1
|
Xu MY, Xu JJ, Kang LJ, Liu ZH, Su MM, Zhao WQ, Wang ZH, Sun L, Xiao JB, Evans PC, Tian XY, Wang L, Huang Y, Liang XM, Weng JP, Xu SW. Urolithin A promotes atherosclerotic plaque stability by limiting inflammation and hypercholesteremia in Apolipoprotein E-deficient mice. Acta Pharmacol Sin 2024; 45:2277-2289. [PMID: 38886550 PMCID: PMC11489441 DOI: 10.1038/s41401-024-01317-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 05/16/2024] [Indexed: 06/20/2024] Open
Abstract
Urolithin A (UroA), a dietary phytochemical, is produced by gut bacteria from fruits rich in natural polyphenols ellagitannins (ETs). The efficiency of ETs metabolism to UroA in humans depends on gut microbiota. UroA has shown a variety of pharmacological activities. In this study we investigated the effects of UroA on atherosclerotic lesion development and stability. Apolipoprotein E-deficient (ApoE-/-) mice were fed a high-fat and high-cholesterol diet for 3 months to establish atherosclerosis model. Meanwhile the mice were administered UroA (50 mg·kg-1·d-1, i.g.). We showed that UroA administration significantly decreased diet-induced atherosclerotic lesions in brachiocephalic arteries, macrophage content in plaques, expression of endothelial adhesion molecules, intraplaque hemorrhage and size of necrotic core, while increased the expression of smooth muscle actin and the thickness of fibrous cap, implying features of plaque stabilization. The underlying mechanisms were elucidated using TNF-α-stimulated human endothelial cells. Pretreatment with UroA (10, 25, 50 μM) dose-dependently inhibited TNF-α-induced endothelial cell activation and monocyte adhesion. However, the anti-inflammatory effects of UroA in TNF-α-stimulated human umbilical vein endothelial cells (HUVECs) were independent of NF-κB p65 pathway. We conducted RNA-sequencing profiling analysis to identify the differential expression of genes (DEGs) associated with vascular function, inflammatory responses, cell adhesion and thrombosis in UroA-pretreated HUVECs. Human disease enrichment analysis revealed that the DEGs were significantly correlated with cardiovascular diseases. We demonstrated that UroA pretreatment mitigated endothelial inflammation by promoting NO production and decreasing YAP/TAZ protein expression and TEAD transcriptional activity in TNF-α-stimulated HUVECs. On the other hand, we found that UroA administration modulated the transcription and cleavage of lipogenic transcription factors SREBP1/2 in the liver to ameliorate cholesterol metabolism in ApoE-/- mice. This study provides an experimental basis for new dietary therapeutic option to prevent atherosclerosis.
Collapse
Affiliation(s)
- Meng-Yun Xu
- Department of Endocrinology, Institute of Endocrine and Metabolic Disease, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, 230000, China
| | - Jing-Jing Xu
- Department of Endocrinology, Institute of Endocrine and Metabolic Disease, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, 230000, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Li-Jing Kang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Zheng-Hong Liu
- Department of Endocrinology, Institute of Endocrine and Metabolic Disease, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, 230000, China
| | - Mei-Ming Su
- Department of Endocrinology, Institute of Endocrine and Metabolic Disease, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, 230000, China
| | - Wen-Qi Zhao
- Department of Endocrinology, Institute of Endocrine and Metabolic Disease, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, 230000, China
| | - Zhi-Hua Wang
- Department of Endocrinology, Institute of Endocrine and Metabolic Disease, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, 230000, China
| | - Lu Sun
- Department of Endocrinology, Institute of Endocrine and Metabolic Disease, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, 230000, China
| | - Jian-Bo Xiao
- Universidade de Vigo, Department of Analytical and Food Chemistry, Faculty of Sciences, Ourense, 32004, Spain
| | - Paul C Evans
- Centre for Biochemical Pharmacology, William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Xiao-Yu Tian
- School of Biomedical Sciences, Heart and Vascular Institute, Faculty of Medicine, The Chinese University of Hong Kong, Shatin NT, Hong Kong SAR, 999077, China
| | - Li Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Xin-Miao Liang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116000, China.
| | - Jian-Ping Weng
- Department of Endocrinology, Institute of Endocrine and Metabolic Disease, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, 230000, China.
| | - Suo-Wen Xu
- Department of Endocrinology, Institute of Endocrine and Metabolic Disease, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, 230000, China.
| |
Collapse
|
2
|
Ma Y, Wang L, He J, Ma X, Wang J, Yan R, Ma W, Ma H, Liu Y, Sun H, Zhang X, Jia S, Wang H. Sodium Selenite Ameliorates Silver Nanoparticles Induced Vascular Endothelial Cytotoxic Injury by Antioxidative Properties and Suppressing Inflammation Through Activating the Nrf2 Signaling Pathway. Biol Trace Elem Res 2024; 202:4567-4585. [PMID: 38150116 PMCID: PMC11339151 DOI: 10.1007/s12011-023-04014-2] [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: 11/03/2023] [Accepted: 12/11/2023] [Indexed: 12/28/2023]
Abstract
Silver nanoparticles (AgNP) are the dominant nanomaterials in commercial products and the medical field, but the widespread occurrence of AgNP has become a global threat to human health. Growing studies indicate that AgNP exposure can induce vascular endothelial toxicity by excessive oxidative stress and inflammation, which is closely related to cardiovascular disease (CVD), but the potential intrinsic mechanism remains poorly elucidated. Thus, it has been crucial to control the toxicological effects of AgNP in order to improve their safety and increase the outcome of their applications.Multiple researches have demonstrated that sodium selenite (Se) possesses the capability to counteract the toxicity of AgNP, but the functional role of Se in AgNP-induced CVD is largely unexplored. The aim of this study was to explore the potential protective effect of Se on AgNP-induced vascular endothelial lesion and elucidate the underlying mechanisms. An in vivo model of toxicity in animals was established by the instillation of 200 µL of AgNP into the trachea of rats both with (0.2 mg/kg/day) and without Se treated. In vitro experiments, human umbilical vein endothelial cells (HUVECs) were incubated with AgNP (0.3 µg/mL ) and Se for a duration of 24 h. Utilizing transmission electron microscopy, we observed that the internalization of AgNP-induced endothelial cells was desquamated from the internal elastic lamina, the endoplasmic reticulum was dilated, and the medullary vesicle formed. Se treatment reduced the levels of vascular cell adhesion molecule-1 (VCAM-1) and intercellular adhesion molecule-1 (ICAM-1), inhibited the release of pro-inflammatory cytokines (specifically tumor necrosis factor (TNF)-α, interleukin (IL)-1β and IL-6), improved endothelial cell permeability, integrity, and dysfunction, and prevented damage to the aortic endothelium caused by AgNP. Importantly, we found that Se showed the capacity against AgNP with biological functions in guiding the intracellular reactive oxygen species (ROS) scavenging and meanwhile exhibiting anti-inflammation effects. Se supplementation decreased the intracellular ROS release and suppressed NOD-like receptor protein 3 (NLRP3) and nuclear factor kappa-B (NF-κB) mediated inflammation within AgNP-intoxicated rats and HUVECs. The anti-oxidant stress and anti-inflammatory effects of Se were at least partly dependent on nuclear factor erythroid 2-related factor 2 (Nrf2). Overall, our results indicated that the protectiveness of Se against AgNP-induced vascular endothelial toxicity injury was at least attributed to the inhibition of oxidative ROS and pro-inflammatory NF-κB/NLRP3 inflammasome by activating the Nrf2 and antioxidant enzyme (HO-1) signal pathway.
Collapse
Affiliation(s)
- Yunyun Ma
- General Hospital of Ningxia Medical University (the First Clinical Medical College of Ningxia Medical University), Yinchuan, 750004, Ningxia, China
- Heart Centre &, Department of Cardiovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Lei Wang
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Jing He
- Heart Centre &, Department of Cardiovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Xueping Ma
- Heart Centre &, Department of Cardiovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Jingjing Wang
- Heart Centre &, Department of Cardiovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Ru Yan
- Heart Centre &, Department of Cardiovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Wanrui Ma
- Department of General Medicine, The First Dongguan Affiliated Hospital of Guangdong Medical University, Dongguan, China
| | - Huiyan Ma
- Heart Centre &, Department of Cardiovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Yajuan Liu
- Heart Centre &, Department of Cardiovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Hongqian Sun
- Heart Centre &, Department of Cardiovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Xiaoxia Zhang
- College of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan, Ningxia, China.
| | - Shaobin Jia
- Heart Centre &, Department of Cardiovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China.
| | - Hao Wang
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia, China.
| |
Collapse
|
3
|
Yalaz C, Bridges E, Alham NK, Zois CE, Chen J, Bensaad K, Miar A, Pires E, Muschel RJ, McCullagh JSO, Harris AL. Cone photoreceptor phosphodiesterase PDE6H inhibition regulates cancer cell growth and metabolism, replicating the dark retina response. Cancer Metab 2024; 12:5. [PMID: 38350962 PMCID: PMC10863171 DOI: 10.1186/s40170-023-00326-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 11/24/2023] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND PDE6H encodes PDE6γ', the inhibitory subunit of the cGMP-specific phosphodiesterase 6 in cone photoreceptors. Inhibition of PDE6, which has been widely studied for its role in light transduction, increases cGMP levels. The purpose of this study is to characterise the role of PDE6H in cancer cell growth. METHODS From an siRNA screen for 487 genes involved in metabolism, PDE6H was identified as a controller of cell cycle progression in HCT116 cells. Role of PDE6H in cancer cell growth and metabolism was studied through the effects of its depletion on levels of cell cycle controllers, mTOR effectors, metabolite levels, and metabolic energy assays. Effect of PDE6H deletion on tumour growth was also studied in a xenograft model. RESULTS PDE6H knockout resulted in an increase of intracellular cGMP levels, as well as changes to the levels of nucleotides and key energy metabolism intermediates. PDE6H knockdown induced G1 cell cycle arrest and cell death and reduced mTORC1 signalling in cancer cell lines. Both knockdown and knockout of PDE6H resulted in the suppression of mitochondrial function. HCT116 xenografts revealed that PDE6H deletion, as well as treatment with the PDE5/6 inhibitor sildenafil, slowed down tumour growth and improved survival, while sildenafil treatment did not have an additive effect on slowing the growth of PDE6γ'-deficient tumours. CONCLUSIONS Our results indicate that the changes in cGMP and purine pools, as well as mitochondrial function which is observed upon PDE6γ' depletion, are independent of the PKG pathway. We show that in HCT116, PDE6H deletion replicates many effects of the dark retina response and identify PDE6H as a new target in preventing cancer cell proliferation and tumour growth.
Collapse
Affiliation(s)
- Ceren Yalaz
- Molecular Oncology Laboratories, Department of Medical Oncology, John Radcliffe Hospital, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK.
| | - Esther Bridges
- Molecular Oncology Laboratories, Department of Medical Oncology, John Radcliffe Hospital, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Nasullah K Alham
- Department of Engineering Science, Institute of Biomedical Engineering (IBME), University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Christos E Zois
- Molecular Oncology Laboratories, Department of Medical Oncology, John Radcliffe Hospital, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Jianzhou Chen
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Karim Bensaad
- Molecular Oncology Laboratories, Department of Medical Oncology, John Radcliffe Hospital, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Ana Miar
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Elisabete Pires
- Department of Chemistry, University of Oxford, Mansfield Road, Oxford, OX1 3TA, UK
| | - Ruth J Muschel
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - James S O McCullagh
- Department of Chemistry, University of Oxford, Mansfield Road, Oxford, OX1 3TA, UK
| | - Adrian L Harris
- Molecular Oncology Laboratories, Department of Medical Oncology, John Radcliffe Hospital, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| |
Collapse
|
4
|
Zhang X, Peng B, Zhang S, Wang J, Yuan X, Peled S, Chen W, Ding J, Li W, Zhang A, Wu Q, Stavrovskaya IG, Luo C, Sinha B, Tu Y, Yuan X, Li M, Liu S, Fu J, Aziz-Sultan A, Kristal BS, Alterovitz G, Du R, Zhou S, Wang X. The MT1 receptor as the target of ramelteon neuroprotection in ischemic stroke. J Pineal Res 2024; 76:e12925. [PMID: 37986632 PMCID: PMC10872556 DOI: 10.1111/jpi.12925] [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: 04/04/2023] [Revised: 10/31/2023] [Accepted: 11/02/2023] [Indexed: 11/22/2023]
Abstract
Stroke is the leading cause of death and disability worldwide. Novel and effective therapies for ischemic stroke are urgently needed. Here, we report that melatonin receptor 1A (MT1) agonist ramelteon is a neuroprotective drug candidate as demonstrated by comprehensive experimental models of ischemic stroke, including a middle cerebral artery occlusion (MCAO) mouse model of cerebral ischemia in vivo, organotypic hippocampal slice cultures ex vivo, and cultured neurons in vitro; the neuroprotective effects of ramelteon are diminished in MT1-knockout (KO) mice and MT1-KO cultured neurons. For the first time, we report that the MT1 receptor is significantly depleted in the brain of MCAO mice, and ramelteon treatment significantly recovers the brain MT1 losses in MCAO mice, which is further explained by the Connectivity Map L1000 bioinformatic analysis that shows gene-expression signatures of MCAO mice are negatively connected to melatonin receptor agonist like Ramelteon. We demonstrate that ramelteon improves the cerebral blood flow signals in ischemic stroke that is potentially mediated, at least, partly by mechanisms of activating endothelial nitric oxide synthase. Our results also show that the neuroprotection of ramelteon counteracts reactive oxygen species-induced oxidative stress and activates the nuclear factor erythroid 2-related factor 2/heme oxygenase-1 pathway. Ramelteon inhibits the mitochondrial and autophagic death pathways in MCAO mice and cultured neurons, consistent with gene set enrichment analysis from a bioinformatics perspective angle. Our data suggest that Ramelteon is a potential neuroprotective drug candidate, and MT1 is the neuroprotective target for ischemic stroke, which provides new insights into stroke therapy. MT1-KO mice and cultured neurons may provide animal and cellular models of accelerated ischemic damage and neuronal cell death.
