experimental Laboratory, Yichang Central People's Hospital, Yichang, 443003, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, 443003, China
| | - Yi Li
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, 443003, China
- Department of Central Experimental Laboratory, Yichang Central People's Hospital, Yichang, 443003, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, 443003, China
| |
Collapse
|
18
|
Liang Y, Fu J, Shi Y, Jiang X, Lu F, Liu S. Integration of 16S rRNA sequencing and metabolomics to investigate the modulatory effect of ginsenoside Rb1 on atherosclerosis. Heliyon 2024; 10:e27597. [PMID: 38500998 PMCID: PMC10945261 DOI: 10.1016/j.heliyon.2024.e27597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 03/04/2024] [Accepted: 03/04/2024] [Indexed: 03/20/2024] Open
Abstract
Background /aims: Atherosclerosis (AS) is the common pathological basis of a variety of cardiovascular diseases (CVD), and has become the main cause of human death worldwide, and the incidence is increasing and younger trend. Ginsenoside Rb1 (Rb1), an important monomer component of the traditional Chinese herb ginseng, known for its ability to improve blood lipid disorders and anti-inflammatory. In addition, Rb1 was proved to be an effective treatment for AS. However, the effect of Rb1 on AS remains to be elucidated. The aim of this study was to investigate the mechanisms of Rb1 in ameliorating AS induced by high-fat diet (HFD). Materials and methods In this study, we developed an experimental AS model in Sprague-Dawley rats by feeding HFD with intraperitoneal injection of vitamin D3. The potential therapeutic mechanism of Rb1 in AS rats was investigated by detecting the expression of inflammatory factors, microbiome 16S rRNA gene sequencing, short-chain fatty acids (SCFAs) targeted metabolomics and untargeted metabolomics. Results Rb1 could effectively alleviate the symptoms of AS and suppress the overexpression of inflammation-related factors. Meanwhile, Rb1 altered gut microbial composition and concentration of SCFAs characterized by Bacteroidetes, Actinobacteria, Lactobacillus, Prevotella, Oscillospira enrichment and Desulfovibrio depletion, accompanied by increased production of acetic acid and propionic acid. Moreover, untargeted metabolomics showed that Rb1 considerably improved faecal metabolite profiles, particularly arachidonic acid metabolism and primary bile acid biosynthesis. Conclusion Rb1 ameliorated the HFD-induced AS, and the mechanism is related to improving intestinal metabolic homeostasis and inhibiting systemic inflammation by regulating gut microbiota.
Collapse
Affiliation(s)
- Yuqin Liang
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Jiaqi Fu
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Yunhe Shi
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Xin Jiang
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Fang Lu
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Shumin Liu
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| |
Collapse
|
19
|
Vazirian F, Darroudi S, Rahimi HR, Latifi M, Shakeri B, Abolbashari S, Mohammadpour AH, Esmaily H, Mouhebati M, Samadi S, Mobarhan MG. Non-HDL cholesterol and long-term follow-up outcomes in patients with metabolic syndrome. Lipids Health Dis 2023; 22:165. [PMID: 37794473 PMCID: PMC10548659 DOI: 10.1186/s12944-023-01923-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 09/11/2023] [Indexed: 10/06/2023] Open
Abstract
BACKGROUND Non-high-density lipoprotein-cholesterol (non-HDL-C) has been identified as a potential biomarker for metabolic syndrome (MetS). However, its predictive capability for MetS varies among different ethnic groups, necessitating further investigation. This study aimed to assess the role of non-HDL-C in the early diagnosis of MetS in the Iranian population through a longitudinal study with a 10-year follow-up period. METHODS Our study enrolled 4684 individuals from the MASHAD (Mashhad Stroke and Heart Atherosclerotic Disorder) cohort who were followed for 10 years to examine the association between non-HDL-C and the incidence of MetS. Additionally, the contribution of individual MetS components to the overall burden was evaluated. RESULTS A total of 1599 subjects developed MetS, while 3085 did not. Non-HDL-C levels ≥ 130 were associated with a 42% higher risk of developing MetS (relative risk (RR), 1.42; 95% confidence interval (CI), 1.25-1.62). Regarding MetS components, elevated waist circumference (WC) showed the strongest association with MetS incidence (RR, 2.32; 95% CI, 1.45-2.9), whereas triglyceride (TG) levels ≥ 150 mg/dL demonstrated the weakest association (RR, 1.23; 95% CI, 1.04-1.46). Additionally, higher HDL-C levels were reported to be 20% protective against the risk of MetS (RR, 0.8; 95% CI, 0.73-0.86). Moreover, fasting blood glucose (FBG) levels ≥ 100 mg/dL were not significantly linked to MetS burden, while systolic blood pressure (BP) levels ≥ 130 mmHg or diastolic BP levels ≥ 85 mmHg increased the risk of MetS incidence (RR, 1.25; 95% CI: 1.11-1.41). CONCLUSIONS Elevated non-HDL-C and increased WC serve as significant predictors of MetS in Iranians. Strategies targeting non-HDL-C levels and weight loss should be emphasized to mitigate the risk of MetS development.
Collapse
Affiliation(s)
- Fatemeh Vazirian
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Susan Darroudi
- International UNESCO center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Reza Rahimi
- International UNESCO center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran
- Human Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - MohamadReza Latifi
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Behrouz Shakeri
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Samaneh Abolbashari
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Hooshang Mohammadpour
- Department of Clinical Pharmacy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Habibollah Esmaily
- Department of Biostatistics, School of Health, Mashhad University of Medical Sciences, Mashhad, Iran
- Social Determinants of Health Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohsen Mouhebati
- Cardiovascular Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sara Samadi
- Department of Internal Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Majid Ghayour Mobarhan
- International UNESCO center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran.
