1
|
Veliz AL, Hughes L, Carrillo D, Pecaut MJ, Kearns-Jonker M. Immunization induces inflammation in the mouse heart during spaceflight. BMC Genomics 2025; 26:229. [PMID: 40065216 PMCID: PMC11892206 DOI: 10.1186/s12864-025-11426-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
Space travel is a growing area of interest and includes initiatives such as NASA's Moon-to-Mars Mission. Reports on the cardiovascular effects of space travel reveal changes in morphology, metabolism, and function of the cardiovascular system. In this study, the cardiovascular response to immunization in space was studied in mice which were housed and immunized while on the International Space Station (ISS). Mice were immunized with tetanus toxoid combined with the adjuvant CpG (TT + CpG) and the effects of vaccination in space were studied using transcriptomics. Analysis of the mouse heart transcriptome was performed on flight control and flight-immunized mice. The results show that immunization aboard the ISS stimulates heightened inflammation in the heart via induction of the nuclear factor kappa B (NF-κB) signaling pathway to promote the release of the pro-inflammatory cytokines IFNγ, IL-17 and IL-6. Additional transcriptomic changes included alterations in the cytoskeleton and in the expression of transcripts associated with protection from oxidative stress. In summary, inflammation in the heart can occur following immunization in space. This investigation explores the impact of immune challenges on the heart and lays the groundwork for future research into additional cardiac alterations which can occur during spaceflight.
Collapse
Affiliation(s)
- Alicia L Veliz
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Lorelei Hughes
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Delia Carrillo
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Michael J Pecaut
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Mary Kearns-Jonker
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.
| |
Collapse
|
2
|
Alkhatib B, Ciarelli J, Ghnenis A, Pallas B, Olivier N, Padmanabhan V, Vyas AK. Early- to mid-gestational testosterone excess leads to adverse cardiac outcomes in postpartum sheep. Am J Physiol Heart Circ Physiol 2024; 327:H315-H330. [PMID: 38819385 PMCID: PMC11687963 DOI: 10.1152/ajpheart.00763.2023] [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: 12/07/2023] [Revised: 05/13/2024] [Accepted: 05/22/2024] [Indexed: 06/01/2024]
Abstract
Cardiovascular dysfunctions complicate 10-20% of pregnancies, increasing the risk for postpartum mortality. Various gestational insults, including preeclampsia are reported to be associated with adverse maternal cardiovascular outcomes. One such insult, gestational hyperandrogenism increases the risk for preeclampsia and other gestational morbidities but its impact on postpartum maternal health is not well known. We hypothesize that gestational hyperandrogenism such as testosterone (T) excess will adversely impact the maternal heart in the postpartum period. Pregnant ewes were injected with T propionate from day 30 to day 90 of gestation (term 147 days). Three months postpartum, echocardiograms, plasma cytokine profiles, cardiac morphometric, and molecular analysis were conducted [control (C) n = 6, T-treated (T) n = 7 number of animals]. Data were analyzed by two-tailed Student's t test and Cohen's effect size (d) analysis. There was a nonsignificant large magnitude decrease in cardiac output (7.64 ± 1.27 L/min vs. 10.19 ± 1.40, P = 0.22, d = 0.81) and fractional shortening in the T ewes compared with C (35.83 ± 2.33% vs. 41.50 ± 2.84, P = 0.15, d = 0.89). T treatment significantly increased 1) left ventricle (LV) weight-to-body weight ratio (2.82 ± 0.14 g/kg vs. 2.46 ± 0.08) and LV thickness (14.56 ± 0.52 mm vs. 12.50 ± 0.75), 2) proinflammatory marker [tumor necrosis factor-alpha (TNF-α)] in LV (1.66 ± 0.35 vs. 1.06 ± 0.18), 3) LV collagen (Masson's Trichrome stain: 3.38 ± 0.35 vs. 1.49 ± 0.15 and Picrosirius red stain: 5.50 ± 0.32 vs. 3.01 ± 0.23), 4) markers of LV apoptosis, including TUNEL (8.3 ± 1.1 vs. 0.9 ± 0.18), bcl-2-associated X protein (Bax)+-to-b-cell lymphoma 2 (Bcl2)+ ratio (0.68 ± 0.30 vs. 0.13 ± 0.02), and cleaved caspase 3 (15.4 ± 1.7 vs. 4.4 ± 0.38). These findings suggest that gestational testosterone excess adversely programs the maternal LV, leading to adverse structural and functional consequences in the postpartum period.NEW & NOTEWORTHY Using a sheep model of human translational relevance, this study provides evidence that excess gestational testosterone exposure such as that seen in hyperandrogenic disorders adversely impacts postpartum maternal hearts.
Collapse
Affiliation(s)
- Bashar Alkhatib
- Department of Pediatrics, Washington University, St Louis, Missouri, United States
| | - Joseph Ciarelli
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, United States
| | - Adel Ghnenis
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, United States
| | - Brooke Pallas
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, Michigan, United States
| | - Nicholas Olivier
- Department of Veterinary Medicine, Michigan State University, Lansing, Michigan, United States
| | - Vasantha Padmanabhan
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, United States
| | - Arpita Kalla Vyas
- Department of Pediatrics, Washington University, St Louis, Missouri, United States
| |
Collapse
|
3
|
Parletta AC, Cerri GC, Gasparini CRB, Panico K, Vieira-Junior DN, Zacarias-Rodrigues LM, Senger N, de Almeida Silva A, Fevereiro M, Diniz GP, Irigoyen MCC, Barreto-Chaves MLM. Cardiac hypertrophy that affects hyperthyroidism occurs independently of the NLRP3 inflammasome. Pflugers Arch 2024; 476:1065-1075. [PMID: 38679646 DOI: 10.1007/s00424-024-02965-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 03/26/2024] [Accepted: 04/16/2024] [Indexed: 05/01/2024]
Abstract
Cardiac hypertrophy (CH) is an adaptive response to maintain cardiac function; however, persistent stress responses lead to contractile dysfunction and heart failure. Although inflammation is involved in these processes, the mechanisms that control cardiac inflammation and hypertrophy still need to be clarified. The NLRP3 inflammasome is a cytosolic multiprotein complex that mediates IL-1β production. The priming step of NLRP3 is essential for increasing the expression of its components and occurs following NF-κB activation. Hyperthyroidism triggers CH, which can progress to maladaptive CH and even heart failure. We have shown in a previous study that thyroid hormone (TH)-induced CH is linked to the upregulation of S100A8, leading to NF-κB activation. Therefore, we aimed to investigate whether the NLRP3 inflammasome is involved in TH-induced CH and its potential role in CH pathophysiology. Hyperthyroidism was induced in NLRP3 knockout (NLRP3-KO), Caspase-1-KO and Wild Type (WT) male mice of the C57Bl/6J strain, aged 8-12 weeks, by triiodothyronine (7 μg/100 g BW, i.p.) administered daily for 14 days. Morphological and cardiac functional analysis besides molecular assays showed, for the first time, that TH-induced CH is accompanied by reduced NLRP3 expression in the heart and that it occurs independently of the NLRP3 inflammasome and caspase 1-related pathways. However, NLRP3 is important for the maintenance of basal cardiac function since NLRP3-KO mice had impaired diastolic function and reduced heart rate, ejection fraction, and fractional shortening compared with WT mice.
Collapse
Affiliation(s)
- Aline Cristina Parletta
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 2415, Butanta, Sao Paulo, 05508-000, Brazil
| | - Gabriela Cavazza Cerri
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 2415, Butanta, Sao Paulo, 05508-000, Brazil
| | - Claudia Ribeiro Borba Gasparini
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 2415, Butanta, Sao Paulo, 05508-000, Brazil
| | - Karine Panico
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 2415, Butanta, Sao Paulo, 05508-000, Brazil
| | - Denival Nascimento Vieira-Junior
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 2415, Butanta, Sao Paulo, 05508-000, Brazil
| | - Larissa Maria Zacarias-Rodrigues
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 2415, Butanta, Sao Paulo, 05508-000, Brazil
| | - Nathalia Senger
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 2415, Butanta, Sao Paulo, 05508-000, Brazil
| | - Amanda de Almeida Silva
- Department of Cardiopneumology, Heart Institute, Faculty of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Marina Fevereiro
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 2415, Butanta, Sao Paulo, 05508-000, Brazil
| | - Gabriela Placoná Diniz
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 2415, Butanta, Sao Paulo, 05508-000, Brazil
- Center for Regenerative Medicine, University of South Florida Health Heart Institute, Morsani School of Medicine, University of South Florida, Tampa, FL, USA
| | - Maria Cláudia Costa Irigoyen
- Department of Cardiopneumology, Heart Institute, Faculty of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Maria Luiza Morais Barreto-Chaves
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 2415, Butanta, Sao Paulo, 05508-000, Brazil.
| |
Collapse
|
4
|
Holt M, Lin J, Cicka M, Wong A, Epelman S, Lavine KJ. Dissecting and Visualizing the Functional Diversity of Cardiac Macrophages. Circ Res 2024; 134:1791-1807. [PMID: 38843293 DOI: 10.1161/circresaha.124.323817] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 05/09/2024] [Indexed: 06/11/2024]
Abstract
Cardiac macrophages represent a functionally diverse population of cells involved in cardiac homeostasis, repair, and remodeling. With recent advancements in single-cell technologies, it is possible to elucidate specific macrophage subsets based on transcriptional signatures and cell surface protein expression to gain a deep understanding of macrophage diversity in the heart. The use of fate-mapping technologies and parabiosis studies have provided insight into the ontogeny and dynamics of macrophages identifying subsets derived from embryonic and adult definitive hematopoietic progenitors that include tissue-resident and bone marrow monocyte-derived macrophages, respectively. Within the heart, these subsets have distinct tissue niches and functional roles in the setting of homeostasis and disease, with cardiac resident macrophages representing a protective cell population while bone marrow monocyte-derived cardiac macrophages have a context-dependent effect, triggering both proinflammatory tissue injury, but also promoting reparative functions. With the increased understanding of the clinical relevance of cardiac macrophage subsets, there has been an increasing need to detect and measure cardiac macrophage compositions in living animals and patients. New molecular tracers compatible with positron emission tomography/computerized tomography and positron emission tomography/ magnetic resonance imaging have enabled investigators to noninvasively and serially visualize cardiac macrophage subsets within the heart to define associations with disease and measure treatment responses. Today, advancements within this thriving field are poised to fuel an era of clinical translation.
Collapse
Affiliation(s)
- Megan Holt
- Division of Cardiology, Department of Medicine, Center for Cardiovascular Research, Washington University School of Medicine (M.H., M.C., K.J.L.)
| | - Julia Lin
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada (J.L., A.W., S.E.)
- Department of Immunology, University of Toronto, ON, Canada (J.L., A.W., S.E.)
| | - Markus Cicka
- Division of Cardiology, Department of Medicine, Center for Cardiovascular Research, Washington University School of Medicine (M.H., M.C., K.J.L.)
| | - Anthony Wong
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada (J.L., A.W., S.E.)
- Department of Immunology, University of Toronto, ON, Canada (J.L., A.W., S.E.)
| | - Slava Epelman
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada (J.L., A.W., S.E.)
- Ted Rogers Centre for Heart Research, Translational Biology and Engineering Program, Toronto, ON, Canada (S.E.)
- Department of Immunology, University of Toronto, ON, Canada (J.L., A.W., S.E.)
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada (S.E.)
| | - Kory J Lavine
- Division of Cardiology, Department of Medicine, Center for Cardiovascular Research, Washington University School of Medicine (M.H., M.C., K.J.L.)
| |
Collapse
|
5
|
Nagy A, Börzsei D, Hoffmann A, Török S, Veszelka M, Almási N, Varga C, Szabó R. A Comprehensive Overview on Chemotherapy-Induced Cardiotoxicity: Insights into the Underlying Inflammatory and Oxidative Mechanisms. Cardiovasc Drugs Ther 2024:10.1007/s10557-024-07574-0. [PMID: 38492161 DOI: 10.1007/s10557-024-07574-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/05/2024] [Indexed: 03/18/2024]
Abstract
While oncotherapy has made rapid progress in recent years, side effects of anti-cancer drugs and treatments have also come to the fore. These side effects include cardiotoxicity, which can cause irreversible cardiac damages with long-term morbidity and mortality. Despite the continuous in-depth research on anti-cancer drugs, an improved knowledge of the underlying mechanisms of cardiotoxicity are necessary for early detection and management of cardiac risk. Although most reviews focus on the cardiotoxic effect of a specific individual chemotherapeutic agent, the aim of our review is to provide comprehensive insight into various agents that induced cardiotoxicity and their underlying mechanisms. Characterization of these mechanisms are underpinned by research on animal models and clinical studies. In order to gain insight into these complex mechanisms, we emphasize the role of inflammatory processes and oxidative stress on chemotherapy-induced cardiac changes. A better understanding and identification of the interplay between chemotherapy and inflammatory/oxidative processes hold some promise to prevent or at least mitigate cardiotoxicity-associated morbidity and mortality among cancer survivors.
