1
|
Leiva O, Bohart I, Ahuja T, Park D. Off-Target Effects of Cancer Therapy on Development of Therapy-Induced Arrhythmia: A Review. Cardiology 2023; 148:324-334. [PMID: 36702116 PMCID: PMC10614257 DOI: 10.1159/000529260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 01/17/2023] [Indexed: 01/27/2023]
Abstract
BACKGROUND Advances in cancer therapeutics have improved overall survival and prognosis in this patient population; however, this has come at the expense of cardiotoxicity including arrhythmia. SUMMARY Cancer and its therapies are associated with cardiotoxicity via several mechanisms including inflammation, cardiomyopathy, and off-target effects. Among cancer therapies, anthracyclines and tyrosine kinase inhibitors (TKIs) are particularly known for their pro-arrhythmia effects. In addition to cardiomyopathy, anthracyclines may be pro-arrhythmogenic via reactive oxygen species (ROS) generation and altered calcium handling. TKIs may mediate their cardiotoxicity via inhibition of off-target tyrosine kinases. Ibrutinib-mediated inhibition of CSK may be responsible for the increased prevalence of atrial fibrillation. Further investigation is warranted to further elucidate the mechanisms behind arrhythmias in cancer therapies. KEY MESSAGES Arrhythmias are a common cardiotoxicity of cancer therapies. Cancer therapies may induce arrhythmias via off-target effects. Understanding the mechanisms underlying arrhythmogenesis associated with cancer therapies may help design cancer therapies that can avoid these toxicities.
Collapse
Affiliation(s)
- Orly Leiva
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York City, New York, USA
| | - Isaac Bohart
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York City, New York, USA
| | - Tania Ahuja
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York City, New York, USA
| | - David Park
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York City, New York, USA
| |
Collapse
|
2
|
Peng J, Zeng G, Zhong P, Wang G, Lei C, Tian G, Chen J, Wu J, Shen C. The Akt pathway mediates the protective effects of myeloid differentiation protein 1 in pathological cardiac remodelling. ESC Heart Fail 2021; 8:3214-3222. [PMID: 34041859 PMCID: PMC8318477 DOI: 10.1002/ehf2.13447] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 05/11/2021] [Accepted: 05/16/2021] [Indexed: 12/24/2022] Open
Abstract
Aims Myeloid differentiation protein 1 (MD1) was shown to ameliorate pressure overload‐induced cardiac hypertrophy and fibrosis by negatively regulating the MEK–ERK1/2 and NF‐κB pathways. However, whether MD1 modulates cardiac function and whether the Akt pathway mediates the benefits of MD1 in pressure overload‐induced cardiac remodelling remain unclear. Methods and Results Male cardiac‐specific transgenic MD1 (MD1‐TG) mice, MD1‐knockout (KO) mice and wild‐type (WT) littermates aged 8–10 weeks were subjected to sham operation and aortic banding (AB) for 4 weeks. Then, left ventricular (LV) hypertrophy, fibrosis and function of the mice were assessed. When compared with WT‐AB mice, MD1‐TGs showed decreased cross‐sectional area (CSA) of cardiomyocytes (P < 0.001), mRNA expression of β‐myosin heavy chain (β‐MHC) (P < 0.02), ratios of heart weight/body weight and heart weight/tibia length (P < 0.04) and collagen volume fraction (P < 0.001). The LV end‐diastolic diameter was reduced, and LV ejection fraction and fractional shortening were improved in MD1‐TG‐AB mice than in WT‐AB mice (P < 0.05). In cultured H9C2 cells, adenovirus vector‐mediated MD1 overexpression decreased angiotensin II‐induced mRNA expression of brain natriuretic peptide (BNP) and β‐MHC and cell CSA (P < 0.002), whereas knockdown of MD1 by shRNA exhibited opposite effects (P < 0.04). Mechanistically, MD1 suppressed pathological cardiac remodelling at least partly by blocking Akt pathway. Akt inactivation by MK2206 largely offset the pro‐hypertrophic effects of MD1 deficiency in angiotensin II‐stimulated cardiomyocytes. Conclusions The Akt pathway mediates the protective effects of MD1 in pressure overload‐induced cardiac remodelling in mice. Targeting MD1 may provide therapeutic strategy for the treatment of pathological cardiac remodelling and heart failure.
Collapse
Affiliation(s)
- Jianye Peng
- Department of Cardiovascular Medicine, Key Laboratory of Heart Failure Prevention & Treatment of Hengyang, The Second Affiliated Hospital of the University of South China, Hengyang, China
| | - Gaofeng Zeng
- Department of Cardiovascular Medicine, Key Laboratory of Heart Failure Prevention & Treatment of Hengyang, The Second Affiliated Hospital of the University of South China, Hengyang, China
| | - Peng Zhong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Guangji Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - ChangCheng Lei
- Department of Cardiovascular Medicine, Key Laboratory of Heart Failure Prevention & Treatment of Hengyang, The Second Affiliated Hospital of the University of South China, Hengyang, China
| | - Guoping Tian
- Department of Cardiovascular Medicine, Key Laboratory of Heart Failure Prevention & Treatment of Hengyang, The Second Affiliated Hospital of the University of South China, Hengyang, China
| | - Jingsong Chen
- Department of Cardiovascular Medicine, Key Laboratory of Heart Failure Prevention & Treatment of Hengyang, The Second Affiliated Hospital of the University of South China, Hengyang, China
| | - Jianfeng Wu
- Department of Cardiovascular Medicine, Key Laboratory of Heart Failure Prevention & Treatment of Hengyang, The Second Affiliated Hospital of the University of South China, Hengyang, China
| | - Caijie Shen
- Department of Cardiology, Ningbo First Hospital, Ningbo, China
| |
Collapse
|
3
|
Uncovering the Molecular Mechanism of the Qiang-Xin 1 Formula on Sepsis-Induced Cardiac Dysfunction Based on Systems Pharmacology. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3815185. [PMID: 32908632 PMCID: PMC7474398 DOI: 10.1155/2020/3815185] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/22/2020] [Accepted: 07/27/2020] [Indexed: 12/29/2022]
Abstract
Cardiac dysfunction is a critical manifestation of sepsis-induced multiorgan failure and results in the high mortality of sepsis. Our previous study demonstrated that a traditional Chinese medicine formula, Qiang-Xin 1 (QX1), ameliorates cardiac tissue damage in septic mice; however, the underlying pharmacology mechanism remains to be elucidated. The present study was aimed at clarifying the protective mechanism of the QX1 formula on sepsis-induced cardiac dysfunction. The moderate sepsis model of mice was established by cecal ligation and puncture surgery. Treatment with the QX1 formula improved the 7-day survival outcome, attenuated cardiac dysfunction, and ameliorated the disruption of myocardial structure in septic mice. Subsequent systems pharmacology analysis found that 63 bioactive compounds and the related 79 candidate target proteins were screened from the QX1 formula. The network analysis showed that the QX1 active components quercetin, formononetin, kaempferol, taxifolin, cryptotanshinone, and tanshinone IIA had a good binding activity with screened targets. The integrating pathway analysis indicated the calcium, PI3K/AKT, MAPK, and Toll-like receptor signaling pathways may be involved in the protective effect of the QX1 formula on sepsis-induced cardiac dysfunction. Further, experimental validation showed that the QX1 formula inhibited the activity of calcium/calmodulin-dependent protein kinase II (CaMKII), MAPK (P38, ERK1/2, and JNK), and TLR4/NF-κB signaling pathways but promoted the activation of the PI3K/AKT pathway. A cytokine array found that the QX1 formula attenuated sepsis-induced upregulated levels of serum IFN-γ, IL-1β, IL-3, IL-6, IL-17, IL-4, IL-10, and TNF-α. Our data suggested that QX1 may represent a novel therapeutic strategy for sepsis by suppressing the activity of calcium, MAPK, and TLR4/NF-κB pathways, but promoting the activation of AKT, thus controlling cytokine storm and regulating immune balance. The present study demonstrated the multicomponent, multitarget, and multipathway characteristics of the QX1 formula and provided a novel understanding of the QX1 formula in the clinical application on cardiac dysfunction-related diseases.
