1
|
Gong S, Li Y, Yan K, Shi Z, Leng J, Bao Y, Ning K. The Crosstalk Between Endothelial Cells, Smooth Muscle Cells, and Macrophages in Atherosclerosis. Int J Mol Sci 2025; 26:1457. [PMID: 40003923 PMCID: PMC11855868 DOI: 10.3390/ijms26041457] [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: 12/31/2024] [Revised: 02/02/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory vascular disease closely tied to cellular metabolism. Recent genome-wide association study data have suggested the significant roles of endothelial cells, smooth muscle cells, and macrophages in the regression and exacerbation of AS. However, the impact of cellular crosstalk and cellular metabolic derangements on disease progression in AS is vaguely understood. In this review, we analyze the roles of the three cell types in AS. We also summarize the crosstalk between the two of them, and the associated molecules and consequences involved. In addition, we emphasize potential therapeutic targets and highlight the importance of the three-cell co-culture model and extracellular vesicles in AS-related research, providing ideas for future studies.
Collapse
Affiliation(s)
- Sihe Gong
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; (S.G.); (Y.L.); (K.Y.); (Z.S.)
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Yanni Li
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; (S.G.); (Y.L.); (K.Y.); (Z.S.)
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Kaijie Yan
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; (S.G.); (Y.L.); (K.Y.); (Z.S.)
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Zhonghong Shi
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; (S.G.); (Y.L.); (K.Y.); (Z.S.)
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Jing Leng
- Preclinical Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, China;
| | - Yimin Bao
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; (S.G.); (Y.L.); (K.Y.); (Z.S.)
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Ke Ning
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; (S.G.); (Y.L.); (K.Y.); (Z.S.)
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| |
Collapse
|
2
|
Lowe KO, Tanase CE, Maghami S, Fisher LE, Ghaemmaghami AM. Inflammatory Network of Liver Fibrosis and How It Can Be Targeted Therapeutically. IMMUNO 2023; 3:375-408. [DOI: 10.3390/immuno3040023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
Abstract
Liver fibrosis is a complex, dynamic process associated with a broad spectrum of chronic liver diseases and acute liver failure, characterised by the dysregulated intrahepatic production of extracellular matrix proteins replacing functional liver cells with scar tissue. Fibrosis progresses due to an interrelated cycle of hepatocellular injury, triggering a persistent wound-healing response. The accumulation of scar tissue and chronic inflammation can eventually lead to cirrhosis and hepatocellular carcinoma. Currently, no therapies exist to directly treat or reverse liver fibrosis; hence, it remains a substantial global disease burden. A better understanding of the intricate inflammatory network that drives the initiation and maintenance of liver fibrosis to enable the rationale design of new intervention strategies is required. This review clarifies the most current understanding of the hepatic fibrosis cellular network with a focus on the role of regulatory T cells, and a possible trajectory for T cell immunotherapy in fibrosis treatment. Despite good progress in elucidating the role of the immune system in liver fibrosis, future work to better define the function of different immune cells and their mediators at different fibrotic stages is needed, which will enhance the development of new therapies.
Collapse
Affiliation(s)
- Kirstin O. Lowe
- School of Life Sciences, University of Nottingham, Nottingham NG7 2RD, UK
| | | | - Susan Maghami
- Hull York Medical School, University of York, York YO10 5DD, UK
| | - Leanne E. Fisher
- School of Life Sciences, University of Nottingham, Nottingham NG7 2RD, UK
| | | |
Collapse
|
3
|
Xing D, Hage FG, Feng W, Guo Y, Oparil S, Sanders PW. Endothelial cells overexpressing CXCR1/2 are renoprotective in rats with acute kidney injury. Am J Physiol Renal Physiol 2023; 324:F374-F386. [PMID: 36794755 PMCID: PMC10042609 DOI: 10.1152/ajprenal.00238.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 01/19/2023] [Accepted: 02/11/2023] [Indexed: 02/17/2023] Open
Abstract
Inflammation that develops with the release of chemokines and cytokines during acute kidney injury (AKI) has been shown to participate in functional renal recovery. Although a major research focus has been on the role of macrophages, the family of C-X-C motif chemokines that promote neutrophil adherence and activation also increases with kidney ischemia-reperfusion (I/R) injury. This study tested the hypothesis that intravenous delivery of endothelial cells (ECs) that overexpress (C-X-C motif) chemokine receptors 1 and 2 (CXCR1 and CXCR2, respectively) improves outcomes in kidney I/R injury. Overexpression of CXCR1/2 enhanced homing of endothelial cells to I/R-injured kidneys and limited interstitial fibrosis, capillary rarefaction, and tissue injury biomarkers (serum creatinine concentration and urinary kidney injury molecule-1) following AKI and also reduced expression of P-selectin and the rodent (C-X-C motif) chemokine cytokine-induced neutrophil chemoattractant (CINC)-2β as well as the number of myeloperoxidase-positive cells in the postischemic kidney. The serum chemokine/cytokine profile, including CINC-1, showed similar reductions. These findings were not observed in rats given endothelial cells transduced with an empty adenoviral vector (null-ECs) or a vehicle alone. These data indicate that extrarenal endothelial cells that overexpress CXCR1 and CXCR2, but not null-ECs or vehicle alone, reduce I/R kidney injury and preserve kidney function in a rat model of AKI.NEW & NOTEWORTHY Inflammation facilitates kidney ischemia-reperfusion (I/R) injury. Endothelial cells (ECs) that were modified to overexpress (C-X-C motif) chemokine receptor (CXCR)1/2 (CXCR1/2-ECs) were injected immediately following kidney I/R injury. The interaction of CXCR1/2-ECs, but not ECs transduced with an empty adenoviral vector, with injured kidney tissue preserved kidney function and reduced production of inflammatory markers, capillary rarefaction, and interstitial fibrosis. The study highlights a functional role for the C-X-C chemokine pathway in kidney damage following I/R injury.
Collapse
Affiliation(s)
- Dongqi Xing
- Division of Pulmonary, Allergy and Critical Care Medicine, Lung Health Center, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Fadi G Hage
- Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama, United States
- Birmingham Veterans Affairs Health Care System, Birmingham, Alabama, United States
| | - Wenguang Feng
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Yuanyuan Guo
- Division of Pulmonary, Allergy and Critical Care Medicine, Lung Health Center, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Suzanne Oparil
- Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Paul W Sanders
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, United States
- Birmingham Veterans Affairs Health Care System, Birmingham, Alabama, United States
| |
Collapse
|
4
|
Mahapatra S, Sharma MVR, Brownson B, Gallicano VE, Gallicano GI. Cardiac inducing colonies halt fibroblast activation and induce cardiac/endothelial cells to move and expand via paracrine signaling. Mol Biol Cell 2022; 33:ar96. [PMID: 35653297 DOI: 10.1091/mbc.e22-02-0032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Myocardial fibrosis (MF), a common event that develops after myocardial infarction, initially is a reparative process but eventually leads to heart failure and sudden cardiac arrest. In MF, the infarct area is replaced by a collagenous-based scar induced by "excessive" collagen deposition from activated cardiac fibroblasts. The scar prevents ventricular wall thinning; however, over time it expands to noninfarcted myocardium. Therapies to prevent fibrosis include reperfusion, anti-fibrotic agents, and ACE inhibitors. Paracrine factor (PF)/stem cell research has recently gained significance as a therapy. We consistently find that cardiac inducing colonies (CiCs) (derived from human germline pluripotent stem cells) secrete PFs at physiologically relevant concentrations that suppress cardiac fibroblast activation and excessive extracellular matrix protein secretion. These factors also affect human cardiomyocytes and endothelial cells by inducing migration/proliferation of both populations into a myocardial wound model. Finally, CiC factors modulate matrix turnover and proinflammation. Taking the results together, we show that CiCs could help tip the balance from fibrosis toward repair.
