1
|
Cho JM, Vu K, Park SK, Zhu E, Li YR, Zhao P, Yokota T, Yang L, Lu R, Xiang YK, Shen YH, Chapleau MW, Hsiai TK. Habitual Exercise Modulates Neuroimmune Interaction to Mitigate Aortic Stiffness. Circ Res 2025. [PMID: 40304034 DOI: 10.1161/circresaha.124.325656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 04/04/2025] [Accepted: 04/10/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND Exercise augments hemodynamic shear to activate mechano-sensitive molecular transducers in the vascular endothelium. Recently, the central nervous system has been reported to mediate neuroimmune interaction in the aortic adventitia (AA). Whether exercise modulates the sympathetic nerve interaction with the immune cells to mitigate aortic stiffness remains unknown. METHODS AND RESULTS Four weeks of Ang II (angiotensin II) infusion to C57BL/6 mice increased neural activation to increase the expression of TH (tyrosine hydroxylase) for sympathetic nerve axons and norepinephrine levels along with the colocalization of synapsin and β2-AR (β2-adrenergic receptor)-positive macrophages in the AA. This Ang II-mediated sympathetic nerve and macrophage interaction activated fibroblasts to increase vascular fibrosis and arterial pulse wave velocity. Sympathetic denervation with celiac ganglionectomy or 6-hydroxydopamine treatment abrogated Ang II-mediated TH+, AA thickness, and pulse wave velocity. Single-cell RNA sequencing analyses of the AA revealed that Ang II increased the circulating monocyte-derived macrophages (Ccr2+CD80) but reduced the resident macrophages (Lyve1+CD163). Gene ontology analysis of differentially expressed genes unveiled that voluntary wheel running mitigated Ang II-mediated increase in Ccr2+CD80 macrophages, cytokine-mediated signaling pathways in macrophages, and extracellular matrix deposition in fibroblasts. Macrophage depletion with Ki20227 (colony stimulating factor 1 receptor inhibitor) reduced Ang II-mediated synapsin+ macrophages. Using the Ccr2 knock-in (Ccr2gfp)/knock-out (Ccr2KO) mice, we observed that Ang II-mediated increases in Ccr2+ macrophages were expressed in Ccr2gfp mice but were absent in Ccr2KO mice. Also, Ang II-induced increases in synapsin expression, neighboring Ccr2+ cells, AA thickness, and pulse wave velocity were reduced in Ccr2KO mice. Both Ki20227 and Ccr2KO reduced the Ang II-mediated increase in TH levels. Furthermore, voluntary wheel running-mediated reduction in vascular fibrosis and aortic stiffness were mitigated by a β2-AR agonist, terbutaline, indicating β2-AR in neuroimmune modulation. CONCLUSIONS Exercise mitigates Ang II-mediated sympathetic axon interaction with the circulating monocyte-derived macrophages in the AA to attenuate vascular fibrosis and aortic stiffness.
Collapse
Affiliation(s)
- Jae Min Cho
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles. (J.M.C., K.V., S.-K.P., E.Z., P.Z., T.Y., T.K.H.)
- Department of Medicine, Greater Los Angeles VA Healthcare System, CA (J.M.C., S.-K.P., E.Z., P.Z., T.Y., T.K.H.)
| | - Khoa Vu
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles. (J.M.C., K.V., S.-K.P., E.Z., P.Z., T.Y., T.K.H.)
| | - Seul-Ki Park
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles. (J.M.C., K.V., S.-K.P., E.Z., P.Z., T.Y., T.K.H.)
- Department of Medicine, Greater Los Angeles VA Healthcare System, CA (J.M.C., S.-K.P., E.Z., P.Z., T.Y., T.K.H.)
| | - Enbo Zhu
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles. (J.M.C., K.V., S.-K.P., E.Z., P.Z., T.Y., T.K.H.)
- Department of Medicine, Greater Los Angeles VA Healthcare System, CA (J.M.C., S.-K.P., E.Z., P.Z., T.Y., T.K.H.)
| | - Yan-Ruide Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles. (Y.-R.L., L.Y.)
| | - Peng Zhao
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles. (J.M.C., K.V., S.-K.P., E.Z., P.Z., T.Y., T.K.H.)
- Department of Medicine, Greater Los Angeles VA Healthcare System, CA (J.M.C., S.-K.P., E.Z., P.Z., T.Y., T.K.H.)
| | - Tomohiro Yokota
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles. (J.M.C., K.V., S.-K.P., E.Z., P.Z., T.Y., T.K.H.)
- Department of Medicine, Greater Los Angeles VA Healthcare System, CA (J.M.C., S.-K.P., E.Z., P.Z., T.Y., T.K.H.)
| | - Lili Yang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles. (Y.-R.L., L.Y.)
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles. (L.Y.)
- Molecular Biology Institute, University of California, Los Angeles. (L.Y.)
- Department of Medicine, Greater Los Angeles VA Healthcare System, CA (J.M.C., S.-K.P., E.Z., P.Z., T.Y., T.K.H.)
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, UCLA (L.Y.)
| | - Rong Lu
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles (R.L.)
| | - Yang Kevin Xiang
- Department of Pharmacology, University of California, Davis (Y.K.X.)
- VA Northern California Healthcare System, Mather, CA (Y.K.X.)
| | - Ying H Shen
- Department of Surgery, Baylor College of Medicine, Houston, TX (Y.H.S.)
| | - Mark W Chapleau
- Departments of Internal Medicine and Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine (M.W.C.)
| | - Tzung K Hsiai
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles. (J.M.C., K.V., S.-K.P., E.Z., P.Z., T.Y., T.K.H.)