Collapse
|
4
|
Melatonin as a protective agent in cardiac ischemia-reperfusion injury: Vision/Illusion? Eur J Pharmacol 2020; 885:173506. [PMID: 32858050 DOI: 10.1016/j.ejphar.2020.173506] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 08/20/2020] [Accepted: 08/23/2020] [Indexed: 01/14/2023]
Abstract
Melatonin, an emphatic endogenous molecule exerts protective effects either via activation of G-protein coupled receptors (Melatonin receptors, MTR 1-3), tumor necrosis factor receptor (TNFR), toll like receptors (TLRS), nuclear receptors (NRS) or by directly scavenging the free radicals. MTRs are extensively expressed in the heart as well as in the coronary vasculature. Accumulating evidences have indicated the existence of a strong correlation between reduction in the circulating level of melatonin and precipitation of heart attack. Apparently, melatonin exhibits cardioprotective effects via modulating inextricably interlinked pathways including modulation of mitochondrial metabolism, mitochondrial permeability transition pore formation, nitric oxide release, autophagy, generation of inflammatory cytokines, regulation of calcium transporters, reactive oxygen species, glycosaminoglycans, collagen accumulation, and regulation of apoptosis. Convincingly, this review shall describe the various signaling pathways involved in salvaging the heart against ischemia-reperfusion injury.
Collapse
|
5
|
Rocca C, Pasqua T, Cerra MC, Angelone T. Cardiac Damage in Anthracyclines Therapy: Focus on Oxidative Stress and Inflammation. Antioxid Redox Signal 2020; 32:1081-1097. [PMID: 31928066 DOI: 10.1089/ars.2020.8016] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Despite their serious side effects, anthracyclines (ANTs) are the most prescribed chemotherapeutic drugs because of their strong efficacy in both solid and hematological tumors. A major limitation to ANTs clinical application is the severe cardiotoxicity observed both acutely and chronically. The mechanism underlying cardiac dysfunction under chemotherapy is mainly dependent on the generation of oxidative stress and systemic inflammation, both of which lead to progressive cardiomyopathy and heart failure. Recent Advances: Over the years, the iatrogenic ANTs-induced cardiotoxicity was believed to be simply given by iron metabolism and reactive oxygen species production; however, several experimental data indicate that ANTs may use alternative damaging mechanisms, such as topoisomerase 2β inhibition, inflammation, pyroptosis, immunometabolism, and autophagy. Critical Issues: In this review, we aimed at discussing ANTs-induced cardiac injury from different points of view, updating and focusing on oxidative stress and inflammation, since these pathways are not exclusive or independent from each other but they together importantly contribute to the complexity of ANTs-induced multifactorial cardiotoxicity. Future Directions: A deeper understanding of the mechanistic signaling leading to ANTs side effects could reveal crucial targeting molecules, thus representing strategic knowledge to promote better therapeutic efficacy and lower cardiotoxicity during clinical application.
Collapse
Affiliation(s)
- Carmine Rocca
- Laboratory of Cellular and Molecular Cardiovascular Physiology, Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy
| | - Teresa Pasqua
- Laboratory of Cellular and Molecular Cardiovascular Physiology, Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy
| | - Maria Carmela Cerra
- Laboratory of Organ and System Physiology, Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy.,National Institute of Cardiovascular Research (INRC), Bologna, Italy
| | - Tommaso Angelone
- Laboratory of Cellular and Molecular Cardiovascular Physiology, Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy.,National Institute of Cardiovascular Research (INRC), Bologna, Italy
| |
Collapse
|
6
|
Bakhshi H, Varadarajan V, Ambale-Venkatesh B, Meyghani Z, Ostovaneh MR, Durda P, Wu CO, Tracy RP, Cushman M, Bluemke DA, Lima JAC. Association of soluble interleukin-2 receptor α and tumour necrosis factor receptor 1 with heart failure: The Multi-Ethnic Study of Atherosclerosis. ESC Heart Fail 2020; 7:639-644. [PMID: 32155316 PMCID: PMC7160474 DOI: 10.1002/ehf2.12623] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 12/19/2019] [Accepted: 01/03/2020] [Indexed: 11/24/2022] Open
Abstract
Aims Soluble tumour necrosis factor‐α receptor 1 (sTNF‐αR1) and interleukin‐2 receptor α (sIL‐2Rα) predict incident heart failure (HF) in the elderly population. However, the association of these biomarkers with HF in a multi‐ethnic asymptomatic population is unclear. We aimed to investigate the association of sTNF‐αR1 and sIL‐2Rα with incident HF in a multi‐ethnic population of middle age and older participants. Methods and results The multi‐ethnic study of atherosclerosis is a prospective population‐based study of 6814 participants aged 45–84 years who were free of clinical cardiovascular disease at enrolment. We included 2869 participants with available sTNF‐αR1 or sIL‐2Rα level measurement at baseline multi‐ethnic study of atherosclerosis exam (2000–2002). We used Cox proportional‐hazards model to investigate the association between sTNF‐αR1 and sIL‐2Rα with incident HF after adjusting for traditional cardiovascular risk factors and coronary artery calcium score measured by cardiac computed tomography. Among the included participants, the mean (standard deviation) age was 61.6 (10.2) years and 46.7% were men. The median (interquartile range) sTNF‐αR1 and sIL‐2Rα were 1293 (1107–1547) and 901 (727–1154) pg/mL. During a median follow‐up of 14.2 (interquartile range: 11.7–14.8) years, 130 participants developed HF. In multivariable analysis, the hazard ratio (95% confidence interval, P value) of incident HF for each standard deviation increment of log‐transformed sTNF‐αR1 and sIL‐2Rα was 1.43 (1.21–1.7, P ≤ 0.001) and 1.26 (1.04–1.53, P = 0.02), respectively. Excluding participants with interim coronary heart disease, we found a statistically significant association between sTNF‐αR1 and HF with hazard ratio of 1.39 (95% confidence interval: 1.11 to 1.74, P = 0.005) and sIL‐2Rα and HF showing a hazard ratio of 1.39 (95% confidence interval: 1.09 to 1.76, P = 0.007). Conclusions sTNF‐αR1 and sIL‐2Rα are associated with a higher risk of incident HF in a multi‐ethnic cohort without a previous history of cardiovascular disease.
Collapse
Affiliation(s)
- Hooman Bakhshi
- Department of Medicine, Division of Cardiology, Johns Hopkins University, Wolfe Street/Blalock 524, Baltimore, MD, 21287, USA.,Inova Heart and Vascular Institute, Falls Church, Virginia, USA
| | - Vinithra Varadarajan
- Department of Medicine, Division of Cardiology, Johns Hopkins University, Wolfe Street/Blalock 524, Baltimore, MD, 21287, USA
| | - Bharath Ambale-Venkatesh
- Department of Medicine, Division of Cardiology, Johns Hopkins University, Wolfe Street/Blalock 524, Baltimore, MD, 21287, USA
| | - Zahra Meyghani
- Department of Medicine, Division of Cardiology, Johns Hopkins University, Wolfe Street/Blalock 524, Baltimore, MD, 21287, USA
| | - Mohammad R Ostovaneh
- Department of Medicine, Division of Cardiology, Johns Hopkins University, Wolfe Street/Blalock 524, Baltimore, MD, 21287, USA
| | - Peter Durda
- Department of Pathology and Laboratory Medicine, Larner College of Medicine at the University of Vermont, Burlington, VT, 05405, USA
| | - Colin O Wu
- Office of Biostatistics Research, National Heart Lung and Blood Institute, Bethesda, MD, 20892, USA
| | - Russell P Tracy
- Department of Pathology and Laboratory Medicine, Larner College of Medicine at the University of Vermont, Burlington, VT, 05405, USA
| | - Mary Cushman
- Department of Pathology and Laboratory Medicine, Larner College of Medicine at the University of Vermont, Burlington, VT, 05405, USA.,Department of Medicine, Larner College of Medicine at the University of Vermont, Burlington, VT, 05405, USA
| | - David A Bluemke
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53726, USA
| | - João A C Lima
- Department of Medicine, Division of Cardiology, Johns Hopkins University, Wolfe Street/Blalock 524, Baltimore, MD, 21287, USA
| |
Collapse
|
7
|
Rotter Sopasakis V, Sandstedt J, Johansson M, Lundqvist A, Bergström G, Jeppsson A, Mattsson Hultén L. Toll-like receptor-mediated inflammation markers are strongly induced in heart tissue in patients with cardiac disease under both ischemic and non-ischemic conditions. Int J Cardiol 2019; 293:238-247. [PMID: 31230935 DOI: 10.1016/j.ijcard.2019.06.033] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 05/29/2019] [Accepted: 06/12/2019] [Indexed: 01/04/2023]
Abstract
BACKGROUND A sustained low grade inflammatory state is a recognized feature of various diseases, including cardiovascular disease. This state of chronic inflammation involves activation of Toll-like receptor (TLR) signaling. However, little is known regarding the genetic profile of TLR components in cardiac tissue from patients with cardiac disease. METHODS In this study we investigated the genetic profile of 84 TLR markers in a unique set of cardiac tissue from patients that had undergone either coronary artery bypass grafting (CABG) or aortic valve replacement (AVR). In addition, we compared the gene data from the cardiac tissue with the same gene profile in blood as well as circulating cytokines to elucidate possible targets in blood that could be used to estimate the inflammatory state of the heart in cardiac disease. RESULTS We found a marked upregulation of TLR-induced inflammation in cardiac tissue from both patient groups compared to healthy controls. The inflammation appeared to be primarily mediated through TLR1, 3, 7, 8 and 10, resulting in a marked induction of mediators of the innate immune response. Furthermore, the gene expression data in combination with unbiased multivariate analysis suggested a difference in inflammatory response in ischemic cardiac tissue compared to non-ischemic cardiac tissue. Serum levels of IL-13 were significantly elevated in both CABG and AVR patients compared to controls, whereas other cytokines did not appear to coincide with cardiac TLR-induced inflammation. CONCLUSIONS We propose that cardiac disease in humans may be mediated by local cardiac TLR signaling under both ischemic and non-ischemic conditions.