Collapse
Affiliation(s)
- Samiksha Mahapatra
- Department of Biochemistry and Molecular Biology, Georgetown University Medical Center, Washington, DC 20057-145
| | | | - Breanna Brownson
- Department of Biochemistry and Molecular Biology, Georgetown University Medical Center, Washington, DC 20057-145.,Rye High School, Rye, NY 10580
| | - Vaughn E Gallicano
- Department of Biochemistry and Molecular Biology, Georgetown University Medical Center, Washington, DC 20057-145.,Thomas Edison High School, Alexandria, VA 22310
| | - G Ian Gallicano
- Department of Biochemistry and Molecular Biology, Georgetown University Medical Center, Washington, DC 20057-145
| |
Collapse
|
5
|
YQWY Decoction Improves Myocardial Remodeling via Activating the IL-10/Stat3 Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2020:7532892. [PMID: 33456490 PMCID: PMC7787750 DOI: 10.1155/2020/7532892] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 11/02/2020] [Accepted: 11/04/2020] [Indexed: 11/17/2022]
Abstract
Heart failure (HF) has been known as a global health problem, and cardiac remodeling plays an essential role in the development of HF. We hypothesized that YQWY decoction might exert a cardioprotective effect against myocardium inflammation, fibrosis, and apoptosis via activating the interleukin-10 (IL-10)/Stat3 signaling pathway. To test this hypothesis, the HF model in rats was established by pressure overload through the minimally invasive transverse aortic constriction (MTAC). Echocardiography was performed to assess the left ventricular function of rats. Myocardial fibrosis in rats was observed by Masson and Picrosirius red staining, and the degree of myocardial apoptosis was detected via TUNEL staining. In addition, expression levels of IL-10, tumor necrosis factor-α (TNF-α), Stat3 (P-Stat3), P65 (P-P65), CD68, collagen I, TGF-β, CTGF, Bax, Bcl-2, cleaved caspase-3, and PARP in rat serum and myocardium samples were examined by ELISA, western blot, and immunohistochemistry, respectively. YQWY decoction treatment significantly improved left ventricular function in HF rats, especially in those of the high-dose group (LVEF%: 51.29 ± 5.876 vs. 66.02 ± 1.264, P < 0.01;, LVFS%: 27.75 ± 3.757 vs. 37.76 ± 1.137, P < 0.01). Furthermore, YQWY decoction markedly inhibited MTAC-induced myocardial fibrosis as evidenced by downregulated collagen I, TGF-β, and CTGF in myocardium and alleviated apoptosis (downregulated caspase-3 and PARP and increased Bcl-2/Bax ratio in cardiomyocytes). In addition, YQWY decoction decreased the level of the proinflammatory cytokine TNF-α in both circulating blood and myocardium and attenuated infiltration of inflammatory cells in heart tissue from HF rats. Most importantly, YQWY decoction suppressed MTAC-induced NF-κB activation and phosphorylated Stat3 by upregulating IL-10 in rat heart tissues. Our study showed that YQWY decoction could attenuate MTAC-induced myocardial inflammation, fibrosis, apoptosis, and reverse the impairment of cardiac function in rats by activating the IL-10/Stat3 signaling pathway and improving myocardium remodeling. Our findings suggested a therapeutic potential of YQWY decoction in HF.
Collapse
|
6
|
Mourouzis K, Oikonomou E, Siasos G, Tsalamadris S, Vogiatzi G, Antonopoulos A, Fountoulakis P, Goliopoulou A, Papaioannou S, Tousoulis D. Pro-inflammatory Cytokines in Acute Coronary Syndromes. Curr Pharm Des 2020; 26:4624-4647. [PMID: 32282296 DOI: 10.2174/1381612826666200413082353] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 04/01/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Over the last decades, the role of inflammation and immune system activation in the initiation and progression of coronary artery disease (CAD) has been established. OBJECTIVES The study aimed to present the interplay between cytokines and their actions preceding and shortly after ACS. METHODS We searched in a systemic manner the most relevant articles to the topic of inflammation, cytokines, vulnerable plaque and myocardial infarction in MEDLINE, COCHRANE and EMBASE databases. RESULTS Different classes of cytokines (intereleukin [IL]-1 family, Tumor necrosis factor-alpha (TNF-α) family, chemokines, adipokines, interferons) are implicated in the entire process leading to destabilization of the atherosclerotic plaque, and consequently, to the incidence of myocardial infarction. Especially IL-1 and TNF-α family are involved in inflammatory cell accumulation, vulnerable plaque formation, platelet aggregation, cardiomyocyte apoptosis and adverse remodeling following the myocardial infarction. Several cytokines such as IL-6, adiponectin, interferon-γ, appear with significant prognostic value in ACS patients. Thus, research interest focuses on the modulation of inflammation in ACS to improve clinical outcomes. CONCLUSION Understanding the unique characteristics that accompany each cytokine-cytokine receptor interaction could illuminate the signaling pathways involved in plaque destabilization and indicate future treatment strategies to improve cardiovascular prognosis in ACS patients.
Collapse
Affiliation(s)
- Konstantinos Mourouzis
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Evangelos Oikonomou
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Gerasimos Siasos
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Sotiris Tsalamadris
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Georgia Vogiatzi
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Alexios Antonopoulos
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Petros Fountoulakis
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Athina Goliopoulou
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Spyridon Papaioannou
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Dimitris Tousoulis
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| |
Collapse
|
7
|
Dechkhajorn W, Maneerat Y, Prasongsukarn K, Kanchanaphum P, Kumsiri R. Interleukin-8 in Hyperlipidemia and Coronary Heart Disease in Thai Patients Taking Statin Cholesterol-Lowering Medication While Undergoing Coronary Artery Bypass Grafting Treatment. SCIENTIFICA 2020; 2020:5843958. [PMID: 32676215 PMCID: PMC7334778 DOI: 10.1155/2020/5843958] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 05/20/2020] [Accepted: 05/25/2020] [Indexed: 05/04/2023]
Abstract
The role of interleukin-8 (IL-8), a pivotal chemokine in atherogenesis and coronary heart disease (CHD) development, is diverse and remains unclear. This cross-sectional study investigates the association of the IL-8 expression in hyperlipidemia (H) and CHD patients who have been treated with statin cholesterol-lowering drugs while undergoing coronary artery bypass grafting treatment. Fifty-five Thai volunteers including 13 normal (N), 24 H, and 18 CHD patients were enrolled for the investigation. All the CHD patients had been treated continuously with statin cholesterol-lowering medications since the disease was diagnosed and were undergoing coronary bypass grafting approximately one month later. Therefore, the CHD group was representative of a pathogenesis improvement in CHD. The IL8 mRNA expression was determined by real-time quantitative PCR in the peripheral blood mononuclear cells from heparinized blood. The plasma IL-8 levels were assessed by enzyme-linked immunosorbent assay. The result shows that the IL8 mRNA expression in the H group tended to increase; however, in the CHD group, there was a significant decrease (p=0.0111) compared to the N group. The IL8 mRNA expression and the plasma levels in the CHD group were significantly lower than those in the H group (p < 0.05). A significant negative correlation between the IL8 mRNA (r = -0.499) or plasma IL-8 (r = -0.3875) expression and CHD progression was observed (p < 0.05). In conclusion, the transcriptomic and the phenotypic IL-8 expression decreased significantly in the Thai CHD patients who had continuously received statin-group medications compared to the H and N group participants. Therefore, IL-8 should serve as a feasible marker and could be used to evaluate the therapeutic effects of statins and illustrate the pathology of CHD treatment.
