1
|
Wu T, Li D, Chen Q, Kong D, Zhu H, Zhou H, Zhang Q, Cui G. Identification of VDAC1 as a cardioprotective target of Ginkgolide B. Chem Biol Interact 2025; 406:111358. [PMID: 39716534 DOI: 10.1016/j.cbi.2024.111358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 11/18/2024] [Accepted: 12/17/2024] [Indexed: 12/25/2024]
Abstract
Ginkgolide B (GB), a compound derived from Ginkgo biloba, exhibits significant cardioprotective properties, although its precise molecular target has yet to be identified. In this study, we synthesized a biotin-labeled GB probe (GB-biotin) to identify the molecular targets of GB. Our experiments demonstrated that treatment with GB or GB-biotin reduced mitochondrial injury, restored mitochondrial membrane potential, and decreased cell apoptosis in a concentration-dependent manner. Additionally, GB increased mitochondrial oxygen consumption rate, indicating improved mitochondrial bioenergetics. Proteomic analysis revealed that the voltage-dependent anion channel 1 (VDAC1) is a key protein that interacts with GB-biotin. This finding was further confirmed through cellular thermal shift assay (CETSA) and molecular docking, which revealed hydrogen bond formation between GB and VDAC1. Furthermore, overexpression of VDAC1 diminished the protective effects of GB, highlighting the crucial role of VDAC1 in GB-mediated cardioprotection. These findings identify VDAC1 as a therapeutic target for GB in vitro, providing valuable insights into the cardioprotective mechanisms of GB and the development of novel cardioprotective strategies.
Collapse
Affiliation(s)
- Tingbiao Wu
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519000, China
| | - Deyao Li
- Guangzhou Henovcom Biosciences Co., Ltd., Guangzhou, 510535, China
| | - Qiuyu Chen
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519000, China
| | - Dezhi Kong
- Institute of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, 050000, China
| | - Hongxuan Zhu
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519000, China
| | - Hefeng Zhou
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519000, China
| | - Qingwen Zhang
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, 999078, China
| | - Guozhen Cui
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519000, China.
| |
Collapse
|
2
|
Zhuang B, Zhong C, Ma Y, Wang A, Quan H, Hong L. Innovative Therapeutic Strategies for Myocardial Infarction Across Various Stages: Non-Coding RNA and Stem Cells. Int J Mol Sci 2024; 26:231. [PMID: 39796085 PMCID: PMC11720039 DOI: 10.3390/ijms26010231] [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: 11/05/2024] [Revised: 12/22/2024] [Accepted: 12/27/2024] [Indexed: 01/13/2025] Open
Abstract
Myocardial infarction (MI) is a highly challenging and fatal disease, with diverse challenges arising at different stages of its progression. As such, non-coding RNAs (ncRNAs), which can broadly regulate cell fate, and stem cells with multi-differentiation potential are emerging as novel therapeutic approaches for treating MI across its various stages. NcRNAs, including microRNAs (miRNAs) and long non-coding RNAs (LncRNAs), can directly participate in regulating intracellular signaling pathways, influence cardiac angiogenesis, and promote the repair of infarcted myocardium. Currently, stem cells commonly used in medicine, such as mesenchymal stem cells (MSCs) and induced pluripotent stem cells (iPSCs), can differentiate into various human cell types without ethical concerns. When combined with ncRNAs, these stem cells can more effectively induce directed differentiation, promote angiogenesis in the infarcted heart, and replenish normal cardiac cells. Additionally, stem cell-derived exosomes, which contain various ncRNAs, can improve myocardial damage in the infarcted region through paracrine mechanisms. However, our understanding of the specific roles and mechanisms of ncRNAs, stem cells, and exosomes secreted by stem cells during different stages of MI remains limited. Therefore, this review systematically categorizes the different stages of MI, aiming to summarize the direct regulatory effects of ncRNAs on an infarcted myocardium at different points of disease progression. Moreover, it explores the specific roles and mechanisms of stem cell therapy and exosome therapy in this complex pathological evolution process. The objective of this review was to provide novel insights into therapeutic strategies for different stages of MI and open new research directions for the application of stem cells and ncRNAs in the field of MI repair.