Collapse
Affiliation(s)
- Victoria Rotter Sopasakis
- Department of Clinical Chemistry, Sahlgrenska University Hospital and Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden.
| | - Joakim Sandstedt
- Department of Clinical Chemistry, Sahlgrenska University Hospital and Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden
| | - Michaela Johansson
- Department of Clinical Chemistry, Sahlgrenska University Hospital and Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden
| | - Annika Lundqvist
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden
| | - Göran Bergström
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden
| | - Anders Jeppsson
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden; Department of Cardiothoracic Surgery, Sahlgrenska University Hospital, SE-413 45 Gothenburg, Sweden
| | - Lillemor Mattsson Hultén
- Department of Clinical Chemistry, Sahlgrenska University Hospital and Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden; Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden
| |
Collapse
|
8
|
Xu M, Liu PP, Li H. Innate Immune Signaling and Its Role in Metabolic and Cardiovascular Diseases. Physiol Rev 2019; 99:893-948. [PMID: 30565509 DOI: 10.1152/physrev.00065.2017] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The innate immune system is an evolutionarily conserved system that senses and defends against infection and irritation. Innate immune signaling is a complex cascade that quickly recognizes infectious threats through multiple germline-encoded cell surface or cytoplasmic receptors and transmits signals for the deployment of proper countermeasures through adaptors, kinases, and transcription factors, resulting in the production of cytokines. As the first response of the innate immune system to pathogenic signals, inflammatory responses must be rapid and specific to establish a physical barrier against the spread of infection and must subsequently be terminated once the pathogens have been cleared. Long-lasting and low-grade chronic inflammation is a distinguishing feature of type 2 diabetes and cardiovascular diseases, which are currently major public health problems. Cardiometabolic stress-induced inflammatory responses activate innate immune signaling, which directly contributes to the development of cardiometabolic diseases. Additionally, although the innate immune elements are highly conserved in higher-order jawed vertebrates, lower-grade jawless vertebrates lack several transcription factors and inflammatory cytokine genes downstream of the Toll-like receptors (TLRs) and retinoic acid-inducible gene-I (RIG-I)-like receptors (RLRs) pathways, suggesting that innate immune signaling components may additionally function in an immune-independent way. Notably, recent studies from our group and others have revealed that innate immune signaling can function as a vital regulator of cardiometabolic homeostasis independent of its immune function. Therefore, further investigation of innate immune signaling in cardiometabolic systems may facilitate the discovery of new strategies to manage the initiation and progression of cardiometabolic disorders, leading to better treatments for these diseases. In this review, we summarize the current progress in innate immune signaling studies and the regulatory function of innate immunity in cardiometabolic diseases. Notably, we highlight the immune-independent effects of innate immune signaling components on the development of cardiometabolic disorders.
Collapse
Affiliation(s)
- Meng Xu
- Department of Cardiology, Renmin Hospital of Wuhan University , Wuhan , China ; Medical Research Center, Zhongnan Hospital of Wuhan University , Wuhan , China ; Animal Experiment Center, Wuhan University , Wuhan , China ; Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ontario , Canada
| | - Peter P Liu
- Department of Cardiology, Renmin Hospital of Wuhan University , Wuhan , China ; Medical Research Center, Zhongnan Hospital of Wuhan University , Wuhan , China ; Animal Experiment Center, Wuhan University , Wuhan , China ; Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ontario , Canada
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University , Wuhan , China ; Medical Research Center, Zhongnan Hospital of Wuhan University , Wuhan , China ; Animal Experiment Center, Wuhan University , Wuhan , China ; Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ontario , Canada
| |
Collapse
|
9
|
Bhat G, Yost GL, Ibrahim K, Pappas P, Tatooles A. Risk stratification with longitudinal neutrophil to lymphocyte ratio assessment after left ventricular assist device implantation. Int J Artif Organs 2018; 41:445-451. [DOI: 10.1177/0391398818784276] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Introduction: Inflammatory processes are well-characterized risk factors in cardiovascular disease including advanced heart failure. Previous studies have utilized individual white cell subtypes in risk analysis, and a recent study has focused on the efficacy of the neutrophil-to-lymphocyte ratio in evaluating negative outcomes following left ventricular assist device implantation. To investigate the interaction between the left ventricular assist device and white cell counts, we assessed longitudinal changes in neutrophil-to-lymphocyte ratio following left ventricular assist device implantation. Methods: This retrospective study included 100 patients who underwent left ventricular assist device implantation between 2012 and 2013. The neutrophil-to-lymphocyte ratio was calculated prior to left ventricular assist device implantation, daily for the first 30 postoperative days, and at the first two postoperative outpatient visits. Preoperative demographic and clinical data were collected for all patients. Results: The mean neutrophil-to-lymphocyte ratio immediately before left ventricular assist device implantation was 5.2 ± 4.9. After surgery, the neutrophil-to-lymphocyte ratio decreased asymptotically, from a peak of 29.2 on postoperative day 1 to 4.1 at the second outpatient visit ( p < 0.001). Lack of improvement in the neutrophil-to-lymphocyte ratio at postoperative day 10 was associated with increased length of stay, right heart failure, and a trend toward worsened survival. Conclusion: Our results indicate a significant inflammatory response to implantation of the left ventricular assist device, a known effect. The magnitude of this response may be effectively and easily monitored over time using the neutrophil-to-lymphocyte ratio. In general, approximately 30 days is required for the neutrophil-to-lymphocyte ratio to return to preoperative levels. After several months, the neutrophil-to-lymphocyte ratio improves to below preoperative levels. It is possible that this reduction reflects the reversal of various heart failure–mediated inflammatory processes following left ventricular assist device implantation.
Collapse
Affiliation(s)
- Geetha Bhat
- Advocate Christ Medical Center, Center for Heart Transplant and Assist Devices, Heart and Vascular Institute Administration, Oak Lawn, IL, USA
| | - Gardner L Yost
- Advocate Christ Medical Center, Center for Heart Transplant and Assist Devices, Heart and Vascular Institute Administration, Oak Lawn, IL, USA
| | - Kamel Ibrahim
- Advocate Christ Medical Center, Center for Heart Transplant and Assist Devices, Heart and Vascular Institute Administration, Oak Lawn, IL, USA
| | - Patroklos Pappas
- Advocate Christ Medical Center, Center for Heart Transplant and Assist Devices, Heart and Vascular Institute Administration, Oak Lawn, IL, USA
| | - Antone Tatooles
- Advocate Christ Medical Center, Center for Heart Transplant and Assist Devices, Heart and Vascular Institute Administration, Oak Lawn, IL, USA
| |
Collapse
|
10
|
Alexandre J, Moslehi JJ, Bersell KR, Funck-Brentano C, Roden DM, Salem JE. Anticancer drug-induced cardiac rhythm disorders: Current knowledge and basic underlying mechanisms. Pharmacol Ther 2018; 189:89-103. [PMID: 29698683 DOI: 10.1016/j.pharmthera.2018.04.009] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Significant advances in cancer treatment have resulted in decreased cancer related mortality for many malignancies with some cancer types now considered chronic diseases. Despite these improvements, there is increasing recognition that many cancer patients or cancer survivors can develop cardiovascular diseases, either due to the cancer itself or as a result of anticancer therapy. Much attention has focused on heart failure; however, other cardiotoxicities, notably cardiac rhythm disorders, can occur without underlying cardiomyopathy. Supraventricular tachycardias occur in cancer patients treated with cytotoxic chemotherapy (anthracyclines, gemcitabine, cisplatin and alkylating-agents) or kinase-inhibitors (KIs) such as ibrutinib. Ventricular arrhythmias, with a subset of them being torsades-de-pointes (TdP) favored by QTc prolongation have been reported: this may be the result of direct hERG-channel inhibition or a more recently-described mechanism of phosphoinositide-3-kinase inhibition. The major anticancer drugs responsible for QTc prolongation in this context are KIs, arsenic trioxide, anthracyclines, histone deacetylase inhibitors, and selective estrogen receptor modulators. Anticancer drug-induced cardiac rhythm disorders remain an underappreciated complication even by experienced clinicians. Moreover, the causal relationship of a particular anticancer drug with cardiac arrhythmia occurrence remains challenging due in part to patient comorbidities and complex treatment regimens. For example, any cancer patient may also be diagnosed with common diseases such as hypertension, diabetes or heart failure which increase an individual's arrhythmia susceptibility. Further, anticancer drugs are generally usually used in combination, increasing the challenge around establishing causation. Thus, arrhythmias appear to be an underappreciated adverse effect of anticancer agents and the incidence, significance and underlying mechanisms are now being investigated.
