1
|
Kamel AA, Nassar AY, Meligy FY, Omar YA, Nassar GAY, Ezzat GM. Acetylated oligopeptide and N-acetylcysteine protect against iron overload-induced dentate gyrus hippocampal degeneration through upregulation of Nestin and Nrf2/HO-1 and downregulation of MMP-9/TIMP-1 and GFAP. Cell Biochem Funct 2024; 42:e3958. [PMID: 38396357 DOI: 10.1002/cbf.3958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 01/29/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024]
Abstract
Iron accumulation in the brain causes oxidative stress, blood-brain barrier (BBB) breakdown, and neurodegeneration. We examined the preventive effects of acetylated oligopeptides (AOP) from whey protein on iron-induced hippocampal damage compared to N-acetyl cysteine (NAC). This 5-week study used 40 male albino rats. At the start, all rats received 150 mg/kg/day of oral NAC for a week. The 40 animals were then randomly divided into four groups: Group I (control) received a normal diet; Group II (iron overload) received 60 mg/kg/day intraperitoneal iron dextran 5 days a week for 4 weeks; Group III (NAC group) received 150 mg/kg/day NAC and iron dextran; and Group IV (AOP group) received 150 mg/kg/day AOP and iron dextran. Enzyme-linked immunosorbent assay, spectrophotometry, and qRT-PCR were used to measure MMP-9, tissue inhibitor metalloproteinase-1 (TIMP-1), MDA, reduced glutathione (GSH) levels, and nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) gene expression. Histopathological and immunohistochemical detection of nestin, claudin, caspase, and GFAP was also done. MMP-9, TIMP-1, MDA, caspase, and GFAP rose in the iron overload group, while GSH, Nrf2, HO-1, nestin, and claudin decreased. The NAC and AOP administrations improved iron overload-induced biochemical and histological alterations. We found that AOP and NAC can protect the brain hippocampus from iron overload, improve BBB disruption, and provide neuroprotection with mostly no significant difference from healthy controls.
Collapse
Affiliation(s)
- Amira A Kamel
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Ahmed Y Nassar
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Fatma Y Meligy
- Department of Restorative Dentistry and Basic Medical Sciences, Faculty of Dentistry, University of Petra, Amman, Jordan
- Department of Histology and Cell Biology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Yomna A Omar
- Department of Biochemistry, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| | - Gamal A Y Nassar
- Metabolic and Genetic Disorders Unit, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Ghada M Ezzat
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
2
|
Qu Y, Zhang N, Zhao Y. Resveratrol Inhibits Abdominal Aortic Aneurysm Progression by Reducing Extracellular Matrix Degradation, Apoptosis, Autophagy, and Inflammation of Vascular Smooth Muscle Cells via Upregulation of HMOX1. J Endovasc Ther 2023:15266028231202727. [PMID: 37789605 DOI: 10.1177/15266028231202727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
OBJECTIVE This study aimed to explore the therapeutic effect of resveratrol (RES) against abdominal aortic aneurysm (AAA) and the role of HMOX1 underlying this effect. METHODS Vascular smooth muscle cells (VSMCs) were induced by angiotensin II (Ang II) to construct the microenvironment of AAA. HMOX1 expression was downregulated by the short hairpin ribonucleic acid (RNA) specific to HMOX1 in RES-pretreated VSMCs. The levels of matrix metalloproteinase (MMP)-2, MMP-9, and elastin were measured by quantitative reverse transcription polymerase chain reaction (qRT-PCR) and western blot. Apoptosis rate was detected. The levels of apoptosis-related proteins (caspase-3 and Bax/Bcl-2), inflammatory cytokines (interleukin [IL]-6, tumor necrosis factor [TNF]-α, and IL-1β), and autophagy-related proteins (Beclin 1, light chain 3 [LC3] II/I, and p62) were detected by western blot. The secretion of inflammatory factors in cell supernatant was detected by enzyme-linked immunosorbent assay (ELISA). The number of autophagic vesicles in VSMCs was observed and analyzed by transmission electron microscopy. A rat model of pancreatic elastase-induced AAA was established to verify the effect and action mechanism of RES. RESULTS Stimulation of Ang II increased the messenger RNA (mRNA) and protein levels of MMP-2 and MMP-9, decreased elastin expression, and enhanced apoptosis, secretion of inflammatory factors, and autophagy in VSMCs, whereas RES pretreatment ameliorated Ang II-induced VSMC dysfunction. In addition, HMOX1 mRNA and heme oxygenase-1 (HO-1) protein levels were significantly increased in VSMCs pretreated with RES compared with Ang II treatment alone. Silencing of HMOX1 abolished the effects of RES on VSMC dysfunction. Consistently, RES suppressed the development of AAA in rats by increasing the expression of HMOX1. CONCLUSION Resveratrol protects against AAA by inhibiting extracellular matrix degradation, apoptosis, autophagy, and inflammation of VSMCs via HMOX1 upregulation. CLINICAL IMPACT Our study found that angiotensin II (Ang II) stimulated increased the levels of MMP-2 and MMP-9 in vascular smooth muscle cells (VSMCs), decreased elastin expression, and promoted apoptosis, autophagy occurrence, and secretion of inflammatory factors, while resveratrol (RES) pretreatment improved this effect. In addition, downregulation of HMOX1 expression eliminated the effect of RES on the function of VSMCs. Our study elucidates that RES improves AAA progression through HMOX1 at both cellular and animal levels. This work can help doctors better understand the pathological mechanism of the occurrence and development of AAA, and provide a theoretical basis for clinicians to find better treatment options.