Collapse
Affiliation(s)
- András Nagy
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary
| | - Denise Börzsei
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary
| | - Alexandra Hoffmann
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary
| | - Szilvia Török
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary
| | - Médea Veszelka
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary
| | - Nikoletta Almási
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary
| | - Csaba Varga
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary
| | - Renáta Szabó
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary.
| |
Collapse
|
6
|
Malandish A, Gulati M. The impacts of exercise interventions on inflammaging markers in overweight/obesity patients with heart failure: A systematic review and meta-analysis of randomized controlled trials. IJC HEART & VASCULATURE 2023; 47:101234. [PMID: 37416483 PMCID: PMC10320319 DOI: 10.1016/j.ijcha.2023.101234] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/22/2023] [Accepted: 06/18/2023] [Indexed: 07/08/2023]
Abstract
Objectives The purpose of this meta-analysis was to investigate the association of aerobic, resistance and concurrent exercises vs. control group on inflammaging markers [tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), IL-1-beta, IL-8, and high sensitivity C-reactive protein (hs-CRP)] in overweight or obesity patients with heart failure (HF). Methods The databases of Scopus, PubMed, Web of Science and Google Scholar were searched until August 31, 2022 for exercise interventions vs. control group on circulating inflammaging markers in patients with HF. Only randomized controlled trial (RCT) articles were included. Standardized mean difference (SMD) and 95% confidence intervals (95%CIs) were calculated (registration code = CRD42022347164). Results Forty-six full-text articles (57 intervention arms and 3693 participants) were included. A significant reduction was occurred in inflammaging markers of IL-6 [SMD-0.205(95% CI:-0.332 to -0.078),p = 0.002] and hs-CRP [SMD -0.379 (95% CI:-0.556 to -0.202), p = 0.001] with exercise training in patients with HF. Analysis of subgroup by age, body mass index (BMI), type, intensity, duration of exercise and mean left ventricular ejection fraction (LVEF) revealed that there was a significant reduction in TNF-α for middle-aged (p = 0.031), concurrent training (p = 0.033), high intensity (p = 0.005), and heart failure with reduced ejection fraction (HFrEF) (p = 0.007) compared to the control group. There was a significant reduction in IL-6 for middle-aged (p = 0.006), overweight (p = 0.001), aerobic exercise (p = 0.001), both high and moderate intensities (p = 0.037 and p = 0.034), short-term follow-up (p = 0.001), and heart failure with preserved ejection fraction (HFpEF) (p = 0.001) compared to the control group. There was a significant reduction in hs-CRP for middle-aged (p = 0.004), elderly-aged (p = 0.001), overweight (p = 0.001), aerobic exercise (p = 0.001), concurrent training (p = 0.031), both high and moderate intensities (p = 0.017 and p = 0.001), short-term (p = 0.011), long-term (p = 0.049), and very long-term (p = 0.016) follow-ups, HFrEF (p = 0.003) and heart failure with mildly reduced ejection fraction (HFmrEF) (p = 0.048) compared to the control group. Conclusions The results confirmed that aerobic exercise and concurrent training interventions were effective to improve inflammaging markers of TNF-α, IL-6, and hs-CRP. These exercise-related anti-inflammaging responses were observed across ages (middle-aged and elderly-aged), exercise intensities, duration of follow-ups, and mean LVEFs (HFrEF, HFmrEF and HFpEF) in overweight patients with HF.
Collapse
Affiliation(s)
- Abbas Malandish
- Department of Exercise Physiology, Faculty of Sport Sciences, Urmia University, No. 19, Shams Tabrizi St., Velayat Ave., Keikhali Zone, Yamchi, East Azerbaijan, Urmia, Iran
| | - Martha Gulati
- Barbra Streisand Women’s Heart Center, Smidt Heart Institute, Cedars Sinai Medical Center, 127 S. San Vicente Blvd, Suite A3600, Los Angeles, CA 90048, USA
| |
Collapse
|
7
|
Abubakar M, Rasool HF, Javed I, Raza S, Abang L, Hashim MMA, Saleem Z, Abdullah RM, Faraz MA, Hassan KM, Bhat RR. Comparative Roles of IL-1, IL-6, IL-10, IL-17, IL-18, 1L-22, IL-33, and IL-37 in Various Cardiovascular Diseases With Potential Insights for Targeted Immunotherapy. Cureus 2023; 15:e42494. [PMID: 37637634 PMCID: PMC10455045 DOI: 10.7759/cureus.42494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2023] [Indexed: 08/29/2023] Open
Abstract
In recent years, the study of interleukins (ILs), crucial cytokines involved in immune response and inflammation, has garnered significant attention within the sphere of cardiovascular diseases (CVDs). The research has provided insights into the involvement of ILs in diverse CVDs, including arrhythmias, myocardial infarction, atherosclerosis, and heart failure (HF). ILs have emerged as promising therapeutic targets for drug interventions through their involvement in disease development and progression. This comprehensive review provides a detailed overview of ILs, elucidating their functions within the immune system and offering insights into their specific contributions to various CVDs. Moreover, the article delves into the examination of current and potential drug therapies that selectively target ILs in the management of CVDs, presenting a comprehensive analysis of the advantages and disadvantages associated with these therapeutic approaches. A comprehensive literature review was conducted to investigate the involvement of ILs in CVDs. The relevant articles were searched on PubMed, PubMed Central, Medline, Cochrane, Google Scholar, and ScienceDirect databases. The search encompassed articles published from these databases' inception until July 12, 2023. We first examine generalized aspects of ILs, particularly CVDs. Then, we shift focus towards examining the direct impact of ILs on cardiac cells and tissue; on the immune system and inflammation; endothelial cells and vascular function; and finally, their interactions with other signaling pathways and molecules. Then, we discuss the molecular mechanisms of various ILs. Sequentially, we delve into a comprehensive analysis of the individualized role of each distinct IL in diverse CVDs, examining their specific contributions. Finally, we explore the potential for targeted drug therapy to modulate IL activity, aiming to enhance outcomes for patients burdened with CVD. The objective is the identification of gaps in current knowledge and highlight areas that require further investigation within the context of cardiovascular medicine. Through deepening our comprehension of the intricate involvement of ILs in CVDs and harnessing their potential for targeted drug therapy, novel treatment strategies can be devised, leading to improved patient outcomes in cardiovascular medicine.
Collapse
Affiliation(s)
- Muhammad Abubakar
- Department of Internal Medicine, Ameer-Ud-Din Medical College, Lahore General Hospital, Lahore, PAK
- Department of Internal Medicine, Siddique Sadiq Memorial Trust Hospital, Gujranwala, PAK
| | - Hafiz Fahad Rasool
- Department of Public Health, Nanjing Medical University School of Public Health, Nanjing, CHN
| | - Izzah Javed
- Department of Internal Medicine, Ameer-Ud-Din Medical College, Lahore General Hospital, Lahore, PAK
| | - Saud Raza
- Department of Internal Medicine, Ameer-Ud-Din Medical College, Lahore General Hospital, Lahore, PAK
| | - Lucy Abang
- Department of Biochemistry, All Saints University School of Medicine, Roseau, DMA
| | | | - Zartasha Saleem
- Department of Emergency Medicine, The University of Lahore Teaching Hospital, Lahore, PAK
| | | | - Muhammad Ahmad Faraz
- Department of Forensic Medicine, Post Graduate Medical Institute, Lahore General Hospital, Lahore, PAK
| | - Khawaja Mushammar Hassan
- Department of Internal Medicine, Ameer-Ud-Din Medical College, Lahore General Hospital, Lahore, PAK
| | - Rakshita Ramesh Bhat
- Department of Medical Oncology, Mangalore Institute of Oncology, Mangalore, IND
- Department of Internal Medicine, Bangalore Medical College and Research Institute, Bangalore, IND
| |
Collapse
|
8
|
Rexius-Hall ML, Khalil NN, Escopete SS, Li X, Hu J, Yuan H, Parker SJ, McCain ML. A myocardial infarct border-zone-on-a-chip demonstrates distinct regulation of cardiac tissue function by an oxygen gradient. SCIENCE ADVANCES 2022; 8:eabn7097. [PMID: 36475790 PMCID: PMC9728975 DOI: 10.1126/sciadv.abn7097] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
After a myocardial infarction, the boundary between the injured, hypoxic tissue and the adjacent viable, normoxic tissue, known as the border zone, is characterized by an oxygen gradient. Yet, the impact of an oxygen gradient on cardiac tissue function is poorly understood, largely due to limitations of existing experimental models. Here, we engineered a microphysiological system to controllably expose engineered cardiac tissue to an oxygen gradient that mimics the border zone and measured the effects of the gradient on electromechanical function and the transcriptome. The gradient delayed calcium release, reuptake, and propagation; decreased diastolic and peak systolic stress; and increased expression of inflammatory cascades that are hallmarks of myocardial infarction. These changes were distinct from those observed in tissues exposed to uniform normoxia or hypoxia, demonstrating distinct regulation of cardiac tissue phenotypes by an oxygen gradient. Our border-zone-on-a-chip model advances functional and mechanistic insight into oxygen-dependent cardiac tissue pathophysiology.
Collapse
Affiliation(s)
- Megan L. Rexius-Hall
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | - Natalie N. Khalil
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | - Sean S. Escopete
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Xin Li
- Department of Mechanics and Aerospace Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Jiayi Hu
- Department of Mechanics and Aerospace Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Hongyan Yuan
- Department of Mechanics and Aerospace Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Sarah J. Parker
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Megan L. McCain
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
- Corresponding author.
| |
Collapse
|
9
|
Das SL, Sutherland BP, Lejeune E, Eyckmans J, Chen CS. Mechanical response of cardiac microtissues to acute localized injury. Am J Physiol Heart Circ Physiol 2022; 323:H738-H748. [PMID: 36053751 DOI: 10.1152/ajpheart.00305.2022] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
After a myocardial infarction (MI), the heart undergoes changes including local remodeling that can lead to regional abnormalities in mechanical and electrical properties, ultimately increasing the risk of arrythmias and heart failure. While these responses have been successfully recapitulated in animal models of MI, local changes in tissue and cell-level mechanics caused by MI remain difficult to study in vivo. Here, we developed an in vitro cardiac microtissue (CMT) injury system which through acute focal injury recapitulates aspects of the regional responses seen following an MI. Using a pulsed laser, cell death was induced in the center of the microtissue causing a loss of calcium signaling and a complete loss of contractile function in the injured region and resulting in a 39% reduction in the CMT's overall force production. After 7 days, the injured area remained void of CMs and showed increased expression of vimentin and fibronectin, two markers for fibrotic remodeling. Interestingly, while the injured region showed minimal recovery, calcium amplitudes in uninjured regions returned to levels comparable to control. Furthermore, overall force production returned to pre-injury levels despite the lack of contractile function in the injured region. Instead, uninjured regions exhibited elevated contractile function, compensating for the loss of function in the injured region, drawing parallels to changes in tissue-level mechanics seen in vivo. Overall, this work presents a new in vitro model to study cardiac tissue remodeling and electromechanical changes after injury.
Collapse
Affiliation(s)
- Shoshana L Das
- Harvard-MIT Program in Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, United States.,Department of Biomedical Engineering, Boston University, Boston, MA, United States.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, United States
| | - Bryan P Sutherland
- Department of Biomedical Engineering, Boston University, Boston, MA, United States.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, United States
| | - Emma Lejeune
- Department of Mechanical Engineering, Boston University, Boston, MA, United States
| | - Jeroen Eyckmans
- Department of Biomedical Engineering, Boston University, Boston, MA, United States.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, United States
| | - Christopher S Chen
- Department of Biomedical Engineering, Boston University, Boston, MA, United States.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, United States
| |
Collapse
|
10
|
Siddiqui SW, Ashok T, Patni N, Fatima M, Lamis A, Anne KK. Anemia and Heart Failure: A Narrative Review. Cureus 2022; 14:e27167. [PMID: 36017290 PMCID: PMC9393312 DOI: 10.7759/cureus.27167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2022] [Indexed: 11/06/2022] Open
Abstract
Anemia in heart failure patients is a relatively common finding and has been linked with an increased risk of hospital admissions, morbidities, and significant mortality making its correction a significant factor in improving the quality of life and clinical outcomes in those suffering from it. This review article has discussed the multifactorial pathophysiology, including iron deficiency, longstanding inflammation, abnormal levels of human erythropoietin (Epo), and the abnormal activation of the renin-angiotensin-aldosterone system (RAAS) being the most significant. The diagnostic guidelines as well as research-based management modalities specifically with iron supplements and erythropoietin stimulating agents have also been discussed, although research done in this area has been limited and shown conflicting results.
Collapse
|
11
|
Arhontoulis DC, Kerr C, Richards D, Tjen K, Hyams N, Jones JA, Deleon-Pennell K, Menick D, Lindner D, Westermann D, Mei Y. Human Cardiac Organoids to Model COVID-19 Cytokine Storm Induced Cardiac Injuries. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.01.31.478497. [PMID: 35132419 PMCID: PMC8820666 DOI: 10.1101/2022.01.31.478497] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Acute cardiac injuries occur in 20-25% of hospitalized COVID-19 patients. Despite urgent needs, there is a lack of 3D organotypic models of COVID-19 hearts for mechanistic studies and drug testing. Herein, we demonstrate that human cardiac organoids (hCOs) are a viable platform to model the cardiac injuries caused by COVID-19 hyperinflammation. As IL-1βis an upstream cytokine and a core COVID-19 signature cytokine, it was used to stimulate hCOs to induce the release of a milieu of proinflammatory cytokines that mirror the profile of COVID-19 cytokine storm. The IL-1 β treated hCOs recapitulated transcriptomic, structural, and functional signatures of COVID-19 hearts. The comparison of IL-1β treated hCOs with cardiac tissue from COVID-19 autopsies illustrated the critical roles of hyper-inflammation in COVID-19 cardiac insults and indicated the cardioprotective effects of endothelium. The IL-1β treated hCOs also provide a viable model to assess the efficacy and potential side effects of immunomodulatory drugs, as well as the reversibility of COVID-19 cardiac injuries at baseline and simulated exercise conditions.