- Department of Bioengineering, School of Engineering and Applied Science, University of California, Los Angeles. (T.K.H.)
- Department of Medicine, Greater Los Angeles VA Healthcare System, CA (J.M.C., S.-K.P., E.Z., P.Z., T.Y., T.K.H.)
- Medical Engineering, California Institute of Technology, Pasadena, CA (T.K.H.)
| |
Collapse
|
2
|
Brown TK, Dang C, Del Carmen A, Alharbi S, Chao CL, Xiong L, John NW, Smires A, Ho KJ, Jiang B. Mice Models for Peripheral Denervation to Enhance Vascular Regeneration. Tissue Eng Part C Methods 2025; 31:119-129. [PMID: 40062562 DOI: 10.1089/ten.tec.2025.0014] [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] [Indexed: 03/20/2025] Open
Abstract
Sympathetic innervation plays a critical role in regulating vascular function, yet its influence on vascular regeneration and reinnervation following ischemic injury remains poorly understood. This study develops and validates murine models of localized sympathetic denervation using 6-hydroxydopamine (6-OHDA) to enable study of the sympathetic nervous system's impact on vascular systems during tissue repair. Two methods of 6-OHDA administration were employed: a single topical application during open surgery and minimally invasive weekly subcutaneous injections. The topical application model achieved temporary denervation lasting 1 week without causing vascular damage, while the subcutaneous injection model provided sustained denervation for up to 4 weeks with minimal inflammation and no significant changes to vascular architecture. To investigate the effects of denervation in an ischemic context, these models were combined with a hindlimb ischemia model. Ischemia induced persistent denervation in both 6-OHDA-treated and control limbs, with limited sympathetic nerve regeneration observed over 4 weeks. Despite persistent denervation, microvascular density and perfusion recovery in ischemic muscles were comparable between denervated and control groups. This suggests that ischemia governs vascular regeneration independently of sympathetic input. These results demonstrate that localized 6-OHDA administration provides a versatile tool for achieving controlled sympathetic denervation in peripheral arteries. These models provide a novel platform for studying vascular regeneration and reinnervation under both normal and ischemic conditions, offering novel insights into the interactions between neural regulation and vascular repair processes. This work lays the foundation for future research into neural-vascular crosstalk and new possibilities for developing regenerative therapies targeting the autonomic regulation of vascular health.
Collapse
Affiliation(s)
- Taylor K Brown
- Department of Biomedical Engineering, Northwestern University, Chicago, Illinois, USA
| | - Caitlyn Dang
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Aurea Del Carmen
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Sara Alharbi
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Calvin L Chao
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Liqun Xiong
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Nikita Wilson John
- Department of Biomedical Engineering, Northwestern University, Chicago, Illinois, USA
| | - Aidan Smires
- Department of Biomedical Engineering, Northwestern University, Chicago, Illinois, USA
| | - Karen J Ho
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Bin Jiang
- Department of Biomedical Engineering, Northwestern University, Chicago, Illinois, USA
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
3
|
Puertas-Umbert L, Alonso J, Blanco-Casoliva L, Almendra-Pegueros R, Camacho M, Rodríguez-Sinovas A, Galán M, Roglans N, Laguna JC, Martínez-González J, Rodríguez C. Inhibition of ATP-citrate lyase by bempedoic acid protects against abdominal aortic aneurysm formation in mice. Biomed Pharmacother 2025; 184:117876. [PMID: 39889383 DOI: 10.1016/j.biopha.2025.117876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/16/2025] [Accepted: 01/27/2025] [Indexed: 02/03/2025] Open
Abstract
Abdominal aortic aneurysm (AAA) is a prevalent degenerative disease characterized by an exacerbated inflammation and destructive vascular remodeling. Unfortunately, effective pharmacological tools for the treatment of this disease remain a challenge. ATP-citrate lyase (ACLY), the primary enzyme responsible for acetyl-CoA biosynthesis, is a key regulator of inflammatory signaling in macrophages and lymphocytes. Here, we found increased levels of the active (phosphorylated) form of ACLY (p-ACLY) in the inflammatory infiltrate of AAA from patients and in aneurysmal lesions from angiotensin II (Ang II)-infused apolipoprotein E-deficient mice (ApoE-/-). Furthermore, plasma ACLY levels positively correlates with IL6 and IFNγ levels in patients with AAA, while inflammatory stimuli strongly upregulated ACLY expression in macrophages and Jurkat cells. The administration of the ACLY inhibitor bempedoic acid (BemA) protected against Ang II-induced AAA formation in ApoE-/- mice, limiting the progression of aortic dilatation and reducing mortality due to aortic rupture. BMS-303141, another ACLY inhibitor, also ameliorated AAA formation, although to a lesser extent. BemA attenuated vascular remodeling and the disorganization and rupture of elastic fibers induced by Ang II, as well as vascular inflammation, decreasing the recruitment of macrophages (CD68 +) and neutrophils (Ly-6G+) into the aortic wall. Moreover, BemA shifted splenic monocytes toward a functionally anti-inflammatory phenotype, and increased the percentage of CD4+CD69+ cells. Taken together, these results support the contribution of ACLY to AAA and point to BemA as a promising tool to be considered for future clinical trials addressing the management of this disease which is quite often associated with disorders of lipoprotein metabolism.
