1
|
Su C, Mi X, Ito T, Kato Y, Nishimura A, Nagata R, Mori Y, Nishida M. TRPC6-Mediated Zn 2+ Influx Negatively Regulates Contractile Differentiation of Vascular Smooth Muscle Cells. Biomolecules 2025; 15:267. [PMID: 40001570 PMCID: PMC11853136 DOI: 10.3390/biom15020267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 02/05/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Vascular smooth muscle cells (VSMCs) can dynamically change their phenotype between contractile and synthetic forms in response to environmental stress, which is pivotal in maintaining vascular homeostasis and mediating pathological remodeling of blood vessels. We previously reported that suppression of canonical transient receptor potential 6 (TRPC6) channel-mediated cation entry sustains VSMCs contractile phenotype and promotes the blood flow recovery after hindlimb ischemia in mice. We also reported that Zn2+, a metal biomolecule mobilized by TRPC6 channel activation, exerts potential beneficial effects on cardiac contractility and remodeling. Therefore, we hypothesized that TRPC6-mediated Zn2+ influx participates in phenotype switching of VSMCs and vascular remodeling. We established rat aortic smooth muscle cells (RAoSMCs) stably expressing wild type (WT) and Zn2+ only impermeable TRPC6 (KYD) mutant. Although the resting phenotypes were similar in both RAoSMCs, pharmacological TRPC6 activation by PPZ2 prevented the transforming growth factor (TGF) β-induced reduction in the intracellular Zn2+ amount and contractile differentiation in RAoSMCs (WT), but failed to prevent them in RAoSMCs (KYD). There were no significant differences in TRPC6-dependent cation currents among all RAoSMCs pretreated with or without TGFβ and/or PPZ2, suggesting that TRPC6 channels are functionally expressed in RAoSMCs regardless of their phenotype. Treatment of mice with PPZ2 attenuated the progression of vascular remodeling caused by chronic angiotensin II infusion. These results suggest that Zn2+ influx through TRPC6 channels negatively regulates the TGFβ-induced contractile differentiation of VSMCs and the progression of vascular remodeling in rodents.
Collapse
Affiliation(s)
- Chenlin Su
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan (Y.K.)
| | - Xinya Mi
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan (Y.K.)
| | - Tomoya Ito
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan (Y.K.)
| | - Yuri Kato
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan (Y.K.)
| | - Akiyuki Nishimura
- National Institute for Physiological Science (NIPS), National Institutes of Natural Sciences, Okazaki 444-8787, Japan
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Science, Okazaki 444-8787, Japan
- Department of Physiological Sciences, SOKENDAI (School of Life Science, The Graduate University for Advanced Studies), Okazaki 444-8787, Japan
| | - Ryu Nagata
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan
| | - Yasuo Mori
- Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan
| | - Motohiro Nishida
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan (Y.K.)
- National Institute for Physiological Science (NIPS), National Institutes of Natural Sciences, Okazaki 444-8787, Japan
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Science, Okazaki 444-8787, Japan
- Department of Physiological Sciences, SOKENDAI (School of Life Science, The Graduate University for Advanced Studies), Okazaki 444-8787, Japan
| |
Collapse
|
2
|
Zhong X, Wu F, Gao W, Hu J, Shen B, Zhong K, Peng J, Zhang C, Zhang C. Effects of Extracellular Matrix Changes Induced by a High-Fat Diet on Gallbladder Smooth Muscle Dysfunction. FRONT BIOSCI-LANDMRK 2024; 29:401. [PMID: 39735983 DOI: 10.31083/j.fbl2912401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 09/12/2024] [Accepted: 09/20/2024] [Indexed: 12/31/2024]
Abstract
BACKGROUND Gallstone formation is a common digestive ailment, with unclear mechanisms underlying its development. Dysfunction of the gallbladder smooth muscle (GSM) may play a crucial role, particularly with a high-fat diet (HFD). This study aimed to investigate the effects of an HFD on GSM and assess how it alters contractility through changes in the extracellular matrix (ECM). METHODS Guinea pigs and C57BL/6 mice were fed either an HFD or normal diet (ND). Primary cultures of their (guinea pigs) gallbladder smooth muscle cells (GSMCs) were used for in vitro experiments. Histological stains, RNA-sequencing, bioinformatics analysis, three-dimensional tissue culture, real-time polymerase chain reaction (PCR), Western blot, atomic force microscopy, and muscle tension measurements were performed. RESULTS Histological evidence indicated structural changes in the gallbladder muscle layer and ECM collagen deposition in the HFD group. The HFD group also showed increased expression of collagen, integrin family, and matrix metalloproteinase (MMP) and the phosphoinositide 3-kinase (PI3K)-protein kinase B (PKB/Akt) signaling pathway. Compared with GSMCs cultured on Matrigel containing 1 mg/mL of collagen I, those cultured with 2 mg/mL showed a phenotype change from contractile to synthetic cells. Consistent with these findings, the HFD group also demonstrated increased ECM stiffness and decreased smooth muscle contractility. CONCLUSIONS Our findings reveal a mechanism by which an HFD alters the ECM composition of the gallbladder muscle, activating the integrin/PI3K-Akt/MMP signaling pathway, thereby impacting GSMC phenotype and contractility. These insights enhance the understanding of gallstone formation mechanism and provide potential therapeutic targets to treat gallbladder dysfunction.