Collapse
Affiliation(s)
- Yunfei Qu
- Department of Vascular Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Ning Zhang
- General Practice, Chongqing University Three Gorges Hospital, Chongqing, P.R. China
| | - Yu Zhao
- Department of Vascular Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| |
Collapse
|
3
|
Abaszadeh S, Ahmadipour B, Pirany N, Hassanpour H, Khajali F. Effect of dietary inclusion of atorvastatin, garlic, and dill on growth performance, antioxidant defense, gut, and cardio-pulmonary function, and lipogenesis in broiler chickens. Trop Anim Health Prod 2023; 55:216. [PMID: 37212936 DOI: 10.1007/s11250-023-03639-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 05/16/2023] [Indexed: 05/23/2023]
Abstract
In the present study, we evaluated the antihyperlipidemic and antioxidant effects of garlic and dill in comparison with atorvastatin to combat lipogenesis in broiler chickens. A total of 400 1-day-old chicks (Ross 308 strain) were randomly distributed into four experimental diets. Dietary treatments included a control diet, the control diet plus atorvastatin at 20 mg/kg, the control diet plus garlic dry powder (GDP) at 7.5 g/kg, and the control diet plus dill dry powder (DDP) at 7.5 g/kg. Chicks were maintained on experimental diets for 42 days under the recommended environmental conditions set out by the strain management manual. The results showed that weight gain, feed conversion ratio (FCR), and duodenal, jejunal, and ileal dimensions of villi (height, width, and the surface absorptive area) were improved by in-feed atorvastatin, GDP, or DDP when compared to the control (P < 0.05). The inclusion of atorvastatin or phytobiotic products increased circulatory levels of nitric oxide (NO) but decreased circulatory levels of malondialdehyde (MDA), triacylglycerol (TAG), and low-density lipoproteins cholesterol (LDL), with concomitant reductions in the T, R, and S waves amplitudes in the Lead 2 electrocardiogram (ECG) (P < 0.05). Dietary supplements caused an up-regulation of inducible nitric oxide synthase (iNOS), superoxide dismutase 1 (SOD1), and glutathione peroxidase (GPX) but reduced the expression of key hepatic lipogenic enzymes (fatty acid synthase (FAS) and hydroxy-methylglutaryl-CoA reductase (HMGCR) (P < 0.05). In conclusion, feed supplementation with atorvastatin, GDP, or DDP suppressed lipogenesis, enhanced antioxidant response, and improved gut and cardio-pulmonary function in broiler chicks subjected to hypobaric hypoxia.
Collapse
Affiliation(s)
- Samira Abaszadeh
- Department of Animal Science, Faculty of Agriculture, Shahrekord University, Shahrekord, 88186 34141, Iran
| | - Behnam Ahmadipour
- Department of Animal Science, Faculty of Agriculture, Shahrekord University, Shahrekord, 88186 34141, Iran.
| | - Nasrollah Pirany
- Department of Animal Science, Faculty of Agriculture, Shahrekord University, Shahrekord, 88186 34141, Iran
| | - Hossein Hassanpour
- Department of Basic Sciences, Faculty of Veterinary Medicine, Shahrekord University, Shahrekord, Iran
| | - Fariborz Khajali
- Department of Animal Science, Faculty of Agriculture, Shahrekord University, Shahrekord, 88186 34141, Iran
| |
Collapse
|
4
|
Li D, Shen L, Zhang D, Wang X, Wang Q, Qin W, Gao Y, Li X. Ammonia-induced oxidative stress triggered proinflammatory response and apoptosis in pig lungs. J Environ Sci (China) 2023; 126:683-696. [PMID: 36503793 DOI: 10.1016/j.jes.2022.05.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 05/06/2022] [Accepted: 05/07/2022] [Indexed: 06/17/2023]
Abstract
Ammonia, a common toxic gas, is not only one of the main causes of haze, but also can enter respiratory tract and directly affect the health of humans and animals. Pig was used as an animal model for exploring the molecular mechanism and dose effect of ammonia toxicity to lung. In this study, the apoptosis of type II alveolar epithelial cells was observed in high ammonia exposure group using transmission electron microscopy. Gene and protein expression analysis using transcriptome sequencing and western blot showed that low ammonia exposure induced T-cell-involved proinflammatory response, but high ammonia exposure repressed the expression of DNA repair-related genes and affected ion transport. Moreover, high ammonia exposure significantly increased 8-hydroxy-2-deoxyguanosine (8-OHdG) level, meaning DNA oxidative damage occurred. In addition, both low and high ammonia exposure caused oxidative stress in pig lungs. Integrated analysis of transcriptome and metabolome revealed that the up-regulation of LDHB and ND2 took part in high ammonia exposure-affected pyruvate metabolism and oxidative phosphorylation progress, respectively. Inclusion, oxidative stress mediated ammonia-induced proinflammatory response and apoptosis of porcine lungs. These findings may provide new insights for understanding the ammonia toxicity to workers in livestock farms and chemical fertilizer plants.
