1
|
Gu S, Kopecky BJ, Peña B, Vagnozzi RJ, Lahm T. Sex-dependent Pathophysiology and Therapeutic Considerations in Right Heart Disease. Can J Cardiol 2025:S0828-282X(25)00178-3. [PMID: 40054579 DOI: 10.1016/j.cjca.2025.02.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/12/2025] [Accepted: 02/28/2025] [Indexed: 03/28/2025] Open
Abstract
Right ventricular (RV) adaptation to the increased afterload in the setting of pulmonary hypertension (PH) and other cardiac and pulmonary vascular conditions is a major determinant of survival. Although the RV remains understudied and less well understood than the left ventricle, recent advances have been made in understanding the function and biology of the RV in health and in disease, particularly in PH. RV adaptation in PH exhibits significant sexual dimorphisms in pathophysiology, adaptation, and outcomes. Despite a higher incidence of PH, women consistently demonstrate better RV adaptation and survival rates in the setting of increased RV afterload compared with men. Sexual dimorphisms extend to therapy responsiveness, with women benefiting more from certain pulmonary vasodilators and exhibiting superior RV recovery. In this review we discuss the current literature on sexual dimorphisms in RV structure, function, and molecular pathways in health and disease, as well as in RV-specific clinical manifestations, treatments, and outcomes in PH. Sex steroid-mediated effects as well as emerging studies on sex steroid-independent effects are reviewed. In general, sex steroids such as 17β-estradiol and dehydroepiandrosterone exert RV-protective effects. In contrast, testosterone negatively impacts RV structure and function. Emerging evidence highlights the influence of nonhormonal genetic determinants, such as BMPR1A and DMRT2 loci, which are associated with better RV function in women. A better understanding of the interplay between sex hormones, genetic factors, and RV biology is crucial for advancing and developing RV-directed therapies for patients of either sex.
Collapse
Affiliation(s)
- Sue Gu
- Cardio Vascular Pulmonary Research Laboratory, University of Colorado School of Medicine, Aurora, Colorado, USA; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.
| | - Benjamin J Kopecky
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Gates Institute, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Brisa Peña
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Department of Bioengineering, College of Engineering, Design and Computing, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; CU-Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Ronald J Vagnozzi
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Gates Institute, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Tim Lahm
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado, USA; Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, Colorado, USA.
| |
Collapse
|
2
|
Svedlund Eriksson E, Lantero Rodriguez M, Halvorsen B, Johansson I, Mårtensson AKF, Wilhelmson AS, Huse C, Ueland T, Aukrust P, Broch K, Gullestad L, Amundsen BH, Andersen GØ, Karlsson MCI, Hagberg Thulin M, Camponeschi A, Trompet D, Hammarsten O, Redfors B, Borén J, Omerovic E, Levin MC, Chagin AS, Dahl TB, Tivesten Å. Testosterone exacerbates neutrophilia and cardiac injury in myocardial infarction via actions in bone marrow. Nat Commun 2025; 16:1142. [PMID: 39910039 PMCID: PMC11799197 DOI: 10.1038/s41467-025-56217-x] [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: 03/14/2024] [Accepted: 01/13/2025] [Indexed: 02/07/2025] Open
Abstract
Men develop larger infarct sizes than women after a myocardial infarction (MI), but the mechanism underlying this sex difference is unknown. Here, we demonstrated that blood neutrophil counts post-MI were higher in male than female mice. Castration-induced testosterone deficiency reduced blood neutrophil counts to the level in females and increased survival post-MI. These effects were mimicked by Osterix-directed ablation of the androgen receptor in bone marrow (BM). Mechanistically, androgens downregulated the leukocyte retention factor CXCL12 in BM stromal cells. Post-hoc analysis of clinical trial data showed that neutrophilia was greater in men than women after reperfusion of first-time ST-elevation MI, and tocilizumab, an interleukin-6 receptor inhibitor, reduced blood neutrophil counts and infarct size to a greater extent in men than women. Our work reveals a previously unknown mechanism connecting testosterone with neutrophilia and MI injury via BM and identifies the importance of considering sex when developing anti-inflammatory strategies to treat MI.
Collapse
Affiliation(s)
- Elin Svedlund Eriksson
- Wallenberg Laboratory for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Marta Lantero Rodriguez
- Wallenberg Laboratory for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Inger Johansson
- Wallenberg Laboratory for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Anna K F Mårtensson
- Wallenberg Laboratory for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Anna S Wilhelmson
- Wallenberg Laboratory for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- The Finsen Laboratory, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Biotech Research and Innovation Centre (BRIC), Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Camilla Huse
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Thrombosis Research Center (TREC), Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Kaspar Broch
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway
- K. G. Jebsen Cardiac Research Centre and Centre for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Lars Gullestad
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway
- K. G. Jebsen Cardiac Research Centre and Centre for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Brage Høyem Amundsen
- Clinic of Cardiology, St. Olav's Hospital, Trondheim University Hospital, Trondheim, Norway
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | | | - Mikael C I Karlsson
- Department of Microbiology, Tumor, and Cell Biology, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Malin Hagberg Thulin
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Alessandro Camponeschi
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Dana Trompet
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Ola Hammarsten
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Björn Redfors
- Wallenberg Laboratory for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Jan Borén
- Wallenberg Laboratory for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Elmir Omerovic
- Wallenberg Laboratory for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Malin C Levin
- Wallenberg Laboratory for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Andrei S Chagin
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Tuva B Dahl
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Åsa Tivesten
- Wallenberg Laboratory for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
- Department of Endocrinology, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden.
| |
Collapse
|
3
|
Niu RZ, Xu HY, Tian H, Zhang D, He CY, Li XL, Li YY, He J. Single-cell transcriptome unveils unique transcriptomic signatures of human organ-specific endothelial cells. Basic Res Cardiol 2024; 119:973-999. [PMID: 39508863 DOI: 10.1007/s00395-024-01087-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/23/2024] [Accepted: 10/08/2024] [Indexed: 11/15/2024]
Abstract
The heterogeneity of endothelial cells (ECs) across human tissues remains incompletely inventoried. We constructed an atlas of > 210,000 ECs derived from 38 regions across 24 human tissues. Our analysis reveals significant differences in transcriptome, phenotype, metabolism and transcriptional regulation among ECs from various tissues. Notably, arterial, venous, and lymphatic ECs shared more common markers in multiple tissues than capillary ECs, which exhibited higher heterogeneity. This diversity in capillary ECs suggests their greater potential as targets for drug development. ECs from different tissues and vascular beds were found to be associated with specific diseases. Importantly, tissue specificity of EC senescence is more determined by somatic site than by tissue type (e.g. subcutaneus adipose tissue and visceral adipose tissue). Additionally, sex-specific differences in brain EC senescence were observed. Our EC atlas offers valuble resoursce for identifying EC subclusters in single-cell datasets from body tissues or organoids, facilitating the screen of tissue-specific targeted therapies, and serving as a powerful tool for future discoveries.
Collapse
Affiliation(s)
- Rui-Ze Niu
- Department of Dermatology and Venereology, The Second Affiliated Hospital of Kunming Medical University, No.374 Dianmian Road, Wuhua District, Kunming, Yunnan, China
- Mental Health Centre of Kunming Medical University, Kunming, Yunnan, China
| | - Hong-Yan Xu
- Department of Dermatology and Venereology, The Second Affiliated Hospital of Kunming Medical University, No.374 Dianmian Road, Wuhua District, Kunming, Yunnan, China
| | - Hui Tian
- Department of Radiation Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Dan Zhang
- Department of Dermatology and Venereology, The First Affiliated Hospital of Kunming Medical University, No.295 Xichang Road, Kunming, Yunnan, China
| | - Chun-Yu He
- Institute of Medicine and Nursing, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xiao-Lan Li
- Department of Dermatology and Venereology, The Second Affiliated Hospital of Kunming Medical University, No.374 Dianmian Road, Wuhua District, Kunming, Yunnan, China.
| | - Yu-Ye Li
- Department of Dermatology and Venereology, The First Affiliated Hospital of Kunming Medical University, No.295 Xichang Road, Kunming, Yunnan, China.
| | - Juan He
- Department of Dermatology and Venereology, The Second Affiliated Hospital of Kunming Medical University, No.374 Dianmian Road, Wuhua District, Kunming, Yunnan, China.
| |
Collapse
|
4
|
Huang R, Zhang C, Xiang Z, Lin T, Ling J, Hu H. Role of mitochondria in renal ischemia-reperfusion injury. FEBS J 2024; 291:5365-5378. [PMID: 38567754 DOI: 10.1111/febs.17130] [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] [Received: 10/01/2023] [Revised: 01/30/2024] [Accepted: 03/22/2024] [Indexed: 12/19/2024]
Abstract
Acute kidney injury (AKI) induced by renal ischemia-reperfusion injury (IRI) has a high morbidity and mortality, representing a worldwide problem. The kidney is an essential organ of metabolism that has high blood perfusion and is the second most mitochondria-rich organ after the heart because of the high ATP demands of its essential functions of nutrient reabsorption, acid-base and electrolyte balance, and hemodynamics. Thus, these energy-intensive cells are particularly vulnerable to mitochondrial dysfunction. As the bulk of glomerular ultrafiltrate reabsorption by proximal tubules occurs via active transport, the mitochondria of proximal tubules must be equipped for detecting and responding to fluctuations in energy availability to guarantee efficient basal metabolism. Any insults to mitochondrial quality control mechanisms may lead to biological disruption, blocking the clearance of damaged mitochondria and resulting in morphological change and tissue dysfunction. Extensive research has shown that mitochondria have pivotal roles in acute kidney disease, so in this article, we discuss the role of mitochondria, their dynamics and mitophagy in renal ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Ruizhen Huang
- Department of Urology, The Second Affiliated Hospital of Nanchang University, China
| | - Chiyu Zhang
- Department of Urology, The Second Affiliated Hospital of Nanchang University, China
| | - Zhengjie Xiang
- Department of Urology, The Second Affiliated Hospital of Nanchang University, China
| | - Tao Lin
- Department of Urology, The Second Affiliated Hospital of Nanchang University, China
| | - Jian Ling
- Department of Urology, The Second Affiliated Hospital of Nanchang University, China
| | - Honglin Hu
- Department of Urology, The Second Affiliated Hospital of Nanchang University, China
| |
Collapse
|
5
|
Lin Z, Jiwani Z, Serpooshan V, Aghaverdi H, Yang PC, Aguirre A, Wu JC, Mahmoudi M. Sex Influences the Safety and Therapeutic Efficacy of Cardiac Nanomedicine Technologies. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2305940. [PMID: 37803920 PMCID: PMC10997742 DOI: 10.1002/smll.202305940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/18/2023] [Indexed: 10/08/2023]
Abstract
Nanomedicine technologies are being developed for the prevention, diagnosis, and treatment of cardiovascular disease (CVD), which is the leading cause of death worldwide. Before delving into the nuances of cardiac nanomedicine, it is essential to comprehend the fundamental sex-specific differences in cardiovascular health. Traditionally, CVDs have been more prevalent in males, but it is increasingly evident that females also face significant risks, albeit with distinct characteristics. Females tend to develop CVDs at a later age, exhibit different clinical symptoms, and often experience worse outcomes compared to males. These differences indicate the need for sex-specific approaches in cardiac nanomedicine. This Perspective discusses the importance of considering sex in the safety and therapeutic efficacy of nanomedicine approaches for the prevention, diagnosis, and treatment of CVD.
Collapse
Affiliation(s)
- Zijin Lin
- Department of Radiology and Precision Health Program, Michigan State University, East Lansing, MI 48824 USA
| | - Zahra Jiwani
- Department of Radiology and Precision Health Program, Michigan State University, East Lansing, MI 48824 USA
| | - Vahid Serpooshan
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Haniyeh Aghaverdi
- Department of Radiology and Precision Health Program, Michigan State University, East Lansing, MI 48824 USA
| | - Phillip C Yang
- Department of Medicine, Cardiovascular Medicine and Cardiovascular Institute, Stanford University, Stanford, CA 94309
| | - Aitor Aguirre
- Regenerative Biology and cell Reprogramming Laboratory, Institute for Quantitative Health Sciences and Engineering, Michigan State University, East Lansing, MI 48823, USA
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48823, USA
| | - Joseph C. Wu
- Department of Medicine, Cardiovascular Medicine and Cardiovascular Institute, Stanford University, Stanford, CA 94309
- Department of Medicine, Division of Cardiology, Stanford University, Stanford, CA 94305, USA
| | - Morteza Mahmoudi
- Department of Radiology and Precision Health Program, Michigan State University, East Lansing, MI 48824 USA
- Connors Center for Women’s Health & Gender Biology, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA 02115 USA
| |
Collapse
|
6
|
Lukovic D, Gyöngyösi M, Pavo IJ, Mester-Tonczar J, Einzinger P, Zlabinger K, Kastner N, Spannbauer A, Traxler D, Pavo N, Goliasch G, Pils D, Jakab A, Szankai Z, Michel-Behnke I, Zhang L, Devaux Y, Graf S, Beitzke D, Winkler J. Increased [ 18F]FDG uptake in the infarcted myocardial area displayed by combined PET/CMR correlates with snRNA-seq-detected inflammatory cell invasion. Basic Res Cardiol 2024; 119:807-829. [PMID: 38922408 PMCID: PMC11461641 DOI: 10.1007/s00395-024-01064-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 06/17/2024] [Accepted: 06/17/2024] [Indexed: 06/27/2024]
Abstract
Combined [18F]FDG PET-cardiac MRI imaging (PET/CMR) is a useful tool to assess myocardial viability and cardiac function in patients with acute myocardial infarction (AMI). Here, we evaluated the prognostic value of PET/CMR in a porcine closed-chest reperfused AMI (rAMI) model. Late gadolinium enhancement by PET/CMR imaging displayed tracer uptake defect at the infarction site by 3 days after the rAMI in the majority of the animals (group Match, n = 28). Increased [18F]FDG uptake at the infarcted area (metabolism/contractility mismatch) with reduced tracer uptake in the remote viable myocardium (group Mismatch, n = 12) 3 days after rAMI was observed in the animals with larger infarct size and worse left ventricular ejection fraction (LVEF) (34 ± 8.7 vs 42.0 ± 5.2%), with lower LVEF also at the 1-month follow-up (35.8 ± 9.5 vs 43.0 ± 6.3%). Transcriptome analyses by bulk and single-nuclei RNA sequencing of the infarcted myocardium and border zones (n = 3 of each group, and 3 sham-operated controls) revealed a strong inflammatory response with infiltration of monocytes and macrophages in the infarcted and border areas in Mismatch animals. Our data indicate a high prognostic relevance of combined PET/MRI in the subacute phase of rAMI for subsequent impairment of heart function and underline the adverse effects of an excessive activation of the innate immune system in the initial phase after rAMI.