Collapse
Affiliation(s)
- Bingqi Zhuang
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji 133002, China; (B.Z.); (C.Z.); (Y.M.)
| | - Chongning Zhong
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji 133002, China; (B.Z.); (C.Z.); (Y.M.)
| | - Yuting Ma
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji 133002, China; (B.Z.); (C.Z.); (Y.M.)
| | - Ao Wang
- Experimental Teaching Center, College of Pharmacy, Yanbian University, Yanji 133002, China;
| | - Hailian Quan
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji 133002, China; (B.Z.); (C.Z.); (Y.M.)
| | - Lan Hong
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji 133002, China; (B.Z.); (C.Z.); (Y.M.)
| |
Collapse
|
3
|
Chen J, Zhang G, Guo A, Mou C, Du M, Zhai S, Huang M. Sevoflurane attenuates hypoxia/reoxygenation-induced cardiomyocyte injury by regulating miR-4454. Toxicol Res (Camb) 2024; 13:tfae219. [PMID: 39712638 PMCID: PMC11659642 DOI: 10.1093/toxres/tfae219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/17/2024] [Accepted: 12/11/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Sevoflurane (Sevo) prevents hypoxia/reoxygenation (H/R)-induced cardiomyocytes injury. The expression of miR-4,454 was increased in individuals experiencing an acute myocardial infarction. OBJECTIVE The purpose of current investigation was to delved into whether the effects of Sevo on cardiomyocytes are mediated through regulation of miR-4,454 expression. METHOD In this study, the expression levels of miR-4,454 and BAG5 were detected by real-time quantitative polymerase chain reaction (RT-qPCR). Cell viability was detected by cell counting kit-8 (CCK-8). The levels of creatine kinase-MB (CK-MB), lactate dehydrogenase (LDH) and cardiac troponin I (cTnI) were detected by enzyme-linked immunosorbent assay (ELISA). Reactive oxygen species (ROS), malondialdehyde (MDA), superoxide dismutase (SOD) and catalase (CAT) were detected using various commercially available kits to assess the level of oxidative stress in the cells. The luciferase reporter gene assay was used to verify the interaction of miR-4,454 with downstream target genes. RESULTS There was a notable upregulation of miR-4,454 expression in H/R-induced cardiomyocyte models. This was accompanied by a decrease in the viability of myocardial cells induced by H/R and an intensification of the extent of myocardial injury and oxidative stress. However, the detrimental effects were mitigated by the administration of Sevo. miR-4,454 had a target site for binding to BAG5, and its expression was negatively modulated by miR-4,454. An increase in the expression of BAG5 was shown to directly offset the exacerbation of cardiomyocyte damage induced by the overexpression of miR-4,454. CONCLUSION Sevo may attenuate H/R-induced cardiomyocyte injury by regulating miR-4454.