Collapse
Affiliation(s)
- Xinmu Zhang
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- Department of Biopharmaceutical Sciences, College of Pharmacy, Jilin University, Changchun, Jilin, China
| | - Bin Peng
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Shenqi Zhang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Jian Wang
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Xiong Yuan
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- Department of Orthopedic Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Sharon Peled
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Wu Chen
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- Department of Clinical Laboratory, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Jinyin Ding
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Wei Li
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Andrew Zhang
- Biomedical Cybernetics Laboratory, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Qiaofeng Wu
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Irina G. Stavrovskaya
- Department of Medicine, Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Research Foundation of The City University of New York, New York, NY, USA
| | - Chengliang Luo
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Bharati Sinha
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Yanyang Tu
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Xiaojing Yuan
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Mingchang Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Shuqing Liu
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jianfang Fu
- Department of Endocrinology, Xijing Hospital, Xi'an, Shaanxi, China
- The Joslin Beth Israel Deaconess Foot Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Ali Aziz-Sultan
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Bruce S. Kristal
- Department of Medicine, Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, Massachusetts, USA
| | - Gil Alterovitz
- Biomedical Cybernetics Laboratory, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Rose Du
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Shuanhu Zhou
- Department of Orthopedic Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Xin Wang
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
5
|
Liu ZB, Fan XY, Wang CW, Ye X, Wu CJ. Potentially active compounds that improve PAD through angiogenesis: A review. Biomed Pharmacother 2023; 168:115634. [PMID: 37879211 DOI: 10.1016/j.biopha.2023.115634] [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: 08/08/2023] [Revised: 09/25/2023] [Accepted: 10/03/2023] [Indexed: 10/27/2023] Open
Abstract
Peripheral arterial disease (PAD) has been historically neglected, which has resulted in a lack of effective drugs in clinical practice. However, with the increasing prevalence of diseases like atherosclerosis and diabetes, the incidence of PAD is rising and cannot be ignored. Researchers are exploring the potential of promoting angiogenesis through exogenous compounds to improve PAD. This paper focuses on the therapeutic effect of natural products (Salidroside, Astragaloside IV, etc.) and synthetic compounds (Cilostazol, Dapagliflozin, etc.). Specifically, it examines how they can promote autocrine secretion of vascular endothelial cells, enhance cell paracrine interactions, and regulate endothelial progenitor cell function. The activation of these effects may be closely related to PI3K, AMPK, and other pathways. Overall, these exogenous compounds have promising therapeutic potential for PAD. This study aims to summarize the potential active compounds, provide a variety of options for the search for drugs for the treatment of PAD, and bring light to the treatment of patients.
Collapse
Affiliation(s)
- Zi-Bo Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xin-Yun Fan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Chen-Wei Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xun Ye
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Chun-Jie Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy/Academy for Interdiscipline, Chengdu Univesity of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
6
|
Wang E, Andrade MJ, Smith Q. Vascularized liver-on-a-chip model to investigate nicotine-induced dysfunction. BIOMICROFLUIDICS 2023; 17:064108. [PMID: 38155919 PMCID: PMC10754629 DOI: 10.1063/5.0172677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 11/29/2023] [Indexed: 12/30/2023]
Abstract
The development of physiologically relevant in vitro systems for simulating disease onset and progression and predicting drug metabolism holds tremendous value in reducing drug discovery time and cost. However, many of these platforms lack accuracy in replicating the tissue architecture and multicellular interactions. By leveraging three-dimensional cell culture, biomimetic soft hydrogels, and engineered stimuli, in vitro models have continued to progress. Nonetheless, the incorporation of the microvasculature has been met with many challenges, specifically with the addition of parenchymal cell types. Here, a systematic approach to investigating the initial seeding density of endothelial cells and its effects on interconnected networks was taken and combined with hepatic spheroids to form a liver-on-a-chip model. Leveraging this system, nicotine's effects on microvasculature and hepatic function were investigated. The findings indicated that nicotine led to interrupted adherens junctions, decreased guanosine triphosphate cyclohydrolase 1 expression, impaired angiogenesis, and lowered barrier function, all key factors in endothelial dysfunction. With the combination of the optimized microvascular networks, a vascularized liver-on-a-chip was formed, providing functional xenobiotic metabolism and synthesis of both albumin and urea. This system provides insight into potential hepatotoxicity caused by various drugs and allows for assessing vascular dysfunction in a high throughput manner.
Collapse
Affiliation(s)
- Eric Wang
- Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, California 92697, USA
| | - Melisa J. Andrade
- Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, California 92697, USA
| | | |
Collapse
|
7
|
Robillard S, Trân K, Lachance MS, Brazeau T, Boisvert E, Lizotte F, Auger-Messier M, Boudreault PL, Marsault É, Geraldes P. Apelin prevents diabetes-induced poor collateral vessel formation and blood flow reperfusion in ischemic limb. Front Cardiovasc Med 2023; 10:1191891. [PMID: 37636297 PMCID: PMC10450936 DOI: 10.3389/fcvm.2023.1191891] [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: 03/22/2023] [Accepted: 08/01/2023] [Indexed: 08/29/2023] Open
Abstract
Introduction Peripheral arterial disease (PAD) is a major risk factor for lower-extremity amputation in diabetic patients. Unfortunately, previous clinical studies investigating therapeutic angiogenesis using the vascular endothelial growth factor (VEGF) have shown disappointing results in diabetic patients, which evokes the necessity for novel therapeutic agents. The apelinergic system (APJ receptor/apelin) is highly upregulated under hypoxic condition and acts as an activator of angiogenesis. Apelin treatment improves revascularization in nondiabetic models of ischemia, however, its role on angiogenesis in diabetic conditions remains poorly investigated. This study explored the impact of Pyr-apelin-13 in endothelial cell function and diabetic mouse model of hindlimb ischemia. Methods Nondiabetic and diabetic mice underwent femoral artery ligation to induce limb ischemia. Diabetic mice were implanted subcutaneously with osmotic pumps delivering Pyr-apelin-13 for 28 days. Blood flow reperfusion was measured for 4 weeks post-surgery and exercise willingness was assessed with voluntary wheels. In vitro, bovine aortic endothelial cells (BAECs) were exposed to normal (NG) or high glucose (HG) levels and hypoxia. Cell migration, proliferation and tube formation assays were performed following either VEGF or Pyr-apelin-13 stimulation. Results and Discussion Following limb ischemia, blood flow reperfusion, functional recovery of the limb and vascular density were improved in diabetic mice receiving Pyr-apelin-13 compared to untreated diabetic mice. In cultured BAECs, exposure to HG concentrations and hypoxia reduced VEGF proangiogenic actions, whereas apelin proangiogenic effects remained unaltered. Pyr-apelin-13 induced its proangiogenic actions through Akt/AMPK/eNOS and RhoA/ROCK signaling pathways under both NG or HG concentrations and hypoxia exposure. Our results identified the apelinergic system as a potential therapeutic target for angiogenic therapy in diabetic patients with PAD.
Collapse
Affiliation(s)
- Stéphanie Robillard
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Kien Trân
- Department of Pharmacology and Physiology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Marie-Sophie Lachance
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Tristan Brazeau
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Elizabeth Boisvert
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Farah Lizotte
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Mannix Auger-Messier
- Division of Cardiology, Department of Medicine, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Pierre-Luc Boudreault
- Department of Pharmacology and Physiology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Éric Marsault
- Department of Pharmacology and Physiology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Pedro Geraldes
- Division of Endocrinology, Department of Medicine, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
8
|
Tu J, Liang H, Li C, Huang Y, Wang Z, Chen X, Yuan X. The application and research progress of anti-angiogenesis therapy in tumor immunotherapy. Front Immunol 2023; 14:1198972. [PMID: 37334350 PMCID: PMC10272381 DOI: 10.3389/fimmu.2023.1198972] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 05/15/2023] [Indexed: 06/20/2023] Open
Abstract
Tumor immunotherapy, as the focus of scientific research and clinical tumor treatment in recent years, has received extensive attention. Due to its remarkable curative effect and fewer side effects than traditional treatments, it has significant clinical benefits for the treatment of various advanced cancers and can improve cancer patient survival in the long term. Currently, most patients cannot benefit from immunotherapy, and some patients may experience tumor recurrence and drug resistance even if they achieve remission overcome. Numerous studies have shown that the abnormal angiogenesis state of tumors can lead to immunosuppressive tumor microenvironment, which affects the efficacy of immunotherapy. Actually, to improve the efficacy of immunotherapy, the application of anti-angiogenesis drugs to normalize abnormal tumor vessel has been widely confirmed in basic and clinical research. This review not only discusses the risk factors, mechanisms, and effects of abnormal and normalized tumor angiogenesis state on the immune environment, but summarizes the latest progress of immunotherapy combined with anti-angiogenic therapy. We hope this review provides an applied reference for anti-angiogenesis drugs and synergistic immunotherapy therapy.
Collapse
Affiliation(s)
- Jingyao Tu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hang Liang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chunya Li
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yongbiao Huang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ziqi Wang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinyi Chen
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
9
|
Barnett SD, Asif H, Buxton ILO. Novel identification and modulation of the mechanosensitive Piezo1 channel in human myometrium. J Physiol 2023; 601:1675-1690. [PMID: 35941750 PMCID: PMC9905381 DOI: 10.1113/jp283299] [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: 05/05/2022] [Accepted: 06/14/2022] [Indexed: 11/08/2022] Open
Abstract
Approximately 10% of US births deliver preterm before 37 weeks of completed gestation. Premature infants are at risk for life-long debilitating morbidities and death, and spontaneous preterm labour explains 50% of preterm births. In all cases existing treatments are ineffective, and none are FDA approved. The mechanisms that initiate preterm labour are not well understood but may result from dysfunctional regulation of quiescence mechanisms. Human pregnancy is accompanied by large increases in blood flow, and the uterus must enlarge by orders of magnitude to accommodate the growing fetus. This mechanical strain suggests that stretch-activated channels may constitute a mechanism to explain gestational quiescence. Here we identify for the first time that Piezo1, a mechanosensitive cation channel, is present in the uterine smooth muscle and microvascular endothelium of pregnant myometrium. Piezo is downregulated during preterm labour, and stimulation of myometrial Piezo1 in an organ bath with the agonist Yoda1 relaxes the tissue in a dose-dependent fashion. Further, stimulation of Piezo1 while inhibiting protein kinase A, AKT, or endothelial nitric oxide synthase mutes the negative inotropic effects of Piezo1 activation, intimating that actions on the myocyte and endothelial nitric oxide signalling contribute to Piezo1-mediated contractile dynamics. Taken together, these data highlight the importance of stretch-activated channels in pregnancy maintenance and parturition, and identify Piezo1 as a tocolytic target of interest. KEY POINTS: Spontaneous preterm labour is a serious obstetric dilemma without a known cause or effective treatments. Piezo1 is a stretch-activated channel important to muscle contractile dynamics. Piezo1 is present in the myometrium and is dysregulated in women who experience preterm labour. Activation of Piezo1 by the agonist Yoda1 relaxes the myometrium in a dose-dependent fashion, indicating that Piezo1 modulation may have therapeutic benefits to treat preterm labour.
Collapse
Affiliation(s)
- Scott D Barnett
- Department of Pharmacology, Center for Molecular Medicine, Reno School of Medicine, University of Nevada, Reno, NV, USA
| | - Hazik Asif
- Department of Pharmacology, Center for Molecular Medicine, Reno School of Medicine, University of Nevada, Reno, NV, USA
| | - Iain L O Buxton
- Department of Pharmacology, Center for Molecular Medicine, Reno School of Medicine, University of Nevada, Reno, NV, USA
| |
Collapse
|
10
|
Development of L-Lysine-Loaded PLGA Microparticles as a Controlled Release System for Angiogenesis Enhancement. Pharmaceutics 2023; 15:pharmaceutics15020479. [PMID: 36839801 PMCID: PMC9961840 DOI: 10.3390/pharmaceutics15020479] [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: 12/12/2022] [Revised: 01/24/2023] [Accepted: 01/28/2023] [Indexed: 02/04/2023] Open
Abstract
Vascularization is a highly conserved and considerably complex and precise process that is finely driven by endogenous regulatory processes at the tissue and systemic levels. However, it can reveal itself to be slow and inadequate for tissue repair and regeneration consequent to severe lesions/damages. Several biomaterial-based strategies were developed to support and enhance vasculogenesis by supplying pro-angiogenic agents. Several approaches were adopted to develop effective drug delivery systems for the controlled release of a huge variety of compounds. In this work, a microparticulate system was chosen to be loaded with the essential amino acid L-lysine, a molecule that has recently gained interest due to its involvement in pro-angiogenic, pro-regenerative, and anti-inflammatory mechanisms. Poly (lactic-co-glycolic acid), the most widely used FDA-approved biodegradable synthetic polymer for the development of drug delivery systems, was chosen due to its versatility and ability to promote neovascularization and wound healing. This study dealt with the development and the effectiveness evaluation of a PLGA-based microparticulate system for the controlled release of L-lysine. Therefore, in order to maximize L-lysine encapsulation efficiency and tune its release kinetics, the microparticle synthesis protocol was optimized by varying some processing parameters. All developed formulations were characterized from a morphological and physicochemical point of view. The optimized formulation was further characterized via the evaluation of its preliminary biological efficacy in vitro. The cellular and molecular studies revealed that the L-lysine-loaded PLGA microparticles were non-toxic, biocompatible, and supported cell proliferation and angiogenesis well by stimulating the expression of pro-angiogenic genes such as metalloproteinase-9, focal adhesion kinases, and different growth factors. Thus, this work showed the potential of delivering L-lysine encapsulated in PLGA microparticles as a cost-effective promoter system for angiogenesis enhancement and rapid healing.
Collapse
|
11
|
Zhang Y, Jing M, Cai C, Zhu S, Zhang C, Wang Q, Zhai Y, Ji X, Wu D. Role of hydrogen sulphide in physiological and pathological angiogenesis. Cell Prolif 2022; 56:e13374. [PMID: 36478328 PMCID: PMC9977675 DOI: 10.1111/cpr.13374] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 11/08/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
The role of hydrogen sulphide (H2 S) in angiogenesis has been widely demonstrated. Vascular endothelial growth factor (VEGF) plays an important role in H2 S-induced angiogenesis. H2 S promotes angiogenesis by upregulating VEGF via pro-angiogenic signal transduction. The involved signalling pathways include the mitogen-activated protein kinase pathway, phosphoinositide-3 kinase pathway, nitric oxide (NO) synthase/NO pathway, signal transducer and activator of transcription 3 (STAT3) pathway, and adenosine triphosphate (ATP)-sensitive potassium (KATP ) channels. H2 S has been shown to contribute to tumour angiogenesis, diabetic wound healing, angiogenesis in cardiac and cerebral ischaemic tissues, and physiological angiogenesis during the menstrual cycle and pregnancy. Furthermore, H2 S can exert an anti-angiogenic effect by inactivating Wnt/β-catenin signalling or blocking the STAT3 pathway in tumours. Therefore, H2 S plays a double-edged sword role in the process of angiogenesis. The regulation of H2 S production is a promising therapeutic approach for angiogenesis-associated diseases. Novel H2 S donors and/or inhibitors can be developed in the treatment of angiogenesis-dependent diseases.
