1
|
Zhang B, Liu Z, Chang Y, Lv R, Guo H, Qiang P, Shimosawa T, Xu Q, Yang F. Aldosterone-Induced Transformation of Vascular Smooth Muscle Cells into Macrophage-like Cells Participates in Inflammatory Vascular Lesions. Int J Mol Sci 2025; 26:3345. [PMID: 40244230 PMCID: PMC11989480 DOI: 10.3390/ijms26073345] [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: 01/13/2025] [Revised: 03/23/2025] [Accepted: 03/31/2025] [Indexed: 04/18/2025] Open
Abstract
Vascular smooth muscle cells (VSMCs) are the most abundant cell type in blood vessels, participating in cardiovascular diseases in various ways, among which their transformation into macrophage-like cells has become a research hotspot. In this study, rats were infused with aldosterone for 12 weeks, and VSMCs stimulated with aldosterone in vitro were used to observe aortic injury and the role of VSMC transformation. Vascular changes were detected via small animal ultrasound and H&E staining. Moreover, immunohistochemistry, immunofluorescence, Western blot, and flow cytometry were used to verify that the transformation of VSMCs into macrophage-like cells is regulated by mineralocorticoid receptor (MR) activation and macrophage colony-stimulating factor (M-CSF) and its receptor. Rat vasculature and in vitro cellular experiments revealed that VSMCs transformed into macrophage-like cells and secreted inflammatory factors such as interleukin-1β (IL-1β) and monocyte chemoattractant protein-1 (MCP-1), thereby exacerbating inflammatory vascular lesions, which was inhibited by the MR antagonist esaxerenone. These results reveal that increased levels of aldosterone activate MR, leading to the secretion of M-CSF by VSMCs. This further promotes the transformation of VSMCs into macrophage-like cells, which participate in inflammatory vascular lesions. Therefore, inhibiting the formation of macrophage-like cells can effectively reduce inflammatory vascular lesions.
Collapse
MESH Headings
- Animals
- Aldosterone/pharmacology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/cytology
- Macrophages/metabolism
- Macrophages/drug effects
- Macrophages/pathology
- Rats
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/pathology
- Receptors, Mineralocorticoid/metabolism
- Male
- Inflammation/metabolism
- Inflammation/pathology
- Rats, Sprague-Dawley
- Cells, Cultured
- Macrophage Colony-Stimulating Factor/metabolism
- Interleukin-1beta/metabolism
- Chemokine CCL2/metabolism
Collapse
Affiliation(s)
- Boya Zhang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Ziqian Liu
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Yi Chang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Ruyan Lv
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Haixia Guo
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Panpan Qiang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Tatsuo Shimosawa
- Department of Clinical Laboratory, School of Medicine, International University of Health and Welfare, Narita 286-8686, Japan
| | - Qingyou Xu
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Fan Yang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| |
Collapse
|
2
|
Mahmoud MM, Hassan MM, Elsayed HES, Fares AE, Saber MM, Rashed LA, Abdelwahed OM. Protective effect of Galectin-3 inhibitor against cardiac remodelling in an isoprenaline-induced myocardial infarction in type 2 diabetes. Arch Physiol Biochem 2025; 131:94-107. [PMID: 39101980 DOI: 10.1080/13813455.2024.2387710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 06/15/2024] [Accepted: 07/28/2024] [Indexed: 08/06/2024]
Abstract
Type 2 Diabetes mellitus (T2DM) has the potential to impair cardiac function and cause heart failure. We aimed to study the cardioprotective influence of Galactin-3 (Gal-3) inhibitor; modified citrus pectin (MCP) in isoprenaline induced myocardial infarction (MI) in T2DM rats. Forty rats were allocated into 4 groups; groups I and II served as control. T2DM was provoked in groups III and IV by serving them high fat diet followed by a single low dose of Streptozotocin (STZ), then group IV were administered MCP in drinking water for 6 weeks. Groups III and IV were then subcutaneously injected isoprenaline hydrochloride once daily on the last 2 successive days to induce MI. MCP restored echocardiographic parameters with significant decline in Gal-3 area % in cardiac tissue alongside protection against cardiac remodelling. our data showed that there is a protective potential for Gal-3 inhibitor (MCP) against cardiac injury in isoprenaline induced MI in T2DM.
Collapse
Affiliation(s)
| | - Mai Mohammed Hassan
- Department of Medical Physiology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | | | - Amal E Fares
- Department of Histology & Cell Biology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mona M Saber
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Laila Ahmed Rashed
- Department of Medical Biochemistry, Faculty of Medicine, Cairo University, Cairo, Egypt
| | | |
Collapse
|
3
|
Ismail R, Habib HA, Anter AF, Amin A, Heeba GH. Modified citrus pectin ameliorates methotrexate-induced hepatic and pulmonary toxicity: role of Nrf2, galectin-3/TLR-4/NF-κB/TNF-α and TGF-β signaling pathways. Front Pharmacol 2025; 16:1528978. [PMID: 39917614 PMCID: PMC11798997 DOI: 10.3389/fphar.2025.1528978] [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: 11/15/2024] [Accepted: 01/06/2025] [Indexed: 02/09/2025] Open
Abstract
Introduction Methotrexate (MTX) is a frequently utilized anti-inflammatory and anticancer agent. Its potential liver and lung toxicity often limits its clinical effectiveness. We conducted this study to demonstrate the possible protective impacts of a natural galectin-3 (Gal-3) inhibitor, modified citrus pectin (MCP), against MTX-induced liver and lung toxicity and verify the potential signaling pathways of these suggested effects. In vitro, the cytotoxicity of MCP and its modulatory effect on MTX cytotoxic efficacy were assessed. Methods Four groups of rats were used: control, MTX (40 mg/kg, single intraperitoneal injection on day 9), MTX + MCP (200 mg/kg/day, orally, for 2 weeks), and MCP alone. MCF7, Nalm6, and JEG3 cell lines were used for the in vitro cytotoxicity assay. Results MCP counteracted liver and lung toxicity evidenced by ameliorating the markers of liver and lung functions. Moreover, MCP minimized oxidative stress elicited by MTX in lung and liver tissues, as indicated by reduced malondialdehyde levels, elevated levels of reduced glutathione, increased superoxide dismutase activity, and upregulated Nrf2 protein expression. In hepatic and pulmonary tissues, MCP downregulated the inflammatory signaling pathway, Gal-3/TLR-4/NF-κB/TNF-α. MCP pretreatment decreased TGF-β, collagen content, and cleaved caspase-3 levels. MCP enhanced the cytotoxicity of MTX in Nalm6 and JEG3 and did not interfere with its cytotoxicity in the MCF7 cell lines. Discussion MCP attenuated MTX-induced liver and lung toxicity through antioxidant, anti-fibrotic, anti-inflammatory, and anti-apoptotic influences, as demonstrated by the improved histopathological changes induced by MTX in pulmonary and hepatic tissues. Moreover, it increased MTX cytotoxicity in different human cell lines.
Collapse
Affiliation(s)
- Randa Ismail
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, El-Minia, Egypt
| | - Heba A. Habib
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, El-Minia, Egypt
| | - Aliaa F. Anter
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, El-Minia, Egypt
| | - Amr Amin
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Gehan H. Heeba
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, El-Minia, Egypt
| |
Collapse
|
4
|
Abubakar M, Irfan U, Abdelkhalek A, Javed I, Khokhar MI, Shakil F, Raza S, Salim SS, Altaf MM, Habib R, Ahmed S, Ahmed F. Comprehensive Quality Analysis of Conventional and Novel Biomarkers in Diagnosing and Predicting Prognosis of Coronary Artery Disease, Acute Coronary Syndrome, and Heart Failure, a Comprehensive Literature Review. J Cardiovasc Transl Res 2024; 17:1258-1285. [PMID: 38995611 DOI: 10.1007/s12265-024-10540-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 06/25/2024] [Indexed: 07/13/2024]
Abstract
Coronary artery disease (CAD), acute coronary syndrome (ACS), and heart failure (HF) are major global health issues with high morbidity and mortality rates. Biomarkers like cardiac troponins (cTn) and natriuretic peptides (NPs) are crucial tools in cardiology, but numerous new biomarkers have emerged, proving increasingly valuable in CAD/ACS. These biomarkers are classified based on their mechanisms, such as fibrosis, metabolism, inflammation, and congestion. The integration of established and emerging biomarkers into clinical practice is an ongoing process, and recognizing their strengths and limitations is crucial for their accurate interpretation, incorporation into clinical settings, and improved management of CVD patients. We explored established biomarkers like cTn, NPs, and CRP, alongside newer biomarkers such as Apo-A1, IL-17E, IgA, Gal-3, sST2, GDF-15, MPO, H-FABP, Lp-PLA2, and ncRNAs; provided evidence of their utility in CAD/ACS diagnosis and prognosis; and empowered clinicians to confidently integrate these biomarkers into clinical practice based on solid evidence.
Collapse
Affiliation(s)
- Muhammad Abubakar
- Department of Internal Medicine, Ameer-Ud-Din Medical College, 6 Birdwood Road, Jinnah Town, Lahore, 54000, Punjab, Pakistan.
| | - Umema Irfan
- Department of Internal Medicine, Deccan College of Medical Sciences, Hyderabad, India
| | - Ahmad Abdelkhalek
- Department of Internal Medicine, Zhejiang University, Zhejiang, China
| | - Izzah Javed
- Department of Internal Medicine, Ameer-Ud-Din Medical College, 6 Birdwood Road, Jinnah Town, Lahore, 54000, Punjab, Pakistan
| | | | - Fraz Shakil
- Department of Emergency Medicine, Mayo Hospital, Lahore, Pakistan
| | - Saud Raza
- Department of Anesthesia, Social Security Teaching Hospital, Lahore, Punjab, Pakistan
| | - Siffat Saima Salim
- Department of Surgery, Holy Family Red Crescent Medical College Hospital, Dhaka, Bangladesh
| | - Muhammad Mahran Altaf
- Department of Internal Medicine, Ameer-Ud-Din Medical College, 6 Birdwood Road, Jinnah Town, Lahore, 54000, Punjab, Pakistan
| | - Rizwan Habib
- Department of Internal Medicine and Emergency, Indus Hospital, Lahore, Pakistan
| | - Simra Ahmed
- Department of Internal Medicine, Ziauddin Medical College, Karachi, Pakistan
| | - Farea Ahmed
- Department of Internal Medicine, Ziauddin Medical College, Karachi, Pakistan
| |
Collapse
|
5
|
García-Martín A, Prados ME, Lastres-Cubillo I, Ponce-Diaz FJ, Cerero L, Garrido-Rodríguez M, Navarrete C, Pineda R, Rodríguez AB, Muñoz I, Moya J, Medeot A, Moreno JA, Chacón A, García-Revillo J, Muñoz E. Etrinabdione (VCE-004.8), a B55α activator, promotes angiogenesis and arteriogenesis in critical limb ischemia. J Transl Med 2024; 22:1003. [PMID: 39506809 PMCID: PMC11539538 DOI: 10.1186/s12967-024-05748-w] [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: 06/04/2024] [Accepted: 10/08/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Vasculogenic therapies explored for the treatment of peripheral artery disease (PAD) have encountered minimal success in clinical trials. Addressing this, B55α, an isoform of protein phosphatase 2A (PP2A), emerges as pivotal in vessel remodeling through activation of hypoxia-inducible factor 1α (HIF-1α). This study delves into the pharmacological profile of VCE-004.8 (Etrinabdione) and evaluates its efficacy in a preclinical model of critical limb ischemia, with a focus on its potential as a PP2A/B55α activator to induce angiogenesis and arteriogenesis. METHODS Vascular endothelial cells were used for in vitro experiments. Aorta ring assay was performed to explore sprouting activity. Matrigel plug-in assay was used to assess the angiogenic potential. Critical limb ischemia (CLI) in mice was induced by double ligation in the femoral arteria. Endothelial vascular and fibrotic biomarkers were studied by immunohistochemistry and qPCR. Arteriogenesis was investigated by microvascular casting and micro-CT. Proteomic analysis in vascular tissues was analyzed by LC-MS/MS. Ex-vivo expression of B55α and biomarkers were investigated in artery samples from PAD patients. RESULTS VCE-004.8 exhibited the ability to induce B55α expression and activate the intersecting pathways B55α/AMPK/Sirtuin 1/eNOS and B55α/PHD2/HIF-1α. VCE-004.8 prevented OxLDL and H2O2-induced cytotoxicity, senescence, and inflammation in endothelial cells. Oral VCE-004.8 increased aorta sprouting in vitro and angiogenesis in vivo. In CLI mice VCE-004.8 improved collateral vessel formation and induced endothelial cells proliferation, angiogenic gene expression and prevented fibrosis. The expression of B55α, Caveolin 1 and Sirtuin-1 is reduced in arteries from CLI mice and PAD patient, and the expression of these markers was restored in mice treated with VCE-004.8. CONCLUSIONS The findings presented in this study indicate that Etrinabdione holds promise in mitigating endothelial cell damage and senescence, while concurrently fostering arteriogenesis and angiogenesis. These observations position Etrinabdione as a compelling candidate for the treatment of PAD, and potentially other cardiovascular disorders.
Collapse
Affiliation(s)
- Adela García-Martín
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), University of Córdoba, Avda Menéndez Pidal s/n, 14004, Córdoba, Spain.
- Cellular Biology, Physiology and Immunology Department, University of Córdoba, Córdoba, Spain.
- Hospital Universitario Reina Sofía, Córdoba, Spain.
| | - María E Prados
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), University of Córdoba, Avda Menéndez Pidal s/n, 14004, Córdoba, Spain
- Cellular Biology, Physiology and Immunology Department, University of Córdoba, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Isabel Lastres-Cubillo
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), University of Córdoba, Avda Menéndez Pidal s/n, 14004, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Francisco J Ponce-Diaz
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), University of Córdoba, Avda Menéndez Pidal s/n, 14004, Córdoba, Spain
- Cellular Biology, Physiology and Immunology Department, University of Córdoba, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Laura Cerero
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), University of Córdoba, Avda Menéndez Pidal s/n, 14004, Córdoba, Spain
- Cellular Biology, Physiology and Immunology Department, University of Córdoba, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Martin Garrido-Rodríguez
- Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Heidelberg University, Bioquant, Heidelberg, Germany
| | - Carmen Navarrete
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), University of Córdoba, Avda Menéndez Pidal s/n, 14004, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Rafael Pineda
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), University of Córdoba, Avda Menéndez Pidal s/n, 14004, Córdoba, Spain
- Cellular Biology, Physiology and Immunology Department, University of Córdoba, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Ana B Rodríguez
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), University of Córdoba, Avda Menéndez Pidal s/n, 14004, Córdoba, Spain
- Cellular Biology, Physiology and Immunology Department, University of Córdoba, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Ignacio Muñoz
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), University of Córdoba, Avda Menéndez Pidal s/n, 14004, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Javier Moya
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), University of Córdoba, Avda Menéndez Pidal s/n, 14004, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Antonella Medeot
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), University of Córdoba, Avda Menéndez Pidal s/n, 14004, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - José A Moreno
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), University of Córdoba, Avda Menéndez Pidal s/n, 14004, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Antonio Chacón
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), University of Córdoba, Avda Menéndez Pidal s/n, 14004, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - José García-Revillo
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), University of Córdoba, Avda Menéndez Pidal s/n, 14004, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Eduardo Muñoz
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), University of Córdoba, Avda Menéndez Pidal s/n, 14004, Córdoba, Spain.
- Cellular Biology, Physiology and Immunology Department, University of Córdoba, Córdoba, Spain.