Collapse
Affiliation(s)
- Jianxing Chen
- Department of Anesthesiology, The First Affiliated Hospital, Fujian Medical University, No. 20, Chazhong Road, Fuzhou 350005, China
- Department of Anesthesiology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, No. 999, Wansha Road, Changle District, Fuzhou 350212, China
- Anesthesiology Research Institute, The First Affiliated Hospital, Fujian Medical University, No. 20, Chazhong Road, Fuzhou 350005, China
| | - Gaofeng Zhang
- Department of Critical Care Medicine, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, No. 6, Huanghe Road, Yushan Town, Changshu 215500, China
| | - Aili Guo
- Department of Anesthesiology and Surgery, Shengli Oilfield Central Hospital, No. 31, Jinan Road, Dongying 257034, China
| | - Changliang Mou
- Department of Anesthesiology and Surgery, Shengli Oilfield Central Hospital, No. 31, Jinan Road, Dongying 257034, China
| | - Meiqing Du
- Department of Anesthesiology and Surgery, Shengli Oilfield Central Hospital, No. 31, Jinan Road, Dongying 257034, China
| | - Shuang Zhai
- Department of Anesthesiology and Surgery, Shengli Oilfield Central Hospital, No. 31, Jinan Road, Dongying 257034, China
| | - Mingshan Huang
- Department of Cardiovascular Medicine, Ganzhou People's Hospital, No. 16 Meiguan Avenue, Ganzhou 341000, China
| |
Collapse
|
4
|
Arif T, Shteinfer-Kuzmine A, Shoshan-Barmatz V. Decoding Cancer through Silencing the Mitochondrial Gatekeeper VDAC1. Biomolecules 2024; 14:1304. [PMID: 39456237 PMCID: PMC11506819 DOI: 10.3390/biom14101304] [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: 08/26/2024] [Revised: 10/13/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
Mitochondria serve as central hubs for regulating numerous cellular processes that include metabolism, apoptosis, cell cycle progression, proliferation, differentiation, epigenetics, immune signaling, and aging. The voltage-dependent anion channel 1 (VDAC1) functions as a crucial mitochondrial gatekeeper, controlling the flow of ions, such as Ca2+, nucleotides, and metabolites across the outer mitochondrial membrane, and is also integral to mitochondria-mediated apoptosis. VDAC1 functions in regulating ATP production, Ca2+ homeostasis, and apoptosis, which are essential for maintaining mitochondrial function and overall cellular health. Most cancer cells undergo metabolic reprogramming, often referred to as the "Warburg effect", supplying tumors with energy and precursors for the biosynthesis of nucleic acids, phospholipids, fatty acids, cholesterol, and porphyrins. Given its multifunctional nature and overexpression in many cancers, VDAC1 presents an attractive target for therapeutic intervention. Our research has demonstrated that silencing VDAC1 expression using specific siRNA in various tumor types leads to a metabolic rewiring of the malignant cancer phenotype. This results in a reversal of oncogenic properties that include reduced tumor growth, invasiveness, stemness, epithelial-mesenchymal transition. Additionally, VDAC1 depletion alters the tumor microenvironment by reducing angiogenesis and modifying the expression of extracellular matrix- and structure-related genes, such as collagens and glycoproteins. Furthermore, VDAC1 depletion affects several epigenetic-related enzymes and substrates, including the acetylation-related enzymes SIRT1, SIRT6, and HDAC2, which in turn modify the acetylation and methylation profiles of histone 3 and histone 4. These epigenetic changes can explain the altered expression levels of approximately 4000 genes that are associated with reversing cancer cells oncogenic properties. Given VDAC1's critical role in regulating metabolic and energy processes, targeting it offers a promising strategy for anti-cancer therapy. We also highlight the role of VDAC1 expression in various disease pathologies, including cardiovascular, neurodegenerative, and viral and bacterial infections, as explored through siRNA targeting VDAC1. Thus, this review underscores the potential of targeting VDAC1 as a strategy for addressing high-energy-demand cancers. By thoroughly understanding VDAC1's diverse roles in metabolism, energy regulation, mitochondrial functions, and other cellular processes, silencing VDAC1 emerges as a novel and strategic approach to combat cancer.