Collapse
Affiliation(s)
- Daojie Li
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Long Shen
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Di Zhang
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiaotong Wang
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Qiankun Wang
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Wenhao Qin
- College of Science, Huazhong Agricultural University, Wuhan 430070, China
| | - Yun Gao
- College of Engineering, The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiaoping Li
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
5
|
Wu W, Hendrix A, Nair S, Cui T. Nrf2-Mediated Dichotomy in the Vascular System: Mechanistic and Therapeutic Perspective. Cells 2022; 11:cells11193042. [PMID: 36231004 PMCID: PMC9563590 DOI: 10.3390/cells11193042] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/25/2022] [Accepted: 09/27/2022] [Indexed: 12/14/2022] Open
Abstract
Nuclear factor-erythroid 2-related factor 2 (Nrf2), a transcription factor, controls the expression of more than 1000 genes that can be clustered into different categories with distinct functions ranging from redox balance and metabolism to protein quality control in the cell. The biological consequence of Nrf2 activation can be either protective or detrimental in a context-dependent manner. In the cardiovascular system, most studies have focused on the protective properties of Nrf2, mainly as a key transcription factor of antioxidant defense. However, emerging evidence revealed an unexpected role of Nrf2 in mediating cardiovascular maladaptive remodeling and dysfunction in certain disease settings. Herein we review the role of Nrf2 in cardiovascular diseases with a focus on vascular disease. We discuss the negative effect of Nrf2 on the vasculature as well as the potential underlying mechanisms. We also discuss the clinical relevance of targeting Nrf2 pathways for the treatment of cardiovascular and other diseases.
Collapse
Affiliation(s)
- Weiwei Wu
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao 266071, China
| | - Andrew Hendrix
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - Sharad Nair
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29209, USA
- Columbia VA Health System, Wm. Jennings Bryan Dorn VA Medical Center, Columbia, SC 29209, USA
| | - Taixing Cui
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29209, USA
- Columbia VA Health System, Wm. Jennings Bryan Dorn VA Medical Center, Columbia, SC 29209, USA
- Correspondence: ; Tel.: +1-803-216-3804
| |
Collapse
|
6
|
Hofmann A, Hamann B, Klimova A, Müglich M, Wolk S, Busch A, Frank F, Sabarstinski P, Kapalla M, Nees JA, Brunssen C, Poitz DM, Morawietz H, Reeps C. Pharmacotherapies and Aortic Heme Oxygenase-1 Expression in Patients with Abdominal Aortic Aneurysm. Antioxidants (Basel) 2022; 11:antiox11091753. [PMID: 36139827 PMCID: PMC9495607 DOI: 10.3390/antiox11091753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/29/2022] [Accepted: 08/31/2022] [Indexed: 11/25/2022] Open
Abstract
Background: Treatment of cardiovascular risk factors slows the progression of small abdominal aortic aneurysms (AAA). Heme oxygenase-1 (HO-1) is a stress- and hemin-induced enzyme providing cytoprotection against oxidative stress when overexpressed. However, nothing is known about the effects of cardiometabolic standard therapies on HO-1 expression in aortic walls in patients with end-stage AAA. Methods: The effects of statins, angiotensin-converting enzyme (ACE) inhibitors, angiotensin II receptor blockers (ARBs), calcium channel blockers (CCBs), beta-blockers, diuretics, acetylsalicylic acid (ASA), and therapeutic anticoagulation on HO-1 mRNA and protein expressions were analyzed in AAA patients using multivariate logistic regression analysis and comparison of monotherapy. Results: Analysis of monotherapy revealed that HO-1 mRNA and protein expressions were higher in patients on diuretics and lower in patients on statin therapy. Tests on combinations of antihypertensive medications demonstrated that ACE inhibitors and diuretics, ARBs and diuretics, and beta-blockers and diuretics were associated with increase in HO-1 mRNA expression. ASA and therapeutic anticoagulation were not linked to HO-1 expression. Conclusion: Diuretics showed the strongest association with HO-1 expression, persisting even in combination with other antihypertensive medications. Hence, changes in aortic HO-1 expression in response to different medical therapies and their effects on vessel wall degeneration should be analyzed in future studies.
Collapse
Affiliation(s)
- Anja Hofmann
- Division of Vascular and Endovascular Surgery, Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine, Technische Universität Dresden, Fetscherstr. 74, D-01307 Dresden, Germany
- Correspondence: ; Tel.: +49-351-458-16607
| | - Bianca Hamann
- Division of Vascular and Endovascular Surgery, Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine, Technische Universität Dresden, Fetscherstr. 74, D-01307 Dresden, Germany
| | - Anna Klimova
- National Center for Tumor Diseases, Partner Site Dresden, Institute for Medical Informatics and Biometry, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, D-01307 Dresden, Germany
| | - Margarete Müglich
- Division of Vascular and Endovascular Surgery, Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine, Technische Universität Dresden, Fetscherstr. 74, D-01307 Dresden, Germany
| | - Steffen Wolk
- Division of Vascular and Endovascular Surgery, Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine, Technische Universität Dresden, Fetscherstr. 74, D-01307 Dresden, Germany
| | - Albert Busch
- Division of Vascular and Endovascular Surgery, Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine, Technische Universität Dresden, Fetscherstr. 74, D-01307 Dresden, Germany
| | - Frieda Frank
- Division of Vascular and Endovascular Surgery, Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine, Technische Universität Dresden, Fetscherstr. 74, D-01307 Dresden, Germany
| | - Pamela Sabarstinski