- Hospital Universitario Reina Sofía, Córdoba, Spain.
| |
Collapse
|
6
|
Boutin L, Soussi S, Garcia Lavello A, Masson Fron E, Deniau B, Legrand M, Blot-Chabaud M, Figueroa SM, Chadjichristos CE, Azibani F, Dépret F. Galectin-3 and Soluble CD146 Identify Cardiorenal Injuries in Severe Burn Patients: A Biomarker-Based Approach. Cardiorenal Med 2024; 14:460-472. [PMID: 39134005 DOI: 10.1159/000540845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 08/07/2024] [Indexed: 09/12/2024] Open
Abstract
INTRODUCTION Acute kidney injury (AKI) and myocardial injury (MI) are severe conditions in patients with severe burn injury, and combination of both is even worst and is called the cardiorenal syndrome (CRS). Identifying a distinct cardiorenal phenotype could significantly enhance the management of these patients. Galectin-3 (Gal3) and soluble CD146 (sCD146) are biomarkers for renal and cardiac injuries. This study aims to assess the occurrence and reliability of these biomarkers in recognizing CRS in individuals who have been severely burn. METHODS This study is a single-center prospective proof-of-concept study involving patients with severe burn injuries. Plasma samples for Gal3 and sCD146 measurements were collected daily during the initial 7 days following admission. CRS was defined after 24 h of admission by the association of AKI stage 1 or more (KDIGO definition) and MI defined on high sensitive troponin (hsTnT) (variation >20% baseline value or absolute value >40 ng/mL). RESULTS Forty patients met the inclusion criteria and were included in this study. Thirty-eight patients had CRS. The pooled values of Gal3 or combination of Gal3 and sCD146 values following 7 days after admission were associated with CRS with an odds ratio (OR) of 1.145 (95% CI: 1.081-1.211), p < 0.001, and 1.147 (95% CI: 1.085-1.212), p < 0.001, respectively. Gal3 values at admission (D0) had a predictive performance for CRS with an AUC of 0.78 (95% CI: 0.63-0.93), and this performance improved when using the combination of Gal3 and sCD146 values at admission (D0), with an AUC of 0.81 (95% CI: 0.66-0.96). Gal3 levels during the first 7 days were associated with patients experiencing AKI and no MI, with an OR of 1.129 (95% CI: 1.065-1.195), p < 0.001, and MI without AKI with an OR of 1.095 (95% CI: 1.037-1.167), p < 0.001. sCD146 alone was not associated with AKI without MI or MI without AKI and was poorly associated with CRS. CONCLUSION In severely burned patients, CRS is a frequent and severe condition. Gal3 values during the first 7 days following admission were associated with CRS. The use of sCD146 with Gal3 improved prediction performance for CRS identification. The use of such biomarkers to identify CRS is important and needs to be confirmed in other studies.
Collapse
Affiliation(s)
- Louis Boutin
- Department of Anaesthesiology, Critical Care Medicine and Burn Unit, AP-HP, Saint-Louis Hospital, DMU Parabol, FHU PROMICE, Université de Paris, Paris, France
- INSERM, UMR 942, MASCOT: Cardiovascular Marker in Stress Condition, Lariboisière Hospital, Université de Paris, Paris, France
- INSERM, UMR 1155, CORAKID, Tenon Hospital, Sorbonne Université, Paris, France
| | - Sabri Soussi
- Department of Anesthesia and Pain Management, University Health Network (UHN), Women's College Hospital, University of Toronto, Toronto Western Hospital, Toronto, Ontario, Canada
- St Michael's Hospital, Keenan Research Centre for Biomedical Science and Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Angèle Garcia Lavello
- Department of Anaesthesiology, Critical Care Medicine and Burn Unit, AP-HP, Saint-Louis Hospital, DMU Parabol, FHU PROMICE, Université de Paris, Paris, France
| | - Elisabeth Masson Fron
- Département de Biochimie et Biologie Moléculaire, Hôpitaux Lariboisière-Fernand Widal, Paris, France
| | - Banjamin Deniau
- Department of Anaesthesiology, Critical Care Medicine and Burn Unit, AP-HP, Saint-Louis Hospital, DMU Parabol, FHU PROMICE, Université de Paris, Paris, France
- INSERM, UMR 942, MASCOT: Cardiovascular Marker in Stress Condition, Lariboisière Hospital, Université de Paris, Paris, France
| | - Matthieu Legrand
- Division of Critical Care Medicine, Department of Anesthesiology and Peri-Operative Medicine, University of California - UCSF Medical Center, San Francisco, California, USA
| | | | | | | | - Feriel Azibani
- INSERM, UMR 942, MASCOT: Cardiovascular Marker in Stress Condition, Lariboisière Hospital, Université de Paris, Paris, France
| | - Fançois Dépret
- Department of Anaesthesiology, Critical Care Medicine and Burn Unit, AP-HP, Saint-Louis Hospital, DMU Parabol, FHU PROMICE, Université de Paris, Paris, France
- INSERM, UMR 942, MASCOT: Cardiovascular Marker in Stress Condition, Lariboisière Hospital, Université de Paris, Paris, France
| |
Collapse
|
7
|
Jiménez-González S, Delgado-Valero B, Islas F, Romero-Miranda A, Luaces M, Ramchandani B, Cuesta-Corral M, Montoro-Garrido A, Martínez-Martínez E, Cachofeiro V. The detrimental role of galectin-3 and endoplasmic reticulum stress in the cardiac consequences of myocardial ischemia in the context of obesity. FASEB J 2024; 38:e23818. [PMID: 38989572 DOI: 10.1096/fj.202400747r] [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: 04/03/2024] [Revised: 06/18/2024] [Accepted: 07/01/2024] [Indexed: 07/12/2024]
Abstract
The association between cardiac fibrosis and galectin-3 was evaluated in patients with acute myocardial infarction (MI). The role of galectin-3 and its association with endoplasmic reticulum (ER) stress activation in the progression of cardiovascular fibrosis was also evaluated in obese-infarcted rats. The inhibitor of galectin-3 activity, modified citrus pectin (MCP; 100 mg/kg/day), and the inhibitor of the ER stress activation, 4-phenylbutyric acid (4-PBA; 500 mg/kg/day), were administered for 4 weeks after MI in obese rats. Overweight-obese patients who suffered a first MI showed higher circulating galectin-3 levels, higher extracellular volume, and LV infarcted size, as well as lower E/e'ratio and LVEF compared with normal-weight patients. A correlation was observed between galectin-3 levels and extracellular volume. Obese-infarcted animals presented cardiac hypertrophy and reduction in LVEF, and E/A ratio as compared with control animals. They also showed an increase in galectin-3 gene expression, as well as cardiac fibrosis and reduced autophagic flux. These alterations were associated with ER stress activation characterized by enhanced cardiac levels of binding immunoglobulin protein, which were correlated with those of galectin-3. Both MCP and 4-PBA not only reduced cardiac fibrosis, oxidative stress, galectin-3 levels, and ER stress activation, but also prevented cardiac functional alterations and ameliorated autophagic flux. These results show the relevant role of galectin-3 in the development of diffuse fibrosis associated with MI in the context of obesity in both the animal model and patients. Galectin-3 in tandem with ER stress activation could modulate different downstream mechanisms, including inflammation, oxidative stress, and autophagy.
Collapse
Affiliation(s)
- Sara Jiménez-González
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, Madrid, Spain
| | - Beatriz Delgado-Valero
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, Madrid, Spain
| | - Fabian Islas
- Unidad de Imagen Cardíaca, Hospital General Universitario de Talavera de la Reina, Toledo, Spain
| | - Ana Romero-Miranda
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, Madrid, Spain
| | - María Luaces
- Servicio de Cardiología, Instituto Cardiovascular, Hospital Clínico San Carlos, Madrid, Spain
| | - Bunty Ramchandani
- Servicio de Cirugía Cardiaca Infantil, Hospital La Paz, Madrid, Spain
| | - María Cuesta-Corral
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, Madrid, Spain
| | - Alejandro Montoro-Garrido
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, Madrid, Spain
| | - Ernesto Martínez-Martínez
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, Madrid, Spain
- Ciber de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Majadahonda, Spain
| | - Victoria Cachofeiro
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, Madrid, Spain
- Ciber de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Majadahonda, Spain
| |
Collapse
|
8
|
Grujcic M, Milovanovic M, Nedeljkovic J, Jovanovic D, Arsenijevic D, Solovjova N, Stankovic V, Tanaskovic I, Arsenijevic A, Milovanovic J. The Possible Effects of Galectin-3 on Mechanisms of Renal and Hepatocellular Injury Induced by Intravascular Hemolysis. Int J Mol Sci 2024; 25:8129. [PMID: 39125698 PMCID: PMC11311984 DOI: 10.3390/ijms25158129] [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: 06/05/2024] [Revised: 07/20/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
Intravascular hemolysis is a central feature of congenital and acquired hemolytic anemias, complement disorders, infectious diseases, and toxemias. Massive and/or chronic hemolysis is followed by the induction of inflammation, very often with severe damage of organs, which enhances the morbidity and mortality of hemolytic diseases. Galectin-3 (Gal-3) is a β-galactoside-binding lectin that modulates the functions of many immune cells, thus affecting inflammatory processes. Gal-3 is also one of the main regulators of fibrosis. The role of Gal-3 in the development of different kidney and liver diseases and the potential of therapeutic Gal-3 inhibition have been demonstrated. Therefore, the objective of this review is to discuss the possible effects of Gal-3 on the process of kidney and liver damage induced by intravascular hemolysis, as well as to shed light on the potential therapeutic targeting of Gal-3 in intravascular hemolysis.
Collapse
Affiliation(s)
- Mirjana Grujcic
- Institute for Transfusiology and Hemobiology of Military Medical Academy, 11000 Belgrade, Serbia;
| | - Marija Milovanovic
- Center for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia; (M.M.); (D.A.); (V.S.); (A.A.)
- Department of Microbiology and Immunology, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia
| | - Jelena Nedeljkovic
- Department of Medical Statistics and Informatics, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Danijela Jovanovic
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Dragana Arsenijevic
- Center for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia; (M.M.); (D.A.); (V.S.); (A.A.)
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Natalija Solovjova
- Academy of Applied Studies Belgrade, The College of Health Science, Cara Dušana 254, 11080 Belgrade, Serbia;
| | - Vesna Stankovic
- Center for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia; (M.M.); (D.A.); (V.S.); (A.A.)
- Department of Pathology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Irena Tanaskovic
- Center for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia; (M.M.); (D.A.); (V.S.); (A.A.)
- Department of Histology and Embriology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Aleksandar Arsenijevic
- Center for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia; (M.M.); (D.A.); (V.S.); (A.A.)
- Department of Microbiology and Immunology, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia
| | - Jelena Milovanovic
- Center for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia; (M.M.); (D.A.); (V.S.); (A.A.)
- Department of Histology and Embriology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| |
Collapse
|
9
|
Hao WR, Cheng CH, Liu JC, Chen HY, Chen JJ, Cheng TH. Understanding Galectin-3's Role in Diastolic Dysfunction: A Contemporary Perspective. Life (Basel) 2024; 14:906. [PMID: 39063659 PMCID: PMC11277993 DOI: 10.3390/life14070906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
Diastolic dysfunction, a prevalent condition characterized by impaired relaxation and filling of the left ventricle, significantly contributes to heart failure with preserved ejection fraction (HFpEF). Galectin-3, a β-galactoside-binding lectin, has garnered attention as a potential biomarker and mediator of fibrosis and inflammation in cardiovascular diseases. This comprehensive review investigates the impact of galectin-3 on diastolic dysfunction. We explore its molecular mechanisms, including its involvement in cellular signaling pathways and interaction with components of the extracellular matrix. Evidence from both animal models and clinical studies elucidates galectin-3's role in cardiac remodeling, inflammation, and fibrosis, shedding light on the underlying pathophysiology of diastolic dysfunction. Additionally, we examine the diagnostic and therapeutic implications of galectin-3 in diastolic dysfunction, emphasizing its potential as both a biomarker and a therapeutic target. This review underscores the significance of comprehending galectin-3's role in diastolic dysfunction and its promise in enhancing diagnosis and treatment approaches for HFpEF patients.
Collapse
Affiliation(s)
- Wen-Rui Hao
- Division of Cardiology, Department of Internal Medicine, Shuang Ho Hospital, Ministry of Health and Welfare, Taipei Medical University, New Taipei City 23561, Taiwan; (W.-R.H.); (J.-C.L.)
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei City 11002, Taiwan
| | - Chun-Han Cheng
- Department of Medical Education, Linkou Chang Gung Memorial Hospital, Taoyuan City 33305, Taiwan;
| | - Ju-Chi Liu
- Division of Cardiology, Department of Internal Medicine, Shuang Ho Hospital, Ministry of Health and Welfare, Taipei Medical University, New Taipei City 23561, Taiwan; (W.-R.H.); (J.-C.L.)
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei City 11002, Taiwan
| | - Huan-Yuan Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan; (H.-Y.C.); (J.-J.C.)
| | - Jin-Jer Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan; (H.-Y.C.); (J.-J.C.)
- Division of Cardiology, Department of Internal Medicine and Graduate Institute of Clinical Medical Science, China Medical University, Taichung City 404333, Taiwan
| | - Tzu-Hurng Cheng
- Department of Biochemistry, School of Medicine, College of Medicine, China Medical University, Taichung City 404333, Taiwan
| |
Collapse
|
10
|
Camarda ND, Ibarrola J, Biwer LA, Jaffe IZ. Mineralocorticoid Receptors in Vascular Smooth Muscle: Blood Pressure and Beyond. Hypertension 2024; 81:1008-1020. [PMID: 38426347 PMCID: PMC11023801 DOI: 10.1161/hypertensionaha.123.21358] [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] [Indexed: 03/02/2024]
Abstract
After half a century of evidence suggesting the existence of mineralocorticoid receptors (MR) in the vasculature, the advent of technology to specifically knockout the MR from smooth muscle cells (SMCs) in mice has elucidated contributions of SMC-MR to cardiovascular function and disease, independent of the kidney. This review summarizes the latest understanding of the molecular mechanisms by which SMC-MR contributes to (1) regulation of vasomotor function and blood pressure to contribute to systemic and pulmonary hypertension; (2) vascular remodeling in response to hypertension, vascular injury, obesity, and aging, and the impact on vascular calcification; and (3) cardiovascular pathologies including aortic aneurysm, heart valve dysfunction, and heart failure. Data are reviewed from in vitro studies using SMCs and in vivo findings from SMC-specific MR-knockout mice that implicate target genes and signaling pathways downstream of SMC-MR. By regulating expression of the L-type calcium channel subunit Cav1.2 and angiotensin II type-1 receptor, SMC-MR contributes to myogenic tone and vasoconstriction, thereby contributing to systemic blood pressure. MR activation also promotes SMC proliferation, migration, production and degradation of extracellular matrix, and osteogenic differentiation by regulating target genes including connective tissue growth factor, osteopontin, bone morphogenetic protein 2, galectin-3, and matrix metallopeptidase-2. By these mechanisms, SMC-MR promotes disease progression in models of aging-associated vascular stiffness, vascular calcification, mitral and aortic valve disease, pulmonary hypertension, and heart failure. While rarely tested, when sexes were compared, the mechanisms of SMC-MR-mediated disease were sexually dimorphic. These advances support targeting SMC-MR-mediated mechanisms to prevent and treat diverse cardiovascular disorders.
Collapse
Affiliation(s)
- Nicholas D. Camarda
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Jaime Ibarrola
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Lauren A. Biwer
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
| | - Iris Z. Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
| |
Collapse
|
11
|
Koletsos N, Lazaridis A, Triantafyllou A, Anyfanti P, Lamprou S, Stoimeni A, Papadopoulos NG, Koravou EE, Gkaliagkousi E. Accumulation of Microvascular Target Organ Damage in Systemic Lupus Erythematosus Patients Is Associated with Increased Cardiovascular Risk. J Clin Med 2024; 13:2140. [PMID: 38610905 PMCID: PMC11012611 DOI: 10.3390/jcm13072140] [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: 03/12/2024] [Revised: 03/27/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024] Open
Abstract
Background: Systemic lupus erythematosus (SLE) is a prototype autoimmune disease associated with increased cardiovascular (CV) burden. Besides increased arterial stiffness and subclinical atherosclerosis, microvascular dysfunction is considered an important component in the pathophysiology of CV disease. However, there is a lack of data regarding the effect of multiple target organ damage (TOD) on CV health. Objectives: This study aimed to evaluate (i) the presence of microvascular changes in SLE in various vascular beds, (ii) the possible associations between the accumulation of microvascular TOD and CV risk and (iii) whether Galectin-3 represents a predictor of combined microvascular TOD. Methods: Participants underwent (i) evaluation of skin microvascular perfusion (laser speckle contrast analysis), (ii) fundoscopy (non-mydriatic fundus camera), (iii) indirect assessment of myocardial perfusion (subendocardial viability ratio) and (iv) determination of urine albumin-to-creatinine ratio (UACR). CV risk was calculated using the QResearch Risk Estimator version 3 (QRISK3). Serum Galectin-3 levels were determined. Results: Forty-seven SLE patients and fifty controls were studied. SLE patients demonstrated impaired skin microvascular reactivity (160.2 ± 41.0 vs. 203.6 ± 40.1%), retinal arteriolar narrowing (88.1 ± 11.1 vs. 94.6 ± 13.5 μm) and higher UACR levels compared to controls. Furthermore, SLE individuals had significantly higher Galectin-3 levels [21.5(6.1) vs. 6.6(6.6) ng/dL], QRISK3 scores [7.0(8.6) vs. 1.3(3.6)%] and a greater chance for microvascular dysfunction. In the SLE group, patients with multiple TOD exhibited higher QRISK3. In the multivariate analysis, the accumulation of TOD correlated with disease activity and Galectin-3 (p < 0.05). Conclusions: Our study showed for the first time that SLE patients exhibit a greater number of cases of TOD. The accumulation of TOD was associated with increased CV risk. Clinicians dealing with SLE should be aware and seek microvascular alterations.