Collapse
Affiliation(s)
- Wilanee Dechkhajorn
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Yaowapa Maneerat
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | | | - Panan Kanchanaphum
- Department of Medical Science, Faculty of Science, Rangsit University, Pathumthani 12000, Thailand
| | - Ratchanok Kumsiri
- Department of Medical Science, Faculty of Science, Rangsit University, Pathumthani 12000, Thailand
| |
Collapse
|
8
|
Shetelig C, Limalanathan S, Hoffmann P, Seljeflot I, Gran JM, Eritsland J, Andersen GØ. Association of IL-8 With Infarct Size and Clinical Outcomes in Patients With STEMI. J Am Coll Cardiol 2019; 72:187-198. [PMID: 29976293 DOI: 10.1016/j.jacc.2018.04.053] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 04/12/2018] [Accepted: 04/12/2018] [Indexed: 12/28/2022]
Abstract
BACKGROUND Little is known about the role of interleukin (IL)-8 in patients with acute ST-segment elevation myocardial infarction (STEMI). OBJECTIVES The aims of this study were to evaluate, in STEMI patients, the temporal profile of IL-8 and possible associations with left ventricular (LV) function and remodeling, infarct size, microvascular obstruction, myocardial salvage, and future clinical events. METHODS A total of 258 patients with STEMI were included. Blood samples were drawn before and immediately after percutaneous coronary intervention (PCI), at day 1, and after 4 months. Cardiac magnetic resonance imaging was performed in the acute phase and after 4 months. Clinical events were registered during 12 months' follow-up and all-cause mortality after median 70 months' follow-up. RESULTS Patients with IL-8 levels greater than the median measured both immediately after PCI and at day 1 had larger final infarct size, lower LV ejection fraction, larger increase in LV end-diastolic volume, and higher frequency of microvascular obstruction. After multivariate adjustment, high IL-8 levels at day 1 were associated with an increased risk of developing a large MI and having reduced LV ejection fraction at 4 months, also after adjustment for peak troponin value. Patients with IL-8 levels in the highest quartile measured at all sampling points were more likely to have a clinical event during the first 12 months after the MI and had lower overall survival during long-term follow-up. CONCLUSIONS High levels of circulating IL-8 were associated with large infarct size, impaired recovery of LV function, and adverse clinical outcome in patients with STEMI, suggesting IL-8 as a future therapeutic target based on its important role in post-infarction inflammation.
Collapse
Affiliation(s)
- Christian Shetelig
- Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway; Center for Clinical Heart Research, Oslo University Hospital Ullevål, Oslo, Norway; Center for Heart Failure Research, Oslo, Norway; Faculty of Medicine, University of Oslo, Oslo, Norway.
| | - Shanmuganathan Limalanathan
- Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway; National Association for Heart and Lung Diseases Clinics, Feiring Heart Clinic, Feiring, Norway
| | - Pavel Hoffmann
- Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway; Department of Cardiology, Section of Interventional Cardiology, Oslo University Hospital Ullevål, Oslo, Norway
| | - Ingebjørg Seljeflot
- Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway; Center for Clinical Heart Research, Oslo University Hospital Ullevål, Oslo, Norway; Center for Heart Failure Research, Oslo, Norway; Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Jon M Gran
- Oslo Center for Biostatistics and Epidemiology, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Jan Eritsland
- Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway; Center for Heart Failure Research, Oslo, Norway
| | - Geir Ø Andersen
- Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway; Center for Heart Failure Research, Oslo, Norway
| |
Collapse
|
9
|
Xing D, Wells JM, Giordano SS, Feng W, Gaggar A, Yan J, Hage FG, Li L, Chen YF, Oparil S. Induced pluripotent stem cell-derived endothelial cells attenuate lipopolysaccharide-induced acute lung injury. J Appl Physiol (1985) 2019; 127:444-456. [PMID: 31295064 PMCID: PMC6732441 DOI: 10.1152/japplphysiol.00587.2018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 06/11/2019] [Accepted: 06/27/2019] [Indexed: 02/08/2023] Open
Abstract
The chemokine receptors CXCR1/2 and CCR2/5 play a critical role in neutrophil and monocyte recruitment to sites of injury and/or inflammation. Neutrophil-mediated inflammation and endothelial cell (EC) injury are unifying factors in the pathogenesis of the acute respiratory distress syndrome. This study tested the hypothesis that systemic administration of rat-induced pluripotent stem cell (iPS)-derived ECs (iPS-ECs) overexpressing CXCR1/2 or CCR2/5 attenuates lipopolysaccharide (LPS)-induced acute lung injury. Rat iPS-ECs were transduced with adenovirus containing cDNA of CXCR1/2 or CCR2/5. Ovariectomized Sprague-Dawley rats (10 wk old) received intraperitoneal injection of LPS and intravenous infusion of 1) saline vehicle, 2) AdNull-iPS-ECs (iPS-ECs transduced with empty adenoviral vector), 3) CXCR1/2-iPS-ECs (iPS-ECs overexpressing CXCR1/2), or 4) CCR2/5-iPS-ECs (iPS-ECs overexpressing CCR2/5) at 2 h post-LPS. Rats receiving intraperitoneal injection of saline served as sham controls. Later (4 h), proinflammatory cytokine/chemokine mRNA and protein levels were measured in total lung homogenates by real-time RT-PCR and Luminex multiplex assays, and neutrophil and macrophage infiltration in alveoli was measured by immunohistochemical staining. Pulmonary microvascular permeability was assessed by the Evans blue technique, and pulmonary edema was estimated by wet-to-dry lung weight ratios. Albumin levels and neutrophil counts were assessed in bronchoalveolar lavage fluid at 24 h post-LPS. Both CXCR1/2-iPS-ECs and CCR2/5-iPS-ECs significantly reduced LPS-induced proinflammatory mediator expression, neutrophil and macrophage infiltration, pulmonary edema, and vascular permeability compared with controls. These provocative findings provide strong evidence that targeted delivery of iPS-ECs overexpressing CXCR1/2 or CCR2/5 prevents LPS-induced acute lung injury.NEW & NOTEWORTHY We have developed a novel approach to address neutrophil-mediated inflammation and endothelial damage by targeted delivery of rat-induced pluripotent stem cell (iPS)-derived endothelial cell (ECs)overexpressing chemokine receptors CXCR1/2 and CCR2/5 in injured lung tissue in a model of acute lung injury. We have demonstrated that intravenously transfused CXCR1/2-iPS-ECs and CCR2/5-iPS-ECs are recruited to lipopolysaccharide-injured lungs and attenuate lipopolysaccharide-induced parenchymal lung injury responses, including inflammatory mediator expression, inflammatory cell infiltration, and vascular leakage compared with controls.