Collapse
Affiliation(s)
- Xingguo Zhong
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, 230022 Hefei, Anhui, China
- Department of General Surgery, Anhui No. 2 Provincial People's Hospital, 230041 Hefei, Anhui, China
| | - Feiyang Wu
- Department of General Surgery, Anhui No. 2 Provincial People's Hospital, 230041 Hefei, Anhui, China
- Graduate School, Bengbu Medical University, 233030 Bengbu, Anhui, China
| | - Weicheng Gao
- Department of General Surgery, Anhui No. 2 Provincial People's Hospital, 230041 Hefei, Anhui, China
- Graduate School, Bengbu Medical University, 233030 Bengbu, Anhui, China
| | - Jinlong Hu
- Department of General Surgery, Anhui No. 2 Provincial People's Hospital, 230041 Hefei, Anhui, China
| | - Bing Shen
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, 999078 Taipa, Macao SAR, China
| | - Kaiyuan Zhong
- College of Medical Technology, Qiqihar Medical University, 161003 Qiqihar, Heilongjiang, China
| | - Junbin Peng
- Medical School, Anhui University of Science & Technology, 232001 Huainan, Anhui, China
| | - Chong Zhang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, 230022 Hefei, Anhui, China
| | - Chao Zhang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, 230022 Hefei, Anhui, China
| |
Collapse
|
3
|
Kuwabara S, Mizoguchi T, Ma J, Kanoh T, Ohta Y, Itoh M. Notch signaling pathway suppresses mRNA expression of hexokinase 2 under nutrient-poor conditions in U87-MG glioma cells. Genes Cells 2024. [PMID: 39462157 DOI: 10.1111/gtc.13176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 10/29/2024]
Abstract
Control of nutrient homeostasis plays a central role in cell proliferation/survival during embryonic development and tumor growth. Activation of the Notch signaling pathway, a major contributor to cell-cell interactions, is a potential mechanism for cell adaptation to nutrient-poor conditions. Our previous study also demonstrated that during embryogenesis when nutrients such as glutamine and growth factors are potentially maintained at lower levels, Notch signaling suppresses mRNA expression of hexokinase 2 (hk2), which is a glycolysis-associated gene, in the central nervous system. However, whether and how the genetic regulation of HK2 via Notch signaling contributes to cellular adaptability to nutrient-poor environments remains unknown. In this study, we performed gene expression analysis using a U87-MG human glioma cell line and revealed that under conditions where both glutamine and serum were absent, Notch signaling was activated and HK2 expression was downregulated by Notch signaling. We also found that Notch-mediated HK2 suppression was triggered in a Notch ligand-selective manner. Furthermore, HK2 was shown to inhibit cell proliferation of U87-MG gliomas, which might depend on Notch signaling activity. Together, our findings suggest the involvement of Notch-mediated HK2 suppression in an adaptive mechanism of U87-MG glioma cells to nutrient-poor conditions.
Collapse
Affiliation(s)
- Shuhei Kuwabara
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Takamasa Mizoguchi
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Jiawei Ma
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Tohgo Kanoh
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Yuki Ohta
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Motoyuki Itoh
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
- Research Institute of Disaster Medicine, Chiba University, Chiba, Japan
- Health and Disease Omics Center, Chiba University, Chiba, Japan
| |
Collapse
|
4
|
Qian L, Koval OM, Endoni BT, Juhr D, Stein CS, Allamargot C, Lin LH, Guo DF, Rahmouni K, Boudreau RL, Streeter J, Thiel WH, Grumbach IM. MIRO1 controls energy production and proliferation of smooth muscle cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.13.607854. [PMID: 39185180 PMCID: PMC11343164 DOI: 10.1101/2024.08.13.607854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Background The outer mitochondrial Rho GTPase 1, MIRO1, mediates mitochondrial motility within cells, but implications for vascular smooth muscle cell (VSMC) physiology and its roles invascular diseases, such as neointima formation following vascular injury are widely unknown. Methods An in vivo model of selective Miro1 deletion in VSMCs was generated, and the animals were subjected to carotid artery ligation. The molecular mechanisms relevant to VSMC proliferation were then explored in explanted VSMCs by imaging mitochondrial positioning and cristae structure and assessing the effects on ATP production, metabolic function and interactions with components of the electron transport chain (ETC). Results MIRO1 was robustly expressed in VSMCs within human atherosclerotic plaques and promoted VSMC proliferation and neointima formation in mice by blocking cell-cycle progression at G1/S, mitochondrial positioning, and PDGF-induced ATP production and respiration; overexpression of a MIRO1 mutant lacking the EF hands that are required for mitochondrial mobility did not fully rescue these effects. At the ultrastructural level, Miro1 deletion distorted the mitochondrial cristae and reduced the formation of super complexes and the activity of ETC complex I. Conclusions Mitochondrial motility is essential for VSMC proliferation and relies on MIRO1. The EF-hands of MIRO1 regulate the intracellular positioning of mitochondria. Additionally, the absence of MIRO1 leads to distorted mitochondrial cristae and reduced ATP generation. Our findings demonstrate that motility is linked to mitochondrial ATP production. We elucidated two unrecognized mechanisms through which MIRO1 influences cell proliferation by modulating mitochondria: first, by managing mitochondrial placement via Ca2+-dependent EF hands, and second, by affecting cristae structure and ATP synthesis.