Collapse
Affiliation(s)
- Lídia Puertas-Umbert
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona 08036, Spain; Institut de Recerca Sant Pau (IR SANT PAU), Barcelona 08041, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Judith Alonso
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona 08036, Spain; Institut de Recerca Sant Pau (IR SANT PAU), Barcelona 08041, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Laia Blanco-Casoliva
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona 08036, Spain; Institut de Recerca Sant Pau (IR SANT PAU), Barcelona 08041, Spain
| | | | - Mercedes Camacho
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona 08041, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Antonio Rodríguez-Sinovas
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid 28029, Spain; Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Barcelona 08035, Spain
| | - María Galán
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid 28029, Spain; Facultad de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos, Alcorcón, Madrid 28922, Spain
| | - Nuria Roglans
- Dept. Farmacologia, Toxicologia i Química Terapèutica. Facultat de Farmàcia i Ciències de l'Alimentació, Institut de Biomedicina, Universitat de Barcelona, Barcelona 08028, Spain; CIBER de Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Juan Carlos Laguna
- Dept. Farmacologia, Toxicologia i Química Terapèutica. Facultat de Farmàcia i Ciències de l'Alimentació, Institut de Biomedicina, Universitat de Barcelona, Barcelona 08028, Spain; CIBER de Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - José Martínez-González
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona 08036, Spain; Institut de Recerca Sant Pau (IR SANT PAU), Barcelona 08041, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid 28029, Spain.
| | - Cristina Rodríguez
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona 08041, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid 28029, Spain.
| |
Collapse
|
4
|
Puertas-Umbert L, Alonso J, Roselló-Díez E, Santamaría-Orleans A, Martínez-González J, Rodríguez C. Rolipram impacts on redox homeostasis and cellular signaling in an experimental model of abdominal aortic aneurysm. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2024; 36:108-117. [PMID: 38061958 DOI: 10.1016/j.arteri.2023.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 05/08/2024]
Abstract
INTRODUCTION Cyclic nucleotide phosphodiesterases (PDEs) of the PDE4 subfamily are responsible for the hydrolysis and subcellular compartmentalization of cAMP, a second messenger that modulates vascular functionality. We had shown that PDE4B is induced in abdominal aortic aneurysms (AAA) and that PDE4 inhibition by rolipram limits experimental aneurysms. In this study we have delved into the mechanisms underlying the beneficial effect of rolipram on AAA. METHODS AAA were induced in ApoE-/- mice by angiotensin II (Ang II) infusion. Aneurysm formation was evaluated by ultrasonography. The expression of enzymes involved in rédox homeostasis was analyzed by real-time RT-PCR and the activation of signaling pathways by Western blot. RESULTS Induction of PDE4B in human AAA has been confirmed in a second cohort of patients. In Ang II-infused ApoE-/- mice, rolipram increased the percentage of animals free of aneurysms without affecting the percentage of aortic ruptures. Quantitative analyses determined that this drug significantly attenuated aortic collagen deposition. Additionally, rolipram reduced the increased Nox2 expression triggered by Ang II, exacerbated Sod1 induction, and normalized Sod3 expression. Likewise, PDE4 inhibition decreased the activation of both ERK1/2 and the canonical Wnt pathway, while AKT activity was not altered. CONCLUSIONS The inhibition of PDE4 activity modulates the expression of enzymes involved in rédox homeostasis and affects cell signaling pathways involved in the development of AAA.
Collapse
Affiliation(s)
- Lídia Puertas-Umbert
- Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, España; Institut de Recerca Hospital de la Santa Creu i Sant Pau (IRHSCSP), Barcelona, España; CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, España
| | - Judith Alonso
- Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, España; CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, España; Instituto de Investigaciones Biomédicas de Barcelona (IIBB-CSIC), Barcelona, España
| | - Elena Roselló-Díez
- Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, España; Departamento de Cirugía Cardíaca, Hospital de la Santa Creu i Sant Pau-Universitat Autònoma de Barcelona (HSCSP-UAB), Barcelona, España
| | - Alicia Santamaría-Orleans
- Laboratorios Ordesa S.L., Scientific Communication Department, Sant Boi del Llobregat, Barcelona, España
| | - José Martínez-González
- Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, España; CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, España; Instituto de Investigaciones Biomédicas de Barcelona (IIBB-CSIC), Barcelona, España
| | - Cristina Rodríguez
- Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, España; Institut de Recerca Hospital de la Santa Creu i Sant Pau (IRHSCSP), Barcelona, España; CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, España.
| |
Collapse
|
5
|
Chao C, Dang C, Reddy N, Alharbi S, Doan J, Karthikeyan A, Applewhite B, Jiang B. Characterization of a phenol-based model for denervation of the abdominal aorta and its implications for aortic remodeling. JVS Vasc Sci 2024; 5:100202. [PMID: 38694477 PMCID: PMC11061754 DOI: 10.1016/j.jvssci.2024.100202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 03/11/2024] [Indexed: 05/04/2024] Open
Abstract
Objective Sympathetic innervation plays a pivotal role in regulating cardiovascular health, and its dysregulation is implicated in a wide spectrum of cardiovascular diseases. This study seeks to evaluate the impact of denervation of the abdominal aorta on its morphology and wall homeostasis. Methods Male and female Sprague-Dawley rats (N = 12), aged 3 months, underwent midline laparotomy for infrarenal aorta exposure. Chemical denervation was induced via a one-time topical application of 10% phenol (n = 6), whereas sham controls received phosphate-buffered saline (n = 6). Animals were allowed to recover and subsequently were sacrificed after 6 months for analysis encompassing morphology, histology, and immunohistochemistry. Results At 6 months post-treatment, abdominal aortas subjected to phenol denervation still exhibited a significant reduction in nerve fiber density compared with sham controls. Denervated aortas demonstrated reduced intima-media thickness, increased elastin fragmentation, decreased expression of vascular smooth muscle proteins (α-SMA and MYH11), and elevated adventitial vascular density. Sex-stratified analyses revealed additional dimorphic responses, particularly in aortic collagen and medial cellular density in female animals. Conclusions Single-timepoint phenol-based chemical denervation induces alterations in abdominal aortic morphology and vascular remodeling over a 6-month period. These findings underscore the potential of the sympathetic nervous system as a therapeutic target for aortic pathologies. Clinical Relevance Aortic remodeling remains an important consideration in the pathogenesis of aortic disease, including occlusive, aneurysmal, and dissection disease states. The paucity of medical therapies for the treatment of aortic disease has driven considerable interest in elucidating the pathogenesis of these conditions; new therapeutic targets are critically needed. Here, we show significant remodeling after phenol-induced denervation with morphologic, histologic, and immunohistochemical features. Future investigations should integrate sympathetic dysfunction as a potential driver of pathologic aortic wall changes with additional consideration of the sympathetic nervous system as a therapeutic target.