Collapse
Affiliation(s)
- Dongqi Xing
- Division of Pulmonary, Allergy & Critical Care Medicine, Lung Health Center, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - J Michael Wells
- Division of Pulmonary, Allergy & Critical Care Medicine, Lung Health Center, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Birmingham Veterans Affairs Medical Center, Birmingham, Alabama
| | - Samantha S Giordano
- Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Wenguang Feng
- Division of Nephrology, Nephrology Research and Training Center, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Amit Gaggar
- Division of Pulmonary, Allergy & Critical Care Medicine, Lung Health Center, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Birmingham Veterans Affairs Medical Center, Birmingham, Alabama
| | - Jie Yan
- Department of Pathology, University of New Mexico, Albuquerque, New Mexico
| | - Fadi G Hage
- Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Birmingham Veterans Affairs Medical Center, Birmingham, Alabama
| | - Li Li
- Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Physiology, School of Medicine, Shihezi University, Xinjiang, China
| | - Yiu-Fai Chen
- Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Suzanne Oparil
- Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
10
|
Qian C, Chang D, Li H, Wang Y. Identification of potentially critical genes in the development of heart failure after ST-segment elevation myocardial infarction (STEMI). J Cell Biochem 2019; 120:7771-7777. [PMID: 30485493 DOI: 10.1002/jcb.28051] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Accepted: 10/22/2018] [Indexed: 01/24/2023]
Abstract
Heart failure (HF) remains a common complication after acute ST-segment elevation myocardial infarction (STEMI). Here, we aim to identify critical genes related to the developed HF in patients with STEMI using bioinformatics analysis. The microarray data of GSE59867, including peripheral blood samples from nine patients with post-infarct HF and eight patients without post-infarct HF, were downloaded from the Gene Expression Omnibus database. Differentially expressed genes (DEGs) between HF and non-HF groups were screened by LIMMA package. Functional enrichment analyses of DEGs were conducted, followed by construction of a protein-protein interaction (PPI) network. The dynamic messenger RNA (mRNA) level of the hub genes during the follow-up was analyzed to further elucidate their role in HF development. A total of 58 upregulated and 75 downregulated DEGs were screen out. They were mainly enriched in biological processes about inflammatory response, extracellular matrix organization, response to cAMP, immune response, and positive regulation of cytosolic calcium ion concentration. Pathway analysis revealed that the DEGs were also involved in hematopoietic cell lineage, pathways in cancer, and extracellular matrix-receptor interaction. In the PPI network consisting of 58 nodes and 72 interactions, CXCL8 (degree = 15), THBS1 (degree = 8), FOS (degree = 7), and ITGA2B (degree = 6) were identified as the hub genes. In the comparison of patients with and without post-infarct HF, the mRNA level of these hub genes were all higher within 30 days but reached similar at 6 months after STEMI. In conclusion, CXCL8, THBS1, FOS, and ITGA2B may play important roles in the development of HF after acute STEMI.
Collapse
Affiliation(s)
- Cheng Qian
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Danqi Chang
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Hang Li
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yanggan Wang
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China.,Medical Research Institute of Wuhan University, Wuhan University, Wuhan, China
| |
Collapse
|
11
|
Buoncervello M, Maccari S, Ascione B, Gambardella L, Marconi M, Spada M, Macchia D, Stati T, Patrizio M, Malorni W, Matarrese P, Marano G, Gabriele L. Inflammatory cytokines associated with cancer growth induce mitochondria and cytoskeleton alterations in cardiomyocytes. J Cell Physiol 2019; 234:20453-20468. [PMID: 30982981 PMCID: PMC6767566 DOI: 10.1002/jcp.28647] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 03/19/2019] [Accepted: 03/19/2019] [Indexed: 12/13/2022]
Abstract
Cardiac dysfunction is often observed in patients with cancer also representing a serious problem limiting chemotherapeutic intervention and even patient survival. In view of the recently established role of the immune system in the control of cancer growth, the present work has been undertaken to investigate the effects of a panel of the most important inflammatory cytokines on the integrity and function of mitochondria, as well as of the cytoskeleton, two key elements in the functioning of cardiomyocytes. Either mitochondria features or actomyosin cytoskeleton organization of in vitro‐cultured cardiomyocytes treated with different inflammatory cytokines were analyzed. In addition, to investigate the interplay between tumor growth and cardiac function in an in vivo system, immunocompetent female mice were inoculated with cancer cells and treated with the chemotherapeutic drug doxorubicin at a dosing schedule able to suppress tumor growth without inducing cardiac alterations. Analyses carried out in cardiomyocytes treated with the inflammatory cytokines, such as tumor necrosis factor α (TNF‐α), interferon γ (IFN‐γ), interleukin 6 (IL‐6), IL‐8, and IL‐1β revealed severe phenotypic changes, for example, of contractile cytoskeletal elements, mitochondrial membrane potential, mitochondrial reactive oxygen species production and mitochondria network organization. Accordingly, in immunocompetent mice, the tumor growth was accompanied by increased levels of the inflammatory cytokines TNF‐α, IFN‐γ, IL‐6, and IL‐8, either in serum or in the heart tissue, together with a significant reduction of ventricular systolic function. The alterations of mitochondria and of microfilament system of cardiomyocytes, due to the systemic inflammation associated with cancer growth, could be responsible for remote cardiac injury and impairment of systolic function observed in vivo.