Collapse
Affiliation(s)
- Joachim Alexandre
- CHU Caen, PICARO Cardio-oncology Program, Department of Pharmacology, F-14033 Caen, France; Normandie Univ, UNICAEN, CHU Caen, EA 4650, Signalisation, Électrophysiologie et Imagerie des Lésions d'Ischémie-Reperfusion Myocardique, 14000 Caen, France
| | - Javid J Moslehi
- Vanderbilt University Medical Center, Cardio-oncology Program, Department of Medicine, Nashville, Tennessee, USA
| | - Kevin R Bersell
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Christian Funck-Brentano
- Sorbonne Université, INSERM CIC Paris-Est, AP-HP, ICAN, Pitié-Salpêtrière Hospital, Department of Pharmacology, F-75013 Paris, France
| | - Dan M Roden
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Joe-Elie Salem
- Vanderbilt University Medical Center, Cardio-oncology Program, Department of Medicine, Nashville, Tennessee, USA; Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA; Sorbonne Université, INSERM CIC Paris-Est, AP-HP, ICAN, Pitié-Salpêtrière Hospital, Department of Pharmacology, F-75013 Paris, France.
| |
Collapse
|
11
|
Kołodzińska A, Czarzasta K, Szczepankiewicz B, Główczyńska R, Fojt A, Ilczuk T, Budnik M, Krasuski K, Folta M, Cudnoch-Jędrzejewska A, Górnicka B, Opolski G. Toll-like receptor expression and apoptosis morphological patterns in female rat hearts with takotsubo syndrome induced by isoprenaline. Life Sci 2018; 199:112-121. [PMID: 29501923 DOI: 10.1016/j.lfs.2018.02.042] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 02/16/2018] [Accepted: 02/28/2018] [Indexed: 11/28/2022]
Abstract
AIMS Toll-like receptors (TLR) and apoptosis were indicated as important factors in heart failure. Our aim was to characterize the morphological pattern of apoptosis, TLR2, TLR4, and TLR6 expression in female rat hearts in the model of takotsubo syndrome (TTS). MAIN METHODS 60 Sprague-Dawley female rats were treated with a single dose of 150 mg/kg b.wt. of isoprenaline (ISO) or 0.9% NaCl (controls). Hearts were collected 24, 48, 72 h and 7 days post-ISO injection. 32/60 hearts were used in immunohistopathological studies and 28/60 in real time. KEY FINDINGS Apoptosis was observed 24 h post-ISO in cardiomyocytes, 24, 48, 72 h and 7 days post-ISO in infiltrating inflammatory cells, 7 days post-ISO in endothelial cells of vessels. Diffuse TLR4CD68 (CD68, a macrophage marker) and TLR6CD68 positive cells and TLR2, TLR4, TLR6 mononuclear cells were observed in both acute and recovery phase of TTS. In the foci located in the neighborhood of damaged (necrotic/apoptotic) cardiomyocytes in TTS, high (strong) protein expression of TLR2 (TLR2high) was observed: 24, 48, 72 h post-ISO; TLR4high - 48 and 72 h post-ISO; TLR6high - 48 h post-ISO. Whereas in cardiomyocytes of remote myocardium: TLR2high - 72 h post-ISO; TLR4high - 24 and 72 h post-ISO; TLR6high - 24 h post-ISO. TLR2 mRNA was down-regulated 48 and 72 h post-ISO whereas TLR4 up-regulated 7 days post-ISO. SIGNIFICANCE The expression pattern of apoptosis and TLR differs in the course of TTS in comparison with the control rats. We hypothesize that innate immunity and apoptosis may play a crucial role in TTS pathophysiology.
Collapse
Affiliation(s)
- Agnieszka Kołodzińska
- First Department of Cardiology, Medical University of Warsaw, 1a Banacha St, 02-097 Warsaw, Poland.
| | - Katarzyna Czarzasta
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 1b Banacha St, 02-097 Warsaw, Poland.
| | | | - Renata Główczyńska
- First Department of Cardiology, Medical University of Warsaw, 1a Banacha St, 02-097 Warsaw, Poland.
| | - Anna Fojt
- First Department of Cardiology, Medical University of Warsaw, 1a Banacha St, 02-097 Warsaw, Poland.
| | - Tomasz Ilczuk
- Department of Pathology, Medical University of Warsaw, 7 Pawińskiego St, 02-106 Warsaw, Poland.
| | - Monika Budnik
- First Department of Cardiology, Medical University of Warsaw, 1a Banacha St, 02-097 Warsaw, Poland.
| | - Krzysztof Krasuski
- Department of Medical Informatics and Telemedicine, Medical University of Warsaw, 1a Banacha St, 02-097 Warsaw, Poland; Faculty of Mathematics and Information Science, Warsaw University of Technology, 75 Koszykowa St, 00-662 Warsaw, Poland.
| | - Miłosz Folta
- First Department of Cardiology, Medical University of Warsaw, 1a Banacha St, 02-097 Warsaw, Poland; Department of Pathology, Medical University of Warsaw, 7 Pawińskiego St, 02-106 Warsaw, Poland.
| | - Agnieszka Cudnoch-Jędrzejewska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 1b Banacha St, 02-097 Warsaw, Poland.
| | - Barbara Górnicka
- Department of Pathology, Medical University of Warsaw, 7 Pawińskiego St, 02-106 Warsaw, Poland.
| | - Grzegorz Opolski
- First Department of Cardiology, Medical University of Warsaw, 1a Banacha St, 02-097 Warsaw, Poland.
| |
Collapse
|
12
|
Yu L, Feng Z. The Role of Toll-Like Receptor Signaling in the Progression of Heart Failure. Mediators Inflamm 2018; 2018:9874109. [PMID: 29576748 PMCID: PMC5822798 DOI: 10.1155/2018/9874109] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 11/28/2017] [Accepted: 12/14/2017] [Indexed: 12/14/2022] Open
Abstract
Medical systems worldwide are being faced with a growing need to understand mechanisms behind the pathogenesis of heart failure (HF) that is considered as a leading cause of morbidity and mortality around the world. Elevated levels of inflammatory mediators have been identified in patients with HF, which are primarily manifestations of innate immune responses mediated by pattern recognition receptors (PRRs). Toll-like receptors (TLRs), which belong to PRRs, are subjected to the release of pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs) to generate innate immune responses. More and more emerging data indicate that TLR signaling pathway molecules are involved in the progression of HF. Herein, we present new data with regard to the activation of TLRs in the failing heart, focusing on TLR2, TLR3, TLR4, and TLR9, and suggest the potential use of TLRs in target therapy.
Collapse
Affiliation(s)
- Lili Yu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, China
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA 70808, USA
- Henan Key Laboratory of immunology and Targeted Drugs, Xinxiang, Henan 453003, China
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang, Henan 453003, China
| | - Zhiwei Feng
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, China
| |
Collapse
|
13
|
Zhou D, Zhu Y, Ouyang MZ, Zhang M, Tang K, Niu CC, Li L. Knockout of Toll-like receptor 4 improves survival and cardiac function in a murine model of severe sepsis. Mol Med Rep 2018; 17:5368-5375. [PMID: 29393431 DOI: 10.3892/mmr.2018.8495] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 12/19/2017] [Indexed: 11/06/2022] Open
Abstract
Toll-like receptor 4 (TLR4) is a transmembrane pattern‑recognition receptor expressed in immune cells and the heart. Activation of TLR4 signaling during sepsis results in the release of cardiac depression mediators that may impair heart function. The present study aimed to determine whether TLR4 contributes to development of severe sepsis‑induced myocardial dysfunction. A cecum ligation and puncture (CLP) procedure was employed to establish severe sepsis models. Wild type (WT) and TLR4 knock‑out (TLR4‑KO) mice were divided into four groups: WT‑sham, TLR4‑KO‑sham, WT‑CLP, and TLR4‑KO‑CLP. Cardiac function of these animals was evaluated at various time points following the surgical procedure. The expression levels of proinflammatory cytokines in the heart tissues were detected by reverse transcription‑semi quantitative polymerase chain reaction (RT‑PCR). Myocardial neutrophil and macrophage infiltration were investigated by histopathological examination, as well as a myeloperoxidase activity assay in heart tissue by RT‑PCR. Myocardium Fas cell surface death receptor/Fas ligand and caspase‑3 were also analyzed by RT‑PCR. Additionally, myeloid differentiation primary response 88 M, toll or interleukin‑1 receptor‑domain‑containing adapter‑inducing interferon‑β and nuclear factor‑κB expression levels were observed in the myocardium of all four groups. WT‑CLP mice exhibited increased mortality rates, more severe cardiac dysfunction and had increased levels of interleukin (IL)‑1β, IL‑6 and tumor necrosis factor‑α in heart tissues and increased neutrophil infiltration compared with TRL4‑KO‑CLP mice. The present study reported that TLR4 aggravates severe sepsis‑induced cardiac impairment by promoting the release of proinflammatory cytokines and neutrophil infiltration in hearts.