Collapse
Affiliation(s)
- Dimitrios C Arhontoulis
- Molecular and Cellular Biology and Pathobiology Program, Medical University of South Carolina, Charleston, SC, USA
| | - Charles Kerr
- Molecular and Cellular Biology and Pathobiology Program, Medical University of South Carolina, Charleston, SC, USA
| | - Dylan Richards
- Bioengineering Department, Clemson University, Clemson, SC, USA
| | - Kelsey Tjen
- Molecular and Cellular Biology and Pathobiology Program, Medical University of South Carolina, Charleston, SC, USA
| | - Nathaniel Hyams
- Bioengineering Department, Clemson University, Clemson, SC, USA
| | - Jefferey A. Jones
- Molecular and Cellular Biology and Pathobiology Program, Medical University of South Carolina, Charleston, SC, USA
- Ralph H. Johnson Veterans Affairs Medical Center, Research Service, Charleston, SC, USA
- Division of Cardiothoracic Surgery, Department of Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Kristine Deleon-Pennell
- Molecular and Cellular Biology and Pathobiology Program, Medical University of South Carolina, Charleston, SC, USA
- Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute, Medical University of South Carolina, Charleston, SC, USA
- Ralph H. Johnson Veterans Affairs Medical Center, Research Service, Charleston, SC, USA
| | - Donald Menick
- Molecular and Cellular Biology and Pathobiology Program, Medical University of South Carolina, Charleston, SC, USA
- Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute, Medical University of South Carolina, Charleston, SC, USA
- Ralph H. Johnson Veterans Affairs Medical Center, Research Service, Charleston, SC, USA
| | - Diana Lindner
- Department of Cardiology, University Heart and Vascular Center Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg / Kiel / Lübeck, Germany
| | - Dirk Westermann
- Department of Cardiology, University Heart and Vascular Center Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg / Kiel / Lübeck, Germany
| | - Ying Mei
- Molecular and Cellular Biology and Pathobiology Program, Medical University of South Carolina, Charleston, SC, USA
- Bioengineering Department, Clemson University, Clemson, SC, USA
| |
Collapse
|
12
|
Salerno JA, Torquato T, Temerozo JR, Goto-Silva L, Karmirian K, Mendes MA, Sacramento CQ, Fintelman-Rodrigues N, Souza LRQ, Ornelas IM, Veríssimo CP, Aragão LGHS, Vitória G, Pedrosa CSG, da Silva Gomes Dias S, Cardoso Soares V, Puig-Pijuan T, Salazar V, Dariolli R, Biagi D, Furtado DR, Barreto Chiarini L, Borges HL, Bozza PT, Zaluar P. Guimarães M, Souza TM, Rehen SK. Inhibition of SARS-CoV-2 infection in human iPSC-derived cardiomyocytes by targeting the Sigma-1 receptor disrupts cytoarchitecture and beating. PeerJ 2021; 9:e12595. [PMID: 35036128 PMCID: PMC8697769 DOI: 10.7717/peerj.12595] [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: 09/30/2021] [Accepted: 11/14/2021] [Indexed: 12/21/2022] Open
Abstract
SARS-CoV-2 infects cardiac cells and causes heart dysfunction. Conditions such as myocarditis and arrhythmia have been reported in COVID-19 patients. The Sigma-1 receptor (S1R) is a ubiquitously expressed chaperone that plays a central role in cardiomyocyte function. S1R has been proposed as a therapeutic target because it may affect SARS-CoV-2 replication; however, the impact of the inhibition of S1R in human cardiomyocytes remains to be described. In this study, we investigated the consequences of S1R inhibition in iPSC-derived human cardiomyocytes (hiPSC-CM). SARS-CoV-2 infection in hiPSC-CM was productive and reduced cell survival. S1R inhibition decreased both the number of infected cells and viral particles after 48 hours. S1R inhibition also prevented the release of pro-inflammatory cytokines and cell death. Although the S1R antagonist NE-100 triggered those protective effects, it compromised cytoskeleton integrity by downregulating the expression of structural-related genes and reducing beating frequency. Our findings suggest that the detrimental effects of S1R inhibition in human cardiomyocytes' integrity may abrogate its therapeutic potential against COVID and should be carefully considered.
Collapse
Affiliation(s)
- José Alexandre Salerno
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | - Thayana Torquato
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | - Jairo R. Temerozo
- National Institute for Science and Technology on Neuroimmunomodulation (INCT/NIM), Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil
- Laboratory on Thymus Research, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil
| | - Livia Goto-Silva
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | - Karina Karmirian
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | - Mayara A. Mendes
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | - Carolina Q. Sacramento
- Immunopharmacology Laboratory, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil
- National Institute for Science and Technology on Innovation in Diseases of Neglected Populations (INCT/IDPN), Center for Technological Development in Health (CDTS), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil
| | - Natalia Fintelman-Rodrigues
- Immunopharmacology Laboratory, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil
- National Institute for Science and Technology on Innovation in Diseases of Neglected Populations (INCT/IDPN), Center for Technological Development in Health (CDTS), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil
| | - Letícia R Q. Souza
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | - Isis M. Ornelas
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | - Carla P. Veríssimo
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | | | - Gabriela Vitória
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | | | - Suelen da Silva Gomes Dias
- Immunopharmacology Laboratory, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil
| | - Vinicius Cardoso Soares
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Immunopharmacology Laboratory, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil
| | - Teresa Puig-Pijuan
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
- Carlos Chagas Filho Institute of Biophysics (IBCCF), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Vinícius Salazar
- Department of Systems and Computer Engineering, COPPE, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Rafael Dariolli
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- PluriCell Biotech, São Paulo, Brazil
| | | | - Daniel R. Furtado
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | - Luciana Barreto Chiarini
- Carlos Chagas Filho Institute of Biophysics (IBCCF), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Helena L. Borges
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Patrícia T. Bozza
- Immunopharmacology Laboratory, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil
| | - Marilia Zaluar P. Guimarães
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | - Thiago M.L. Souza
- Immunopharmacology Laboratory, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil
- National Institute for Science and Technology on Innovation in Diseases of Neglected Populations (INCT/IDPN), Center for Technological Development in Health (CDTS), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil
| | - Stevens K. Rehen
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
- Department of Genetics, Institute of Biology, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
13
|
Goswami SK, Ranjan P, Dutta RK, Verma SK. Management of inflammation in cardiovascular diseases. Pharmacol Res 2021; 173:105912. [PMID: 34562603 DOI: 10.1016/j.phrs.2021.105912] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 09/01/2021] [Accepted: 09/20/2021] [Indexed: 12/12/2022]
Abstract
Cardiovascular disease is the leading cause of morbidity and mortality world-wide. Recently, the role of inflammation in the progression of diseases has significantly attracted considerable attention. In addition, various comorbidities, including diabetes, obesity, etc. exacerbate inflammation in the cardiovascular system, which ultimately leads to heart failure. Furthermore, cytokines released from specialized immune cells are key mediators of cardiac inflammation. Here, in this review article, we focused on the role of selected immune cells and cytokines (both pro-inflammatory and anti-inflammatory) in the regulation of cardiac inflammation and ultimately in cardiovascular diseases. While IL-1β, IL-6, TNFα, and IFNγ are associated with cardiac inflammation; IL-10, TGFβ, etc. are associated with resolution of inflammation and cardiac repair. IL-10 reduces cardiovascular inflammation and protects the cardiovascular system via interaction with SMAD2, p53, HuR, miR-375 and miR-21 pathway. In addition, we also highlighted recent advancements in the management of cardiac inflammation, including clinical trials of anti-inflammatory molecules to alleviate cardiovascular diseases.
Collapse
Affiliation(s)
- Sumanta Kumar Goswami
- Department of Medicine, Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Prabhat Ranjan
- Department of Medicine, Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Roshan Kumar Dutta
- Department of Medicine, Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Suresh Kumar Verma
- Department of Medicine, Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL 35233, USA.
| |
Collapse
|
14
|
Farag MM, Khalifa AA, Elhadidy WF, Rashad RM. Thymoquinone dose-dependently attenuates myocardial injury induced by isoproterenol in rats via integrated modulations of oxidative stress, inflammation, apoptosis, autophagy, and fibrosis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2021; 394:1787-1801. [PMID: 34216225 DOI: 10.1007/s00210-021-02087-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/06/2021] [Indexed: 11/30/2022]
Abstract
As rats develop myocardial infarction (MI) like lesions when injected with large doses of isoproterenol (ISO), this investigation was designed to evaluate the dose-dependent effects of thymoquinone (TQ) on ISO-induced myocardial injury in rats. Adult male rats were divided into negative control, TQ20 (20 mg/kg/day), TQ50 (50 mg/kg/day), ISO positive control, TQ20 + ISO, and TQ50 + ISO groups. In these rats, biochemical, immunobiochemical, and histopathological studies were carried out to evaluate myocardial oxidative stress, inflammation, apoptosis, fibrosis, and autophagy, and the changes in serum cardiac biomarkers. The results showed that TQ pretreatment in ISO-administered rats produced a dose-dependent significant reduction of the myocardial infarct size, markedly reduced the ISO-induced elevation in serum cardiac markers and demonstrated several other important findings related to the cardioprotective efficacy of TQ. First, this study is the first reported research work showing that TQ treatment could increase the myocardial reduced glutathione baseline level, adding an indirect antioxidant effect to its known direct free radical scavenging effect. Second, pretreatment with TQ significantly reduced the markers of myocardial oxidative stress, inflammation, fibrosis, and apoptosis. Third, TQ acted as an autophagy enhancer ameliorating myocardial cell damage and dysfunction. Thus, the morphological and biochemical changes associated with ISO-induced myocardial injury were ameliorated with TQ pretreatment. The extent of this improvement was significantly greater in the TQ50 + ISO group than in the TQ20 + ISO group. The present study, for the first time, demonstrates these dose-dependent effects of TQ in experimentally induced myocardial injury. These findings raise the possibility that TQ may serve as a promising prophylactic cardioprotective therapy for patients who are at risk of developing myocardial injury and against the progression of existent myocardial injury as in cases of MI.
Collapse
Affiliation(s)
- Mahmoud M Farag
- Department of Pharmacology, Medical Research Institute, Alexandria University, 165 El-Horria Avenue, P.O. El-Hadara 21561, Alexandria, Egypt.
| | - Asmaa A Khalifa
- Department of Pharmacology, Medical Research Institute, Alexandria University, 165 El-Horria Avenue, P.O. El-Hadara 21561, Alexandria, Egypt
| | - Wessam F Elhadidy
- Department of Pharmacology, Medical Research Institute, Alexandria University, 165 El-Horria Avenue, P.O. El-Hadara 21561, Alexandria, Egypt
| | - Radwa M Rashad
- Department of Pathology, Medical Research Institute, Alexandria University, Alexandria, Egypt
| |
Collapse
|
15
|
Lun Y, Borjini N, Miura NN, Ohno N, Singer NG, Lin F. CDCP1 on Dendritic Cells Contributes to the Development of a Model of Kawasaki Disease. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 206:2819-2827. [PMID: 34099547 PMCID: PMC9011922 DOI: 10.4049/jimmunol.2001406] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 04/09/2021] [Indexed: 11/19/2022]
Abstract
The etiology and pathology of Kawasaki disease (KD) remain elusive. Cub domain-containing protein 1 (CDCP1), a cell-surface protein that confers poor prognosis of patients with certain solid tumors, was recently identified as one of the most significantly upregulated genes in SARS-CoV-2-infected children who developed systemic vasculitis, a hallmark of KD. However, a potential role of CDCP1 in KD has not previously been explored. In this study, we found that CDCP1 knockout (KO) mice exhibited attenuated coronary and aortic vasculitis and decreased serum Candida albicans water-soluble fraction (CAWS)-specific IgM/IgG2a and IL-6 concentrations compared with wild-type mice in an established model of KD induced by CAWS administration. CDCP1 expression was not detectable in cardiomyocytes, cardio fibroblasts, or coronary endothelium, but constitutive expression of CDCP1 was observed on dendritic cells (DCs) and was upregulated by CAWS stimulation. CAWS-induced IL-6 production was significantly reduced in CDCP1 KO DCs, in association with impaired Syk-MAPK signaling pathway activation. These novel findings suggest that CDCP1 might regulate KD development by modulating IL-6 production from DCs via the Syk-MAPK signaling pathway.
Collapse
Affiliation(s)
- Yu Lun
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Vascular Surgery, The First Hospital, China Medical University, Shenyang, China
| | - Nozha Borjini
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Noriko N Miura
- Laboratory for Immunopharmacology of Microbial Products, School of Pharmacy, Tokyo University of Pharmacy and Life Science, Hachioji, Tokyo, Japan; and
| | - Naohito Ohno
- Laboratory for Immunopharmacology of Microbial Products, School of Pharmacy, Tokyo University of Pharmacy and Life Science, Hachioji, Tokyo, Japan; and
| | - Nora G Singer
- Division of Rheumatology, MetroHealth Medical Center, Cleveland, OH
| | - Feng Lin
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA;
| |
Collapse
|
16
|
Gordon HP, Katz MG, Fazal S, Gillespie VL, Fargnoli AS, Gubara SM, Madjarova SJ, Cohen JA. Inflammatory Responses with Left Ventricular Compromise after Induction of Myocardial Infarcts in Sheep (Ovis aries). Comp Med 2021; 71:240-246. [PMID: 34082856 DOI: 10.30802/aalas-cm-21-000005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Ischemic myocardial disease is a major cause of death among humans worldwide; it results in scarring and pallor of the myocardium and triggers an inflammatory response that contributes to impaired left ventricular function. This response includes and is evidenced by the production of several inflammatory cytokines including TNFα, IL1β, IL4, IFNγ, IL10 and IL6. In the current study, myocardial infarcts were induced in 6 mo old male castrated sheep by ligation of the left circumflex obtuse marginal arteries (OM 1 and 2). MRI was used to measure parameters of left ventricular function that include EDV, ESV, EF, SVI, dp/dt max and dp/dt min at baseline and at 4 wk and 3 mo after infarct induction. We also measured serum concentrations of an array of cytokines. Postmortem histologic findings corroborate the existence of left ventricular myocardial injury and deterioration. Our data show a correlation between serum cytokine concentrations and the development of myocardial damage and left ventricular functional compromise.