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
20
|
Naguib M, Magdy M, Yousef OAE, Ibrahim W, Gharib DM. Circulating MicroRNA-30a, Beclin1 and Their Association with Different Variables in Females with Metabolically Healthy /Unhealthy Obesity. Diabetes Metab Syndr Obes 2023; 16:3065-3074. [PMID: 37810570 PMCID: PMC10559787 DOI: 10.2147/dmso.s428844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 09/26/2023] [Indexed: 10/10/2023] Open
Abstract
Background Obesity is associated with metabolic and cardiovascular co-morbidities. It is important to determine the factors associated with metabolic derangement in obesity. Autophagy plays a major role in the pathogenesis of metabolic syndrome. MicroRNA-30a targets beclin1, the main regulator of autophagy. Purpose We assess circulating microRNA-30a and serum beclin1 in women with metabolically unhealthy obesity (MUO), women with metabolically healthy obesity (MHO) and non-obese healthy control and determine their relationship with different clinical and metabolic variables in women with obesity. Patients and Methods This cross-sectional study included 34 women with MHO, 34 with MUO, and 20 healthy non-obese women. Blood pressure, body mass index (BMI), and waist circumference were recorded. Glycemic and lipid indices, urinary albumin-to-creatinine ratio, ALT, AST, microRNA-30a expression in serum were measured using real-time polymerase chain reaction and beclin1 by enzyme-linked immunosorbent assay were measured. Results The expression of microRNA-30a was significantly higher, and beclin1 level was significantly lower in women with MUO compared to those in women with MHO (P<0.001; for both). People with MUO were significantly older (P<0.001) and had higher TSH (P=0.006), HbA1c (P<0.001), triglyceride (P<0.001), and ALT (P<0.001) compared to women with MHO. However, there was no significant difference between the two groups in any anthropometric measurements, HDL-C or LDL-C. In univariate analyses, age, ALT, TSH, microRNA-30a, and beclin1 were significantly correlated with the MUO phenotype (P<0.001; for all). Significance was confirmed in the multivariate analysis for microRNA-30a (95% CI 1.317-28.252; P=0.021). Conclusion MicroRNA-30a, beclin1, age, and ALT and TSH levels were significantly associated with the MUO phenotype, among which microRNA-30a was the best indicator of metabolic syndrome in women with obesity.
Collapse
Affiliation(s)
- Mervat Naguib
- Diabetes and Endocrinology Unite, Internal Medicine Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mohamed Magdy
- Diabetes and Endocrinology Unite, Internal Medicine Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | | | - Walaa Ibrahim
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Doaa Mostafa Gharib
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
21
|
Xu T, Cai J, Wang L, Xu L, Zhao H, Wang F, Meyron-Holtz EG, Missirlis F, Qiao T, Li K. Hormone replacement therapy for postmenopausal atherosclerosis is offset by late age iron deposition. eLife 2023; 12:e80494. [PMID: 37561022 PMCID: PMC10414966 DOI: 10.7554/elife.80494] [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] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 06/30/2023] [Indexed: 08/11/2023] Open
Abstract
Postmenopausal atherosclerosis (AS) has been attributed to estrogen deficiency. However, the beneficial effect of hormone replacement therapy (HRT) is lost in late postmenopausal women with atherogenesis. We asked whether aging-related iron accumulation affects estrogen receptor α (ERα) expression, thus explaining HRT inefficacy. A negative correlation has been observed between aging-related systemic iron deposition and ERα expression in postmenopausal AS patients. In an ovariectomized Apoe-/- mouse model, estradiol treatment had contrasting effects on ERα expression in early versus late postmenopausal mice. ERα expression was inhibited by iron treatment in cell culture and iron-overloaded mice. Combined treatment with estradiol and iron further decreased ERα expression, and the latter effect was mediated by iron-regulated E3 ligase Mdm2. In line with these observations, cellular cholesterol efflux was reduced, and endothelial homeostasis was disrupted. Consequently, AS was aggravated. Accordingly, systemic iron chelation attenuated estradiol-triggered progressive AS in late postmenopausal mice. Thus, iron and estradiol together downregulate ERα through Mdm2-mediated proteolysis, providing a potential explanation for failures of HRT in late postmenopausal subjects with aging-related iron accumulation. This study suggests that immediate HRT after menopause, along with appropriate iron chelation, might provide benefits from AS.
Collapse
Affiliation(s)
- Tianze Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Vascular Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjingChina
| | - Jing Cai
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Vascular Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjingChina
| | - Lei Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Vascular Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjingChina
| | - Li Xu
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing UniversityNanjingChina
| | - Hongting Zhao
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing UniversityNanjingChina
| | - Fudi Wang
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of MedicineHangzhouChina
| | - Esther G Meyron-Holtz
- Faculty of Biotechnology and Food Engineering, Technion Israel Institute of TechnologyHaifaIsrael
| | - Fanis Missirlis
- Department of Physiology, Biophysics and Neuroscience, CinvestavMexicoMexico
| | - Tong Qiao
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Vascular Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjingChina
| | - Kuanyu Li
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Vascular Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjingChina
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing UniversityNanjingChina
| |
Collapse
|
22
|
Metur SP, Lei Y, Zhang Z, Klionsky DJ. Regulation of autophagy gene expression and its implications in cancer. J Cell Sci 2023; 136:jcs260631. [PMID: 37199330 PMCID: PMC10214848 DOI: 10.1242/jcs.260631] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2023] Open
Abstract
Autophagy is a catabolic cellular process that targets and eliminates superfluous cytoplasmic components via lysosomal degradation. This evolutionarily conserved process is tightly regulated at multiple levels as it is critical for the maintenance of homeostasis. Research in the past decade has established that dysregulation of autophagy plays a major role in various diseases, such as cancer and neurodegeneration. However, modulation of autophagy as a therapeutic strategy requires identification of key players that can fine tune the induction of autophagy without complete abrogation. In this Review, we summarize the recent discoveries on the mechanism of regulation of ATG (autophagy related) gene expression at the level of transcription, post transcription and translation. Furthermore, we briefly discuss the role of aberrant expression of ATG genes in the context of cancer.