Collapse
Affiliation(s)
- Nikolaos Koletsos
- 3rd Department of Internal Medicine, Papageorgiou General Hospital, Aristotle University of Thessaloniki, 56429 Thessaloniki, Greece; (A.L.); (A.T.); (S.L.); (A.S.); (E.G.)
| | - Antonios Lazaridis
- 3rd Department of Internal Medicine, Papageorgiou General Hospital, Aristotle University of Thessaloniki, 56429 Thessaloniki, Greece; (A.L.); (A.T.); (S.L.); (A.S.); (E.G.)
| | - Areti Triantafyllou
- 3rd Department of Internal Medicine, Papageorgiou General Hospital, Aristotle University of Thessaloniki, 56429 Thessaloniki, Greece; (A.L.); (A.T.); (S.L.); (A.S.); (E.G.)
| | - Panagiota Anyfanti
- Second Medical Department, Hippokration General Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece;
| | - Stamatina Lamprou
- 3rd Department of Internal Medicine, Papageorgiou General Hospital, Aristotle University of Thessaloniki, 56429 Thessaloniki, Greece; (A.L.); (A.T.); (S.L.); (A.S.); (E.G.)
| | - Anastasia Stoimeni
- 3rd Department of Internal Medicine, Papageorgiou General Hospital, Aristotle University of Thessaloniki, 56429 Thessaloniki, Greece; (A.L.); (A.T.); (S.L.); (A.S.); (E.G.)
| | | | | | - Eugenia Gkaliagkousi
- 3rd Department of Internal Medicine, Papageorgiou General Hospital, Aristotle University of Thessaloniki, 56429 Thessaloniki, Greece; (A.L.); (A.T.); (S.L.); (A.S.); (E.G.)
| |
Collapse
|
12
|
Calvier L, Alexander A, Marckx AT, Kounnas MZ, Durakoglugil M, Herz J. Safety of Anti-Reelin Therapeutic Approaches for Chronic Inflammatory Diseases. Cells 2024; 13:583. [PMID: 38607022 PMCID: PMC11011630 DOI: 10.3390/cells13070583] [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: 02/23/2024] [Revised: 03/18/2024] [Accepted: 03/25/2024] [Indexed: 04/13/2024] Open
Abstract
Reelin, a large extracellular glycoprotein, plays critical roles in neuronal development and synaptic plasticity in the central nervous system (CNS). Recent studies have revealed non-neuronal functions of plasma Reelin in inflammation by promoting endothelial-leukocyte adhesion through its canonical pathway in endothelial cells (via ApoER2 acting on NF-κB), as well as in vascular tone regulation and thrombosis. In this study, we have investigated the safety and efficacy of selectively depleting plasma Reelin as a potential therapeutic strategy for chronic inflammatory diseases. We found that Reelin expression remains stable throughout adulthood and that peripheral anti-Reelin antibody treatment with CR-50 efficiently depletes plasma Reelin without affecting its levels or functionality within the CNS. Notably, this approach preserves essential neuronal functions and synaptic plasticity. Furthermore, in mice induced with experimental autoimmune encephalomyelitis (EAE), selective modulation of endothelial responses by anti-Reelin antibodies reduces pathological leukocyte infiltration without completely abolishing diapedesis. Finally, long-term Reelin depletion under metabolic stress induced by a Western diet did not negatively impact the heart, kidney, or liver, suggesting a favorable safety profile. These findings underscore the promising role of peripheral anti-Reelin therapeutic strategies for autoimmune diseases and conditions where endothelial function is compromised, offering a novel approach that may avoid the immunosuppressive side effects associated with conventional anti-inflammatory therapies.
Collapse
Affiliation(s)
- Laurent Calvier
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas, TX 75390, USA (A.T.M.); (M.D.); (J.H.)
- Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Anna Alexander
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas, TX 75390, USA (A.T.M.); (M.D.); (J.H.)
- Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Austin T. Marckx
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas, TX 75390, USA (A.T.M.); (M.D.); (J.H.)
- Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | | | - Murat Durakoglugil
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas, TX 75390, USA (A.T.M.); (M.D.); (J.H.)
- Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Joachim Herz
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas, TX 75390, USA (A.T.M.); (M.D.); (J.H.)
- Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
13
|
Cao Z, Ramadan A, Tai A, Zetterberg F, Panjwani N. Anti-Angiogenic and Anti-Scarring Dual Effect of Galectin-3 Inhibition in Mouse Models of Corneal Wound Healing. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:447-458. [PMID: 38159722 DOI: 10.1016/j.ajpath.2023.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 11/05/2023] [Accepted: 11/28/2023] [Indexed: 01/03/2024]
Abstract
Corneal scarring is the third leading cause of global blindness. Neovascularization of ocular tissues is a major predisposing factor in scar development. Although corneal transplantation is effective in restoring vision, some patients are at high risk for graft rejection due to the presence of blood vessels in the injured cornea. Current treatment options for controlling corneal scarring are limited, and outcomes are typically poor. In this study, topical application of a small-molecule inhibitor of galectin-3, GB1265, in mouse models of corneal wound healing, led to the reduction of the following in injured corneas: i) corneal angiogenesis; ii) corneal fibrosis; iii) infiltration of immune cells; and iv) expression of the proinflammatory cytokine IL-1β. Four independent techniques (RNA sequencing, NanoString, real-time quantitative RT-PCR, and Western blot analysis) determined that decreased corneal opacity in the galectin-3 inhibitor-treated corneas was associated with decreases in the numbers of genes and signaling pathways known to promote fibrosis. These findings allowed for a high level of confidence in the conclusion that galectin-3 inhibition by the small-molecule inhibitor GB1265 has dual anti-angiogenic and anti-scarring effects. Targeting galectin-3 by GB1265 is, thus, attractive for the development of innovative therapies for a myriad of ocular and nonocular diseases characterized by pathologic angiogenesis and fibrosis.
Collapse
Affiliation(s)
- Zhiyi Cao
- New England Eye Center/Department of Ophthalmology, Tufts University School of Medicine, Boston, Massachusetts
| | - Abdulraouf Ramadan
- New England Eye Center/Department of Ophthalmology, Tufts University School of Medicine, Boston, Massachusetts
| | - Albert Tai
- Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts
| | | | - Noorjahan Panjwani
- New England Eye Center/Department of Ophthalmology, Tufts University School of Medicine, Boston, Massachusetts; Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Boston, Massachusetts.
| |
Collapse
|
14
|
Chan MJ, Liu KD. Acute Kidney Injury and Subsequent Cardiovascular Disease: Epidemiology, Pathophysiology, and Treatment. Semin Nephrol 2024; 44:151515. [PMID: 38849258 DOI: 10.1016/j.semnephrol.2024.151515] [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: 06/09/2024]
Abstract
Cardiovascular disease poses a significant threat to individuals with kidney disease, including those affected by acute kidney injury (AKI). In the short term, AKI has several physiological consequences that can impact the cardiovascular system. These include fluid and sodium overload, activation of the renin-angiotensin-aldosterone system and sympathetic nervous system, and inflammation along with metabolic complications of AKI (acidosis, electrolyte imbalance, buildup of uremic toxins). Recent studies highlight the role of AKI in elevating long-term risks of hypertension, thromboembolism, stroke, and major adverse cardiovascular events, though some of this increased risk may be due to the impact of AKI on the course of chronic kidney disease. Current management strategies involve avoiding nephrotoxic agents, optimizing hemodynamics and fluid balance, and considering renin-angiotensin-aldosterone system inhibition or sodium-glucose cotransporter 2 inhibitors. However, future research is imperative to advance preventive and therapeutic strategies for cardiovascular complications in AKI. This review explores the existing knowledge on the cardiovascular consequences of AKI, delving into epidemiology, pathophysiology, and treatment of various cardiovascular complications following AKI.
Collapse
Affiliation(s)
- Ming-Jen Chan
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Taoyuan, Taiwan; Graduate Institute of Clinical Medical Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Kathleen D Liu
- Divisions of Nephrology and Critical Care Medicine, Departments of Medicine and Anesthesia, University of California, San Francisco, CA.
| |
Collapse
|
15
|
Xiao Y, Vazquez-Padron RI, Martinez L, Singer HA, Woltmann D, Salman LH. Role of platelet factor 4 in arteriovenous fistula maturation failure: What do we know so far? J Vasc Access 2024; 25:390-406. [PMID: 35751379 PMCID: PMC9974241 DOI: 10.1177/11297298221085458] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The rate of arteriovenous fistula (AVF) maturation failure remains unacceptably high despite continuous efforts on technique improvement and careful pre-surgery planning. In fact, half of all newly created AVFs are unable to be used for hemodialysis (HD) without a salvage procedure. While vascular stenosis in the venous limb of the access is the culprit, the underlying factors leading to vascular narrowing and AVF maturation failure are yet to be determined. We have recently demonstrated that AVF non-maturation is associated with post-operative medial fibrosis and fibrotic stenosis, and post-operative intimal hyperplasia (IH) exacerbates the situation. Multiple pathological processes and signaling pathways are underlying the stenotic remodeling of the AVF. Our group has recently indicated that a pro-inflammatory cytokine platelet factor 4 (PF4/CXCL4) is upregulated in veins that fail to mature after AVF creation. Platelet factor 4 is a fibrosis marker and can be detected in vascular stenosis tissue, suggesting that it may contribute to AVF maturation failure through stimulation of fibrosis and development of fibrotic stenosis. Here, we present an overview of the how PF4-mediated fibrosis determines AVF maturation failure.
Collapse
Affiliation(s)
- Yuxuan Xiao
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Roberto I Vazquez-Padron
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Laisel Martinez
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Harold A Singer
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Daniel Woltmann
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Loay H Salman
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
- Division of Nephrology and Hypertension, Albany Medical College, Albany, NY, USA
| |
Collapse
|
16
|
Wang G, Li R, Feng C, Li K, Liu S, Fu Q. Galectin-3 is involved in inflammation and fibrosis in arteriogenic erectile dysfunction via the TLR4/MyD88/NF-κB pathway. Cell Death Discov 2024; 10:92. [PMID: 38378809 PMCID: PMC10879531 DOI: 10.1038/s41420-024-01859-x] [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: 08/10/2023] [Revised: 02/04/2024] [Accepted: 02/08/2024] [Indexed: 02/22/2024] Open
Abstract
Galectin-3 (Gal-3) is a multifunctional protein that has been linked to fibrosis and inflammation in the cardiovascular system. In this study, we examined the impact of Gal-3 on inflammation and fibrosis in patients with arteriogenic erectile dysfunction (A-ED) and the underlying mechanisms involved. To induce arterial injury, we utilized cuffs on the periaqueductal common iliac arteries of Sprague‒Dawley (SD) rats and administered a high-fat diet to co-induce local atherosclerosis. Our results showed that we successfully developed a novel A-ED model that was validated based on histological evidence. In vivo, the vascular lumen of rats subjected to a high-fat diet and cuff placement exhibited significant narrowing, accompanied by the upregulation of Gal-3, Toll-like receptor 4 (TLR4), and myeloid differentiation primary response protein 88 (MyD88) expression in the penile cavernosa. This led to the activation of nuclear factor kappa B 65 (NF-κB-p65), resulting in reduced intracavernosal pressure, endothelial nitric oxide synthase expression, and smooth muscle content, promoting inflammation and fibrosis. However, treatment with Gal-3 inhibitor-modified citrus pectin (MCP) significantly normalized those effects. In vitro, knocking down Gal-3 led to a significant reduction in TLR4, MyD88, and NF-κB-p65 expression in corpus cavernosum smooth muscle cells (CCSMCs), decreasing inflammation levels. In conclusion, inhibiting Gal-3 may improve A-ED by reducing inflammation, endothelial injury, and fibrosis in the penile corpus cavernosum through the TLR4/MyD88/NF-κB pathway. These findings highlight the potential therapeutic target of Gal-3 in A-ED.
Collapse
Affiliation(s)
- Guanbo Wang
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ruiyu Li
- Department of Urology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Chen Feng
- Department of Urology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Kefan Li
- Department of Urology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Shuai Liu
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.
- Department of Urology, Shandong Provincial Hospital, Shandong University, Jinan, China.
- Engineering Laboratory of Urinary Organ and Functional Reconstruction of Shandong Province, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.
| | - Qiang Fu
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.
- Department of Urology, Shandong Provincial Hospital, Shandong University, Jinan, China.
- Engineering Laboratory of Urinary Organ and Functional Reconstruction of Shandong Province, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.
| |
Collapse
|
17
|
Hua R, Gao H, He C, Xin S, Wang B, Zhang S, Gao L, Tao Q, Wu W, Sun F, Xu J. An emerging view on vascular fibrosis molecular mediators and relevant disorders: from bench to bed. Front Cardiovasc Med 2023; 10:1273502. [PMID: 38179503 PMCID: PMC10764515 DOI: 10.3389/fcvm.2023.1273502] [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: 08/06/2023] [Accepted: 11/27/2023] [Indexed: 01/06/2024] Open
Abstract
Vascular fibrosis is a widespread pathologic condition that arises during vascular remodeling in cardiovascular dysfunctions. According to previous studies, vascular fibrosis is characterized by endothelial matrix deposition and vascular wall thickening. The RAAS and TGF-β/Smad signaling pathways have been frequently highlighted. It is, however, far from explicit in terms of understanding the cause and progression of vascular fibrosis. In this review, we collected and categorized a large number of molecules which influence the fibrosing process, in order to acquire a better understanding of vascular fibrosis, particularly of pathologic dysfunction. Furthermore, several mediators that prevent vascular fibrosis are discussed in depth in this review, with the aim that this will contribute to the future prevention and treatment of related conditions.
Collapse
Affiliation(s)
- Rongxuan Hua
- Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Han Gao
- Department of Clinical Laboratory, Aerospace Center Hospital, Peking University, Beijing, China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Chengwei He
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Shuzi Xin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Boya Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, Beijing, China
| | - Sitian Zhang
- Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Lei Gao
- Department of Biomedical Informatics, School of Biomedical Engineering, Capital Medical University, Beijing, China
| | - Qiang Tao
- Department of Biomedical Informatics, School of Biomedical Engineering, Capital Medical University, Beijing, China
| | - Wenqi Wu
- Experimental Center for Morphological Research Platform, Capital Medical University, Beijing, China
| | - Fangling Sun
- Department of Experimental Animal Laboratory, Xuan-Wu Hospital of Capital Medical University, Beijing, China
| | - Jingdong Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
18
|
Seropian IM, Cassaglia P, Miksztowicz V, González GE. Unraveling the role of galectin-3 in cardiac pathology and physiology. Front Physiol 2023; 14:1304735. [PMID: 38170009 PMCID: PMC10759241 DOI: 10.3389/fphys.2023.1304735] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 12/05/2023] [Indexed: 01/05/2024] Open
Abstract
Galectin-3 (Gal-3) is a carbohydrate-binding protein with multiple functions. Gal-3 regulates cell growth, proliferation, and apoptosis by orchestrating cell-cell and cell-matrix interactions. It is implicated in the development and progression of cardiovascular disease, and its expression is increased in patients with heart failure. In atherosclerosis, Gal-3 promotes monocyte recruitment to the arterial wall boosting inflammation and atheroma. In acute myocardial infarction (AMI), the expression of Gal-3 increases in infarcted and remote zones from the beginning of AMI, and plays a critical role in macrophage infiltration, differentiation to M1 phenotype, inflammation and interstitial fibrosis through collagen synthesis. Genetic deficiency of Gal-3 delays wound healing, impairs cardiac remodeling and function after AMI. On the contrary, Gal-3 deficiency shows opposite results with improved remodeling and function in other cardiomyopathies and in hypertension. Pharmacologic inhibition with non-selective inhibitors is also protective in cardiac disease. Finally, we recently showed that Gal-3 participates in normal aging. However, genetic absence of Gal-3 in aged mice exacerbates pathological hypertrophy and increases fibrosis, as opposed to reduced fibrosis shown in cardiac disease. Despite some gaps in understanding its precise mechanisms of action, Gal-3 represents a potential therapeutic target for the treatment of cardiovascular diseases and the management of cardiac aging. In this review, we summarize the current knowledge regarding the role of Gal-3 in the pathophysiology of heart failure, atherosclerosis, hypertension, myocarditis, and ischemic heart disease. Furthermore, we describe the physiological role of Gal-3 in cardiac aging.