Collapse
Affiliation(s)
- Calvin Chao
- Division of Vascular Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Caitlyn Dang
- Division of Vascular Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Nidhi Reddy
- Division of Vascular Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Sara Alharbi
- Division of Vascular Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Jimmy Doan
- Department of Biomedical Engineering, Northwestern University McCormick School of Engineering, Evanston, IL
| | - Akashraj Karthikeyan
- Department of Biomedical Engineering, Northwestern University McCormick School of Engineering, Evanston, IL
| | - Brandon Applewhite
- Department of Biomedical Engineering, Northwestern University McCormick School of Engineering, Evanston, IL
| | - Bin Jiang
- Division of Vascular Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL
- Department of Biomedical Engineering, Northwestern University McCormick School of Engineering, Evanston, IL
| |
Collapse
|
6
|
Ballester-Servera C, Cañes L, Alonso J, Puertas-Umbert L, Vázquez-Sufuentes P, Taurón M, Roselló-Díez E, Marín F, Rodríguez C, Martínez-González J. Upregulation of NOR-1 in calcified human vascular tissues: impact on osteogenic differentiation and calcification. Transl Res 2024; 264:1-14. [PMID: 37690706 DOI: 10.1016/j.trsl.2023.09.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 09/04/2023] [Accepted: 09/05/2023] [Indexed: 09/12/2023]
Abstract
Cardiovascular calcification is a significant public health issue whose pathophysiology is not fully understood. NOR-1 regulates critical processes in cardiovascular remodeling, but its contribution to ectopic calcification is unknown. NOR-1 was overexpressed in human calcific aortic valves and calcified atherosclerotic lesions colocalizing with RUNX2, a factor essential for osteochondrogenic differentiation and calcification. NOR-1 and osteogenic markers were upregulated in calcifying human valvular interstitial cells (VICs) and human vascular smooth muscle cells (VSMCs). Gain- and loss-of-function approaches demonstrated that NOR-1 negatively modulates the expression of osteogenic genes relevant for the osteogenic transdifferentiation (RUNX2, IL-6, BMP2, and ALPL) and calcification of VICs. VSMCs from transgenic mice overexpressing NOR-1 in these cells (TgNOR-1VSMC) expressed lower basal levels of osteogenic genes (IL-6, BMP2, ALPL, OPN) than cells from WT littermates, and their upregulation by a high-phosphate osteogenic medium (OM) was completely prevented by NOR-1 transgenesis. Consistently, this was associated with a dramatic reduction in the calcification of both transgenic VSMCs and aortic rings from TgNOR-1VSMC mice exposed to OM. Atherosclerosis and calcification were induce in mice by the administration of AAV-PCSK9D374Y and a high-fat/high-cholesterol diet. Challenged-TgNOR-1VSMC mice exhibited decreased vascular expression of osteogenic markers, and both less atherosclerotic burden (assessed in whole aorta and lesion size in aortic arch and brachiocephalic artery) and less vascular calcification (assessed either by near-infrared fluorescence imaging or histological analysis) than WT mice. Our data indicate that NOR-1 negatively modulates the expression of genes critically involved in the osteogenic differentiation of VICs and VSMCs, thereby restraining ectopic cardiovascular calcification.
Collapse
Affiliation(s)
- Carme Ballester-Servera
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona, Spain; CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Laia Cañes
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona, Spain; CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Judith Alonso
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona, Spain; CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Lidia Puertas-Umbert
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain; Institut de Recerca Hospital de la Santa Creu i Sant Pau (IRHSCSP), Barcelona, Spain
| | - Paula Vázquez-Sufuentes
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Manel Taurón
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain; Departamento de Cirugía Cardíaca, Hospital de la Santa Creu i Sant Pau-Universitat Autònoma de Barcelona (HSCSP-UAB), Barcelona, Spain
| | - Elena Roselló-Díez
- Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain; Departamento de Cirugía Cardíaca, Hospital de la Santa Creu i Sant Pau-Universitat Autònoma de Barcelona (HSCSP-UAB), Barcelona, Spain
| | - Francisco Marín
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Departamento de Cardiología, Hospital Clínico Universitario Virgen de la Arrixaca-Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), Murcia, Spain
| | - Cristina Rodríguez
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain; Institut de Recerca Hospital de la Santa Creu i Sant Pau (IRHSCSP), Barcelona, Spain
| | - José Martínez-González
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona, Spain; CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain.