Collapse
Affiliation(s)
- Shree Padma Metur
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yuchen Lei
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Zhihai Zhang
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Daniel J. Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
23
|
Wu Q, Lv Q, Liu X, Ye X, Cao L, Wang M, Li J, Yang Y, Li L, Wang S. Natural compounds from botanical drugs targeting mTOR signaling pathway as promising therapeutics for atherosclerosis: A review. Front Pharmacol 2023; 14:1083875. [PMID: 36744254 PMCID: PMC9894899 DOI: 10.3389/fphar.2023.1083875] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 01/05/2023] [Indexed: 01/22/2023] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease that is a major cause of cardiovascular diseases (CVDs), including coronary artery disease, hypertension, myocardial infarction, and heart failure. Hence, the mechanisms of AS are still being explored. A growing compendium of evidence supports that the activity of the mechanistic/mammalian target of rapamycin (mTOR) is highly correlated with the risk of AS. The mTOR signaling pathway contributes to AS progression by regulating autophagy, cell senescence, immune response, and lipid metabolism. Various botanical drugs and their functional compounds have been found to exert anti- AS effects by modulating the activity of the mTOR signaling pathway. In this review, we summarize the pathogenesis of AS based on the mTOR signaling pathway from the aspects of immune response, autophagy, cell senescence, and lipid metabolism, and comb the recent advances in natural compounds from botanical drugs to inhibit the mTOR signaling pathway and delay AS development. This review will provide a new perspective on the mechanisms and precision treatments of AS.
Collapse
Affiliation(s)
- Qian Wu
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Qianyu Lv
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Xiao’an Liu
- Capital University of Medical, Beijing, China
| | - Xuejiao Ye
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Linlin Cao
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Manshi Wang
- Beijing Xicheng District Guangwai Hospital, Beijing, China
| | - Junjia Li
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Yingtian Yang
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Lanlan Li
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Shihan Wang
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
24
|
Bourebaba L, Serwotka-Suszczak A, Pielok A, Sikora M, Mularczyk M, Marycz K. The PTP1B inhibitor MSI-1436 ameliorates liver insulin sensitivity by modulating autophagy, ER stress and systemic inflammation in Equine metabolic syndrome affected horses. Front Endocrinol (Lausanne) 2023; 14:1149610. [PMID: 37020593 PMCID: PMC10067883 DOI: 10.3389/fendo.2023.1149610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 03/08/2023] [Indexed: 04/07/2023] Open
Abstract
BACKGROUND Equine metabolic syndrome (EMS) is a multifactorial pathology gathering insulin resistance, low-grade inflammation and past or chronic laminitis. Among the several molecular mechanisms underlying EMS pathogenesis, increased negative insulin signalling regulation mediated by protein tyrosine phosphatase 1 B (PTP1B) has emerged as a critical axis in the development of liver insulin resistance and general metabolic distress associated to increased ER stress, inflammation and disrupted autophagy. Thus, the use of PTP1B selective inhibitors such as MSI-1436 might be considered as a golden therapeutic tool for the proper management of EMS and associated conditions. Therefore, the present investigation aimed at verifying the clinical efficacy of MSI-1436 systemic administration on liver metabolic balance, insulin sensitivity and inflammatory status in EMS affected horses. Moreover, the impact of MSI-1436 treatment on liver autophagy machinery and associated ER stress in liver tissue has been analysed. METHODS Liver explants isolated from healthy and EMS horses have been treated with MSI-1436 prior to gene and protein expression analysis of main markers mediating ER stress, mitophagy and autophagy. Furthermore, EMS horses have been intravenously treated with a single dose of MSI-1436, and evaluated for their metabolic and inflammatory status. RESULTS Clinical application of MSI-1436 to EMS horses restored proper adiponectin levels and attenuated the typical hyperinsulinemia and hyperglycemia. Moreover, administration of MSI-1436 further reduced the circulating levels of key pro-inflammatory mediators including IL-1β, TNF-α and TGF-β and triggered the Tregs cells activation. At the molecular level, PTP1B inhibition resulted in a noticeable mitigation of liver ER stress, improvement of mitochondrial dynamics and consequently, a regulation of autophagic response. Similarly, short-term ex vivo treatment of EMS liver explants with trodusquemine (MSI-1436) substantially enhanced autophagy by upregulating the levels of HSC70 and Beclin-1 at both mRNA and protein level. Moreover, the PTP1B inhibitor potentiated mitophagy and associated expression of MFN2 and PINK1. Interestingly, inhibition of PTP1B resulted in potent attenuation of ER stress key mediators' expression namely, CHOP, ATF6, HSPA5 and XBP1. CONCLUSION Presented findings shed for the first time promising new insights in the development of an MSI-1436-based therapy for proper equine metabolic syndrome intervention and may additionally find potential translational application to human metabolic syndrome treatment.
Collapse
Affiliation(s)
- Lynda Bourebaba
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
- International Institute of Translational Medicine, Wisznia Mała, Poland
| | - Anna Serwotka-Suszczak
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Ariadna Pielok
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Mateusz Sikora
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Malwina Mularczyk
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
- International Institute of Translational Medicine, Wisznia Mała, Poland
| | - Krzysztof Marycz
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
- *Correspondence: Krzysztof Marycz,
| |
Collapse
|
25
|
Ohba K, Iwaki T. Role of thyroid hormone in an experimental model of atherosclerosis: the potential mediating role of immune response and autophagy. Endocr J 2022; 69:1043-1052. [PMID: 35871569 DOI: 10.1507/endocrj.ej22-0177] [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] [Indexed: 11/23/2022] Open
Abstract
Accumulating evidence has revealed that several conditions related to abnormal thyroid hormone status, such as dyslipidemia, hypertension, or hypercoagulable state, can exacerbate atherosclerotic vascular disease. Thyroid hormone effects on vascular smooth muscle cells and endothelial cells have also been studied extensively. However, only limited information is available on thyroid hormone-mediated immune response in current review articles on the pathophysiology of atherosclerosis. This report thus presents an overview of the recent advances in the understanding of the dynamic interactions taking place between thyroid hormone status and immune response in the pathogenesis of atherosclerosis. In particular, we focus on macrophages and T-lymphocytes, which have been recognized as important determinants for the initiation and development of atherosclerosis. Numerous studies have revealed the role of autophagy in immune cells produced in atherosclerosis. In addition, thyroid hormones induce autophagy in several cells and tissues, such as liver, skeletal muscles, lungs, and brown adipose tissue. Our research group, among others, have reported different targets of thyroid hormone-mediated autophagy, including lipid droplets (lipophagy), mitochondria (mitophagy), and aggregated proteins (aggrephagy). Based on these findings, thyroid hormone-mediated autophagy could serve as a novel therapeutic approach for atherosclerosis. We also consider the limitations of the current murine models for studies on atherosclerosis, especially in relation to low-density lipoprotein-cholesterol driven atherosclerotic plaque.