Collapse
Affiliation(s)
- Dan Zhou
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Yun Zhu
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Min-Zhi Ouyang
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Ming Zhang
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Kui Tang
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Cheng-Cheng Niu
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Ling Li
- Medical Basic Teaching Experiment Center, College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| |
Collapse
|
14
|
Accornero F, Schips TG, Petrosino JM, Gu SQ, Kanisicak O, van Berlo JH, Molkentin JD. BEX1 is an RNA-dependent mediator of cardiomyopathy. Nat Commun 2017; 8:1875. [PMID: 29192139 PMCID: PMC5709413 DOI: 10.1038/s41467-017-02005-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 11/01/2017] [Indexed: 01/29/2023] Open
Abstract
Regulation of mRNA splicing, processing and stability is increasingly recognized as a critical control point in dynamically altering gene expression during stress or disease. Very little is understood of this process in heart failure. Here, we show that BEX1 is a heart failure-induced gene functioning as an mRNA-associated protein that enhances expression of a subset of cardiac disease-promoting genes. Modeling the increase in BEX1 that occurs in disease, cardiac-specific BEX1 transgenic mice show worse cardiac disease with stress stimulation, whereas Bex1 gene-deleted mice are protected from heart failure-promoting insults. Proteomic and interactive screening assays show that BEX1 is part of a large ribonucleoprotein processing complex involved in regulating proinflammatory mRNA expression in the heart. Specifically, induction of BEX1 augments the stability and expression of AU-rich element containing mRNAs typically found within proinflammatory genes. Thus, BEX1 functions as an mRNA-dependent effector that augments pathology-promoting gene expression during heart failure. Little is known about the changes in mRNA splicing, processing and stability that can alter gene expression during heart failure. Here, the authors show that BEX1 is induced during heart failure and is part of a ribonucleoprotein complex enhancing the expression and stability of proinflammatory genes.
Collapse
Affiliation(s)
- Federica Accornero
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA. .,Dorothy M. Davis Heart and Lung Research Institute, Department of Physiology and Cell Biology, Ohio State University, Columbus, OH, 43210, USA.
| | - Tobias G Schips
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Jennifer M Petrosino
- Dorothy M. Davis Heart and Lung Research Institute, Department of Physiology and Cell Biology, Ohio State University, Columbus, OH, 43210, USA
| | - Shan-Qing Gu
- Dorothy M. Davis Heart and Lung Research Institute, Department of Physiology and Cell Biology, Ohio State University, Columbus, OH, 43210, USA
| | - Onur Kanisicak
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Jop H van Berlo
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA.,Department of Medicine, Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Jeffery D Molkentin
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA. .,Howard Hughes Medical Institute, Cincinnati, OH, 45229, USA.
| |
Collapse
|
15
|
Gröschel C, Sasse A, Röhrborn C, Monecke S, Didié M, Elsner L, Kruse V, Bunt G, Lichtman AH, Toischer K, Zimmermann WH, Hasenfuß G, Dressel R. T helper cells with specificity for an antigen in cardiomyocytes promote pressure overload-induced progression from hypertrophy to heart failure. Sci Rep 2017; 7:15998. [PMID: 29167489 PMCID: PMC5700082 DOI: 10.1038/s41598-017-16147-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 11/08/2017] [Indexed: 12/13/2022] Open
Abstract
We investigated whether CD4+-T cells with specificity for an antigen in cardiomyocytes promote the progression from hypertrophy to heart failure in mice with increased pressure load due to transverse aortic constriction (TAC). OT-II mice expressing a transgenic T cell receptor (TCR) with specificity for ovalbumin (OVA) on CD4+-T cells and cMy-mOVA mice expressing OVA on cardiomyocytes were crossed. The resulting cMy-mOVA-OT-II mice did not display signs of spontaneous autoimmunity despite the fact that their OVA-specific CD4+-T cells were not anergic. After TAC, progression to heart failure was significantly accelerated in cMy-mOVA-OT-II compared to cMy-mOVA mice. No OVA-specific antibodies were induced in response to TAC in cMy-mOVA-OT-II mice, yet more CD3+ T cells infiltrated their myocardium when compared with TAC-operated cMy-mOVA mice. Systemically, the proportion of activated CD4+-T cells with a Th1 and Th17 cytokine profile was increased in cMy-mOVA-OT-II mice after TAC. Thus, T helper cells with specificity for an antigen in cardiomyocytes can directly promote the progression of heart failure in response to pressure overload independently of autoantibodies.
Collapse
Affiliation(s)
- Carina Gröschel
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany
| | - André Sasse
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Charlotte Röhrborn
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Sebastian Monecke
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany
| | - Michael Didié
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany.,Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany.,Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Leslie Elsner
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Vanessa Kruse
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Gertrude Bunt
- Clinical Optical Microscopy, Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Andrew H Lichtman
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Karl Toischer
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany.,Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Wolfram-Hubertus Zimmermann
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany.,Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany
| | - Gerd Hasenfuß
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany.,Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Ralf Dressel
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany. .,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany.
| |
Collapse
|
16
|
Patel MD, Mohan J, Schneider C, Bajpai G, Purevjav E, Canter CE, Towbin J, Bredemeyer A, Lavine KJ. Pediatric and adult dilated cardiomyopathy represent distinct pathological entities. JCI Insight 2017; 2:94382. [PMID: 28724792 DOI: 10.1172/jci.insight.94382] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 06/06/2017] [Indexed: 01/15/2023] Open
Abstract
Pediatric dilated cardiomyopathy (DCM) is the most common indication for heart transplantation in children. Despite similar genetic etiologies, medications routinely used in adult heart failure patients do not improve outcomes in the pediatric population. The mechanistic basis for these observations is unknown. We hypothesized that pediatric and adult DCM comprise distinct pathological entities, in that children do not undergo adverse remodeling, the target of adult heart failure therapies. To test this hypothesis, we examined LV specimens obtained from pediatric and adult donor controls and DCM patients. Consistent with the established pathophysiology of adult heart failure, adults with DCM displayed marked cardiomyocyte hypertrophy and myocardial fibrosis compared with donor controls. In contrast, pediatric DCM specimens demonstrated minimal cardiomyocyte hypertrophy and myocardial fibrosis compared with both age-matched controls and adults with DCM. Strikingly, RNA sequencing uncovered divergent gene expression profiles in pediatric and adult patients, including enrichment of transcripts associated with adverse remodeling and innate immune activation in adult DCM specimens. Collectively, these findings reveal that pediatric and adult DCM represent distinct pathological entities, provide a mechanistic basis to explain why children fail to respond to adult heart failure therapies, and suggest the need to develop new approaches for pediatric DCM.