Collapse
Affiliation(s)
- Ignacio M. Seropian
- Laboratorio de Patología Cardiovascular Experimental e Hipertensión Arterial, Instituto de Investigaciones Biomédicas (UCA-CONICET), Facultad de Ciencias Médicas Universidad Católica Argentina, Buenos Aires, Argentina
- Servicio de Hemodinamia, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Pablo Cassaglia
- Departamento de Patología, Instituto de Salud Comunitaria, Universidad Nacional de Hurlingham, Buenos Aires, Argentina
| | - Verónica Miksztowicz
- Laboratorio de Patología Cardiovascular Experimental e Hipertensión Arterial, Instituto de Investigaciones Biomédicas (UCA-CONICET), Facultad de Ciencias Médicas Universidad Católica Argentina, Buenos Aires, Argentina
| | - Germán E. González
- Laboratorio de Patología Cardiovascular Experimental e Hipertensión Arterial, Instituto de Investigaciones Biomédicas (UCA-CONICET), Facultad de Ciencias Médicas Universidad Católica Argentina, Buenos Aires, Argentina
- Departamento de Patología, Instituto de Salud Comunitaria, Universidad Nacional de Hurlingham, Buenos Aires, Argentina
| |
Collapse
|
19
|
Lequain H, Dégletagne C, Streichenberger N, Valantin J, Simonet T, Schaeffer L, Sève P, Leblanc P. Spatial Transcriptomics Reveals Signatures of Histopathological Changes in Muscular Sarcoidosis. Cells 2023; 12:2747. [PMID: 38067175 PMCID: PMC10706822 DOI: 10.3390/cells12232747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 11/25/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Sarcoidosis is a multisystemic disease characterized by non-caseating granuloma infiltrating various organs. The form with symptomatic muscular involvement is called muscular sarcoidosis. The impact of immune cells composing the granuloma on the skeletal muscle is misunderstood. Here, we investigated the granuloma-skeletal muscle interactions through spatial transcriptomics on two patients affected by muscular sarcoidosis. Five major transcriptomic clusters corresponding to perigranuloma, granuloma, and three successive muscle tissue areas (proximal, intermediate, and distal) around the granuloma were identified. Analyses revealed upregulated pathways in the granuloma corresponding to the activation of T-lymphocytes and monocytes/macrophages cytokines, the upregulation of extracellular matrix signatures, and the induction of the TGF-β signaling in the perigranuloma. A comparison between the proximal and distal muscles to the granuloma revealed an inverse correlation between the distance to the granuloma and the upregulation of cellular response to interferon-γ/α, TNF-α, IL-1,4,6, fibroblast proliferation, epithelial to mesenchymal cell transition, and the downregulation of muscle gene expression. These data shed light on the intercommunications between granulomas and the muscle tissue and provide pathophysiological mechanisms by showing that granuloma immune cells have a direct impact on proximal muscle tissue by promoting its progressive replacement by fibrosis via the expression of pro-inflammatory and profibrosing signatures. These data could possibly explain the evolution towards a state of disability for some patients.
Collapse
Affiliation(s)
- Hippolyte Lequain
- Département de Médecine Interne, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, 69004 Lyon, France;
- Institut NeuroMyoGène INMG-PGNM, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, Inserm U1315, Faculté de Médecine Rockefeller, Université Claude Bernard UCBL-Lyon 1, 69008 Lyon, France; (N.S.); (T.S.)
| | - Cyril Dégletagne
- CRCL Core Facilities, Centre de Recherche en Cancérologie de Lyon (CRCL) INSERM U1052-CNRS UMR5286, Université de Lyon, Université Claude Bernard Lyon1, Centre Léon Bérard, 69008 Lyon, France; (C.D.); (J.V.)
| | - Nathalie Streichenberger
- Institut NeuroMyoGène INMG-PGNM, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, Inserm U1315, Faculté de Médecine Rockefeller, Université Claude Bernard UCBL-Lyon 1, 69008 Lyon, France; (N.S.); (T.S.)
- Service d’Anatomopathologie, Centre de Biologie et Pathologie Est (CBPE), Hospices Civils de Lyon, 69500 Bron, France
| | - Julie Valantin
- CRCL Core Facilities, Centre de Recherche en Cancérologie de Lyon (CRCL) INSERM U1052-CNRS UMR5286, Université de Lyon, Université Claude Bernard Lyon1, Centre Léon Bérard, 69008 Lyon, France; (C.D.); (J.V.)
| | - Thomas Simonet
- Institut NeuroMyoGène INMG-PGNM, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, Inserm U1315, Faculté de Médecine Rockefeller, Université Claude Bernard UCBL-Lyon 1, 69008 Lyon, France; (N.S.); (T.S.)
| | - Laurent Schaeffer
- Institut NeuroMyoGène INMG-PGNM, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, Inserm U1315, Faculté de Médecine Rockefeller, Université Claude Bernard UCBL-Lyon 1, 69008 Lyon, France; (N.S.); (T.S.)
- Centre de Biotechnologie Cellulaire, CHU de Lyon—HCL Groupement Est, 69677 Bron, France
| | - Pascal Sève
- Département de Médecine Interne, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, 69004 Lyon, France;
- Pôle IMER, HESPER EA 7425, 69002 Lyon, France
| | - Pascal Leblanc
- Institut NeuroMyoGène INMG-PGNM, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, Inserm U1315, Faculté de Médecine Rockefeller, Université Claude Bernard UCBL-Lyon 1, 69008 Lyon, France; (N.S.); (T.S.)
| |
Collapse
|
20
|
Fries R. Hypertrophic Cardiomyopathy-Advances in Imaging and Diagnostic Strategies. Vet Clin North Am Small Anim Pract 2023; 53:1325-1342. [PMID: 37423845 DOI: 10.1016/j.cvsm.2023.05.010] [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: 07/11/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is the most important and prevalent cardiac disease in cats. Due to the highly variable nature of HCM, a multimodal approach including physical examination, genetic evaluation, cardiac biomarkers, and imaging are all essential elements to appropriate and timely diagnosis. These foundational elements are advancing rapidly in veterinary medicine. Newer biomarkers such as galectin-3 are currently being researched and advances in tissue speckle-tracking and contrast-enhanced echocardiography are readily available. Advanced imaging techniques, such as cardiac MRI, are providing previously unavailable information about myocardial fibrosis and paving the way for enhanced diagnostic capabilities and risk-stratification in cats with HCM.
Collapse
Affiliation(s)
- Ryan Fries
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, 1008 West Hazelwood Drive, Urbana, IL 61802, USA.
| |
Collapse
|
21
|
Zhou Z, Feng Z, Sun X, Wang Y, Dou G. The Role of Galectin-3 in Retinal Degeneration and Other Ocular Diseases: A Potential Novel Biomarker and Therapeutic Target. Int J Mol Sci 2023; 24:15516. [PMID: 37958500 PMCID: PMC10649114 DOI: 10.3390/ijms242115516] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 11/15/2023] Open
Abstract
Galectin-3 is the most studied member of the Galectin family, with a large range of mediation in biological activities such as cell growth, proliferation, apoptosis, differentiation, cell adhesion, and tissue repair, as well as in pathological processes such as inflammation, tissue fibrosis, and angiogenesis. As is known to all, inflammation, aberrant cell apoptosis, and neovascularization are the main pathophysiological processes in retinal degeneration and many ocular diseases. Therefore, the review aims to conclude the role of Gal3 in the retinal degeneration of various diseases as well as the occurrence and development of the diseases and discuss its molecular mechanisms according to research in systemic diseases. At the same time, we summarized the predictive role of Gal3 as a biomarker and the clinical application of its inhibitors to discuss the possibility of Gal3 as a novel target for the treatment of ocular diseases.
Collapse
Affiliation(s)
| | | | | | - Yusheng Wang
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China; (Z.Z.); (Z.F.); (X.S.)
| | - Guorui Dou
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China; (Z.Z.); (Z.F.); (X.S.)
| |
Collapse
|
22
|
Matilla L, Martín-Núñez E, Garaikoetxea M, Navarro A, Tamayo I, Fernández-Celis A, Gainza A, Fernández-Irigoyen J, Santamaría E, Muntendam P, Álvarez V, Sádaba R, Jover E, López-Andrés N. Sex-specific role of galectin-3 in aortic stenosis. Biol Sex Differ 2023; 14:72. [PMID: 37875993 PMCID: PMC10598900 DOI: 10.1186/s13293-023-00556-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 10/10/2023] [Indexed: 10/26/2023] Open
Abstract
BACKGROUND Aortic stenosis (AS) is characterized by inflammation, fibrosis, osteogenesis and angiogenesis. Men and women develop these mechanisms differently. Galectin-3 (Gal-3) is a pro-inflammatory and pro-osteogenic lectin in AS. In this work, we aim to analyse a potential sex-differential role of Gal-3 in AS. METHODS 226 patients (61.50% men) with severe AS undergoing surgical aortic valve (AV) replacement were recruited. In AVs, Gal-3 expression and its relationship with inflammatory, osteogenic and angiogenic markers was assessed. Valve interstitial cells (VICs) were primary cultured to perform in vitro experiments. RESULTS Proteomic analysis revealed that intracellular Gal-3 was over-expressed in VICs of male AS patients. Gal-3 secretion was also higher in men's VICs as compared to women's. In human AVs, Gal-3 protein levels were significantly higher in men, with stronger immunostaining in VICs with myofibroblastic phenotype and valve endothelial cells. Gal-3 levels in AVs were positively correlated with inflammatory markers in both sexes. Gal-3 expression was also positively correlated with osteogenic markers mainly in men AVs, and with angiogenic molecules only in this sex. In vitro, Gal-3 treatment induced expression of inflammatory, osteogenic and angiogenic markers in male's VICs, while it only upregulated inflammatory and osteogenic molecules in women-derived cells. Gal-3 blockade with pharmacological inhibitors (modified citrus pectin and G3P-01) prevented the upregulation of inflammatory, osteogenic and angiogenic molecules. CONCLUSIONS Gal-3 plays a sex-differential role in the setting of AS, and it could be a new sex-specific therapeutic target controlling pathological features of AS in VICs.
Collapse
Affiliation(s)
- Lara Matilla
- Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IdiSNA, C/Irunlarrea 3., 31008, Pamplona, Spain
| | - Ernesto Martín-Núñez
- Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IdiSNA, C/Irunlarrea 3., 31008, Pamplona, Spain
| | - Mattie Garaikoetxea
- Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IdiSNA, C/Irunlarrea 3., 31008, Pamplona, Spain
| | - Adela Navarro
- Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IdiSNA, C/Irunlarrea 3., 31008, Pamplona, Spain
| | - Ibai Tamayo
- Research Methodology Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain
| | - Amaya Fernández-Celis
- Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IdiSNA, C/Irunlarrea 3., 31008, Pamplona, Spain
| | - Alicia Gainza
- Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IdiSNA, C/Irunlarrea 3., 31008, Pamplona, Spain
| | - Joaquín Fernández-Irigoyen
- Clinical Neuroproteomics Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain
| | - Enrique Santamaría
- Clinical Neuroproteomics Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain
| | | | - Virginia Álvarez
- Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IdiSNA, C/Irunlarrea 3., 31008, Pamplona, Spain
| | - Rafael Sádaba
- Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IdiSNA, C/Irunlarrea 3., 31008, Pamplona, Spain
| | - Eva Jover
- Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IdiSNA, C/Irunlarrea 3., 31008, Pamplona, Spain.
| | - Natalia López-Andrés
- Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IdiSNA, C/Irunlarrea 3., 31008, Pamplona, Spain.
| |
Collapse
|
23
|
Anyfanti P, Dimitriadou A, Dara A, Angeloudi E, Gavriilaki E, Nikolaidou B, Triantafyllou A, Dimitroulas T, Gkaliagkousi E. Circulating levels of galectin-3 and coronary microvascular perfusion in rheumatoid arthritis patients with suppressed inflammation. Clin Rheumatol 2023; 42:2881-2887. [PMID: 37418035 DOI: 10.1007/s10067-023-06685-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/18/2023] [Accepted: 06/24/2023] [Indexed: 07/08/2023]
Abstract
OBJECTIVE Cardiovascular manifestations are the leading cause of mortality in rheumatoid arthritis (RA). Galectin-3, a lectin protein with major role in cellular, inflammatory, and fibrotic processes, has been introduced as a novel cardiac biomarker. We hypothesized that patients with RA present increased levels of galectin-3, and investigated potential associations with arterial stiffness and coronary microvascular dysfunction. METHODS This cross-sectional study enrolled RA patients and non-RA individuals without cardiovascular comorbidities. Galectin-3 and high-sensitivity C-reactive protein (hsCRP) were measured with enzyme-linked immunosorbent assay (ELISA) in serum samples. Subendocardial viability ratio (SEVR), an index of microvascular myocardial perfusion, and pulse wave velocity (PWV), the gold-standard measure of vascular stiffness, were estimated with applanation tonometry. RESULTS Cardiovascular risk factors and hsCRP were comparable between patients (n = 24) and controls (n = 24). However, galectin-3 was increased [6.9 (6.7) vs 4.6 (4.7)] ng/dl, p = 0.015], and coronary microvascular perfusion was decreased (142.6 ± 22.8 vs 159.7 ± 23.2%, p = 0.028) in RA patients compared to controls, whereas PWV did not significantly differ. Galectin-3 correlated with both PWV and SEVR in univariate analysis. However, after adjustment for cardiovascular risk factors and subclinical inflammation, these associations were rendered non-significant. CONCLUSION Galectin-3 appears increased in RA, even among patients with suppressed inflammation in the absence of cardiovascular comorbidities. The observed association of galectin-3 with coronary microvascular perfusion in our study was non-significant after adjustment for cardiovascular risk factors and inflammation. The potential role of galectin-3 as a cardiac biomarker in RA warrants further investigation. Key Points • Galectin-3 has emerged as a novel cardiac biomarker but remains understudied in RA. • Patients with RA present elevated levels of galectin-3 and impaired coronary microvascular perfusion compared to non-RA individuals. • These differences were observed in patients with suppressed inflammation, even in the absence of CVD. • The association of galectin-3 with coronary microvascular impairment in RA warrants further investigation.