| |
Collapse
|
7
|
Puertas-Umbert L, Almendra-Pegueros R, Jiménez-Altayó F, Sirvent M, Galán M, Martínez-González J, Rodríguez C. Novel pharmacological approaches in abdominal aortic aneurysm. Clin Sci (Lond) 2023; 137:1167-1194. [PMID: 37559446 PMCID: PMC10415166 DOI: 10.1042/cs20220795] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/05/2023] [Accepted: 07/28/2023] [Indexed: 08/11/2023]
Abstract
Abdominal aortic aneurysm (AAA) is a severe vascular disease and a major public health issue with an unmet medical need for therapy. This disease is featured by a progressive dilation of the abdominal aorta, boosted by atherosclerosis, ageing, and smoking as major risk factors. Aneurysm growth increases the risk of aortic rupture, a life-threatening emergency with high mortality rates. Despite the increasing progress in our knowledge about the etiopathology of AAA, an effective pharmacological treatment against this disorder remains elusive and surgical repair is still the unique available therapeutic approach for high-risk patients. Meanwhile, there is no medical alternative for patients with small aneurysms but close surveillance. Clinical trials assessing the efficacy of antihypertensive agents, statins, doxycycline, or anti-platelet drugs, among others, failed to demonstrate a clear benefit limiting AAA growth, while data from ongoing clinical trials addressing the benefit of metformin on aneurysm progression are eagerly awaited. Recent preclinical studies have postulated new therapeutic targets and pharmacological strategies paving the way for the implementation of future clinical studies exploring these novel therapeutic strategies. This review summarises some of the most relevant clinical and preclinical studies in search of new therapeutic approaches for AAA.
Collapse
Affiliation(s)
- Lídia Puertas-Umbert
- Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
- CIBER de Enfermedades Cardiovasculares, ISCIII, Madrid, Spain
| | | | - Francesc Jiménez-Altayó
- Department of Pharmacology, Therapeutics and Toxicology, School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
- Neuroscience Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Marc Sirvent
- CIBER de Enfermedades Cardiovasculares, ISCIII, Madrid, Spain
- Departamento de Angiología y Cirugía Vascular del Hospital Universitari General de Granollers, Granollers, Barcelona, Spain
| | - María Galán
- Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
- CIBER de Enfermedades Cardiovasculares, ISCIII, Madrid, Spain
- Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos, Alcorcón, Spain
| | - José Martínez-González
- Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
- CIBER de Enfermedades Cardiovasculares, ISCIII, Madrid, Spain
- Instituto de Investigaciones Biomédicas de Barcelona (IIBB-CSIC), Barcelona, Spain
| | - Cristina Rodríguez
- Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
- CIBER de Enfermedades Cardiovasculares, ISCIII, Madrid, Spain
| |
Collapse
|
8
|
Zhu H, Tan J, Wang Z, Wu Z, Zhou W, Zhang Z, Li M, Zhao Y. Bioinformatics analysis constructs potential ferroptosis-related ceRNA network involved in the formation of intracranial aneurysm. Front Cell Neurosci 2022; 16:1016682. [PMCID: PMC9612944 DOI: 10.3389/fncel.2022.1016682] [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: 08/11/2022] [Accepted: 09/29/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundIntracranial aneurysm (IA) causes more than 80% of nontraumatic subarachnoid hemorrhages (SAHs). The mechanism of ferroptosis involved in IA formation remains unclear. The roles played by competitive endogenous RNA (ceRNA) regulation networks in many diseases are becoming clearer. The goal of this study was to understand more fully the ferroptosis-related ceRNA regulation network in IA.Materials and methodsTo identify differentially expressed genes (DEGs), differentially expressed miRNAs (DEMs), and differentially expressed lncRNAs (DELs) across IA and control samples, the GEO datasets GSE122897 and GSE66239 were downloaded and analyzed with the aid of R. Ferroptosis DEGs were discovered by exploring the DEGs of ferroptosis-related genes of the ferroptosis database. Potentially interacting miRNAs and lncRNAs were predicted using miRWalk and StarBase. Enrichment analysis was also performed. We utilized the STRING database and Cytoscape software to identify protein-protein interactions and networks. DAB-enhanced Prussian blue staining was used to detect iron in IA tissues.ResultsIron deposition was evident in IA tissue. In all, 30 ferroptosis DEGs, 5 key DEMs, and 17 key DELs were screened out for constructing a triple regulatory network. According to expression regulation of DELs, DEMs, and DEGs, a hub triple regulatory network was built. As the functions of lncRNAs are determined by their cellular location, PVT1-hsa-miR-4644-SLC39A14 ceRNA and DUXAP8-hsa-miR-378e/378f-SLC2A3 ceRNA networks were constructed.ConclusionCeRNA (PVT1-hsa-miR-4644-SLC39A14 and DUXAP8-hsa-miR-378e/378f-SLC2A3) overexpression networks associated with ferroptosis in IA were established.