Collapse
Affiliation(s)
- Maria Buoncervello
- Research Coordination and Support Service, Istituto Superiore di Sanità, Rome, Italy
| | - Sonia Maccari
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Barbara Ascione
- Center for Gender-Specific Medicine, Oncology Unit, Istituto Superiore di Sanità, Rome, Italy
| | - Lucrezia Gambardella
- Center for Gender-Specific Medicine, Oncology Unit, Istituto Superiore di Sanità, Rome, Italy
| | - Matteo Marconi
- Center for Gender-Specific Medicine, Oncology Unit, Istituto Superiore di Sanità, Rome, Italy
| | - Massimo Spada
- National Centre of Animal Research and Welfare, Istituto Superiore di Sanità, Rome, Italy
| | - Daniele Macchia
- National Centre of Animal Research and Welfare, Istituto Superiore di Sanità, Rome, Italy
| | - Tonino Stati
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Mario Patrizio
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Walter Malorni
- Center for Gender-Specific Medicine, Oncology Unit, Istituto Superiore di Sanità, Rome, Italy.,Department of Biology, University of Tor Vergata, Rome, Italy
| | - Paola Matarrese
- Center for Gender-Specific Medicine, Oncology Unit, Istituto Superiore di Sanità, Rome, Italy
| | - Giuseppe Marano
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Lucia Gabriele
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
12
|
Bartekova M, Radosinska J, Jelemensky M, Dhalla NS. Role of cytokines and inflammation in heart function during health and disease. Heart Fail Rev 2019; 23:733-758. [PMID: 29862462 DOI: 10.1007/s10741-018-9716-x] [Citation(s) in RCA: 178] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
By virtue of their actions on NF-κB, an inflammatory nuclear transcription factor, various cytokines have been documented to play important regulatory roles in determining cardiac function under both physiological and pathophysiological conditions. Several cytokines including TNF-α, TGF-β, and different interleukins such as IL-1 IL-4, IL-6, IL-8, and IL-18 are involved in the development of various inflammatory cardiac pathologies, namely ischemic heart disease, myocardial infarction, heart failure, and cardiomyopathies. In ischemia-related pathologies, most of the cytokines are released into the circulation and serve as biological markers of inflammation. Furthermore, there is an evidence of their direct role in the pathogenesis of ischemic injury, suggesting cytokines as potential targets for the development of some anti-ischemic therapies. On the other hand, certain cytokines such as IL-2, IL-4, IL-6, IL-8, and IL-10 are involved in the post-ischemic tissue repair and thus are considered to exert beneficial effects on cardiac function. Conflicting reports regarding the role of some cytokines in inducing cardiac dysfunction in heart failure and different types of cardiomyopathies seem to be due to differences in the nature, duration, and degree of heart disease as well as the concentrations of some cytokines in the circulation. In spite of extensive research work in this field of investigation, no satisfactory anti-cytokine therapy for improving cardiac function in any type of heart disease is available in the literature.
Collapse
Affiliation(s)
- Monika Bartekova
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, Bratislava, Slovak Republic.,Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovak Republic
| | - Jana Radosinska
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, Bratislava, Slovak Republic.,Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovak Republic
| | - Marek Jelemensky
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Naranjan S Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Center, 351 Tache Avenue, Winnipeg, MB, R2H 2A6, Canada. .,Department of Physiology and Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada.
| |
Collapse
|
13
|
Hartman MHT, Groot HE, Leach IM, Karper JC, van der Harst P. Translational overview of cytokine inhibition in acute myocardial infarction and chronic heart failure. Trends Cardiovasc Med 2018. [PMID: 29519701 DOI: 10.1016/j.tcm.2018.02.003] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Many cytokines are currently under investigation as potential target to improve cardiac function and outcome in the setting of acute myocardial infarction (MI) or chronic heart failure (HF). Here we aim to provide a translational overview of cytokine inhibiting therapies tested in experimental models and clinical studies. In various experimental studies, inhibition of interleukin-1 (IL-1), -6 (IL-6), -8 (IL-8), monocyte chemoattractant protein-1 (MCP-1), CC- and CXC chemokines, and tumor necrosis factor-α (TNF-α) had beneficial effects on cardiac function and outcome. On the other hand, neutral or even detrimental results have been reported for some (IL-1, IL-6, IL-8, and MCP-1). Ambivalence of cytokine function, differences in study designs, treatment regimens and chosen endpoints hamper the translation of experimental research into clinical practice. Human studies are currently limited to IL-1β inhibition, IL-1 receptor antagonists (IL-1RA), IL-6 receptor antagonists (IL-6RA) or TNF inhibition. Despite favorable effects on cardiovascular events observed in retrospective cohort studies of rheumatoid arthritis patients treated with TNF inhibition or IL-1RA, most prospective studies reported disappointing and inconsistent results. Smaller studies (n < 100) generally reported favorable results of anticytokine therapy on cardiac function, but only one of the larger studies (n > 100) evaluating IL-1β inhibition presented positive results on outcome. In conclusion, of the 10 anticytokine therapies tested in animals models beneficial effects have been reported in at least one setting. In larger clinical studies, findings were unsatisfactory in all but one. Many anticytokine therapies with promising animal experimental data continue to require further evaluation in humans.
Collapse
Affiliation(s)
- Minke H T Hartman
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9700 RB, Groningen, the Netherlands.
| | - Hilde E Groot
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9700 RB, Groningen, the Netherlands
| | - Irene Mateo Leach
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9700 RB, Groningen, the Netherlands
| | - Jacco C Karper
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9700 RB, Groningen, the Netherlands
| | - Pim van der Harst
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9700 RB, Groningen, the Netherlands
| |
Collapse
|
14
|
Giordano S, Zhao X, Chen YF, Litovsky SH, Hage FG, Townes TM, Sun CW, Wu LC, Oparil S, Xing D. Induced Pluripotent Stem Cell-Derived Endothelial Cells Overexpressing Interleukin-8 Receptors A/B and/or C-C Chemokine Receptors 2/5 Inhibit Vascular Injury Response. Stem Cells Transl Med 2017; 6:1168-1177. [PMID: 28233474 PMCID: PMC5442847 DOI: 10.1002/sctm.16-0316] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 09/14/2016] [Accepted: 10/31/2016] [Indexed: 12/24/2022] Open
Abstract