Collapse
Affiliation(s)
- Hylton P Gordon
- Center for Comparative Medicine and Surgery, Icahn School of Medicine at Mt. Sinai, New York, New York;,
| | - Michael G Katz
- Department of Cardiology, Icahn School of Medicine at Mt. Sinai, New York, New York
| | - Shahood Fazal
- Department of Cardiology, Icahn School of Medicine at Mt. Sinai, New York, New York
| | - Virginia L Gillespie
- Center for Comparative Medicine and Surgery, Icahn School of Medicine at Mt. Sinai, New York, New York
| | - Anthony S Fargnoli
- Department of Cardiology, Icahn School of Medicine at Mt. Sinai, New York, New York
| | - Sarah M Gubara
- Department of Cardiology, Icahn School of Medicine at Mt. Sinai, New York, New York
| | - Sophia J Madjarova
- Department of Biology, Columbia University School of Medicine, New York, New York
| | - Jonathan A Cohen
- Center for Comparative Medicine and Surgery, Icahn School of Medicine at Mt. Sinai, New York, New York
| |
Collapse
|
17
|
Impact of SGLT2 Inhibitors on Heart Failure: From Pathophysiology to Clinical Effects. Int J Mol Sci 2021; 22:ijms22115863. [PMID: 34070765 PMCID: PMC8199383 DOI: 10.3390/ijms22115863] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 12/11/2022] Open
Abstract
Heart failure (HF) affects up to over 20% of patients with type 2 diabetes (T2DM), even more in the elderly. Although, in T2DM, both hyperglycemia and the proinflammatory status induced by insulin resistance are crucial in cardiac function impairment, SGLT2i cardioprotective mechanisms against HF are several. In particular, these beneficial effects seem attributable to the significant reduction of intracellular sodium levels, well-known to exert a cardioprotective role in the prevention of oxidative stress and consequent cardiomyocyte death. From a molecular perspective, patients’ exposure to gliflozins’ treatment mimics nutrient and oxygen deprivation, with consequent autophagy stimulation. This allows to maintain the cellular homeostasis through different degradative pathways. Thus, since their introduction in the clinical practice, the hypotheses on SGLT2i mechanisms of action have changed: from simple glycosuric drugs, with consequent glucose lowering, erythropoiesis enhancing and ketogenesis stimulating, to intracellular sodium-lowering molecules. This provides their consequent cardioprotective effect, which justifies its significant reduction in CV events, especially in populations at higher risk. Finally, the updated clinical evidence of SGLT2i benefits on HF was summarized. Thus, this review aimed to analyze the cardioprotective mechanisms of sodium glucose transporter 2 inhibitors (SGLT2i) in patients with HF, as well as their clinical impact on cardiovascular events.
Collapse
|
18
|
Panizo S, Martínez-Arias L, Alonso-Montes C, Cannata P, Martín-Carro B, Fernández-Martín JL, Naves-Díaz M, Carrillo-López N, Cannata-Andía JB. Fibrosis in Chronic Kidney Disease: Pathogenesis and Consequences. Int J Mol Sci 2021; 22:E408. [PMID: 33401711 PMCID: PMC7795409 DOI: 10.3390/ijms22010408] [Citation(s) in RCA: 183] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/18/2020] [Accepted: 12/29/2020] [Indexed: 02/07/2023] Open
Abstract
Fibrosis is a process characterized by an excessive accumulation of the extracellular matrix as a response to different types of tissue injuries, which leads to organ dysfunction. The process can be initiated by multiple and different stimuli and pathogenic factors which trigger the cascade of reparation converging in molecular signals responsible of initiating and driving fibrosis. Though fibrosis can play a defensive role, in several circumstances at a certain stage, it can progressively become an uncontrolled irreversible and self-maintained process, named pathological fibrosis. Several systems, molecules and responses involved in the pathogenesis of the pathological fibrosis of chronic kidney disease (CKD) will be discussed in this review, putting special attention on inflammation, renin-angiotensin system (RAS), parathyroid hormone (PTH), fibroblast growth factor 23 (FGF23), Klotho, microRNAs (miRs), and the vitamin D hormonal system. All of them are key factors of the core and regulatory pathways which drive fibrosis, having a great negative kidney and cardiac impact in CKD.
Collapse
Affiliation(s)
- Sara Panizo
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Retic REDinREN-ISCIII, Universidad de Oviedo, 33011 Oviedo, Spain; (S.P.); (L.M.-A.); (C.A.-M.); (B.M.-C.); (J.L.F.-M.); (N.C.-L.)
| | - Laura Martínez-Arias
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Retic REDinREN-ISCIII, Universidad de Oviedo, 33011 Oviedo, Spain; (S.P.); (L.M.-A.); (C.A.-M.); (B.M.-C.); (J.L.F.-M.); (N.C.-L.)
| | - Cristina Alonso-Montes
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Retic REDinREN-ISCIII, Universidad de Oviedo, 33011 Oviedo, Spain; (S.P.); (L.M.-A.); (C.A.-M.); (B.M.-C.); (J.L.F.-M.); (N.C.-L.)
| | - Pablo Cannata
- Pathology Department, Fundación Instituto de Investigaciones Sanitarias-Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid (UAM), Retic REDinREN-ISCIII, 28040 Madrid, Spain;
| | - Beatriz Martín-Carro
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Retic REDinREN-ISCIII, Universidad de Oviedo, 33011 Oviedo, Spain; (S.P.); (L.M.-A.); (C.A.-M.); (B.M.-C.); (J.L.F.-M.); (N.C.-L.)
| | - José L. Fernández-Martín
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Retic REDinREN-ISCIII, Universidad de Oviedo, 33011 Oviedo, Spain; (S.P.); (L.M.-A.); (C.A.-M.); (B.M.-C.); (J.L.F.-M.); (N.C.-L.)
| | - Manuel Naves-Díaz
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Retic REDinREN-ISCIII, Universidad de Oviedo, 33011 Oviedo, Spain; (S.P.); (L.M.-A.); (C.A.-M.); (B.M.-C.); (J.L.F.-M.); (N.C.-L.)
| | - Natalia Carrillo-López
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Retic REDinREN-ISCIII, Universidad de Oviedo, 33011 Oviedo, Spain; (S.P.); (L.M.-A.); (C.A.-M.); (B.M.-C.); (J.L.F.-M.); (N.C.-L.)
| | - Jorge B. Cannata-Andía
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Retic REDinREN-ISCIII, Universidad de Oviedo, 33011 Oviedo, Spain; (S.P.); (L.M.-A.); (C.A.-M.); (B.M.-C.); (J.L.F.-M.); (N.C.-L.)
| |
Collapse
|
19
|
Kashiwagi A, Araki S, Maegawa H. Sodium-glucose cotransporter 2 inhibitors represent a paradigm shift in the prevention of heart failure in type 2 diabetes patients. J Diabetes Investig 2021; 12:6-20. [PMID: 32563214 PMCID: PMC7779279 DOI: 10.1111/jdi.13329] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/12/2020] [Accepted: 06/16/2020] [Indexed: 02/06/2023] Open
Abstract
Recent major clinical trials of the use of sodium-glucose cotransporter 2 (SGLT2) inhibitors in patients with type 2 diabetes have shown that they reduce three-point major adverse cardiovascular events, cardiovascular death, hospitalization for heart failure (HF) and a composite renal outcome. These beneficial effects of SGLT2 inhibitors are also evident in type 2 diabetes patients with a previous history of atherosclerotic cardiovascular disease or advanced renal disease. HF is a major determinant of the prognosis of diabetes patients. Although HF with low ejection fraction can be effectively treated with antihypertensive drugs, these treatments do not reduce mortality in HF patients with preserved ejection fraction (HFpEF). HFpEF is clinically characterized by left ventricular diastolic dysfunction, perivascular fibrosis and stiffness of cardiomyocytes, defined as "cardiomyopathy". Therefore, HFpEF is considered to be an entirely separate entity to HF with low ejection fraction. Recent studies have suggested that HFpEF might be treatable using SGLT2 inhibitors, which ameliorate visceral adiposity, insulin resistance, hyperglycemia, hyperlipidemia, volume overload, hypertension and cardiac inflammation. In the final part of the present review, we discuss the biochemical and molecular mechanisms of the effects of SGLT2 inhibitors in type 2 diabetes patients with HFpEF. These involve amelioration of the low nitric oxide production and oxidative stress, a reduction in cardiac inflammatory cytokine signaling, inhibition of Ca2+ overload, and an improvement in cardiac energy metabolism as a result of ketone body production. Investigations of the beneficial effects of SGLT2 inhibitors on cardiorenal outcomes, including hospitalization for HF, are now being carried out in preclinical and clinical studies.
Collapse
Affiliation(s)
| | - Shinchi Araki
- Department of MedicineShiga University of Medical ScienceOtsuJapan
| | - Hiroshi Maegawa
- Department of MedicineShiga University of Medical ScienceOtsuJapan
| |
Collapse
|
20
|
Riley LA, Merryman WD. Cadherin-11 and cardiac fibrosis: A common target for a common pathology. Cell Signal 2020; 78:109876. [PMID: 33285242 DOI: 10.1016/j.cellsig.2020.109876] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 12/01/2020] [Accepted: 12/02/2020] [Indexed: 02/06/2023]
Abstract
Cardiac fibrosis represents an enormous health concern as it is prevalent in nearly every form of cardiovascular disease, the leading cause of death worldwide. Fibrosis is characterized by the activation of fibroblasts into myofibroblasts, a contractile cell type that secretes significant amounts of extracellular matrix components; however, the onset of this condition is also due to persistent inflammation and the cellular responses to a changing mechanical environment. In this review, we provide an overview of the pro-fibrotic, pro-inflammatory, and biomechanical mechanisms that lead to cardiac fibrosis in cardiovascular diseases. We then discuss cadherin-11, an intercellular adhesion protein present on both myofibroblasts and inflammatory cells, as a potential link for all three of the fibrotic mechanisms. Since experimentally blocking cadherin-11 dimerization prevents fibrotic diseases including cardiac fibrosis, understanding how this protein can be targeted for therapeutic use could lead to better treatments for patients with heart disease.
Collapse
Affiliation(s)
- Lance A Riley
- Department of Biomedical Engineering, Vanderbilt University, USA
| | - W David Merryman
- Department of Biomedical Engineering, Vanderbilt University, USA.
| |
Collapse
|
21
|
Denning NL, Aziz M, Diao L, Prince JM, Wang P. Targeting the eCIRP/TREM-1 interaction with a small molecule inhibitor improves cardiac dysfunction in neonatal sepsis. Mol Med 2020; 26:121. [PMID: 33276725 PMCID: PMC7716442 DOI: 10.1186/s10020-020-00243-6] [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: 09/08/2020] [Accepted: 11/12/2020] [Indexed: 02/06/2023] Open
Abstract
Background Neonatal sepsis and the associated myocardial dysfunction remain a leading cause of infant mortality. Extracellular cold-inducible RNA-binding protein (eCIRP) acts as a ligand of triggering receptor expressed on myeloid cells-1 (TREM-1). M3 is a small CIRP-derived peptide that inhibits the eCIRP/TREM-1 interaction. We hypothesize that the eCIRP/TREM-1 interaction in cardiomyocytes contributes to sepsis-induced cardiac dysfunction in neonatal sepsis, while M3 is cardioprotective. Methods Serum was collected from neonates in the Neonatal Intensive Care Unit (NICU). 5–7-day old C57BL/6 mouse pups were used in this study. Primary murine neonatal cardiomyocytes were stimulated with recombinant murine (rm) CIRP with M3. TREM-1 mRNA and supernatant cytokine levels were assayed. Mitochondrial oxidative stress, ROS, and membrane potential were assayed. Neonatal mice were injected with rmCIRP and speckle-tracking echocardiography was conducted to measure cardiac strain. Sepsis was induced by i.p. cecal slurry. Mouse pups were treated with M3 or vehicle. After 16 h, echocardiography was performed followed by euthanasia for tissue analysis. A 7-day survival study was conducted. Results Serum eCIRP levels were elevated in septic human neonates. rmCIRP stimulation of cardiomyocytes increased TREM-1 gene expression. Stimulation of cardiomyocytes with rmCIRP upregulated TNF-α and IL-6 in the supernatants, while this upregulation was inhibited by M3. Stimulation of cardiomyocytes with rmCIRP resulted in a reduction in mitochondrial membrane potential (MMP) while M3 treatment returned MMP to near baseline. rmCIRP caused mitochondrial calcium overload; this was inhibited by M3. rmCIRP injection impaired longitudinal and radial cardiac strain. Sepsis resulted in cardiac dysfunction with a reduction in cardiac output and left ventricular end diastolic diameter. Both were improved by M3 treatment. Treatment with M3 attenuated serum, cardiac, and pulmonary levels of pro-inflammatory cytokines compared to vehicle-treated septic neonates. M3 dramatically increased sepsis survival. Conclusions Inhibition of eCIRP/TREM-1 interaction with M3 is cardioprotective, decreases inflammation, and improves survival in neonatal sepsis. Trial registration Retrospectively registered.