Collapse
Affiliation(s)
- Kenji Ohba
- Medical Education Center, Hamamatsu University School of Medicine, Shizuoka 431-3192, Japan
| | - Takayuki Iwaki
- Department of Pharmacology, Hamamatsu University School of Medicine, Shizuoka 431-3192, Japan
| |
Collapse
|
26
|
Yu Q, Gong X, Tong Y, Wang M, Duan K, Zhang X, Ge F, Yu X, Li S. Phosphorylation of Jhd2 by the Ras-cAMP-PKA(Tpk2) pathway regulates histone modifications and autophagy. Nat Commun 2022; 13:5675. [PMID: 36167807 PMCID: PMC9515143 DOI: 10.1038/s41467-022-33423-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/16/2022] [Indexed: 11/12/2022] Open
Abstract
Cells need to coordinate gene expression with their metabolic states to maintain cell homeostasis and growth. How cells transduce nutrient availability to appropriate gene expression remains poorly understood. Here we show that glycolysis regulates histone modifications and gene expression by activating protein kinase A (PKA) via the Ras-cyclic AMP pathway. The catalytic subunit of PKA, Tpk2 antagonizes Jhd2-catalyzed H3K4 demethylation by phosphorylating Jhd2 at Ser321 and Ser340 in response to glucose availability. Tpk2-catalyzed Jhd2 phosphorylation impairs its nuclear localization, reduces its binding to chromatin, and promotes its polyubiquitination and degradation by the proteasome. Tpk2-catalyzed Jhd2 phosphorylation also maintains H3K14 acetylation by preventing the binding of histone deacetylase Rpd3 to chromatin. By phosphorylating Jhd2, Tpk2 regulates gene expression, maintains normal chronological life span and promotes autophagy. These results provide a direct connection between metabolism and histone modifications and shed lights on how cells rewire their biological responses to nutrient signals.
Collapse
Affiliation(s)
- Qi Yu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei, 430062, China
| | - Xuanyunjing Gong
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei, 430062, China
| | - Yue Tong
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei, 430062, China
| | - Min Wang
- Key Laboratory of Algal Biology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei, 430072, China
| | - Kai Duan
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei, 430062, China
| | - Xinyu Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei, 430062, China
| | - Feng Ge
- Key Laboratory of Algal Biology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei, 430072, China
| | - Xilan Yu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei, 430062, China.
| | - Shanshan Li
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei, 430062, China.
| |
Collapse
|
27
|
Hong S, Ghandriz R, Siddiqi S, Zhu XY, Saadiq IM, Jordan KL, Tang H, Ali KA, Lerman A, Eirin A, Lerman LO. Effects of Elamipretide on Autophagy in Renal Cells of Pigs with Metabolic Syndrome. Cells 2022; 11:cells11182891. [PMID: 36139466 PMCID: PMC9496989 DOI: 10.3390/cells11182891] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/26/2022] [Accepted: 09/13/2022] [Indexed: 11/24/2022] Open
Abstract
Autophagy eliminates excessive nutrients and maintains homeostasis. Obesity and metabolic syndrome (MetS) dysregulate autophagy, possibly partly due to mitochondria injury and inflammation. Elamipretide (ELAM) improves mitochondrial function. We hypothesized that MetS blunts kidney autophagy, which ELAM would restore. Domestic pigs were fed a control or MetS-inducing diet for 16 weeks. During the 4 last weeks, MetS pigs received subcutaneous injections of ELAM (0.1 mg/kg/day, MetS + ELAM) or vehicle (MetS), and kidneys were then harvested to measure protein expression of autophagy mediators and apoptosis. Systemic and renal venous levels of inflammatory cytokines were measured to calculate renal release. The function of isolated mitochondria was assessed by oxidative stress, energy production, and pro-apoptotic activity. MetS slightly downregulated renal expression of autophagy mediators including p62, ATG5-12, mTOR, and AMPK vs. control. Increased mitochondrial H2O2 production accompanied decreased ATP production, elevated apoptosis, and renal fibrosis. In MetS + ELAM, mito-protection restored autophagic protein expression, improved mitochondrial energetics, and blunted renal cytokine release and fibrosis. In vitro, mitoprotection restored mitochondrial membrane potential and reduced oxidative stress in injured proximal tubular epithelial cells. Our study suggests that swine MetS mildly affects renal autophagy, possibly secondary to mitochondrial damage, and may contribute to kidney structural damage in MetS.
Collapse
Affiliation(s)
- Siting Hong
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Ramyar Ghandriz
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA
| | - Sarosh Siddiqi
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA
| | - Xiang-Yang Zhu
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA
| | - Ishran M. Saadiq
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA
| | - Kyra L. Jordan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA
| | - Hui Tang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA
| | - Khaled A. Ali
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA
| | - Amir Lerman
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN 55905, USA
| | - Alfonso Eirin
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA
| | - Lilach O. Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA
- Correspondence: ; Tel.: +1-507-293-0890
| |
Collapse
|
28
|
Molecular Framework of Mouse Endothelial Cell Dysfunction during Inflammation: A Proteomics Approach. Int J Mol Sci 2022; 23:ijms23158399. [PMID: 35955534 PMCID: PMC9369400 DOI: 10.3390/ijms23158399] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/18/2022] [Accepted: 07/25/2022] [Indexed: 02/01/2023] Open
Abstract
A key aspect of cytokine-induced changes as observed in sepsis is the dysregulated activation of endothelial cells (ECs), initiating a cascade of inflammatory signaling leading to leukocyte adhesion/migration and organ damage. The therapeutic targeting of ECs has been hampered by concerns regarding organ-specific EC heterogeneity and their response to inflammation. Using in vitro and in silico analysis, we present a comprehensive analysis of the proteomic changes in mouse lung, liver and kidney ECs following exposure to a clinically relevant cocktail of proinflammatory cytokines. Mouse lung, liver and kidney ECs were incubated with TNF-α/IL-1β/IFN-γ for 4 or 24 h to model the cytokine-induced changes. Quantitative label-free global proteomics and bioinformatic analysis performed on the ECs provide a molecular framework for the EC response to inflammatory stimuli over time and organ-specific differences. Gene Ontology and PANTHER analysis suggest why some organs are more susceptible to inflammation early on, and show that, as inflammation progresses, some protein expression patterns become more uniform while additional organ-specific proteins are expressed. These findings provide an in-depth understanding of the molecular changes involved in the EC response to inflammation and can support the development of drugs targeting ECs within different organs. Data are available via ProteomeXchange (identifier PXD031804).