Collapse
Affiliation(s)
| | - Jayaram Mohan
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Caralin Schneider
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Geetika Bajpai
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Enkhsaikhan Purevjav
- Department of Pediatrics, Division of Cardiology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | | | - Jeffrey Towbin
- Department of Pediatrics, Division of Cardiology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Andrea Bredemeyer
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Kory J Lavine
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA.,Department of Developmental Biology, and.,Department of Immunology and Pathology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
17
|
Loss of MD1 exacerbates pressure overload-induced left ventricular structural and electrical remodelling. Sci Rep 2017; 7:5116. [PMID: 28698617 PMCID: PMC5505950 DOI: 10.1038/s41598-017-05379-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 05/30/2017] [Indexed: 12/24/2022] Open
Abstract
Myeloid differentiation protein 1 (MD1) has been implicated in numerous pathophysiological processes, including immune regulation, obesity, insulin resistance, and inflammation. However, the role of MD1 in cardiac remodelling remains incompletely understood. We used MD1-knockout (KO) mice and their wild-type littermates to determine the functional significance of MD1 in the regulation of aortic banding (AB)-induced left ventricular (LV) structural and electrical remodelling and its underlying mechanisms. After 4 weeks of AB, MD1-KO hearts showed substantial aggravation of LV hypertrophy, fibrosis, LV dilation and dysfunction, and electrical remodelling, which resulted in overt heart failure and increased electrophysiological instability. Moreover, MD1-KO-AB cardiomyocytes showed increased diastolic sarcoplasmic reticulum (SR) Ca2+ leak, reduced Ca2+ transient amplitude and SR Ca2+ content, decreased SR Ca2+-ATPase2 expression, and increased phospholamban and Na+/Ca2+-exchanger 1 protein expression. Mechanistically, the adverse effects of MD1 deletion on LV remodelling were related to hyperactivated CaMKII signalling and increased impairment of intracellular Ca2+ homeostasis, whereas the increased electrophysiological instability was partly attributed to exaggerated prolongation of cardiac repolarisation, decreased action potential duration alternans threshold, and increased diastolic SR Ca2+ leak. Therefore, our study on MD1 could provide new therapeutic strategies for preventing/treating heart failure.
Collapse
|
18
|
Ottaviani L, da Costa Martins PA. Non-coding RNAs in cardiac hypertrophy. J Physiol 2017; 595:4037-4050. [PMID: 28233323 PMCID: PMC5471409 DOI: 10.1113/jp273129] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 02/21/2017] [Indexed: 12/23/2022] Open
Abstract
Heart failure is one of the largest contributors to disease burden and healthcare outflow in the Western world. Despite significant progress in the treatment of heart failure, disease prognosis remains very poor, with the only curative therapy still being heart transplantation. To counteract the current situation, efforts have been made to better understand the underlying molecular pathways in the progression of cardiac disease towards heart failure, and to link the disease to novel therapeutic targets such as non‐coding RNAs. The non‐coding part of the genome has gained prominence over the last couple of decades, opening a completely new research field and establishing different non‐coding RNAs species as fundamental regulators of cellular functions. Not surprisingly, their dysregulation is increasingly being linked to pathology, including to cardiac disease. Pre‐clinically, non‐coding RNAs have been shown to be of great value as therapeutic targets in pathological cardiac remodelling and also as diagnostic/prognostic biomarkers for heart failure. Therefore, it is to be expected that non‐coding RNA‐based therapeutic strategies will reach the bedside in the future and provide new and more efficient treatments for heart failure. Here, we review recent discoveries linking the function and molecular interactions of non‐coding RNAs with the pathophysiology of cardiac hypertrophy and heart failure.
![]()
Collapse
Affiliation(s)
- Lara Ottaviani
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Paula A da Costa Martins
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands.,Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
19
|
Abstract
Heart failure represents a systemic disease with profound effects on multiple peripheral tissues including skeletal muscle. Within the context of heart failure, perturbations in skeletal muscle physiology, structure, and function strongly contribute to exercise intolerance and the morbidity of this devastating disease. There is growing evidence that chronic heart failure imparts specific pathological changes within skeletal muscle beds resulting in muscle dysfunction and tissue atrophy. Mechanistically, systemic and local inflammatory responses drive critical aspects of this pathology. In this review, we will discuss pathological mechanisms that drive skeletal muscle inflammation and highlight emerging roles for distinct innate immune subsets that reside within damage muscle tissue focusing on the recently described embryonic and monocyte-derived macrophage lineages. Within this context, we will discuss how immune mechanisms can be differentially targeted to stimulate skeletal muscle inflammation, catabolism, fiber atrophy, and regeneration.
Collapse
Affiliation(s)
- Kory J Lavine
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine St. Louis, 660 S. Euclid Ave, Campus Box 8086, St. Louis, MO, 63110, USA.
- Department of Developmental Biology, Washington University School of Medicine St. Louis, St. Louis, MO, 63110, USA.
- Department of Immunology and Pathology, Washington University School of Medicine St. Louis, St. Louis, MO, 63110, USA.
| | - Oscar L Sierra
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine St. Louis, 660 S. Euclid Ave, Campus Box 8086, St. Louis, MO, 63110, USA
| |
Collapse
|
20
|
Rienks M, Papageorgiou A, Wouters K, Verhesen W, Leeuwen RV, Carai P, Summer G, Westermann D, Heymans S. A novel 72-kDa leukocyte-derived osteoglycin enhances the activation of toll-like receptor 4 and exacerbates cardiac inflammation during viral myocarditis. Cell Mol Life Sci 2016; 74:1511-1525. [PMID: 27878326 PMCID: PMC5357299 DOI: 10.1007/s00018-016-2423-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Revised: 10/20/2016] [Accepted: 11/14/2016] [Indexed: 01/04/2023]
Abstract
Background Viral myocarditis can severely damage the myocardium through excessive infiltration of immune cells. Osteoglycin (OGN) is part of the small leucine-rich repeat proteoglycan (SLRP) family. SLRP’s may affect inflammatory and fibrotic processes, but the implication of OGN in cardiac inflammation and the resulting injury upon viral myocarditis is unknown. Methods and results This study uncovered a previously unidentified 72-kDa variant of OGN that is predominant in cardiac human and mouse samples of viral myocarditis. Its absence in mice significantly decreased cardiac inflammation and injury in Coxsackievirus-B3-induced myocarditis. It also delayed mortality in lipopolysaccharide-induced endotoxemia going along with a reduced systemic production of pro-inflammatory cytokines. This 72-kDa OGN is expressed in the cell membrane of circulating and resident cardiac macrophages and neutrophils. Co-immunoprecipitation and OGN siRNA experiments revealed that this 72-kDa variant activates the toll-like receptor-4 (TLR4) with a concomitant increase in IL-6, TNF-α, IL-1β, and IL-12 expression. This immune cell activation by OGN occurred via MyD88 and increased phosphorylation of c-jun. Finally, the 72-kDa chondroitin sulfate is the result of O-linked glycosylation of the 32-kDa protein core of OGN. In contrast, the 34-kDa dermatan sulfate-OGN, involved in collagen cross linking, was also the result of O-linked glycosylation. Conclusion The current study discovered a novel 72-kDa chondroitin sulfate-OGN that is specific for innate immune cells. This variant is able to bind and activate TLR4. The absence of OGN decreases cytokine production by both circulating and cardiac leukocytes upon (systemic) LPS exposure, and reduces cardiac inflammation and injury in viral myocarditis. Electronic supplementary material The online version of this article (doi:10.1007/s00018-016-2423-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marieke Rienks
- Center for Heart Failure Research, Cardiovascular Research Institute Maastricht, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands.
| | - Anna Papageorgiou
- Center for Heart Failure Research, Cardiovascular Research Institute Maastricht, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands.,Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Hamburg, Germany
| | - Kristiaan Wouters
- Center for Heart Failure Research, Cardiovascular Research Institute Maastricht, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
| | - Wouter Verhesen
- Center for Heart Failure Research, Cardiovascular Research Institute Maastricht, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
| | - Rick van Leeuwen
- Center for Heart Failure Research, Cardiovascular Research Institute Maastricht, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
| | - Paolo Carai
- Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Hamburg, Germany
| | - Georg Summer
- Center for Heart Failure Research, Cardiovascular Research Institute Maastricht, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
| | - Dirk Westermann
- Centre for Cardiology Research, Hamburg University, Leuven, Belgium
| | - Stephane Heymans
- Center for Heart Failure Research, Cardiovascular Research Institute Maastricht, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands.,Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Hamburg, Germany
| |
Collapse
|
21
|
Turner NA. Inflammatory and fibrotic responses of cardiac fibroblasts to myocardial damage associated molecular patterns (DAMPs). J Mol Cell Cardiol 2015; 94:189-200. [PMID: 26542796 DOI: 10.1016/j.yjmcc.2015.11.002] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 10/30/2015] [Accepted: 11/01/2015] [Indexed: 02/07/2023]
Abstract
Cardiac fibroblasts (CF) are well-established as key regulators of extracellular matrix (ECM) turnover in the context of myocardial remodelling and fibrosis. Recently, this cell type has also been shown to act as a sensor of myocardial damage by detecting and responding to damage-associated molecular patterns (DAMPs) upregulated with cardiac injury. CF express a range of innate immunity pattern recognition receptors (TLRs, NLRs, IL-1R1, RAGE) that are stimulated by a host of different DAMPs that are evident in the injured or remodelling myocardium. These include intracellular molecules released by necrotic cells (heat shock proteins, high mobility group box 1 protein, S100 proteins), proinflammatory cytokines (interleukin-1α), specific ECM molecules up-regulated in response to tissue injury (fibronectin-EDA, tenascin-C) or molecules modified by a pathological environment (advanced glycation end product-modified proteins observed with diabetes). DAMP receptor activation on fibroblasts is coupled to altered cellular function including changes in proliferation, migration, myofibroblast transdifferentiation, ECM turnover and production of fibrotic and inflammatory paracrine factors, which directly impact on the heart's ability to respond to injury. This review gives an overview of the important role played by CF in responding to myocardial DAMPs and how the DAMP/CF axis could be exploited experimentally and therapeutically.