Collapse
Affiliation(s)
- Yan‐Xia Zhang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical SciencesHenan UniversityKaifengHenanChina,Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular MedicineHenan UniversityKaifengHenanChina
| | - Mi‐Rong Jing
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical SciencesHenan UniversityKaifengHenanChina,Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular MedicineHenan UniversityKaifengHenanChina
| | - Chun‐Bo Cai
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical SciencesHenan UniversityKaifengHenanChina,Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular MedicineHenan UniversityKaifengHenanChina
| | - Shuai‐Gang Zhu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical SciencesHenan UniversityKaifengHenanChina,Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular MedicineHenan UniversityKaifengHenanChina
| | - Chao‐Jing Zhang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical SciencesHenan UniversityKaifengHenanChina,Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular MedicineHenan UniversityKaifengHenanChina
| | - Qi‐Meng Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical SciencesHenan UniversityKaifengHenanChina,Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular MedicineHenan UniversityKaifengHenanChina
| | - Yuan‐Kun Zhai
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical SciencesHenan UniversityKaifengHenanChina,School of StomatologyHenan UniversityKaifengHenanChina
| | - Xin‐Ying Ji
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical SciencesHenan UniversityKaifengHenanChina,Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular MedicineHenan UniversityKaifengHenanChina,Kaifeng Key Laboratory of Infection and Biological Safety, School of Basic Medical SciencesHenan UniversityKaifengHenanChina
| | - Dong‐Dong Wu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical SciencesHenan UniversityKaifengHenanChina,Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular MedicineHenan UniversityKaifengHenanChina,School of StomatologyHenan UniversityKaifengHenanChina
| |
Collapse
|
12
|
Nakayama A, Roquid KA, Iring A, Strilic B, Günther S, Chen M, Weinstein LS, Offermanns S. Suppression of CCL2 angiocrine function by adrenomedullin promotes tumor growth. J Exp Med 2022; 220:213682. [PMID: 36374225 PMCID: PMC9665902 DOI: 10.1084/jem.20211628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/19/2022] [Accepted: 10/13/2022] [Indexed: 11/16/2022] Open
Abstract
Within the tumor microenvironment, tumor cells and endothelial cells regulate each other. While tumor cells induce angiogenic responses in endothelial cells, endothelial cells release angiocrine factors, which act on tumor cells and other stromal cells. We report that tumor cell-derived adrenomedullin has a pro-angiogenic as well as a direct tumor-promoting effect, and that endothelium-derived CC chemokine ligand 2 (CCL2) suppresses adrenomedullin-induced tumor cell proliferation. Loss of the endothelial adrenomedullin receptor CALCRL or of the G-protein Gs reduced endothelial proliferation. Surprisingly, tumor cell proliferation was also reduced after endothelial deletion of CALCRL or Gs. We identified CCL2 as a critical angiocrine factor whose formation is inhibited by adrenomedullin. Furthermore, CCL2 inhibited adrenomedullin formation in tumor cells through its receptor CCR2. Consistently, loss of endothelial CCL2 or tumor cell CCR2 normalized the reduced tumor growth seen in mice lacking endothelial CALCRL or Gs. Our findings show tumor-promoting roles of adrenomedullin and identify CCL2 as an angiocrine factor controlling adrenomedullin formation by tumor cells.
Collapse
Affiliation(s)
- Akiko Nakayama
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany,Correspondence to Akiko Nakayama:
| | - Kenneth Anthony Roquid
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - András Iring
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Boris Strilic
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Stefan Günther
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Min Chen
- Metabolic Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MA
| | - Lee S. Weinstein
- Metabolic Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MA
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany,Center for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany,Cardiopulmonary Institute, Bad Nauheim, Germany,German Center for Cardiovascular Research, Bad Nauheim, Germany,Stefan Offermanns:
| |
Collapse
|
13
|
Schley G, Goppelt-Struebe M. A PHD inhibitor prevents changes in the phosphoproteome and capillary rarefaction by CsA: treatment option for CKD? Kidney Int 2022; 102:686-688. [PMID: 36150758 DOI: 10.1016/j.kint.2022.06.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/08/2022] [Accepted: 06/10/2022] [Indexed: 11/24/2022]
Abstract
Labes et al. analyze the phosphoproteome in a mouse model of chronic cyclosporine A nephrotoxicity and detect significant changes in the angiogenic pathway. Furthermore, they observe reduced hemoglobin levels and capillary rarefaction in the kidney. The authors show that coadministration of the hypoxia-inducible factor prolyl hydroxylase inhibitor daprodustat almost completely prevents changes of the phosphoproteome and capillary rarefaction, suggesting that prolyl hydroxylase domain enzyme inhibitors may preserve microvasculature of the kidney, which is commonly impaired in chronic kidney disease.
Collapse
Affiliation(s)
- Gunnar Schley
- Department of Nephrology and Hypertension, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Margarete Goppelt-Struebe
- Department of Nephrology and Hypertension, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
14
|
The spectrin cytoskeleton integrates endothelial mechanoresponses. Nat Cell Biol 2022; 24:1226-1238. [PMID: 35817960 DOI: 10.1038/s41556-022-00953-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 06/01/2022] [Indexed: 12/13/2022]
Abstract
Physiological blood flow induces the secretion of vasoactive compounds, notably nitric oxide, and promotes endothelial cell elongation and reorientation parallel to the direction of applied shear. How shear is sensed and relayed to intracellular effectors is incompletely understood. Here, we demonstrate that an apical spectrin network is essential to convey the force imposed by shear to endothelial mechanosensors. By anchoring CD44, spectrins modulate the cell surface density of hyaluronan and sense and translate shear into changes in plasma membrane tension. Spectrins also regulate the stability of apical caveolae, where the mechanosensitive PIEZO1 channels are thought to reside. Accordingly, shear-induced PIEZO1 activation and the associated calcium influx were absent in spectrin-deficient cells. As a result, cell realignment and flow-induced endothelial nitric oxide synthase stimulation were similarly dependent on spectrin. We conclude that the apical spectrin network is not only required for shear sensing but also transmits and distributes the resulting tensile forces to mechanosensors that elicit protective and vasoactive responses.
Collapse
|
15
|
Zhou Z, Chai W, Liu Y, Liu Y, Pan H, Wu Q, Zhang X. Connexin 43 overexpression induces lung cancer angiogenesis in vitro following phosphorylation at Ser279 in its C‑terminus. Oncol Lett 2022; 24:293. [PMID: 35949588 PMCID: PMC9353244 DOI: 10.3892/ol.2022.13413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 04/27/2022] [Indexed: 12/24/2022] Open
Abstract
Blocking angiogenesis can inhibit tumor growth and metastasis. However, the mechanism underlying regulation of lung cancer angiogenesis remains unclear. The gap junction protein connexin 43 (Cx43) is implicated in angiogenesis. The aim of the present study was to determine the role of Cx43 in angiogenesis in vitro and its signaling pathways. Human pulmonary microvascular endothelial cells were transfected with Cx43-targeting siRNA or Cx43-overexpressing recombinant plasmid vector. Reverse transcription-quantitative polymerase chain reaction and western blotting were performed to determine Cx43, zonula occludens-1 (ZO-1), E-cadherin, β-catenin, von Willebrand factor (vWF), and plasminogen activator inhibitor-1 (PAI-1) mRNA and protein expression levels, respectively. Tyr265, Ser279, Ser368, and Ser373 phosphorylation levels in the C-terminus of Cx43 and intracellular and membranal Cx43 contents were determined using western blotting. Additionally, immunofluorescence, tube formation, Cell Counting Kit-8, and Transwell migration assays were performed. The results revealed that compared with that in the control samples, Cx43, ZO-1, E-cadherin, β-catenin, vWF, and PAI-1 mRNA and protein expression were significantly increased in the Cx43 overexpression group and significantly decreased in the Cx43-knockdown group. Moreover, the phosphorylation level of Ser279 as well as cell proliferation and migration rates were markedly increased in the Cx43 overexpression group, and tube formation revealed that the potential of angiogenesis was also increased. Conversely, in the Cx43-knockdown group, the phosphorylation level of Ser279 and cell proliferation and migration rates were reduced, and the potential of angiogenesis was greatly impaired. Under Cx43 overexpression, membranal Cx43 content was significantly increased, whereas under Cx43 knockdown, it was significantly reduced. Therefore, Cx43 overexpression could induce pulmonary angiogenesis in vitro by promoting cell proliferation and migration and activating ZO-1, E-cadherin, β-catenin, vWF, and PAI-1. This may be achieved by promoting phosphorylation and activation of the intracellular signal site Ser279 at the C-terminus of Cx43.
Collapse
Affiliation(s)
- Zizi Zhou
- Department of Cardio‑Thoracic Surgery, Shenzhen University General Hospital, Shenzhen, Guangdong 518055, P.R. China
| | - Wenxiang Chai
- Department of Cardio‑Thoracic Surgery, Shenzhen University General Hospital, Shenzhen, Guangdong 518055, P.R. China
| | - Yi Liu
- Department of Cardio‑Thoracic Surgery, Shenzhen University General Hospital, Shenzhen, Guangdong 518055, P.R. China
| | - Yao Liu
- Department of Cardio‑Thoracic Surgery, Shenzhen University General Hospital, Shenzhen, Guangdong 518055, P.R. China
| | - Huiyu Pan
- Department of Cardio‑Thoracic Surgery, Shenzhen University General Hospital, Shenzhen, Guangdong 518055, P.R. China
| | - Qiang Wu
- Department of Cardio‑Thoracic Surgery, Shenzhen University General Hospital, Shenzhen, Guangdong 518055, P.R. China
| | - Xiaoming Zhang
- Department of Cardio‑Thoracic Surgery, Shenzhen University General Hospital, Shenzhen, Guangdong 518055, P.R. China
| |
Collapse
|
16
|
Labes R, Brinkmann L, Kulow VA, Roegner K, Mathia S, Balcerek B, Persson PB, Rosenberger C, Fähling M. Daprodustat prevents Cyclosporine-A mediated anemia and peritubular capillary loss. Kidney Int 2022; 102:750-765. [DOI: 10.1016/j.kint.2022.04.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 04/06/2022] [Accepted: 04/18/2022] [Indexed: 12/24/2022]
|
17
|
Ma X, Ji C. Remote Ischemic Conditioning: A Potential Treatment for Chronic Cerebral Hypoperfusion. Eur Neurol 2022; 85:253-259. [PMID: 35104816 DOI: 10.1159/000521803] [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: 06/03/2021] [Accepted: 12/19/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND Chronic cerebral hypoperfusion (CCH) is a clinical syndrome, which is characterized by significantly decreased cerebral blood flow (CBF). CCH is a common consequence of cerebrovascular and cardiovascular diseases and the elderly. CCH results in a series of pathological damages, increasing cell death, autophagy dysfunction, amyloid β (Aβ) peptide accumulation, blood-brain barrier (BBB) disruption, and endothelial damage, which are found in CCH models. In addition, CCH is a prominent risk factor of cognitive impairment, such as vascular dementia, and CCH contributes to the occurrence and development of Alzheimer's disease. Therefore, the treatment of patients with CCH is of great value. It has been confirmed that remote ischemic conditioning (RIC) is a safe, promising treatment for acute and chronic cerebrovascular diseases. RIC significantly increases CBF in both CCH models and patients, inhibits neuronal apoptosis, reduces Aβ deposition, protects BBB integrity and endothelial function, alleviates neuroinflammation, improves cognitive impairment, and exerts neuroprotection. SUMMARY With the development of animal models, the pathophysiological mechanisms of CCH and RIC are increasingly revealed. Key Messages: We discuss the mechanisms related to hypoperfusion in the brain and explore the potential treatment of RIC for CCH to promote its transformation and application in humans.
Collapse
Affiliation(s)
- Xiao Ma
- Department of General Practice Medicine, Dalian Municipal Central Hospital, Dalian, China
| | - Chenhua Ji
- Department of General Practice Medicine, Dalian Municipal Central Hospital, Dalian, China
| |
Collapse
|
18
|
Systematic Understanding of Mechanism of Danggui Shaoyao San against Ischemic Stroke Using a Network Pharmacology Approach. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:3747285. [PMID: 35035503 PMCID: PMC8754614 DOI: 10.1155/2022/3747285] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 12/07/2021] [Indexed: 11/30/2022]
Abstract
Purpose Danggui Shaoyao San (DSS) was developed to treat the ischemic stroke (IS) in patients and animal models. The purpose of this study was to explore its active compounds and demonstrate its mechanism against IS through network pharmacology, molecular docking, and animal experiment. Methods All the components of DSS were retrieved from the pharmacology database of TCM system. The genes corresponding to the targets were retrieved using OMIM, CTD database, and TTD database. The herb-compound-target network was constructed by Cytoscape software. The target protein-protein interaction network was built using the STRING database. The core targets of DSS were analyzed by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). Then, we achieved molecular docking between the hub proteins and the key active compounds. Finally, animal experiments were performed to verify the core targets. Triphenyltetrazolium chloride (TTC) staining was used to calculate the infarct size in mice. The protein expression was determined using the Western blot. Results Compound-target network mainly contained 51 compounds and 315 corresponding targets. Key targets contained MAPK1, SRC, PIK3R1, HRAS, AKT1, RHOA, RAC1, HSP90AA1, and RXRA FN1. There were 417 GO items in GO enrichment analysis (p < 0.05) and 119 signaling pathways (p < 0.05) in KEGG, mainly including negative regulation of apoptosis, steroid hormone-mediated signaling pathway, neutrophil activation, cellular response to oxidative stress, and VEGF signaling pathway. MAPK1, SRC, and PIK3R1 docked with small molecule compounds. According to the Western blot, the expression of p-MAPK 1, p-AKT, and p-SRC was regulated by DSS. Conclusions This study showed that DSS can treat IS through multiple targets and routes and provided new insights to explore the mechanisms of DSS against IS.