Collapse
|
29
|
Brondeel KC, Lakatta AC, Torres GB, Hurley JJ, Kunik IL, Haney KF, Cornett EM, Kaye AD. Physiologic and pharmacologic considerations in morbid obesity and bariatric anesthesia. Saudi J Anaesth 2022; 16:306-313. [PMID: 35898535 PMCID: PMC9311176 DOI: 10.4103/sja.sja_185_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 02/26/2022] [Indexed: 11/15/2022] Open
Abstract
Obesity is a growing worldwide health hazard that is characterized by excess malnutrition. Excess food intake leads to dysregulated energy homeostasis and increased adiposity, activating pro-inflammatory physiologic pathways that can contribute to the chronic inflammatory state associated with many chronic illnesses. Obesity is a preventable illness, but its multifaceted etiology, including genetic, behavioral, and environmental variables, is critical to understanding its epidemiology and pathophysiology. Obesity is a critical predisposing factor for illnesses including type II diabetes, cardiovascular disease, and cancer, with higher morbidity and death. Obesity rates are rising, and so will the need for perioperative anesthesia for subjects with obesity. Obesity epidemiology, biochemistry, and pathophysiology are significant concepts in perioperative anesthesia management for subjects with obesity. To provide optimal intraoperative care for subjects with obesity, preoperative cardiovascular assessment for coronary artery disease and drug monitoring is required. Individuals suffering from obesity have significantly higher oxygen consumption rates and a higher risk of desaturation and surgical complications. Individuals suffering from obesity require specialized perioperative treatment related to higher prevalence of perioperative complications.
Collapse
Affiliation(s)
- Kimberley C. Brondeel
- Department of Medicine, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA
| | - Alexis C. Lakatta
- Department of Medicine, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA
| | - Grant B. Torres
- Department of Medicine, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA
| | - Joshua J. Hurley
- Anesthesiology Resident, LSU Health New Orleans, 433 Bolivar St, New Orleans, LA, USA
| | - Illan L. Kunik
- Department of Medicine, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA
| | - Kaley F. Haney
- Department of Medicine, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA
| | - Elyse M. Cornett
- Department of Anesthesiology, LSU Health Shreveport, 1501 Kings Highway, Shreveport, LA, USA
| | - Alan D. Kaye
- Department of Anesthesiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| |
Collapse
|
30
|
Zhang R, Hao Y, Zhang J. The lncRNA DANCR promotes development of atherosclerosis by regulating the miR-214-5p/COX20 signaling pathway. Cell Mol Biol Lett 2022; 27:15. [PMID: 35177003 PMCID: PMC8903577 DOI: 10.1186/s11658-022-00310-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 01/07/2022] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Although long non-coding RNA differentiation antagonizing non-protein coding RNA (DANCR) has been reported to be involved in atherosclerosis (AS) development, its specific mechanism remains unclear. METHODS DANCR expression levels in blood samples of AS patients and oxidized low-density lipoprotein (ox-LDL) treated vascular smooth muscle cells (VSMCs) and human umbilical vein endothelial cells (HUVECs) were detected by quantitative real-time polymerase chain reaction (qRT-PCR). The small interfering RNA targeting DANCR (si-DANCR) was used to silence DANCR expression. Cell viability was assessed by CCK-8 assay. Cell apoptosis was evaluated by flow cytometry. Levels of inflammatory cytokines, anti-oxidative enzyme superoxide dismutase (SOD) activity, and malonaldehyde (MDA) were detected by specific commercial kits. An animal AS model was established to confirm the role of DANCR/microR-214-5p/COX20 (the chaperone of cytochrome c oxidase subunit II COX2) in AS development. RESULTS DANCR was significantly increased in the blood samples of AS patients and ox-LDL treated VSMCs and HUVECs. DANCR downregulation obviously increased viability and reduced apoptosis of ox-LDL-treated VSMCs and HUVECs. Meanwhile, DANCR downregulation reduced the levels of inflammatory cytokines, including interleukin (IL)-6 (IL-6), IL-1beta (IL-1β), IL-6 and tumor necrosis factor (TNF)-alpha (TNF-α) and MDA while increasing the SOD level in ox-LDL-treated VSMCs and HUVECs. DANCR regulated COX20 expression by acting as a competing endogenous RNA (ceRNA) of miR-214-5p. Rescue experiments demonstrated that miR-214-5p downregulation obviously attenuated si-DANCR-induced protective effects on ox-LDL-caused endothelial injury. CONCLUSIONS Our results revealed that DANCR promoted AS progression by targeting the miR-214-5p/COX20 axis, suggesting that DANCR might be a potential therapeutic target for AS.