Recruitment of neutrophils and monocytes/macrophages to the site of vascular injury is mediated by binding of chemoattractants to interleukin (IL) 8 receptors RA and RB (IL8RA/B) C‐C chemokine receptors (CCR) 2 and 5 expressed on neutrophil and monocyte/macrophage membranes. Endothelial cells (ECs) derived from rat‐induced pluripotent stem cells (RiPS) were transduced with adenovirus containing cDNA of IL8RA/B and/or CCR2/5. We hypothesized that RiPS‐ECs overexpressing IL8RA/B (RiPS‐IL8RA/B‐ECs), CCR2/5 (RiPS‐CCR2/5‐ECs), or both receptors (RiPS‐IL8RA/B+CCR2/5‐ECs) will inhibit inflammatory responses and neointima formation in balloon‐injured rat carotid artery. Twelve‐week‐old male Sprague‐Dawley rats underwent balloon injury of the right carotid artery and intravenous infusion of (a) saline vehicle, (b) control RiPS‐Null‐ECs (ECs transduced with empty virus), (c) RiPS‐IL8RA/B‐ECs, (d) RiPS‐CCR2/5‐ECs, or (e) RiPS‐IL8RA/B+CCR2/5‐ECs. Inflammatory mediator expression and leukocyte infiltration were measured in injured and uninjured arteries at 24 hours postinjury by enzyme‐linked immunosorbent assay (ELISA) and immunohistochemistry, respectively. Neointima formation was assessed at 14 days postinjury. RiPS‐ECs expressing the IL8RA/B or CCR2/5 homing device targeted the injured arteries and decreased injury‐induced inflammatory cytokine expression, neutrophil/macrophage infiltration, and neointima formation. Transfused RiPS‐ECs overexpressing IL8RA/B and/or CCR2/5 prevented inflammatory responses and neointima formation after vascular injury. Targeted delivery of iPS‐ECs with a homing device to inflammatory mediators in injured arteries provides a novel strategy for the treatment of cardiovascular diseases. Stem Cells Translational Medicine2017;6:1168–1177
Collapse
Affiliation(s)
- Samantha Giordano
- Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Xiangmin Zhao
- Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Yiu-Fai Chen
- Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Silvio H Litovsky
- Division of Anatomic Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Fadi G Hage
- Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Section of Cardiology, Birmingham Veterans Affairs Medical Center, Birmingham, Alabama, USA
| | - Tim M Townes
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Chiao-Wang Sun
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Li-Chen Wu
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Suzanne Oparil
- Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Dongqi Xing
- Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
15
|
Pasquier J, Gupta R, Rioult D, Hoarau-Véchot J, Courjaret R, Machaca K, Al Suwaidi J, Stanley EG, Rafii S, Elliott DA, Abi Khalil C, Rafii A. Coculturing with endothelial cells promotes in vitro maturation and electrical coupling of human embryonic stem cell-derived cardiomyocytes. J Heart Lung Transplant 2017; 36:684-693. [PMID: 28169114 DOI: 10.1016/j.healun.2017.01.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Revised: 12/22/2016] [Accepted: 01/04/2017] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Pluripotent human embryonic stem cells (hESC) are a promising source of repopulating cardiomyocytes. We hypothesized that we could improve maturation of cardiomyocytes and facilitate electrical interconnections by creating a model that more closely resembles heart tissue; that is, containing both endothelial cells (ECs) and cardiomyocytes. METHODS We induced cardiomyocyte differentiation in the coculture of an hESC line expressing the cardiac reporter NKX2.5-green fluorescent protein (GFP), and an Akt-activated EC line (E4+ECs). We quantified spontaneous beating rates, synchrony, and coordination between different cardiomyocyte clusters using confocal imaging of Fura Red-detected calcium transients and computer-assisted image analysis. RESULTS After 8 days in culture, 94% ± 6% of the NKX2-5GFP+ cells were beating when hESCs embryonic bodies were plated on E4+ECs compared with 34% ± 12.9% for controls consisting of hESCs cultured on BD Matrigel (BD Biosciences) without ECs at Day 11 in culture. The spatial organization of beating areas in cocultures was different. The GFP+ cardiomyocytes were close to the E4+ECs. The average beats/min of the cardiomyocytes in coculture was faster and closer to physiologic heart rates compared with controls (50 ± 14 [n = 13] vs 25 ± 9 [n = 8]; p < 0.05). The coculture with ECs led to synchronized beating relying on the endothelial network, as illustrated by the loss of synchronization upon the disruption of endothelial bridges. CONCLUSIONS The coculturing of differentiating cardiomyocytes with Akt-activated ECs but not EC-conditioned media results in (1) improved efficiency of the cardiomyocyte differentiation protocol and (2) increased maturity leading to better intercellular coupling with improved chronotropy and synchrony.
Collapse
Affiliation(s)
- Jennifer Pasquier
- Stem Cell and Microenvironment Laboratory, Weill Cornell Medicine-Qatar, Doha, Qatar; Department of Genetic Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Renuka Gupta
- Stem Cell and Microenvironment Laboratory, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Damien Rioult
- UMR_I 02 INERIS-URCA-ULH Unité Stress Environnementaux et BIOsurveillance des milieux aquatiques, Université de Reims, Reims, France
| | - Jessica Hoarau-Véchot
- Stem Cell and Microenvironment Laboratory, Weill Cornell Medicine-Qatar, Doha, Qatar; Cardiovascular Epigenetics Laboratory, Department of Genetic Medicine, Doha, Qatar
| | - Raphael Courjaret
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Khaled Machaca
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Doha, Qatar
| | | | - Edouard G Stanley
- Department of Medicine, Weill Cornell Medicine, New York, New York; Murdoch Childrens Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Shahin Rafii
- Department of Medicine, Weill Cornell Medicine, New York, New York
| | - David A Elliott
- Department of Medicine, Weill Cornell Medicine, New York, New York; Murdoch Childrens Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Charbel Abi Khalil
- Department of Genetic Medicine, Weill Cornell Medicine, New York, New York, USA; Cardiovascular Epigenetics Laboratory, Department of Genetic Medicine, Doha, Qatar; Heart Hospital, Hamad Medical Corporation, Doha, Qatar; Department of Medicine, Weill Cornell Medicine, New York, New York, USA.
| | - Arash Rafii
- Stem Cell and Microenvironment Laboratory, Weill Cornell Medicine-Qatar, Doha, Qatar; Department of Genetic Medicine, Weill Cornell Medicine, New York, New York, USA.
| |
Collapse
|
16
|
Jose Corbalan J, Vatner DE, Vatner SF. Myocardial apoptosis in heart disease: does the emperor have clothes? Basic Res Cardiol 2016; 111:31. [PMID: 27043720 DOI: 10.1007/s00395-016-0549-2] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 03/24/2016] [Indexed: 01/06/2023]
Abstract
Since the discovery of a novel mechanism of cell death that differs from traditional necrosis, i.e., apoptosis, there have been numerous studies concluding that increased apoptosis augments myocardial infarction and heart failure and that limiting apoptosis protects the heart. Importantly, the vast majority of cells in the heart are non-myocytes with only roughly 30 % myocytes, yet almost the entire field studying apoptosis in the heart has disregarded non-myocyte apoptosis, e.g., only 4.7 % of 423 studies on myocardial apoptosis in the past 3 years quantified non-myocyte apoptosis. Accordingly, we reviewed the history of apoptosis in the heart focusing first on myocyte apoptosis, followed by the history of non-myocyte apoptosis in myocardial infarction and heart failure. Apoptosis of several of the major non-myocyte cell types in the heart (cardiac fibroblasts, endothelial cells, vascular smooth muscle cells, macrophages and leukocytes) may actually be responsible for affecting the severity of myocardial infarction and heart failure. In summary, even though it is now known that the majority of apoptosis in the heart occurs in non-myocytes, very little work has been done to elucidate the mechanisms by which non-myocyte apoptosis might be responsible for the adverse effects of apoptosis in myocardial infarction and heart failure. The goal of this review is to provide an impetus for future work in this field on non-myocyte apoptosis that will be required for a better understanding of the role of apoptosis in the heart.
Collapse
Affiliation(s)
- J Jose Corbalan
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, Newark, NJ, 07103, USA
| | - Dorothy E Vatner
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, Newark, NJ, 07103, USA
| | - Stephen F Vatner
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, Newark, NJ, 07103, USA.