Collapse
Affiliation(s)
- Lan Qian
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City IA 52242, USA
| | - Olha M. Koval
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City IA 52242, USA
| | - Benney T. Endoni
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City IA 52242, USA
- Interdisciplinary Program in Molecular Medicine, University of Iowa
| | - Denise Juhr
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City IA 52242, USA
| | - Colleen S. Stein
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City IA 52242, USA
| | | | - Li-Hsien Lin
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City IA 52242, USA
| | - Deng-Fu Guo
- Department of Neuroscience and Pharmacology, University of Iowa
| | - Kamal Rahmouni
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City IA 52242, USA
- Interdisciplinary Program in Molecular Medicine, University of Iowa
- Department of Neuroscience and Pharmacology, University of Iowa
- Veterans Affairs Healthcare System, Iowa City, IA 52246, USA
| | - Ryan L. Boudreau
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City IA 52242, USA
| | - Jennifer Streeter
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City IA 52242, USA
| | - William H. Thiel
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City IA 52242, USA
| | - Isabella M. Grumbach
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City IA 52242, USA
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City IA 52242, USA
- Veterans Affairs Healthcare System, Iowa City, IA 52246, USA
| |
Collapse
|
5
|
Muderrisoglu AE, Ciotkowska A, Rutz B, Hu S, Qian S, Tamalunas A, Stief CG, Hennenberg M. Dynamic phenotypic shifts and M2 receptor downregulation in bladder smooth muscle cells induced by mirabegron. Front Pharmacol 2024; 15:1446831. [PMID: 39114356 PMCID: PMC11303193 DOI: 10.3389/fphar.2024.1446831] [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: 06/10/2024] [Accepted: 07/11/2024] [Indexed: 08/10/2024] Open
Abstract
Introduction Mirabegron is available for treatment of overactive bladder (OAB). However, mechanisms underlying symptom improvements and long-term effects on bladder smooth muscle cells are uncertain. Contractility and growth of bladder smooth muscle contribute to OAB, and depend on smooth muscle phenotypes, and on muscarinic receptor expression. Here, we examined prolonged exposure to mirabegron (20-48 h) on phenotype markers, muscarinic receptor expression, and phenotype-dependent functions in human bladder smooth muscle cells (hBSMC). Methods Expression of markers for contractile (calponin, MYH11) and proliferative (MYH10, vimentin) phenotypes, proliferation (Ki-67), and of muscarinic receptors were assessed by RT-PCR. Proliferation, viability, actin organization and contractions in cultured hBSMC were examined by EdU, CCK-8, phalloidin staining and matrix contraction assays. Results Calponin-1 mRNA decreased with 100 nM and 150 nM mirabegron applied for 20 h (0.56-0.6 fold of controls). Decreases were resistant to the β3-AR antagonist L-748,337 (0.34-0.55 fold, 100-150 nM, 20 h). After 40 h, decreases occured in the presence of L-748,337, but not without L-748,337. MYH11 mRNA increased with 150 nM mirabegron (40 h, 1.9 fold). This was partly preserved with L-748,337, but not observed after 20 h mirabegron exposure. Vimentin mRNA reduced with 150 nM mirabegron after 20 h, but not after 40 h, with and without L-748,337 (0.71-0.63 fold). MYH10 mRNA expression remained unaffected by mirabegron. Exposure to 150 nM mirabegron increased Ki-67 mRNA after 20 h in the presence of, but not without L-748,337, and after 40 h without, but not with L-748,337. Proliferation rates and actin organization were stable with 50-150 nM mirabegron (24 h, 48 h). Viability increased significantly after mirabegron exposure for 20 h, and by trend after 40 h, which was fully sensitive to L-748,337. M2 mRNA was reduced by 20 h mirabegron, which was resistant to L-748,337. Carbachol (3 µM) enhanced time-dependent contractions of hBSMC, which was inhibited by mirabegron (150 nM) in late phases (24 h), but not in early phases of contractions. Conclusion: Mirabegron induces dynamic phenotype alterations and M2 downregulation in hBSMC, which is paralleled by time-shifted anticontractile effects. Phenotype transitions may be involved in improvements of storage symptoms in OAB by mirabegron.