Collapse
Affiliation(s)
- Tasleem Arif
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Anna Shteinfer-Kuzmine
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel;
| | - Varda Shoshan-Barmatz
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel;
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| |
Collapse
|
5
|
Qi K, Cao F, Wang J, Wang Y, Li G. miR-652-3p Suppressed the Protective Effects of Isoflurane Against Myocardial Injury in Hypoxia/Reoxygenation by Targeting ISL1. Cardiovasc Toxicol 2024; 24:646-655. [PMID: 38801481 DOI: 10.1007/s12012-024-09870-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 05/10/2024] [Indexed: 05/29/2024]
Abstract
This research is concentrated on investigating the role and mechanism of miR-652-3p in the protective effects of isoflurane (ISO) against myocardial ischemia-reperfusion (I/R) injury. H9c2 cells underwent pretreatment with varying concentrations of ISO, and subsequently, a hypoxia/reoxygenation (H/R) model was constructed. The levels of miR-652-3p, ISL LIM homeobox 1 (ISL1), and inflammatory cytokines interleukin (IL)-6 and tumor necrosis factor-alpha (TNF-α) were evaluated through reverse transcription polymerase chain reaction (RT-qPCR). Enzyme-linked immunosorbent assay was employed to investigate concentrations of myocardial injury markers, such as creatine kinase-MB (CK-MB) and cardiac troponin I (cTnI). Cell counting kit-8 was used to evaluate cell viability, while flow cytometry was utilized to measure apoptosis. Additionally, a dual luciferase reporter assay was conducted to validate the targeting relationship between ISL1 and miR-652-3p. Herein, we confirmed that the level of miR-652-3p was gradually increased with prolonged hypoxia; nevertheless, this increase was suppressed by ISO pretreatment (P < 0.05). Additionally, ISO pretreatment prevented the decrease in cell viability, increase in apoptosis, and overproduction of IL-6, TNF-α, CK-MB, and cTnI induced by H/R (P < 0.05). However, the inhibitory effects of ISO were counteracted by the increased levels of miR-652-3p (P < 0.05). ISL1 is a potential target of miR-652-3p. H/R induction suppressed ISL1 levels compared to the control, but ISO treatment increased its expression (P < 0.05). Overexpression of ISL1 inhibited the elimination of the protective effect of ISO on myocardial damage induced by the elevation of miR-652-3p (P < 0.05). The findings of this research confirm that miR-652-3p attenuated the protective effect of ISO on cardiomyocytes in myocardial ischemia by targeting ISL1.
Collapse
Affiliation(s)
- Kaikai Qi
- Department of Anesthesiology, The Second Affiliated Hospital of Shandong First Medical University, 366, Taishan Street, Taishan District, Taian, 271000, Shandong, China
| | - Fang Cao
- Department of Orthopedics, The Second Affiliated Hospital of Shandong First Medical University, Taian, 271000, Shandong, China
| | - Jing Wang
- Operating Room, The Second Affiliated Hospital of Shandong First Medical University, Taian, 271000, Shandong, China
| | - Yu Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Shandong First Medical University, 366, Taishan Street, Taishan District, Taian, 271000, Shandong, China
| | - Guohua Li
- Department of Anesthesiology, The Second Affiliated Hospital of Shandong First Medical University, 366, Taishan Street, Taishan District, Taian, 271000, Shandong, China.
| |
Collapse
|
6
|
Jiao Y, Zhang YH, Wang CY, Yu Y, Li YZ, Cui W, Li Q, Yu YH. MicroRNA-7a-5p ameliorates diabetic peripheral neuropathy by regulating VDAC1/JNK/c-JUN pathway. Diabet Med 2023; 40:e14890. [PMID: 35616949 DOI: 10.1111/dme.14890] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 05/04/2022] [Indexed: 12/24/2022]
Abstract
AIMS The pathogenesis of diabetic peripheral neuropathy (DPN) is complex, and its treatment is extremely challenging. MicroRNA-7a-5p (miR-7a-5p) has been widely reported to alleviate apoptosis and oxidative stress in various diseases. This study aimed to investigate the mechanism of miR-7a-5p in DPN. METHODS DPN cell model was constructed with high-glucose-induced RSC96 cells. Cell apoptosis and viability were detected by flow cytometry analysis and cell counting kit-8 (CCK-8) assay respectively. The apoptosis and Jun N-terminal kinase (JNK)/c-JUN signalling pathway-related proteins expression were detected by Western blotting. The intracellular calcium content and oxidative stress levels