Collapse
Affiliation(s)
- Ruolan Zhang
- Department of Cardiology, Harrison International Peace Hospital, No. 180 Renmin Road, Hengshui City, 053000, Hebei Province, People's Republic of China.
| | - Yuming Hao
- Department of Cardiology, Second Affiliated Hospital of Hebei Medical University, Shijiazhuang City, 05000, Hebei Province, People's Republic of China
| | - Jinrong Zhang
- Department of Cardiology, Harrison International Peace Hospital, No. 180 Renmin Road, Hengshui City, 053000, Hebei Province, People's Republic of China
| |
Collapse
|
31
|
Meng LB, Zhang YM, Luo Y, Gong T, Liu DP. Chronic Stress A Potential Suspect Zero of Atherosclerosis: A Systematic Review. Front Cardiovasc Med 2022; 8:738654. [PMID: 34988123 PMCID: PMC8720856 DOI: 10.3389/fcvm.2021.738654] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 11/29/2021] [Indexed: 12/11/2022] Open
Abstract
Atherosclerosis (AS) is a chronic vascular inflammatory disease, in which the lipid accumulation in the intima of the arteries shows yellow atheromatous appearance, which is the pathological basis of many diseases, such as coronary artery disease, peripheral artery disease and cerebrovascular disease. In recent years, it has become the main cause of death in the global aging society, which seriously endangers human health. As a result, research on AS is increasing. Lesions of atherosclerosis contain macrophages, T cells and other cells of the immune response, together with cholesterol that infiltrates from the blood. Recent studies have shown that chronic stress plays an important role in the occurrence and development of AS. From the etiology of disease, social, environmental and genetic factors jointly determine the occurrence of disease. Atherosclerotic cardio-cerebrovascular disease (ASCVD) is often caused by chronic stress (CS). If it cannot be effectively prevented, there will be biological changes in the body environment successively, and then the morphological changes of the corresponding organs. If the patient has a genetic predisposition and a combination of environmental factors triggers the pathogenesis, then chronic stress can eventually lead to AS. Therefore, this paper discusses the influence of chronic stress on AS in the aspects of inflammation, lipid metabolism, endothelial dysfunction, hemodynamics and blood pressure, plaque stability, autophagy, ferroptosis, and cholesterol efflux.
Collapse
Affiliation(s)
- Ling-Bing Meng
- Department of Cardiology, National Center of Gerontology, Institute of Geriatric Medicine, Beijing Hospital, Chinese Academy of Medical Sciences, Beijing, China.,Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yuan-Meng Zhang
- Department of Internal Medicine, The Third Medical Centre of Chinese People's Liberation Army (PLA) General Hospital, The Training Site for Postgraduate of Jinzhou Medical University, Beijing, China
| | - Yue Luo
- Department of Respiratory, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Tao Gong
- Department of Neurology, National Center of Gerontology, National Center of Gerontology, Institute of Geriatric Medicine, Beijing Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - De-Ping Liu
- Department of Cardiology, National Center of Gerontology, Institute of Geriatric Medicine, Beijing Hospital, Chinese Academy of Medical Sciences, Beijing, China.,Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
32
|
Effects of SGLT2 Inhibitors on Atherosclerosis: Lessons from Cardiovascular Clinical Outcomes in Type 2 Diabetic Patients and Basic Researches. J Clin Med 2021; 11:jcm11010137. [PMID: 35011882 PMCID: PMC8745121 DOI: 10.3390/jcm11010137] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/21/2021] [Accepted: 12/24/2021] [Indexed: 12/16/2022] Open
Abstract
Atherosclerosis-caused cardiovascular diseases (CVD) are the leading cause of mortality in type 2 diabetes mellitus (T2DM). Sodium-glucose cotransporter 2 (SGLT2) inhibitors are effective oral drugs for the treatment of T2DM patients. Multiple pre-clinical and clinical studies have indicated that SGLT2 inhibitors not only reduce blood glucose but also confer benefits with regard to body weight, insulin resistance, lipid profiles and blood pressure. Recently, some cardiovascular outcome trials have demonstrated the safety and cardiovascular benefits of SGLT2 inhibitors beyond glycemic control. The SGLT2 inhibitors empagliflozin, canagliflozin, dapagliflozin and ertugliflozin reduce the rates of major adverse cardiovascular events and of hospitalization for heart failure in T2DM patients regardless of CVD. The potential mechanisms of SGLT2 inhibitors on cardioprotection may be involved in improving the function of vascular endothelial cells, suppressing oxidative stress, inhibiting inflammation and regulating autophagy, which further protect from the progression of atherosclerosis. Here, we summarized the pre-clinical and clinical evidence of SGLT2 inhibitors on cardioprotection and discussed the potential molecular mechanisms of SGLT2 inhibitors in preventing the pathogenesis of atherosclerosis and CVD.
Collapse
|
33
|
Zhu W, Wei Z, Han C, Weng X. Nanomaterials as Promising Theranostic Tools in Nanomedicine and Their Applications in Clinical Disease Diagnosis and Treatment. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:3346. [PMID: 34947695 PMCID: PMC8707825 DOI: 10.3390/nano11123346] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/28/2021] [Accepted: 11/30/2021] [Indexed: 12/12/2022]
Abstract
In recent decades, with the rapid development of nanotechnology, nanomaterials have been widely used in the medical field, showing great potential due to their unique physical and chemical properties including minimal size and functionalized surface characteristics. Nanomaterials such as metal nanoparticles and polymeric nanoparticles have been extensively studied in the diagnosis and treatment of diseases that seriously threaten human life and health, and are regarded to significantly improve the disadvantages of traditional diagnosis and treatment platforms, such as poor effectiveness, low sensitivity, weak security and low economy. In this review, we report and discuss the development and application of nanomaterials in the diagnosis and treatment of diseases based mainly on published research in the last five years. We first briefly introduce the improvement of several nanomaterials in imaging diagnosis and genomic sequencing. We then focus on the application of nanomaterials in the treatment of diseases, and select three diseases that people are most concerned about and that do the most harm: tumor, COVID-19 and cardiovascular diseases. First, we introduce the characteristics of nanoparticles according to the excellent effect of nanoparticles as delivery carriers of anti-tumor drugs. We then review the application of various nanoparticles in tumor therapy according to the classification of nanoparticles, and emphasize the importance of functionalization of nanomaterials. Second, COVID-19 has been the hottest issue in the health field in the past two years, and nanomaterials have also appeared in the relevant treatment. We enumerate the application of nanomaterials in various stages of viral pathogenesis according to the molecular mechanism of the complete pathway of viral infection, pathogenesis and transmission, and predict the application prospect of nanomaterials in the treatment of COVID-19. Third, aiming at the most important causes of human death, we focus on atherosclerosis, aneurysms and myocardial infarction, three of the most common and most harmful cardiovascular diseases, and prove that nanomaterials could be involved in a variety of therapeutic approaches and significantly improve the therapeutic effect in cardiovascular diseases. Therefore, we believe nanotechnology will become more widely involved in the diagnosis and treatment of diseases in the future, potentially helping to overcome bottlenecks under existing medical methods.