Collapse
|
19
|
Sobrinho JEM, Aguiar MT, Machado LC, Carlos ACAM, Nunes Alves APN, Mesquita KC, de Miranda Candeiro GT, de Barros Silva PG. Intense orthodontic forces induces the three dental pulp nitric oxide synthases isoforms and leads to orofacial discomfort in rats. Orthod Craniofac Res 2021; 25:485-493. [DOI: 10.1111/ocr.12560] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/13/2021] [Accepted: 12/20/2021] [Indexed: 12/01/2022]
Affiliation(s)
| | - Maria Thaynara Aguiar
- Christus University Center Department of Dentistry Laboratory of Pathology Fortaleza, Ceará Brazil
| | - Larissa Carvalho Machado
- Christus University Center Department of Dentistry Laboratory of Pathology Fortaleza, Ceará Brazil
| | | | | | - Karine Cestaro Mesquita
- Christus University Center Department of Dentistry Laboratory of Pathology Fortaleza, Ceará Brazil
| | | | | |
Collapse
|
20
|
Nguyen H, Koh JY, Li H, Islas-Robles A, Meda Venkata SP, Wang JM, Monks TJ. A novel imidazolinone metformin-methylglyoxal metabolite promotes endothelial cell angiogenesis via the eNOS/HIF-1α pathway. FASEB J 2021; 35:e21645. [PMID: 34105824 PMCID: PMC8237315 DOI: 10.1096/fj.202002674rr] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 04/13/2021] [Accepted: 04/20/2021] [Indexed: 12/28/2022]
Abstract
Peripheral arterial disease (PAD) is one of the major complications of diabetes due to an impairment in angiogenesis. Since there is currently no drug with satisfactory efficacy to enhance blood vessel formation, discovering therapies to improve angiogenesis is critical. An imidazolinone metabolite of the metformin‐methylglyoxal scavenging reaction, (E)‐1,1‐dimethyl‐2‐(5‐methyl‐4‐oxo‐4,5‐dihydro‐1H‐imidazol‐2‐yl) guanidine (IMZ), was recently characterized and identified in the urine of type‐2 diabetic patients. Here, we report the pro‐angiogenesis effect of IMZ (increased aortic sprouting, cell migration, network formation, and upregulated multiple pro‐angiogenic factors) in human umbilical vein endothelial cells. Using genetic and pharmacological approaches, we showed that IMZ augmented angiogenesis by activating the endothelial nitric oxide synthase (eNOS)/hypoxia‐inducible factor‐1 alpha (HIF‐1α) pathway. Furthermore, IMZ significantly promoted capillary density in the in vivo Matrigel plug angiogenesis model. Finally, the role of IMZ in post‐ischemic angiogenesis was examined in a chronic hyperglycemia mouse model subjected to hind limb ischemia. We observed improved blood perfusion, increased capillary density, and reduced tissue necrosis in mice receiving IMZ compared to control mice. Our data demonstrate the pro‐angiogenic effects of IMZ, its underlying mechanism, and provides a structural basis for the development of potential pro‐angiogenic agents for the treatment of PAD.
Collapse
Affiliation(s)
- Huong Nguyen
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
| | - Jia Yi Koh
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
| | - Hainan Li
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
| | | | - Sai Pranathi Meda Venkata
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
| | - Jie-Mei Wang
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA.,Centers for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
| | - Terrence J Monks
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
| |
Collapse
|
21
|
Yun W, Dan W, Liu J, Guo X, Li M, He Q. Investigation of the Mechanism of Traditional Chinese Medicines in Angiogenesis through Network Pharmacology and Data Mining. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:5539970. [PMID: 34007289 PMCID: PMC8102115 DOI: 10.1155/2021/5539970] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/20/2021] [Accepted: 04/15/2021] [Indexed: 12/11/2022]
Abstract
Although traditional Chinese medicine is effective and safe for the treatment of angiogenesis, the in vivo intervention mechanism is diverse, complex, and largely unknown. Therefore, we aimed to explore the active ingredients of traditional Chinese medicine and their mechanisms of action against angiogenesis. Data on angiogenesis-related targets were collected from GeneCards, Therapeutic Target Database, Online Mendelian Inheritance in Man, DrugBank, and DisGeNET. These were matched to related molecular compounds and ingredients in the traditional Chinese medicine system pharmacology platform. The data were integrated and based on the condition of degree > 1, and relevant literature, target-compound, compound-medicine, and target-compound-medicine networks were constructed using Cytoscape. Molecular docking was used to predict the predominant binding combination of core targets and components. We obtained 79 targets for angiogenesis; 41 targets were matched to 3839 compounds, of which 110 compounds were selected owing to their high correlation with angiogenesis. Fifty-five combinations in the network were obtained by molecular docking, among which PTGS2-astragalin (-9.18 kcal/mol), KDR-astragalin (-7.94 kcal/mol), PTGS2-quercetin (-7.41 kcal/mol), and PTGS2-myricetin (-7.21 kcal/mol) were top. These results indicated that the selected potential core compounds have good binding activity with the core targets. Eighty new combinations were obtained from the network, and the top combinations based on affinity were KDR-beta-carotene (-10.13 kcal/mol), MMP9-beta-sitosterol (-8.04 kcal/mol), MMP9-astragalin (-7.82 kcal/mol), and MMP9-diosgenin (-7.51 kcal/mol). The core targets included PTGS2, KDR, VEGFA, and MMP9. The essential components identified were astragalin, kaempferol, myricetin, quercetin, and β-sitosterol. The crucial Chinese medicines identified included Polygoni Cuspidati Rhizoma et Radix, Morus alba Root Bark, and Forsythiae Fructus. By systematically analysing the ingredients of traditional Chinese medicine and their targets, it is possible to determine their potential mechanisms of action against pathological angiogenesis. Our study provides a basis for further research and the development of new therapeutics for angiogenesis.
Collapse
Affiliation(s)
- Wingyan Yun
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
- Graduate School of Beijing University of Chinese Medicine, Beijing 100029, China
| | - Wenchao Dan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
- Graduate School of Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jinlei Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Xinyuan Guo
- Cancer Hospital Chinese Academy of Medical Sciences, Beijing 100021, China
| | - Min Li
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Qingyong He
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| |
Collapse
|
22
|
Zhu HY, Hong FF, Yang SL. The Roles of Nitric Oxide Synthase/Nitric Oxide Pathway in the Pathology of Vascular Dementia and Related Therapeutic Approaches. Int J Mol Sci 2021; 22:ijms22094540. [PMID: 33926146 PMCID: PMC8123648 DOI: 10.3390/ijms22094540] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/18/2021] [Accepted: 04/21/2021] [Indexed: 12/16/2022] Open
Abstract
Vascular dementia (VaD) is the second most common form of dementia worldwide. It is caused by cerebrovascular disease, and patients often show severe impairments of advanced cognitive abilities. Nitric oxide synthase (NOS) and nitric oxide (NO) play vital roles in the pathogenesis of VaD. The functions of NO are determined by its concentration and bioavailability, which are regulated by NOS activity. The activities of different NOS subtypes in the brain are partitioned. Pathologically, endothelial NOS is inactivated, which causes insufficient NO production and aggravates oxidative stress before inducing cerebrovascular endothelial dysfunction, while neuronal NOS is overactive and can produce excessive NO to cause neurotoxicity. Meanwhile, inflammation stimulates the massive expression of inducible NOS, which also produces excessive NO and then induces neuroinflammation. The vicious circle of these kinds of damage having impacts on each other finally leads to VaD. This review summarizes the roles of the NOS/NO pathway in the pathology of VaD and also proposes some potential therapeutic methods that target this pathway in the hope of inspiring novel ideas for VaD therapeutic approaches.
Collapse
Affiliation(s)
- Han-Yan Zhu
- Department of Physiology, College of Medicine, Nanchang University, 461 Bayi Avenue, Nanchang 330006, China;
- Queen Marry College, College of Medicine, Nanchang University, 461 Bayi Avenue, Nanchang 330006, China
| | - Fen-Fang Hong
- Teaching Center, Department of Experimental, Nanchang University, 461 Bayi Avenue, Nanchang 330006, China
- Correspondence: (F.-F.H.); (S.-L.Y.)
| | - Shu-Long Yang
- Department of Physiology, College of Medicine, Nanchang University, 461 Bayi Avenue, Nanchang 330006, China;
- Correspondence: (F.-F.H.); (S.-L.Y.)
| |
Collapse
|
23
|
Roy B, Palaniyandi SS. A role for aldehyde dehydrogenase (ALDH) 2 in angiotensin II-mediated decrease in angiogenesis of coronary endothelial cells. Microvasc Res 2021; 135:104133. [PMID: 33428883 DOI: 10.1016/j.mvr.2021.104133] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 01/06/2021] [Accepted: 01/06/2021] [Indexed: 11/17/2022]
Abstract
Diabetes-induced coronary endothelial cell (CEC) dysfunction contributes to diabetic heart diseases. Angiotensin II (Ang II), a vasoactive hormone, is upregulated in diabetes, and is reported to increase oxidative stress in CECs. 4-hydroxy-2-nonenal (4HNE), a key lipid peroxidation product, causes cellular dysfunction by forming adducts with proteins. By detoxifying 4HNE, aldehyde dehydrogenase (ALDH) 2 reduces 4HNE mediated proteotoxicity and confers cytoprotection. Thus, we hypothesize that ALDH2 improves Ang II-mediated defective CEC angiogenesis by decreasing 4HNE-mediated cytotoxicity. To test our hypothesis, we treated the cultured mouse CECs (MCECs) with Ang II (0.1, 1 and 10 μM) for 2, 4 and 6 h. Next, we treated MCECs with Alda-1 (10 μM), an ALDH2 activator or disulfiram (2.5 μM)/ALDH2 siRNA (1.25 nM), the ALDH2 inhibitors, or blockers of angiotensin II type-1 and 2 receptors i.e. Losartan and PD0123319 respectively before challenging MCECs with 10 μM Ang II. We found that 10 μM Ang II decreased tube formation in MCECs with in vitro angiogenesis assay (P < .0005 vs control). 10 μM Ang II downregulated the levels of vascular endothelial growth factor receptor 1 (VEGFR1) (p < .005 for mRNA and P < .05 for protein) and VEGFR2 (p < .05 for mRNA and P < .005 for protein) as well as upregulated the levels of angiotensin II type-2 receptor (AT2R) (p < .05 for mRNA and P < .005 for protein) and 4HNE-adducts (P < .05 for protein) in cultured MCECs, compared to controls. ALDH2 inhibition with disulfiram/ALDH2 siRNA exacerbated 10 μM Ang II-induced decrease in coronary angiogenesis (P < .005) by decreasing the levels of VEGFR1 (P < .005 for mRNA and P < .05 for protein) and VEGFR2 (P < .05 for both mRNA and protein) and increasing the levels of AT2R (P < .05 for both mRNA and protein) and 4HNE-adducts (P < .05 for protein) relative to Ang II alone. AT2R inhibition per se improved angiogenesis in MCECs. Additionally, enhancing ALDH2 activity with Alda 1 rescued Ang II-induced decrease in angiogenesis by increasing the levels of VEGFR1, VEGFR2 and decreasing the levels of AT2R. In summary, ALDH2 can be an important target in reducing 4HNE-induced proteotoxicity and improving angiogenesis in MCECs. Finally, we conclude ALDH2 activation can be a therapeutic strategy to improve coronary angiogenesis to ameliorate cardiometabolic diseases.
Collapse
Affiliation(s)
- Bipradas Roy
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Health System, Detroit, MI 48202, United States of America; Department of Physiology, Wayne State University, Detroit, MI 48202, United States of America
| | - Suresh Selvaraj Palaniyandi
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Health System, Detroit, MI 48202, United States of America; Department of Physiology, Wayne State University, Detroit, MI 48202, United States of America.
| |
Collapse
|
24
|
Co-Culture of Primary Human Coronary Artery and Internal Thoracic Artery Endothelial Cells Results in Mutually Beneficial Paracrine Interactions. Int J Mol Sci 2020; 21:ijms21218032. [PMID: 33126651 PMCID: PMC7663246 DOI: 10.3390/ijms21218032] [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: 09/24/2020] [Revised: 10/21/2020] [Accepted: 10/27/2020] [Indexed: 12/12/2022] Open
Abstract
Although saphenous veins (SVs) are commonly used as conduits for coronary artery bypass grafting (CABG), internal thoracic artery (ITA) grafts have significantly higher long-term patency. As SVs and ITA endothelial cells (ECs) have a considerable level of heterogeneity, we suggested that synergistic paracrine interactions between CA and ITA ECs (HCAECs and HITAECs, respectively) may explain the increased resistance of ITA grafts and adjacent CAs to atherosclerosis and restenosis. In this study, we measured the gene and protein expression of the molecules responsible for endothelial homeostasis, pro-inflammatory response, and endothelial-to-mesenchymal transition in HCAECs co-cultured with either HITAECs or SV ECs (HSaVECs) for an ascending duration. Upon the co-culture, HCAECs and HITAECs showed augmented expression of endothelial nitric oxide synthase (eNOS) and reduced expression of endothelial-to-mesenchymal transition transcription factors Snail and Slug when compared to the HCAEC–HSaVEC model. HCAECs co-cultured with HITAECs demonstrated an upregulation of HES1, a master regulator of arterial specification, of which the expression was also exclusively induced in HSaVECs co-cultured with HCAECs, suggestive of their arterialisation. In addition, co-culture of HCAECs and HITAECs promoted the release of pro-angiogenic molecules. To conclude, co-culture of HCAECs and HITAECs results in reciprocal and beneficial paracrine interactions that might contribute to the better performance of ITA grafts upon CABG.
Collapse
|
25
|
McKay EC, Counts SE. Oxytocin Receptor Signaling in Vascular Function and Stroke. Front Neurosci 2020; 14:574499. [PMID: 33071746 PMCID: PMC7544744 DOI: 10.3389/fnins.2020.574499] [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: 06/22/2020] [Accepted: 08/31/2020] [Indexed: 12/13/2022] Open
Abstract
The oxytocin receptor (OXTR) is a G protein-coupled receptor with a diverse repertoire of intracellular signaling pathways, which are activated in response to binding oxytocin (OXT) and a similar nonapeptide, vasopressin. This review summarizes the cell and molecular biology of the OXTR and its downstream signaling cascades, particularly focusing on the vasoactive functions of OXTR signaling in humans and animal models, as well as the clinical applications of OXTR targeting cerebrovascular accidents.