Collapse
Affiliation(s)
- A. E. Muderrisoglu
- Department of Urology, LMU University Hospital, LMU Munich, Munich, Germany
- Department of Medical Pharmacology, Istanbul Medipol University, Istanbul, Türkiye
| | - A. Ciotkowska
- Department of Urology, LMU University Hospital, LMU Munich, Munich, Germany
| | - B. Rutz
- Department of Urology, LMU University Hospital, LMU Munich, Munich, Germany
| | - S. Hu
- Department of Urology, LMU University Hospital, LMU Munich, Munich, Germany
| | - S. Qian
- Department of Urology, LMU University Hospital, LMU Munich, Munich, Germany
| | - A. Tamalunas
- Department of Urology, LMU University Hospital, LMU Munich, Munich, Germany
| | - C. G. Stief
- Department of Urology, LMU University Hospital, LMU Munich, Munich, Germany
| | - M. Hennenberg
- Department of Urology, LMU University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
6
|
Sarkar A, Pawar SV, Chopra K, Jain M. Gamut of glycolytic enzymes in vascular smooth muscle cell proliferation: Implications for vascular proliferative diseases. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167021. [PMID: 38216067 DOI: 10.1016/j.bbadis.2024.167021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 01/05/2024] [Accepted: 01/05/2024] [Indexed: 01/14/2024]
Abstract
Vascular smooth muscle cells (VSMCs) are the predominant cell type in the media of the blood vessels and are responsible for maintaining vascular tone. Emerging evidence confirms that VSMCs possess high plasticity. During vascular injury, VSMCs switch from a "contractile" phenotype to an extremely proliferative "synthetic" phenotype. The balance between both strongly affects the progression of vascular remodeling in many cardiovascular pathologies such as restenosis, atherosclerosis and aortic aneurism. Proliferating cells demand high energy requirements and to meet this necessity, alteration in cellular bioenergetics seems to be essential. Glycolysis, fatty acid metabolism, and amino acid metabolism act as a fuel for VSMC proliferation. Metabolic reprogramming of VSMCs is dynamically variable that involves multiple mechanisms and encompasses the coordination of various signaling molecules, proteins, and enzymes. Here, we systemically reviewed the metabolic changes together with the possible treatments that are still under investigation underlying VSMC plasticity which provides a promising direction for the treatment of diseases associated with VSMC proliferation. A better understanding of the interaction between metabolism with associated signaling may uncover additional targets for better therapeutic strategies in vascular disorders.
Collapse
Affiliation(s)
- Ankan Sarkar
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Sandip V Pawar
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Kanwaljit Chopra
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Manish Jain
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India.
| |
Collapse
|
7
|
Liu D, Jing Y, Peng G, Wei L, Zheng L, Chang G, Wang M. MiR-199a-5p Deficiency Promotes Artery Restenosis in Peripheral Artery Disease by Regulating ASMCs Function via Targeting HIF-1α and E2F3. Curr Vasc Pharmacol 2024; 22:342-354. [PMID: 38910413 DOI: 10.2174/0115701611280634240616062413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 04/12/2024] [Accepted: 05/15/2024] [Indexed: 06/25/2024]
Abstract
BACKGROUND Restenosis (RS) poses a significant concern, leading to recurrent ischemia and the potential for amputation following intraluminal angioplasty in the treatment of Peripheral Artery Disease (PAD). Through microRNA microarray analysis, the study detected a significant downregulation of miR-199a-5p within arterial smooth muscle cells (ASMCs) associated with RS. OBJECTIVE This research aims to explore the possible function and the underlying mechanisms of miR-199a-5p in the context of RS. METHODS Primary ASMCs were extracted from the femoral arteries of both healthy individuals and patients with PAD or RS. The expression levels of miR-199a-5p were assessed using both qRT-PCR and in situ hybridization techniques. To examine the impacts of miR-199a-5p, a series of experiments were performed, including flow cytometry, TUNEL assay, EdU assay, CCK8 assay, Transwell assay, and wound closure assay. A rat carotid balloon injury model was employed to elucidate the mechanism through which miR-199a-5p mitigated neointimal hyperplasia. RESULTS MiR-199a-5p exhibited downregulation in RS patients and was predominantly expressed within ASMCs. Elevated the expression of miR-199a-5p resulted in an inhibitory effect of proliferation and migration in ASMCs. Immunohistochemistry and a dual-luciferase reporter assay uncovered that RS exhibited elevated expression levels of both HIF-1α and E2F3, and they were identified as target genes regulated by miR-199a-5p. The co-transfection of lentiviruses carrying HIF-1α and E2F3 alongside miR-199a-5p further elucidated their role in the cellular responses mediated by miR-199a-5p. In vivo, the delivery of miR-199a-5p via lentivirus led to the mitigation of neointimal formation following angioplasty, achieved by targeting HIF-1α and E2F3. CONCLUSION MiR-199a-5p exhibits promise as a prospective therapeutic target for RS since it alleviates the condition by inhibiting the proliferation and migration of ASMCs via its regulation of HIF-1α and E2F3.