Collapse
Affiliation(s)
- Wei Zhu
- Department of Orthopaedics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China; (W.Z.); (Z.W.); (C.H.)
| | - Zhanqi Wei
- Department of Orthopaedics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China; (W.Z.); (Z.W.); (C.H.)
- School of Medicine, Tsinghua University, Haidian District, Beijing 100084, China
| | - Chang Han
- Department of Orthopaedics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China; (W.Z.); (Z.W.); (C.H.)
| | - Xisheng Weng
- Department of Orthopaedics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China; (W.Z.); (Z.W.); (C.H.)
- Department of State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
34
|
Kountouras J, Papaefthymiou A, Polyzos SA, Deretzi G, Vardaka E, Soteriades ES, Tzitiridou-Chatzopoulou M, Gkolfakis P, Karafyllidou K, Doulberis M. Impact of Helicobacter pylori-Related Metabolic Syndrome Parameters on Arterial Hypertension. Microorganisms 2021; 9:microorganisms9112351. [PMID: 34835476 PMCID: PMC8618184 DOI: 10.3390/microorganisms9112351] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/28/2021] [Accepted: 11/10/2021] [Indexed: 12/12/2022] Open
Abstract
Arterial hypertension is a risk factor for several pathologies, mainly including cardio-cerebrovascular diseases, which rank as leading causes of morbidity and mortality worldwide. Arterial hypertension also constitutes a fundamental component of the metabolic syndrome. Helicobacter pylori infection is one of the most common types of chronic infection globally and displays a plethora of both gastric and extragastric effects. Among other entities, Helicobacter pylori has been implicated in the pathogenesis of the metabolic syndrome. Within this review, we illustrate the current state-of-the-art evidence, which may link several components of the Helicobacter pylori-related metabolic syndrome, including non-alcoholic fatty liver disease and arterial hypertension. In particular, current knowledge of how Helicobacter pylori exerts its virulence through dietary, inflammatory and metabolic pathways will be discussed. Although there is still no causative link between these entities, the emerging evidence from both basic and clinical research supports the proposal that several components of the Helicobacter pylori infection-related metabolic syndrome present an important risk factor in the development of arterial hypertension. The triad of Helicobacter pylori infection, the metabolic syndrome, and hypertension represents a crucial worldwide health problem on a pandemic scale with high morbidity and mortality, like COVID-19, thereby requiring awareness and appropriate management on a global scale.
Collapse
Affiliation(s)
- Jannis Kountouras
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (A.P.); (M.T.-C.); (M.D.)
- Correspondence:
| | - Apostolis Papaefthymiou
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (A.P.); (M.T.-C.); (M.D.)
- Department of Gastroenterology, University Hospital of Larisa, 41110 Larisa, Greece
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Stergios A. Polyzos
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Georgia Deretzi
- Multiple Sclerosis Unit, Department of Neurology, Papageorgiou General Hospital, 56403 Thessaloniki, Greece;
| | - Elisabeth Vardaka
- Department of Nutritional Sciences and Dietetics, School of Health Sciences, International Hellenic University, 57400 Thessaloniki, Greece;
| | - Elpidoforos S. Soteriades
- Healthcare Management Program, School of Economics and Management, Open University of Cyprus, Nicosia 2252, Cyprus;
- Department of Environmental Health, Environmental and Occupational Medicine and Epidemiology (EOME), Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Maria Tzitiridou-Chatzopoulou
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (A.P.); (M.T.-C.); (M.D.)
- School of Healthcare Sciences, Midwifery Department, University of West Macedonia, Koila, 50100 Kozani, Greece
| | - Paraskevas Gkolfakis
- Department of Gastroenterology, Hepatopancreatology and Digestive Oncology, Erasme University Hospital, 1070 Brussels, Belgium;
- Department of Medical Oncology, Institut Jules Bordet, 1000 Brussels, Belgium
| | - Kyriaki Karafyllidou
- Department of Pediatrics, University Children’s Hospital of Zurich, 8032 Zurich, Switzerland;
| | - Michael Doulberis
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (A.P.); (M.T.-C.); (M.D.)
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
- Division of Gastroenterology and Hepatology, Medical University Department, Kantonsspital Aarau, 5001 Aarau, Switzerland
| |
Collapse
|
35
|
Zhang N, Xue M, Sun T, Yang J, Pei Z, Qin K. Fucoidan as an Autophagy Regulator: Mechanisms and Therapeutic Potentials for Cancer and Other Diseases. Nutr Cancer 2021; 74:1568-1579. [PMID: 34477470 DOI: 10.1080/01635581.2021.1973045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Fucoidan, a natural polysaccharide with a variety of classical bioactivities mainly sourced from brown algae, has been extensively studied owing to its favorable pharmacological effects, including anti-inflammatory, anti-tumor, anticoagulant and liver protection. Recently it has been found to play a regulatory role in the processes of autophagy. Autophagy is an important cellular process that effectively protects cells and organisms from stimulating factors such as nutrient deficiency, low cellular ATP levels, metabolic stress, growth factor deprivation and hypoxic conditions. In recent years, many studies have shown that fucoidan can treat human diseases by regulating autophagy process though cell signaling pathways. In this review, we summarize the latest progress in the discovery of natural autophagy regulator of fucoidan for the therapeutic application in cardiac diseases, cancers and liver diseases, aiming to provide the new pharmacological application that fucoidan may treat human diseases by regulating autophagy. Furthermore, we look forward to seeing more diseases that would be treated by autophagy modulator of fucoidan and the discovery of more elaborate autophagy regulation mechanism.