Collapse
|
9
|
Jiménez-Fernández M, Rodríguez-Sinovas C, Cañes L, Ballester-Servera C, Vara A, Requena S, de la Fuente H, Martínez-González J, Sánchez-Madrid F. CD69-oxLDL ligand engagement induces Programmed Cell Death 1 (PD-1) expression in human CD4 + T lymphocytes. Cell Mol Life Sci 2022; 79:468. [PMID: 35930205 PMCID: PMC9355928 DOI: 10.1007/s00018-022-04481-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 06/28/2022] [Accepted: 07/12/2022] [Indexed: 11/24/2022]
Abstract
The mechanisms that control the inflammatory–immune response play a key role in tissue remodelling in cardiovascular diseases. T cell activation receptor CD69 binds to oxidized low-density lipoprotein (oxLDL), inducing the expression of anti-inflammatory NR4A nuclear receptors and modulating inflammation in atherosclerosis. To understand the downstream T cell responses triggered by the CD69-oxLDL binding, we incubated CD69-expressing Jurkat T cells with oxLDL. RNA sequencing revealed a differential gene expression profile dependent on the presence of CD69 and the degree of LDL oxidation. CD69-oxLDL binding induced the expression of NR4A receptors (NR4A1 and NR4A3), but also of PD-1. These results were confirmed using oxLDL and a monoclonal antibody against CD69 in CD69-expressing Jurkat and primary CD4 + lymphocytes. CD69-mediated induction of PD-1 and NR4A3 was dependent on NFAT activation. Silencing NR4A3 slightly increased PD-1 levels, suggesting a potential regulation of PD-1 by this receptor. Moreover, expression of PD-1, CD69 and NR4A3 was increased in human arteries with chronic inflammation compared to healthy controls, with a strong correlation between PD-1 and CD69 mRNA expression (r = 0.655 P < 0.0001). Moreover, PD-1 was expressed in areas enriched in CD3 infiltrating T cells. Our results underscore a novel mechanism of PD-1 induction independent of TCR signalling that might contribute to the role of CD69 in the modulation of inflammation and vascular remodelling in cardiovascular diseases.
Collapse
Affiliation(s)
- María Jiménez-Fernández
- Hospital Universitario de la Princesa, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa (IIS-IP), c/ Diego de León, 62, 28006, Madrid, Spain.,Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Cristina Rodríguez-Sinovas
- Institut de Recerca Hospital de la Santa Creu i Sant Pau (IRHSCSP), IIB-Sant Pau, Barcelona, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Laia Cañes
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.,Instituto de Investigaciones Biomédicas de Barcelona - Consejo Superior de Investigaciones Científicas (IIBB-CSIC), IIB-Sant Pau, C/ Rosselló, 161, 08036, Barcelona, Spain
| | - Carme Ballester-Servera
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.,Instituto de Investigaciones Biomédicas de Barcelona - Consejo Superior de Investigaciones Científicas (IIBB-CSIC), IIB-Sant Pau, C/ Rosselló, 161, 08036, Barcelona, Spain
| | - Alicia Vara
- Hospital Universitario de la Princesa, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa (IIS-IP), c/ Diego de León, 62, 28006, Madrid, Spain
| | - Silvia Requena
- Hospital Universitario de la Princesa, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa (IIS-IP), c/ Diego de León, 62, 28006, Madrid, Spain
| | - Hortensia de la Fuente
- Hospital Universitario de la Princesa, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa (IIS-IP), c/ Diego de León, 62, 28006, Madrid, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - José Martínez-González
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain. .,Instituto de Investigaciones Biomédicas de Barcelona - Consejo Superior de Investigaciones Científicas (IIBB-CSIC), IIB-Sant Pau, C/ Rosselló, 161, 08036, Barcelona, Spain.
| | - Francisco Sánchez-Madrid
- Hospital Universitario de la Princesa, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa (IIS-IP), c/ Diego de León, 62, 28006, Madrid, Spain. .,Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain. .,CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.
| |
Collapse
|
10
|
Ballester-Servera C, Cañes L, Alonso J, Puertas L, Taurón M, Rodríguez C, Martínez-González J. Nuclear receptor NOR-1 (Neuron-derived Orphan Receptor-1) in pathological vascular remodelling and vascular remodelling. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2022; 34:229-243. [PMID: 35581107 DOI: 10.1016/j.arteri.2022.03.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/28/2022] [Accepted: 03/01/2022] [Indexed: 06/15/2023]
Abstract
Vascular cells and their interaction with inflammatory cells and the immune system play a key role in pathological vascular remodeling. A large number of genes and proteins regulated in a coordinated manner by a small number of transcription factors are involved in this process. In recent years, research on a small subfamily of transcription factors, the NR4A subfamily, has had a major impact on our understanding of vascular biology. The NR4A1 (Nur77), NR4A2 (Nurr1) and NR4A3 (NOR-1) receptors are products of early response genes whose expression is induced by multiple pathophysiological and physical stimuli. Their wide distribution in different tissues and cells places them in the control of numerous processes such as cell differentiation, proliferation, survival and apoptosis, as well as inflammation and the metabolism of lipids and carbohydrates. This review analyzes the role of these receptors, particularly NOR-1, in pathological vascular remodeling associated with atherosclerosis, abdominal aortic aneurysm and pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Carme Ballester-Servera
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona, España; CIBER de Enfermedades Cardiovasculares, ISCIII, Madrid, España; Instituto de Investigación Biomédica Sant Pau, Barcelona, España
| | - Laia Cañes
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona, España; CIBER de Enfermedades Cardiovasculares, ISCIII, Madrid, España
| | - Judith Alonso
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona, España; CIBER de Enfermedades Cardiovasculares, ISCIII, Madrid, España; Instituto de Investigación Biomédica Sant Pau, Barcelona, España
| | - Lidia Puertas
- Instituto de Investigación Biomédica Sant Pau, Barcelona, España; Institut de Recerca Hospital de la Santa Creu i Sant Pau (IRHSCSP), Barcelona, España
| | - Manel Taurón
- Servicio de Cirugía Cardiovascular, Hospital de la Santa Creu i Sant Pau, Barcelona, España
| | - Cristina Rodríguez
- CIBER de Enfermedades Cardiovasculares, ISCIII, Madrid, España; Instituto de Investigación Biomédica Sant Pau, Barcelona, España; Institut de Recerca Hospital de la Santa Creu i Sant Pau (IRHSCSP), Barcelona, España
| | - José Martínez-González
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona, España; CIBER de Enfermedades Cardiovasculares, ISCIII, Madrid, España; Instituto de Investigación Biomédica Sant Pau, Barcelona, España.