Collapse
Affiliation(s)
- Dominika Lukovic
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Mariann Gyöngyösi
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria.
| | - Imre J Pavo
- Division of Pediatric Cardiology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Julia Mester-Tonczar
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Patrick Einzinger
- Institute of Information Systems Engineering, Research Unit of Information and Software Engineering, Vienna University of Technology, 1040, Vienna, Austria
| | - Katrin Zlabinger
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Nina Kastner
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Andreas Spannbauer
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Denise Traxler
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Noemi Pavo
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Georg Goliasch
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Dietmar Pils
- Division of General Surgery, Department of Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - Andras Jakab
- Center for MR-Research, University Children's Hospital Zurich, Zurich, Switzerland
| | | | - Ina Michel-Behnke
- Division of Pediatric Cardiology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Lu Zhang
- Cardiovascular Research Unit, Department of Population Health, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Yvan Devaux
- Cardiovascular Research Unit, Department of Population Health, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Senta Graf
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Dietrich Beitzke
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Johannes Winkler
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
7
|
Mouton AJ, Aitken NM, Morato JG, O'Quinn KR, do Carmo JM, da Silva AA, Omoto ACM, Li X, Wang Z, Schrimpe-Rutledge AC, Codreanu SG, Sherrod SD, McLean JA, Stanford JK, Brown JA, Hall JE. Glutamine metabolism improves left ventricular function but not macrophage-mediated inflammation following myocardial infarction. Am J Physiol Cell Physiol 2024; 327:C571-C586. [PMID: 38981605 PMCID: PMC11427008 DOI: 10.1152/ajpcell.00272.2024] [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: 04/25/2024] [Revised: 06/07/2024] [Accepted: 06/21/2024] [Indexed: 07/11/2024]
Abstract
Glutamine is a critical amino acid that serves as an energy source, building block, and signaling molecule for the heart tissue and the immune system. However, the role of glutamine metabolism in regulating cardiac remodeling following myocardial infarction (MI) is unknown. In this study, we show in adult male mice that glutamine metabolism is altered both in the remote (contractile) area and in infiltrating macrophages in the infarct area after permanent left anterior descending artery occlusion. We found that metabolites related to glutamine metabolism were differentially altered in macrophages at days 1, 3, and 7 after MI using untargeted metabolomics. Glutamine metabolism in live cells was increased after MI relative to no MI controls. Gene expression in the remote area of the heart indicated a loss of glutamine metabolism. Glutamine administration improved left ventricle (LV) function at days 1, 3, and 7 after MI, which was associated with improved contractile and metabolic gene expression. Conversely, administration of BPTES, a pharmacological inhibitor of glutaminase-1, worsened LV function after MI. Neither glutamine nor BPTES administration impacted gene expression or bioenergetics of macrophages isolated from the infarct area. Our results indicate that glutamine metabolism plays a critical role in maintaining LV contractile function following MI and that glutamine administration improves LV function. Glutamine metabolism may also play a role in regulating macrophage function, but macrophages are not responsive to exogenous pharmacological manipulation of glutamine metabolism.NEW & NOTEWORTHY Glutamine metabolism is altered in both infarct macrophages and the remote left ventricle (LV) following myocardial infarction (MI). Supplemental glutamine improves LV function following MI while inhibiting glutamine metabolism with BPTES worsens LV function. Supplemental glutamine or BPTES does not impact macrophage immunometabolic phenotypes after MI.
Collapse
Affiliation(s)
- Alan J Mouton
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, United States
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Nikaela M Aitken
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Jemylle G Morato
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Katherine R O'Quinn
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Jussara M do Carmo
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, United States
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Alexandre A da Silva
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, United States
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Ana C M Omoto
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, United States
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Xuan Li
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, United States
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Zhen Wang
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, United States
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Alexandra C Schrimpe-Rutledge
- Department of Chemistry and Center for Innovative Technology, Vanderbilt University, Nashville, Tennessee, United States
| | - Simona G Codreanu
- Department of Chemistry and Center for Innovative Technology, Vanderbilt University, Nashville, Tennessee, United States
| | - Stacy D Sherrod
- Department of Chemistry and Center for Innovative Technology, Vanderbilt University, Nashville, Tennessee, United States
| | - John A McLean
- Department of Chemistry and Center for Innovative Technology, Vanderbilt University, Nashville, Tennessee, United States
| | - Joshua K Stanford
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Jordan A Brown
- Jackson State University, Jackson, Mississippi, United States
| | - John E Hall
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, United States
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi, United States
| |
Collapse
|
8
|
Pilling D, Consalvo KM, Kirolos SA, Gomer RH. Differences between human male and female neutrophils in mRNA, translation efficiency, protein, and phosphoprotein profiles. RESEARCH SQUARE 2024:rs.3.rs-4284171. [PMID: 38746380 PMCID: PMC11092807 DOI: 10.21203/rs.3.rs-4284171/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Background Human males and females show differences in the incidence of neutrophil-associated diseases such as systemic lupus erythematosus, rheumatoid arthritis, and reactive arthritis, and differences in neutrophil physiological responses such as a faster response to the chemorepellent SLIGKV. Little is known about the basis of sex-based differences in human neutrophils. Methods Starting with human neutrophils from healthy donors, we used RNA-seq to examine total mRNA profiles, mRNAs not associated with ribosomes and thus not being translated, mRNAs in monosomes, and mRNAs in polysomes and thus heavily translated. We used mass spectrometry systems to identify proteins and phosphoproteins. Results There were sex-based differences in the translation of 24 mRNAs. There were 132 proteins with higher levels in male neutrophils; these tended to be associated with RNA regulation, ribosome, and phosphoinositide signaling pathways, whereas 30 proteins with higher levels in female neutrophils were associated with metabolic processes, proteosomes, and phosphatase regulatory proteins. Male neutrophils had increased phosphorylation of 32 proteins. After exposure to SLIGKV, male neutrophils showed a faster response in terms of protein phosphorylation compared to female neutrophils. Conclusions Male neutrophils have higher levels of proteins and higher phosphorylation of proteins associated with RNA processing and signaling pathways, while female neutrophils have higher levels of proteins associated with metabolism and proteolytic pathways. This suggests that male neutrophils might be more ready to adapt to a new environment, and female neutrophils might be more effective at responding to pathogens. This may contribute to the observed sex-based differences in neutrophil behavior and neutrophil-associated disease incidence and severity.
Collapse
Affiliation(s)
- Darrell Pilling
- Department of Biology, Texas A&M University, College Station, TX 77843-3474 USA
| | - Kristen M Consalvo
- Department of Biology, Texas A&M University, College Station, TX 77843-3474 USA
| | - Sara A Kirolos
- Department of Biology, Texas A&M University, College Station, TX 77843-3474 USA
| | - Richard H Gomer
- Department of Biology, Texas A&M University, College Station, TX 77843-3474 USA
| |
Collapse
|
9
|
Antipenko S, Mayfield N, Jinno M, Gunzer M, Ismahil MA, Hamid T, Prabhu SD, Rokosh G. Neutrophils are indispensable for adverse cardiac remodeling in heart failure. J Mol Cell Cardiol 2024; 189:1-11. [PMID: 38387309 PMCID: PMC10997476 DOI: 10.1016/j.yjmcc.2024.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 01/18/2024] [Accepted: 02/13/2024] [Indexed: 02/24/2024]
Abstract
Persistent immune activation contributes significantly to left ventricular (LV) dysfunction and adverse remodeling in heart failure (HF). In contrast to their well-known essential role in acute myocardial infarction (MI) as first responders that clear dead cells and facilitate subsequent reparative macrophage polarization, the role of neutrophils in the pathobiology of chronic ischemic HF is poorly defined. To determine the importance of neutrophils in the progression of ischemic cardiomyopathy, we measured their production, levels, and activation in a mouse model of chronic HF 8 weeks after permanent coronary artery ligation and large MI. In HF mice, neutrophils were more abundant both locally in failing myocardium (more in the border zone) and systemically in the blood, spleen, and bone marrow, together with increased BM granulopoiesis. There were heightened stimuli for neutrophil recruitment and trafficking in HF, with increased myocardial expression of the neutrophil chemoattract chemokines CXCL1 and CXCL5, and increased neutrophil chemotactic factors in the circulation. HF neutrophil NETotic activity was increased in vitro with coordinate increases in circulating neutrophil extracellular traps (NETs) in vivo. Neutrophil depletion with either antibody-based or genetic approaches abrogated the progression of LV remodeling and fibrosis at both intermediate and late stages of HF. Moreover, analogous to murine HF, the plasma milieu in human acute decompensated HF strongly promoted neutrophil trafficking. Collectively, these results support a key tissue-injurious role for neutrophils and their associated cytotoxic products in ischemic cardiomyopathy and suggest that neutrophils are potential targets for therapeutic immunomodulation in this disease.
Collapse
Affiliation(s)
- Sergey Antipenko
- Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nicolas Mayfield
- Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Miki Jinno
- Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Matthias Gunzer
- Institute for Experimental Immunology and Imaging, University Hospital, University of Duisburg-Essen, Essen, Germany; Leibniz-Institute fur Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany
| | - Mohamed Ameen Ismahil
- Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA; Division of Cardiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tariq Hamid
- Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA; Division of Cardiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Sumanth D Prabhu
- Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA; Division of Cardiology, Washington University School of Medicine, St. Louis, MO, USA.
| | - Gregg Rokosh
- Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA; Division of Cardiology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
10
|
Rahunen R, Tulppo M, Rinne V, Lepojärvi S, Perkiömäki JS, Huikuri HV, Ukkola O, Junttila J, Hukkanen J. Liver X Receptor Agonist 4β-Hydroxycholesterol as a Prognostic Factor in Coronary Artery Disease. J Am Heart Assoc 2024; 13:e031824. [PMID: 38390795 PMCID: PMC10944077 DOI: 10.1161/jaha.123.031824] [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: 09/15/2023] [Accepted: 01/05/2024] [Indexed: 02/24/2024]
Abstract
BACKGROUND Regardless of progress in treatment of coronary artery disease (CAD), there is still a significant residual risk of death in patients with CAD, highlighting the need for additional risk stratification markers. Our previous study provided evidence for a novel blood pressure-regulating mechanism involving 4β-hydroxycholesterol (4βHC), an agonist for liver X receptors, as a hypotensive factor. The aim was to determine the role of 4βHC as a prognostic factor in CAD. METHODS AND RESULTS The ARTEMIS (Innovation to Reduce Cardiovascular Complications of Diabetes at the Intersection) cohort consists of 1946 patients with CAD. Men and women were analyzed separately in quartiles according to plasma 4βHC. Basic characteristics, medications, ECG, and echocardiography parameters as well as mortality rate were analyzed. At baseline, subjects with a beneficial cardiovascular profile, as assessed with traditional markers such as body mass index, exercise capacity, prevalence of diabetes, and use of antihypertensives, had the highest plasma 4βHC concentrations. However, in men, high plasma 4βHC was associated with all-cause death, cardiac death, and especially sudden cardiac death (SCD) in a median follow-up of 8.8 years. Univariate and comprehensively adjusted hazard ratios for SCD in the highest quartile were 3.76 (95% CI, 1.6-8.7; P=0.002) and 4.18 (95% CI, 1.5-11.4; P=0.005), respectively. In contrast, the association of cardiac death and SCD in women showed the lowest risk in the highest 4βHC quartile. CONCLUSIONS High plasma 4βHC concentration was associated with death and especially SCD in men, while an inverse association was detected in women. Our results suggest 4βHC as a novel sex-specific risk marker of cardiac death and especially SCD in chronic CAD. REGISTRATION INFORMATION clinicaltrials.gov. Identifier NCT01426685.
Collapse
Affiliation(s)
- Roosa Rahunen
- Research Unit of Biomedicine and Internal MedicineUniversity of OuluOuluFinland
- Biocenter OuluUniversity of OuluOuluFinland
- Medical Research Center OuluOulu University Hospital and University of OuluOuluFinland
| | - Mikko Tulppo
- Research Unit of Biomedicine and Internal MedicineUniversity of OuluOuluFinland
- Medical Research Center OuluOulu University Hospital and University of OuluOuluFinland
| | | | - Samuli Lepojärvi
- Research Unit of Biomedicine and Internal MedicineUniversity of OuluOuluFinland
- Medical Research Center OuluOulu University Hospital and University of OuluOuluFinland
| | - Juha S. Perkiömäki
- Research Unit of Biomedicine and Internal MedicineUniversity of OuluOuluFinland
- Medical Research Center OuluOulu University Hospital and University of OuluOuluFinland
| | - Heikki V. Huikuri
- Research Unit of Biomedicine and Internal MedicineUniversity of OuluOuluFinland
- Medical Research Center OuluOulu University Hospital and University of OuluOuluFinland
| | - Olavi Ukkola
- Research Unit of Biomedicine and Internal MedicineUniversity of OuluOuluFinland
- Medical Research Center OuluOulu University Hospital and University of OuluOuluFinland
| | - Juhani Junttila
- Research Unit of Biomedicine and Internal MedicineUniversity of OuluOuluFinland
- Biocenter OuluUniversity of OuluOuluFinland
- Medical Research Center OuluOulu University Hospital and University of OuluOuluFinland
| | - Janne Hukkanen
- Research Unit of Biomedicine and Internal MedicineUniversity of OuluOuluFinland
- Biocenter OuluUniversity of OuluOuluFinland
- Medical Research Center OuluOulu University Hospital and University of OuluOuluFinland
| |
Collapse
|
11
|
Hegemann N, Barth L, Döring Y, Voigt N, Grune J. Implications for neutrophils in cardiac arrhythmias. Am J Physiol Heart Circ Physiol 2024; 326:H441-H458. [PMID: 38099844 PMCID: PMC11219058 DOI: 10.1152/ajpheart.00590.2023] [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: 09/22/2023] [Revised: 12/08/2023] [Accepted: 12/13/2023] [Indexed: 02/03/2024]
Abstract
Cardiac arrhythmias commonly occur as a result of aberrant electrical impulse formation or conduction in the myocardium. Frequently discussed triggers include underlying heart diseases such as myocardial ischemia, electrolyte imbalances, or genetic anomalies of ion channels involved in the tightly regulated cardiac action potential. Recently, the role of innate immune cells in the onset of arrhythmic events has been highlighted in numerous studies, correlating leukocyte expansion in the myocardium to increased arrhythmic burden. Here, we aim to call attention to the role of neutrophils in the pathogenesis of cardiac arrhythmias and their expansion during myocardial ischemia and infectious disease manifestation. In addition, we will elucidate molecular mechanisms associated with neutrophil activation and discuss their involvement as direct mediators of arrhythmogenicity.