Collapse
Affiliation(s)
- Naomi-Liza Denning
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA.,Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, USA.,Department of Surgery, Donald and Barbara Zucker School of Medicine At Hofstra/Northwell, Hempstead, NY, USA
| | - Monowar Aziz
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA.,Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, USA
| | - Li Diao
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA
| | - Jose M Prince
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA.,Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, USA.,Department of Surgery, Donald and Barbara Zucker School of Medicine At Hofstra/Northwell, Hempstead, NY, USA.,Division of Pediatric Surgery, Cohen Children's Medical Center At Hofstra/Northwell, New Hyde Park, NY, USA
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA. .,Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, USA. .,Department of Surgery, Donald and Barbara Zucker School of Medicine At Hofstra/Northwell, Hempstead, NY, USA.
| |
Collapse
|
22
|
Sun J, Niu C, Ye W, An N, Chen G, Huang X, Wang J, Chen X, Shen Y, Huang S, Wang Y, Wang X, Wang Y, Jin L, Cong W, Li X. FGF13 Is a Novel Regulator of NF-κB and Potentiates Pathological Cardiac Hypertrophy. iScience 2020; 23:101627. [PMID: 33089113 PMCID: PMC7567043 DOI: 10.1016/j.isci.2020.101627] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 08/13/2020] [Accepted: 09/25/2020] [Indexed: 12/31/2022] Open
Abstract
FGF13 is an intracellular FGF factor. Its role in cardiomyopathies has been rarely investigated. We revealed that endogenous FGF13 is up-regulated in cardiac hypertrophy accompanied by increased nuclear localization. The upregulation of FGF13 plays a deteriorating role both in hypertrophic cardiomyocytes and mouse hearts. Mechanistically, FGF13 directly interacts with p65 by its nuclear localization sequence and co-localizes with p65 in the nucleus in cardiac hypertrophy. FGF13 deficiency inhibits NF-κB activation in ISO-treated NRCMs and TAC-surgery mouse hearts, whereas FGF13 overexpression shows the opposite trend. Moreover, FGF13 overexpression alone is sufficient to activate NF-κB in cardiomyocytes. The interaction between FGF13 and p65 or the effects of FGF13 on NF-κB have nothing to do with IκB. Together, an IκB-independent mechanism for NF-κB regulation has been revealed in cardiomyocytes both under basal and stressful conditions, suggesting the promising application of FGF13 as a therapeutic target for pathological cardiac hypertrophy and heart failure. Endogenous FGF13 is up-regulated in cardiomyocytes under pressure overload FGF13 directly interacts with p65 Forced FGF13 overexpression activates NF-κB in cardiomyocytes
Collapse
Affiliation(s)
- Jia Sun
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Chao Niu
- Pediatric Research Institute, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Weijian Ye
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Ning An
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Gen Chen
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Xiaozhong Huang
- Department of Pediatric Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Jianan Wang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Xixi Chen
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Yingjie Shen
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Shuai Huang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Ying Wang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Xu Wang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Yang Wang
- Department of Histology and Embryology, Institute of Neuroscience, Wenzhou Medical University Wenzhou, 325000, China
| | - Litai Jin
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Weitao Cong
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Xiaokun Li
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| |
Collapse
|
23
|
Yang J, Zhang H, Wang X, Guo J, Wei L, Song Y, Luo Y, Zhao Y, Subramaniam M, Spelsberg TC, Wang L, Xu W, Li M. Kruppel-like factor 10 protects against acute viral myocarditis by negatively regulating cardiac MCP-1 expression. Cell Mol Immunol 2020; 18:2236-2248. [PMID: 32895486 DOI: 10.1038/s41423-020-00539-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 12/18/2022] Open
Abstract
Viral myocarditis (VMC) is a cardiac disease associated with myocardial inflammation and injury induced by virus infection. Cardiomyocytes have recently been regarded as key players in eliciting and modulating inflammation within the myocardium. Kruppel-like factor 10 (KLF10) is a crucial regulator of various pathological processes and plays different roles in a variety of diseases. However, its role in VMC induced by coxsackievirus B3 (CVB3) infection remains unknown. In this study, we report that cardiac KLF10 confers enhanced protection against viral myocarditis. We found that KLF10 expression was downregulated upon CVB3 infection. KLF10 deficiency enhanced cardiac viral replication and aggravated VMC progress. Bone marrow chimera experiments indicated that KLF10 expression in nonhematopoietic cells was involved in the pathogenesis of VMC. We further identified MCP-1 as a novel target of KLF10 in cardiomyocytes, and KLF10 cooperated with histone deacetylase 1 (HDAC1) to negatively regulate MCP-1 expression by binding its promoter, leading to activation of MCP-1 transcription and recruitment of Ly6Chigh monocytes/macrophages into the myocardium. This novel mechanism of MCP-1 regulation by KLF10 might provide new insights into the pathogenesis of VMC and a potential therapeutic target for VMC.
Collapse
Affiliation(s)
- Jie Yang
- Institute of Biology and Medical Sciences, Soochow University, Building 703, 199 Ren-ai Road, 215123, Suzhou, China
| | - Hongkai Zhang
- Institute of Biology and Medical Sciences, Soochow University, Building 703, 199 Ren-ai Road, 215123, Suzhou, China
| | - Xuelian Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Jing Guo
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Lin Wei
- Institute of Biology and Medical Sciences, Soochow University, Building 703, 199 Ren-ai Road, 215123, Suzhou, China
| | - Yahui Song
- Institute of Biology and Medical Sciences, Soochow University, Building 703, 199 Ren-ai Road, 215123, Suzhou, China
| | - Yuan Luo
- Institute of Biology and Medical Sciences, Soochow University, Building 703, 199 Ren-ai Road, 215123, Suzhou, China
| | - YinXia Zhao
- Central Laboratory, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, 200031, Shanghai, China
| | | | - Thomas C Spelsberg
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Lie Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China.
| | - Wei Xu
- Institute of Biology and Medical Sciences, Soochow University, Building 703, 199 Ren-ai Road, 215123, Suzhou, China.
| | - Min Li
- Institute of Biology and Medical Sciences, Soochow University, Building 703, 199 Ren-ai Road, 215123, Suzhou, China.
| |
Collapse
|
24
|
VEGF-A in Cardiomyocytes and Heart Diseases. Int J Mol Sci 2020; 21:ijms21155294. [PMID: 32722551 PMCID: PMC7432634 DOI: 10.3390/ijms21155294] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 12/11/2022] Open
Abstract
The vascular endothelial growth factor (VEGF), a homodimeric vasoactive glycoprotein, is the key mediator of angiogenesis. Angiogenesis, the formation of new blood vessels, is responsible for a wide variety of physio/pathological processes, including cardiovascular diseases (CVD). Cardiomyocytes (CM), the main cell type present in the heart, are the source and target of VEGF-A and express its receptors, VEGFR1 and VEGFR2, on their cell surface. The relationship between VEGF-A and the heart is double-sided. On the one hand, VEGF-A activates CM, inducing morphogenesis, contractility and wound healing. On the other hand, VEGF-A is produced by CM during inflammation, mechanical stress and cytokine stimulation. Moreover, high concentrations of VEGF-A have been found in patients affected by different CVD, and are often correlated with an unfavorable prognosis and disease severity. In this review, we summarized the current knowledge about the expression and effects of VEGF-A on CM and the role of VEGF-A in CVD, which are the most important cause of disability and premature death worldwide. Based on clinical studies on angiogenesis therapy conducted to date, it is possible to think that the control of angiogenesis and VEGF-A can lead to better quality and span of life of patients with heart disease.
Collapse
|
25
|
Firoozi S, Pahlavan S, Ghanian MH, Rabbani S, Tavakol S, Barekat M, Yakhkeshi S, Mahmoudi E, Soleymani M, Baharvand H. A Cell-Free SDKP-Conjugated Self-Assembling Peptide Hydrogel Sufficient for Improvement of Myocardial Infarction. Biomolecules 2020; 10:E205. [PMID: 32019267 PMCID: PMC7072713 DOI: 10.3390/biom10020205] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/12/2020] [Accepted: 01/25/2020] [Indexed: 01/01/2023] Open
Abstract
Biomaterials in conjunction with stem cell therapy have recently attracted attention as a new therapeutic approach for myocardial infarction (MI), with the aim to solve the delivery challenges that exist with transplanted cells. Self-assembling peptide (SAP) hydrogels comprise a promising class of synthetic biomaterials with cardiac-compatible properties such as mild gelation, injectability, rehealing ability, and potential for sequence modification. Herein, we developed an SAP hydrogel composed of a self-assembling gel-forming core sequence (RADA) modified with SDKP motif with pro-angiogenic and anti-fibrotic activity to be used as a cardioprotective scaffold. The RADA-SDKP hydrogel was intramyocardially injected into the infarct border zone of a rat model of MI induced by left anterior descending artery (LAD) ligation as a cell-free or a cell-delivering scaffold for bone marrow mesenchymal stem cells (BM-MSCs). The left ventricular ejection fraction (LVEF) was markedly improved after transplantation of either free hydrogel or cell-laden hydrogel. This cardiac functional repair coincided very well with substantially lower fibrotic tissue formation, expanded microvasculature, and lower inflammatory response in the infarct area. Interestingly, BM-MSCs alone or in combination with hydrogel could not surpass the cardiac repair effects of the SDKP-modified SAP hydrogel. Taken together, we suggest that the RADA-SDKP hydrogel can be a promising cell-free construct that has the capability for functional restoration in the instances of acute myocardial infarction (AMI) that might minimize the safety concerns of cardiac cell therapy and facilitate clinical extrapolation.
Collapse
Affiliation(s)
- Saman Firoozi
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran;
| | - Sara Pahlavan
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran; (S.P.); (S.Y.)
| | - Mohammad-Hossein Ghanian
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran;
| | - Shahram Rabbani
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran 1416753955, Iran;
| | - Shima Tavakol
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran;
| | - Maryam Barekat
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran;
| | - Saeed Yakhkeshi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran; (S.P.); (S.Y.)
| | - Elena Mahmoudi
- Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA;
| | - Mansoureh Soleymani
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran;
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran; (S.P.); (S.Y.)
- Department of Developmental Biology, University of Science and Culture, ACECR, Tehran 1461968151, Iran
| |
Collapse
|
26
|
Aluganti Narasimhulu C, Singla DK. The Role of Bone Morphogenetic Protein 7 (BMP-7) in Inflammation in Heart Diseases. Cells 2020; 9:cells9020280. [PMID: 31979268 PMCID: PMC7073173 DOI: 10.3390/cells9020280] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 01/17/2020] [Accepted: 01/21/2020] [Indexed: 12/31/2022] Open
Abstract
Bone morphogenetic protein-7 is (BMP-7) is a potent anti-inflammatory growth factor belonging to the Transforming Growth Factor Beta (TGF-β) superfamily. It plays an important role in various biological processes, including embryogenesis, hematopoiesis, neurogenesis and skeletal morphogenesis. BMP-7 stimulates the target cells by binding to specific membrane-bound receptor BMPR 2 and transduces signals through mothers against decapentaplegic (Smads) and mitogen activated protein kinase (MAPK) pathways. To date, rhBMP-7 has been used clinically to induce the differentiation of mesenchymal stem cells bordering the bone fracture site into chondrocytes, osteoclasts, the formation of new bone via calcium deposition and to stimulate the repair of bone fracture. However, its use in cardiovascular diseases, such as atherosclerosis, myocardial infarction, and diabetic cardiomyopathy is currently being explored. More importantly, these cardiovascular diseases are associated with inflammation and infiltrated monocytes where BMP-7 has been demonstrated to be a key player in the differentiation of pro-inflammatory monocytes, or M1 macrophages, into anti-inflammatory M2 macrophages, which reduces developed cardiac dysfunction. Therefore, this review focuses on the molecular mechanisms of BMP-7 treatment in cardiovascular disease and its role as an anti-fibrotic, anti-apoptotic and anti-inflammatory growth factor, which emphasizes its potential therapeutic significance in heart diseases.
Collapse
|
27
|
Ranjan P, Kumari R, Verma SK. Cardiac Fibroblasts and Cardiac Fibrosis: Precise Role of Exosomes. Front Cell Dev Biol 2019; 7:318. [PMID: 31867328 PMCID: PMC6904280 DOI: 10.3389/fcell.2019.00318] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 11/20/2019] [Indexed: 12/12/2022] Open
Abstract
Exosomes are a group of extracellular microvesicles that deliver biologically active RNAs, proteins, lipids and other signaling molecules to recipient cells. Classically, exosomes act as a vehicle by which cells or organs communicate with each other to maintain cellular/tissue homeostasis and to respond to pathological stress. Most multicellular systems, including the cardiovascular system, use exosomes for intercellular communication. In heart, endogenous exosomes from cardiac cells or stem cells aid in regulation of cell survival, cell proliferation and cell death; and thus tightly regulate cardiac biology and repair processes. Pathological stimulus in heart alters secretion and molecular composition of exosomes, thus influencing the above processes. The past decade has yielded increasing interest in the role of exosomes in the cardiovascular system and significant contribution of cardiac fibroblast (CF) and mediated cardiac fibrosis in heart failure, in this review we had overviewed the relevant literatures about fibroblast exosomes, its effect in the cardiovascular biology and its impact on cardiovascular disease (CVD). This review briefly describes the communication between fibroblasts and other cardiac cells via exosomes, the influence of such on myocardial fibrosis and remodeling, and the possibilities to use exosomes as biomarkers for acute and chronic heart diseases.
Collapse
Affiliation(s)
- Prabhat Ranjan
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Rajesh Kumari
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Suresh Kumar Verma
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL, United States.,Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
28
|
Post-TTM Rebound Pyrexia after Ischemia-Reperfusion Injury Results in Sterile Inflammation and Apoptosis in Cardiomyocytes. Mediators Inflamm 2019; 2019:6431957. [PMID: 31871429 PMCID: PMC6906799 DOI: 10.1155/2019/6431957] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 11/04/2019] [Indexed: 01/25/2023] Open
Abstract
Introduction Fever is frequently observed after acute ischemic events and is associated with poor outcome and higher mortality. Targeted temperature management (TTM) is recommended for neuroprotection in comatose cardiac arrest survivors, but pyrexia after rewarming is proven to be detrimental in clinical trials. However, the cellular mechanisms and kinetics of post-TTM rebound pyrexia remain to be elucidated. Therefore, we investigated the effects of cooling and post-TTM pyrexia on the inflammatory response and apoptosis in a cardiomyocyte ischemia-reperfusion (IR) injury model. Methods HL-1 cardiomyocytes were divided into the following groups to investigate the effect of oxygen-glucose deprivation/reperfusion (OGD/R), hypothermia (33.5°C), and pyrexia (40°C): normoxia controls maintained at 37°C and warmed to 40°C, OGD/R groups maintained at 37°C and cooled to 33.5°C for 24 h with rewarming to 37°C, and OGD/R pyrexia groups further warmed from 37 to 40°C. Caspase-3 and RBM3 were assessed by Western blot and TNF-α, IL-6, IL-1β, SOCS3, iNOS, and RBM3 transcriptions by RT-qPCR. Results OGD-induced oxidative stress (iNOS) in cardiomyocytes was attenuated post-TTM by cooling. Cytokine transcriptions were suppressed by OGD, while reperfusion induced significant TNF-α transcription that was exacerbated by cooling. Significant inductions of TNF-α, IL-6, IL-1β, and SOCS3 were observed in noncooled, but not in cooled and rewarmed, OGD/R-injured cardiomyocytes. Further warming to pyrexia induced a sterile inflammatory response in OGD/R-injured groups that was attenuated by previous cooling, but no inflammation was observed in pyrexic normoxia groups. Moreover, cytoprotective RBM3 expression was induced by cooling but suppressed by pyrexia, correlating with apoptotic caspase-3 activation. Conclusion Our findings show that maintaining a period of post-TTM “therapeutic normothermia” is effective in preventing secondary apoptosis-driven myocardial cell death, thus minimizing the infarct area and further release of mediators of the innate sterile inflammatory response after acute IR injury.