| |
Collapse
|
11
|
Lv J, Li X, Wu H, Li J, Luan B, Li Y, Li Y, Yang D, Wen H. Icariside II Restores Vascular Smooth Muscle Cell Contractile Phenotype by Enhancing the Focal Adhesion Signaling Pathway in the Rat Vascular Remodeling Model. Front Pharmacol 2022; 13:897615. [PMID: 35770073 PMCID: PMC9234455 DOI: 10.3389/fphar.2022.897615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 05/03/2022] [Indexed: 11/13/2022] Open
Abstract
Vascular smooth muscle cell (VSMC) phenotypic transition represents the fundamental pathophysiological alteration in the vascular remodeling process during the initiation and progression of cardiovascular diseases. Recent studies have revealed that Icariside II (ICS-II), a flavonol glycoside derived from the traditional Chinese medicine Herba Epimedii, exhibited therapeutic effects in various cardiovascular diseases. However, the therapeutic efficacy and underlying mechanisms of ICS-II regarding VSMC phenotypic transition were unknown. In this study, we investigated the therapeutic effects of ICS-Ⅱ on vascular remodeling with a rat’s balloon injury model in vivo. The label-free proteomic analysis was further implemented to identify the differentially expressed proteins (DEPs) after ICS-II intervention. Gene ontology and the pathway enrichment analysis were performed based on DEPs. Moreover, platelet-derived growth factor (PDGF-BB)-induced primary rat VSMC was implemented to verify the restoration effects of ICS-II on the VSMC contractile phenotype. Results showed that ICS-II could effectively attenuate the vascular remodeling process, promote SMA-α protein expression, and inhibit OPN expression in vivo. The proteomic analysis identified 145 differentially expressed proteins after ICS-II intervention. Further, the bioinformatics analysis indicated that the focal adhesion signaling pathway was enriched in the ICS-II group. In vitro studies showed that ICS-II suppressed VSMC proliferation and migration, and promoted VSMC contractile phenotype by modulating the focal adhesion signaling pathway. Taken together, our results suggest that ICS-II attenuates the vascular remodeling process and restores the VSMC contractile phenotype by promoting the focal adhesion pathway.
Collapse
Affiliation(s)
- Junyuan Lv
- Breast and Thyroid Surgery, Department of General Surgery, The Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Xintong Li
- Department of Vascular Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Hongyu Wu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Jiayang Li
- Drug Clinical Trial Institution, The Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Boyang Luan
- Department of Trauma Center, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yiqi Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Yeli Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Danli Yang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Hao Wen
- Department of Trauma Center, The First Affiliated Hospital of China Medical University, Shenyang, China
- *Correspondence: Hao Wen,
| |
Collapse
|
12
|
Exploring the Effect and Mechanism of Si-Miao-Yong-An Decoction on Abdominal Aortic Aneurysm Based on Mice Experiment and Bioinformatics Analysis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:4766987. [PMID: 35685724 PMCID: PMC9173986 DOI: 10.1155/2022/4766987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 04/26/2022] [Accepted: 05/19/2022] [Indexed: 12/02/2022]
Abstract
Background Abdominal aortic aneurysm (AAA) is a fatal disease characterized by high morbidity and mortality in old population. Globally, effective drugs for AAA are still limited. Si-Miao-Yong-An decoction (SMYAD), a traditional Chinese medicine (TCM) formula with a high medical value, was reported to be successfully used in an old AAA patient. Thus, we reason that SMYAD may serve as a potential anti-AAA regime. Objective The exact effects and detailed mechanisms of SMYAD on AAA were explored by using the experimental study and bioinformatics analysis. Methods Firstly, C57BL/6N mice induced by Bap and Ang II were utilized to reproduce the AAA model, and the effects of SMYAD were systematically assessed according to histology, immunohistochemistry, and enzyme-linked immunosorbent assay (ELISA). Then, network pharmacology was applied to identify the biological processes, pathways, and hub targets of SMYAD against AAA; moreover, molecular docking was utilized to identify the binding ability and action targets. Results In an animal experiment, SMYAD was found to effectively alleviate the degree of pathological expansion of abdominal aorta and reduce the incidence of Bap/Ang II-induced AAA, along with reducing the damage to elastic lamella, attenuating infiltration of macrophage, and lowering the circulating IL-6 level corresponding to the animal study, and network pharmacology revealed the detailed mechanisms of SMYAD on AAA that were related to pathways of inflammatory response, defense response, apoptotic, cell migration and adhesion, and reactive oxygen species metabolic process. Then, seven targets, IL-6, TNF, HSP90AA1, RELA, PTGS2, ESR1, and MMP9, were identified as hub targets of SMYAD against AAA. Furthermore, molecular docking verification revealed that the active compounds of SMYAD had good binding ability and clear binding site with core targets related to AAA formation. Conclusion SMYAD can suppress AAA development through multicompound, multitarget, and multipathway, which provides a research direction for further study.