Collapse
Affiliation(s)
- Panagiota Anyfanti
- Second Medical Department, Hippokration General Hospital, Aristotle University of Thessaloniki, 49, Konstantinoupoleos Str, 54642, Thessaloniki, Greece.
| | - Antonia Dimitriadou
- 3rd Department of Internal Medicine, Papageorgiou Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Athanasia Dara
- Fourth Department of Internal Medicine, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Elena Angeloudi
- Second Medical Department, Hippokration General Hospital, Aristotle University of Thessaloniki, 49, Konstantinoupoleos Str, 54642, Thessaloniki, Greece
| | - Eleni Gavriilaki
- 2nd Propedeutic Department of Internal Medicine, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Barbara Nikolaidou
- 3rd Department of Internal Medicine, Papageorgiou Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Areti Triantafyllou
- 3rd Department of Internal Medicine, Papageorgiou Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Theodoros Dimitroulas
- Fourth Department of Internal Medicine, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Eugenia Gkaliagkousi
- 3rd Department of Internal Medicine, Papageorgiou Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
24
|
Liang T, Zhu Z, Gong F, Yang X, Lei X, Lu L. Galectin-3 promotes brain injury by modulating the phenotype of microglia via binding TLR-4 after intracerebral hemorrhage. Aging (Albany NY) 2023; 15:9041-9058. [PMID: 37698533 PMCID: PMC10522396 DOI: 10.18632/aging.205014] [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: 04/17/2023] [Accepted: 08/20/2023] [Indexed: 09/13/2023]
Abstract
BACKGROUND Intracerebral hemorrhage (ICH) is a stroke subtype with high mortality and disability rate, and neuroinflammation is involved in secondary brain injury. Galectin-3 (Gal-3) is one of the scaffold proteins of Galectins. Studies have indicated that Gal-3 plays an important role in the physiological and pathological state of the nervous system. Here we focus on the role of Gal-3 in ICH, especially in neuroinflammation. METHODS Injection of autologous blood into the right basal ganglia was used to simulate ICH injury, and the level of Gal-3 in brain was regulated by related means. The changes of Gal-3 were detected by western blot and immunofluorescence, the level of neuroinflammation by immunofluorescence staining and ELISA. Apoptosis and neuron loss were detected by TUNEL staining FJB staining and Nissl staining, and neurological deficits were judged by neurobehavioral tests. RESULTS The protein level of Gal-3 increased at 24 h after ICH. Downregulation of Gal-3 level can reduce the infiltration of M1-type microglia and peripheral inflammatory cells, thus alleviating post-ICH neuroinflammation, and reducing cell apoptosis and neuron loss in brain tissue. ICH-induced neurological damage was rescued. Meanwhile, the promotion in the expression level of Gal-3 increased neuroinflammatory activation and nerve cell death, aggravating ICH-induced brain injury. CONCLUSIONS This study proves that Gal-3 is involved in neuroinflammation and nerve damage after ICH. Gal-3 expression should not be encouraged early on to prevent neuroinflammation. which provides a new possibility for clinical treatment for ICH patients.
Collapse
Affiliation(s)
- Tianyu Liang
- Emergency and Critical Care Center, Intensive Care Unit, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zheng Zhu
- Emergency and Critical Care Center, Intensive Care Unit, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Fangxiao Gong
- Emergency and Critical Care Center, Intensive Care Unit, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xiaobo Yang
- Center for General Practice Medicine, Department of Nursing, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xiaoju Lei
- Center for General Practice Medicine, Department of Nursing, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Ling Lu
- Center for Rehabilitation Medicine, Department of Anesthesiology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
25
|
Affiliation(s)
- Iacopo Fabiani
- Cardiology and Cardiovascular Medicine Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Michela Chianca
- Health Science Interdisciplinary Center, Scuola Superiore Sant’Anna, Pisa, Italy
| | - Michele Emdin
- Cardiology and Cardiovascular Medicine Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
- Health Science Interdisciplinary Center, Scuola Superiore Sant’Anna, Pisa, Italy
| |
Collapse
|
26
|
Chumakova SP, Urazova OI, Shipulin VM, Andreev SL, Denisenko OA, Gladkovskaya MV, Litvinova LS, Bubenchikov MA. Role of Angiopoietic Coronary Endothelial Dysfunction in the Pathogenesis of Ischemic Cardiomyopathy. Biomedicines 2023; 11:1950. [PMID: 37509589 PMCID: PMC10377729 DOI: 10.3390/biomedicines11071950] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/05/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND The angiopoietic endothelial dysfunction in ischemic cardiomyopathy (ICMP) remains unexplored. AIM The identification of the imbalance of endothelial dysfunction mediators and the number of endothelial progenitor (EPC) and desquamated (EDC) cells in patients with coronary heart disease (CHD) with and without ICMP. METHODS A total of 87 patients (47 with ICMP and 40 without ICMP) were observed. The content of EPCs (CD14+CD34+VEGFR2+) in vein blood and EDCs (CD45-CD146+) in the blood from the coronary sinus and cubital vein was determined by flow cytometry. The contents of HIF-1α and HIF-2α in vein blood as well as that of ADMA and endothelin-1 in sinus plasma and angiopoietin-2, MMP-9 and galectin-3 in both samples were assessed using ELISA, and VEGF, PDGF, SDF-1 and MCP-1 contents using immunofluorescence. RESULTS ADMA and endothelin-1 levels in the sinus blood were comparable between the patient groups; a deficiency of HIF-1α and excess of HIF-2α were detected in the vein blood of ICMP patients. The EDC content in the vein blood increased in CHD patients regardless of ICMP, and the concentrations of VEGF-A, VEGF-B, PDGF, MCP-1, angiopoietin-2, and MMP-9 were normal. In ICMP patients, vein blood was characterized by an excess of galectin-3 and sinus blood by an excess of EDCs, angiopoietin-2, MMP-9 and galectin-3. CONCLUSION ICMP is accompanied by angiopoietic endothelial dysfunction.
Collapse
Affiliation(s)
- Svetlana P Chumakova
- Pathophysiology Division, Siberian State Medical University, Tomsk 634050, Russia
- Central Research Laboratory, Siberian State Medical University, Tomsk 634050, Russia
| | - Olga I Urazova
- Pathophysiology Division, Siberian State Medical University, Tomsk 634050, Russia
- Central Research Laboratory, Siberian State Medical University, Tomsk 634050, Russia
- Department of Complex Information Security of Computer Systems, Tomsk State University of Control Systems and Radioelectronics, Tomsk 634050, Russia
| | - Vladimir M Shipulin
- Cardiovascular Surgery Unit, Cardiology Research Institute, Tomsk National Medical Research Center, Russian Academy of Sciences, Tomsk 634050, Russia
| | - Sergey L Andreev
- Cardiovascular Surgery Unit, Cardiology Research Institute, Tomsk National Medical Research Center, Russian Academy of Sciences, Tomsk 634050, Russia
| | - Olga A Denisenko
- Pathophysiology Division, Siberian State Medical University, Tomsk 634050, Russia
| | | | - Larisa S Litvinova
- Immunology and Cell Biotechnology Center, Immanuel Kant Baltic Federal University, Kaliningrad 236041, Russia
| | - Mikhail A Bubenchikov
- Department of Theoretical Mechanics, National Research Tomsk State University, Tomsk 634050, Russia
| |
Collapse
|
27
|
Coulis G, Jaime D, Guerrero-Juarez C, Kastenschmidt JM, Farahat PK, Nguyen Q, Pervolarakis N, McLinden K, Thurlow L, Movahedi S, Hughes BS, Duarte J, Sorn A, Montoya E, Mozaffar I, Dragan M, Othy S, Joshi T, Hans CP, Kimonis V, MacLean AL, Nie Q, Wallace LM, Harper SQ, Mozaffar T, Hogarth MW, Bhattacharya S, Jaiswal JK, Golann DR, Su Q, Kessenbrock K, Stec M, Spencer MJ, Zamudio JR, Villalta SA. Single-cell and spatial transcriptomics identify a macrophage population associated with skeletal muscle fibrosis. SCIENCE ADVANCES 2023; 9:eadd9984. [PMID: 37418531 PMCID: PMC10328414 DOI: 10.1126/sciadv.add9984] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 06/05/2023] [Indexed: 07/09/2023]
Abstract
Macrophages are essential for skeletal muscle homeostasis, but how their dysregulation contributes to the development of fibrosis in muscle disease remains unclear. Here, we used single-cell transcriptomics to determine the molecular attributes of dystrophic and healthy muscle macrophages. We identified six clusters and unexpectedly found that none corresponded to traditional definitions of M1 or M2 macrophages. Rather, the predominant macrophage signature in dystrophic muscle was characterized by high expression of fibrotic factors, galectin-3 (gal-3) and osteopontin (Spp1). Spatial transcriptomics, computational inferences of intercellular communication, and in vitro assays indicated that macrophage-derived Spp1 regulates stromal progenitor differentiation. Gal-3+ macrophages were chronically activated in dystrophic muscle, and adoptive transfer assays showed that the gal-3+ phenotype was the dominant molecular program induced within the dystrophic milieu. Gal-3+ macrophages were also elevated in multiple human myopathies. These studies advance our understanding of macrophages in muscular dystrophy by defining their transcriptional programs and reveal Spp1 as a major regulator of macrophage and stromal progenitor interactions.
Collapse
Affiliation(s)
- Gerald Coulis
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
- Institute for Immunology, University of California Irvine, Irvine, CA, USA
| | - Diego Jaime
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
- Institute for Immunology, University of California Irvine, Irvine, CA, USA
| | | | - Jenna M. Kastenschmidt
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
- Institute for Immunology, University of California Irvine, Irvine, CA, USA
| | - Philip K. Farahat
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
- Institute for Immunology, University of California Irvine, Irvine, CA, USA
| | - Quy Nguyen
- Department of Biological Chemistry, University of California Irvine, Irvine, CA USA
| | | | - Katherine McLinden
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
| | - Lauren Thurlow
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
| | - Saba Movahedi
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
| | - Brandon S. Hughes
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
| | - Jorge Duarte
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
| | - Andrew Sorn
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
| | - Elizabeth Montoya
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
| | - Izza Mozaffar
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
| | - Morgan Dragan
- Department of Biological Chemistry, University of California Irvine, Irvine, CA USA
| | - Shivashankar Othy
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
- Institute for Immunology, University of California Irvine, Irvine, CA, USA
| | - Trupti Joshi
- Department of Health Management and Informatics, University of Missouri, Columbia, MO, USA
| | - Chetan P. Hans
- Department of Cardiovascular Medicine, University of Missouri, Columbia, MO USA
| | - Virginia Kimonis
- Department of Pediatrics, University of California Irvine, Irvine, CA, USA
| | - Adam L. MacLean
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA, USA
| | - Qing Nie
- Department of Mathematics, Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA
| | - Lindsay M. Wallace
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
| | - Scott Q. Harper
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University, Columbus, OH, USA
| | - Tahseen Mozaffar
- Department of Neurology, University of California Irvine, Irvine, CA, USA
- Department of Pathology and Laboratory Medicine, University of California Irvine, Irvine, CA, USA
| | - Marshall W. Hogarth
- Children’s National Hospital, Research Center for Genetic Medicine, Washington, DC, USA
| | - Surajit Bhattacharya
- Children’s National Hospital, Research Center for Genetic Medicine, Washington, DC, USA
| | - Jyoti K. Jaiswal
- Children’s National Hospital, Research Center for Genetic Medicine, Washington, DC, USA
| | | | - Qi Su
- Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA
| | - Kai Kessenbrock
- Department of Biological Chemistry, University of California Irvine, Irvine, CA USA
| | - Michael Stec
- Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA
| | - Melissa J. Spencer
- Department of Neurology, University of California Los Angeles, Los Angeles, CA, USA
| | - Jesse R. Zamudio
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
| | - S. Armando Villalta
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
- Institute for Immunology, University of California Irvine, Irvine, CA, USA
- Department of Neurology, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
28
|
Stack JP, Fries RC, Kruckman L, Kadotani S, Wallace G. Galectin-3 as a novel biomarker in cats with hypertrophic cardiomyopathy. J Vet Cardiol 2023; 48:54-62. [PMID: 37480722 DOI: 10.1016/j.jvc.2023.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 06/13/2023] [Accepted: 06/23/2023] [Indexed: 07/24/2023]
Abstract
INTRODUCTION/OBJECTIVES Galectin-3 (Gal-3) is a circulating biomarker of fibrosis. In humans, increased Gal-3 is predictive of myocardial fibrosis and adverse cardiac events. The aim of this study was to evaluate the potential for Gal-3 as a cardiac biomarker in cats with hypertrophic cardiomyopathy (HCM). MATERIALS AND METHODS Eighty cats were enrolled (25 healthy cats with normal hearts, 35 with HCM American College of Veterinary Internal Medicine (ACVIM) stage B, and 21 with HCM ACVIM stage C). Each cat received a full echocardiogram, health panel, and total thyroxin level. Galectin-3 levels were measured for each enrolled patient. Troponin I and N-terminal pro-brain natriuretic peptide (NT-proBNP) were obtained for the majority of cats. Additionally, 17 ACVIM stage B cats underwent cardiac-gated magnetic resonance (CMR) imaging to assess myocardial extracellular volume (ECV), a noninvasive measure of myocardial fibrosis. RESULTS Galectin-3 levels are increased in cats with HCM ACVIM stage B and C compared to healthy cats; however, no significant differences were detected between ACVIM stage B and ACVIM stage C cats. In HCM-affected cats, Galectin-3 showed statistically significant correlations with left atrial dimensions, left atrial:aorta ratio, and CMR-derived ECV. Quantitative NT-proBNP showed excellent discrimination between all groups and troponin I was able to discriminate between ACVIM stage C and normal cats, but not between other groups. CONCLUSIONS Circulating Gal-3 levels are increased in cats with HCM and is positively correlated with left atrial dimensions and ECV in affected cats. Further studies evaluating the relationship between Gal-3, myocardial fibrosis, and clinical outcomes are warranted.
Collapse
Affiliation(s)
- J P Stack
- VCA Loomis Basin Veterinary Clinic, Loomis, CA, USA
| | - R C Fries
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign College of Veterinary Medicine, Urbana, IL, USA.
| | - L Kruckman
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign College of Veterinary Medicine, Urbana, IL, USA
| | - S Kadotani
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign College of Veterinary Medicine, Urbana, IL, USA
| | - G Wallace
- Pacific Northwest Pet ER and Specialty Center, Vancouver, WA, USA
| |
Collapse
|
29
|
Calvier L, Drelich A, Hsu J, Tseng CT, Mina Y, Nath A, Kounnas MZ, Herz J. Circulating Reelin promotes inflammation and modulates disease activity in acute and long COVID-19 cases. Front Immunol 2023; 14:1185748. [PMID: 37441066 PMCID: PMC10333573 DOI: 10.3389/fimmu.2023.1185748] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 06/08/2023] [Indexed: 07/15/2023] Open
Abstract
Thromboembolic complications and excessive inflammation are frequent in severe COVID-19, potentially leading to long COVID. In non-COVID studies, we and others demonstrated that circulating Reelin promotes leukocyte infiltration and thrombosis. Thus, we hypothesized that Reelin participates in endothelial dysfunction and hyperinflammation during COVID-19. We showed that Reelin was increased in COVID-19 patients and correlated with the disease activity. In the severe COVID-19 group, we observed a hyperinflammatory state, as judged by increased concentration of cytokines (IL-1α, IL-4, IL-6, IL-10 and IL-17A), chemokines (IP-10 and MIP-1β), and adhesion markers (E-selectin and ICAM-1). Reelin level was correlated with IL-1α, IL-4, IP-10, MIP-1β, and ICAM-1, suggesting a specific role for Reelin in COVID-19 progression. Furthermore, Reelin and all of the inflammatory markers aforementioned returned to normal in a long COVID cohort, showing that the hyperinflammatory state was resolved. Finally, we tested Reelin inhibition with the anti-Reelin antibody CR-50 in hACE2 transgenic mice infected with SARS-CoV-2. CR-50 prophylactic treatment decreased mortality and disease severity in this model. These results demonstrate a direct proinflammatory function for Reelin in COVID-19 and identify it as a drug target. This work opens translational clinical applications in severe SARS-CoV-2 infection and beyond in auto-inflammatory diseases.