Collapse
Affiliation(s)
- Niklas Hegemann
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - Lukas Barth
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - Yannic Döring
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg August University Göttingen, Göttingen, Germany
- German Centre for Cardiovascular Research (DZHK), Göttingen, Germany
| | - Niels Voigt
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg August University Göttingen, Göttingen, Germany
- German Centre for Cardiovascular Research (DZHK), Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Jana Grune
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| |
Collapse
|
12
|
Kanuri B, Biswas P, Dahdah A, Murphy AJ, Nagareddy PR. Impact of age and sex on myelopoiesis and inflammation during myocardial infarction. J Mol Cell Cardiol 2024; 187:80-89. [PMID: 38163742 PMCID: PMC10922716 DOI: 10.1016/j.yjmcc.2023.11.011] [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: 05/26/2023] [Revised: 11/16/2023] [Accepted: 11/21/2023] [Indexed: 01/03/2024]
Abstract
Of all the different risk factors known to cause cardiovascular disease (CVD), age and sex are considered to play a crucial role. Aging follows a continuum from birth to death, and therefore it inevitably acts as a risk for CVD. Along with age, sex differences have also been shown to demonstrate variations in immune system responses to pathological insults. It has been widely perceived that females are protected against myocardial infarction (MI) and the protection is quite apparent in young vs. old women. Acute MI leads to changes in the population of myeloid and lymphoid cells at the injury site with myeloid bias being observed in the initial inflammation and the lymphoid in the late-resolution phases of the pathology. Multiple evidence demonstrates that aging enhances damage to various cellular processes through inflamm-aging, an inflammatory process identified to increase pro-inflammatory markers in circulation and tissues. Following MI, marked changes were observed in different sub-sets of major myeloid cell types viz., neutrophils, monocytes, and macrophages. There is a paucity of information regarding the tissue and site-specific functions of these sub-sets. In this review, we highlight the importance of age and sex as crucial risk factors by discussing their role during MI-induced myelopoiesis while emphasizing the current status of myeloid cell sub-sets. We further put forth the need for designing and executing age and sex interaction studies aimed to determine the appropriate age and sex to develop personalized therapeutic strategies post-MI.
Collapse
Affiliation(s)
- Babunageswararao Kanuri
- Department of Internal Medicine, Section of Cardiovascular Diseases, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, OK, USA
| | - Priosmita Biswas
- Department of Internal Medicine, Section of Cardiovascular Diseases, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, OK, USA
| | - Albert Dahdah
- Department of Internal Medicine, Section of Cardiovascular Diseases, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, OK, USA
| | - Andrew J Murphy
- Baker Heart and Diabetes Institute, Division of Immunometabolism, Melbourne, Australia
| | - Prabhakara R Nagareddy
- Department of Internal Medicine, Section of Cardiovascular Diseases, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, OK, USA.
| |
Collapse
|
13
|
Usselman CW, Lindsey ML, Robinson AT, Habecker BA, Taylor CE, Merryman WD, Kimmerly D, Bender JR, Regensteiner JG, Moreau KL, Pilote L, Wenner MM, O'Brien M, Yarovinsky TO, Stachenfeld NS, Charkoudian N, Denfeld QE, Moreira-Bouchard JD, Pyle WG, DeLeon-Pennell KY. Guidelines on the use of sex and gender in cardiovascular research. Am J Physiol Heart Circ Physiol 2024; 326:H238-H255. [PMID: 37999647 PMCID: PMC11219057 DOI: 10.1152/ajpheart.00535.2023] [Citation(s) in RCA: 40] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/02/2023] [Accepted: 11/21/2023] [Indexed: 11/25/2023]
Abstract
In cardiovascular research, sex and gender have not typically been considered in research design and reporting until recently. This has resulted in clinical research findings from which not only all women, but also gender-diverse individuals have been excluded. The resulting dearth of data has led to a lack of sex- and gender-specific clinical guidelines and raises serious questions about evidence-based care. Basic research has also excluded considerations of sex. Including sex and/or gender as research variables not only has the potential to improve the health of society overall now, but it also provides a foundation of knowledge on which to build future advances. The goal of this guidelines article is to provide advice on best practices to include sex and gender considerations in study design, as well as data collection, analysis, and interpretation to optimally establish rigor and reproducibility needed to inform clinical decision-making and improve outcomes. In cardiovascular physiology, incorporating sex and gender is a necessary component when optimally designing and executing research plans. The guidelines serve as the first guidance on how to include sex and gender in cardiovascular research. We provide here a beginning path toward achieving this goal and improve the ability of the research community to interpret results through a sex and gender lens to enable comparison across studies and laboratories, resulting in better health for all.
Collapse
Affiliation(s)
- Charlotte W Usselman
- Cardiovascular Health and Autonomic Regulation Laboratory, Department of Kinesiology and Physical Education, McGill University, Montreal, Quebec, Canada
| | - Merry L Lindsey
- School of Graduate Studies, Meharry Medical College, Nashville, Tennessee, United States
- Research Service, Nashville Veterans Affairs Medical Center, Nashville, Tennessee, United States
| | - Austin T Robinson
- Neurovascular Physiology Laboratory, School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | - Beth A Habecker
- Department of Chemical Physiology and Biochemistry and Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon, United States
| | - Chloe E Taylor
- School of Health Sciences, Western Sydney University, Sydney, New South Wales, Australia
| | - W David Merryman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, United States
| | - Derek Kimmerly
- Autonomic Cardiovascular Control and Exercise Laboratory, Division of Kinesiology, School of Health and Human Performance, Faculty of Health, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Jeffrey R Bender
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, New Haven, Connecticut, United States
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, United States
| | - Judith G Regensteiner
- Divisions of General Internal Medicine and Cardiology, Department of Medicine, Ludeman Family Center for Women's Health Research, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Kerrie L Moreau
- Division of Geriatrics, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Eastern Colorado Health Care System, Geriatric Research Education and Clinical Center, Aurora, Colorado, United States
| | - Louise Pilote
- Centre for Outcomes Research and Evaluation, Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada
| | - Megan M Wenner
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, Delaware, United States
| | - Myles O'Brien
- School of Physiotherapy and Department of Medicine, Faculty of Health, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Timur O Yarovinsky
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, New Haven, Connecticut, United States
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, United States
| | - Nina S Stachenfeld
- John B. Pierce Laboratory, New Haven, Connecticut, United States
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, United States
| | - Nisha Charkoudian
- Thermal and Mountain Medicine Division, United States Army Research Institute of Environmental Medicine, Natick, Massachusetts, United States
| | - Quin E Denfeld
- School of Nursing and Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon, United States
| | - Jesse D Moreira-Bouchard
- Q.U.E.E.R. Lab, Programs in Human Physiology, Department of Health Sciences, Boston University College of Health and Rehabilitation Sciences: Sargent College, Boston, Massachusetts, United States
| | - W Glen Pyle
- IMPART Team Canada Network, Dalhousie Medicine, Saint John, New Brunswick, Canada
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Kristine Y DeLeon-Pennell
- School of Medicine, Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States
- Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina, United States
| |
Collapse
|
14
|
Xie W, Huang T, Guo Y, Zhang Y, Chen W, Li Y, Chen C, Li P. Neutrophil-derived cathelicidin promotes cerebral angiogenesis after ischemic stroke. J Cereb Blood Flow Metab 2023; 43:1503-1518. [PMID: 37194247 PMCID: PMC10414012 DOI: 10.1177/0271678x231175190] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/10/2023] [Accepted: 04/09/2023] [Indexed: 05/18/2023]
Abstract
Neutrophils play critical roles in the evolving of brain injuries following ischemic stroke. However, how they impact the brain repair in the late phase after stroke remain uncertain. Using a prospective clinical stroke patient cohort, we found significantly increased cathelicidin antimicrobial peptide (CAMP) in the peripheral blood of stroke patients compared to that of healthy controls. While in the mouse stroke model, CAMP was present in the peripheral blood, brain ischemic core and significantly increased at day 1, 3, 7, 14 after middle cerebral artery occlusion (MCAO). CAMP-/- mice exhibited significantly increased infarct volume, exacerbated neurological outcome, reduced cerebral endothelial cell proliferation and vascular density at 7 and 14 days after MCAO. Using bEND3 cells subjected to oxygen-glucose deprivation (OGD), we found significantly increased angiogenesis-related gene expression with the treatment of recombinant CAMP peptide (rCAMP) after reoxygenation. Intracerebroventricular injection (ICV) of AZD-5069, the antagonist of CAMP receptor CXCR2, or knockdown of CXCR2 by shCXCR2 recombinant adeno-associated virus (rAAV) impeded angiogenesis and neurological recovery after MCAO. Administration of rCAMP promoted endothelial proliferation and angiogenesis and attenuated neurological deficits 14 days after MCAO. In conclusion, neutrophil derived CAMP represents an important mediator that could promote post-stroke angiogenesis and neurological recovery in the late phase after stroke.
Collapse
Affiliation(s)
| | | | | | - Yueman Zhang
- Department of Anesthesiology, Key Laboratory of the Ministry of Education of Anesthesia Medicine, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weijie Chen
- Department of Anesthesiology, Key Laboratory of the Ministry of Education of Anesthesia Medicine, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Li
- Department of Anesthesiology, Key Laboratory of the Ministry of Education of Anesthesia Medicine, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Chen
- Department of Anesthesiology, Key Laboratory of the Ministry of Education of Anesthesia Medicine, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peiying Li
- Department of Anesthesiology, Key Laboratory of the Ministry of Education of Anesthesia Medicine, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
15
|
Liu Y, Wang P, Jin G, Shi P, Zhao Y, Guo J, Yin Y, Shao Q, Li P, Yang P. The novel function of bexarotene for neurological diseases. Ageing Res Rev 2023; 90:102021. [PMID: 37495118 DOI: 10.1016/j.arr.2023.102021] [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: 05/28/2023] [Revised: 07/13/2023] [Accepted: 07/20/2023] [Indexed: 07/28/2023]
Abstract
Bexarotene, a retinoid X receptor (RXR) agonist, is approved by FDA to treat cutaneous T-cell lymphoma. However, it has also demonstrated promising therapeutic potential for neurological diseases such as stroke, traumatic brain injury, Parkinson's disease, and particularly Alzheimer's disease(AD). In AD, bexarotene inhibits the production and aggregation of amyloid β (Aβ), activates Liver X Receptor/RXR heterodimers to increase lipidated apolipoprotein E to remove Aβ, mitigates the negative impact of Aβ, regulates neuroinflammation, and ultimately improves cognitive function. For other neurological diseases, its mechanisms of action include inhibiting inflammatory responses, up-regulating microglial phagocytosis, and reducing misfolded protein aggregation, all of which aid in alleviating neurological damage. Here, we briefly discuss the characteristics, applications, and adverse effects of bexarotene, summarize its pharmacological mechanisms and therapeutic results in various neurological diseases, and elaborate on the problems encountered in preclinical research, with the aim of providing help for the further application of bexarotene in central nervous system diseases.
Collapse
Affiliation(s)
- Yangtao Liu
- College of Pharamacy, Xinxiang Medical University, Xinxiang, China; Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China; College of Third Clinical, Xinxiang Medical University, Xinxiang, China
| | - Pengwei Wang
- Department of Pharmacy, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang Medical University, Weihui, China
| | - Guofang Jin
- College of Pharamacy, Xinxiang Medical University, Xinxiang, China; Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China
| | - Peijie Shi
- College of Pharamacy, Xinxiang Medical University, Xinxiang, China; Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China; Xinxiang First People's Hospital, The Affiliated People's Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yonghui Zhao
- Xinxiang First People's Hospital, The Affiliated People's Hospital of Xinxiang Medical University, Xinxiang, China
| | - Jiayi Guo
- College of Pharamacy, Xinxiang Medical University, Xinxiang, China; Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China
| | - Yaling Yin
- School of Basic Medical Sciences, Department of Physiology and Pathophysiology, Sino-UK Joint Laboratory of Brain Function and Injury and Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, China
| | - Qianhang Shao
- Department of Pharmacy, People's Hospital of Peking University, Beijing, China.
| | - Peng Li
- College of Pharamacy, Xinxiang Medical University, Xinxiang, China; Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.
| | - Pengfei Yang
- College of Pharamacy, Xinxiang Medical University, Xinxiang, China; Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.
| |
Collapse
|
16
|
Zhou W, Cao X, Xu Q, Qu J, Sun Y. The double-edged role of neutrophil heterogeneity in inflammatory diseases and cancers. MedComm (Beijing) 2023; 4:e325. [PMID: 37492784 PMCID: PMC10363828 DOI: 10.1002/mco2.325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 05/31/2023] [Accepted: 06/09/2023] [Indexed: 07/27/2023] Open
Abstract
Neutrophils are important immune cells act as the body's first line of defense against infection and respond to diverse inflammatory cues. Many studies have demonstrated that neutrophils display plasticity in inflammatory diseases and cancers. Clarifying the role of neutrophil heterogeneity in inflammatory diseases and cancers will contribute to the development of novel treatment strategies. In this review, we have presented a review on the development of the understanding on neutrophil heterogeneity from the traditional perspective and a high-resolution viewpoint. A growing body of evidence has confirmed the double-edged role of neutrophils in inflammatory diseases and tumors. This may be due to a lack of precise understanding of the role of specific neutrophil subsets in the disease. Thus, elucidating specific neutrophil subsets involved in diseases would benefit the development of precision medicine. Thusly, we have summarized the relevance and actions of neutrophil heterogeneity in inflammatory diseases and cancers comprehensively. Meanwhile, we also discussed the potential intervention strategy for neutrophils. This review is intended to deepen our understanding of neutrophil heterogeneity in inflammatory diseases and cancers, while hold promise for precise treatment of neutrophil-related diseases.
Collapse
Affiliation(s)
- Wencheng Zhou
- Department of PharmacyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine)HangzhouChina
| | - Xinran Cao
- State Key Laboratory of Pharmaceutical BiotechnologyDepartment of Biotechnology and Pharmaceutical SciencesSchool of Life ScienceNanjing UniversityNanjingChina
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical BiotechnologyDepartment of Biotechnology and Pharmaceutical SciencesSchool of Life ScienceNanjing UniversityNanjingChina
| | - Jiao Qu
- State Key Laboratory of Pharmaceutical BiotechnologyDepartment of Biotechnology and Pharmaceutical SciencesSchool of Life ScienceNanjing UniversityNanjingChina
| | - Yang Sun
- State Key Laboratory of Pharmaceutical BiotechnologyDepartment of Biotechnology and Pharmaceutical SciencesSchool of Life ScienceNanjing UniversityNanjingChina
| |
Collapse
|
17
|
Ma L, Shao M, Cheng W, Jiang J, Chen X, Tan N, Ling G, Yang Y, Wang Q, Yang R, Li C, Wang Y. Neocryptotanshinone ameliorates insufficient energy production in heart failure by targeting retinoid X receptor alpha. Biomed Pharmacother 2023; 163:114868. [PMID: 37201263 DOI: 10.1016/j.biopha.2023.114868] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/08/2023] [Accepted: 05/10/2023] [Indexed: 05/20/2023] Open
Abstract
Retinoid X receptor alpha (RXRα) is a nuclear transcription factor that extensively regulates energy metabolism in cardiovascular diseases. Identification of targeted RXRα drugs for heart failure (HF) therapy is urgently needed. Neocryptotanshinone (NCTS) is a component derived from Salvia miltiorrhiza Bunge, the effect and mechanism of which for treating HF have not been reported. The goal of this study was to explore the pharmacological effects of NCTS on energy metabolism to protect against HF post-acute myocardial infarction (AMI) via RXRα. We established a left anterior descending artery ligation-induced HF post-AMI model in mice and an oxygen-glucose deprivation-reperfusion-induced H9c2 cell model to investigate the cardioprotective effect of NCTS. Component-target binding techniques, surface plasmon resonance (SPR), microscale thermophoresis (MST) and small interfering RNA (siRNA) transfection were applied to explore the potential mechanism by which NCTS targets RXRα. The results showed that NCTS protects the heart against ischaemic damage, evidenced by improvement of cardiac dysfunction and attenuation of cellular hypoxic injury. Importantly, the SPR and MST results showed that NCTS has a high binding affinity for RXRα. Meanwhile, the critical downstream target genes of RXRα/PPARα, which are involved in fatty acid metabolism, including Cd36 and Cpt1a, were upregulated under NCTS treatment. Moreover, NCTS enhanced TFAM levels, promoted mitochondrial biogenesis and increased myocardial adenosine triphosphate levels by activating RXRα. In conclusion, we confirmed that NCTS improves myocardial energy metabolism, including fatty acid oxidation and mitochondrial biogenesis, by regulating the RXRα/PPARα pathway in mice with HF post-AMI.