- Division of Vascular and Endovascular Surgery, Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine, Technische Universität Dresden, Fetscherstr. 74, D-01307 Dresden, Germany
| | - Marvin Kapalla
- Division of Vascular and Endovascular Surgery, Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine, Technische Universität Dresden, Fetscherstr. 74, D-01307 Dresden, Germany
| | - Josef Albin Nees
- Clinic for Internal Medicine, Asklepios-ASB Klinik Radeberg, D-01454 Radeberg, Germany
| | - Coy Brunssen
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, University Hospital Carl Gustav Carus Dresden, Technische Universität Dresden, D-01307 Dresden, Germany
| | - David M. Poitz
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, D-01307 Dresden, Germany
| | - Henning Morawietz
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, University Hospital Carl Gustav Carus Dresden, Technische Universität Dresden, D-01307 Dresden, Germany
| | - Christian Reeps
- Division of Vascular and Endovascular Surgery, Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine, Technische Universität Dresden, Fetscherstr. 74, D-01307 Dresden, Germany
| |
Collapse
|
7
|
Wu R, Zuo W, Xu X, Bi L, Zhang C, Chen H, Liu H. MCU That Is Transcriptionally Regulated by Nrf2 Augments Malignant Biological Behaviors in Oral Squamous Cell Carcinoma Cells. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6650791. [PMID: 34189138 PMCID: PMC8195654 DOI: 10.1155/2021/6650791] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 05/10/2021] [Indexed: 01/10/2023]
Abstract
OBJECTIVE To clarify the role and molecular mechanism of mitochondrial calcium uniporter (MCU) in the malignant biological behaviors of oral squamous cell carcinoma (OSCC) cells through clinical and cellular experiments. METHODS Immunohistochemistry and qRT-PCR techniques were used to observe the expression of MCU, nuclear factor erythroid 2-related factor 2 (Nrf2), mitochondrial calcium uptake 1 (MICU1), and MICU2 in OSCC and normal tissues. After treatment with si-MCU, spermine, and/or sh-Nrf2, malignant biological behaviors of OSCC cells including proliferation, migration, and apoptosis were detected by clone formation, migration, and mitochondrial membrane potential (MMP) assays. Furthermore, MCU, MICU1, MICU2, Nrf2, and other proteins related to malignant biological behaviors were examined using western blot, immunohistochemistry, and immunofluorescence assays. RESULTS MCU, Nrf2, and MICU1 were strongly expressed in OSCC as compared to normal tissues, while MICU2 was relatively weakly expressed in OSCC tissues. Knockdown of MCU distinctly weakened proliferation and migration and lowered MMP level in CAL 27 cells. Conversely, its activation reinforced migrated capacity and increased MMP level in CAL 27 cells, which was reversed after cotransfection with sh-Nrf2. After treatment with si-MCU or spermine, Nrf2 expression was not affected in CAL 27 cells. However, MCU expression was distinctly suppressed in CAL 27 cells transfected with sh-Nrf2. Furthermore, knockdown of Nrf2 significantly reversed the increase in expression of MICU1 and MICU2 induced by MCU activation in CAL 27 cells. CONCLUSION MCU, as a novel oncogene of OSCC, augments malignant biological behaviors of OSCC cells, which could be transcriptionally regulated by Nrf2.
Collapse
Affiliation(s)
- Ran Wu
- Department of Stomatology, North China University of Science and Technology Affiliated Hospital, Tangshan, 063000 Hebei, China
| | - Weiwen Zuo
- Department of Stomatology, Tangshan Vocational and Technical College, Tangshan, 063000 Hebei, China
| | - Xiaoliang Xu
- Department of Stomatology, The Second Hospital of Tangshan, Tangshan, 063000 Hebei, China
| | - Lei Bi
- Department of Stomatology, North China University of Science and Technology Affiliated Hospital, Tangshan, 063000 Hebei, China
| | - Chunguang Zhang
- Department of Stomatology, North China University of Science and Technology Affiliated Hospital, Tangshan, 063000 Hebei, China
| | - Hui Chen
- Department of Stomatology, North China University of Science and Technology Affiliated Hospital, Tangshan, 063000 Hebei, China
| | - Hui Liu
- Department of Stomatology, North China University of Science and Technology Affiliated Hospital, Tangshan, 063000 Hebei, China
| |
Collapse
|
8
|
Kopacz A, Klóska D, Werner E, Hajduk K, Grochot-Przęczek A, Józkowicz A, Piechota-Polańczyk A. A Dual Role of Heme Oxygenase-1 in Angiotensin II-Induced Abdominal Aortic Aneurysm in the Normolipidemic Mice. Cells 2021; 10:cells10010163. [PMID: 33467682 PMCID: PMC7830394 DOI: 10.3390/cells10010163] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/09/2021] [Accepted: 01/12/2021] [Indexed: 11/16/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) bears a high risk of rupture and sudden death of the patient. The pathogenic mechanisms of AAA remain elusive, and surgical intervention represents the only treatment option. Heme oxygenase-1 (HO-1), a heme degrading enzyme, is induced in AAA, both in mice and humans. HO-1 was reported to mitigate AAA development in an angiotensin II (AngII)-induced model of AAA in hyperlipidemic ApoE-/- mice. Since the role of hyperlipidaemia in the pathogenesis of AAA remains controversial, we aimed to evaluate the significance of HO-1 in the development and progression of AAA in normolipidemic animals. The experiments were performed in HO-1-deficient mice and their wild-type counterparts. We demonstrated in non-hypercholesterolemic mice that the high-dose of AngII leads to the efficient formation of AAA, which is attenuated by HO-1 deficiency. Yet, if formed, they are significantly more prone to rupture upon HO-1 shortage. Differential susceptibility to AAA formation does not rely on enhanced inflammatory response or oxidative stress. AAA-resistant mice are characterized by an increase in regulators of aortic remodeling and angiotensin receptor-2 expression, significant medial thickening, and delayed blood pressure elevation in response to AngII. To conclude, we unveil a dual role of HO-1 deficiency in AAA in normolipidemic mice, where it protects against AAA development, but exacerbates the state of formed AAA.