| |
Collapse
|
17
|
Giordano S, Zhao X, Xing D, Hage F, Oparil S, Cooke JP, Lee J, Nakayama KH, Huang NF, Chen YF. Targeted delivery of human iPS-ECs overexpressing IL-8 receptors inhibits neointimal and inflammatory responses to vascular injury in the rat. Am J Physiol Heart Circ Physiol 2016; 310:H705-15. [PMID: 26801304 PMCID: PMC4865064 DOI: 10.1152/ajpheart.00587.2015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 01/14/2016] [Indexed: 12/24/2022]
Abstract
Interleukin-8 (IL8) is highly expressed by injured arteries in a variety of diseases and is a chemoattractant for neutrophils which express IL8 receptors IL8RA and RB (IL8RA/B) on their membranes. Neutrophils interact with the damaged endothelium and initiate an inflammatory cascade at the site of injury. We have generated a novel translational targeted cell therapy for acute vascular injury using adenoviral vectors to overexpress IL8RA/B and green fluorescent protein (GFP) on the surface of endothelial cells (ECs) derived from human induced pluripotent stem cells (HiPS-IL8RA/B-ECs). We hypothesize that HiPS-IL8RA/B-ECs transfused intravenously into rats with balloon injury of the carotid artery will target to the injured site and compete with neutrophils, thus inhibiting inflammation and neointima formation. Young adult male Sprague-Dawley rats underwent balloon injury of the right carotid artery and received intravenous transfusion of saline vehicle, 1.5 × 10(6) HiPS-ECs, 1.5 × 10(6) HiPS-Null-ECs, or 1.5 × 10(6) HiPS-IL8RA/B-ECs immediately after endoluminal injury. Tissue distribution of HiPS-IL8RA/B-ECs was analyzed by a novel GFP DNA qPCR method. Cytokine and chemokine expression and leukocyte infiltration were measured in injured and uninjured arteries at 24 h postinjury by ELISA and immunohistochemistry, respectively. Neointimal, medial areas, and reendothelialization were measured 14 days postinjury. HiPS-IL8RA/B-ECs homed to injured arteries, inhibited inflammatory mediator expression and inflammatory cell infiltration, accelerated reendothelialization, and attenuated neointima formation after endoluminal injury while control HiPS-ECs and HiPS-Null-ECs did not. HiPS-IL8RA/B-ECs transfused into rats with endoluminal carotid artery injury target to the injured artery and provide a novel strategy to treat vascular injury.
Collapse
Affiliation(s)
- Samantha Giordano
- Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Xiangmin Zhao
- Department of Pulmonary, Critical Care, Sleep and Allergy, College of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Daisy Xing
- Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Fadi Hage
- Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; Division of Cardiology, Birmingham Veterans Affairs Medical Center, Birmingham, Alabama
| | - Suzanne Oparil
- Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - John P Cooke
- Houston Methodist Research Institute, Houston, Texas
| | - Jieun Lee
- Division of Cardiovascular Medicine, Stanford University, Stanford, California
| | - Karina H Nakayama
- Cardiovascular Institute, Stanford University, Stanford, California; Veterans Affairs Palo Alto Health Care System, Palo Alto, California; and
| | - Ngan F Huang
- Cardiovascular Institute, Stanford University, Stanford, California; Veterans Affairs Palo Alto Health Care System, Palo Alto, California; and Department of Cardiothoracic Surgery, Stanford University, Stanford, California
| | - Yiu-Fai Chen
- Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama;
| |
Collapse
|
18
|
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
|
19
|
Suss PH, Capriglione LGA, Barchiki F, Miyague L, Jackowski D, Fracaro L, Schittini AV, Senegaglia AC, Rebelatto CLK, Olandoski M, Correa A, Brofman PRS. Direct intracardiac injection of umbilical cord-derived stromal cells and umbilical cord blood-derived endothelial cells for the treatment of ischemic cardiomyopathy. Exp Biol Med (Maywood) 2015; 240:969-78. [PMID: 25576340 DOI: 10.1177/1535370214565077] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 11/12/2014] [Indexed: 01/09/2023] Open
Abstract
The development of new therapeutic strategies is necessary to reduce the worldwide social and economic impact of cardiovascular disease, which produces high rates of morbidity and mortality. A therapeutic option that has emerged in the last decade is cell therapy. The aim of this study was to compare the effect of transplanting human umbilical cord-derived stromal cells (UCSCs), human umbilical cord blood-derived endothelial cells (UCBECs) or a combination of these two cell types for the treatment of ischemic cardiomyopathy (IC) in a Wistar rat model. IC was induced by left coronary artery ligation, and baseline echocardiography was performed seven days later. Animals with a left ventricular ejection fraction (LVEF) of ≤40% were selected for the study. On the ninth day after IC was induced, the animals were randomized into the following experimental groups: UCSCs, UCBECs, UCSCs plus UCBECs, or vehicle (control). Thirty days after treatment, an echocardiographic analysis was performed, followed by euthanasia. The animals in all of the cell therapy groups, regardless of the cell type transplanted, had less collagen deposition in their heart tissue and demonstrated a significant improvement in myocardial function after IC. Furthermore, there was a trend of increasing numbers of blood vessels in the infarcted area. The median value of LVEF increased by 7.19% to 11.77%, whereas the control group decreased by 0.24%. These results suggest that UCSCs and UCBECs are promising cells for cellular cardiomyoplasty and can be an effective therapy for improving cardiac function following IC.
Collapse
Affiliation(s)
- Paula H Suss
- Core for Cell Technology, Pontifícia Universidade Católica do Paraná, Curitiba 80.215-901, Brazil
| | | | - Fabiane Barchiki
- Core for Cell Technology, Pontifícia Universidade Católica do Paraná, Curitiba 80.215-901, Brazil
| | - Lye Miyague
- Core for Cell Technology, Pontifícia Universidade Católica do Paraná, Curitiba 80.215-901, Brazil
| | - Danielle Jackowski
- Core for Cell Technology, Pontifícia Universidade Católica do Paraná, Curitiba 80.215-901, Brazil
| | - Letícia Fracaro
- Core for Cell Technology, Pontifícia Universidade Católica do Paraná, Curitiba 80.215-901, Brazil
| | - Andressa V Schittini
- Carlos Chagas Institute, Oswaldo Cruz Foundation, FIOCRUZ/PR, Curitiba 81.350-010, Brazil
| | - Alexandra C Senegaglia
- Core for Cell Technology, Pontifícia Universidade Católica do Paraná, Curitiba 80.215-901, Brazil
| | - Carmen L K Rebelatto
- Core for Cell Technology, Pontifícia Universidade Católica do Paraná, Curitiba 80.215-901, Brazil
| | - Márcia Olandoski
- Core for Cell Technology, Pontifícia Universidade Católica do Paraná, Curitiba 80.215-901, Brazil
| | - Alejandro Correa
- Carlos Chagas Institute, Oswaldo Cruz Foundation, FIOCRUZ/PR, Curitiba 81.350-010, Brazil
| | - Paulo R S Brofman
- Core for Cell Technology, Pontifícia Universidade Católica do Paraná, Curitiba 80.215-901, Brazil
| |
Collapse
|
20
|
Rogers NM, Isenberg JS. Endothelial cell global positioning system for pulmonary arterial hypertension: homing in on vascular repair. Arterioscler Thromb Vasc Biol 2014; 34:1336-8. [PMID: 24951651 DOI: 10.1161/atvbaha.114.303877] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Natasha M Rogers
- From the Vascular Medicine Institute, Department of Medicine (N.M.R., J.S.I.), Starzl Transplantation Institute, Department of Surgery (N.M.R., J.S.I.), and Division of Pulmonary Allergy and Critical Care Medicine (J.S.I.), University of Pittsburgh School of Medicine, PA
| | - Jeffrey S Isenberg
- From the Vascular Medicine Institute, Department of Medicine (N.M.R., J.S.I.), Starzl Transplantation Institute, Department of Surgery (N.M.R., J.S.I.), and Division of Pulmonary Allergy and Critical Care Medicine (J.S.I.), University of Pittsburgh School of Medicine, PA.