Collapse
MESH Headings
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Animals
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Cell Proliferation
- Peripheral Arterial Disease/genetics
- Peripheral Arterial Disease/pathology
- Peripheral Arterial Disease/metabolism
- Peripheral Arterial Disease/therapy
- Disease Models, Animal
- Male
- Cell Movement
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Rats, Sprague-Dawley
- Cells, Cultured
- E2F3 Transcription Factor/genetics
- E2F3 Transcription Factor/metabolism
- Middle Aged
- Signal Transduction
- Case-Control Studies
- Femoral Artery/pathology
- Femoral Artery/metabolism
- Femoral Artery/surgery
- Femoral Artery/physiopathology
- Neointima
- Female
- Carotid Artery Injuries/genetics
- Carotid Artery Injuries/pathology
- Carotid Artery Injuries/metabolism
- Aged
- Angioplasty, Balloon/adverse effects
- Apoptosis/genetics
Collapse
Affiliation(s)
- Duan Liu
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
- Division of Vascular Surgery, Xuanwu Hospital, Capital Medical University and Institute of Vascular Surgery, Capital Medical University, Beijing, 100053, China
| | - Yexiang Jing
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Guiyan Peng
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Litai Wei
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Liang Zheng
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Guangqi Chang
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Mian Wang
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| |
Collapse
|
8
|
Li S, Li J, Cheng W, He W, Dai SS. Independent and Interactive Roles of Immunity and Metabolism in Aortic Dissection. Int J Mol Sci 2023; 24:15908. [PMID: 37958896 PMCID: PMC10647240 DOI: 10.3390/ijms242115908] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 10/22/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Aortic dissection (AD) is a cardiovascular disease that seriously endangers the lives of patients. The mortality rate of this disease is high, and the incidence is increasing annually, but the pathogenesis of AD is complicated. In recent years, an increasing number of studies have shown that immune cell infiltration in the media and adventitia of the aorta is a novel hallmark of AD. These cells contribute to changes in the immune microenvironment, which can affect their own metabolism and that of parenchymal cells in the aortic wall, which are essential factors that induce degeneration and remodeling of the vascular wall and play important roles in the formation and development of AD. Accordingly, this review focuses on the independent and interactive roles of immunity and metabolism in AD to provide further insights into the pathogenesis, novel ideas for diagnosis and new strategies for treatment or early prevention of AD.
Collapse
Affiliation(s)
- Siyu Li
- School of Medicine, Chongqing University, Chongqing 400044, China
- Department of Biochemistry and Molecular Biology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jun Li
- Department of Cardiac Surgery, The First Affiliated Hospital of Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Wei Cheng
- Department of Cardiac Surgery, The First Affiliated Hospital of Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Wenhui He
- Department of Biochemistry and Molecular Biology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Shuang-Shuang Dai
- School of Medicine, Chongqing University, Chongqing 400044, China
- Department of Biochemistry and Molecular Biology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| |
Collapse
|
9
|
Tang W, Zhang L, Li J, Guan Y. KCNQ1OT1 promotes retinoblastoma progression by targeting miR-339-3p that suppresses KIF23. Int Ophthalmol 2023:10.1007/s10792-023-02641-1. [PMID: 37198502 DOI: 10.1007/s10792-023-02641-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 01/19/2023] [Indexed: 05/19/2023]
Abstract
BACKGROUND Long noncoding RNAs (lncRNAs) are involved in tumor formation and development. KCNQ1OT1 regulates the malignant proliferation of retinoblastoma (RB), but the specific mechanism remains to be further investigated. METHODS The KCNQ1OT1, miR-339-3p and KIF23 expression levels in RB were detected by qRT-PCR and western blotting. The cell viability, proliferation, migration ability and caspase-3 activity of RB cells were evaluated by CCK-8, BrdU, transwell and caspase-3 activity analysis. Western blot was used to detect the Bax and Bcl-2 protein expression in RB cells. The binding relationship between KCNQ1OT1, miR-339-3p and KIF23 was detected by luciferase, RIP and RNA pull-down assay. RESULTS KCNQ1OT1 and KIF23 were up-regulated frequently in RB, and miR-339-3p was down-regulated. Functional studies showed that downregulation of KCNQ1OT1 or KIF23 inhibited the survival and migration of RB cells, and facilitated apoptosis. Interference with miR-339-3p showed the opposite effect. Mechanisms suggested that KCNQ1OT1 exited its oncogenic activity by positively regulating the expression of KIF23 and sponging miR-339-3p. CONCLUSION KCNQ1OT1/miR-339-3p/KIF23 may be a new biomarker for the diagnosis and treatment of RB.