Collapse
Affiliation(s)
- Aleksandra Kopacz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-392 Krakow, Poland; (A.K.); (D.K.); (E.W.); (K.H.); (A.G.-P.); (A.J.)
| | - Damian Klóska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-392 Krakow, Poland; (A.K.); (D.K.); (E.W.); (K.H.); (A.G.-P.); (A.J.)
| | - Ewa Werner
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-392 Krakow, Poland; (A.K.); (D.K.); (E.W.); (K.H.); (A.G.-P.); (A.J.)
- Department of Animal Reproduction, Anatomy and Genomic, Faculty of Animal Science, University of Agriculture, 30-059 Krakow, Poland
| | - Karolina Hajduk
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-392 Krakow, Poland; (A.K.); (D.K.); (E.W.); (K.H.); (A.G.-P.); (A.J.)
| | - Anna Grochot-Przęczek
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-392 Krakow, Poland; (A.K.); (D.K.); (E.W.); (K.H.); (A.G.-P.); (A.J.)
| | - Alicja Józkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-392 Krakow, Poland; (A.K.); (D.K.); (E.W.); (K.H.); (A.G.-P.); (A.J.)
| | - Aleksandra Piechota-Polańczyk
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-392 Krakow, Poland; (A.K.); (D.K.); (E.W.); (K.H.); (A.G.-P.); (A.J.)
- Correspondence:
| |
Collapse
|
9
|
Freitas F, Tibiriçá E, Singh M, Fraser PA, Mann GE. Redox Regulation of Microvascular Permeability: IL-1β Potentiation of Bradykinin-Induced Permeability Is Prevented by Simvastatin. Antioxidants (Basel) 2020; 9:antiox9121269. [PMID: 33327440 PMCID: PMC7764912 DOI: 10.3390/antiox9121269] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/08/2020] [Accepted: 12/09/2020] [Indexed: 12/27/2022] Open
Abstract
Antioxidant effects of statins have been implicated in the reduction in microvascular permeability and edema formation in experimental and clinical studies. Bradykinin (Bk)-induced increases in microvascular permeability are potentiated by IL-1β; however, no studies have examined the protection afforded by statins against microvascular hyperpermeability. We investigated the effects of simvastatin pretreatment on albumin–fluorescein isothiocyanate conjugate (FITC-albumin) permeability in post-capillary venules in rat cremaster muscle. Inhibition of nitric oxide synthase with L-NAME (10µM) increased basal permeability to FITC-albumin, which was abrogated by superoxide dismutase and catalase. Histamine-induced (1 µM) permeability was blocked by L-NAME but unaffected by scavenging reactive oxygen species with superoxide dismutase (SOD) and catalase. In contrast, bradykinin-induced (1–100 nM) permeability increases were unaffected by L-NAME but abrogated by SOD and catalase. Acute superfusion of the cremaster muscle with IL-1β (30 pM, 10 min) resulted in a leftward shift of the bradykinin concentration–response curve. Potentiation by IL-1β of bradykinin-induced microvascular permeability was prevented by the nicotinamide adenine dinucleotide phosphate oxidase (NADPH oxidase) inhibitor apocynin (1 µM). Pretreatment of rats with simvastatin (5 mg·kg−1, i.p.) 24 h before permeability measurements prevented the potentiation of bradykinin permeability responses by IL-1β, which was not reversed by inhibition of heme oxygenase-1 with tin protoporphyrin IX (SnPP). This study highlights a novel mechanism by which simvastatin prevents the potentiation of bradykinin-induced permeability by IL-1β, possibly by targeting the assembly of NADPH oxidase subunits. Our findings highlight the therapeutic potential of statins in the prevention and treatment of patients predisposed to inflammatory diseases.
Collapse
Affiliation(s)
- Felipe Freitas
- Centre of Research Excellence, King’s College London British Heart Foundation, School of Cardiovascular Medicine & Sciences, Faculty of Life Sciences & Medicine, King’s College London, 150 Stamford Street, London SE1 9NH, UK.; (F.F.); (M.S.)
| | - Eduardo Tibiriçá
- National Institute of Cardiology, Ministry of Health, Rio de Janeiro 22240-006, Brazil;
| | - Mita Singh
- Centre of Research Excellence, King’s College London British Heart Foundation, School of Cardiovascular Medicine & Sciences, Faculty of Life Sciences & Medicine, King’s College London, 150 Stamford Street, London SE1 9NH, UK.; (F.F.); (M.S.)
| | - Paul A. Fraser
- Centre of Research Excellence, King’s College London British Heart Foundation, School of Cardiovascular Medicine & Sciences, Faculty of Life Sciences & Medicine, King’s College London, 150 Stamford Street, London SE1 9NH, UK.; (F.F.); (M.S.)
- Correspondence: (P.A.F.); (G.E.M.); Tel.: +44-(0)20-78484306 (G.E.M.)
| | - Giovanni E. Mann
- Centre of Research Excellence, King’s College London British Heart Foundation, School of Cardiovascular Medicine & Sciences, Faculty of Life Sciences & Medicine, King’s College London, 150 Stamford Street, London SE1 9NH, UK.; (F.F.); (M.S.)