Collapse
Affiliation(s)
- Erin C McKay
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States.,Neuroscience Program, Michigan State University, East Lansing, MI, United States
| | - Scott E Counts
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States.,Neuroscience Program, Michigan State University, East Lansing, MI, United States.,Department of Family Medicine, Michigan State University, Grand Rapids, MI, United States.,Hauenstein Neurosciences Center, Mercy Health Saint Mary's Hospital, Grand Rapids, MI, United States.,Michigan Alzheimer's Disease Research Center, Ann Arbor, MI, United States
| |
Collapse
|
26
|
Ullah M, Liu DD, Rai S, Razavi M, Concepcion W, Thakor AS. Pulsed focused ultrasound enhances the therapeutic effect of mesenchymal stromal cell-derived extracellular vesicles in acute kidney injury. Stem Cell Res Ther 2020; 11:398. [PMID: 32928310 PMCID: PMC7490886 DOI: 10.1186/s13287-020-01922-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/24/2020] [Accepted: 09/02/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Acute kidney injury (AKI) is characterized by rapid failure of renal function and has no curative therapies. Mesenchymal stromal cell (MSC)-derived extracellular vesicles (EVs) are known to carry therapeutic factors, which have shown promise in regenerative medicine applications, including AKI. However, there remains an unmet need to optimize their therapeutic effect. One potential avenue of optimization lies in pulsed focused ultrasound (pFUS), where tissues-of-interest are treated with sound waves. pFUS has been shown to enhance MSC therapy via increased cell homing, but its effects on cell-free EV therapy remain largely unexplored. METHODS We combine pFUS pretreatment of the kidney with MSC-derived EV therapy in a mouse model of cisplatin-induced AKI. RESULTS EVs significantly improved kidney function, reduced injury markers, mediated increased proliferation, and reduced inflammation and apoptosis. While pFUS did not enhance EV homing to the kidney, the combined treatment resulted in a superior therapeutic effect compared to either treatment alone. We identified several molecular mechanisms underlying this synergistic therapeutic effect, including upregulation of proliferative signaling (MAPK/ERK, PI3K/Akt) and regenerative pathways (eNOS, SIRT3), as well as suppression of inflammation. CONCLUSION Taken together, pFUS may be a strategy for enhancing the therapeutic efficacy of MSC-derived EV treatment for the treatment of AKI.
Collapse
Affiliation(s)
- Mujib Ullah
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University, Palo Alto, CA, 94304, USA
| | - Daniel D Liu
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University, Palo Alto, CA, 94304, USA
| | - Sravanthi Rai
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University, Palo Alto, CA, 94304, USA
| | - Mehdi Razavi
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University, Palo Alto, CA, 94304, USA
| | - Waldo Concepcion
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University, Palo Alto, CA, 94304, USA
| | - Avnesh S Thakor
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University, Palo Alto, CA, 94304, USA.
| |
Collapse
|
27
|
Beghi S, Cavaliere F, Buschini A. Gene polymorphisms in calcium-calmodulin pathway: Focus on cardiovascular disease. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2020; 786:108325. [PMID: 33339582 DOI: 10.1016/j.mrrev.2020.108325] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 07/22/2020] [Accepted: 07/24/2020] [Indexed: 12/30/2022]
Abstract
Cardiovascular disease is the leading cause of death in industrialized countries and affects an increasing number of people. Several risk factors play an important role in the etiology of this disease, such as an unhealthy lifestyle. It is increasingly clear that genetic factors influencing the molecular basis of excitation-contraction mechanisms in the heart could contribute to modify the individual's risk. Thanks to the progress that has been made in understanding calcium signaling in the heart, it is assumed that calmodulin can play a crucial role in the excitation-contraction coupling. In fact, calmodulin (CaM) binds calcium and consequently regulates calcium channels. Several works show how some polymorphic variants can be considered predisposing factors to complex pathologies. Therefore, we hypothesize that the identification of polymorphic variants of proteins involved in the CaM pathway could be important for understanding how genetic traits can influence predisposition to myocardial infarction. This review considers each pathway of the three different isoforms of calmodulin (CaM1; CaM2; CaM3) and focuses on some common proteins involved in the three pathways, with the aim of analyzing the polymorphisms studied in the literature and understanding if they are associated with cardiovascular disease.
Collapse
Affiliation(s)
- Sofia Beghi
- University of Parma, Department of Chemistry, Life Sciences and Environmental Sustainability, Parco Area Delle Scienze 11A, 43124, Parma, Italy
| | - Francesca Cavaliere
- University of Parma, Department of Food and Drug, Parco Area Delle Scienze 17A, 43124, Parma, Italy
| | - Annamaria Buschini
- University of Parma, Department of Chemistry, Life Sciences and Environmental Sustainability, Parco Area Delle Scienze 11A, 43124, Parma, Italy.
| |
Collapse
|
28
|
Ren C, Li N, Gao C, Zhang W, Yang Y, Li S, Ji X, Ding Y. Ligustilide provides neuroprotection by promoting angiogenesis after cerebral ischemia. Neurol Res 2020; 42:683-692. [PMID: 32584207 DOI: 10.1080/01616412.2020.1782122] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Changhong Ren
- Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- BeijingKey Laboratory of Hypoxia Translational Medicine, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorder, Beijing, China
| | - Ning Li
- Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- BeijingKey Laboratory of Hypoxia Translational Medicine, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorder, Beijing, China
| | - Chen Gao
- Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- BeijingKey Laboratory of Hypoxia Translational Medicine, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorder, Beijing, China
| | - Wei Zhang
- Department of Herbal Formula Science Medicine, Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, China
| | - Yong Yang
- Department of Herbal Formula Science Medicine, Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, China
| | - Sijie Li
- BeijingKey Laboratory of Hypoxia Translational Medicine, Beijing, China
| | - Xunming Ji
- Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- BeijingKey Laboratory of Hypoxia Translational Medicine, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorder, Beijing, China
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
29
|
Kuo KL, Zhao JF, Huang PH, Guo BC, Tarng DC, Lee TS. Indoxyl sulfate impairs valsartan-induced neovascularization. Redox Biol 2020; 30:101433. [PMID: 31972507 PMCID: PMC6974788 DOI: 10.1016/j.redox.2020.101433] [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: 09/12/2019] [Revised: 12/21/2019] [Accepted: 01/13/2020] [Indexed: 12/30/2022] Open
Abstract
Studies revealed that the use of renin-angiotensin-aldosterone system antagonism is not associated with a statistically significant reduction in the risk of cardiovascular events in patients with chronic kidney disease (CKD) compared with that in the general population. We tested the hypothesis that indoxyl sulfate (IS) can interfere with the protective effect of valsartan-mediated on endothelial function in vitro and neovascularization in mice underwent subtotal nephrectomy. In human aortic endothelial cells, we first demonstrated that IS impaired the valsartan-mediated phosphorylation of eNOSThr495, nitric oxide production and tube formation via NADPH oxidase (NOX) and protein kinase C (PKC) phosphorylation, but this effect was suppressed by cotreatment with apocynin and calphostin C. In vivo, IS attenuated valsartan-induced angiogenesis in Matrigel plugs in mice. Moreover, in subtotal nephrectomy mice who underwent hindlimb ischemic surgery, valsartan significantly increased the mobilization of endothelial progenitor cells in circulation as well as the reperfusion of blood flow and density of CD31+ capillaries in ischemic limbs. However, IS attenuated the protective effect of valsartan-induced neovascularization and increased the expression of p-PKCαSer657 and p-eNOSThr497 in ischemic limbs. Cotreatment of apocynin and calphostin C reversed the IS impaired-neovascularization and decreased the expression of p-PKCαSer657 and p-eNOSThr497 in ischemic limbs. Our study suggests that the NOX/PKC/eNOS signaling pathway plays a pivotal role in the IS-mediated inhibition of valsartan-conferred beneficial effects on endothelial function in vitro and neovascularization in subtotal nephrectomy mice. We proposed a novel causative role for IS in cardiovascular complications in CKD patients. The use of renin-angiotensin-aldosterone system antagonism fails to reduce in the risk cardiovascular events in patients with CKD. Indoxyl sulfate interferes with the protective effect of angiotensin II receptor blocker-mediated neovascularization in CKD mice. Indoxyl sulfate interferes with the beneficial effect of of valsartan on endothelial function by activating the NOX/PKC signaling pathway. This article proposed a novel causative role for indoxyl sulfate in cardiovascular complications in CKD patients.
Collapse
Affiliation(s)
- Ko-Lin Kuo
- Division of Nephrology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan; School of Medicine, Buddhist Tzu Chi University, Hualien, Taiwan
| | - Jin-Feng Zhao
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK
| | - Po-Hsun Huang
- Institutes of Clinical Medicine, Taipei, Taiwan; Cardiovascular Research Center, School of Medicine, National Yang-Ming University, Taipei, Taiwan; Divisions of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Bei-Chia Guo
- Graduate Institute and Department of Physiology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Der-Cherng Tarng
- Institutes of Clinical Medicine, Taipei, Taiwan; Department and Institute of Physiology, National Yang-Ming University, Taipei, Taiwan; Division of Nephrology, Department of Medicine and Immunology Research Centre, Taipei Veterans General Hospital, Taipei, Taiwan.
| | - Tzong-Shyuan Lee
- Graduate Institute and Department of Physiology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan.
| |
Collapse
|
30
|
Icli B, Wu W, Ozdemir D, Li H, Cheng HS, Haemmig S, Liu X, Giatsidis G, Avci SN, Lee N, Guimaraes RB, Manica A, Marchini JF, Rynning SE, Risnes I, Hollan I, Croce K, Yang X, Orgill DP, Feinberg MW. MicroRNA-615-5p Regulates Angiogenesis and Tissue Repair by Targeting AKT/eNOS (Protein Kinase B/Endothelial Nitric Oxide Synthase) Signaling in Endothelial Cells. Arterioscler Thromb Vasc Biol 2019; 39:1458-1474. [PMID: 31092013 PMCID: PMC6594892 DOI: 10.1161/atvbaha.119.312726] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 04/29/2019] [Indexed: 12/16/2022]
Abstract
Objective- In response to tissue injury, the appropriate progression of events in angiogenesis is controlled by a careful balance between pro and antiangiogenic factors. We aimed to identify and characterize microRNAs that regulate angiogenesis in response to tissue injury. Approach and Results- We show that in response to tissue injury, microRNA-615-5p (miR-615-5p) is rapidly induced and serves as an antiangiogenic microRNA by targeting endothelial cell VEGF (vascular endothelial growth factor)-AKT (protein kinase B)/eNOS (endothelial nitric oxide synthase) signaling in vitro and in vivo. MiR-615-5p expression is increased in wounds of diabetic db/db mice, in plasma of human subjects with acute coronary syndromes, and in plasma and skin of human subjects with diabetes mellitus. Ectopic expression of miR-615-5p markedly inhibited endothelial cell proliferation, migration, network tube formation in Matrigel, and the release of nitric oxide, whereas miR-615-5p neutralization had the opposite effects. Mechanistic studies using transcriptomic profiling, bioinformatics, 3' untranslated region reporter and microribonucleoprotein immunoprecipitation assays, and small interfering RNA dependency studies demonstrate that miR-615-5p inhibits the VEGF-AKT/eNOS signaling pathway in endothelial cells by targeting IGF2 (insulin-like growth factor 2) and RASSF2 (Ras-associating domain family member 2). Local delivery of miR-615-5p inhibitors, markedly increased angiogenesis, granulation tissue thickness, and wound closure rates in db/db mice, whereas miR-615-5p mimics impaired these effects. Systemic miR-615-5p neutralization improved skeletal muscle perfusion and angiogenesis after hindlimb ischemia in db/db mice. Finally, modulation of miR-615-5p expression dynamically regulated VEGF-induced AKT signaling and angiogenesis in human skin organoids as a model of tissue injury. Conclusions- These findings establish miR-615-5p as an inhibitor of VEGF-AKT/eNOS-mediated endothelial cell angiogenic responses and that manipulating miR-615-5p expression could provide a new target for angiogenic therapy in response to tissue injury. Visual Overview- An online visual overview is available for this article.
Collapse
Affiliation(s)
- Basak Icli
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Winona Wu
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Denizhan Ozdemir
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
- Department of Medical Biology, Hacettepe University, Ankara, Turkey
| | - Hao Li
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Henry S. Cheng
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Stefan Haemmig
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Xin Liu
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Giorgio Giatsidis
- Division of Plastic Surgery, Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Seyma Nazli Avci
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Nathan Lee
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Raphael Boesch Guimaraes
- Instituto de Cardiologia do Rio Grande do Sul, Fundação Universitária de Cardiologia (ICFUC), Porto Alegre, RS, Brazil
| | - Andre Manica
- Instituto de Cardiologia do Rio Grande do Sul, Fundação Universitária de Cardiologia (ICFUC), Porto Alegre, RS, Brazil
| | - Julio F Marchini
- Heart Institute, University of São Paulo Medical School, São Paulo, Brazil
| | - Stein Erik Rynning
- Rheumatology, Lillehamer Hospital for Rheumatic Diseases, Lillehamer, Norway
| | - Ivar Risnes
- Rheumatology, Lillehamer Hospital for Rheumatic Diseases, Lillehamer, Norway
| | - Ivana Hollan
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
- Research Department, Lillehamer Hospital for Rheumatic Diseases, Lillehamer, Norway
| | - Kevin Croce
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | | | - Dennis P. Orgill
- Division of Plastic Surgery, Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Mark W. Feinberg
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
31
|
McKay EC, Beck JS, Khoo SK, Dykema KJ, Cottingham SL, Winn ME, Paulson HL, Lieberman AP, Counts SE. Peri-Infarct Upregulation of the Oxytocin Receptor in Vascular Dementia. J Neuropathol Exp Neurol 2019; 78:436-452. [PMID: 30990880 PMCID: PMC6467199 DOI: 10.1093/jnen/nlz023] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Vascular dementia (VaD) is cognitive decline linked to reduced cerebral blood perfusion, yet there are few therapeutic options to protect cognitive function following cerebrovascular accidents. The purpose of this study was to profile gene expression changes unique to VaD to identify and characterize disease relevant changes that could offer clues for future therapeutic direction. Microarray-based profiling and validation studies of postmortem frontal cortex samples from VaD, Alzheimer disease, and age-matched control subjects revealed that the oxytocin receptor (OXTR) was strongly and differentially upregulated in VaD. Further characterization in fixed tissue from the same cases showed that OXTR upregulation occurs de novo around and within microinfarcts in peri-infarct reactive astrocytes as well as within vascular profiles, likely on microvascular endothelial cells. These results indicate that increased OXTR expression in peri-infarct regions may be a specific response to microvascular insults. Given the established OXTR signaling cascades that elicit antioxidant, anti-inflammatory, and pro-angiogenic responses, the present findings suggest that de novo OXTR expression in the peri-infarct space is a tissue-protective response by astroglial and vascular cells in the wake of ischemic damage that could be exploited as a therapeutic option for the preservation of cognition following cerebrovascular insults.