Collapse
Affiliation(s)
- Neil A Turner
- Division of Cardiovascular & Diabetes Research, and Multidisciplinary Cardiovascular Research Centre (MCRC), University of Leeds, Leeds, UK.
| |
Collapse
|
22
|
Karper JC, Westenbrink BD. BNP in heart failure: even leucocytes cannot escape its influence. Eur J Heart Fail 2015; 17:536-8. [DOI: 10.1002/ejhf.295] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 03/30/2015] [Accepted: 04/08/2015] [Indexed: 11/09/2022] Open
Affiliation(s)
- Jacco C. Karper
- Department of Cardiology; University Medical Center Groningen, University of Groningen; Groningen The Netherlands
| | - B. Daan Westenbrink
- Department of Cardiology; University Medical Center Groningen, University of Groningen; Groningen The Netherlands
| |
Collapse
|
23
|
Expression of Adiponectin Receptors on Peripheral Blood Leukocytes of Hypertensive Children Is Associated with the Severity of Hypertension. BIOMED RESEARCH INTERNATIONAL 2015; 2015:742646. [PMID: 26146630 PMCID: PMC4471253 DOI: 10.1155/2015/742646] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Revised: 01/30/2015] [Accepted: 02/25/2015] [Indexed: 12/16/2022]
Abstract
The aim of the study was to find out whether peripheral blood leukocyte adiponectin receptors 1 and 2 (AdipoR1, AdipoR2) protein expression patterns (flow cytometry) differ between the primary hypertension children (n = 57) and healthy controls (n = 19) and if their expression levels are related to selected clinical parameters. The group of 26 patients [AdipoR(-)] showed lower and the group of 31 patients [AdipoR(+)] showed higher AdipoRs protein expression than the control and each other (P < 0.01 for neutrophils, P < 0.05 for monocytes). The AdipoR(+) leukocytes expressed higher AdipoR1 mRNA levels (RT-PCR) than AdipoR(-) ones and controls (P = 0.022 and P = 0.007, resp.). Despite greater BMI, the AdipoR(-) patients had unchanged serum adiponectin levels. In contrast, AdipoR(+) patients had lower serum adiponectin concentrations than the AdipoR(-) ones and controls (P < 0.001). The AdipoR(+) patients had higher blood pressure (P = 0.042) and greater carotid intima-media thickness (P = 0.017) than the AdipoR(-) ones. The stage of hypertension was associated with increased neutrophil but not monocyte AdipoR1 density (AdipoR1 MFI) (P < 0.05). Severe ambulatory hypertension was presented more often in AdipoR(+) patients than in AdipoR(-) ones (51.6% versus 26.9%, resp.; P < 0.01). In conclusion, neutrophil AdipoRs upregulation was associated with early stages of vascular injury, hypertension severity, and low serum levels of adiponectin.
Collapse
|
24
|
Abstract
SIGNIFICANCE Pattern-recognition receptors (PRRs) are a family of receptors that are used to detect pathogen-associated molecular patterns or damage-associated molecular patterns, which initiate immune responses to resolve infections and repair damaged tissues. Abnormalities in PRR activation will unavoidably lead to excessive inflammation. RECENT ADVANCES Although multiple pathophysiological processes are involved in cardiovascular disease, recent studies have highlighted the importance of innate PRRs, in particular, Toll-like receptors and nucleotide-binding oligomerization domain-like receptors, in mediating inflammatory responses and cardiovascular function. CRITICAL ISSUES The functional roles and regulatory mechanisms of PRRs in cardiovascular diseases are still largely unknown. In particular, controversies exist on the certainty of these detrimental or beneficial effects of some PRRs in different diseased states or different experimental animal models. FUTURE DIRECTIONS Considering that the molecular mechanisms for individual PRR to regulate cellular function are complex and multiple PRRs are activated simultaneously or synergistically, a better understanding of the function of individual PRRs and the interplay of PRRs will provide unexpected opportunities to develop new therapies for cardiovascular disease by modulation of an innate immune system.
Collapse
Affiliation(s)
- Xiaojie Wang
- Department of Pharmacology, Shandong University School of Medicine , Jinan, China
| | | |
Collapse
|
25
|
Wagner KB, Felix SB, Riad A. Innate immune receptors in heart failure: Side effect or potential therapeutic target? World J Cardiol 2014; 6:791-801. [PMID: 25228958 PMCID: PMC4163708 DOI: 10.4330/wjc.v6.i8.791] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 04/18/2014] [Accepted: 06/11/2014] [Indexed: 02/06/2023] Open
Abstract
Heart failure (HF) is a leading cause of mortality and morbidity in western countries and occasions major expenses for public health systems. Although optimal medical treatment is widely available according to current guidelines, the prognosis of patients with HF is still poor. Despite the etiology of the disease, increased systemic or cardiac activation of the innate immune system is well documented in several types of HF. In some cases there is evidence of an association between innate immune activation and clinical outcome of patients with this disease. However, the few large trials conducted with the use of anti-inflammatory medication in HF have not revealed its benefits. Thus, greater understanding of the relationship between alteration in the immune system and development and progression of HF is urgently necessary: prior to designing therapeutic interventions that target pathological inflammatory processes in preventing harmful cardiac effects of immune modulatory therapy. In this regard, relatively recently discovered receptors of the innate immune system, i.e., namely toll-like receptors (TLRs) and nod-like receptors (NLRs)-are the focus of intense cardiovascular research. These receptors are main up-stream regulators of cytokine activation. This review will focus on current knowledge of the role of TLRs and NLRs, as well as on downstream cytokine activation, and will discuss potential therapeutic implications.
Collapse
|
26
|
Cardiopulmonary exercise capacity and preoperative markers of inflammation. Mediators Inflamm 2014; 2014:727451. [PMID: 25061264 PMCID: PMC4098894 DOI: 10.1155/2014/727451] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 06/10/2014] [Accepted: 06/11/2014] [Indexed: 12/18/2022] Open
Abstract
Explanatory mechanisms for the association between poor exercise capacity and infections following surgery are underexplored. We hypothesized that aerobic fitness—assessed by cardiopulmonary exercise testing (CPET)—would be associated with circulating inflammatory markers, as quantified by the neutrophil-lymphocyte ratio (NLR) and monocyte subsets. The association between cardiopulmonary reserve and inflammation was tested by multivariable regression analysis with covariates including anaerobic threshold (AT) and malignancy. In a first cohort of 240 colorectal patients, AT was identified as the sole factor associated with higher NLR (P = 0.03) and absolute and relative lymphopenia (P = 0.01). Preoperative leukocyte subsets and monocyte CD14+ expression (downregulated by endotoxin and indicative of chronic inflammation) were also assessed in two further cohorts of age-matched elective gastrointestinal and orthopaedic surgical patients. Monocyte CD14+ expression was lower in gastrointestinal patients (n = 43) compared to age-matched orthopaedic patients (n = 31). The circulating CD14+CD16− monocyte subset was reduced in patients with low cardiopulmonary reserve. Poor exercise capacity in patients without a diagnosis of heart failure is independently associated with markers of inflammation. These observations suggest that preoperative inflammation associated with impaired cardiorespiratory performance may contribute to the pathophysiology of postoperative outcome.