Collapse
Affiliation(s)
- Lin Ma
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Mingyan Shao
- School of Life Science, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Wenkun Cheng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jinchi Jiang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xu Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Nannan Tan
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Guanjing Ling
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Ye Yang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Qian Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Ran Yang
- Guang'anmen Hospital China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Chun Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Yong Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; School of Life Science, Beijing University of Chinese Medicine, Beijing 100029, China; Beijing Key Laboratory of TCM Syndrome and Formula, Beijing 100029, China; Key Laboratory of Beijing University of Chinese Medicine, Ministry of Education, Beijing 100029, China.
| |
Collapse
|
18
|
Pearce DP, Nemcek MT, Witzenburg CM. Don't go breakin' my heart: cardioprotective alterations to the mechanical and structural properties of reperfused myocardium during post-infarction inflammation. Biophys Rev 2023; 15:329-353. [PMID: 37396449 PMCID: PMC10310682 DOI: 10.1007/s12551-023-01068-3] [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: 05/01/2023] [Accepted: 05/21/2023] [Indexed: 07/04/2023] Open
Abstract
Myocardial infarctions (MIs) kickstart an intense inflammatory response resulting in extracellular matrix (ECM) degradation, wall thinning, and chamber dilation that leaves the heart susceptible to rupture. Reperfusion therapy is one of the most effective strategies for limiting adverse effects of MIs, but is a challenge to administer in a timely manner. Late reperfusion therapy (LRT; 3 + hours post-MI) does not limit infarct size, but does reduce incidences of post-MI rupture and improves long-term patient outcomes. Foundational studies employing LRT in the mid-twentieth century revealed beneficial reductions in infarct expansion, aneurysm formation, and left ventricle dysfunction. The mechanism by which LRT acts, however, is undefined. Structural analyses, relying largely on one-dimensional estimates of ECM composition, have found few differences in collagen content between LRT and permanently occluded animal models when using homogeneous samples from infarct cores. Uniaxial testing, on the other hand, revealed slight reductions in stiffness early in inflammation, followed soon after by an enhanced resistance to failure for cases of LRT. The use of one-dimensional estimates of ECM organization and gross mechanical function have resulted in a poor understanding of the infarct's spatially variable mechanical and structural anisotropy. To resolve these gaps in literature, future work employing full-field mechanical, structural, and cellular analyses is needed to better define the spatiotemporal post-MI alterations occurring during the inflammatory phase of healing and how they are impacted following reperfusion therapy. In turn, these studies may reveal how LRT affects the likelihood of rupture and inspire novel approaches to guide scar formation.
Collapse
Affiliation(s)
- Daniel P. Pearce
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706 USA
| | - Mark T. Nemcek
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706 USA
| | - Colleen M. Witzenburg
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706 USA
| |
Collapse
|
19
|
Mouton AJ, Aitken NM, Moak SP, do Carmo JM, da Silva AA, Omoto ACM, Li X, Wang Z, Schrimpe-Rutledge AC, Codreanu SG, Sherrod SD, McLean JA, Hall JE. Temporal changes in glucose metabolism reflect polarization in resident and monocyte-derived macrophages after myocardial infarction. Front Cardiovasc Med 2023; 10:1136252. [PMID: 37215542 PMCID: PMC10196495 DOI: 10.3389/fcvm.2023.1136252] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 03/28/2023] [Indexed: 05/24/2023] Open
Abstract
Introduction Metabolic reprogramming from glycolysis to the mitochondrial tricarboxylic acid (TCA) cycle and oxidative phosphorylation may mediate macrophage polarization from the pro-inflammatory M1 to the anti-inflammatory M2 phenotype. We hypothesized that changes in cardiac macrophage glucose metabolism would reflect polarization status after myocardial infarction (MI), ranging from the early inflammatory phase to the later wound healing phase. Methods MI was induced by permanent ligation of the left coronary artery in adult male C57BL/6J mice for 1 (D1), 3 (D3), or 7 (D7) days. Infarct macrophages were subjected to metabolic flux analysis or gene expression analysis. Monocyte versus resident cardiac macrophage metabolism was assessed using mice lacking the Ccr2 gene (CCR2 KO). Results By flow cytometry and RT-PCR, D1 macrophages exhibited an M1 phenotype while D7 macrophages exhibited an M2 phenotype. Macrophage glycolysis (extracellular acidification rate) was increased at D1 and D3, returning to basal levels at D7. Glucose oxidation (oxygen consumption rate) was decreased at D3, returning to basal levels at D7. At D1, glycolytic genes were elevated (Gapdh, Ldha, Pkm2), while TCA cycle genes were elevated at D3 (Idh1 and Idh2) and D7 (Pdha1, Idh1/2, Sdha/b). Surprisingly, Slc2a1 and Hk1/2 were increased at D7, as well as pentose phosphate pathway (PPP) genes (G6pdx, G6pd2, Pgd, Rpia, Taldo1), indicating increased PPP activity. Macrophages from CCR2 KO mice showed decreased glycolysis and increased glucose oxidation at D3, and decreases in Ldha and Pkm2 expression. Administration of dichloroacetate, a pyruvate dehydrogenase kinase inhibitor, robustly decreased pyruvate dehydrogenase phosphorylation in the non-infarcted remote zone, but did not affect macrophage phenotype or metabolism in the infarct zone. Discussion Our results indicate that changes in glucose metabolism and the PPP underlie macrophage polarization following MI, and that metabolic reprogramming is a key feature of monocyte-derived but not resident macrophages.
Collapse
Affiliation(s)
- Alan J. Mouton
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, United States
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, United States
| | - Nikaela M. Aitken
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, United States
| | - Sydney P. Moak
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, United States
| | - Jussara M. do Carmo
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, United States
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, United States
| | - Alexandre A. da Silva
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, United States
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, United States
| | - Ana C. M. Omoto
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, United States
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, United States
| | - Xuan Li
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, United States
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, United States
| | - Zhen Wang
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, United States
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, United States
| | | | - Simona G. Codreanu
- Department of Chemistry and Center for Innovative Technology, Vanderbilt University, Nashville, TN, United States
| | - Stacy D. Sherrod
- Department of Chemistry and Center for Innovative Technology, Vanderbilt University, Nashville, TN, United States
| | - John A. McLean
- Department of Chemistry and Center for Innovative Technology, Vanderbilt University, Nashville, TN, United States
| | - John E. Hall
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, United States
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, United States
| |
Collapse
|
20
|
Corker A, Learmonth M, Patrick DM, DeLeon-Pennell KY, Van Beusecum JP. Cardiac and vascular complications in lupus: Is there a role for sex? Front Immunol 2023; 14:1098383. [PMID: 37063843 PMCID: PMC10090292 DOI: 10.3389/fimmu.2023.1098383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 03/07/2023] [Indexed: 03/30/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a common systemic autoimmune disorder and is characterized by autoantibody formation and subsequent immune complex deposition into target organs. SLE affects nearly nine women to every one man worldwide. Patients with SLE are at an enhanced risk for cardiovascular disease (CVD) morbidity and mortality. CVD is the leading cause of death worldwide and includes heart and blood vessel disorders, cerebrovascular disease, and rheumatic heart disease. Specific mechanisms by which cardiac and vascular pathophysiology develops in patients with SLE are still not fully known. Not only do we not understand this correlation between SLE and CVD, but there is also a critical gap in scientific knowledge on the contribution of sex. In this review, we will discuss the cardiac and vascular pathological disease states that are present in some patients with SLE. More importantly, we will discuss the potential mechanisms for the role of sex and sex hormones in the development of CVD with SLE.
Collapse
Affiliation(s)
- Alexa Corker
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Maya Learmonth
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - David M. Patrick
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Research Service, Tennessee Valley Healthcare Veterans Affairs (VA) Medical Center, Nashville, TN, United States
| | - Kristine Y. DeLeon-Pennell
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, SC, United States
- Department of Research Service, Ralph H. Johnson Veterans Affairs (VA) Healthcare System, Charleston, SC, United States
| | - Justin P. Van Beusecum
- Department of Research Service, Ralph H. Johnson Veterans Affairs (VA) Healthcare System, Charleston, SC, United States
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
21
|
Chalise U, Becirovic‐Agic M, Lindsey ML. The cardiac wound healing response to myocardial infarction. WIREs Mech Dis 2023; 15:e1584. [PMID: 36634913 PMCID: PMC10077990 DOI: 10.1002/wsbm.1584] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 03/31/2022] [Accepted: 05/18/2022] [Indexed: 01/14/2023]
Abstract
Myocardial infarction (MI) is defined as evidence of myocardial necrosis consistent with prolonged ischemia. In response to MI, the myocardium undergoes a series of wound healing events that initiate inflammation and shift to anti-inflammation before transitioning to tissue repair that culminates in scar formation to replace the region of the necrotic myocardium. The overall response to MI is determined by two major steps, the first of which is the secretion of proteases by infiltrating leukocytes to breakdown extracellular matrix (ECM) components, a necessary step to remove necrotic cardiomyocytes. The second step is the generation of new ECM that comprises the scar; and this step is governed by the cardiac fibroblasts as the major source of new ECM synthesis. The leukocyte component resides in the middle of the two-step process, contributing to both sides as the leukocytes transition from pro-inflammatory to anti-inflammatory and reparative cell phenotypes. The balance between the two steps determines the final quantity and quality of scar formed, which in turn contributes to chronic outcomes following MI, including the progression to heart failure. This review will summarize our current knowledge regarding the cardiac wound healing response to MI, primarily focused on experimental models of MI in mice. This article is categorized under: Cardiovascular Diseases > Molecular and Cellular Physiology Immune System Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Upendra Chalise
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular ResearchUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Research ServiceNebraska‐Western Iowa Health Care SystemOmahaNebraskaUSA
| | - Mediha Becirovic‐Agic
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular ResearchUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Research ServiceNebraska‐Western Iowa Health Care SystemOmahaNebraskaUSA
| | - Merry L. Lindsey
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular ResearchUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Research ServiceNebraska‐Western Iowa Health Care SystemOmahaNebraskaUSA
| |
Collapse
|
22
|
Joshua J, Caswell J, O’Sullivan ML, Wood G, Fonfara S. Feline myocardial transcriptome in health and in hypertrophic cardiomyopathy-A translational animal model for human disease. PLoS One 2023; 18:e0283244. [PMID: 36928240 PMCID: PMC10019628 DOI: 10.1371/journal.pone.0283244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 03/03/2023] [Indexed: 03/18/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common heart disease in cats, characterized by primary left ventricular hypertrophy. Feline HCM closely resembles human HCM and is suggested as translational animal model for the human disease. A genetic cause is established in humans and suspected for cats, but little is known about the gene expression and pathways involved in the pathogenesis of HCM. To investigate the myocardial transcriptome changes in HCM, RNA sequencing was conducted on left ventricle (LV) and left atrium (LA) samples of healthy cats and cats with HCM (each n = 5; 20 samples). Ingenuity Pathway Analysis was used to determine functional pathways, regulators, and networks. Distinct gene expression profiles were identified in the LV and LA of the feline healthy and HCM myocardium. Analysis of differentially expressed mRNAs (>2 fold; FDR < 0.01) found chamber-specific (LV vs. LA) expression in both healthy and HCM groups, with higher transcriptional activity in the LA. Genes that contribute to the distinct structure and function of each chamber in health and HCM were identified in the regional comparison. The gene expression profiles of HCM compared to healthy hearts revealed disease related genes, including THBS4 and KLHL33 (LV), FAM177B and THRSP (LA), the latter 3 have not been reported for the myocardium so far, as the top differently expressed genes in the HCM heart. Differently expressed genes and functional pathways found in the HCM heart are associated with cardiac remodeling and fibrosis, inflammation, microvascular changes, calcium signaling and cardiac metabolism, with some regional differences. RhoGDI-RhoGTPase signaling, integrin and ILK signaling pathways, the LXR/RXR pathway in the LA, and the PPARα/RXRα, HIF1α and CXCR4 pathways in the LV might be of particular importance in the HCM disease process. This study identified region-specific myocardial gene transcription patterns as well as novel genes and pathways associated with HCM.