Collapse
|
29
|
Nystoriak MA, Kilfoil PJ, Lorkiewicz PK, Ramesh B, Kuehl PJ, McDonald J, Bhatnagar A, Conklin DJ. Comparative effects of parent and heated cinnamaldehyde on the function of human iPSC-derived cardiac myocytes. Toxicol In Vitro 2019; 61:104648. [PMID: 31518667 DOI: 10.1016/j.tiv.2019.104648] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 08/19/2019] [Accepted: 09/09/2019] [Indexed: 12/31/2022]
Abstract
Many e-cigarette products contain cinnamaldehyde as a primary constituent of cinnamon flavorings. When used as a food additive, cinnamaldehyde is generally regarded as safe for ingestion. However, little is known about the effects of cinnamaldehyde or its degradation products, generated after heating and inhalation, which may lead to elevated circulatory exposure to the heart. Hence, in this study, we tested the in vitro cardiac toxicity of cinnamaldehyde and its thermal degradation products generated by heating at low (200 ± 50 °C) and high temperatures (700 ± 50 °C) on the contractility, rhythmicity and electrical signaling properties of human induced pluripotent stem cell-derived cardiac myocytes (hiPSC-CMs). Cellular impedance measurements on spontaneously beating hiPSC-CMs revealed that cinnamaldehyde significantly alters contraction-dependent signal amplitude, beating rate, and cell morphology. These effects were attenuated after cinnamaldehyde was subjected to heating at low or high temperatures. Current clamp analysis of hiPSC-CM action potentials (APs) showed only modest effects of acute application of 1-100 μM cinnamaldehyde on resting membrane potential, while prolonged (~20 min) application of 100 μM cinnamaldehyde resulted in progressive depolarization and loss of rhythmic AP spiking activity. Collectively, these results suggest that micromolar levels of cinnamaldehyde could alter cardiac excitability, in part by impairing the processes that regulate membrane potential and depolarization. Our results further suggest that heating cinnamaldehyde by itself does not directly lead to the formation of products with greater cardiotoxicity in vitro.
Collapse
Affiliation(s)
- Matthew A Nystoriak
- American Heart Association Tobacco Regulation and Addiction Center, University of Louisville, Louisville, KY 40202, United States of America; Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, KY 40202, United States of America; Diabetes and Obesity Center, Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY, 40202, United States of America; Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, United States of America.
| | - Peter J Kilfoil
- Diabetes and Obesity Center, Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY, 40202, United States of America
| | - Pawel K Lorkiewicz
- American Heart Association Tobacco Regulation and Addiction Center, University of Louisville, Louisville, KY 40202, United States of America; Diabetes and Obesity Center, Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY, 40202, United States of America
| | - Bhargav Ramesh
- Diabetes and Obesity Center, Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY, 40202, United States of America
| | - Philip J Kuehl
- Lovelace Biomedical, Albuquerque, NM 87108-5127, United States of America
| | - Jacob McDonald
- Lovelace Biomedical, Albuquerque, NM 87108-5127, United States of America
| | - Aruni Bhatnagar
- American Heart Association Tobacco Regulation and Addiction Center, University of Louisville, Louisville, KY 40202, United States of America; Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, KY 40202, United States of America; Diabetes and Obesity Center, Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY, 40202, United States of America; Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, United States of America
| | - Daniel J Conklin
- American Heart Association Tobacco Regulation and Addiction Center, University of Louisville, Louisville, KY 40202, United States of America; Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, KY 40202, United States of America; Diabetes and Obesity Center, Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY, 40202, United States of America; Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, United States of America
| |
Collapse
|
30
|
Kuo FY, Lee SP, Cheng JT, Wu MC. The direct effect of lipopolysaccharide on an isolated heart is different from the effect on cardiac myocytes in vitro. Arch Med Sci 2019; 19:216-228. [PMID: 36817673 PMCID: PMC9897085 DOI: 10.5114/aoms.2019.86976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Accepted: 07/04/2019] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Lipopolysaccharide (LPS) is widely used to induce experimental animals. However, its effects on cardiac contraction is controversial. Although LPS probably induces its influence in vivo both directly and indirectly, we focused on the direct effects of LPS in this report. MATERIAL AND METHODS Isolated ventricular myocytes mounted on a Langendorff apparatus were perfused with LPS. The changes in cultured H9c2 cells incubated with LPS over a 3-h exposure were compared with the changes after a 24-h incubation. Apoptosis was identified using flow cytometry and Western blotting. The mRNA levels were also determined. RESULTS LPS directly stimulated cardiac contractility at low doses, although it produced inhibition at higher doses. The TLR4-coupled JAK2/STAT3 pathway was identified in H9c2 cells after LPS treatment, with an increase in intracellular calcium levels. LPS dose-dependently activated hypertrophic signals in H9c2 cells and induced apoptosis at the high dose. However, apoptosis was observed in H9c2 cells after a 24-h exposure to LPS, even at low doses. This observation appears to be associated with the level of paracrine cytokines. Changes in H9c2 cells by LPS were diminished by NPS2390, an inhibitor of the calcium-sensing receptor (CaSR). LPS also promoted CaSR mRNA expression in H9c2 cells, which may be unrelated to the changes in cytokine expression influenced by an inflammasome inhibitor. CONCLUSIONS In contrast to the isolated hearts, LPS activated hypertrophic signals prior to apoptotic signals in cardiac cells. Thus, LPS injury appears to be associated with CaSR, which was not markedly influenced by an inflammasome inhibitor.
Collapse
Affiliation(s)
- Feng Yu Kuo
- Department of Food Science, College of Agriculture, National Pingtung University of Science and Technology, Pingtung, Taiwan
- Cardiovascular Centre, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Shu Ping Lee
- Department of Food Science, College of Agriculture, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Juei-Tang Cheng
- Department of Medical Research, Chi-Mei Medical Centre, Tainan, Taiwan
- Institute of Medical Science, College of Health Science, Chang Jung Christian University, Tainan, Taiwan
| | - Ming Chang Wu
- Department of Food Science, College of Agriculture, National Pingtung University of Science and Technology, Pingtung, Taiwan
| |
Collapse
|
31
|
Nguyen AH, Marsh P, Schmiess-Heine L, Burke PJ, Lee A, Lee J, Cao H. Cardiac tissue engineering: state-of-the-art methods and outlook. J Biol Eng 2019; 13:57. [PMID: 31297148 PMCID: PMC6599291 DOI: 10.1186/s13036-019-0185-0] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 06/03/2019] [Indexed: 12/17/2022] Open
Abstract
The purpose of this review is to assess the state-of-the-art fabrication methods, advances in genome editing, and the use of machine learning to shape the prospective growth in cardiac tissue engineering. Those interdisciplinary emerging innovations would move forward basic research in this field and their clinical applications. The long-entrenched challenges in this field could be addressed by novel 3-dimensional (3D) scaffold substrates for cardiomyocyte (CM) growth and maturation. Stem cell-based therapy through genome editing techniques can repair gene mutation, control better maturation of CMs or even reveal its molecular clock. Finally, machine learning and precision control for improvements of the construct fabrication process and optimization in tissue-specific clonal selections with an outlook of cardiac tissue engineering are also presented.
Collapse
Affiliation(s)
- Anh H. Nguyen
- Electrical and Computer Engineering Department, University of Alberta, Edmonton, Alberta Canada
- Electrical Engineering and Computer Science Department, University of California Irvine, Irvine, CA USA
| | - Paul Marsh
- Electrical Engineering and Computer Science Department, University of California Irvine, Irvine, CA USA
| | - Lauren Schmiess-Heine
- Electrical Engineering and Computer Science Department, University of California Irvine, Irvine, CA USA
| | - Peter J. Burke
- Electrical Engineering and Computer Science Department, University of California Irvine, Irvine, CA USA
- Biomedical Engineering Department, University of California Irvine, Irvine, CA USA
- Chemical Engineering and Materials Science Department, University of California Irvine, Irvine, CA USA
| | - Abraham Lee
- Biomedical Engineering Department, University of California Irvine, Irvine, CA USA
- Mechanical and Aerospace Engineering Department, University of California Irvine, Irvine, CA USA
| | - Juhyun Lee
- Bioengineering Department, University of Texas at Arlington, Arlington, TX USA
| | - Hung Cao
- Electrical Engineering and Computer Science Department, University of California Irvine, Irvine, CA USA
- Biomedical Engineering Department, University of California Irvine, Irvine, CA USA
- Henry Samueli School of Engineering, University of California, Irvine, USA
| |
Collapse
|
32
|
Gardner GT, Travers JG, Qian J, Liu GS, Haghighi K, Robbins N, Jiang M, Li Y, Fan GC, Rubinstein J, Blaxall BC, Kranias EG. Phosphorylation of Hsp20 Promotes Fibrotic Remodeling and Heart Failure. ACTA ACUST UNITED AC 2019; 4:188-199. [PMID: 31061921 PMCID: PMC6488766 DOI: 10.1016/j.jacbts.2018.11.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 09/27/2018] [Accepted: 11/14/2018] [Indexed: 01/28/2023]
Abstract
Cardiomyocyte-specific increases in phosphorylated Hsp20 (S16D-Hsp20) to levels similar to those observed in human failing hearts are associated with early fibrotic remodeling and depressed left ventricular function, symptoms which progress to heart failure and early death. The underlying mechanisms appear to involve translocation of phosphorylated Hsp20 to the nucleus and upregulation of interleukin (IL)-6, which subsequently activates cardiac fibroblasts in a paracrine fashion through transcription factor STAT3 signaling. Accordingly, treatment of S16D-Hsp20 mice with a rat anti-mouse IL-6 receptor monoclonal antibody (MR16-1) attenuated interstitial fibrosis and preserved cardiac function. These findings suggest that phosphorylated Hsp20 may be a potential therapeutic target in heart failure.
Collapse
Key Words
- Ccl2, C-C motif chemokine ligand 2
- Ccl3, C-C motif chemokine ligand 3
- Col1a1, collagen 1A1
- Col3A1, collagen 3A1
- ECM, extra-cellular matrix
- Hsp, heat shock protein
- Hsp20
- I/R, ischemia/reperfusion
- IL, interleukin
- IL-6
- Postn, periostin
- SMA, smooth muscle actin
- STAT3, signal transducer and activator of transcription 3
- TG, transgenic
- TGF, transforming growth factor
- TNF, tumor necrosis factor
- TUNEL, terminal deoxynucleotidyl transferase dUTP nick end labeling
- WT, wild type
- fibroblast
- heart failure
- remodeling
Collapse
Affiliation(s)
- George T Gardner
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Joshua G Travers
- Department of Pediatrics, Division of Molecular Cardiovascular Biology, The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Jiang Qian
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Guan-Sheng Liu
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Kobra Haghighi
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Nathan Robbins
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Min Jiang
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Yutian Li
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Guo-Chang Fan
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Jack Rubinstein
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Burns C Blaxall
- Department of Pediatrics, Division of Molecular Cardiovascular Biology, The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Evangelia G Kranias
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio.,Molecular Biology Division, Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| |
Collapse
|
33
|
Yang X, Li X, Yuan M, Tian C, Yang Y, Wang X, Zhang X, Sun Y, He T, Han S, Chen G, Liu N, Gao Y, Hu D, Xing Y, Shang H. Anticancer Therapy-Induced Atrial Fibrillation: Electrophysiology and Related Mechanisms. Front Pharmacol 2018; 9:1058. [PMID: 30386232 PMCID: PMC6198283 DOI: 10.3389/fphar.2018.01058] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 09/03/2018] [Indexed: 12/19/2022] Open
Abstract
Some well-established immunotherapy, radiotherapy, postoperation, anticancer drugs such as anthracyclines, antimetabolites, human epidermal growth factor receptor 2 blockers, tyrosine kinase inhibitors, alkylating agents, checkpoint inhibitors, and angiogenesis inhibitors, are significantly linked to cardiotoxicity. Cardiotoxicity is a common complication of several cancer treatments. Some studies observed complications of cardiac arrhythmia associated with the treatment of cancer, including atrial fibrillation (AF), supraventricular arrhythmias, and cardiac repolarization abnormalities. AF increases the risk of cardiovascular morbidity and mortality; it is associated with an almost doubled risk of mortality and a nearly 5-fold increase in the risk of stroke. The occurrence of AF is also usually researched in patients with advanced cancer and those undergoing active cancer treatments. During cancer treatments, the incidence rate of AF affects the prognosis of tumor treatment and challenges the treatment strategy. The present article is mainly focused on the cardiotoxicity of cancer treatments. In our review, we discuss these anticancer therapies and how they induce AF and consequently provide information on the precaution of AF during cancer treatment.