Collapse
Affiliation(s)
- Wenting Tang
- Department of Ophthalmology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, Sichuan, China
| | - Li Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, Sichuan, China
| | - Jing Li
- Department of Ophthalmology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, Sichuan, China
| | - Yu Guan
- Department of Ophthalmology, The 2nd Affiliated Hospital of Chengdu Medical College, Nuclear Industry 416 Hospital, No. 4, North 4th Erhuan Street, Chengdu, 610051, Sichuan, China.
| |
Collapse
|
10
|
Cao G, Xuan X, Li Y, Hu J, Zhang R, Jin H, Dong H. Single-cell RNA sequencing reveals the vascular smooth muscle cell phenotypic landscape in aortic aneurysm. Cell Commun Signal 2023; 21:113. [PMID: 37189183 DOI: 10.1186/s12964-023-01120-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 03/30/2023] [Indexed: 05/17/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Phenotypic switching in vascular smooth muscle cells (VSMCs) has been linked to aortic aneurysm, but the phenotypic landscape in aortic aneurysm is poorly understood. The present study aimed to analyse the phenotypic landscape, phenotypic differentiation trajectory, and potential functions of various VSMCs phenotypes in aortic aneurysm. METHODS Single-cell sequencing data of 12 aortic aneurysm samples and 5 normal aorta samples (obtained from GSE166676 and GSE155468) were integrated by the R package Harmony. VSMCs were identified according to the expression levels of ACTA2 and MYH11. VSMCs clustering was determined by the R package 'Seurat'. Cell annotation was determined by the R package 'singleR' and background knowledge of VSMCs phenotypic switching. The secretion of collagen, proteinases, and chemokines by each VSMCs phenotype was assessed. Cell‒cell junctions and cell-matrix junctions were also scored by examining the expression of adhesion genes. Trajectory analysis was performed by the R package 'Monocle2'. qPCR was used to quantify VSMCs markers. RNA fluorescence in situ hybridization (RNA FISH) was performed to determine the spatial localization of vital VSMCs phenotypes in aortic aneurysms. RESULTS A total of 7150 VSMCs were categorize into 6 phenotypes: contractile VSMCs, fibroblast-like VSMCs, T-cell-like VSMCs, adipocyte-like VSMCs, macrophage-like VSMCs, and mesenchymal-like VSMCs. The proportions of T-cell-like VSMCs, adipocyte-like VSMCs, macrophage-like VSMCs, and mesenchymal-like VSMCs were significantly increased in aortic aneurysm. Fibroblast-like VSMCs secreted abundant amounts of collagens. T-cell-like VSMCs and macrophage-like VSMCs were characterized by high chemokine levels and proinflammatory effects. Adipocyte-like VSMCs and mesenchymal-like VSMCs were associated with high proteinase levels. RNA FISH validated the presence of T-cell-like VSMCs and macrophage-like VSMCs in the tunica media and the presence of mesenchymal-like VSMCs in the tunica media and tunica adventitia. CONCLUSION A variety of VSMCs phenotypes are involved in the formation of aortic aneurysm. T-cell-like VSMCs, macrophage-like VSMCs, and mesenchymal-like VSMCs play pivotal roles in this process. Video Abstract.
Collapse
Affiliation(s)
- Genmao Cao
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, No. 382, Wuyi Road, Taiyuan, China
| | - Xuezhen Xuan
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, No. 382, Wuyi Road, Taiyuan, China
| | - Yaling Li
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, No. 382, Wuyi Road, Taiyuan, China
| | - Jie Hu
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, No. 382, Wuyi Road, Taiyuan, China
| | - Ruijing Zhang
- Department of Nephrology, The Second Hospital of Shanxi Medical University, No. 382, Wuyi Road, Taiyuan, China
| | - Haijiang Jin
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, No. 382, Wuyi Road, Taiyuan, China
| | - Honglin Dong
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, No. 382, Wuyi Road, Taiyuan, China.
| |
Collapse
|
11
|
Tang HY, Chen AQ, Zhang H, Gao XF, Kong XQ, Zhang JJ. Vascular Smooth Muscle Cells Phenotypic Switching in Cardiovascular Diseases. Cells 2022; 11:cells11244060. [PMID: 36552822 PMCID: PMC9777337 DOI: 10.3390/cells11244060] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/02/2022] [Accepted: 12/13/2022] [Indexed: 12/16/2022] Open
Abstract
Vascular smooth muscle cells (VSMCs), the major cell type in the arterial vessel wall, have a contractile phenotype that maintains the normal vessel structure and function under physiological conditions. In response to stress or vascular injury, contractile VSMCs can switch to a less differentiated state (synthetic phenotype) to acquire the proliferative, migratory, and synthetic capabilities for tissue reparation. Imbalances in VSMCs phenotypic switching can result in a variety of cardiovascular diseases, including atherosclerosis, in-stent restenosis, aortic aneurysms, and vascular calcification. It is very important to identify the molecular mechanisms regulating VSMCs phenotypic switching to prevent and treat cardiovascular diseases with high morbidity and mortality. However, the key molecular mechanisms and signaling pathways participating in VSMCs phenotypic switching have still not been fully elucidated despite long-term efforts by cardiovascular researchers. In this review, we provide an updated summary of the recent studies and systematic knowledge of VSMCs phenotypic switching in atherosclerosis, in-stent restenosis, aortic aneurysms, and vascular calcification, which may help guide future research and provide novel insights into the prevention and treatment of related diseases.