Collapse
Affiliation(s)
- Jessica Joshua
- University of Guelph, Ontario Veterinary College, Department of Pathobiology, Guelph, Ontario, Canada
- University of Guelph, Ontario Veterinary College, Department of Clinical Studies, Guelph, Ontario, Canada
| | - Jeff Caswell
- University of Guelph, Ontario Veterinary College, Department of Pathobiology, Guelph, Ontario, Canada
| | - M. Lynne O’Sullivan
- University of Prince Edward Island, Department of Companion Animals, Charlottetown, Prince Edward Island, Canada
| | - Geoffrey Wood
- University of Guelph, Ontario Veterinary College, Department of Pathobiology, Guelph, Ontario, Canada
| | - Sonja Fonfara
- University of Guelph, Ontario Veterinary College, Department of Clinical Studies, Guelph, Ontario, Canada
- * E-mail:
| |
Collapse
|
23
|
Song BY, Chen C, Xu WH, Cong BL, Guo ZY, Zhao ZH, Cui L, Zhang YH. Gender Differences in the Correlations Between Immune Cells and Organ Damage Indexes of Acute Myocardial Infarction Patients. Vasc Health Risk Manag 2022; 18:839-850. [DOI: 10.2147/vhrm.s374157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 10/12/2022] [Indexed: 12/03/2022] Open
|
24
|
Broughton P, Troncoso M, Corker A, Williams A, Bolus D, Munoz G, McWhorter C, Roerden H, Huebsch P, DeLeon-Pennell KY. Riding the wave: a quantitative report of electrocardiogram utilization for myocardial infarction confirmation. Am J Physiol Heart Circ Physiol 2022; 323:H378-H387. [PMID: 35802516 PMCID: PMC9359650 DOI: 10.1152/ajpheart.00201.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/22/2022] [Accepted: 07/07/2022] [Indexed: 11/22/2022]
Abstract
The purpose of this study was to generate a quantitative profile of electrocardiograms (ECGs) for confirming surgical success of permanent coronary artery ligation. An ECG was recorded at baseline, and 0, 1, and 5 min after ligation and analyzed using iWorkx LabScribe software. Cohort 1 (C57Bl6/J, n = 8/sex) was enrolled to determine ECG characteristics that were confirmed in cohort 2 (C57Bl6/J, n = 6/sex; CD8-/-n = 6 males/4 females). Of the 16 mice in cohort 1, 12 (6/sex) had an infarct ≥35% and four mice (2/sex) had <35% based on 2,3,5-triphenyltetrazolium chloride staining. After ligation, the QRS complex and R-S amplitude were significantly different compared with baseline. No differences were observed in the R-S amplitude between mice with infarcts ≥35% versus <35% at any time point, whereas the QRS complex was significant 1 min after ligation. Receiver operating characteristic (ROC) curve linked changes in the QRS complex but not the R-S amplitude at 1 and 5 min with surgical success. Data were normalized to baseline values to calculate fold change. ROC analysis of the normalized QRS data indicated strong sensitivity and specificity for infarcts ≥35%; normalized R-S amplitude remained nonsignificant. With a cutoff generated by ROC analysis of cohort 1 (>80% sensitivity; >90% specificity), the non-normalized QRS complex of cohort 2 had an 86% success rate (2 false positives; 1 false negative). The normalized data had a 77% success rate (2 false positives; 3 false negatives). Neither sex nor genotype was associated with false predictions (P = 0.18). Our data indicate that the area under the QRS complex 1 min after ligation can improve reproducibility in MI surgeries.NEW & NOTEWORTHY Our study describes a quantitative method for using an electrocardiogram (ECG) to determine which animals have infarcts that reflect coronary artery ligation. Using a quantitative ECG, investigators will have the benefit of having real-time feedback during the procedure, which will ultimately decrease the amount of time investigators spend performing experiments. This overall increase in efficiency will help investigators decrease animal numbers used due to better surgical outcomes.
Collapse
Affiliation(s)
- Philip Broughton
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Miguel Troncoso
- Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| | - Alexa Corker
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Alexus Williams
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Dawson Bolus
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Gualberto Munoz
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Caroline McWhorter
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Hallie Roerden
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Penny Huebsch
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Kristine Y DeLeon-Pennell
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
- Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| |
Collapse
|
25
|
Zorrilla Veloz RI, McKenzie T, Palacios BE, Hu J. Nuclear hormone receptors in demyelinating diseases. J Neuroendocrinol 2022; 34:e13171. [PMID: 35734821 PMCID: PMC9339486 DOI: 10.1111/jne.13171] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 04/20/2022] [Accepted: 05/27/2022] [Indexed: 11/28/2022]
Abstract
Demyelination results from the pathological loss of myelin and is a hallmark of many neurodegenerative diseases. Despite the prevalence of demyelinating diseases, there are no disease modifying therapies that prevent the loss of myelin or promote remyelination. This review aims to summarize studies in the field that highlight the importance of nuclear hormone receptors in the promotion and maintenance of myelination and the relevance of nuclear hormone receptors as potential therapeutic targets for demyelinating diseases. These nuclear hormone receptors include the estrogen receptor, progesterone receptor, androgen receptor, vitamin D receptor, thyroid hormone receptor, peroxisome proliferator-activated receptor, liver X receptor, and retinoid X receptor. Pre-clinical studies in well-established animal models of demyelination have shown a prominent role of these nuclear hormone receptors in myelination through their promotion of oligodendrocyte maturation and development. The activation of the nuclear hormone receptors by their ligands also promotes the synthesis of myelin proteins and lipids in mouse models of demyelination. There are limited clinical studies that focus on how the activation of these nuclear hormone receptors could alleviate demyelination in patients with diseases such as multiple sclerosis (MS). However, the completed clinical trials have reported improved clinical outcome in MS patients treated with the ligands of some of these nuclear hormone receptors. Together, the positive results from both clinical and pre-clinical studies point to nuclear hormone receptors as promising therapeutic targets to counter demyelination.
Collapse
Affiliation(s)
- Rocío I Zorrilla Veloz
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Cancer Biology Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Takese McKenzie
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Neuroscience Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Bridgitte E Palacios
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Cancer Biology Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
- Neuroscience Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Jian Hu
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Cancer Biology Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
- Neuroscience Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| |
Collapse
|
26
|
Sex Differences in Cardiovascular Diseases: A Matter of Estrogens, Ceramides, and Sphingosine 1-Phosphate. Int J Mol Sci 2022; 23:ijms23074009. [PMID: 35409368 PMCID: PMC8999971 DOI: 10.3390/ijms23074009] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/01/2022] [Accepted: 04/04/2022] [Indexed: 12/30/2022] Open
Abstract
The medical community recognizes sex-related differences in pathophysiology and cardiovascular disease outcomes (CVD), culminating with heart failure. In general, pre-menopausal women tend to have a better prognosis than men. Explaining why this occurs is not a simple matter. For decades, sex hormones like estrogens (Es) have been identified as one of the leading factors driving these sex differences. Indeed, Es seem protective in women as their decline, during and after menopause, coincides with an increased CV risk and HF development. However, clinical trials demonstrated that E replacement in post-menopause women results in adverse cardiac events and increased risk of breast cancer. Thus, a deeper understanding of E-related mechanisms is needed to provide a vital gateway toward better CVD prevention and treatment in women. Of note, sphingolipids (SLs) and their metabolism are strictly related to E activities. Among the SLs, ceramide and sphingosine 1-phosphate play essential roles in mammalian physiology, particularly in the CV system, and appear differently modulated in males and females. In keeping with this view, here we explore the most recent experimental and clinical observations about the role of E and SL metabolism, emphasizing how these factors impact the CV system.
Collapse
|
27
|
Chalise U, Daseke MJ, Kalusche WJ, Konfrst SR, Rodriguez-Paar JR, Flynn ER, Cook LM, Becirovic-Agic M, Lindsey ML. Macrophages secrete murinoglobulin-1 and galectin-3 to regulate neutrophil degranulation after myocardial infarction. Mol Omics 2022; 18:186-195. [PMID: 35230372 PMCID: PMC8963000 DOI: 10.1039/d1mo00519g] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 02/11/2022] [Indexed: 02/03/2023]
Abstract
Inflammation presides early after myocardial infarction (MI) as a key event in cardiac wound healing. Ischemic cardiomyocytes secrete inflammatory cues to stimulate infiltration of leukocytes, predominantly macrophages and neutrophils. Infiltrating neutrophils degranulate to release a series of proteases including matrix metalloproteinase (MMP)-9 to break down extracellular matrix and remove necrotic myocytes to create space for the infarct scar to form. While neutrophil to macrophage communication has been explored, the reverse has been understudied. We used a proteomics approach to catalogue the macrophage secretome at MI day 1. Murinoglobulin-1 (MUG1) was the highest-ranked secreted protein (4.1-fold upregulated at MI day 1 vs. day 0 pre-MI cardiac macrophages, p = 0.004). By transcriptomics evaluation, galectin-3 (Lgals3) was 2.2-fold upregulated (p = 0.008) in MI day 1 macrophages. We explored the direct roles of MUG1 and Lgals3 on neutrophil degranulation. MUG1 blunted while Lgals3 amplified neutrophil degranulation in response to phorbol 12-myristate 13-acetate or interleukin-1β, as measured by MMP-9 secretion. Lgals3 itself also stimulated MMP-9 secretion. To determine if MUG1 regulated Lgals3, we co-stimulated neutrophils with MUG1 and Lgals3. MUG1 limited degranulation stimulated by Lgals3 by 64% (p < 0.001). In vivo, MUG1 was elevated in the infarct region at MI days 1 and 3, while Lgals3 increased at MI day 7. The ratio of MUG1 to Lgals3 positively correlated with infarct wall thickness, revealing that MUG1 attenuated infarct wall thinning. In conclusion, macrophages at MI day 1 secrete MUG1 to limit and Lgals3 to accentuate neutrophil degranulation to regulate infarct wall thinning.
Collapse
Affiliation(s)
- Upendra Chalise
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE, 68105, USA
| | - Michael J Daseke
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE, 68105, USA
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - William J Kalusche
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Shelby R Konfrst
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE, 68105, USA
| | - Jocelyn R Rodriguez-Paar
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE, 68105, USA
| | - Elizabeth R Flynn
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Leah M Cook
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Mediha Becirovic-Agic
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE, 68105, USA
| | - Merry L Lindsey
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE, 68105, USA
| |
Collapse
|
28
|
Zalesak-Kravec S, Huang W, Jones JW, Yu J, Alloush J, Defnet AE, Moise AR, Kane MA. Role of cellular retinol-binding protein, type 1 and retinoid homeostasis in the adult mouse heart: A multi-omic approach. FASEB J 2022; 36:e22242. [PMID: 35253263 DOI: 10.1096/fj.202100901rrr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 02/15/2022] [Accepted: 02/17/2022] [Indexed: 12/14/2022]
Abstract
The main active metabolite of Vitamin A, all-trans retinoic acid (RA), is required for proper cellular function and tissue organization. Heart development has a well-defined requirement for RA, but there is limited research on the role of RA in the adult heart. Homeostasis of RA includes regulation of membrane receptors, chaperones, enzymes, and nuclear receptors. Cellular retinol-binding protein, type 1 (CRBP1), encoded by retinol-binding protein, type 1 (Rbp1), regulates RA homeostasis by delivering vitamin A to enzymes for RA synthesis and protecting it from non-specific oxidation. In this work, a multi-omics approach was used to characterize the effect of CRBP1 loss using the Rbp1-/- mouse. Retinoid homeostasis was disrupted in Rbp1-/- mouse heart tissue, as seen by a 33% and 24% decrease in RA levels in the left and right ventricles, respectively, compared to wild-type mice (WT). To further inform on the effect of disrupted RA homeostasis, we conducted high-throughput targeted metabolomics. A total of 222 metabolite and metabolite combinations were analyzed, with 33 having differential abundance between Rbp1-/- and WT hearts. Additionally, we performed global proteome profiling to further characterize the impact of CRBP1 loss in adult mouse hearts. More than 2606 unique proteins were identified, with 340 proteins having differential expression between Rbp1-/- and WT hearts. Pathway analysis performed on metabolomic and proteomic data revealed pathways related to cellular metabolism and cardiac metabolism were the most disrupted in Rbp1-/- mice. Together, these studies characterize the effect of CRBP1 loss and reduced RA in the adult heart.
Collapse
Affiliation(s)
- Stephanie Zalesak-Kravec
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| | - Weiliang Huang
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| | - Jace W Jones
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| | - Jianshi Yu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| | - Jenna Alloush
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| | - Amy E Defnet
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| | - Alexander R Moise
- Medical Sciences Division, Northern Ontario School of Medicine, Sudbury, Ontario, Canada
| | - Maureen A Kane
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| |
Collapse
|
29
|
Lindsey ML, Brunt KR, Kirk JA, Kleinbongard P, Calvert JW, de Castro Brás LE, DeLeon-Pennell KY, Del Re DP, Frangogiannis NG, Frantz S, Gumina RJ, Halade GV, Jones SP, Ritchie RH, Spinale FG, Thorp EB, Ripplinger CM, Kassiri Z. Guidelines for in vivo mouse models of myocardial infarction. Am J Physiol Heart Circ Physiol 2021; 321:H1056-H1073. [PMID: 34623181 PMCID: PMC8834230 DOI: 10.1152/ajpheart.00459.2021] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 10/05/2021] [Accepted: 10/05/2021] [Indexed: 12/11/2022]
Abstract
Despite significant improvements in reperfusion strategies, acute coronary syndromes all too often culminate in a myocardial infarction (MI). The consequent MI can, in turn, lead to remodeling of the left ventricle (LV), the development of LV dysfunction, and ultimately progression to heart failure (HF). Accordingly, an improved understanding of the underlying mechanisms of MI remodeling and progression to HF is necessary. One common approach to examine MI pathology is with murine models that recapitulate components of the clinical context of acute coronary syndrome and subsequent MI. We evaluated the different approaches used to produce MI in mouse models and identified opportunities to consolidate methods, recognizing that reperfused and nonreperfused MI yield different responses. The overall goal in compiling this consensus statement is to unify best practices regarding mouse MI models to improve interpretation and allow comparative examination across studies and laboratories. These guidelines will help to establish rigor and reproducibility and provide increased potential for clinical translation.
Collapse
Affiliation(s)
- Merry L Lindsey
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, Nebraska
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, Nebraska
| | - Keith R Brunt
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Saint John, New Brunswick, Canada
| | - Jonathan A Kirk
- Department of Cell and Molecular Physiology, Loyola University Chicago Stritch School of Medicine, Chicago, Illinois
| | - Petra Kleinbongard
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany
| | - John W Calvert
- Carlyle Fraser Heart Center of Emory University Hospital Midtown, Atlanta, Georgia
- Division of Cardiothoracic Surgery, Department of Surgery, Emory University School of Medicine, Atlanta, Georgia
| | - Lisandra E de Castro Brás
- Department of Physiology, The Brody School of Medicine, East Carolina University, Greenville, North Carolina
| | - Kristine Y DeLeon-Pennell
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
- Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| | - Dominic P Del Re
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Nikolaos G Frangogiannis
- Division of Cardiology, Department of Medicine, The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York
| | - Stefan Frantz
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - Richard J Gumina
- Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, Ohio
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Ganesh V Halade
- Division of Cardiovascular Sciences, Department of Medicine, University of South Florida, Tampa, Florida
| | - Steven P Jones
- Department of Medicine, Diabetes and Obesity Center, University of Louisville, Louisville, Kentucky
| | - Rebecca H Ritchie
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), Victoria, Australia
| | - Francis G Spinale
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine and the Columbia Veteran Affairs Medical Center, Columbia, South Carolina
| | - Edward B Thorp
- Department of Pathology and Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Crystal M Ripplinger
- Department of Pharmacology, University of California Davis School of Medicine, Davis, California
| | - Zamaneh Kassiri
- Department of Physiology, Cardiovascular Research Center, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
30
|
Nolan E, Malanchi I. Connecting the dots: Neutrophils at the interface of tissue regeneration and cancer. Semin Immunol 2021; 57:101598. [PMID: 35221216 PMCID: PMC9232712 DOI: 10.1016/j.smim.2022.101598] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/19/2022] [Accepted: 02/08/2022] [Indexed: 11/28/2022]
Abstract
Knowledge about neutrophil biology has exponentially grown over the past decades. A high volume of investigations focusing on the characterization of their initially unappreciated multifaceted functions have grown in parallel with the immunity and the cancer fields. This has led to a significant gain in knowledge about their functions not only in tissue defence against pathogens and the collateral damage their overactivation can cause, but also their role in tissue repair and regeneration especially in the context of sterile injuries. On the other hand, the cancer field has also intensively focused its attention on neutrophil engagement in the many steps of the tumorigenic process. This review aims to draw the readers' attention to the similar functions described for neutrophils in tissue repair and in cancer. By bridging the two fields, we provide support for the hypothesis that the underlying program driving cancer-dependent exploitation of neutrophils is rooted in their physiologic tissue protection functions. In this view, cross-fertilization between the two fields will expedite the discovery of therapeutic interventions based on neutrophil targeting or their manipulation.