Collapse
Affiliation(s)
- Xinyu Yang
- Guang'an men Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China.,Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Xinye Li
- Guang'an men Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China.,Beijing University of Chinese Medicine, Beijing, China
| | - Mengchen Yuan
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Chao Tian
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Yihan Yang
- Guang'an men Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China.,Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Xiaofeng Wang
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoyu Zhang
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Yang Sun
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Tianmai He
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Songjie Han
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Guang Chen
- Guang'an men Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Nian Liu
- Department of Cardiology, Beijing An Zhen Hospital of the Capital University of Medical Sciences, Beijing, China
| | - Yonghong Gao
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Dan Hu
- Department of Cardiology and Cardiovascular Research Institute, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yanwei Xing
- Guang'an men Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Hongcai Shang
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China.,Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
34
|
Willeford A, Suetomi T, Nickle A, Hoffman HM, Miyamoto S, Heller Brown J. CaMKIIδ-mediated inflammatory gene expression and inflammasome activation in cardiomyocytes initiate inflammation and induce fibrosis. JCI Insight 2018; 3:97054. [PMID: 29925681 DOI: 10.1172/jci.insight.97054] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 05/14/2018] [Indexed: 01/08/2023] Open
Abstract
Inflammation accompanies heart failure and is a mediator of cardiac fibrosis. CaMKIIδ plays an essential role in adverse remodeling and decompensation to heart failure. We postulated that inflammation is the mechanism by which CaMKIIδ contributes to adverse remodeling in response to nonischemic interventions. We demonstrate that deletion of CaMKIIδ in the cardiomyocyte (CKO) significantly attenuates activation of NF-κB, expression of inflammatory chemokines and cytokines, and macrophage accumulation induced by angiotensin II (Ang II) infusion. The inflammasome was activated by Ang II, and this response was also diminished in CKO mice. These events occurred prior to any evidence of Ang II-induced cell death. In addition, CaMKII-dependent inflammatory gene expression and inflammasome priming were observed as early as the third hour of infusion, a time point at which macrophage recruitment was not evident. Inhibition of either the inflammasome or monocyte chemoattractant protein 1 (MCP1) signaling attenuated macrophage accumulation, and these interventions, like cardiomyocyte CaMKIIδ deletion, diminished the fibrotic response to Ang II. Thus, activation of CaMKIIδ in the cardiomyocyte represents what we believe to be a novel mechanism for initiating inflammasome activation and an inflammatory gene program that leads to macrophage recruitment and ultimately to development of fibrosis.
Collapse
Affiliation(s)
| | | | | | - Hal M Hoffman
- Department of Medicine, and.,Department of Pediatrics, UCSD, La Jolla, California, USA
| | | | | |
Collapse
|
35
|
An Injectable Oxygen Release System to Augment Cell Survival and Promote Cardiac Repair Following Myocardial Infarction. Sci Rep 2018; 8:1371. [PMID: 29358595 PMCID: PMC5778078 DOI: 10.1038/s41598-018-19906-w] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 01/10/2018] [Indexed: 01/15/2023] Open
Abstract
Oxygen deficiency after myocardial infarction (MI) leads to massive cardiac cell death. Protection of cardiac cells and promotion of cardiac repair are key therapeutic goals. These goals may be achieved by re-introducing oxygen into the infarcted area. Yet current systemic oxygen delivery approaches cannot efficiently diffuse oxygen into the infarcted area that has extremely low blood flow. In this work, we developed a new oxygen delivery system that can be delivered specifically to the infarcted tissue, and continuously release oxygen to protect the cardiac cells. The system was based on a thermosensitive, injectable and fast gelation hydrogel, and oxygen releasing microspheres. The fast gelation hydrogel was used to increase microsphere retention in the heart tissue. The system was able to continuously release oxygen for 4 weeks. The released oxygen significantly increased survival of cardiac cells under the hypoxic condition (1% O2) mimicking that of the infarcted hearts. It also reduced myofibroblast formation under hypoxic condition (1% O2). After implanting into infarcted hearts for 4 weeks, the released oxygen significantly augmented cell survival, decreased macrophage density, reduced collagen deposition and myofibroblast density, and stimulated tissue angiogenesis, leading to a significant increase in cardiac function.
Collapse
|
36
|
Boerrigter D, Weickert TW, Lenroot R, O'Donnell M, Galletly C, Liu D, Burgess M, Cadiz R, Jacomb I, Catts VS, Fillman SG, Weickert CS. Using blood cytokine measures to define high inflammatory biotype of schizophrenia and schizoaffective disorder. J Neuroinflammation 2017; 14:188. [PMID: 28923068 PMCID: PMC5604300 DOI: 10.1186/s12974-017-0962-y] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 09/07/2017] [Indexed: 12/11/2022] Open
Abstract
Background Increases in pro-inflammatory cytokines are found in the brain and blood of people with schizophrenia. However, increased cytokines are not evident in all people with schizophrenia, but are found in a subset. The cytokine changes that best define this subset, termed the “elevated inflammatory biotype”, are still being identified. Methods Using quantitative RT-PCR, we measured five cytokine mRNAs (IL-1β, IL-2 IL-6, IL-8 and IL-18) from peripheral blood of healthy controls and of people with schizophrenia or schizoaffective disorder (n = 165). We used a cluster analysis of the transcript levels to define those with low and those with elevated levels of cytokine expression. From the same cohort, eight cytokine proteins (IL-1β, IL-2, IL-6, IL-8, IL-10, IL-12, IFNγ and TNFα) were measured in serum and plasma using a Luminex Magpix-based assay. We compared peripheral mRNA and protein levels across diagnostic groups and between those with low and elevated levels of cytokine expression according to our transcription-based cluster analysis. Results We found an overall decrease in the anti-inflammatory IL-2 mRNA (p = 0.006) and an increase in three serum cytokines, IL-6 (p = 0.010), IL-8 (p = 0.024) and TNFα (p < 0.001) in people with schizophrenia compared to healthy controls. A greater percentage of people with schizophrenia (48%) were categorised into the elevated inflammatory biotype compared to healthy controls (33%). The magnitude of increase in IL-1β, IL-6, IL-8 and IL-10 mRNAs in people in the elevated inflammation biotype ranged from 100 to 220% of those in the non-elevated inflammatory biotype and was comparable between control and schizophrenia groups. Blood cytokine protein levels did not correlate with cytokine mRNA levels, and plasma levels of only two cytokines distinguished the elevated and low inflammatory biotypes, with IL-1β significantly increased in the elevated cytokine control group and IL-8 significantly increased in the elevated cytokine schizophrenia group. Conclusions Our results confirm that individuals with schizophrenia are more likely to have elevated levels of inflammation compared to controls. We suggest that efforts to define inflammatory status based on peripheral measures need to consider both mRNA and protein measures as each have distinct advantages and disadvantages and can yield different results. Electronic supplementary material The online version of this article (10.1186/s12974-017-0962-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Danny Boerrigter
- Neuroscience Research Australia, and Schizophrenia Research Institute, Barker Street, Randwick, New South Wales, 2031, Australia
| | - Thomas W Weickert
- Neuroscience Research Australia, and Schizophrenia Research Institute, Barker Street, Randwick, New South Wales, 2031, Australia.,School of Psychiatry, University of New South Wales, Kensington, New South Wales, Australia
| | - Rhoshel Lenroot
- Neuroscience Research Australia, and Schizophrenia Research Institute, Barker Street, Randwick, New South Wales, 2031, Australia.,School of Psychiatry, University of New South Wales, Kensington, New South Wales, Australia
| | - Maryanne O'Donnell
- School of Psychiatry, University of New South Wales, Kensington, New South Wales, Australia
| | - Cherrie Galletly
- Discipline of Psychiatry, School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia.,Northern Adelaide Local Health Network, Adelaide, South Australia, Australia.,Ramsay Health Care (SA) Mental Health, Adelaide, South Australia, Australia
| | - Dennis Liu
- Discipline of Psychiatry, School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia.,Northern Adelaide Local Health Network, Adelaide, South Australia, Australia
| | - Martin Burgess
- Neuroscience Research Australia, and Schizophrenia Research Institute, Barker Street, Randwick, New South Wales, 2031, Australia
| | - Roxanne Cadiz
- Neuroscience Research Australia, and Schizophrenia Research Institute, Barker Street, Randwick, New South Wales, 2031, Australia
| | - Isabella Jacomb
- Neuroscience Research Australia, and Schizophrenia Research Institute, Barker Street, Randwick, New South Wales, 2031, Australia
| | - Vibeke S Catts
- Neuroscience Research Australia, and Schizophrenia Research Institute, Barker Street, Randwick, New South Wales, 2031, Australia.,School of Psychiatry, University of New South Wales, Kensington, New South Wales, Australia
| | - Stu G Fillman
- Neuroscience Research Australia, and Schizophrenia Research Institute, Barker Street, Randwick, New South Wales, 2031, Australia.,School of Psychiatry, University of New South Wales, Kensington, New South Wales, Australia
| | - Cynthia Shannon Weickert
- Neuroscience Research Australia, and Schizophrenia Research Institute, Barker Street, Randwick, New South Wales, 2031, Australia. .,School of Psychiatry, University of New South Wales, Kensington, New South Wales, Australia.
| |
Collapse
|
37
|
Freeman ML, Shive CL, Nguyen TP, Younes SA, Panigrahi S, Lederman MM. Cytokines and T-Cell Homeostasis in HIV Infection. J Infect Dis 2017; 214 Suppl 2:S51-7. [PMID: 27625431 DOI: 10.1093/infdis/jiw287] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Untreated human immunodeficiency virus (HIV) infection is characterized by progressive CD4(+) T-cell depletion and CD8(+) T-cell expansion, and CD4(+) T-cell depletion is linked directly to the risk for opportunistic infections and infection-associated mortality. With suppression of HIV replication by antiretroviral therapy, circulating CD4(+) Tcell numbers typically improve while CD8(+) T-cell expansion persists, and both CD4(+) T-cell cytopenia and CD8(+) T-cell expansion are associated with morbidity and mortality. In this brief review, we report on the role that selected homeostatic and inflammatory cytokines may play both in the failure of CD4(+) T-cell restoration and the CD8(+) T-cell expansion that characterize HIV infection.
Collapse
Affiliation(s)
- Michael L Freeman
- Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, University Hospitals/Case Medical Center
| | - Carey L Shive
- Department of Veterans Affairs, Cleveland VA Medical Center, Ohio
| | - Thao P Nguyen
- Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, University Hospitals/Case Medical Center
| | - Souheil-Antoine Younes
- Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, University Hospitals/Case Medical Center
| | - Soumya Panigrahi
- Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, University Hospitals/Case Medical Center
| | - Michael M Lederman
- Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, University Hospitals/Case Medical Center
| |
Collapse
|
38
|
Rivera-Serrano EE, Sherry B. NF-κB activation is cell type-specific in the heart. Virology 2016; 502:133-143. [PMID: 28043025 DOI: 10.1016/j.virol.2016.12.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 12/16/2016] [Accepted: 12/19/2016] [Indexed: 01/31/2023]
Abstract
Viral myocarditis is common and can progress to cardiac failure. Cardiac cell pro-inflammatory responses are critical for viral clearance, however sustained inflammatory responses contribute to cardiac damage. The transcription factor NF-κB regulates expression of many pro-inflammatory cytokines, but basal and induced activation of NF-κB in different cardiac cell types have not been compared. Here, we used primary cultures of cardiac myocytes and cardiac fibroblasts to identify cardiac cell type-specific events. We show that while viral infection readily stimulates activation of NF-κB in cardiac fibroblasts, cardiac myocytes are largely recalcitrant to activation of NF-κB. Moreover, we show that cardiac myocyte subpopulations differ in their NF-κB subcellular localization and identify the cis-Golgi as a cardiac myocyte-specific host compartment. Together, results indicate that NF-κB-dependent signaling in the heart is cardiac cell type-specific, likely reflecting mechanisms that have evolved to balance responses that can be either protective or damaging to the heart.
Collapse
Affiliation(s)
- Efraín E Rivera-Serrano
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, USA; Comparative Medicine Institute, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - Barbara Sherry
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, USA; Comparative Medicine Institute, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA.
| |
Collapse
|
39
|
Al-Rasheed NM, Al-Rasheed NM, Hasan IH, Al-Amin MA, Al-Ajmi HN, Mahmoud AM. Sitagliptin attenuates cardiomyopathy by modulating the JAK/STAT signaling pathway in experimental diabetic rats. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:2095-107. [PMID: 27418808 PMCID: PMC4933570 DOI: 10.2147/dddt.s109287] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Sitagliptin, a dipeptidyl peptidase-4 inhibitor, has been reported to promote cardioprotection in diabetic hearts by limiting hyperglycemia and hyperlipidemia. However, little is known about the involvement of the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway modulation in the cardioprotective effects of sitagliptin. The current study aimed to investigate the protective effects of sitagliptin against diabetic cardiomyopathy (DCM), focusing on the modulation of the JAK/STAT pathway. Diabetes was induced by streptozotocin injection, and rats received sitagliptin orally and daily for 90 days. Diabetic rats exhibited hyperglycemia, hyperlipidemia, and a significant increase in heart-to-body weight (HW/BW) ratio. Serum troponin I and creatine kinase MB, cardiac interleukin-6 (IL-6), lipid peroxidation, and nitric oxide levels showed significant increase in diabetic rats. In contrast, both enzymatic and nonenzymatic antioxidant defenses were significantly declined in the heart of diabetic rats. Histopathological study revealed degenerations, increased collagen deposition in the heart of diabetic rats. Sitagliptin alleviated hyperglycemia, hyperlipidemia, HW/BW ratio, histological architecture, oxidative stress, and inflammation, and rejuvenated the antioxidant defenses. In addition, cardiac levels of pJAK2 and pSTAT3 were increased in diabetic rats, an effect which was remarkably decreased after sitagliptin treatment. In conclusion, these results confer an evidence that sitagliptin has great therapeutic potential on DCM through down-regulation of the JAK/STAT signaling pathway.