- Correspondence: (P.A.F.); (G.E.M.); Tel.: +44-(0)20-78484306 (G.E.M.)
| |
Collapse
|
10
|
Therapeutic Potential of Heme Oxygenase-1 in Aneurysmal Diseases. Antioxidants (Basel) 2020; 9:antiox9111150. [PMID: 33228202 PMCID: PMC7699558 DOI: 10.3390/antiox9111150] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 12/15/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) and intracranial aneurysm (IA) are serious arterial diseases in the aorta and brain, respectively. AAA and IA are associated with old age in males and females, respectively, and if rupture occurs, they carry high morbidity and mortality. Aneurysmal subarachnoid hemorrhage (SAH) due to IA rupture has a high rate of complication and fatality. Despite these severe clinical outcomes, preventing or treating these devastating diseases remains an unmet medical need. Inflammation and oxidative stress are shared pathologies of these vascular diseases. Therefore, therapeutic strategies have focused on reducing inflammation and reactive oxygen species levels. Interestingly, in response to cellular stress, the inducible heme oxygenase-1 (HO-1) is highly upregulated and protects against tissue injury. HO-1 degrades the prooxidant heme and generates molecules with antioxidative and anti-inflammatory properties, resulting in decreased oxidative stress and inflammation. Therefore, increasing HO-1 activity is an attractive option for therapy. Several HO-1 inducers have been identified and tested in animal models for preventing or alleviating AAA, IA, and SAH. However, clinical trials have shown conflicting results. Further research and the development of highly selective HO-1 regulators may be needed to prevent the initiation and progression of AAA, IA, or SAH.
Collapse
|
11
|
Budbazar E, Rodriguez F, Sanchez JM, Seta F. The Role of Sirtuin-1 in the Vasculature: Focus on Aortic Aneurysm. Front Physiol 2020; 11:1047. [PMID: 32982786 PMCID: PMC7477329 DOI: 10.3389/fphys.2020.01047] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 07/30/2020] [Indexed: 12/19/2022] Open
Abstract
Sirtuin-1 (SirT1) is a nicotinamide adenine dinucleotide-dependent deacetylase and the best characterized member of the sirtuins family in mammalians. Sirtuin-1 shuttles between the cytoplasm and the nucleus, where it deacetylates histones and non-histone proteins involved in a plethora of cellular processes, including survival, growth, metabolism, senescence, and stress resistance. In this brief review, we summarize the current knowledge on the anti-oxidant, anti-inflammatory, anti-apoptotic, and anti-senescence effects of SirT1 with an emphasis on vascular diseases. Specifically, we describe recent research advances on SirT1-mediated molecular mechanisms in aortic aneurysm (AA), and how these processes relate to oxidant stress and the heme-oxygenase (HO) system. HO-1 and HO-2 catalyze the rate-limiting step of cellular heme degradation and, similar to SirT1, HO-1 exerts beneficial effects in the vasculature through the activation of anti-oxidant, anti-inflammatory, anti-apoptotic, and anti-proliferative signaling pathways. SirT1 and HO-1 are part of an integrated system for cellular stress tolerance, and may positively interact to regulate vascular function. We further discuss sex differences in HO-1 and SirT1 activity or expression, and the potential interactions between the two proteins, in relation to the progression and severity of AA, as well as the ongoing efforts for translational applications of SirT1 activation and HO-1 induction in the treatment of cardiovascular diseases including AA.
Collapse
Affiliation(s)
- Enkhjargal Budbazar
- Vascular Biology Section, Boston University School of Medicine, Boston, MA, United States
| | - Francisca Rodriguez
- Department of Physiology, University of Murcia and Biomedical Research Institute in Murcia (IMIB), Murcia, Spain
| | - José M Sanchez
- Department of Physiology, University of Murcia and Biomedical Research Institute in Murcia (IMIB), Murcia, Spain
| | - Francesca Seta
- Vascular Biology Section, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
12
|
Simvastatin Attenuates Abdominal Aortic Aneurysm Formation Favoured by Lack of Nrf2 Transcriptional Activity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6340190. [PMID: 32617140 PMCID: PMC7315306 DOI: 10.1155/2020/6340190] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 05/19/2020] [Accepted: 05/29/2020] [Indexed: 12/21/2022]
Abstract
Surgical intervention is currently the only option for an abdominal aortic aneurysm (AAA), preventing its rupture and sudden death of a patient. Therefore, it is crucial to determine the pathogenic mechanisms of this disease for the development of effective pharmacological therapies. Oxidative stress is said to be one of the pivotal factors in the pathogenesis of AAAs. Thus, we aimed to evaluate the significance of nuclear factor erythroid 2-related factor 2 (Nrf2) transcriptional activity in the development of AAA and to verify if simvastatin, administered as pre- and cotreatment, may counteract this structural malformation. Experiments were performed on mice with inhibited transcriptional activity of Nrf2 (tKO) and wild-type (WT) counterparts. We used a model of angiotensin II- (AngII-) induced AAA, combined with a fat-enriched diet. Mice were administered with AngII or saline for up to 28 days via osmotic minipumps. Simvastatin administration was started 7 days before the osmotic pump placement and then continued until the end of the experiment. We found that Nrf2 inactivation increased the risk of development and rupture of AAA. Importantly, these effects were reversed by simvastatin in tKO mice, but not in WT. The abrupt blood pressure rise induced by AngII was mitigated in simvastatin-treated animals regardless of the genotype. Simvastatin-affected parameters that differed between the healthy structure of the aorta and aneurysmal tissue included immune cell infiltration of the aortic wall, VCAM1 mRNA and protein level, extracellular matrix degradation, TGF-β1 mRNA level, and ERK phosphorylation, but neither oxidative stress nor the level of Angiotensin II Type 1 Receptor (AT1R). Taken together, the inhibition of Nrf2 transcriptional activity facilitates AAA formation in mice, which can be prevented by simvastatin. It suggests that statin treatment of patients with hypercholesterolemia might have not only a beneficial effect in terms of controlling atherosclerosis but also potential AAA prevention.