Collapse
Affiliation(s)
- Emma Nolan
- Tumour Host Interaction Laboratory, The Francis Crick Institute, 1 Midland Road, NW1 1AT London, United Kingdom
| | - Ilaria Malanchi
- Tumour Host Interaction Laboratory, The Francis Crick Institute, 1 Midland Road, NW1 1AT London, United Kingdom.
| |
Collapse
|
31
|
Zalesak-Kravec S, Huang W, Wang P, Yu J, Liu T, Defnet AE, Moise AR, Farese AM, MacVittie TJ, Kane MA. Multi-omic Analysis of Non-human Primate Heart after Partial-body Radiation with Minimal Bone Marrow Sparing. HEALTH PHYSICS 2021; 121:352-371. [PMID: 34546217 PMCID: PMC8554778 DOI: 10.1097/hp.0000000000001478] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
ABSTRACT High-dose radiation exposure results in hematopoietic and gastrointestinal acute radiation syndromes followed by delayed effects of acute radiation exposure, which encompasses multiple organs, including heart, kidney, and lung. Here we sought to further characterize the natural history of radiation-induced heart injury via determination of differential protein and metabolite expression in the heart. We quantitatively profiled the proteome and metabolome of left and right ventricle from non-human primates following 12 Gy partial body irradiation with 2.5% bone marrow sparing over a time period of 3 wk. Global proteome profiling identified more than 2,200 unique proteins, with 220 and 286 in the left and right ventricles, respectively, showing significant responses across at least three time points compared to baseline levels. High-throughput targeted metabolomics analyzed a total of 229 metabolites and metabolite combinations, with 18 and 22 in the left and right ventricles, respectively, showing significant responses compared to baseline levels. Bioinformatic analysis performed on metabolomic and proteomic data revealed pathways related to inflammation, energy metabolism, and myocardial remodeling were dysregulated. Additionally, we observed dysregulation of the retinoid homeostasis pathway, including significant post-radiation decreases in retinoic acid, an active metabolite of vitamin A. Significant differences between left and right ventricles in the pathology of radiation-induced injury were identified. This multi-omic study characterizes the natural history and molecular mechanisms of radiation-induced heart injury in NHP exposed to PBI with minimal bone marrow sparing.
Collapse
Affiliation(s)
- Stephanie Zalesak-Kravec
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD
| | - Weiliang Huang
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD
| | - Pengcheng Wang
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD
| | - Jianshi Yu
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD
| | - Tian Liu
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD
| | - Amy E. Defnet
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD
| | - Alexander R. Moise
- Medical Sciences Division, Northern Ontario School of Medicine, Sudbury, ON, Canada; Departments of Chemistry and Biochemistry, and Biology and Biomolecular Sciences Program, Laurentian University, Sudbury, ON, Canada
| | - Ann M. Farese
- University of Maryland, School of Medicine, Department of Radiation Oncology, Baltimore, MD
| | - Thomas J. MacVittie
- University of Maryland, School of Medicine, Department of Radiation Oncology, Baltimore, MD
| | - Maureen A. Kane
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD
| |
Collapse
|
32
|
Aimo A, Panichella G, Barison A, Maffei S, Cameli M, Coiro S, D'Ascenzi F, Di Mario C, Liga R, Marcucci R, Morrone D, Olivotto I, Tritto I, Emdin M. Sex-related differences in ventricular remodeling after myocardial infarction. Int J Cardiol 2021; 339:62-69. [PMID: 34314766 DOI: 10.1016/j.ijcard.2021.07.036] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 07/11/2021] [Accepted: 07/15/2021] [Indexed: 11/24/2022]
Abstract
The epidemiology, clinical features and outcome of myocardial infarction (MI) display significant differences between men and women. Prominent sex differences have also been suggested in left ventricular (LV) remodeling after MI. Ventricular remodeling refers to a deterioration of LV geometry and function often leading to heart failure (HF) development and an increased risk of adverse cardiovascular events. Women have a lower propensity to the acquisition of a spherical geometry and LV dysfunction. These differences can be attributed at least partially to a lower frequency of transmural infarction and smaller areas of microvascular obstruction in women, as well as to a less prominent activation of neuroendocrine systems and apoptotic, inflammatory and profibrotic pathways in women. Estrogens might play a role in this difference, which could partially persist even after the menopause because of a persisting intramyocardial synthesis of estrogens in women. Conversely, androgens may exert a detrimental influence. Future studies should better clarify sex differences in the predictors, clinical correlates, prognostic impact and disease mechanisms of remodeling, as well as the existence of sex-specific therapeutic targets. This research effort should hopefully allow to optimize the treatment of MI during the acute and post-acute phase, possibly through different therapeutic strategies in men and women, with the goal of reducing the risk of HF development and improving patient outcome.
Collapse
Affiliation(s)
- Alberto Aimo
- Istituto di Scienze della Vita, Scuola Superiore Sant'Anna, Pisa, Italy; Fondazione Toscana Gabriele Monasterio, Pisa, Italy.
| | | | - Andrea Barison
- Istituto di Scienze della Vita, Scuola Superiore Sant'Anna, Pisa, Italy; Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | | | - Matteo Cameli
- Department of Medical Biotechnologies, Section of Cardiology, University of Siena, Italy
| | - Stefano Coiro
- Division of Cardiology, University of Perugia, Italy
| | - Flavio D'Ascenzi
- Department of Medical Biotechnologies, Section of Cardiology, University of Siena, Italy
| | - Carlo Di Mario
- Structural Interventional Cardiology, Careggi University Hospital, Florence, Italy
| | - Riccardo Liga
- Cardio-Thoracic and Vascular Department, University Hospital, Pisa, Italy
| | - Rossella Marcucci
- Experimental and Clinical Medicine, University of Florence, Atherothrombotic Center, AOU Careggi, Florence, Italy
| | - Doralisa Morrone
- Cardio-Thoracic and Vascular Department, University Hospital, Pisa, Italy
| | - Iacopo Olivotto
- Cardiomiopathy Unit, AOU Careggi, Florence, Italy. Società Italiana di Cardiologia, Sezione Regionale Tosco-Umbra
| | | | - Michele Emdin
- Istituto di Scienze della Vita, Scuola Superiore Sant'Anna, Pisa, Italy; Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| |
Collapse
|
33
|
Paredes A, Santos-Clemente R, Ricote M. Untangling the Cooperative Role of Nuclear Receptors in Cardiovascular Physiology and Disease. Int J Mol Sci 2021; 22:ijms22157775. [PMID: 34360540 PMCID: PMC8346021 DOI: 10.3390/ijms22157775] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/13/2021] [Accepted: 07/16/2021] [Indexed: 12/12/2022] Open
Abstract
The heart is the first organ to acquire its physiological function during development, enabling it to supply the organism with oxygen and nutrients. Given this early commitment, cardiomyocytes were traditionally considered transcriptionally stable cells fully committed to contractile function. However, growing evidence suggests that the maintenance of cardiac function in health and disease depends on transcriptional and epigenetic regulation. Several studies have revealed that the complex transcriptional alterations underlying cardiovascular disease (CVD) manifestations such as myocardial infarction and hypertrophy is mediated by cardiac retinoid X receptors (RXR) and their partners. RXRs are members of the nuclear receptor (NR) superfamily of ligand-activated transcription factors and drive essential biological processes such as ion handling, mitochondrial biogenesis, and glucose and lipid metabolism. RXRs are thus attractive molecular targets for the development of effective pharmacological strategies for CVD treatment and prevention. In this review, we summarize current knowledge of RXR partnership biology in cardiac homeostasis and disease, providing an up-to-date view of the molecular mechanisms and cellular pathways that sustain cardiomyocyte physiology.
Collapse
|
34
|
Xi H, He Z, Lv C. FOXG1 improves mitochondrial function and promotes the progression of nasopharyngeal carcinoma. Mol Med Rep 2021; 24:651. [PMID: 34278485 PMCID: PMC8299199 DOI: 10.3892/mmr.2021.12290] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 05/14/2021] [Indexed: 12/28/2022] Open
Abstract
Forkhead‑box gene 1 (FOXG1) has been reported to serve an important role in various malignancies, but its effects on nasopharyngeal cancer (NPC) remain unknown. Thus, the present study aimed to investigate the specific regulatory relationship between FOXG1 and NPC progression. Tumor tissues and matching para‑carcinoma tissues were obtained from patients with NPC. Small interfering (si)RNA‑FOXG1 and pcDNA3.1‑FOXG1 were transfected into SUNE‑1 and C666‑1 cells to knockdown and overexpress FOXG1 expression, respectively. FOXG1 expression was detected using reverse transcription‑quantitative PCR and immunohistochemistry. Cell proliferation was detected using MTT and 5‑ethynyl‑20‑deoxyuridine assays. Transwell invasion assay, wound healing assay and flow cytometry were used to detect cell invasion, migration and apoptosis, respectively. Western blotting was conducted to detect the expression levels of mitochondrial markers (succinate dehydrogenase complex flavoprotein subunit A, heat shock protein 60 and pyruvate dehydrogenase), epithelial‑mesenchymal transition (EMT) related proteins (N‑cadherin, Snail and E‑cadherin) and apoptosis‑related proteins [Bax, Bcl‑2, poly(ADP‑ribose) polymerase 1 (PARP), cleaved PARP, cleaved caspase‑3, cleaved caspase‑8, cleaved caspase‑9, caspase‑3, caspase‑8 and caspase‑9]. The mitochondrial membrane potential was detected via flow cytometry, while the ATP/ADP ratio was determined using the ADP/ATP ratio assay kit. The present results demonstrated that FOXG1 expression was upregulated in NPC tissues and cells, and was associated with distant metastasis and TNM stage. Moreover, knockdown of FOXG1 inhibited the proliferation, migration, invasion, EMT and mitochondrial function of SUNE‑1 cells, as well as promoted cell apoptosis, while the opposite results were observed in C666‑1 cells. In conclusion, FOXG1 enhanced proliferation, migration and invasion, induced EMT and improved mitochondrial function in NPC cells. The current findings provide an adequate theoretical basis for the treatment of NPC.
Collapse
Affiliation(s)
- Huajun Xi
- Department of Otolaryngology and Stomatology, Shouguang People's Hospital, Shouguang, Shandong 262700, P.R. China
| | - Zhengxiang He
- Department of Otolaryngology and Maxillofacial Surgery, Wuwei People's Hospital, Wuwei, Gansu 733000, P.R. China
| | - Cao Lv
- Department of Otolaryngology, Head and Neck Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| |
Collapse
|
35
|
Takioku M, Takamura Y, Fujihara M, Watanabe M, Yamada S, Kawasaki M, Ito S, Nakano S, Kakuta H. Creation of Fluorescent RXR Antagonists Based on CBTF-EE and Application to a Fluorescence Polarization Binding Assay. ACS Med Chem Lett 2021; 12:1024-1029. [PMID: 34141088 DOI: 10.1021/acsmedchemlett.1c00201] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 05/05/2021] [Indexed: 01/08/2023] Open
Abstract
Retinoid X receptor (RXR) ligands often bind in modes in which the carboxy group forms a hydrogen bond inside the ligand-binding pocket (LBP). However, our previously reported RXR antagonist, CBTF-EE (4a), binds with its carboxy group directed outside the LBP and its alkoxy side chain located inside the LBP. Here, we examined the binding modes of 4b and 4c bearing a nitrobenzoxadiazole (NBD) or boron-dipyrromethene (BODIPY) fluorophore, respectively, at the end of the alkoxy chain of 4a. Both compounds function as RXR antagonists. 4c, but not 4b, was available for a fluorescence polarization binding assay, indicating that rotation of BODIPY, but not NBD, is restricted in the bound state. The fluorescence findings, supported by docking simulations, suggest the fluorophores are located outside the LBP, so that the binding mode of 4b and 4c is different from that of 4a. The assay results were highly correlated with those of a [3H]9-cis-retinoic acid assay.
Collapse
Affiliation(s)
- Maho Takioku
- Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 1-1-1, Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
| | - Yuta Takamura
- Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 1-1-1, Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
| | - Michiko Fujihara
- Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 1-1-1, Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
- AIBIOS Co. Ltd., Tri-Seven Roppongi 8F 7-7-7 Roppongi, Minato-ku, Tokyo 106-0032, Japan
| | - Masaki Watanabe
- Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 1-1-1, Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
| | - Shoya Yamada
- Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 1-1-1, Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
- Research Fellowship Division, Japan Society for the Promotion of Science, Sumitomo-Ichibancho FS Bldg., 8 Ichibancho, Chiyoda-ku, Tokyo 102-8472, Japan
| | - Mayu Kawasaki
- Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52- 1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Sohei Ito
- Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52- 1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Shogo Nakano
- Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52- 1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Hiroki Kakuta
- Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 1-1-1, Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
| |
Collapse
|
36
|
LXR directly regulates glycosphingolipid synthesis and affects human CD4+ T cell function. Proc Natl Acad Sci U S A 2021; 118:2017394118. [PMID: 34006637 DOI: 10.1073/pnas.2017394118] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The liver X receptor (LXR) is a key transcriptional regulator of cholesterol, fatty acid, and phospholipid metabolism. Dynamic remodeling of immunometabolic pathways, including lipid metabolism, is a crucial step in T cell activation. Here, we explored the role of LXR-regulated metabolic processes in primary human CD4+ T cells and their role in controlling plasma membrane lipids (glycosphingolipids and cholesterol), which strongly influence T cell immune signaling and function. Crucially, we identified the glycosphingolipid biosynthesis enzyme glucosylceramide synthase as a direct transcriptional LXR target. LXR activation by agonist GW3965 or endogenous oxysterol ligands significantly altered the glycosphingolipid:cholesterol balance in the plasma membrane by increasing glycosphingolipid levels and reducing cholesterol. Consequently, LXR activation lowered plasma membrane lipid order (stability), and an LXR antagonist could block this effect. LXR stimulation also reduced lipid order at the immune synapse and accelerated activation of proximal T cell signaling molecules. Ultimately, LXR activation dampened proinflammatory T cell function. Finally, compared with responder T cells, regulatory T cells had a distinct pattern of LXR target gene expression corresponding to reduced lipid order. This suggests LXR-driven lipid metabolism could contribute to functional specialization of these T cell subsets. Overall, we report a mode of action for LXR in T cells involving the regulation of glycosphingolipid and cholesterol metabolism and demonstrate its relevance in modulating T cell function.