Collapse
Affiliation(s)
- Laurent Calvier
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas, TX, United States
- Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX, United States
| | - Aleksandra Drelich
- Department of Biochemistry, Cellular, and Molecular Biology, University of Texas Medical Branch (UTMB) Health, Galveston, TX, United States
| | - Jason Hsu
- Department of Biochemistry, Cellular, and Molecular Biology, University of Texas Medical Branch (UTMB) Health, Galveston, TX, United States
| | - Chien-Te Tseng
- Department of Biochemistry, Cellular, and Molecular Biology, University of Texas Medical Branch (UTMB) Health, Galveston, TX, United States
| | - Yair Mina
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Avindra Nath
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | | | - Joachim Herz
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas, TX, United States
- Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX, United States
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, United States
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
30
|
Ahmed R, Anam K, Ahmed H. Development of Galectin-3 Targeting Drugs for Therapeutic Applications in Various Diseases. Int J Mol Sci 2023; 24:8116. [PMID: 37175823 PMCID: PMC10179732 DOI: 10.3390/ijms24098116] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 04/24/2023] [Accepted: 04/29/2023] [Indexed: 05/15/2023] Open
Abstract
Galectin-3 (Gal3) is one of the most studied members of the galectin family that mediate various biological processes such as growth regulation, immune function, cancer metastasis, and apoptosis. Since Gal3 is pro-inflammatory, it is involved in many diseases that are associated with chronic inflammation such as cancer, organ fibrosis, and type 2 diabetes. As a multifunctional protein involved in multiple pathways of many diseases, Gal3 has generated significant interest in pharmaceutical industries. As a result, several Gal3-targeting therapeutic drugs are being developed to address unmet medical needs. Based on the PubMed search of Gal3 to date (1987-2023), here, we briefly describe its structure, carbohydrate-binding properties, endogenous ligands, and roles in various diseases. We also discuss its potential antagonists that are currently being investigated clinically or pre-clinically by the public and private companies. The updated knowledge on Gal3 function in various diseases could initiate new clinical or pre-clinical investigations to test therapeutic strategies, and some of these strategies could be successful and recognized as novel therapeutics for unmet medical needs.
Collapse
Affiliation(s)
| | | | - Hafiz Ahmed
- GlycoMantra Inc., Biotechnology Center, University of Maryland Baltimore County, Baltimore, MD 21250, USA
| |
Collapse
|
31
|
Coulis G, Jaime D, Guerrero-Juarez C, Kastenschmidt JM, Farahat PK, Nguyen Q, Pervolarakis N, McLinden K, Thurlow L, Movahedi S, Duarte J, Sorn A, Montoya E, Mozaffar I, Dragan M, Othy S, Joshi T, Hans CP, Kimonis V, MacLean AL, Nie Q, Wallace LM, Harper SQ, Mozaffar T, Hogarth MW, Bhattacharya S, Jaiswal JK, Golann DR, Su Q, Kessenbrock K, Stec M, Spencer MJ, Zamudio JR, Villalta SA. Single-cell and spatial transcriptomics identify a macrophage population associated with skeletal muscle fibrosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.18.537253. [PMID: 37131694 PMCID: PMC10153153 DOI: 10.1101/2023.04.18.537253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The monocytic/macrophage system is essential for skeletal muscle homeostasis, but its dysregulation contributes to the pathogenesis of muscle degenerative disorders. Despite our increasing knowledge of the role of macrophages in degenerative disease, it still remains unclear how macrophages contribute to muscle fibrosis. Here, we used single-cell transcriptomics to determine the molecular attributes of dystrophic and healthy muscle macrophages. We identified six novel clusters. Unexpectedly, none corresponded to traditional definitions of M1 or M2 macrophage activation. Rather, the predominant macrophage signature in dystrophic muscle was characterized by high expression of fibrotic factors, galectin-3 and spp1. Spatial transcriptomics and computational inferences of intercellular communication indicated that spp1 regulates stromal progenitor and macrophage interactions during muscular dystrophy. Galectin-3 + macrophages were chronically activated in dystrophic muscle and adoptive transfer assays showed that the galectin-3 + phenotype was the dominant molecular program induced within the dystrophic milieu. Histological examination of human muscle biopsies revealed that galectin-3 + macrophages were also elevated in multiple myopathies. These studies advance our understanding of macrophages in muscular dystrophy by defining the transcriptional programs induced in muscle macrophages, and reveal spp1 as a major regulator of macrophage and stromal progenitor interactions.
Collapse
Affiliation(s)
- Gerald Coulis
- Department of Physiology and Biophysics, University of California Irvine, USA
- Institute for Immunology, University of California Irvine, USA
| | - Diego Jaime
- Department of Physiology and Biophysics, University of California Irvine, USA
- Institute for Immunology, University of California Irvine, USA
| | - Christian Guerrero-Juarez
- Department of Mathematics, University of California Irvine, USA
- Department of Developmental and Cell Biology, University of California Irvine, USA
| | - Jenna M. Kastenschmidt
- Department of Physiology and Biophysics, University of California Irvine, USA
- Institute for Immunology, University of California Irvine, USA
| | - Philip K. Farahat
- Department of Physiology and Biophysics, University of California Irvine, USA
- Institute for Immunology, University of California Irvine, USA
| | - Quy Nguyen
- Department of Biological Chemistry, University of California Irvine, USA
| | | | - Katherine McLinden
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, USA
| | - Lauren Thurlow
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, USA
| | - Saba Movahedi
- Department of Physiology and Biophysics, University of California Irvine, USA
| | - Jorge Duarte
- Department of Physiology and Biophysics, University of California Irvine, USA
| | - Andrew Sorn
- Department of Physiology and Biophysics, University of California Irvine, USA
| | - Elizabeth Montoya
- Department of Physiology and Biophysics, University of California Irvine, USA
| | - Izza Mozaffar
- Department of Physiology and Biophysics, University of California Irvine, USA
| | - Morgan Dragan
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, USA
| | - Shivashankar Othy
- Department of Physiology and Biophysics, University of California Irvine, USA
- Institute for Immunology, University of California Irvine, USA
| | - Trupti Joshi
- Department of Health Management and Informatics, University of Missouri, Columbia, USA
| | - Chetan P. Hans
- Department of Cardiovascular Medicine, University of Missouri, Columbia, USA
| | | | - Adam L. MacLean
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, USA
| | - Qing Nie
- Department of Mathematics, University of California Irvine, USA
- Department of Developmental and Cell Biology, University of California Irvine, USA
| | - Lindsay M. Wallace
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital
| | - Scott Q. Harper
- Department of Pediatrics, The Ohio State University, Columbus, OH, USA
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital
| | - Tahseen Mozaffar
- Department of Neurology, University of California Irvine, USA
- Department of Pathology and Laboratory Medicine, University of California Irvine, USA
| | - Marshall W. Hogarth
- Children’s National Hospital, Research Center for Genetic Medicine, Washington, DC, USA
| | - Surajit Bhattacharya
- Children’s National Hospital, Research Center for Genetic Medicine, Washington, DC, USA
| | - Jyoti K. Jaiswal
- Children’s National Hospital, Research Center for Genetic Medicine, Washington, DC, USA
| | | | - Qi Su
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | - Kai Kessenbrock
- Department of Biological Chemistry, University of California Irvine, USA
| | - Michael Stec
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | | | - Jesse R. Zamudio
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, USA
| | - S. Armando Villalta
- Department of Physiology and Biophysics, University of California Irvine, USA
- Institute for Immunology, University of California Irvine, USA
- Department of Neurology, University of California Irvine, USA
| |
Collapse
|
32
|
Parksook WW, Williams GH. Aldosterone and cardiovascular diseases. Cardiovasc Res 2023; 119:28-44. [PMID: 35388416 DOI: 10.1093/cvr/cvac027] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 12/07/2021] [Accepted: 12/28/2021] [Indexed: 11/12/2022] Open
Abstract
Aldosterone's role in the kidney and its pathophysiologic actions in hypertension are well known. However, its role or that of its receptor [minieralocorticoid receptor (MR)] in other cardiovascular (CV) disease are less well described. To identify their potential roles in six CV conditions (heart failure, myocardial infarction, atrial fibrillation, stroke, atherosclerosis, and thrombosis), we assessed these associations in the following four areas: (i) mechanistic studies in rodents and humans; (ii) pre-clinical studies of MR antagonists; (iii) clinical trials of MR antagonists; and (iv) genetics. The data were acquired from an online search of the National Library of Medicine using the PubMed search engine from January 2011 through June 2021. There were 3702 publications identified with 200 publications meeting our inclusion and exclusion criteria. Data strongly supported an association between heart failure and dysregulated aldosterone/MR. This association is not surprising given aldosterone/MR's prominent role in regulating sodium/volume homeostasis. Atrial fibrillation and myocardial infarction are also associated with dysregulated aldosterone/MR, but less strongly. For the most part, the data were insufficient to determine whether there was a relationship between atherosclerosis, stroke, or thrombosis and aldosterone/MR dysregulation. This review clearly documented an expanding role for aldosterone/MR's dysregulation in CV diseases beyond hypertension. How expansive it might be is limited by the currently available data. It is anticipated that with an increased focus on aldosterone/MR's potential roles in these diseases, additional clinical and pre-clinical data will clarify these relationships, thereby, opening approaches to use modulators of aldosterone/MR's action to more precisely treat these CV conditions.
Collapse
Affiliation(s)
- Wasita W Parksook
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Division of Endocrinology and Metabolism, Faculty of Medicine, Chulalongkorn University, and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
- Division of General Internal Medicine, Faculty of Medicine, Chulalongkorn University, and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Gordon H Williams
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
33
|
Fan M, Lan X, Wang Q, Shan M, Fang X, Zhang Y, Wu D, Luo H, Gao W, Zhu D. Renal function protection and the mechanism of ginsenosides: Current progress and future perspectives. Front Pharmacol 2023; 14:1070738. [PMID: 36814491 PMCID: PMC9939702 DOI: 10.3389/fphar.2023.1070738] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 01/24/2023] [Indexed: 02/08/2023] Open
Abstract
Nephropathy is a general term for kidney diseases, which refers to changes in the structure and function of the kidney caused by various factors, resulting in pathological damage to the kidney, abnormal blood or urine components, and other diseases. The main manifestations of kidney disease include hematuria, albuminuria, edema, hypertension, anemia, lower back pain, oliguria, and other symptoms. Early detection, diagnosis, and active treatment are required to prevent chronic renal failure. The concept of nephropathy encompasses a wide range of conditions, including acute renal injury, chronic kidney disease, nephritis, renal fibrosis, and diabetic nephropathy. Some of these kidney-related diseases are interrelated and may lead to serious complications without effective control. In serious cases, it can also develop into chronic renal dysfunction and eventually end-stage renal disease. As a result, it seriously affects the quality of life of patients and places a great economic burden on society and families. Ginsenoside is one of the main active components of ginseng, with anti-inflammatory, anti-tumor, antioxidant, and other pharmacological activities. A variety of monomers in ginsenosides can play protective roles in multiple organs. According to the difference of core structure, ginsenosides can be divided into protopanaxadiol-type (including Rb1, Rb3, Rg3, Rh2, Rd and CK, etc.), and protopanaxatriol (protopanaxatriol)- type (including Rg1, Rg2 and Rh1, etc.), and other types (including Rg5, Rh4, Rh3, Rk1, and Rk3, etc.). All of these ginsenosides showed significant renal function protection, which can reduce renal damage in renal injury, nephritis, renal fibrosis, and diabetic nephropathy models. This review summarizes reports on renal function protection and the mechanisms of action of these ginsenosides in various renal injury models.
Collapse
Affiliation(s)
- Meiling Fan
- The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Xintian Lan
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China,Key Laboratory of Effective Components of Traditional Chinese Medicine, Changchun, China
| | - Qunling Wang
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China,Key Laboratory of Effective Components of Traditional Chinese Medicine, Changchun, China
| | - Mengyao Shan
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China,Key Laboratory of Effective Components of Traditional Chinese Medicine, Changchun, China
| | - Xiaoxue Fang
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China,Key Laboratory of Effective Components of Traditional Chinese Medicine, Changchun, China
| | - Yegang Zhang
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China,Key Laboratory of Effective Components of Traditional Chinese Medicine, Changchun, China
| | - Donglu Wu
- Key Laboratory of Effective Components of Traditional Chinese Medicine, Changchun, China,School of Clinical Medical, Changchun University of Chinese Medicine, Changchun, China
| | - Haoming Luo
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China,Key Laboratory of Effective Components of Traditional Chinese Medicine, Changchun, China
| | - Wenyi Gao
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China,*Correspondence: Wenyi Gao, ; Difu Zhu,
| | - Difu Zhu
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China,Key Laboratory of Effective Components of Traditional Chinese Medicine, Changchun, China,*Correspondence: Wenyi Gao, ; Difu Zhu,
| |
Collapse
|
34
|
Cardiovascular Disease in Obstructive Sleep Apnea: Putative Contributions of Mineralocorticoid Receptors. Int J Mol Sci 2023; 24:ijms24032245. [PMID: 36768567 PMCID: PMC9916750 DOI: 10.3390/ijms24032245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/12/2023] [Accepted: 01/20/2023] [Indexed: 01/24/2023] Open
Abstract
Obstructive sleep apnea (OSA) is a chronic and highly prevalent condition that is associated with oxidative stress, inflammation, and fibrosis, leading to endothelial dysfunction, arterial stiffness, and vascular insulin resistance, resulting in increased cardiovascular disease and overall mortality rates. To date, OSA remains vastly underdiagnosed and undertreated, with conventional treatments yielding relatively discouraging results for improving cardiovascular outcomes in OSA patients. As such, a better mechanistic understanding of OSA-associated cardiovascular disease (CVD) and the development of novel adjuvant therapeutic targets are critically needed. It is well-established that inappropriate mineralocorticoid receptor (MR) activation in cardiovascular tissues plays a causal role in a multitude of CVD states. Clinical studies and experimental models of OSA lead to increased secretion of the MR ligand aldosterone and excessive MR activation. Furthermore, MR activation has been associated with worsened OSA prognosis. Despite these documented relationships, there have been no studies exploring the causal involvement of MR signaling in OSA-associated CVD. Further, scarce clinical studies have exclusively assessed the beneficial role of MR antagonists for the treatment of systemic hypertension commonly associated with OSA. Here, we provide a comprehensive overview of overlapping mechanistic pathways recruited in the context of MR activation- and OSA-induced CVD and propose MR-targeted therapy as a potential avenue to abrogate the deleterious cardiovascular consequences of OSA.
Collapse
|
35
|
Pang ZD, Sun X, Bai RY, Han MZ, Zhang YJ, Wu W, Zhang Y, Lai BC, Zhang Y, Wang Y, Du XJ, Deng XL. YAP-galectin-3 signaling mediates endothelial dysfunction in angiotensin II-induced hypertension in mice. Cell Mol Life Sci 2023; 80:38. [PMID: 36629913 PMCID: PMC11072047 DOI: 10.1007/s00018-022-04623-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 10/29/2022] [Accepted: 11/03/2022] [Indexed: 01/12/2023]
Abstract
BACKGROUND Vascular endothelial dysfunction is regarded as an early event of hypertension. Galectin-3 (Gal-3) is known to participate in various pathological processes. Whilst previous studies showed that inhibition of Gal-3 effectively ameliorates angiotensin II (Ang II)-induced atherosclerosis or hypertension, it remains unclear whether Ang II regulates Gal-3 expression and actions in vascular endothelium. METHODS Using techniques of molecular biology and myograph, we investigated Ang II-mediated changes in Gal-3 expression and activity in thoracic aortas and mesenteric arteries from wild-type and Gal-3 gene deleted (Gal-3-/-) mice and cultured endothelial cells. RESULTS The serum level of Gal-3 was significantly higher in hypertensive patients or in mice with chronic Ang II-infusion. Ang II infusion to wild-type mice enhanced Gal-3 expression in the aortic and mesenteric arteries, elevated systolic blood pressure and impaired endothelium-dependent relaxation of the thoracic aortas and mesenteric arteries, changes that were abolished in Gal-3-/- mice. In human umbilical vein endothelial cells, Ang II significantly upregulated Gal-3 expression by promoting nuclear localization of Yes-associated protein (YAP) and its interaction with transcription factor Tead1 with enhanced YAP/Tead1 binding to Gal-3 gene promoter region. Furthermore, Gal-3 deletion augmented the bioavailability of nitric oxide, suppressed oxidative stress, and alleviated inflammation in the thoracic aorta of Ang II-infused mice or endothelial cells exposed to Ang II. CONCLUSIONS Our results demonstrate for the first time that Ang II upregulates Gal-3 expression via increment in YAP nuclear localization in vascular endothelium, and that Gal-3 mediates endothelial dysfunction contributing to the development of hypertension.