Collapse
Affiliation(s)
- Hao-Yue Tang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing 210006, China
| | - Ai-Qun Chen
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing 210006, China
| | - Huan Zhang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing 210006, China
| | - Xiao-Fei Gao
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing 210006, China
- Department of Cardiology, Nanjing Heart Centre, No. 68 Changle Road, Nanjing 210006, China
| | - Xiang-Quan Kong
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing 210006, China
| | - Jun-Jie Zhang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing 210006, China
- Department of Cardiology, Nanjing Heart Centre, No. 68 Changle Road, Nanjing 210006, China
- Correspondence: or ; Tel./Fax: +86-25-52208048
| |
Collapse
|
12
|
Ai X, Yu P, Peng L, Luo L, Liu J, Li S, Lai X, Luan F, Meng X. Berberine: A Review of its Pharmacokinetics Properties and Therapeutic Potentials in Diverse Vascular Diseases. Front Pharmacol 2022; 12:762654. [PMID: 35370628 PMCID: PMC8964367 DOI: 10.3389/fphar.2021.762654] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/20/2021] [Indexed: 12/11/2022] Open
Abstract
Traditional Chinese medicine plays a significant role in the treatment of various diseases and has attracted increasing attention for clinical applications. Vascular diseases affecting vasculature in the heart, cerebrovascular disease, atherosclerosis, and diabetic complications have compromised quality of life for affected individuals and increase the burden on health care services. Berberine, a naturally occurring isoquinoline alkaloid form Rhizoma coptidis, is widely used in China as a folk medicine for its antibacterial and anti-inflammatory properties. Promisingly, an increasing number of studies have identified several cellular and molecular targets for berberine, indicating its potential as an alternative therapeutic strategy for vascular diseases, as well as providing novel evidence that supports the therapeutic potential of berberine to combat vascular diseases. The purpose of this review is to comprehensively and systematically describe the evidence for berberine as a therapeutic agent in vascular diseases, including its pharmacological effects, molecular mechanisms, and pharmacokinetics. According to data published so far, berberine shows remarkable anti-inflammatory, antioxidant, antiapoptotic, and antiautophagic activity via the regulation of multiple signaling pathways, including AMP-activated protein kinase (AMPK), nuclear factor κB (NF-κB), mitogen-activated protein kinase silent information regulator 1 (SIRT-1), hypoxia-inducible factor 1α (HIF-1α), vascular endothelial growth factor phosphoinositide 3-kinase (PI3K), protein kinase B (Akt), janus kinase 2 (JAK-2), Ca2+ channels, and endoplasmic reticulum stress. Moreover, we discuss the existing limitations of berberine in the treatment of vascular diseases, and give corresponding measures. In addition, we propose some research perspectives and challenges, and provide a solid evidence base from which further studies can excavate novel effective drugs from Chinese medicine monomers.
Collapse
Affiliation(s)
- Xiaopeng Ai
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Peiling Yu
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Lixia Peng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Liuling Luo
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jia Liu
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shengqian Li
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xianrong Lai
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fei Luan
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xianli Meng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
13
|
Metabolism in atherosclerotic plaques: immunoregulatory mechanisms in the arterial wall. Clin Sci (Lond) 2022; 136:435-454. [PMID: 35348183 PMCID: PMC8965849 DOI: 10.1042/cs20201293] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 03/02/2022] [Accepted: 03/16/2022] [Indexed: 02/05/2023]
Abstract
Over the last decade, there has been a growing interest to understand the link between metabolism and the immune response in the context of metabolic diseases but also beyond, giving then birth to a new field of research. Termed 'immunometabolism', this interdisciplinary field explores paradigms of both immunology and metabolism to provided unique insights into different disease pathogenic processes, and the identification of new potential therapeutic targets. Similar to other inflammatory conditions, the atherosclerotic inflammatory process in the artery has been associated with a local dysregulated metabolic response. Thus, recent studies show that metabolites are more than just fuels in their metabolic pathways, and they can act as modulators of vascular inflammation and atherosclerosis. In this review article, we describe the most common immunometabolic pathways characterised in innate and adaptive immune cells, and discuss how macrophages' and T cells' metabolism may influence phenotypic changes in the plaque. Moreover, we discuss the potential of targeting immunometabolism to prevent and treat cardiovascular diseases (CVDs).