Collapse
Affiliation(s)
- Erin C McKay
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, Michigan
- Neuroscience Program, Michigan State University, East Lansing, Michigan
| | - John S Beck
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, Michigan
| | - Sok Kean Khoo
- Department of Cell and Molecular Biology, Grand Valley State University, Grand Rapids, Michigan
| | - Karl J Dykema
- Bioinformatics and Biostatistics Core, Van Andel Research Institute, Grand Rapids, Michigan
| | - Sandra L Cottingham
- Department of Pathology, Spectrum Health and Helen DeVos Children’s Hospital, Grand Rapids, Michigan
| | - Mary E Winn
- Bioinformatics and Biostatistics Core, Van Andel Research Institute, Grand Rapids, Michigan
| | - Henry L Paulson
- Department of Neurology, University of Michigan, Ann Arbor, Michigan
- Michigan Alzheimer’s Disease Core Center, Ann Arbor, Michigan
| | - Andrew P Lieberman
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- Michigan Alzheimer’s Disease Core Center, Ann Arbor, Michigan
| | - Scott E Counts
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, Michigan
- Neuroscience Program, Michigan State University, East Lansing, Michigan
- Michigan Alzheimer’s Disease Core Center, Ann Arbor, Michigan
- Department of Family Medicine, Michigan State University, Grand Rapids, Michigan
- Hauenstein Neurosciences Center, Mercy Health Saint Mary’s Hospital, Grand Rapids, Michigan
| |
Collapse
|
32
|
MacKeil JL, Brzezinska P, Burke-Kleinman J, Craig AW, Nicol CJB, Maurice DH. A PKA/cdc42 Signaling Axis Restricts Angiogenic Sprouting by Regulating Podosome Rosette Biogenesis and Matrix Remodeling. Sci Rep 2019; 9:2385. [PMID: 30787359 PMCID: PMC6382826 DOI: 10.1038/s41598-018-37805-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 12/12/2018] [Indexed: 12/17/2022] Open
Abstract
Angiogenic sprouting can contribute adaptively, or mal-adaptively, to a myriad of conditions including ischemic heart disease and cancer. While the cellular and molecular systems that regulate tip versus stalk endothelial cell (EC) specification during angiogenesis are known, those systems that regulate their distinct actions remain poorly understood. Pre-clinical and clinical findings support sustained adrenergic signaling in promoting angiogenesis, but links between adrenergic signaling and angiogenesis are lacking; importantly, adrenergic agents alter the activation status of the cAMP signaling system. Here, we show that the cAMP effector, PKA, acts in a cell autonomous fashion to constitutively reduce the in vitro and ex vivo angiogenic sprouting capacity of ECs. At a cellular level, we observed that silencing or inhibiting PKA in human ECs increased their invasive capacity, their generation of podosome rosettes and, consequently, their ability to degrade a collagen matrix. While inhibition of either Src-family kinases or of cdc42 reduced these events in control ECs, only cdc42 inhibition, or silencing, significantly impacted them in PKA(Cα)-silenced ECs. Consistent with these findings, cell-based measurements of cdc42 activity revealed that PKA activation inhibits EC cdc42 activity, at least in part, by promoting its interaction with the inhibitory regulator, guanine nucleotide dissociation inhibitor-α (RhoGDIα).
Collapse
Affiliation(s)
- J L MacKeil
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, K7L 3N6, Canada
| | - P Brzezinska
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, K7L 3N6, Canada
| | - J Burke-Kleinman
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, K7L 3N6, Canada
| | - A W Craig
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, K7L 3N6, Canada
| | - C J B Nicol
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, K7L 3N6, Canada
| | - D H Maurice
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, K7L 3N6, Canada. .,Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, K7L 3N6, Canada.
| |
Collapse
|
33
|
α-Melanocyte-Stimulating Hormone Attenuates Neovascularization by Inducing Nitric Oxide Deficiency via MC-Rs/PKA/NF-κB Signaling. Int J Mol Sci 2018; 19:ijms19123823. [PMID: 30513637 PMCID: PMC6321109 DOI: 10.3390/ijms19123823] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 11/27/2018] [Accepted: 11/27/2018] [Indexed: 12/13/2022] Open
Abstract
α-melanocyte-stimulating hormone (α-MSH) has been characterized as a novel angiogenesis inhibitor. The homeostasis of nitric oxide (NO) plays an important role in neovascularization. However, it remains unclear whether α-MSH mitigates angiogenesis through modulation of NO and its signaling pathway. The present study elucidated the function and mechanism of NO signaling in α-MSH-induced angiogenesis inhibition using cultured human umbilical vein endothelial cells (HUVECs), rat aorta rings, and transgenic zebrafish. By Griess reagent assay, it was found α-MSH dose-dependently reduced the NO release in HUVECs. Immunoblotting and immunofluorescence analysis revealed α-MSH potently suppressed endothelial and inducible nitric oxide synthase (eNOS/iNOS) expression, which was accompanied with inhibition of nuclear factor kappa B (NF-κB) activities. Excessive supply of NO donor l-arginine reversed the α-MSH-induced angiogenesis inhibition in vitro and in vivo. By using antibody neutralization and RNA interference, it was delineated that melanocortin-1 receptor (MC1-R) and melanocortin-2 receptor (MC2-R) participated in α-MSH-induced inhibition of NO production and NF-κB/eNOS/iNOS signaling. This was supported by pharmaceutical inhibition of protein kinase A (PKA), the downstream effector of MC-Rs signaling, using H89 abolished the α-MSH-mediated suppression of NO release and eNOS/iNOS protein level. Therefore, α-MSH exerts anti-angiogenic function by perturbing NO bioavailability and eNOS/iNOS expression in endothelial cells.
Collapse
|
34
|
Maniyar R, Chakraborty S, Suriano R. Ethanol Enhances Estrogen Mediated Angiogenesis in Breast Cancer. J Cancer 2018; 9:3874-3885. [PMID: 30410590 PMCID: PMC6218769 DOI: 10.7150/jca.25581] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 08/20/2018] [Indexed: 01/15/2023] Open
Abstract
Angiogenesis, a highly regulated process, is exploited by tumors like breast cancer to ensure a constant supply of oxygen and nutrients and is key for tumor survival and progression. Estrogen and alcohol independently have been observed to contribute to angiogenesis in breast cancer but their combinatorial effects have never been evaluated. The exact mechanism by which estrogen and alcohol contribute to breast cancer angiogenesis remains to be elucidated. In this study, we defined the in vitro effects of the combination of estrogen and alcohol in breast cancer angiogenesis using the tubulogenesis and scratch wound assays. Conditioned media, generated by culturing the murine mammary cancer cell line, TG1-1, in estrogen and ethanol, enhanced tubule formation and migration as well as modulated the MAP Kinase pathway in the murine endothelial cell line, SVEC4-10. Additionally, estrogen and ethanol in combination enhanced the expression of the pro-angiogenic factors VEGF, MMP-9, and eNOS, and modulated Akt activation. These observations suggest that TG1-1 cells secrete pro-angiogenic molecules in response to the combination of estrogen and ethanol that modulate the morphological and migratory properties of endothelial cells. The data presented in this study, is the first in attempting to link the cooperative activity between estrogen and ethanol in breast cancer progression, underscoring correlations first made by epidemiological observations linking the two.
Collapse
Affiliation(s)
- Rachana Maniyar
- Department of Microbiology and Immunology, New York Medical College, Valhalla, New York, United States of America
| | - Sanjukta Chakraborty
- Department of Microbiology and Immunology, New York Medical College, Valhalla, New York, United States of America
| | - Robert Suriano
- Department of Microbiology and Immunology, New York Medical College, Valhalla, New York, United States of America
- Division of Natural Sciences, College of Mount Saint Vincent, Bronx. New York, United States of America
| |
Collapse
|
35
|
Ren C, Li N, Li S, Han R, Huang Q, Hu J, Jin K, Ji X. Limb Ischemic Conditioning Improved Cognitive Deficits via eNOS-Dependent Augmentation of Angiogenesis after Chronic Cerebral Hypoperfusion in Rats. Aging Dis 2018; 9:869-879. [PMID: 30271664 PMCID: PMC6147592 DOI: 10.14336/ad.2017.1106] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 11/06/2017] [Indexed: 12/27/2022] Open
Abstract
Intracranial and extracranial arterial stenosis, the primary cause of chronic cerebral hypoperfusion (CCH), is a critical reason for the pathogenesis of vascular dementia and Alzheimer’s disease characterized by cognitive impairments. Our previous study demonstrated that limb remote ischemic conditioning (LRIC) improved cerebral perfusion in intracranial arterial stenosis patients. The current study aimed to test whether LRIC promotes angiogenesis and increases phosphorylated endothelial nitric oxide synthase (p-eNOS) activity in CCH rat model. Adult male Sprague-Dawley rats were randomly assigned to three different groups: sham group, bilateral carotid artery occlusion (2VO) group and 2VO+LRIC group. Cerebral Blood Flow (CBF) was measured with laser speckle contrast imager at 4 weeks. Cognitive testing was performed at four and six weeks after 2VO surgery. We demonstrated that LRIC treatment increased cerebral perfusion and improved the CCH induced spatial learning and memory impairment. Immunohistochemistry confirmed that LRIC prevented cell death in the CA1 region, and increased the number of vessels and angiogenesis in the hippocampus after 2VO. Western blot analysis shows that LRIC therapy significantly increased p-eNOS expression in the hippocampus when compared with 2VO rats. Moreover, eNOS inhibitor reduced the effect of LRIC on angiogenesis in the hippocampus and spatial learning and memory function. Our data suggested that LRIC promoted angiogenesis, which is mediated, in part, by eNOS/NO.
Collapse
Affiliation(s)
- Changhong Ren
- 1Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.,2Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, TX Texas 76107, USA.,3Center of Stroke, Beijing Institute for Brain Disorder, Beijing 100069, China.,4Beijing Key Laboratory of Hypoxia Translational Medicine, Beijing 100053, China
| | - Ning Li
- 1Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.,5Department of Neurobiology, Capital Medical University, Beijing 10069, China
| | - Sijie Li
- 1Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.,4Beijing Key Laboratory of Hypoxia Translational Medicine, Beijing 100053, China
| | - Rongrong Han
- 1Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.,4Beijing Key Laboratory of Hypoxia Translational Medicine, Beijing 100053, China
| | - Qingjian Huang
- 1Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.,2Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, TX Texas 76107, USA.,4Beijing Key Laboratory of Hypoxia Translational Medicine, Beijing 100053, China
| | - Jiangnan Hu
- 2Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, TX Texas 76107, USA
| | - Kunlin Jin
- 2Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, TX Texas 76107, USA
| | - Xunming Ji
- 1Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.,3Center of Stroke, Beijing Institute for Brain Disorder, Beijing 100069, China.,4Beijing Key Laboratory of Hypoxia Translational Medicine, Beijing 100053, China
| |
Collapse
|
36
|
Yu W, Gu Y, Chen P, Luo J, Liu P, Chao Y, Chen SL, Zhang H. Norepinephrine stimulation downregulates the β 2 -adrenergic receptor-nitric oxide pathway in human pulmonary artery endothelial cells. J Cell Physiol 2018; 234:1842-1850. [PMID: 30132872 DOI: 10.1002/jcp.27057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 06/25/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND Norepinephrine (NE)-mediated vasoconstriction plays an important role in pulmonary hypertension associated with left heart disease (PH-LHD). However, the role of NE-mediated endothelial cell dysfunction in the pathogenesis of PH-LHD remains to be elucidated. METHODS AND RESULTS An enzyme-linked immunosorbent assay showed that the NE concentration in the plasma of patients with PH-LHD was higher and the nitrate-nitrite concentration was lower than those in the control group. NE treatment decreased phospho-Ser633-eNOS and β2 -adrenergic receptor (β2 -AR) levels in the membrane of human pulmonary artery endothelial cells (HPAECs) analysed by western blot analysis. Consistently, fluorescence microscopy and flow cytometry showed that nitric oxide (NO) production was also decreased in HPAECs. Coimmunoprecipitation confirmed a direct interaction between β2 -AR and endothelial NO synthase (eNOS). Overexpression of β2 -AR attenuated the decline in phospho-Ser633-eNOS and NO production. Additionally, the expression of phospho-Ser633-eNOS and β2 -AR was decreased in human pulmonary artery endothelium. Finally, our results indicate that NE stimulated HPAEC proliferation, which was blocked by protein kinase A inhibitor or protein kinase B (PKB-AKT) inhibitor. CONCLUSIONS These data provide a novel mechanism for NE-decreased endothelium-derived NO and NE-induced HPAEC proliferation that leads to PH-LHD, suggesting a potential therapeutic target for PH-LHD.
Collapse
Affiliation(s)
- Wande Yu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yue Gu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - PeiP Chen
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jie Luo
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Pengfei Liu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yuelin Chao
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Shao-Liang Chen
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Hang Zhang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
37
|
Kho J, Tian X, Wong WT, Bertin T, Jiang MM, Chen S, Jin Z, Shchelochkov OA, Burrage LC, Reddy AK, Jiang H, Abo-Zahrah R, Ma S, Zhang P, Bissig KD, Kim JJ, Devaraj S, Rodney GG, Erez A, Bryan NS, Nagamani SC, Lee BH. Argininosuccinate Lyase Deficiency Causes an Endothelial-Dependent Form of Hypertension. Am J Hum Genet 2018; 103:276-287. [PMID: 30075114 DOI: 10.1016/j.ajhg.2018.07.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 07/12/2018] [Indexed: 02/07/2023] Open
Abstract
Primary hypertension is a major risk factor for ischemic heart disease, stroke, and chronic kidney disease. Insights obtained from the study of rare Mendelian forms of hypertension have been invaluable in elucidating the mechanisms causing primary hypertension and development of antihypertensive therapies. Endothelial cells play a key role in the regulation of blood pressure; however, a Mendelian form of hypertension that is primarily due to endothelial dysfunction has not yet been described. Here, we show that the urea cycle disorder, argininosuccinate lyase deficiency (ASLD), can manifest as a Mendelian form of endothelial-dependent hypertension. Using data from a human clinical study, a mouse model with endothelial-specific deletion of argininosuccinate lyase (Asl), and in vitro studies in human aortic endothelial cells and induced pluripotent stem cell-derived endothelial cells from individuals with ASLD, we show that loss of ASL in endothelial cells leads to endothelial-dependent vascular dysfunction with reduced nitric oxide (NO) production, increased oxidative stress, and impaired angiogenesis. Our findings show that ASLD is a unique model for studying NO-dependent endothelial dysfunction in human hypertension.