Collapse
|
13
|
Biliverdin reductase deficiency triggers an endothelial-to-mesenchymal transition in human endothelial cells. Arch Biochem Biophys 2019; 678:108182. [DOI: 10.1016/j.abb.2019.108182] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 10/05/2019] [Accepted: 11/04/2019] [Indexed: 02/07/2023]
|
14
|
Statin-Induced Nitric Oxide Signaling: Mechanisms and Therapeutic Implications. J Clin Med 2019; 8:jcm8122051. [PMID: 31766595 PMCID: PMC6947613 DOI: 10.3390/jcm8122051] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 11/16/2019] [Accepted: 11/20/2019] [Indexed: 12/27/2022] Open
Abstract
In addition to their cholesterol-lowering effects, statins are associated with pleiotropic effects including improvements in heart failure (HF), reduced blood pressure, prevention of the rupture of atherosclerotic plaques and improved angiogenesis. In addition to these cardiovascular benefits, statins have been implicated in the treatment of neurological injuries, cancer, sepsis, and cirrhosis. These cholesterol-independent beneficial effects of statins are predominantly mediated through signaling pathways leading to increased production and bioavailability of nitric oxide (NO). In this review, the mechanistic pathways and therapeutic effects of statin-mediated elevations of NO are described and discussed.
Collapse
|
15
|
Polanczyk A, Podgorski M, Polanczyk M, Piechota-Polanczyk A, Stefanczyk L, Strzelecki M. A novel vision-based system for quantitative analysis of abdominal aortic aneurysm deformation. Biomed Eng Online 2019; 18:56. [PMID: 31088563 PMCID: PMC6518716 DOI: 10.1186/s12938-019-0681-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 05/06/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In clinical diagnostics, combination of different imaging techniques is applied to assess spatial configuration of the abdominal aortic aneurysm (AAA) and deformation of its wall. As deformation of aneurysm wall is crucial parameter in assessing wall rupture, we aimed to develop and validate a Non-Invasive Vision-Based System (NIVBS) for the analysis of 3D elastic artificial abdominal aortic models. 3D-printed elastic AAA models from four patients were applied for the reconstruction of real hemodynamic. During experiments, the inlet boundary conditions included the injection volume and frequency of pulsation averaged from electrocardiography traces. NIVBS system was equipped with nine cameras placed at a constant distance to record wall movement from 360o angle and a dedicated set of artificial lights providing coherent illumination. Additionally, self-prepared algorithms for image acquisition, processing, segmentation, and contour detection were used to analyze wall deformation. Finally, the shape deformation factor was applied to evaluate aorta's deformation. Experimental results were confronted with medical data from AngioCT and 2D speckle-tracking echocardiography (2DSTE). RESULTS Image square analyses indicated that the optimal distance between the camera's lens and the investigated object was in the range of 0.30-0.35 m. There was approximately 1.44% difference observed in aneurysm diameters between NIVBS (86.57 ± 5.86 mm) and AngioCT (87.82 ± 6.04 mm) (p = 0.7764). The accuracy of developed algorithm for the reconstruction of the AAA deformation was equal to 98.56%. Bland-Altman analysis showed that the difference between clinical data (2DSTE) and predicted wall deformation (NIVBS) for all patients was 0.00 mm (confidence interval equal to 0.12 mm) for aneurysm size, 0.01 mm (confidence interval equal to 0.13 mm) and 0.00 mm (confidence interval equal to 0.09 mm) for the anterior and posterior side, as well as 0.01 mm (confidence interval equal to 0.18 mm) and 0.01 mm (confidence interval equal to 0.11 mm) for the left and right side. The optimal range of camera's lens did not affect acquired values. CONCLUSIONS The NIVBS with proposed algorithm that reconstructs the pressure from surrounding organs is appropriate to analyze the AAAs in water environment. Moreover, NIVBS allowed detailed quantitative analysis of aneurysm sac wall deformation.
Collapse
Affiliation(s)
- Andrzej Polanczyk
- Faculty of Process and Environmental Engineering, Department of Heat and Mass Transfer, Lodz University of Technology, Łódź, Poland.