were detected by flow cytometry and reagent kits. Mitochondrial membrane potential was evaluated by tetrechloro-tetraethylbenzimidazol carbocyanine iodide (JC-1) staining. The targeting relationship between miR-7a-5p and voltage-dependent anion-selective channel protein 1 (VDAC1) was determined by RNA pull-down assay and dual-luciferase reporter gene assay. The streptozotocin (STZ) rat model was constructed to simulate DPN in vivo. The paw withdrawal mechanical threshold (PTW) was measured by Frey capillary line, and the motor nerve conduction velocity (MNCV) was measured by electromyography. RESULTS MiR-7a-5p expression was decreased, while VDAC1 expression was increased in HG-induced RSC96 cells and STZ rats. In HG-induced RSC96 cells, miR-7a-5p overexpression promoted cell proliferation, inhibited apoptosis, down-regulated calcium release, improved mitochondrial membrane potential and repressed oxidative stress response. MiR-7a-5p negatively regulated VDAC1 expression. VDAC1 knockdown improved cell proliferation activity, suppressed cell apoptosis and mitochondrial dysfunction by inhibiting JNK/c-JUN pathway activation. MiR-7a-5p overexpression raised PTW, restored MNCV and reduced oxidative stress levels and nerve cell apoptosis in STZ rats. CONCLUSION MiR-7a-5p overexpression ameliorated mitochondrial dysfunction and inhibited apoptosis in DPN by regulating VDAC1/JNK/c-JUN pathway.
Collapse
Affiliation(s)
- Yang Jiao
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Research Institute of Anesthesiology, Tianjin, China
| | - Yue-Hua Zhang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Research Institute of Anesthesiology, Tianjin, China
| | - Chun-Yan Wang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Research Institute of Anesthesiology, Tianjin, China
| | - Yang Yu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Research Institute of Anesthesiology, Tianjin, China
| | - Yi-Ze Li
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Research Institute of Anesthesiology, Tianjin, China
| | - Wei Cui
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Research Institute of Anesthesiology, Tianjin, China
| | - Qing Li
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Research Institute of Anesthesiology, Tianjin, China
| | - Yong-Hao Yu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Research Institute of Anesthesiology, Tianjin, China
| |
Collapse
|
7
|
Kansakar U, Varzideh F, Mone P, Jankauskas SS, Santulli G. Functional Role of microRNAs in Regulating Cardiomyocyte Death. Cells 2022; 11:983. [PMID: 35326433 PMCID: PMC8946783 DOI: 10.3390/cells11060983] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 03/08/2022] [Accepted: 03/09/2022] [Indexed: 02/08/2023] Open
Abstract
microRNAs (miRNA, miRs) play crucial roles in cardiovascular disease regulating numerous processes, including inflammation, cell proliferation, angiogenesis, and cell death. Herein, we present an updated and comprehensive overview of the functional involvement of miRs in the regulation of cardiomyocyte death, a central event in acute myocardial infarction, ischemia/reperfusion, and heart failure. Specifically, in this systematic review we are focusing on necrosis, apoptosis, and autophagy.
Collapse
Affiliation(s)
- Urna Kansakar
- Department of Medicine (Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY 10461, USA; (U.K.); (F.V.); (P.M.); (S.S.J.)
| | - Fahimeh Varzideh
- Department of Medicine (Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY 10461, USA; (U.K.); (F.V.); (P.M.); (S.S.J.)
| | - Pasquale Mone
- Department of Medicine (Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY 10461, USA; (U.K.); (F.V.); (P.M.); (S.S.J.)
| | - Stanislovas S. Jankauskas
- Department of Medicine (Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY 10461, USA; (U.K.); (F.V.); (P.M.); (S.S.J.)
- Department of Molecular Pharmacology, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Fleischer Institute for Diabetes and Metabolism (FIDAM), Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Gaetano Santulli
- Department of Medicine (Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY 10461, USA; (U.K.); (F.V.); (P.M.); (S.S.J.)
- Department of Molecular Pharmacology, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Fleischer Institute for Diabetes and Metabolism (FIDAM), Albert Einstein College of Medicine, New York, NY 10461, USA
| |
Collapse
|