Collapse
Affiliation(s)
- Zheng-Da Pang
- Department of Physiology and Pathophysiology, Cardiovascular Research Centre, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an, 710061, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Xia Sun
- Department of Physiology and Pathophysiology, Cardiovascular Research Centre, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an, 710061, Shaanxi, China
- School of Basic and Medical Sciences, Xi'an Medical University, 1 Xinwang Road, Xi'an, 710021, Shaanxi, China
| | - Ru-Yue Bai
- Department of Physiology and Pathophysiology, Cardiovascular Research Centre, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Meng-Zhuan Han
- Department of Physiology and Pathophysiology, Cardiovascular Research Centre, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Yong-Jian Zhang
- Department of Physiology and Pathophysiology, Cardiovascular Research Centre, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an, 710061, Shaanxi, China
- Department of Cardiac Surgery, The First Affiliated Hospital, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Wei Wu
- Department of Physiology and Pathophysiology, Cardiovascular Research Centre, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Yu Zhang
- Department of Physiology and Pathophysiology, Cardiovascular Research Centre, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Bao-Chang Lai
- Department of Physiology and Pathophysiology, Cardiovascular Research Centre, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Yi Zhang
- Department of Physiology and Pathophysiology, Cardiovascular Research Centre, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Yan Wang
- Department of Physiology and Pathophysiology, Cardiovascular Research Centre, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Xiao-Jun Du
- Department of Physiology and Pathophysiology, Cardiovascular Research Centre, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an, 710061, Shaanxi, China.
| | - Xiu-Ling Deng
- Department of Physiology and Pathophysiology, Cardiovascular Research Centre, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an, 710061, Shaanxi, China.
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
36
|
Wang T, Ou L, Li X, Zhang P, Miao Q, Niu R, Chen Y. Inhibition of Galectin-3 attenuates silica particles-induced silicosis via regulating the GSK-3β/β-catenin signal pathway-mediated epithelial-mesenchymal transition. Chem Biol Interact 2022; 368:110218. [DOI: 10.1016/j.cbi.2022.110218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/28/2022] [Accepted: 10/06/2022] [Indexed: 11/28/2022]
|
37
|
Phosphorylation of CaMK and CREB-Mediated Cardiac Aldosterone Synthesis Induced by Arginine Vasopressin in Rats with Myocardial Infarction. Int J Mol Sci 2022; 23:ijms232315061. [PMID: 36499387 PMCID: PMC9738971 DOI: 10.3390/ijms232315061] [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: 11/01/2022] [Revised: 11/22/2022] [Accepted: 11/27/2022] [Indexed: 12/02/2022] Open
Abstract
Both aldosterone and arginine vasopressin (AVP) are produced in the heart and may participate in cardiac fibrosis. However, their relationship remains unknown. This study aims to demonstrate the regulation and role of AVP in aldosterone synthesis in the heart. Rats were subjected to a sham operation or myocardial infarction (MI) by ligating the coronary artery. Cardiac function and fibrosis were assessed using echocardiography and immunohistochemical staining, respectively. In addition, the effects of AVP stimulation on cardiac microvascular endothelial cells (CMECs) were studied using ELISA, real-time PCR, and Western blotting. Compared with the rats having undergone a sham operation, the MI rats had an increased LVMI, type I collagen composition, and concentrations of aldosterone and AVP in the heart but decreased cardiac function. As the MI rats aged, the LVMI, type I collagen, aldosterone, and AVP increased, while the LVMI decreased. Furthermore, AVP time-dependently induced aldosterone secretion and CYP11B2 mRNA expression in CMECs. The p-CREB levels were significantly increased by AVP. Nevertheless, these effects were completely blocked by SR49059 or partially inhibited by KN93. This study demonstrated that AVP could induce the secretion of local cardiac aldosterone, which may involve CaMK and CREB phosphorylation and CYP11B2 upregulation through V1 receptor activation.
Collapse
|
38
|
Mansour AA, Krautter F, Zhi Z, Iqbal AJ, Recio C. The interplay of galectins-1, -3, and -9 in the immune-inflammatory response underlying cardiovascular and metabolic disease. Cardiovasc Diabetol 2022; 21:253. [PMID: 36403025 PMCID: PMC9675972 DOI: 10.1186/s12933-022-01690-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 11/08/2022] [Indexed: 11/21/2022] Open
Abstract
Galectins are β-galactoside-binding proteins that bind and crosslink molecules via their sugar moieties, forming signaling and adhesion networks involved in cellular communication, differentiation, migration, and survival. Galectins are expressed ubiquitously across immune cells, and their function varies with their tissue-specific and subcellular location. Particularly galectin-1, -3, and -9 are highly expressed by inflammatory cells and are involved in the modulation of several innate and adaptive immune responses. Modulation in the expression of these proteins accompany major processes in cardiovascular diseases and metabolic disorders, such as atherosclerosis, thrombosis, obesity, and diabetes, making them attractive therapeutic targets. In this review we consider the broad cellular activities ascribed to galectin-1, -3, and -9, highlighting those linked to the progression of different inflammatory driven pathologies in the context of cardiovascular and metabolic disease, to better understand their mechanism of action and provide new insights into the design of novel therapeutic strategies.
Collapse
Affiliation(s)
- Adel Abo Mansour
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Franziska Krautter
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Zhaogong Zhi
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Asif Jilani Iqbal
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.
| | - Carlota Recio
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Farmacología Molecular y Traslacional -BIOPharm, Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Las Palmas, Spain.
| |
Collapse
|
39
|
Nangia-Makker P, Hogan V, Balan V, Raz A. Chimeric galectin-3 and collagens: Biomarkers and potential therapeutic targets in fibroproliferative diseases. J Biol Chem 2022; 298:102622. [PMID: 36272642 PMCID: PMC9706532 DOI: 10.1016/j.jbc.2022.102622] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 11/27/2022] Open
Abstract
Fibrosis, stiffening and scarring of an organ/tissue due to genetic abnormalities, environmental factors, infection, and/or injury, is responsible for > 40% of all deaths in the industrialized world, and to date, there is no cure for it despite extensive research and numerous clinical trials. Several biomarkers have been identified, but no effective therapeutic targets are available. Human galectin-3 is a chimeric gene product formed by the fusion of the internal domain of the collagen alpha gene [N-terminal domain (ND)] at the 5'-end of galectin-1 [C-terminal domain (CRD)] that appeared during evolution together with vertebrates. Due to the overlapping structural similarities between collagen and galectin-3 and their shared susceptibility to cleavage by matrix metalloproteases to generate circulating collagen-like peptides, this review will discuss present knowledge on the role of collagen and galectin-3 as biomarkers of fibrosis. We will also highlight the need for transformative approaches targeting both the ND and CRD domains of galectin-3, since glycoconjugate binding by the CRD is triggered by ND-mediated oligomerization and the therapies targeted only at the CRD have so far achieved limited success.
Collapse
Affiliation(s)
- Pratima Nangia-Makker
- Barbara Ann Karmanos Cancer Institute, Department of Oncology, School of Medicine, Redwood City, California, USA,For correspondence: Pratima Nangia-Makker; Avraham Raz
| | - Victor Hogan
- Barbara Ann Karmanos Cancer Institute, Department of Oncology, School of Medicine, Redwood City, California, USA
| | - Vitaly Balan
- Guardant Health, Bioinformatics, Redwood City, California, USA
| | - Avraham Raz
- Barbara Ann Karmanos Cancer Institute, Department of Oncology, School of Medicine, Redwood City, California, USA,Department of Pathology, School of Medicine, Wayne State University, Detroit, Michigan, USA,For correspondence: Pratima Nangia-Makker; Avraham Raz
| |
Collapse
|
40
|
Maupin KA, Diegel CR, Stevens PD, Dick D, Williams BO. Mutation of the galectin-3 glycan-binding domain (Lgals3-R200S) enhances cortical bone expansion in male mice and trabecular bone mass in female mice. FEBS Open Bio 2022; 12:1717-1728. [PMID: 36062328 PMCID: PMC9527582 DOI: 10.1002/2211-5463.13483] [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: 05/04/2022] [Revised: 07/14/2022] [Accepted: 08/02/2022] [Indexed: 12/14/2022] Open
Abstract
We previously observed that genomic loss of galectin-3 (Gal-3; encoded by Lgals3) in mice has a significant protective effect on age-related bone loss. Gal-3 has both intracellular and extracellular functionality, and we wanted to assess whether the affect we observed in the Lgals3 knockout (KO) mice could be attributed to the ability of Gal-3 to bind glycoproteins. Mutation of a highly conserved arginine to a serine in human Gal-3 (LGALS3-R186S) blocks glycan binding and secretion. We generated mice with the equivalent mutation (Lgals3-R200S) and observed a subsequent reduction in Gal-3 secretion from mouse embryonic fibroblasts and in circulating blood. When examining bone structure in aged mice, we noticed some similarities to the Lgals3-KO mice and some differences. First, we observed greater bone mass in Lgals3-R200S mutant mice, as was previously observed in Lgals3-KO mice. Like Lgals3-KO mice, significantly increased trabecular bone mass was only observed in female Lgals3-R200S mice. These results suggest that the greater bone mass observed is driven by the loss of extracellular Gal-3 functionality. However, the results from our cortical bone expansion data showed a sex-dependent difference, with only male Lgals3-KO mice having an increased response, contrasting with our earlier study. These notable sex differences suggest a potential role for sex hormones, most likely androgen signaling, being involved. In summary, our results suggest that targeting extracellular Gal-3 function may be a suitable treatment for age-related loss of bone mass.
Collapse
|
41
|
Abstract
Besides the physiological regulation of water, sodium, and potassium homeostasis, aldosterone modulates several physiological and pathological processes in the cardiovascular system. At the vascular level, aldosterone excess stimulates endothelial dysfunction and infiltration of inflammatory cells, enhances the development of the atherosclerotic plaque, and favors plaque instability, arterial stiffness, and calcification. At the cardiac level, aldosterone increases cardiac inflammation, fibrosis, and myocardial hypertrophy. As a clinical consequence, high aldosterone levels are associated with enhanced risk of cardiovascular events and mortality, especially when aldosterone secretion is inappropriate for renin levels and sodium intake, as in primary aldosteronism. Several clinical trials showed that mineralocorticoid receptor antagonists reduce cardiovascular mortality in patients with heart failure and reduced ejection fraction, but inconclusive results were reported for other cardiovascular conditions, such as heart failure with preserved ejection fraction, myocardial infarction, and atrial fibrillation. In patients with primary aldosteronism, adrenalectomy or treatment with mineralocorticoid receptor antagonists significantly mitigate adverse aldosterone effects, reducing the risk of cardiovascular events, mortality, and incident atrial fibrillation. In this review, we will summarize the major preclinical and clinical studies investigating the cardiovascular damage mediated by aldosterone and the protective effect of mineralocorticoid receptor antagonists for the reduction of cardiovascular risk in patients with cardiovascular diseases and primary aldosteronism.
Collapse
Affiliation(s)
- Fabrizio Buffolo
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences, University of Torino, Italy
| | - Martina Tetti
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences, University of Torino, Italy
| | - Paolo Mulatero
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences, University of Torino, Italy
| | - Silvia Monticone
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences, University of Torino, Italy
| |
Collapse
|
42
|
Owen A, Patel JM, Parekh D, Bangash MN. Mechanisms of Post-critical Illness Cardiovascular Disease. Front Cardiovasc Med 2022; 9:854421. [PMID: 35911546 PMCID: PMC9334745 DOI: 10.3389/fcvm.2022.854421] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 06/22/2022] [Indexed: 11/13/2022] Open
Abstract
Prolonged critical care stays commonly follow trauma, severe burn injury, sepsis, ARDS, and complications of major surgery. Although patients leave critical care following homeostatic recovery, significant additional diseases affect these patients during and beyond the convalescent phase. New cardiovascular and renal disease is commonly seen and roughly one third of all deaths in the year following discharge from critical care may come from this cluster of diseases. During prolonged critical care stays, the immunometabolic, inflammatory and neurohumoral response to severe illness in conjunction with resuscitative treatments primes the immune system and parenchymal tissues to develop a long-lived pro-inflammatory and immunosenescent state. This state is perpetuated by persistent Toll-like receptor signaling, free radical mediated isolevuglandin protein adduct formation and presentation by antigen presenting cells, abnormal circulating HDL and LDL isoforms, redox and metabolite mediated epigenetic reprogramming of the innate immune arm (trained immunity), and the development of immunosenescence through T-cell exhaustion/anergy through epigenetic modification of the T-cell genome. Under this state, tissue remodeling in the vascular, cardiac, and renal parenchymal beds occurs through the activation of pro-fibrotic cellular signaling pathways, causing vascular dysfunction and atherosclerosis, adverse cardiac remodeling and dysfunction, and proteinuria and accelerated chronic kidney disease.
Collapse
Affiliation(s)
- Andrew Owen
- Department of Critical Care, Queen Elizabeth Hospital, University Hospitals Birmingham, Birmingham, United Kingdom
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Jaimin M. Patel
- Department of Critical Care, Queen Elizabeth Hospital, University Hospitals Birmingham, Birmingham, United Kingdom
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Dhruv Parekh
- Department of Critical Care, Queen Elizabeth Hospital, University Hospitals Birmingham, Birmingham, United Kingdom
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Mansoor N. Bangash
- Department of Critical Care, Queen Elizabeth Hospital, University Hospitals Birmingham, Birmingham, United Kingdom
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
- *Correspondence: Mansoor N. Bangash
| |
Collapse
|
43
|
Calvier L, Kökény G, Martinez-Martinez E. Editorial: New Advances in Cardiorenal Syndrome. Front Cardiovasc Med 2022; 9:976846. [PMID: 35911551 PMCID: PMC9331892 DOI: 10.3389/fcvm.2022.976846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 06/27/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Laurent Calvier
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, TX, United States
- *Correspondence: Laurent Calvier
| | - Gábor Kökény
- Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
- International Nephrology Research and Training Center, Semmelweis University, Budapest, Hungary
| | - Ernesto Martinez-Martinez
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense de Madrid, Madrid, Spain
- Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
44
|
Bayes-Genis A, Cediel G, Domingo M, Codina P, Santiago E, Lupón J. Biomarkers in Heart Failure with Preserved Ejection Fraction. Card Fail Rev 2022; 8:e20. [PMID: 35815256 PMCID: PMC9253965 DOI: 10.15420/cfr.2021.37] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/02/2022] [Indexed: 12/23/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a heterogeneous disorder developing from multiple aetiologies with overlapping pathophysiological mechanisms. HFpEF diagnosis may be challenging, as neither cardiac imaging nor physical examination are sensitive in this situation. Here, we review biomarkers of HFpEF, of which the best supported are related to myocardial stretch and injury, including natriuretic peptides and cardiac troponins. An overview of biomarkers of inflammation, extracellular matrix derangements and fibrosis, senescence, vascular dysfunction, anaemia/iron deficiency and obesity is also provided. Finally, novel biomarkers from -omics technologies, including plasma metabolites and circulating microRNAs, are outlined briefly. A cardiac-centred approach to HFpEF diagnosis using natriuretic peptides seems reasonable at present in clinical practice. A holistic approach including biomarkers that provide information on the non-cardiac components of the HFpEF syndrome may enrich our understanding of the disease and may be useful in classifying HFpEF phenotypes or endotypes that may guide patient selection in HFpEF trials.
Collapse
Affiliation(s)
- Antoni Bayes-Genis
- Heart Institute, University Hospital Germans Trias i Pujol, Badalona, Spain; Department of Medicine, Universitat Autonoma de Barcelona, Barcelona, Spain; CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Germán Cediel
- Heart Institute, University Hospital Germans Trias i Pujol, Badalona, Spain; CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Mar Domingo
- Heart Institute, University Hospital Germans Trias i Pujol, Badalona, Spain; CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Pau Codina
- Heart Institute, University Hospital Germans Trias i Pujol, Badalona, Spain; CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Evelyn Santiago
- Heart Institute, University Hospital Germans Trias i Pujol, Badalona, Spain; CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Josep Lupón
- Heart Institute, University Hospital Germans Trias i Pujol, Badalona, Spain; CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
45
|
Di Fusco SA, Cianfrocca C, Bisceglia I, Spinelli A, Alonzo A, Mocini E, Gulizia MM, Gabrielli D, Oliva F, Imperoli G, Colivicchi F. Potential pathophysiologic mechanisms underlying the inherent risk of cancer in patients with atherosclerotic cardiovascular disease. Int J Cardiol 2022; 363:190-195. [PMID: 35724799 DOI: 10.1016/j.ijcard.2022.06.048] [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: 01/06/2022] [Revised: 05/22/2022] [Accepted: 06/15/2022] [Indexed: 11/25/2022]
Abstract
Emerging evidence demonstrates an intimate interplay between cardiovascular disease and cancer pathophysiology. The aim of this review is to shed light on the common biological pathways underlying cardiovascular disease and cancer. These common pathways form the basis of "reverse cardio-oncology". We focus on the role of inflammation, stress response, cell proliferation, angiogenesis and tissue remodeling, neurohormonal system activation, and genomic instability as pathogenic pathways shared by cardiovascular disease and cancer. We also discuss shared mediators that may have a potential role as biomarkers for risk prediction in both diseases. Furthermore, we highlight current knowledge on biological pathways and mediators that are upregulated in diabetes and myocardial infarction and may be involved in tumorigenesis. On the basis of the shared pathophysiologic mechanisms, we also suggest an integrated approach to reduce the global burden of both cardiovascular disease and cancer.