| | - Michal Podgorski
- Department of Radiology and Diagnostic Imaging, Medical University of Lodz, Łódź, Poland
| | - Maciej Polanczyk
- Faculty of Process and Environmental Engineering, Department of Heat and Mass Transfer, Lodz University of Technology, Łódź, Poland
| | | | - Ludomir Stefanczyk
- Department of Radiology and Diagnostic Imaging, Medical University of Lodz, Łódź, Poland
| | - Michal Strzelecki
- Institute of Electronics, Lodz University of Technology, Łódź, Poland
| |
Collapse
|
16
|
Li F, Wang X, Zhang Z, Gao P, Zhang X. Breviscapine provides a neuroprotective effect after traumatic brain injury by modulating the Nrf2 signaling pathway. J Cell Biochem 2019; 120:14899-14907. [PMID: 31042302 DOI: 10.1002/jcb.28751] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 12/29/2018] [Accepted: 01/09/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Fayin Li
- Department of Anesthesiology The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University Huai'an Jiangsu China
| | - Xiaodong Wang
- Department of Neurosurgery The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University Huai'an Jiangsu China
| | - Zhijie Zhang
- Department of Anesthesiology The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University Huai'an Jiangsu China
| | - Pengfei Gao
- Department of Anesthesiology The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University Huai'an Jiangsu China
| | - Xianlong Zhang
- Department of Anesthesiology The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University Huai'an Jiangsu China
| |
Collapse
|
17
|
Xia D, Zhai X, Wang H, Chen Z, Fu C, Zhu M. Alpha lipoic acid inhibits oxidative stress-induced apoptosis by modulating of Nrf2 signalling pathway after traumatic brain injury. J Cell Mol Med 2019; 23:4088-4096. [PMID: 30989783 PMCID: PMC6533507 DOI: 10.1111/jcmm.14296] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 02/04/2019] [Accepted: 03/07/2019] [Indexed: 11/29/2022] Open
Abstract
Alpha lipoic acid (ALA) is a powerful antioxidant which has been widely used in the treatment of different system diseases, such as cardiovascular and cerebrovascular diseases. But, there are few studies that refer to protective effects and potential mechanisms on traumatic brain injury (TBI). This study was carried out to investigate the neuroprotective effect following TBI and illuminate the underlying mechanism. Weight drop‐injured model in rats was induced by weight‐drop. ALA was administrated via intraperitoneal injection after TBI. Neurologic scores were examined following several tests. Neurological score was performed to measure behavioural outcomes. Nissl staining and TUNEL were performed to evaluate the neuronal apoptosis. Western blotting was engaged to analyse the protein content of the Nuclear factor erythroid 2‐related factor 2 (Nrf2) and its downstream protein factors, including hemeoxygenase‐1 (HO‐1) and quinine oxidoreductase‐1 (NQO1). ALA treatment alleviated TBI‐induced neuron cell apoptosis and improved neurobehavioural function by up‐regulation of Nrf2 expression and its downstream protein factors after TBI. This study presents new perspective of the mechanisms responsible for the neuronal apoptosis of ALA, with possible involvement of Nrf2 pathway.
Collapse
Affiliation(s)
- Dayong Xia
- Department of Neurosurgery, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Xiaofu Zhai
- Department of Neurosurgery, Huai'an Second People's Hospital, Xuzhou Medical College, Huai'an, Jiangsu Province, China
| | - Honglian Wang
- Department of Radiology, Huai'an Fourth people's Hospital, Huai'an, Jiangsu Province, China
| | - Zhiyong Chen
- Department of Anesthesiology, the Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Chuanjing Fu
- Department of Neurosurgery, Jiangsu Hospital of Traditional Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Meihua Zhu
- Department of Anesthesiology, the Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| |
Collapse
|
18
|
Shetty AK, Kodali M, Upadhya R, Madhu LN. Emerging Anti-Aging Strategies - Scientific Basis and Efficacy. Aging Dis 2018; 9:1165-1184. [PMID: 30574426 PMCID: PMC6284760 DOI: 10.14336/ad.2018.1026] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 11/30/2018] [Indexed: 12/11/2022] Open
Abstract
The prevalence of age-related diseases is in an upward trend due to increased life expectancy in humans. Age-related conditions are among the leading causes of morbidity and death worldwide currently. Therefore, there is an urgent need to find apt interventions that slow down aging and reduce or postpone the incidence of debilitating age-related diseases. This review discusses the efficacy of emerging anti-aging approaches for maintaining better health in old age. There are many anti-aging strategies in development, which include procedures such as augmentation of autophagy, elimination of senescent cells, transfusion of plasma from young blood, intermittent fasting, enhancement of adult neurogenesis, physical exercise, antioxidant intake, and stem cell therapy. Multiple pre-clinical studies suggest that administration of autophagy enhancers, senolytic drugs, plasma from young blood, drugs that enhance neurogenesis and BDNF are promising approaches to sustain normal health during aging and also to postpone age-related neurodegenerative diseases such as Alzheimer's disease. Stem cell therapy has also shown promise for improving regeneration and function of the aged or Alzheimer's disease brain. Several of these approaches are awaiting critical appraisal in clinical trials to determine their long-term efficacy and possible adverse effects. On the other hand, procedures such as intermittent fasting, physical exercise, intake of antioxidants such as resveratrol and curcumin have shown considerable promise for improving function in aging, some of which are ready for large-scale clinical trials, as they are non-invasive, and seem to have minimal side effects. In summary, several approaches are at the forefront of becoming mainstream therapies for combating aging and postponing age-related diseases in the coming years.
Collapse
Affiliation(s)
- Ashok K. Shetty
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center College of Medicine, College Station, Texas 77843, USA
- Olin E. Teague Veterans’ Medical Center, Central Texas Veterans Health Care System, Temple, Texas 76504, USA
| | - Maheedhar Kodali
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center College of Medicine, College Station, Texas 77843, USA
- Olin E. Teague Veterans’ Medical Center, Central Texas Veterans Health Care System, Temple, Texas 76504, USA
| | - Raghavendra Upadhya
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center College of Medicine, College Station, Texas 77843, USA
- Olin E. Teague Veterans’ Medical Center, Central Texas Veterans Health Care System, Temple, Texas 76504, USA
| | - Leelavathi N. Madhu
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center College of Medicine, College Station, Texas 77843, USA
| |
Collapse
|