Collapse
|
14
|
Chen X, Luo J, Liu J, Chen T, Sun J, Zhang Y, Xi Q. Exploration of the Effect on Genome-Wide DNA Methylation by miR-143 Knock-Out in Mice Liver. Int J Mol Sci 2021; 22:13075. [PMID: 34884879 PMCID: PMC8658369 DOI: 10.3390/ijms222313075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/30/2021] [Accepted: 12/01/2021] [Indexed: 12/12/2022] Open
Abstract
MiR-143 play an important role in hepatocellular carcinoma and liver fibrosis via inhibiting hepatoma cell proliferation. DNA methyltransferase 3 alpha (DNMT3a), as a target of miR-143, regulates the development of primary organic solid tumors through DNA methylation mechanisms. However, the effect of miR-143 on DNA methylation profiles in liver is unclear. In this study, we used Whole-Genome Bisulfite Sequencing (WGBS) to detect the differentially methylated regions (DMRs), and investigated DMR-related genes and their enriched pathways by miR-143. We found that methylated cytosines increased 0.19% in the miR-143 knock-out (KO) liver fed with high-fat diet (HFD), compared with the wild type (WT). Furthermore, compared with the WT group, the CG methylation patterns of the KO group showed lower CG methylation levels in CG islands (CGIs), promoters and hypermethylation in CGI shores, 5'UTRs, exons, introns, 3'UTRs, and repeat regions. A total of 984 DMRs were identified between the WT and KO groups consisting of 559 hypermethylation and 425 hypomethylation DMRs. Furthermore, DMR-related genes were enriched in metabolism pathways such as carbon metabolism (serine hydroxymethyltransferase 2 (Shmt2), acyl-Coenzyme A dehydrogenase medium chain (Acadm)), arginine and proline metabolism (spermine synthase (Sms), proline dehydrogenase (Prodh2)) and purine metabolism (phosphoribosyl pyrophosphate synthetase 2 (Prps2)). In summary, we are the first to report the change in whole-genome methylation levels by miR-143-null through WGBS in mice liver, and provide an experimental basis for clinical diagnosis and treatment in liver diseases, indicating that miR-143 may be a potential therapeutic target and biomarker for liver damage-associated diseases and hepatocellular carcinoma.
Collapse
Affiliation(s)
| | | | | | | | | | - Yongliang Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou 510642, China; (X.C.); (J.L.); (J.L.); (T.C.); (J.S.)
| | - Qianyun Xi
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou 510642, China; (X.C.); (J.L.); (J.L.); (T.C.); (J.S.)
| |
Collapse
|
15
|
Sorrentino S, Polini A, Arima V, Romano A, Quattrini A, Gigli G, Mozetic P, Moroni L. Neurovascular signals in amyotrophic lateral sclerosis. Curr Opin Biotechnol 2021; 74:75-83. [PMID: 34800850 DOI: 10.1016/j.copbio.2021.10.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/24/2021] [Accepted: 10/22/2021] [Indexed: 12/21/2022]
Abstract
The neurovascular system (NVS) is a complex anatomic-functional unit that synergically works to maintain organs/tissues homeostasis of the entire body. NVS alterations have recently emerged as a common distinct feature in the pathogenesis of several neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). Despite their undeniable involvement, neurovascular signalling pathways remain still far unknown in ALS. This review underlines the importance of endothelial, mural, and fibroblast cells as novel targets for ALS investigation and identifies in the interplay between neuronal and vascular systems the way to disclose novel molecular mechanisms behind the pathogenesis of ALS.
Collapse
Affiliation(s)
- Stefano Sorrentino
- CNR Nanotec - Institute of Nanotechnology, Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Alessandro Polini
- CNR Nanotec - Institute of Nanotechnology, Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Valentina Arima
- CNR Nanotec - Institute of Nanotechnology, Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Alessandro Romano
- San Raffaele Hospital, Division of Neuroscience, Institute of Experimental Neurology, San Rafaele Scientifc Institute, Via Olgettina, 60, 20132, Milan, Italy
| | - Angelo Quattrini
- San Raffaele Hospital, Division of Neuroscience, Institute of Experimental Neurology, San Rafaele Scientifc Institute, Via Olgettina, 60, 20132, Milan, Italy
| | - Giuseppe Gigli
- CNR Nanotec - Institute of Nanotechnology, Campus Ecotekne, via Monteroni, Lecce, 73100, Italy; Department of Mathematics and Physics "Ennio De Giorgi", University of Salento, via Arnesano, 73100, Lecce, Italy
| | - Pamela Mozetic
- CNR Nanotec - Institute of Nanotechnology, Campus Ecotekne, via Monteroni, Lecce, 73100, Italy; San Raffaele Hospital, Division of Neuroscience, Institute of Experimental Neurology, San Rafaele Scientifc Institute, Via Olgettina, 60, 20132, Milan, Italy
| | - Lorenzo Moroni
- CNR Nanotec - Institute of Nanotechnology, Campus Ecotekne, via Monteroni, Lecce, 73100, Italy; Maastricht University, MERLN Institute for Technology-Inspired Regenerative Medicine, Complex Tissue Regeneration Department, Universiteitssingel 40, 6229ER, Maastricht, The Netherlands.
| |
Collapse
|