Collapse
Affiliation(s)
- Nouf M Al-Rasheed
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University
| | - Nawal M Al-Rasheed
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University; Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Iman H Hasan
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University
| | - Maha A Al-Amin
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University
| | - Hanaa N Al-Ajmi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University
| | - Ayman M Mahmoud
- Physiology Division, Department of Zoology, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| |
Collapse
|
40
|
Liu J, Yang S, Zhang X, Liu G, Yue X. Isoflurane reduces oxygen-glucose deprivation-induced oxidative, inflammatory, and apoptotic responses in H9c2 cardiomyocytes. Am J Transl Res 2016; 8:2597-2608. [PMID: 27398143 PMCID: PMC4931154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 06/13/2016] [Indexed: 06/06/2023]
Abstract
Isoflurane (ISO) protects the heart from hypoxia-reperfusion injury. However, the molecular mechanisms of ISO in oxygen-glucose deprivation (OGD)-induced H9c2 cardiomyocyte injury is yet to be understood. Using H9c2 cells cultured in vitro, we examined the cytotoxicity of different doses of ISO (0.7%, 1.4%, and 2.1%) to H9c2 cells and found that 2.1% ISO had significant toxicity to the cell. Thus, 1.4% ISO was selected for the subsequent experiments. ISO notably ameliorated cell viability loss, lactate dehydrogenase release, and creatine kinase activity of H9c2 cells that were treated with OGD. ISO suppressed OGD-induced pro-inflammatory tumor necrosis factor-α, interleukin (IL)-1β, IL-6, IL-8 production, and nuclear factor (NF)-κB activation in H9c2 cells. ISO reduced the reactive oxygen species and malondialdehyde generation, but it enhanced the superoxide dismutase activity in OGD-stimulated H9c2 cells. In addition, diminished OGD-induced cell apoptosis and preserved mitochondrial membrane potential were observed in ISO-treated H9c2 cells. ISO markedly up-regulated the anti-apoptotic Bcl-2 expression but inhibited the pro-apoptotic expressions of Bax, procaspase-3, cleaved caspase-3, and caspase-3 activity. Mechanistically, the cardioprotective effects of ISO on OGD-induced H9c2 cell injury were mediated by the Akt signaling pathway. These findings suggest that ISO alleviates OGD-induced H9c2 cell injury and may therefore be used to prevent and treat ischemic heart diseases.
Collapse
Affiliation(s)
- Jun Liu
- Department of Anesthesiology, The First Affiliated Hospital of Xinxiang Medical University Weihui 453100, Xinxiang, Henan, People's Republic of China
| | - Shuangmei Yang
- Department of Anesthesiology, The First Affiliated Hospital of Xinxiang Medical University Weihui 453100, Xinxiang, Henan, People's Republic of China
| | - Xiaoran Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Xinxiang Medical University Weihui 453100, Xinxiang, Henan, People's Republic of China
| | - Guoze Liu
- Department of Anesthesiology, The First Affiliated Hospital of Xinxiang Medical University Weihui 453100, Xinxiang, Henan, People's Republic of China
| | - Xiuqin Yue
- Department of Anesthesiology, The First Affiliated Hospital of Xinxiang Medical University Weihui 453100, Xinxiang, Henan, People's Republic of China
| |
Collapse
|
41
|
Roberts MA, Tran D, Coulombe KL, Razumova M, Regnier M, Murry CE, Zheng Y. Stromal Cells in Dense Collagen Promote Cardiomyocyte and Microvascular Patterning in Engineered Human Heart Tissue. Tissue Eng Part A 2016; 22:633-44. [PMID: 26955856 PMCID: PMC4840925 DOI: 10.1089/ten.tea.2015.0482] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 02/29/2016] [Indexed: 01/08/2023] Open
Abstract
Cardiac tissue engineering is a strategy to replace damaged contractile tissue and model cardiac diseases to discover therapies. Current cardiac and vascular engineering approaches independently create aligned contractile tissue or perfusable vasculature, but a combined vascularized cardiac tissue remains to be achieved. Here, we sought to incorporate a patterned microvasculature into engineered heart tissue, which balances the competing demands from cardiomyocytes to contract the matrix versus the vascular lumens that need structural support. Low-density collagen hydrogels (1.25 mg/mL) permit human embryonic stem cell-derived cardiomyocytes (hESC-CMs) to form a dense contractile tissue but cannot support a patterned microvasculature. Conversely, high collagen concentrations (density ≥6 mg/mL) support a patterned microvasculature, but the hESC-CMs lack cell-cell contact, limiting their electrical communication, structural maturation, and tissue-level contractile function. When cocultured with matrix remodeling stromal cells, however, hESC-CMs structurally mature and form anisotropic constructs in high-density collagen. Remodeling requires the stromal cells to be in proximity with hESC-CMs. In addition, cocultured cardiac constructs in dense collagen generate measurable active contractions (on the order of 0.1 mN/mm(2)) and can be paced up to 2 Hz. Patterned microvascular networks in these high-density cocultured cardiac constructs remain patent through 2 weeks of culture, and hESC-CMs show electrical synchronization. The ability to maintain microstructural control within engineered heart tissue enables generation of more complex features, such as cellular alignment and a vasculature. Successful incorporation of these features paves the way for the use of large scale engineered tissues for myocardial regeneration and cardiac disease modeling.
Collapse
Affiliation(s)
- Meredith A. Roberts
- Department of Bioengineering, University of Washington, Seattle, Washington
- Center for Cardiovascular Biology, University of Washington, Seattle, Washington
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington
| | - Dominic Tran
- Department of Bioengineering, University of Washington, Seattle, Washington
| | - Kareen L.K. Coulombe
- Center for Cardiovascular Biology, University of Washington, Seattle, Washington
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington
- Department of Pathology, University of Washington, Seattle, Washington
| | - Maria Razumova
- Department of Bioengineering, University of Washington, Seattle, Washington
- Center for Cardiovascular Biology, University of Washington, Seattle, Washington
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington
| | - Michael Regnier
- Department of Bioengineering, University of Washington, Seattle, Washington
- Center for Cardiovascular Biology, University of Washington, Seattle, Washington
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington
| | - Charles E. Murry
- Department of Bioengineering, University of Washington, Seattle, Washington
- Center for Cardiovascular Biology, University of Washington, Seattle, Washington
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington
- Department of Pathology, University of Washington, Seattle, Washington
- Department of Medicine/Cardiology, University of Washington, Seattle, Washington
| | - Ying Zheng
- Department of Bioengineering, University of Washington, Seattle, Washington
- Center for Cardiovascular Biology, University of Washington, Seattle, Washington
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
42
|
Jahng JWS, Song E, Sweeney G. Crosstalk between the heart and peripheral organs in heart failure. Exp Mol Med 2016; 48:e217. [PMID: 26964833 PMCID: PMC4892881 DOI: 10.1038/emm.2016.20] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 12/10/2015] [Accepted: 12/11/2015] [Indexed: 12/31/2022] Open
Abstract
Mediators from peripheral tissues can influence the development and progression of heart failure (HF). For example, in obesity, an altered profile of adipokines secreted from adipose tissue increases the incidence of myocardial infarction (MI). Less appreciated is that heart remodeling releases cardiokines, which can strongly impact various peripheral tissues. Inflammation, and, in particular, activation of the nucleotide-binding oligomerization domain-like receptors with pyrin domain (NLRP3) inflammasome are likely to have a central role in cardiac remodeling and mediating crosstalk with other organs. Activation of the NLRP3 inflammasome in response to cardiac injury induces the production and secretion of the inflammatory cytokines interleukin (IL)-1β and IL-18. In addition to having local effects in the myocardium, these pro-inflammatory cytokines are released into circulation and cause remodeling in the spleen, kidney, skeletal muscle and adipose tissue. The collective effects of various cardiokines on peripheral organs depend on the degree and duration of myocardial injury, with systematic inflammation and peripheral tissue damage observed as HF progresses. In this article, we review mechanisms regulating myocardial inflammation in HF and the role of factors secreted by the heart in communication with peripheral tissues.
Collapse
Affiliation(s)
| | - Erfei Song
- Department of Biology, York University, Toronto, ON, Canada
| | - Gary Sweeney
- Department of Biology, York University, Toronto, ON, Canada
| |
Collapse
|
43
|
Altara R, Manca M, Sabra R, Eid AA, Booz GW, Zouein FA. Temporal cardiac remodeling post-myocardial infarction: dynamics and prognostic implications in personalized medicine. Heart Fail Rev 2015; 21:25-47. [PMID: 26498937 DOI: 10.1007/s10741-015-9513-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Despite dramatic improvements in short-term mortality rates following myocardial infarction (MI), long-term survival for MI patients who progress to heart failure remains poor. MI occurs when the left ventricle (LV) is deprived of oxygen for a sufficient period of time to induce irreversible necrosis of the myocardium. The LV response to MI involves significant tissue, cellular, and molecular level modifications, as well as substantial hemodynamic changes that feedback negatively to amplify the response. Inflammation to remove necrotic myocytes and fibroblast activation to form a scar are key wound healing responses that are highly variable across individuals. Few biomarkers of early remodeling stages are currently clinically adopted. The discovery of underlying pathophysiological mechanisms and associated novel biomarkers has the potential of improving prognostic capability and therapeutic monitoring. Combining these biomarkers with other prominent ones could constitute a powerful diagnostic and prognostic tool that directly reflects the pathophysiological remodeling of the LV. Understanding temporal remodeling at the tissue, cellular, and molecular level and its link to a well-defined set of biomarkers at early stages post-MI is a prerequisite for improving personalized care and devising more successful therapeutic interventions. Here we summarize the integral mechanisms that occur during early cardiac remodeling in the post-MI setting and highlight the most prominent biomarkers for assessing disease progression.
Collapse
Affiliation(s)
- Raffaele Altara
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA.,Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Marco Manca
- DG-DI, Medical Applications, CERN, Geneva, Switzerland
| | - Ramzi Sabra
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Assaad A Eid
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - George W Booz
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Fouad A Zouein
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, USA. .,Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.
| |
Collapse
|
44
|
Interleukin-27 Protects Cardiomyocyte-Like H9c2 Cells against Metabolic Syndrome: Role of STAT3 Signaling. BIOMED RESEARCH INTERNATIONAL 2015; 2015:689614. [PMID: 26339633 PMCID: PMC4538580 DOI: 10.1155/2015/689614] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 03/06/2015] [Accepted: 03/13/2015] [Indexed: 12/12/2022]
Abstract
The present results demonstrated that high glucose (G), salt (S), and cholesterol C (either alone or in combination), as mimicking extracellular changes in metabolic syndrome, damage cardiomyocyte-like H9c2 cells and reduce their viability in a time-dependent manner. However, the effects were greatest when cells were exposed to all three agents (GSC). The mRNA of glycoprotein (gp) 130 and WSX-1, both components of the interleukin (IL)-27 receptor, were present in H9c2 cells. Although mRNA expression was not affected by exogenous treatment with IL-27, the expression of gp130 mRNA (but not that of WSX-1 mRNA) was attenuated by GSC. Treatment of IL-27 to H9c2 cells increased activation of signal transducer and activator of transcription 3 (STAT3) and protected cells from GSC-induced cytochrome c release and cell damage. The protective effects of IL-27 were abrogated by the STAT3 inhibitor, stattic. The results of the present study clearly demonstrate that the STAT3 pathway triggered by anti-inflammatory IL-27 plays a role in protecting cardiomyocytes against GSC-mediated damage.
Collapse
|
45
|
Aoyagi T, Higa JK, Aoyagi H, Yorichika N, Shimada BK, Matsui T. Cardiac mTOR rescues the detrimental effects of diet-induced obesity in the heart after ischemia-reperfusion. Am J Physiol Heart Circ Physiol 2015; 308:H1530-9. [PMID: 25888508 DOI: 10.1152/ajpheart.00008.2015] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 04/10/2015] [Indexed: 01/22/2023]
Abstract
Diet-induced obesity deteriorates the recovery of cardiac function after ischemia-reperfusion (I/R) injury. While mechanistic target of rapamycin (mTOR) is a key mediator of energy metabolism, the effects of cardiac mTOR in ischemic injury under metabolic syndrome remains undefined. Using cardiac-specific transgenic mice overexpressing mTOR (mTOR-Tg mice), we studied the effect of mTOR on cardiac function in both ex vivo and in vivo models of I/R injury in high-fat diet (HFD)-induced obese mice. mTOR-Tg and wild-type (WT) mice were fed a HFD (60% fat by calories) for 12 wk. Glucose intolerance and insulin resistance induced by the HFD were comparable between WT HFD-fed and mTOR-Tg HFD-fed mice. Functional recovery after I/R in the ex vivo Langendorff perfusion model was significantly lower in HFD-fed mice than normal chow diet-fed mice. mTOR-Tg mice demonstrated better cardiac function recovery and had less of the necrotic markers creatine kinase and lactate dehydrogenase in both feeding conditions. Additionally, mTOR overexpression suppressed expression of proinflammatory cytokines, including IL-6 and TNF-α, in both feeding conditions after I/R injury. In vivo I/R models showed that at 1 wk after I/R, HFD-fed mice exhibited worse cardiac function and larger myocardial scarring along myofibers compared with normal chow diet-fed mice. In both feeding conditions, mTOR overexpression preserved cardiac function and prevented myocardial scarring. These findings suggest that cardiac mTOR overexpression is sufficient to prevent the detrimental effects of diet-induced obesity on the heart after I/R, by reducing cardiac dysfunction and myocardial scarring.
Collapse
Affiliation(s)
- Toshinori Aoyagi
- Department of Anatomy, Biochemistry and Physiology, Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Jason K Higa
- Department of Anatomy, Biochemistry and Physiology, Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Hiroko Aoyagi
- Department of Anatomy, Biochemistry and Physiology, Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Naaiko Yorichika
- Department of Anatomy, Biochemistry and Physiology, Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Briana K Shimada
- Department of Anatomy, Biochemistry and Physiology, Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Takashi Matsui
- Department of Anatomy, Biochemistry and Physiology, Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| |
Collapse
|