Collapse
|
38
|
Park KH, Choi Y, Yoon DS, Lee KM, Kim D, Lee JW. Zinc Promotes Osteoblast Differentiation in Human Mesenchymal Stem Cells Via Activation of the cAMP-PKA-CREB Signaling Pathway. Stem Cells Dev 2018; 27:1125-1135. [PMID: 29848179 DOI: 10.1089/scd.2018.0023] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The crucial trace element zinc stimulates osteogenesis in vitro and in vivo. However, the pathways mediating these effects remain poorly understood. This study aimed to investigate the effects of zinc on osteoblast differentiation in human bone marrow-derived mesenchymal stem cells (hBMSCs) and to identify the molecular mechanisms of these effects. In hBMSCs, zinc exposure resulted in a dose-dependent increase in osteogenesis and increased mRNA and protein levels of the master transcriptional factor RUNX2. Analyzing the upstream signaling pathways of RUNX2, we found that protein kinase A (PKA) signaling inhibition blocked zinc-induced osteogenic effects. Zinc exposure increased transcriptional activity and protein levels of phospho-CREB and enhanced translocation of phospho-CREB into the nucleus. These effects were reversed by H-89, a potent inhibitor of PKA. Moreover, zinc exposure led to dose-dependent increases in levels of intracellular cyclic adenosine monophosphate (cAMP). These findings indicate that zinc activates the PKA signaling pathway by triggering an increase in intracellular cAMP, leading to enhanced osteogenic differentiation in hBMSCs. Our results suggest that zinc exerts osteogenic effects in hBMSCs by activation of RUNX2 via the cAMP-PKA-CREB signaling pathway. Zinc supplementation may offer a promise as a potential pharmaceutical therapy for osteoporosis and other bone loss conditions.
Collapse
Affiliation(s)
- Kwang Hwan Park
- 1 Department of Orthopaedic Surgery, Yonsei University College of Medicine , Seoul, South Korea
| | - Yoorim Choi
- 1 Department of Orthopaedic Surgery, Yonsei University College of Medicine , Seoul, South Korea .,2 Brain Korea 21 Plus Project for Medical Sciences, Yonsei University College of Medicine , Seoul, South Korea
| | - Dong Suk Yoon
- 3 Department of Internal Medicine, Brody School of Medicine at East Carolina University , Greenville, North Carolina
| | - Kyoung-Mi Lee
- 1 Department of Orthopaedic Surgery, Yonsei University College of Medicine , Seoul, South Korea .,4 Severance Biomedical Science Institute, Yonsei University College of Medicine , South Korea
| | - Dohyun Kim
- 1 Department of Orthopaedic Surgery, Yonsei University College of Medicine , Seoul, South Korea
| | - Jin Woo Lee
- 1 Department of Orthopaedic Surgery, Yonsei University College of Medicine , Seoul, South Korea .,2 Brain Korea 21 Plus Project for Medical Sciences, Yonsei University College of Medicine , Seoul, South Korea .,4 Severance Biomedical Science Institute, Yonsei University College of Medicine , South Korea
| |
Collapse
|
39
|
Browne S, Jha AK, Ameri K, Marcus SG, Yeghiazarians Y, Healy KE. TGF-β1/CD105 signaling controls vascular network formation within growth factor sequestering hyaluronic acid hydrogels. PLoS One 2018; 13:e0194679. [PMID: 29566045 PMCID: PMC5864059 DOI: 10.1371/journal.pone.0194679] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 03/07/2018] [Indexed: 12/17/2022] Open
Abstract
Cell-based strategies for the treatment of ischemic diseases are at the forefront of tissue engineering and regenerative medicine. Cell therapies purportedly can play a key role in the neovascularization of ischemic tissue; however, low survival and poor cell engraftment with the host vasculature following implantation limits their potential to treat ischemic diseases. To overcome these limitations, we previously developed a growth factor sequestering hyaluronic acid (HyA)-based hydrogel that enhanced transplanted mouse cardiosphere-derived cell survival and formation of vasculature that anastomosed with host vessels. In this work, we examined the mechanism by which HyA hydrogels presenting transforming growth factor beta-1 (TGF-β1) promoted proliferation of more clinically relevant human cardiosphere-derived cells (hCDC), and their formation of vascular-like networks in vitro. We observed hCDC proliferation and enhanced formation of vascular-like networks occurred in the presence of TGF-β1. Furthermore, production of nitric oxide (NO), VEGF, and a host of angiogenic factors were increased in the presence of TGF-β1. This response was dependent on the co-activity of CD105 (Endoglin) with the TGF-βR2 receptor, demonstrating its role in the process of angiogenic differentiation and vascular organization of hCDC. These results demonstrated that hCDC form vascular-like networks in vitro, and that the induction of vascular networks by hCDC within growth factor sequestering HyA hydrogels was mediated by TGF-β1/CD105 signaling.
Collapse
Affiliation(s)
- Shane Browne
- Departments of Bioengineering and Materials Science and Engineering, University of California, Berkeley, CA, United States of America
- Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Galway, Ireland
| | - Amit K. Jha
- Departments of Bioengineering and Materials Science and Engineering, University of California, Berkeley, CA, United States of America
| | - Kurosh Ameri
- Department of Medicine, University of California, San Francisco, CA, United States of America
| | - Sivan G. Marcus
- Departments of Bioengineering and Materials Science and Engineering, University of California, Berkeley, CA, United States of America
| | - Yerem Yeghiazarians
- Department of Medicine, University of California, San Francisco, CA, United States of America
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, United States of America
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States of America
| | - Kevin E. Healy
- Departments of Bioengineering and Materials Science and Engineering, University of California, Berkeley, CA, United States of America
- * E-mail:
| |
Collapse
|
40
|
Ribeiro A, Abreu RM, Dias MM, Barreiro MF, Ferreira IC. Antiangiogenic compounds: well-established drugs versus emerging natural molecules. Cancer Lett 2018; 415:86-105. [DOI: 10.1016/j.canlet.2017.12.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 11/17/2017] [Accepted: 12/01/2017] [Indexed: 12/19/2022]
|
41
|
Ricciuti B, Foglietta J, Bianconi V, Sahebkar A, Pirro M. Enzymes involved in tumor-driven angiogenesis: A valuable target for anticancer therapy. Semin Cancer Biol 2017; 56:87-99. [PMID: 29128510 DOI: 10.1016/j.semcancer.2017.11.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 11/01/2017] [Accepted: 11/04/2017] [Indexed: 02/07/2023]
Abstract
Angiogenesis plays a pivotal role in cancer progression and is required for tissue invasion and metastasis. Starting with Folkman's initial observations in 1971, basic research continued to shed new molecular insight into this multifaceted process, leading to the development of several anti-angiogenic drugs. To date, anti-vascular endothelial growth factor monoclonal antibodies, such as bevacizumab and ramucirumab, and receptor tyrosine kinase inhibitors (e.g., sorafenib, sunitinib, regorafenib and axitinib) have had a profound impact on the way we treat patients with advanced cancer, providing in some cases unprecedented clinical benefit. The molecular mechanisms underlying tumor-driven angiogenesis have been explored extensively and have unveiled a number of potential clinically relevant targets, including several novel enzymes. In this review, we summarized the current strategies to target tumor-driven angiogenesis through the inhibition of relevant and selected classes of enzymes involved in this process.
Collapse
Affiliation(s)
- Biagio Ricciuti
- Department of Medical Oncology, Santa Maria della Misericordia Hospital, Piazzale L. Severi n. 1, 06132, Perugia, Italy
| | - Jennifer Foglietta
- Department of Medical Oncology, Santa Maria della Misericordia Hospital, Piazzale L. Severi n. 1, 06132, Perugia, Italy
| | - Vanessa Bianconi
- Department of Medicine, University of Perugia, Piazzale L. Severi n. 1, 06132, Perugia, Italy
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Matteo Pirro
- Department of Medicine, University of Perugia, Piazzale L. Severi n. 1, 06132, Perugia, Italy.
| |
Collapse
|
42
|
Chen D, Tang J, Wan Q, Zhang J, Wang K, Shen Y, Yu Y. E-Prostanoid 3 Receptor Mediates Sprouting Angiogenesis Through Suppression of the Protein Kinase A/β-Catenin/Notch Pathway. Arterioscler Thromb Vasc Biol 2017; 37:856-866. [DOI: 10.1161/atvbaha.116.308587] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 02/16/2017] [Indexed: 01/14/2023]
Abstract
Objective—
Angiogenesis is a hallmark of embryonic development and various ischemic and inflammatory diseases. Prostaglandin E2 receptor subtype 3 (EP3) plays an important role in pathophysiologic angiogenesis; however, the precise mechanisms remain unknown. Here, we investigated the role of EP3 in zebra fish embryo and mouse retina angiogenesis and evaluated the underlying mechanisms.
Approach and Results—
The EP3 receptor was highly expressed in the vasculature in both zebra fish embryos and murine fetal retinas. Pharmacological inhibition or genetic deletion of EP3 significantly reduced vasculature formation in zebra fish embryos and mouse retinas. Further characterization revealed reduced filopodia extension of tip cells in embryonic retinas in EP3-deficient mice. EP3 deletion activated Notch activity by upregulation of delta-like ligand 4 expression in endothelial cells (ECs). Inhibition of Notch signaling rescued the angiogenic defects in EP3-deficient mouse retinas. Moreover, EP3 deficiency led to a significant increase in β-catenin phosphorylation at Ser675 and nuclear accumulation of β-catenin in ECs. Knockdown or inhibition of β-catenin restored the impaired sprouting angiogenesis resulting from EP3 deficiency in ECs. The EP3 receptor depressed protein kinase A activity in ECs by coupling to Gαi. Inhibition of protein kinase A activity significantly reduced Ser675 phosphorylation and nuclear translocation of β-catenin, abolished the increased delta-like ligand 4 expression, and subsequently restored the impaired angiogenic capacity of EP3-deficient ECs both in vitro and in vivo.
Conclusions—
Activation of the EP3 receptor facilitates sprouting angiogenesis through protein kinase A–dependent Notch signaling, suggesting that EP3 and its downstream pathways maybe potential therapeutic targets in the treatment of ischemic diseases.
Collapse
Affiliation(s)
- Di Chen
- From the Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (D.C., J.Z., Y.S., Y.Y.); and Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China (D.C., J.T., Q.W., K.W., Y.Y.)
| | - Juan Tang
- From the Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (D.C., J.Z., Y.S., Y.Y.); and Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China (D.C., J.T., Q.W., K.W., Y.Y.)
| | - Qiangyou Wan
- From the Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (D.C., J.Z., Y.S., Y.Y.); and Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China (D.C., J.T., Q.W., K.W., Y.Y.)
| | - Jian Zhang
- From the Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (D.C., J.Z., Y.S., Y.Y.); and Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China (D.C., J.T., Q.W., K.W., Y.Y.)
| | - Kai Wang
- From the Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (D.C., J.Z., Y.S., Y.Y.); and Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China (D.C., J.T., Q.W., K.W., Y.Y.)
| | - Yujun Shen
- From the Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (D.C., J.Z., Y.S., Y.Y.); and Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China (D.C., J.T., Q.W., K.W., Y.Y.)
| | - Ying Yu
- From the Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (D.C., J.Z., Y.S., Y.Y.); and Key Laboratory of Food Safety Research, CAS Center for Excellence in Molecular Cell Science, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China (D.C., J.T., Q.W., K.W., Y.Y.)
| |
Collapse
|
43
|
Barui AK, Nethi SK, Patra CR. Investigation of the role of nitric oxide driven angiogenesis by zinc oxide nanoflowers. J Mater Chem B 2017; 5:3391-3403. [DOI: 10.1039/c6tb03323g] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Zinc oxide nanoflowers (ZONF) induce the generation of intracellular ROS that stimulates the phosphorylation of Akt and p38MAPK leading to activation of eNOS to produce NO, triggering angiogenesis in a cGMP dependent manner.
Collapse
Affiliation(s)
- Ayan Kumar Barui
- Department of Chemical Biology
- CSIR-Indian Institute of Chemical Technology
- Hyderabad – 500007
- India
- Academy of Scientific and Innovative Research (AcSIR)
| | - Susheel Kumar Nethi
- Department of Chemical Biology
- CSIR-Indian Institute of Chemical Technology
- Hyderabad – 500007
- India
- Academy of Scientific and Innovative Research (AcSIR)
| | - Chitta Ranjan Patra
- Department of Chemical Biology
- CSIR-Indian Institute of Chemical Technology
- Hyderabad – 500007
- India
- Academy of Scientific and Innovative Research (AcSIR)
| |
Collapse
|
44
|
Yuan S, Kevil CG. Nitric Oxide and Hydrogen Sulfide Regulation of Ischemic Vascular Remodeling. Microcirculation 2016; 23:134-45. [PMID: 26381654 DOI: 10.1111/micc.12248] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 09/13/2015] [Indexed: 12/22/2022]
Abstract
Blockage or restriction of blood flow through conduit arteries results in tissue ischemia downstream of the disturbed area. Local tissues can adapt to this challenge by stimulating vascular remodeling through angiogenesis and arteriogenesis thereby restoring blood perfusion and removal of wastes. Multiple molecular mechanisms of vascular remodeling during ischemia have been identified and extensively studied. However, therapeutic benefits from these findings and insights are limited due to the complexity of various signaling networks and a lack of understanding central metabolic regulators governing these responses. The gasotransmitters NO and H2 S have emerged as master regulators that influence multiple molecular targets necessary for ischemic vascular remodeling. In this review, we discuss how NO and H2 S are individually regulated under ischemia, what their roles are in angiogenesis and arteriogenesis, and how their interaction controls ischemic vascular remodeling.