| |
Collapse
|
21
|
Fu J, Chen YF, Zhao X, Creighton JR, Guo Y, Hage FG, Oparil S, Xing DD. Targeted delivery of pulmonary arterial endothelial cells overexpressing interleukin-8 receptors attenuates monocrotaline-induced pulmonary vascular remodeling. Arterioscler Thromb Vasc Biol 2014; 34:1539-47. [PMID: 24790141 DOI: 10.1161/atvbaha.114.303821] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Interleukin-8 (IL-8) receptors IL8RA and IL8RB (IL8RA/B) on neutrophil membranes bind to IL-8 with high affinity and play a critical role in neutrophil recruitment to sites of injury and inflammation. This study tested the hypothesis that administration of rat pulmonary arterial endothelial cells (ECs) overexpressing IL8RA/B can accelerate the adhesion of ECs to the injured lung and inhibit monocrotaline-induced pulmonary inflammation, arterial thickening and hypertension, and right ventricular hypertrophy. APPROACH AND RESULTS The treatment groups included 10-week-old ovariectomized Sprague-Dawley rats that received subcutaneous injection of PBS (vehicle), a single injection of monocrotaline (monocrotaline alone, 60 mg/kg, SC), monocrotaline followed by intravenous transfusion of ECs transduced with the empty adenoviral vector (null-EC), and monocrotaline followed by intravenous transfusion of ECs overexpressing IL8RA/B (1.5 × 10(6) cells/rat). Two days or 4 weeks after monocrotaline treatment, endothelial nitric oxide synthase, inducible nitric oxide synthase, cytokine-induced neutrophil chemoattractant-2β (IL-8 equivalent in rat), and monocyte chemoattractant protein-1 expression, neutrophil and macrophage infiltration into pulmonary arterioles, and arteriolar and alveolar morphology were measured by histological and immunohistochemical techniques. Proinflammatory cytokine/chemokine protein levels were measured by Multiplex rat-specific magnetic bead-based sandwich immunoassay in total lung homogenates. Transfusion of ECs overexpressing IL8RA/B significantly reduced monocrotaline-induced neutrophil infiltration and proinflammatory mediator (IL-8, monocyte chemoattractant protein-1, inducible nitric oxide synthase, cytokine-induced neutrophil chemoattractant, and macrophage inflammatory protein-2) expression in lungs and pulmonary arterioles and alveoli, pulmonary arterial pressure, and pulmonary arterial and right ventricular hypertrophy and remodeling. CONCLUSIONS These provocative findings suggest that targeted delivery of ECs overexpressing IL8RA/B is effective in repairing the injured pulmonary vasculature.
Collapse
Affiliation(s)
- Jinyan Fu
- From the Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine (J.F., Y.-F.C., X.Z., Y.G., F.G.H., S.O., D.D.X.) and Department of Anesthesiology, University of Alabama at Birmingham (J.R.C.); Department of Biochemistry and the Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Xinjiang, China (J.F.); and Section of Cardiology, Birmingham Veteran's Administration Medical Center, AL (F.G.H.)
| | - Yiu-Fai Chen
- From the Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine (J.F., Y.-F.C., X.Z., Y.G., F.G.H., S.O., D.D.X.) and Department of Anesthesiology, University of Alabama at Birmingham (J.R.C.); Department of Biochemistry and the Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Xinjiang, China (J.F.); and Section of Cardiology, Birmingham Veteran's Administration Medical Center, AL (F.G.H.)
| | - Xiangmin Zhao
- From the Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine (J.F., Y.-F.C., X.Z., Y.G., F.G.H., S.O., D.D.X.) and Department of Anesthesiology, University of Alabama at Birmingham (J.R.C.); Department of Biochemistry and the Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Xinjiang, China (J.F.); and Section of Cardiology, Birmingham Veteran's Administration Medical Center, AL (F.G.H.)
| | - Judy R Creighton
- From the Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine (J.F., Y.-F.C., X.Z., Y.G., F.G.H., S.O., D.D.X.) and Department of Anesthesiology, University of Alabama at Birmingham (J.R.C.); Department of Biochemistry and the Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Xinjiang, China (J.F.); and Section of Cardiology, Birmingham Veteran's Administration Medical Center, AL (F.G.H.)
| | - Yuanyuan Guo
- From the Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine (J.F., Y.-F.C., X.Z., Y.G., F.G.H., S.O., D.D.X.) and Department of Anesthesiology, University of Alabama at Birmingham (J.R.C.); Department of Biochemistry and the Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Xinjiang, China (J.F.); and Section of Cardiology, Birmingham Veteran's Administration Medical Center, AL (F.G.H.)
| | - Fadi G Hage
- From the Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine (J.F., Y.-F.C., X.Z., Y.G., F.G.H., S.O., D.D.X.) and Department of Anesthesiology, University of Alabama at Birmingham (J.R.C.); Department of Biochemistry and the Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Xinjiang, China (J.F.); and Section of Cardiology, Birmingham Veteran's Administration Medical Center, AL (F.G.H.)
| | - Suzanne Oparil
- From the Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine (J.F., Y.-F.C., X.Z., Y.G., F.G.H., S.O., D.D.X.) and Department of Anesthesiology, University of Alabama at Birmingham (J.R.C.); Department of Biochemistry and the Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Xinjiang, China (J.F.); and Section of Cardiology, Birmingham Veteran's Administration Medical Center, AL (F.G.H.)
| | - Daisy D Xing
- From the Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine (J.F., Y.-F.C., X.Z., Y.G., F.G.H., S.O., D.D.X.) and Department of Anesthesiology, University of Alabama at Birmingham (J.R.C.); Department of Biochemistry and the Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Xinjiang, China (J.F.); and Section of Cardiology, Birmingham Veteran's Administration Medical Center, AL (F.G.H.).
| |
Collapse
|
22
|
Inhibiting C-reactive protein for the treatment of cardiovascular disease: promising evidence from rodent models. Mediators Inflamm 2014; 2014:353614. [PMID: 24803739 PMCID: PMC3996300 DOI: 10.1155/2014/353614] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 02/28/2014] [Indexed: 11/17/2022] Open
Abstract
Raised blood C-reactive protein (CRP) level is a predictor of cardiovascular events, but whether blood CRP is causal in the disease process is unknown. The latter would best be defined by pharmacological inhibition of the protein in the context of a randomized case-control study. However, no CRP specific drug is currently available so such a prospective study cannot be performed. Blood CRP is synthesized primarily in the liver and the liver is an organ where antisense oligonucleotide (ASO) drugs accumulate. Taking advantage of this we evaluated the efficacy of CRP specific ASOs in rodents with experimentally induced cardiovascular damage. Treating rats for 4 weeks with a rat CRP-specific ASO achieved >60% reduction of blood CRP. Notably, this effect was associated with improved heart function and pathology following myocardial infarction (induced by ligation of the left anterior descending artery). Likewise in human CRP transgenic mice treated for 2 weeks with a human CRP-specific ASO, blood human CRP was reduced by >70% and carotid artery patency was improved (2 weeks after surgical ligation). CRP specific ASOs might pave the way towards a placebo-controlled trial that could clarify the role of CRP in cardiovascular disease.
Collapse
|