Collapse
Affiliation(s)
- Nan Zhang
- Basic Medical College, Qingdao University of Medicine, Qingdao, PR China
| | - Meilan Xue
- Basic Medical College, Qingdao University of Medicine, Qingdao, PR China
| | - Ting Sun
- Basic Medical College, Qingdao University of Medicine, Qingdao, PR China
| | - Jia Yang
- Basic Medical College, Qingdao University of Medicine, Qingdao, PR China
| | - Zhongqian Pei
- Basic Medical College, Qingdao University of Medicine, Qingdao, PR China
| | - Kunpeng Qin
- Basic Medical College, Qingdao University of Medicine, Qingdao, PR China
| |
Collapse
|
36
|
Ni D, Mo Z, Yi G. Recent insights into atherosclerotic plaque cell autophagy. Exp Biol Med (Maywood) 2021; 246:2553-2558. [PMID: 34407677 DOI: 10.1177/15353702211038894] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Cardiovascular and cerebrovascular diseases, such as coronary heart disease and stroke, caused by atherosclerosis have become the "number one killer", seriously endangering human health in developing and developed countries. Atherosclerosis mainly occurs in large and medium-sized arteries and involves intimal thickening, accumulation of foam cells, and formation of atheromatous plaques. Autophagy is a cellular catabolic process that has evolved to defend cells from the turnover of intracellular molecules. Autophagy is thought to play an important role in the development of plaques. This review focuses on studies on autophagy in cells involved in the formation of atherosclerotic plaques, such as monocytes, macrophages, endothelial cells, dendritic cells, and vascular smooth muscle cells, indicating that autophagy plays an important role in plaque development. We mainly discuss the roles of autophagy in these cells in maintaining the stability of atherosclerotic plaques, providing a reference for the next steps to unravel the mechanisms of atherogenesis.
Collapse
Affiliation(s)
- Dan Ni
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang 421001, China
| | - Zhongcheng Mo
- Guangxi Key Laboratory of Diabetic Systems Medicine, Institute of Basic Medical Sciences, Guilin Medical University, Guilin 541000, China
| | - Guanghui Yi
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang 421001, China
| |
Collapse
|
37
|
He XY, Ou CL. Clinical significance of serum miR-129-5p in patients with diabetes mellitus presenting macrovascular complications. World J Diabetes 2021; 12:1282-1291. [PMID: 34512893 PMCID: PMC8394230 DOI: 10.4239/wjd.v12.i8.1282] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/24/2021] [Accepted: 07/06/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Diabetic macrovascular complications (DMCs) are the most common complications encountered during the course of diabetes mellitus (DM) with extremely high mortality rates. Therefore, there is an urgent need to identify specific and sensitive biomarkers for the early diagnosis of DMCs. AIM To investigate the expression and significance of serum miR-129-5p in patients with DM and macrovascular complications. METHODS Serum samples were collected from 36 healthy controls, 58 patients with DM presenting no macrovascular complications, and 62 patients with DMCs. The expression of miR-129-5p was detected using quantitative real-time polymerase chain reaction. Pearson's correlation assay was performed to analyze the correlation between serum miR-129-5p levels and clinical indicators. Receiver operator characteristic (ROC) analysis was conducted to analyze the diagnostic value of serum miR-129-5p in patients with DM or DMCs. RESULTS There was a 4.378-fold and 7.369-fold increase in serum miR-129-5p expression in the DM (5.346 ± 0.405) and DMCs (8.998 ± 0.631) groups, respectively (P < 0.001), compared with the control group (1.221±0.090). In addition, the expression of serum miR-129-5p in patients with DMCs was higher than that in patients with DM, revealing a 1.683-fold increase (P < 0.001). Additionally, serum miR-129-5p expression significantly correlated with smoking history, disease duration, and glycated hemoglobin (HbA1c) in patients with DMCs (P < 0.001). The area under the ROC curve (AUC) of miR-129-5p as a serum marker was 0.964 (95% confidence interval [CI]: 0.930-0.997, P < 0.001) in distinguishing between patients with DM and healthy controls, whereas the AUC of miR-129-5p as a serum marker was 0.979 (95%CI: 0.959-0.999, P < 0.001) in distinguishing between patients with DMCs and healthy controls. CONCLUSION Elevated serum miR-129-5p expression levels correlate with the development of DMCs and can be utilized as a novel early diagnostic biomarker for DM combined with macrovascular complications.
Collapse
Affiliation(s)
- Xiao-Yun He
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- Department of Endocrinology, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, China
| | - Chun-Lin Ou
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| |
Collapse
|
38
|
Kao TW, Huang CC. Recent Progress in Metabolic Syndrome Research and Therapeutics. Int J Mol Sci 2021; 22:6862. [PMID: 34202257 PMCID: PMC8269131 DOI: 10.3390/ijms22136862] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 06/22/2021] [Accepted: 06/22/2021] [Indexed: 02/07/2023] Open
Abstract
Metabolic syndrome (MetS) is a well-defined yet difficult-to-manage disease entity. Both the precipitous rise in its incidence due to contemporary lifestyles and the growing heterogeneity among affected populations present unprecedented challenges. Moreover, the predisposed risk for developing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in populations with MetS, and the viral impacts on host metabolic parameters, underscores the need to investigate this mechanism thoroughly. Recent investigations of metabolomics and proteomics have revealed not only differentially expressed substances in MetS, but also the consequences of diet consumption and physical activity on energy metabolism. These variations in metabolites, as well as protein products, also influence a wide spectrum of host characteristics, from cellular behavior to phenotype. Research on the dysregulation of gut microbiota and the resultant inflammatory status has also contributed to our understanding of the underlying pathogenic mechanisms. As for state-of-the-art therapies, advancing depictions of the bio-molecular landscape of MetS have emerged and now play a key role in individualized precision medicine. Fecal microbiota transplantation, aiming to restore the host's homeostasis, and targeting of the bile acid signaling pathway are two approaches to combatting MetS. Comprehensive molecular inquiries about MetS by omics measures are mandatory to facilitate the development of novel therapeutic modalities.
Collapse
Affiliation(s)
- Ting-Wei Kao
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 100, Taiwan;
| | - Chin-Chou Huang
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Institute of Pharmacology, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| |
Collapse
|