Collapse
Affiliation(s)
- Shuai Yuan
- Departments of Pathology, Molecular and Cellular Physiology, and Cell Biology and Anatomy, LSU Health Shreveport, Shreveport, Louisiana, USA
| | - Christopher G Kevil
- Departments of Pathology, Molecular and Cellular Physiology, and Cell Biology and Anatomy, LSU Health Shreveport, Shreveport, Louisiana, USA
| |
Collapse
|
45
|
Ren C, Li S, Wang B, Han R, Li N, Gao J, Li X, Jin K, Ji X. Limb remote ischemic conditioning increases Notch signaling activity and promotes arteriogenesis in the ischemic rat brain. Behav Brain Res 2016; 340:87-93. [PMID: 27780723 DOI: 10.1016/j.bbr.2016.10.036] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 10/19/2016] [Accepted: 10/21/2016] [Indexed: 01/20/2023]
Abstract
BACKGROUND AND PURPOSE We tested the hypothesis that limb remote ischemic conditioning (LRIC) treatment promotes arteriogenesis and increases Notch signaling activity during stroke recovery. METHODS Adult male Sprague Dawley rats were subjected to middle cerebral artery occlusion (MCAO). LRIC was applied after the onset of focal ischemia (per-conditioning), followed by repeated short episodes of remote ischemia 24h after reperfusion (post-conditioning). Cerebral blood flow (CBF) was measured by Laser Doppler Flowmetry. Immunohistochemistry was used to reveal α-smooth muscle actin (α-SMA) immunopositive cells in the arteries of the brain. The cerebral angioarchitecture was visualized with a latex perfusion technique. RESULTS LRIC treatment significantly elevated local cerebral blood flow and increased arteriogenesis as indicated by increased arterial diameter and vascular smooth muscle cell proliferation in the ischemic brain. The increased arteriogenesis significantly correlated with the functional outcome after stroke. Furthermore, LRIC treatment upregulated the expressions of Notch1 and Notch intracellular domain (NICD) in arteries surrounding the ischemic area. CONCLUSION These results suggest that the therapeutic effects of LRIC may involve the promotion of arteriogenesis during the recovery phase after focal cerebral ischemia and that Notch1 signaling seems to be an important player in limb remote ischemia-mediated arteriogenesis.
Collapse
Affiliation(s)
- Changhong Ren
- Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Center for Neuroscience Discovery, Institute for Healthy Aging, University of North Texas Health Science Center at Fort Worth, TX 76107, USA; Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Beijing 100053, China; Center for Stroke, Beijing Institute for Brain Disorder, Beijing 100069, China
| | - Sijie Li
- Emergency Department, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Beijing 100053, China
| | - Brian Wang
- Center for Neuroscience Discovery, Institute for Healthy Aging, University of North Texas Health Science Center at Fort Worth, TX 76107, USA
| | - Rongrong Han
- Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Beijing 100053, China; Center for Stroke, Beijing Institute for Brain Disorder, Beijing 100069, China
| | - Ning Li
- Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Beijing 100053, China; Center for Stroke, Beijing Institute for Brain Disorder, Beijing 100069, China
| | - Jinhuan Gao
- Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Xiaohua Li
- Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Beijing 100053, China; Center for Stroke, Beijing Institute for Brain Disorder, Beijing 100069, China
| | - Kunlin Jin
- Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Center for Neuroscience Discovery, Institute for Healthy Aging, University of North Texas Health Science Center at Fort Worth, TX 76107, USA
| | - Xunming Ji
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Beijing 100053, China.
| |
Collapse
|
46
|
Varicella-Zoster Virus Activates CREB, and Inhibition of the pCREB-p300/CBP Interaction Inhibits Viral Replication In Vitro and Skin Pathogenesis In Vivo. J Virol 2016; 90:8686-97. [PMID: 27440893 PMCID: PMC5021407 DOI: 10.1128/jvi.00920-16] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 07/13/2016] [Indexed: 12/12/2022] Open
Abstract
UNLABELLED Varicella-zoster virus (VZV) is an alphaherpesvirus that causes varicella upon primary infection and zoster upon reactivation from latency in sensory ganglion neurons. The replication of herpesviruses requires manipulation of cell signaling pathways. Notably, CREB, a factor involved in the regulation of several cellular processes, is activated upon infection of T cells with VZV. Here, we report that VZV infection also induced CREB phosphorylation in fibroblasts and that XX-650-23, a newly identified inhibitor of the phosphorylated-CREB (pCREB) interaction with p300/CBP, restricted cell-cell spread of VZV in vitro CREB phosphorylation did not require the viral open reading frame 47 (ORF47) and ORF66 kinases encoded by VZV. Evaluating the biological relevance of these observations during VZV infection of human skin xenografts in the SCID mouse model of VZV pathogenesis showed both that pCREB was upregulated in infected skin and that treatment with XX-650-23 reduced infectious-virus production and limited lesion formation compared to treatment with a vehicle control. Thus, processes of CREB activation and p300/CBP binding are important for VZV skin infection and may be targeted for antiviral drug development. IMPORTANCE Varicella-zoster virus (VZV) is a common pathogen that causes chicken pox and shingles. As with all herpesviruses, the infection is acquired for life, and the virus can periodically reactivate from latency. Although VZV infection is usually benign with few or no deleterious consequences, infection can be life threatening in immunocompromised patients. Otherwise healthy elderly individuals who develop zoster as a consequence of viral reactivation are at risk for postherpetic neuralgia (PHN), a painful and long-lasting complication. Current vaccines use a live attenuated virus that is usually safe but cannot be given to many immunodeficient patients and retains the capacity to establish latency and reactivate, causing zoster. Antiviral drugs are effective against severe VZV infections but have little impact on PHN. A better understanding of virus-host cell interactions is relevant for developing improved therapies to safely interfere with cellular processes that are crucial for VZV pathogenesis.
Collapse
|
47
|
Chiu WC, Chiou TJ, Chung MJ, Chiang AN. β2-Glycoprotein I Inhibits Vascular Endothelial Growth Factor-Induced Angiogenesis by Suppressing the Phosphorylation of Extracellular Signal-Regulated Kinase 1/2, Akt, and Endothelial Nitric Oxide Synthase. PLoS One 2016; 11:e0161950. [PMID: 27579889 PMCID: PMC5006999 DOI: 10.1371/journal.pone.0161950] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 08/15/2016] [Indexed: 02/07/2023] Open
Abstract
Angiogenesis is the process of new blood vessel formation, and it plays a key role in various physiological and pathological conditions. The β2-glycoprotein I (β2-GPI) is a plasma glycoprotein with multiple biological functions, some of which remain to be elucidated. This study aimed to identify the contribution of 2-GPI on the angiogenesis induced by vascular endothelial growth factor (VEGF), a pro-angiogenic factor that may regulate endothelial remodeling, and its underlying mechanism. Our results revealed that β2-GPI dose-dependently decreased the VEGF-induced increase in endothelial cell proliferation, using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and the bromodeoxyuridine (BrdU) incorporation assays. Furthermore, incubation with both β2-GPI and deglycosylated β2-GPI inhibited the VEGF-induced tube formation. Our results suggest that the carbohydrate residues of β2-GPI do not participate in the function of anti-angiogenesis. Using in vivo Matrigel plug and angioreactor assays, we show that β2-GPI remarkably inhibited the VEGF-induced angiogenesis at a physiological concentration. Moreover, β2-GPI inhibited the VEGF-induced phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2), Akt, and endothelial nitric oxide synthase (eNOS). In summary, our in vitro and in vivo data reveal for the first time that β2-GPI inhibits the VEGF-induced angiogenesis and highlights the potential for β2-GPI in anti-angiogenic therapy.
Collapse
Affiliation(s)
- Wen-Chin Chiu
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
- Division of Thoracic Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tzeon-Jye Chiou
- Division of Transfusion Medicine, Department of Medicine, Taipei Veterans General Hospital and National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Meng-Ju Chung
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
| | - An-Na Chiang
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
48
|
EphrinB2/EphB4 pathway in postnatal angiogenesis: a potential therapeutic target for ischemic cardiovascular disease. Angiogenesis 2016; 19:297-309. [PMID: 27216867 DOI: 10.1007/s10456-016-9514-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 05/13/2016] [Indexed: 01/12/2023]
Abstract
Ischemic cardiovascular disease remains one of the leading causes of morbidity and mortality in the world. Proangiogenic therapy appears to be a promising and feasible strategy for the patients with ischemic cardiovascular disease, but the results of preclinical and clinical trials are limited due to the complicated mechanisms of angiogenesis. Facilitating the formation of functional vessels is important in rescuing the ischemic cardiomyocytes. EphrinB2/EphB4, a novel pathway in angiogenesis, plays a critical role in both microvascular growth and neovascular maturation. Hence, investigating the mechanisms of EphrinB2/EphB4 pathway in angiogenesis may contribute to the development of novel therapeutics for ischemic cardiovascular disease. Previous reviews mainly focused on the role of EphrinB2/EphB4 pathway in embryo vascular development, but their role in postnatal angiogenesis in ischemic heart disease has not been fully illustrated. Here, we summarized the current knowledge of EphrinB2/EphB4 in angiogenesis and their interaction with other angiogenic pathways in ischemic cardiovascular disease.
Collapse
|
49
|
Wu JR, Hsu JH, Dai ZK, Wu BN, Chen IJ, Liou SF, Yeh JL. Activation of endothelial NO synthase by a xanthine derivative ameliorates hypoxia-induced apoptosis in endothelial progenitor cells. ACTA ACUST UNITED AC 2016; 68:810-8. [PMID: 27109251 DOI: 10.1111/jphp.12555] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 03/13/2016] [Indexed: 01/05/2023]
Abstract
OBJECTIVES Endothelial damage is strongly associated with cardiovascular diseases such as atherosclerosis, thrombosis and hypertension. Endothelial progenitor cells (EPCs) are primitive bone marrow (BM) cells that possess the capacity to mature into endothelial cells and play a role in neovascularization and vascular remodelling. This study aimed to investigate whether KMUP-1, a synthetic xanthine-based derivative, atorvastatin and simvastatin, can prevent endothelial dysfunction and apoptosis induced by hypoxia and to elucidate the underlying mechanisms. METHODS Mononuclear cells were separated and were induced to differentiate into EPCs. KMUP-1, atorvastatin or simvastatin were administered prior to hypoxia. KEY FINDINGS We found that EPCs exposed to hypoxia increased apoptosis as well as diminished proliferation. Pretreatment with KMUP-1, atorvastatin and simvastatin significantly prevented hypoxia-induced EPCs death and apoptosis, with associated increased of the Bcl-2/Bax ratio, and reduced caspase-3 and caspase-9 expression. We also assessed the nitrite production and Ser(1177)-phospho-eNOS expression and found that KMUP-1, atorvastatin and simvastatin not only increased the secretion of NO compared with the hypoxia group but also upregulated the eNOS activation. CONCLUSIONS KMUP-1 inhibited hypoxia-induced dysfunction and apoptosis in EPCs, which may be mediated through suppressing oxidative stress, upregulating eNOS and downregulating the caspase-3 signalling pathway.
Collapse
Affiliation(s)
- Jiunn-Ren Wu
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Department of Paediatrics, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jong-Hau Hsu
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Department of Paediatrics, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Zen-Kong Dai
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Department of Paediatrics, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Bin-Nan Wu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department and Graduate Institute of Pharmacology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ing-Jun Chen
- Department and Graduate Institute of Pharmacology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shu-Fen Liou
- Department of Pharmacy, Chia-Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Jwu-Lai Yeh
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department and Graduate Institute of Pharmacology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
| |
Collapse
|
50
|
Alonso F, Domingos-Pereira S, Le Gal L, Derré L, Meda P, Jichlinski P, Nardelli-Haefliger D, Haefliger JA. Targeting endothelial connexin40 inhibits tumor growth by reducing angiogenesis and improving vessel perfusion. Oncotarget 2016; 7:14015-28. [PMID: 26883111 PMCID: PMC4924695 DOI: 10.18632/oncotarget.7370] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 01/29/2016] [Indexed: 01/09/2023] Open
Abstract
Endothelial connexin40 (Cx40) contributes to regulate the structure and function of vessels. We have examined whether the protein also modulates the altered growth of vessels in tumor models established in control mice (WT), mice lacking Cx40 (Cx40-/-), and mice expressing the protein solely in endothelial cells (Tie2-Cx40). Tumoral angiogenesis and growth were reduced, whereas vessel perfusion, smooth muscle cell (SMC) coverage and animal survival were increased in Cx40-/- but not Tie2-Cx40 mice, revealing a critical involvement of endothelial Cx40 in transformed tissues independently of the hypertensive status of Cx40-/- mice. As a result, Cx40-/- mice bearing tumors survived significantly longer than corresponding controls, including after a cytotoxic administration. Comparable observations were made in WT mice injected with a peptide targeting Cx40, supporting the Cx40 involvement. This involvement was further confirmed in the absence of Cx40 or by peptide-inhibition of this connexin in aorta-sprouting, matrigel plug and SMC migration assays, and associated with a decreased expression of the phosphorylated form of endothelial nitric oxide synthase. The data identify Cx40 as a potential novel target in cancer treatment.
Collapse
MESH Headings
- Animals
- Aorta/pathology
- Apoptosis
- Biomarkers, Tumor/metabolism
- Blood Vessels/physiology
- Cell Proliferation
- Connexins/antagonists & inhibitors
- Connexins/metabolism
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Female
- Humans
- Lung Neoplasms/blood supply
- Lung Neoplasms/pathology
- Lung Neoplasms/prevention & control
- Melanoma, Experimental/blood supply
- Melanoma, Experimental/pathology
- Melanoma, Experimental/prevention & control
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Neoplasm Invasiveness
- Neovascularization, Pathologic/prevention & control
- Perfusion
- Tumor Cells, Cultured
- Urinary Bladder Neoplasms/blood supply
- Urinary Bladder Neoplasms/pathology
- Urinary Bladder Neoplasms/prevention & control
- Gap Junction alpha-5 Protein
Collapse
Affiliation(s)
- Florian Alonso
- Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | | | - Loïc Le Gal
- Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Laurent Derré
- Department of Urology, Lausanne University Hospital, Lausanne, Switzerland
| | - Paolo Meda
- Department of Cell Physiology and Metabolism, University of Geneva, Medical Center, Geneva, Switzerland
| | - Patrice Jichlinski
- Department of Urology, Lausanne University Hospital, Lausanne, Switzerland
| | | | | |
Collapse
|