Collapse
|
37
|
Abstract
The thrombospondin family comprises of five multifunctional glycoproteins, whose best-studied member is thrombospondin 1 (TSP1). This matricellular protein is a potent antiangiogenic agent that inhibits endothelial migration and proliferation, and induces endothelial apoptosis. Studies have demonstrated a regulatory role of TSP1 in cell migration and in activation of the latent transforming growth factor beta 1 (TGFβ1). These functions of TSP1 translate into its broad modulation of immune processes. Further, imbalances in immune regulation have been increasingly linked to pathological conditions such as obesity and diabetes mellitus. While most studies in the past have focused on the role of TSP1 in cancer and inflammation, recently published data have revealed new insights about the role of TSP1 in physiological and metabolic disorders. Here, we highlight recent findings that associate TSP1 and its receptors to obesity, diabetes, and cardiovascular diseases. TSP1 regulates nitric oxide, activates latent TGFβ1, and interacts with receptors CD36 and CD47, to play an important role in cell metabolism. Thus, TSP1 and its major receptors may be considered a potential therapeutic target for metabolic diseases.
Collapse
Affiliation(s)
- Linda S. Gutierrez
- Department of Biology, Wilkes University, Wilkes Barre, PA, United States
| | | |
Collapse
|
38
|
Zhou Z, Liu Z, Gao X, Long Q. Mitochondrial respiration in C57BL/6 substrains varies in response to myocardial infarction. J Bioenerg Biomembr 2021; 53:119-127. [PMID: 33630237 DOI: 10.1007/s10863-021-09884-6] [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: 09/09/2020] [Accepted: 02/18/2021] [Indexed: 10/22/2022]
Abstract
The C57BL/6 mouse strain have been commonly used for the genetic background animal models and experimental research. There are several major sources of C57BL/6 substrains for the biomedical research community which display genetic and phenotypic differences. Previous studies have suggested that the varies in baseline of cardiovascular phenotypes as well as in response to pressure overload by transverse aortic constriction (TAC). To investigate whether there exist substrain specific differences in response to heart failure post myocardial infarction (MI), consequently the impaired mitochondrial respiration, we performed MI surgery on two commonly used C57BL/6 substrains: C57BL/6J (BL/6J) and C57BL/6NCrl (BL/6N) mice. Subsequently, measurements about cardiac function, histology and mitochondrial respiration capacities were conducted to evaluate the differences. The data showed that C57BL/6J(BL/6J) mice is more resistant to the attack of MI, evidenced by lower mortality, less infarct size and better preserved cardiac function after MI, especially exhibited better mitochondrial respiration capacities, compared with the C57BL/6NCrl(BL/6N) mice.
Collapse
Affiliation(s)
- Zhou Zhou
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
| | - Zhiheng Liu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
| | - Xu Gao
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
| | - Qinqiang Long
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China.
| |
Collapse
|
39
|
Xiao T, Huang J, Liu Y, Zhao Y, Wei M. Matrine Protects Cardiomyocytes Against Hyperglycemic Stress by Promoting Mitofusin 2-Induced Mitochondrial Fusion. Front Physiol 2021; 11:597429. [PMID: 33613300 PMCID: PMC7888534 DOI: 10.3389/fphys.2020.597429] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 12/07/2020] [Indexed: 12/13/2022] Open
Abstract
Matrine, an active component of Sophora flavescens Ait root extracts, has been used in China for years to treat cancer and viral hepatitis. In the present study, we explored the effects of matrine on hyperglycemia-treated cardiomyocytes. Cardiomyocyte function, oxidative stress, cellular viability, and mitochondrial fusion were assessed through immunofluorescence, quantitative real-time PCR (qRT-PCR), enzyme-linked immunosorbent assays, and RNA interference. Matrine treatment suppressed hyperglycemia-induced oxidative stress in cardiomyocytes by upregulating transcription of nuclear factor erythroid 2-like 2 and heme oxygenase-1. Matrine also improved cardiomyocyte contractile and relaxation function during hyperglycemia, and it reduced hyperglycemia-induced cardiomyocyte death by inhibiting mitochondrial apoptosis. Matrine treatment increased the transcription of mitochondrial fusion-related genes and thus attenuated the proportion of fragmented mitochondria in cardiomyocytes. Inhibiting mitochondrial fusion by knocking down mitofusin 2 (Mfn2) abolished the cardioprotective effects of matrine during hyperglycemia. These results demonstrate that matrine could be an effective drug to alleviate hyperglycemia-induced cardiomyocyte damage by activating Mfn2-induced mitochondrial fusion.
Collapse
Affiliation(s)
- Tong Xiao
- Endocrinology and Geriatric Department, Henan Provincial Chest Hospital, Zhengzhou, China
| | - Jie Huang
- Department of Ultrasonography, Affiliated Tumor Hospital of Zhengzhou University (Henan Cancer Hospital), Zhengzhou, China
| | - Yuan Liu
- Endocrinology and Geriatric Department, Henan Provincial Chest Hospital, Zhengzhou, China
| | - Yujie Zhao
- Endocrinology and Geriatric Department, Henan Provincial Chest Hospital, Zhengzhou, China
| | - Manman Wei
- Department of Cardiovascular, Henan Provincial Chest Hospital, Zhengzhou, China
| |
Collapse
|
40
|
Daseke MJ, Chalise U, Becirovic-Agic M, Salomon JD, Cook LM, Case AJ, Lindsey ML. Neutrophil signaling during myocardial infarction wound repair. Cell Signal 2021; 77:109816. [PMID: 33122000 PMCID: PMC7718402 DOI: 10.1016/j.cellsig.2020.109816] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/21/2020] [Accepted: 10/21/2020] [Indexed: 12/12/2022]
Abstract
Neutrophils are key effector cells of the innate immune system, serving as a first line of defense in the response to injury and playing essential roles in the wound healing process. Following myocardial infarction (MI), neutrophils infiltrate into the infarct region to propagate inflammation and begin the initial phase of cardiac wound repair. Pro-inflammatory neutrophils release proteases to degrade extracellular matrix (ECM), a necessary step for the removal of necrotic myocytes as a prelude for scar formation. Neutrophils transition their phenotype over time to regulate MI inflammation resolution and stabilize scar formation. Neutrophils contribute to the evolution from inflammation to resolution and scar formation by serving anti-inflammatory and repair functions. As anti-inflammatory cells, neutrophils contribute ECM proteins during scar formation, in particular fibronectin, galectin-3, and vimentin. The diverse and polarizing functions that contribute to MI wound repair make this innate immune cell a viable target to improve MI outcomes. Thus, understanding the signaling involved in neutrophil physiology in the context of MI may help to identify novel therapeutic targets.
Collapse
Affiliation(s)
- Michael J Daseke
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE 68198, USA; Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, USA; Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68198, USA
| | - Upendra Chalise
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE 68198, USA; Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68198, USA
| | - Mediha Becirovic-Agic
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Jeffrey D Salomon
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE 68198, USA; Departments of Pediatrics, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Leah M Cook
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Division of Pediatric Critical Care, Center for Heart and Vascular Research, Omaha, NE 68198, USA
| | - Adam J Case
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Merry L Lindsey
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE 68198, USA; Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68198, USA.
| |
Collapse
|
41
|
Zhang Q, Cheng X, Zhang H, Zhang T, Wang Z, Zhang W, Yu W. Dissecting molecular mechanisms underlying H 2O 2-induced apoptosis of mouse bone marrow mesenchymal stem cell: role of Mst1 inhibition. Stem Cell Res Ther 2020; 11:526. [PMID: 33298178 PMCID: PMC7724846 DOI: 10.1186/s13287-020-02041-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 11/19/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Bone marrow mesenchymal stem cell (BM-MSC) has been shown to treat pulmonary arterial hypertension (PAH). However, excessive reactive oxygen species (ROS) increases the apoptosis of BM-MSCs, leading to poor survival and engraft efficiency. Thus, improving the ability of BM-MSCs to scavenge ROS may considerably enhance the effectiveness of transplantation therapy. Mammalian Ste20-like kinase 1 (Mst1) is a pro-apoptotic molecule which increases ROS production. The aim of this study is to uncover the underlying mechanisms the effect of Mst1 inhibition on the tolerance of BM-MSCs under H2O2 condition. METHODS Mst1 expression in BM-MSCs was inhibited via transfection with adenoviruses expressing a short hairpin (sh) RNA directed against Mst1 (Ad-sh-Mst1) and exposure to H2O2. Cell viability was detected by Cell Counting Kit 8 (CCK-8) assay, and cell apoptosis was analyzed by Annexin V-FITC/PI, Caspase 3 Activity Assay kits, and pro caspase 3 expression. ROS level was evaluated by the ROS probe DCFH-DA, mitochondrial membrane potential (ΔΨm) assay, SOD1/2, CAT, and GPx expression. Autophagy was assessed using transmission electron microscopy, stubRFP-sensGFP-LC3 lentivirus, and autophagy-related protein expression. The autophagy/Keap1/Nrf2 signal in H2O2-treated BM-MSC/sh-Mst1 was also measured. RESULTS Mst1 inhibition reduced ROS production; increased antioxidant enzyme SOD1/2, CAT, and GPx expression; maintained ΔΨm; and alleviated cell apoptosis in H2O2-treated BM-MSCs. In addition, this phenomenon was closely correlated with the autophagy/Keap1/Nrf2 signal pathway. Moreover, the antioxidant pathway Keap1/Nrf2 was also blocked when autophagy was inhibited by the autophagy inhibitor 3-MA. However, Keap1 or Nrf2 knockout via siRNA had no effect on autophagy activation or suppression. CONCLUSION Mst1 inhibition mediated the cytoprotective action of mBM-MSCs against H2O2-induced oxidative stress injury. The underlying mechanisms involve autophagy activation and the Keap1/Nrf2 signal pathway.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250062, Shandong, China
| | - Xianfeng Cheng
- Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250062, Shandong, China.,Department of Cardiovascular Surgery, Weifang People's Hospital, Weifang, 261000, Shandong, China
| | - Haizhou Zhang
- Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250062, Shandong, China
| | - Tao Zhang
- Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250062, Shandong, China
| | - Zhengjun Wang
- Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250062, Shandong, China
| | - Wenlong Zhang
- Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250062, Shandong, China
| | - Wancheng Yu
- Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250062, Shandong, China.
| |
Collapse
|
42
|
Shabbir A, Rathod KS, Khambata RS, Ahluwalia A. Sex Differences in the Inflammatory Response: Pharmacological Opportunities for Therapeutics for Coronary Artery Disease. Annu Rev Pharmacol Toxicol 2020; 61:333-359. [PMID: 33035428 DOI: 10.1146/annurev-pharmtox-010919-023229] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Coordinated molecular responses are key to effective initiation and resolution of both acute and chronic inflammation. Vascular inflammation plays an important role in initiating and perpetuating atherosclerotic disease, specifically at the site of plaque and subsequent fibrous cap rupture. Both men and women succumb to this disease process, and although management strategies have focused on revascularization and pharmacological therapies in the acute situation to reverse vessel closure and prevent thrombogenesis, data now suggest that regulation of host inflammation may improve both morbidity and mortality, thus supporting the notion that prevention is better than cure. There is a clear sex difference in the incidence of vascular disease, and data confirm biological differences in inflammatory initiation and resolution between men and women. This article reviews contemporary opinions describing the sex difference in the initiation and resolution of inflammatory responses, with a view to explore potential targets for pharmacological intervention.
Collapse
Affiliation(s)
- Asad Shabbir
- The William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom;
| | - Krishnaraj Sinhji Rathod
- The William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom;
| | - Rayomand Syrus Khambata
- The William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom;
| | - Amrita Ahluwalia
- The William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom;
| |
Collapse
|
43
|
Lindsey ML, Deleon-Pennell KY, Bradshaw AD, Larue RAC, Anderson DR, Thiele GM, Baicu CF, Jones JA, Menick DR, Zile MR, Spinale FG. Focusing Heart Failure Research on Myocardial Fibrosis to Prioritize Translation. J Card Fail 2020; 26:876-884. [PMID: 32446948 PMCID: PMC7584737 DOI: 10.1016/j.cardfail.2020.05.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 05/06/2020] [Accepted: 05/12/2020] [Indexed: 01/05/2023]
Abstract
Heart failure (HF) has traditionally been defined by symptoms of fluid accumulation and poor perfusion, but it is now recognized that specific HF classifications hold prognostic and therapeutic relevance. Specifically, HF with reduced ejection fraction is characterized by reduced left ventricular systolic pump function and dilation and HF with preserved ejection fraction is characterized primarily by abnormal left ventricular filling (diastolic failure) with relatively preserved left ventricular systolic function. These forms of HF are distributed equally among patients with HF and likely require distinctly different strategies to mitigate the morbidity, mortality, and medical resource utilization of this disease. In particular, HF is a significant medical issue within the US Department of Veterans Affairs (VA) hospital system and constitutes a major translational research priority for the VA. Because a common underpinning of both HF with reduced ejection fraction and HF with preserved ejection fraction seems to be changes in the structure and function of the myocardial extracellular matrix, a conference was convened sponsored by the VA, entitled, "Targeting Myocardial Fibrosis in Heart Failure" to explore the extracellular matrix as a potential therapeutic target and to propose specific research directions. The conference was conceptually framed around the hypothesis that although HF with reduced ejection fraction and HF with preserved ejection fraction clearly have distinct mechanisms, they may share modifiable pathways and biological mediators in common. Inflammation and extracellular matrix were identified as major converging themes. A summary of our discussion on unmet challenges and possible solutions to move the field forward, as well as recommendations for future research opportunities, are provided.
Collapse
Affiliation(s)
- Merry L Lindsey
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska; Research Service, Nebraska-Western Iowa Health Care System, Omaha, Nebraska.
| | - Kristine Y Deleon-Pennell
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina; Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| | - Amy D Bradshaw
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina; Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| | - R Amanda C Larue
- Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina; Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Daniel R Anderson
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Geoffrey M Thiele
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, Nebraska; Division of Rheumatology and Immunology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Catalin F Baicu
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Jeffrey A Jones
- Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina; Department of Surgery, Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Donald R Menick
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina; Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| | - Michael R Zile
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina; Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| | - Francis G Spinale
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine, Columbia, SC and William Jennings Bryan Dorn VA Medical Center, Columbia, South Carolina
| |
Collapse
|
44
|
Eicosanoid production varies by sex in mesenteric ischemia reperfusion injury. Clin Immunol 2020; 220:108596. [PMID: 32961332 DOI: 10.1016/j.clim.2020.108596] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 09/13/2020] [Accepted: 09/16/2020] [Indexed: 01/18/2023]
Abstract
Intestinal ischemia/reperfusion (I/R)-induced injury is an inflammatory response with significant morbidity and mortality. The early inflammatory response includes neutrophil infiltration. However, the majority of rodent studies utilize male mice despite a sexual dimorphism in intestinal I/R-related diseases. We hypothesized that sex may alter inflammation by changing neutrophil infiltration and eicosanoid production. To test this hypothesis, male and female C57Bl/6 mice were subjected to sham treatment or 30 min intestinal ischemia followed by a time course of reperfusion. We demonstrate that compared to male mice, females sustain significantly less intestinal I/R-induced tissue damage and produced significant LTB4 concentrations. Male mice release PGE2. Finally, treatment with a COX-2 specific inhibitor, NS-398, attenuated I/R-induced injury, total peroxidase level, and PGE2 production in males, but not in similarly treated female mice. Thus, I/R-induced eicosanoid production and neutrophil infiltration varies between sexes suggesting that distinct therapeutic intervention may be needed in clinical ischemic diseases.