Collapse
|
27
|
Dong RQ, Wang ZF, Zhao C, Gu HR, Hu ZW, Xie J, Wu YQ. Toll-Like Receptor 4 Knockout Protects Against Isoproterenol-Induced Cardiac Fibrosis. J Cardiovasc Pharmacol Ther 2014; 20:84-92. [PMID: 24950765 DOI: 10.1177/1074248414539564] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: Toll-like receptor 4 participates in the process of acute heart injury. The underlying mechanisms of its protection are multifactorial, but we hypothesized that toll-like receptor-mediated autophagy control plays a vital role. The purpose of this study was to clarify the effect of autophagy on cardiac fibrosis. Methods: Cardiac fibrosis was induced by subcutaneous isoproterenol (ISO) injection, and rapamycin was simultaneously administered orally for 14 days. Animal echocardiography was then used to evaluate the success of the cardiac fibrosis model, and the mice were killed after the echocardiography examination. Results: Toll-like receptor 4 knockout (TLR4 KO) mice had better heart function than did wild-type (WT) mice ( P < .05). Rapamycin treatment reduced the left ventricular ejection fraction to 23.5% ( P < .05), and the collagen volume fraction of the ISO and ISO plus rapamycin groups was 5.9% and 25.9%, respectively, in TLR4 KO mice. Compared with the WT mice, Beclin 1 and autophagy were downregulated in TLR4 KO mice ( P < .05); however, the ISO plus rapamycin group had higher autophagy activity than did the ISO group in TLR4 KO mice ( P < .05). Conclusions: Our results suggest that TLR4 KO-induced cardioprotection against ISO-induced cardiac fibrosis is associated with reduced autophagy induction. Cardiac fibroblast autophagy participates in its own activation. The moderate inhibition of autophagic activity may be a new strategy for treating cardiac fibrosis.
Collapse
Affiliation(s)
- Rui-qing Dong
- Department of Cardiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Ze-feng Wang
- Department of Cardiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Can Zhao
- Department of Cardiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Hai-rong Gu
- Department of Cardiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhuo-wei Hu
- Molecular Immunology and Pharmacology Laboratory, State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Xie
- Molecular Immunology and Pharmacology Laboratory, State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yong-quan Wu
- Department of Cardiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
28
|
Okun E, Griffioen KJ, Sarah R, Wan R, Cong WN, De Cabo R, Montalvo AM, Levette A, Maudsley S, Martin B, Arumugam TV, Mattson MP. Toll-like receptors 2 and 4 modulate autonomic control of heart rate and energy metabolism. Brain Behav Immun 2014; 36:90-100. [PMID: 24145051 PMCID: PMC3947180 DOI: 10.1016/j.bbi.2013.10.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Revised: 09/24/2013] [Accepted: 10/12/2013] [Indexed: 02/07/2023] Open
Abstract
Toll-like receptors (TLR) are innate immune receptors typically activated by microbial-associated molecular patterns (MAMPs) during infection or damage-associated molecular patterns (DAMPs) as a result of tissue injury. Recent findings suggest that TLR2 and TLR4 signaling play important roles in developmental and adult neuroplasticity, and in learning and memory. In addition, activation of TLR2 and TLR4 worsens ischemic injury to the heart and brain in animal models of myocardial infarction and stroke. TLR activation is also implicated in thermoregulation and fever in response to infection. However, it is not known whether TLRs participate in the regulation of the sympathetic and/or parasympathetic components of the autonomic nervous system (ANS). Here we provide evidence that TLR2 and TLR4 influence autonomic regulation of heart rate (HR) body temperature and energy metabolism in mice. We show that mice lacking TLR2 or TLR4 exhibit reduced basal HR, which results from an increase of parasympathetic tone. In addition, thermoregulatory responses to stress are altered in TLR2-/- and TLR4-/- mice, and brown fat-dependent thermoregulation is altered in TLR4-/- mice. Moreover, TLR2-/- and TLR4-/- mice consume less food and exhibit a greater mass compared to wild type mice. Collectively, our findings suggest important roles for TLR2 and TLR4 in the ANS regulation of cardiovascular function, thermoregulation, and energy metabolism.
Collapse
Affiliation(s)
- Eitan Okun
- The Mina and Everard Goodman Faculty of Life Sciences, The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan 52900, Israel.
| | - Kathleen J. Griffioen
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD, 21224, USA
| | - Rothman Sarah
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD, 21224, USA
| | - Ruiqian Wan
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD, 21224, USA
| | - Wei-Na Cong
- Laboratory of Clinical Investigation, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Rafael De Cabo
- Translational Gerontology Branch, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Alejandro Martin Montalvo
- Translational Gerontology Branch, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Andrew Levette
- Translational Gerontology Branch, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Stuart Maudsley
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD, 21224, USA
| | - Bronwen Martin
- Laboratory of Clinical Investigation, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224, USA
| | | | - Mark P. Mattson
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD, 21224, USA,Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| |
Collapse
|
29
|
Liu Y, Jiang XL, Liu Y, Jiang DS, Zhang Y, Zhang R, Chen Y, Yang Q, Zhang XD, Fan GC, Li H. Toll-interacting protein (Tollip) negatively regulates pressure overload-induced ventricular hypertrophy in mice. Cardiovasc Res 2013; 101:87-96. [PMID: 24285748 DOI: 10.1093/cvr/cvt232] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
AIMS Toll-interacting protein (Tollip) is a critical regulator of the Toll-like receptor-mediated signalling pathway. However, the role of Tollip in chronic pressure overload-induced cardiac hypertrophy remains unclear. This study aimed to determine the functional significance of Tollip in the regulation of aortic banding-induced cardiac remodelling and its underlying mechanisms. METHODS AND RESULTS First, we observed that Tollip was down-regulated in human failing hearts and murine hypertrophic hearts, as determined by western blotting and RT-PCR. Using cultured neonatal rat cardiomyocytes, we found that adenovirus vector-mediated overexpression of Tollip limited angiotensin II-induced cell hypertrophy; whereas knockdown of Tollip by shRNA exhibited the opposite effects. We then generated a transgenic (TG) mouse model with cardiac specific-overexpression of Tollip and subjected them to aortic banding (AB) for 8 weeks. When compared with AB-treated wild-type mouse hearts, Tollip-TGs showed a significant attenuation of cardiac hypertrophy, fibrosis, and dysfunction, as measured by echocardiography, immune-staining, and molecular/biochemical analysis. Conversely, a global Tollip-knockout mouse model revealed an aggravated cardiac hypertrophy and accelerated maladaptation to chronic pressure overloading. Mechanistically, we discovered that Tollip interacted with AKT and suppressed its downstream signalling pathway. Pre-activation of AKT in cardiomyocytes largely offset the Tollip-elicited anti-hypertrophic effects. CONCLUSION Our results provide the first evidence that Tollip serves as a negative regulator of pathological cardiac hypertrophy by blocking the AKT signalling pathway.
Collapse
Affiliation(s)
- Yi Liu
- College of Life Sciences, Wuhan University, Wuhan 430072, PR China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Therapeutic role of toll-like receptor modification in cardiovascular dysfunction. Vascul Pharmacol 2012; 58:231-9. [PMID: 23070056 DOI: 10.1016/j.vph.2012.10.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Revised: 09/28/2012] [Accepted: 10/05/2012] [Indexed: 01/04/2023]
Abstract
Toll-like receptors (TLR) are key pattern recognition receptors in the innate immune system. The TLR-mediated immune response against pathogens is usually protective however inappropriate TLR activation may lead to excessive tissue damage. It is well recognised that TLRs respond to a variety of endogenous as well as exogenous ligands. By responding to endogenous ligands that are exposed during cellular damage, TLRs have been implicated in a range of pathological conditions associated with cardiovascular dysfunction. Increasing knowledge on the mechanisms involved in TLR signalling has encouraged the exploration of therapeutic pharmacological modulation of TLR activation in conditions such as atherosclerosis, ischaemic heart disease, heart failure and ischaemic reperfusion injury. The aim of this review is to explore the translational potentials of TLR modification in cardiovascular dysfunction, where these agents have been studied.
Collapse
|
31
|
Simpson PC. Where are the new drugs to treat heart failure? Introduction to the special issue on "key signaling molecules in hypertrophy and heart failure". J Mol Cell Cardiol 2011; 51:435-7. [PMID: 21851824 DOI: 10.1016/j.yjmcc.2011.08.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Accepted: 08/10/2011] [Indexed: 12/30/2022]
|
32
|
Abstract
Recent studies suggest that the heart possesses an innate immune system that is intended to delimit tissue injury, as well as orchestrate homoeostatic responses, within the heart. The extant literature suggests that this intrinsic stress response system is mediated, at least in part, by a family of pattern recognition receptors, most notably the Toll-like receptors. Although the innate immune system provides a short-term adaptive response to tissue injury, the beneficial effects of this phylogenetically ancient system may be lost if innate immune signaling becomes sustained and/or excessive; in which case, the salutary effects of activation of these pathways are contravened by the known deleterious effects of inflammatory signaling. Herein, the biology of innate immune signaling in the heart is reviewed, as well as the literature suggesting that the innate immune system is involved in the pathogenesis of atherosclerosis, acute coronary syndromes, stroke, viral myocarditis, sepsis, ischemia/reperfusion injury, and heart failure. The review concludes by discussing new therapies that are being developed to modulate the innate immune system.
Collapse
Affiliation(s)
- Douglas L Mann
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, St Louis, MO 63110, USA.
| |
Collapse
|