Collapse
|
13
|
Paapstel K, Kals J. Metabolomics of Arterial Stiffness. Metabolites 2022; 12:370. [PMID: 35629874 PMCID: PMC9146333 DOI: 10.3390/metabo12050370] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/15/2022] [Accepted: 04/18/2022] [Indexed: 12/18/2022] Open
Abstract
Arterial stiffness (AS) is one of the earliest detectable signs of structural and functional alterations of the vessel wall and an independent predictor of cardiovascular events and death. The emerging field of metabolomics can be utilized to detect a wide spectrum of intermediates and products of metabolism in body fluids that can be involved in the pathogenesis of AS. Research over the past decade has reinforced this idea by linking AS to circulating acylcarnitines, glycerophospholipids, sphingolipids, and amino acids, among other metabolite species. Some of these metabolites influence AS through traditional cardiovascular risk factors (e.g., high blood pressure, high blood cholesterol, diabetes, smoking), while others seem to act independently through both known and unknown pathophysiological mechanisms. We propose the term 'arteriometabolomics' to indicate the research that applies metabolomics methods to study AS. The 'arteriometabolomics' approach has the potential to allow more personalized cardiovascular risk stratification, disease monitoring, and treatment selection. One of its major goals is to uncover the causal metabolic pathways of AS. Such pathways could represent valuable treatment targets in vascular ageing.
Collapse
Affiliation(s)
- Kaido Paapstel
- Endothelial Research Centre, University of Tartu, 8 Puusepa Street, 51014 Tartu, Estonia;
- Department of Cardiology, Institute of Clinical Medicine, University of Tartu, 8 Puusepa Street, 51014 Tartu, Estonia
- Heart Clinic, Tartu University Hospital, 8 Puusepa Street, 51014 Tartu, Estonia
| | - Jaak Kals
- Endothelial Research Centre, University of Tartu, 8 Puusepa Street, 51014 Tartu, Estonia;
- Department of Surgery, Institute of Clinical Medicine, University of Tartu, 8 Puusepa Street, 51014 Tartu, Estonia
- Surgery Clinic, Tartu University Hospital, 8 Puusepa Street, 51014 Tartu, Estonia
- Department of Biochemistry, Institute of Biomedicine and Translational Medicine, Centre of Excellence for Genomics and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia
| |
Collapse
|
14
|
Martínez-González J, Cañes L, Alonso J, Ballester-Servera C, Rodríguez-Sinovas A, Corrales I, Rodríguez C. NR4A3: A Key Nuclear Receptor in Vascular Biology, Cardiovascular Remodeling, and Beyond. Int J Mol Sci 2021; 22:ijms222111371. [PMID: 34768801 PMCID: PMC8583700 DOI: 10.3390/ijms222111371] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 12/14/2022] Open
Abstract
The mechanisms committed in the activation and response of vascular and inflammatory immune cells play a major role in tissue remodeling in cardiovascular diseases (CVDs) such as atherosclerosis, pulmonary arterial hypertension, and abdominal aortic aneurysm. Cardiovascular remodeling entails interrelated cellular processes (proliferation, survival/apoptosis, inflammation, extracellular matrix (ECM) synthesis/degradation, redox homeostasis, etc.) coordinately regulated by a reduced number of transcription factors. Nuclear receptors of the subfamily 4 group A (NR4A) have recently emerged as key master genes in multiple cellular processes and vital functions of different organs, and have been involved in a variety of high-incidence human pathologies including atherosclerosis and other CVDs. This paper reviews the major findings involving NR4A3 (Neuron-derived Orphan Receptor 1, NOR-1) in the cardiovascular remodeling operating in these diseases.
Collapse
Affiliation(s)
- José Martínez-González
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), 08036 Barcelona, Spain; (L.C.); (J.A.); (C.B.-S.)
- CIBER de Enfermedades Cardiovasculares, ISCIII, 28029 Madrid, Spain;
- Instituto de Investigación Biomédica Sant Pau, 08041 Barcelona, Spain
- Correspondence: (J.M.-G.); (C.R.); Tel.: +34-93-5565896 (J.M.-G.); +34-93-5565897 (C.R.)
| | - Laia Cañes
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), 08036 Barcelona, Spain; (L.C.); (J.A.); (C.B.-S.)
- CIBER de Enfermedades Cardiovasculares, ISCIII, 28029 Madrid, Spain;
- Instituto de Investigación Biomédica Sant Pau, 08041 Barcelona, Spain
| | - Judith Alonso
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), 08036 Barcelona, Spain; (L.C.); (J.A.); (C.B.-S.)
- CIBER de Enfermedades Cardiovasculares, ISCIII, 28029 Madrid, Spain;
- Instituto de Investigación Biomédica Sant Pau, 08041 Barcelona, Spain
| | - Carme Ballester-Servera
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), 08036 Barcelona, Spain; (L.C.); (J.A.); (C.B.-S.)
- CIBER de Enfermedades Cardiovasculares, ISCIII, 28029 Madrid, Spain;
- Instituto de Investigación Biomédica Sant Pau, 08041 Barcelona, Spain
| | - Antonio Rodríguez-Sinovas
- CIBER de Enfermedades Cardiovasculares, ISCIII, 28029 Madrid, Spain;
- Cardiovascular Diseases Research Group, Vall d’Hebron Institut de Recerca, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
| | - Irene Corrales
- Laboratorio de Coagulopatías Congénitas, Banc de Sang i Teixits (BST), 08005 Barcelona, Spain;
- Medicina Transfusional, Vall d’Hebron Institut de Recerca-Universitat Autònoma de Barcelona (VHIR-UAB), 08035 Barcelona, Spain
| | - Cristina Rodríguez
- CIBER de Enfermedades Cardiovasculares, ISCIII, 28029 Madrid, Spain;
- Instituto de Investigación Biomédica Sant Pau, 08041 Barcelona, Spain
- Institut de Recerca Hospital de la Santa Creu i Sant Pau (IRHSCSP), 08041 Barcelona, Spain
- Correspondence: (J.M.-G.); (C.R.); Tel.: +34-93-5565896 (J.M.-G.); +34-93-5565897 (C.R.)
| |
Collapse
|