Collapse
|
45
|
Hanna A, Shinde AV, Frangogiannis NG. Validation of diagnostic criteria and histopathological characterization of cardiac rupture in the mouse model of nonreperfused myocardial infarction. Am J Physiol Heart Circ Physiol 2020; 319:H948-H964. [PMID: 32886000 DOI: 10.1152/ajpheart.00318.2020] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
In patients with myocardial infarction (MI), cardiac rupture is an uncommon but catastrophic complication. In the mouse model of nonreperfused MI, reported rupture rates are highly variable and depend not only on the genetic background and sex of animals but also on the method used for documentation of rupture. In most studies, diagnosis of cardiac rupture is based on visual inspection during autopsy; however, criteria are poorly defined. We performed systematic histopathological analysis of whole hearts from C57BL/6J mice dying after nonreperfused MI and evaluated the reliability of autopsy-based criteria in identification of rupture. Moreover, we compared the cell biological environment of the infarct between rupture-related and rupture-independent deaths. Histopathological analysis documented rupture in 50% of mice dying during the first week post-MI. Identification of a gross rupture site was highly specific but had low sensitivity; in contrast, hemothorax had high sensitivity but low specificity. Mice with rupture had lower myofibroblast infiltration, accentuated macrophage influx, and a trend toward reduced collagen content in the infarct. Male mice had increased mortality and higher incidence of rupture. However, infarct myeloid cells harvested from male and female mice at the peak of the incidence of rupture had comparable inflammatory gene expression. In conclusion, the reliability of autopsy in documentation of rupture in infarcted mice is dependent on the specific criteria used. Macrophage-driven inflammation and reduced activation of collagen-secreting reparative myofibroblasts may be involved in the pathogenesis of post-MI cardiac rupture.NEW & NOTEWORTHY We show that cardiac rupture accounts for 50% of deaths in C57BL/6J mice undergoing nonreperfused myocardial infarction protocols. Overestimation of rupture events in published studies likely reflects the low specificity of hemothorax as a criterion for documentation of rupture. In contrast, identification of a gross rupture site has high specificity and low sensitivity. We also show that mice dying of rupture have increased macrophage influx and attenuated myofibroblast infiltration in the infarct. These findings are consistent with a role for perturbations in the balance between inflammatory and reparative responses in the pathogenesis of postinfarction cardiac rupture. We also report that the male predilection for rupture in infarcted mice is not associated with increased inflammatory activation of myeloid cells.
Collapse
Affiliation(s)
- Anis Hanna
- Division of Cardiology, Department of Medicine, The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York
| | - Arti V Shinde
- Division of Cardiology, Department of Medicine, The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York
| | - Nikolaos G Frangogiannis
- Division of Cardiology, Department of Medicine, The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
46
|
Bradshaw AD, DeLeon-Pennell KY. T-cell regulation of fibroblasts and cardiac fibrosis. Matrix Biol 2020; 91-92:167-175. [PMID: 32438054 PMCID: PMC7434661 DOI: 10.1016/j.matbio.2020.04.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 03/30/2020] [Accepted: 04/13/2020] [Indexed: 02/06/2023]
Abstract
Inflammation contributes to the development of heart failure (HF) through multiple mechanisms including regulating extracellular matrix (ECM) degradation and deposition. Interactions between cells in the myocardium orchestrates the magnitude and duration of inflammatory cell recruitment and ECM remodeling events associated with HF. More recently, studies have shown T-cells have signficant roles in post-MI wound healing. T-cell biology in HF illustrates the complexity of cross-talk between inflammatory cell types and resident fibroblasts. This review will focus on T-cell recruitment to the myocardium and T-cell specific factors that might influence cardiac wound healing and fibrosis in the heart with consideration of age and sex as important factors in T-cell activity.
Collapse
Affiliation(s)
- Amy D Bradshaw
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, Ralph H. Johnson Veterans Affairs Medical Center, 109 Bee Street Charleston, SC 29401, United States
| | - Kristine Y DeLeon-Pennell
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, Ralph H. Johnson Veterans Affairs Medical Center, 109 Bee Street Charleston, SC 29401, United States.
| |
Collapse
|
47
|
Li C, Tan Y, Wu J, Ma Q, Bai S, Xia Z, Wan X, Liang J. Resveratrol Improves Bnip3-Related Mitophagy and Attenuates High-Fat-Induced Endothelial Dysfunction. Front Cell Dev Biol 2020; 8:796. [PMID: 32923443 PMCID: PMC7457020 DOI: 10.3389/fcell.2020.00796] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 07/28/2020] [Indexed: 12/16/2022] Open
Abstract
Statin treatment reduces cardiovascular risk. However, individuals with well-controlled low-density lipoprotein (LDL) levels may remain at increased risk owing to persistent high triglycerides and low high-density lipoprotein cholesterol. Because resveratrol promotes glucose metabolism and mitigates cardiovascular disorders, we explored its mechanism of protective action on high-fat-induced endothelial dysfunction. Human umbilical venous endothelial cells were treated with oxidized LDL (ox-LDL) in vitro. Endothelial function, cell survival, proliferation, migration, and oxidative stress were analyzed through western blots, quantitative polymerase chain reaction, ELISA, and immunofluorescence. ox-LDL induced endothelial cell apoptosis, proliferation arrest, and mobilization inhibition, all of which resveratrol reduced. ox-LDL suppressed the activities of mitochondrial respiration complex I and III and reduced levels of intracellular antioxidative enzymes, resulting in reactive oxygen species overproduction and mitochondrial dysfunction. Resveratrol treatment upregulated Bnip3-related mitophagy and prevented ox-LDL-mediated mitochondrial respiration complexes inactivation, sustaining mitochondrial membrane potential and favoring endothelial cell survival. We found that resveratrol enhanced Bnip3 transcription through hypoxia-inducible factor 1 (HIF1) and 5' AMP-activated protein kinase (AMPK). Inhibition of AMPK and HIF1 abolished resveratrol-mediated protection of mitochondrial redox balance and endothelial viability. Together, these data demonstrate resveratrol reduces hyperlipemia-related endothelial damage by preserving mitochondrial homeostasis.
Collapse
Affiliation(s)
- Chen Li
- Department of Cardiology, Foshan Hospital Affiliated with Southern Medical University (The Second People's Hospital of Foshan), Foshan, China
| | - Ying Tan
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiandi Wu
- Department of Cardiology, Foshan Hospital Affiliated with Southern Medical University (The Second People's Hospital of Foshan), Foshan, China
| | - Qinghui Ma
- Department of Oncology Hematology, Foshan Hospital Affiliated with Southern Medical University (The Second People's Hospital of Foshan), Foshan, China
| | - Shuchang Bai
- Department of Cardiology, Foshan Hospital Affiliated with Southern Medical University (The Second People's Hospital of Foshan), Foshan, China
| | - Zhangqing Xia
- Department of Cardiology, Foshan Hospital Affiliated with Southern Medical University (The Second People's Hospital of Foshan), Foshan, China
| | - Xiaoliang Wan
- Department of Cardiology, Foshan Hospital Affiliated with Southern Medical University (The Second People's Hospital of Foshan), Foshan, China
| | - Jianqiu Liang
- Department of Cardiology, Foshan Hospital Affiliated with Southern Medical University (The Second People's Hospital of Foshan), Foshan, China
| |
Collapse
|
48
|
Wang J, Zhu P, Toan S, Li R, Ren J, Zhou H. Pum2-Mff axis fine-tunes mitochondrial quality control in acute ischemic kidney injury. Cell Biol Toxicol 2020; 36:365-378. [PMID: 31993882 DOI: 10.1007/s10565-020-09513-9] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 01/24/2020] [Indexed: 12/31/2022]
Abstract
Mitochondrial fission factor (Mff) has been demonstrated to play a role in the activation of mitochondrial cleavage and mitochondrial death, denoting its role in the regulation of mitochondrial quality control. Recent evidence suggested that the mRNA translation of Mff is under the negative regulation by the RNA-binding protein Pumilio2 (Pum2). This study was designed to examine the role of Pum2 and Mff in the governance of mitochondrial quality control in a murine model of acute ischemic kidney injury. Our results indicated that genetic deletion of Mff overtly attenuated ischemic acute kidney injury (AKI)-induced renal failure through inhibition of pro-inflammatory response, tubular oxidative stress, and ultimately cell death in the kidney. Furthermore, Mff inhibition effectively preserved mitochondrial homeostasis through amelioration of mitochondrial mitosis, restoration of Sirt1/3 expression, and boost of mitochondrial respiration. Western blot analysis revealed that levels of Pum2 were significantly downregulated by ischemic AKI, inversely coinciding with levels of Mff. Overexpression of Pum2 reduced ischemic AKI-mediated Mff upregulation and offered protection on renal tubules through modulation of mitochondrial quality control. Taken together, our data have unveiled the molecular mechanism of the Pum2-Mff axis in mitochondrial quality control in a mouse model of ischemic AKI. These data indicated the therapeutic potential of Pum2 activation and Mff inhibition in the management of ischemic AKI.
Collapse
Affiliation(s)
- Jin Wang
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, China
| | - Pingjun Zhu
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, China
| | - Sam Toan
- Department of Chemical Engineering, University of Minnesota-Duluth, Duluth, MN, 55812, USA
| | - Ruibing Li
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, China
| | - Jun Ren
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming, Laramie, WY, 82071, USA.
| | - Hao Zhou
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, China.
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming, Laramie, WY, 82071, USA.
| |
Collapse
|
49
|
Daseke MJ, Tenkorang-Impraim MAA, Ma Y, Chalise U, Konfrst SR, Garrett MR, DeLeon-Pennell KY, Lindsey ML. Exogenous IL-4 shuts off pro-inflammation in neutrophils while stimulating anti-inflammation in macrophages to induce neutrophil phagocytosis following myocardial infarction. J Mol Cell Cardiol 2020; 145:112-121. [PMID: 32574573 PMCID: PMC7483959 DOI: 10.1016/j.yjmcc.2020.06.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 06/08/2020] [Accepted: 06/11/2020] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Macrophages and neutrophils are primary leukocytes involved in the inflammatory response to myocardial infarction (MI). While interleukin (IL)-4 is an in vitro anti-inflammatory stimulus, the MI myocardium does not express a considerable amount of IL-4 but does express IL4 receptors. We hypothesized that continuous exogenous IL-4 infusion starting 24 h after MI would promote a polarization switch in inflammatory cells towards a reparative phenotype. METHODS C57BL/6J male mice (3-6 months of age) were subcutaneously infused with either saline (n = 17) or IL-4 (20 ng/g/day; n = 17) beginning 24 h after MI and evaluated at MI day 3. RESULTS Macrophages and neutrophils were isolated ex vivo from the infarct region and examined. Exogenous IL-4 decreased pro-inflammatory Ccl3, Il12a, Tnfa, and Tgfb1 in neutrophils and increased anti-inflammatory Arg1 and Ym1 in macrophages (all p < .05). Tissue clearance by IL-4 treated neutrophils was not different, while selective phagocytosis of neutrophils doubled in IL-4 treated macrophages (p < .05). Of 24,339 genes examined by RNA-sequencing, 2042 genes were differentially expressed in macrophages from IL-4 stimulated infarct (all FDR p < .05). Pdgfc gene expression was ranked first, increasing 3-fold in macrophages stimulated with IL-4 (p = 1 × 10-9). Importantly, changes in macrophage physiology and transcriptome occurred in the absence of global LV effects. Bone marrow derived monocytes stimulated with mouse recombinant PDGF-CC protein (10 μg/ml) or PDGF-CC blocking antibody (200 ng/ml) did not change Arg1 or Ym1 expression, indicating the in vivo effect of IL-4 to stimulate macrophage anti-inflammatory gene expression was independent of PDGF-CC. CONCLUSIONS Our results indicate that exogenous IL-4 promotes inflammation resolution by turning off pro-inflammation in neutrophils while stimulating anti-inflammation in macrophages to mediate removal of apoptotic neutrophils.
Collapse
Affiliation(s)
- Michael J Daseke
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, USA; Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA; Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Mavis A A Tenkorang-Impraim
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, USA
| | - Yonggang Ma
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, USA
| | - Upendra Chalise
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, USA; Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Shelby R Konfrst
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, USA; Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Michael R Garrett
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Kristine Y DeLeon-Pennell
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC, USA; Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Merry L Lindsey
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, USA; Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE, USA.
| |
Collapse
|
50
|
Yang D, Liu HQ, Liu FY, Tang N, Guo Z, Ma SQ, An P, Wang MY, Wu HM, Yang Z, Fan D, Tang QZ. The Roles of Noncardiomyocytes in Cardiac Remodeling. Int J Biol Sci 2020; 16:2414-2429. [PMID: 32760209 PMCID: PMC7378633 DOI: 10.7150/ijbs.47180] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 06/16/2020] [Indexed: 02/07/2023] Open
Abstract
Cardiac remodeling is a common characteristic of almost all forms of heart disease, including cardiac infarction, valvular diseases, hypertension, arrhythmia, dilated cardiomyopathy and other conditions. It is not merely a simple outcome induced by an increase in the workload of cardiomyocytes (CMs). The remodeling process is accompanied by abnormalities of cardiac structure as well as disturbance of cardiac function, and emerging evidence suggests that a wide range of cells in the heart participate in the initiation and development of cardiac remodeling. Other than CMs, there are numerous noncardiomyocytes (non-CMs) that regulate the process of cardiac remodeling, such as cardiac fibroblasts and immune cells (including macrophages, lymphocytes, neutrophils, and mast cells). In this review, we summarize recent knowledge regarding the definition and significant effects of various non-CMs in the pathogenesis of cardiac remodeling, with a particular emphasis on the involved signaling mechanisms. In addition, we discuss the properties of non-CMs, which serve as targets of many cardiovascular drugs that reduce adverse cardiac remodeling.
Collapse
Affiliation(s)
- Dan Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| | - Han-Qing Liu
- Department of Thyroid and Breast, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
| | - Fang-Yuan Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| | - Nan Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| | - Zhen Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| | - Shu-Qing Ma
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| | - Peng An
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| | - Ming-Yu Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| | - Hai-Ming Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| | - Zheng Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| | - Di Fan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| |
Collapse
|