Collapse
Affiliation(s)
| | - Cinzia Cianfrocca
- Clinical and Rehabilitation Cardiology Unit, P.O. San Filippo Neri, ASL Roma 1, Rome, Italy
| | - Irma Bisceglia
- Integrated Cardiology Services, Cardio-Thoracic-Vascular Department, San Camillo Hospital, Rome, Italy
| | - Antonella Spinelli
- Clinical and Rehabilitation Cardiology Unit, P.O. San Filippo Neri, ASL Roma 1, Rome, Italy
| | - Alessandro Alonzo
- Clinical and Rehabilitation Cardiology Unit, P.O. San Filippo Neri, ASL Roma 1, Rome, Italy
| | - Edoardo Mocini
- Department of Experimental Medicine, Sapienza University, Rome
| | - Michele Massimo Gulizia
- Cardiology Division, Ospedale Garibaldi-Nesima, Azienda di Rilievo Nazionale e Alta Specializzazione "Garibaldi" Catania, Italy; Fondazione per il Tuo Cuore, Heart Care Foundation, Florence, Italy
| | | | - Fabrizio Oliva
- De Gasperis Cardio Center, Niguarda Hospital, Milano, Italy
| | - Giuseppe Imperoli
- Medicine Unit, Emergency Department, P.O San Filippo Neri, ASL Roma 1, Rome, Italy
| | - Furio Colivicchi
- Clinical and Rehabilitation Cardiology Unit, P.O. San Filippo Neri, ASL Roma 1, Rome, Italy
| |
Collapse
|
46
|
The Role of Circulating Collagen Turnover Biomarkers and Late Gadolinium Enhancement in Patients with Non-Ischemic Dilated Cardiomyopathy. Diagnostics (Basel) 2022; 12:diagnostics12061435. [PMID: 35741245 PMCID: PMC9222171 DOI: 10.3390/diagnostics12061435] [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: 05/08/2022] [Revised: 06/08/2022] [Accepted: 06/08/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Myocardial scarring is a primary pathogenetic process in nonischemic dilated cardiomyopathy (NIDCM) that is responsible for progressive cardiac remodeling and heart failure, severely impacting the survival of these patients. Although several collagen turnover biomarkers have been associated with myocardial fibrosis, their clinical utility is still limited. Late gadolinium enhancement (LGE) determined by cardiac magnetic resonance imaging (CMR) has become a feasible method to detect myocardial replacement fibrosis. We sought to evaluate the association between collagen turnover biomarkers and replacement myocardial scarring by CMR and, also, to test their ability to predict outcome in conjunction with LGE in patients with NIDCM. Method: We conducted a prospective study on 194 patients (48.7 ± 14.3 years of age; 74% male gender) with NIDCM. The inclusion criteria were similar to those for the definition of NIDCM, performed exclusively by CMR: (1) LV dilation with an LV end-diastolic volume (LVEDV) of over 97 mL/m2; (2) global LV dysfunction, expressed as a decreased LVEF of under 45%. CMR was used to determine the presence and extent of LGE. Several collagen turnover biomarkers were determined at diagnosis, comprising galectin-3 (Gal3), procollagen type I carboxy-terminal pro-peptide (PICP) and N-terminal pro-peptide of procollagen type III (PIIINP). A composite outcome (all-cause mortality, ventricular tachyarrhythmias, heart failure hospitalization) was ascertained over a median of 26 months. Results: Gal3, PICP and PIIINP were considerably increased in those with LGE+ (p < 0.001), also being directly correlated with LGE mass (r2 = 0.42; r2 = 0.44; r2 = 0.31; all p < 0.001). Receiver operating characteristic (ROC) analysis revealed a significant ability to diagnose LGE, with an area under the ROC of 0.816 for Gal3, 0.705 for PICP, and 0.757 for PIIINP (all p < 0.0001). Kaplan−Meier analysis showed that at a threshold of >13.8 ng/dL for Gal3 and >97 ng/dL for PICP, they were able to significantly predict outcome (HR = 2.66, p < 0.001; HR = 1.93, p < 0.002). Of all patients, 17% (n = 33) reached the outcome. In multivariate analysis, after adjustment for covariates, only LGE+ and Gal3+ remained independent predictors for outcome (p = 0.008; p = 0.04). Nonetheless, collagen turnover biomarkers were closely related to HF severity, providing incremental predictive value for severely decreased LVEF of under 30% in patients with NIDCM, beyond that with LGE alone. Conclusions: In patients with NIDCM, circulating collagen turnover biomarkers such as Gal3, PICP and PIIINP are closely related to the presence and extent of LGE and can significantly predict cardiovascular outcome. The joint use of LGE with Gal3 and PICP significantly improved outcome prediction.
Collapse
|
47
|
Aksan G, Yanık A, Yontar OC, Boyacı F, Uçar M, Şahin MK, Soylu K. The predictive value of galectin-3 levels on left atrial low voltage areas assessed by high-density mapping in patients with paroxysmal atrial fibrillation. J Arrhythm 2022; 38:353-362. [PMID: 35785368 PMCID: PMC9237302 DOI: 10.1002/joa3.12703] [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: 09/09/2021] [Revised: 02/20/2022] [Accepted: 03/13/2022] [Indexed: 11/12/2022] Open
Abstract
Aims Galectin-3 is an inflammation biomarker that is associated with atrial fibrosis and plays a role in the development of atrial fibrillation (AF). Low voltage areas (LVAs) identified using an electroanatomical mapping system represent the presence of fibrotic tissue. The present study aimed to determine the relationship between coronary sinus (CS) serum sampling of galectin-3 levels and the presence and extent of LVA in patients with paroxysmal AF. Methods A total of 115 consecutive paroxysmal AF patients underwent pulmonary vein isolation (PVI) included prospectively in the study. Voltage mapping was performed before PVI during sinus rhythm guided by multipolar high-density mapping catheter and LVAs were defined as regions where bipolar peak to peak voltage was <0.5 mV. Galectin-3 levels were measured via enzyme-linked immunosorbent assay. Results CS serum sampling of galectin-3 levels was significantly higher in paroxysmal AF patients with LVA than those without LVA (16.5 ± 3.7 ng/ml vs. 10.2 ±2.7 ng/ml, respectively, p < .001). CS serum sampling of galectin-3 levels was significantly higher in paroxysmal AF patients with moderate and severe LVA than in paroxysmal AF patients with mild LVA (17 ± 3.5 ng/ml and 20.1 ± 1.3 ng/ml vs. 13.3 ± 2.3 ng/ml, respectively, p = .002). In the multivariate analysis female gender (odds ratio [OR] = 7.537, 95% confidence interval [CI]: 1.011-56.195; p = .049), left atrium volume (OR = 1.326, 95% CI: 1.052-1.67; p = .017), and CS serum sampling of galectin-3 levels (OR = 1.704, 95% CI: 1.169-2.483; p = .006) were significant and independent predictors for LVAs. Conclusion In this study, we found that the CS serum sampling of galectin-3 levels increased with the extent of LVA and was an independent predictor for the presence of LVA.
Collapse
Affiliation(s)
- Gökhan Aksan
- Department of CardiologySamsun Education and Research HospitalSamsunTurkey
| | - Ahmet Yanık
- Department of CardiologySamsun Education and Research HospitalSamsunTurkey
| | - Osman Can Yontar
- Department of CardiologySamsun Education and Research HospitalSamsunTurkey
| | - Faruk Boyacı
- Department of CardiologySamsun Education and Research HospitalSamsunTurkey
| | - Melisa Uçar
- Department of CardiologySamsun Education and Research HospitalSamsunTurkey
| | - Mustafa Kürşat Şahin
- Department of Family MedicineFaculty of Medicine Ondokuz Mayıs UniversitySamsunTurkey
| | - Korhan Soylu
- Department of Cardiology, Faculty of MedicineOndokuz Mayis UniversitySamsunTurkey
| |
Collapse
|
48
|
Eklund M, Hellberg O, Furuland H, Cao Y, Wall K, Nilsson E. Effect of spironolactone on vascular stiffness in hemodialysis patients: a randomized crossover trial. Ups J Med Sci 2022; 127:8594. [PMID: 35722182 PMCID: PMC9169545 DOI: 10.48101/ujms.v127.8594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/29/2022] [Accepted: 03/30/2022] [Indexed: 12/02/2022] Open
Abstract
Background The role of spironolactone treatment in hemodialysis patients is debated, but a survival benefit is suggested. Mineralocorticoids and chronic kidney disease have been linked to cardiovascular fibrosis. Therefore, we hypothesized that spironolactone would affect vascular stiffness, cardiac systolic, and diastolic function in hemodialysis patients. Methods This was a randomized crossover study in hemodialysis patients supplemented with an echocardiographic case series. All outcomes reported here were secondary in the trial and were assessed without blinding. Block randomization and allocation determined treatment order. Participants received 50 mg spironolactone daily for 12 weeks and untreated observation for another 12 weeks. Pulse wave velocity (PWV) was measured before and after treatment and observation. Doppler-echocardiography was conducted before and after treatment. Systemic arterial compliance indexed to body surface area (SACi), left ventricular ejection fraction (LVEF), the peak early diastolic mitral inflow velocity (E), the peak late diastolic mitral inflow velocity (A), and the peak early diastolic myocardial lengthening velocity (E') were measured. E/A and E/E' were then calculated. Statistical analyses were conducted per protocol. A generalized linear mixed model with random participant effects was used for PWV. The Wilcoxon signed-rank test was used for echocardiographic variables. Results Thirty participants were recruited, 18 completed follow-up, and 17 were included in PWV-analyses. Spironolactone treatment showed a tendency toward an increase in PWV of 1.34 (95% confidence interval: -0.11 to 2.78) m/s, which was not statistically significant (P = 0.07). There were no significant changes in any of the other variables (LVEF, E/A, E/E', or SACi). Conclusions We found no evidence supporting an effect of 12-week administration of spironolactone 50 mg daily on vascular stiffness, cardiac systolic, or diastolic function in hemodialysis patients.
Collapse
Affiliation(s)
- Michael Eklund
- Department of Internal Medicine, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Olof Hellberg
- Department of Internal Medicine, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Hans Furuland
- Department of Medical Sciences, Uppsala University Hospital, Uppsala, Sweden
| | - Yang Cao
- Clinical Epidemiology and Biostatistics, School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Kent Wall
- Department of Clinical Physiology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Erik Nilsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| |
Collapse
|
49
|
Circulating Galectin-3 and Aldosterone for Predicting Atrial Fibrillation Recurrence after Radiofrequency Catheter Ablation. Cardiovasc Ther 2022; 2022:6993904. [PMID: 35692374 PMCID: PMC9151002 DOI: 10.1155/2022/6993904] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/22/2022] [Accepted: 05/06/2022] [Indexed: 12/29/2022] Open
Abstract
Background Circulating galectin-3 (Gal-3) and aldosterone (ALD) are involved in fibrosis and inflammation. However, their potential value as predictors of atrial fibrillation (AF) recurrence after radiofrequency catheter ablation (RFCA) is unknown or controversial. Therefore, the aim of this study was to assess the relationship between baseline Gal-3, ALD levels, and AF recurrence in patients performing RFCA. Methods 153 consecutive patients undergoing RFCA were included. Gal-3 and ALD were measured at baseline. Univariate and multivariate Cox regressions were performed to determine the predictors of AF recurrence. Receiver operating characteristic (ROC) curve and Kaplan-Meier (K-M) curve were used to assess the value of predictors. Results There were 35 (22.88%) cases of AF recurrence after RFCA. The recurrence group had significantly higher preoperative serum levels of Gal-3 and ALD than the nonrecurrence group. Univariate and multivariate analysis showed that Gal-3 (HR = 1.28, 95% CI: 1.04-1.56, p = 0.02) and ALD (OR = 1.02, 95% CI: 1.00-1.03, p < 0.03) were significantly associated with AF recurrence after RFCA. The area under the curve (AUC) of preoperative serum Gal-3, ALD, and 2 combined to predict the recurrence of AF patients after RFCA was 0.636, 0.798, and 0.893, respectively, while sensitivity was 65.32%, 71.69%, and 88.61%, respectively and specificity was 77.46%, 78.53%, and 86.0%, respectively. Patients with Gal-3 above the cutoff value of 14.57 pg/ml had higher frequent AF recurrence than the patients with Gal − 3 ≤ 14.57 pg/ml (35% vs. 12%, p < 0.001) during a follow-up. Meanwhile, patients with ALD above the cutoff value of 243.61 pg/ml also had a higher AF recurrence rate than those with ALD ≤ 243.61 pg/ml (37% vs. 11%, p < 0.001) during a follow-up. The recurrence rate in patients with Gal − 3 > 14.57 pg/ml + ALD > 243.61 pg/ml was higher than that in patients with baseline Gal − 3 > 14.57 pg/ml or ALD > 243.61 pg/ml and patients with Gal − 3 ≤ 14.57 pg/ml + ALD ≤ 243.61 pg/ml (57% vs. 14% vs. 9%, p < 0.01, respectively). Conclusion AF recurrence after RFCA had higher baseline Gal-3 and ALD levels, and higher preoperative circulating Gal-3 and ALD levels were independent predictors of AF recurrence for patients undergoing RFCA, while combination of preoperative Gal-3 and ALD levels has higher prediction accuracy.
Collapse
|
50
|
Bioletto F, Bollati M, Lopez C, Arata S, Procopio M, Ponzetto F, Ghigo E, Maccario M, Parasiliti-Caprino M. Primary Aldosteronism and Resistant Hypertension: A Pathophysiological Insight. Int J Mol Sci 2022; 23:ijms23094803. [PMID: 35563192 PMCID: PMC9100181 DOI: 10.3390/ijms23094803] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/19/2022] [Accepted: 04/25/2022] [Indexed: 12/10/2022] Open
Abstract
Primary aldosteronism (PA) is a pathological condition characterized by an excessive aldosterone secretion; once thought to be rare, PA is now recognized as the most common cause of secondary hypertension. Its prevalence increases with the severity of hypertension, reaching up to 29.1% in patients with resistant hypertension (RH). Both PA and RH are "high-risk phenotypes", associated with increased cardiovascular morbidity and mortality compared to non-PA and non-RH patients. Aldosterone excess, as occurs in PA, can contribute to the development of a RH phenotype through several mechanisms. First, inappropriate aldosterone levels with respect to the hydro-electrolytic status of the individual can cause salt retention and volume expansion by inducing sodium and water reabsorption in the kidney. Moreover, a growing body of evidence has highlighted the detrimental consequences of "non-classical" effects of aldosterone in several target tissues. Aldosterone-induced vascular remodeling, sympathetic overactivity, insulin resistance, and adipose tissue dysfunction can further contribute to the worsening of arterial hypertension and to the development of drug-resistance. In addition, the pro-oxidative, pro-fibrotic, and pro-inflammatory effects of aldosterone may aggravate end-organ damage, thereby perpetuating a vicious cycle that eventually leads to a more severe hypertensive phenotype. Finally, neither the pathophysiological mechanisms mediating aldosterone-driven blood pressure rise, nor those mediating aldosterone-driven end-organ damage, are specifically blocked by standard first-line anti-hypertensive drugs, which might further account for the drug-resistant phenotype that frequently characterizes PA patients.
Collapse
|