1
|
Tran DT, Yeung ESH, Hong LYQ, Kaur H, Advani SL, Liu Y, Syeda MZ, Batchu SN, Advani A. Finerenone attenuates downregulation of the kidney GLP-1 receptor and glucagon receptor and cardiac GIP receptor in mice with comorbid diabetes. Diabetol Metab Syndr 2024; 16:283. [PMID: 39582036 PMCID: PMC11587750 DOI: 10.1186/s13098-024-01525-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 11/12/2024] [Indexed: 11/26/2024] Open
Abstract
BACKGROUND Several new treatments have recently been shown to have heart and kidney protective benefits in people with diabetes. Because these treatments were developed in parallel, it is unclear how the different molecular pathways affected by the therapies may overlap. Here, we examined the effects of the mineralocorticoid receptor antagonist finerenone in mice with comorbid diabetes, focusing on the regulation of expression of the glucagon-like peptide-1 receptor (GLP-1R), gastric inhibitory polypeptide receptor (GIPR) and glucagon receptor (GCGR), which are targets of approved or investigational therapies in diabetes. METHODS Male C57BL/6J mice were fed a high fat diet for 26 weeks. Twelve weeks into the high fat diet feeding period, mice received an intraperitoneal injection of streptozotocin before being followed for the remaining 14 weeks (DMHFD mice). After 26 weeks, mice were fed a high fat diet containing finerenone (100 mg/kg diet) or high fat diet alone for a further 2 weeks. Cell culture experiments were performed in primary vascular smooth muscle cells (VSMCs), NRK-49 F fibroblasts, HK-2 cells, and MDCK cells. RESULTS DMHFD mice developed albuminuria, glomerular mesangial expansion, and diastolic dysfunction (decreased E/A ratio). Glp1r and Gcgr were predominantly expressed in arteriolar VSMCs and distal nephron structures of mouse kidneys respectively, whereas Gipr was the predominant of the three transcripts in mouse hearts. Kidney Glp1r and Gcgr and cardiac Gipr mRNA levels were reduced in DMHFD mice and this reduction was negated or attenuated with finerenone. Mechanistically, finerenone attenuated upregulation of the profibrotic growth factor Ccn2 in DMHFD kidneys, whereas recombinant CCN2 downregulated Glp1r and Gcgr in VSMCs and MDCK cells respectively. CONCLUSIONS Through its anti-fibrotic actions, finerenone reverses Glp1r and Gcgr downregulation in the diabetic kidney. Both finerenone and GLP-1R agonists have proven cardiorenal benefits, whereas receptor co-agonists are approved or under development. The current findings provide preclinical rationale for the combined use of finerenone with the GLP-1R agonist family. They also provide mechanism of action insights into the potential benefit of finerenone in people with diabetes for whom GLP-1R agonists or co-agonists may not be indicated.
Collapse
Affiliation(s)
- Duc Tin Tran
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 6-151 61 Queen Street East, Toronto, ON, M5C 2T2, Canada
| | - Emily S H Yeung
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 6-151 61 Queen Street East, Toronto, ON, M5C 2T2, Canada
| | - Lisa Y Q Hong
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 6-151 61 Queen Street East, Toronto, ON, M5C 2T2, Canada
| | - Harmandeep Kaur
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 6-151 61 Queen Street East, Toronto, ON, M5C 2T2, Canada
| | - Suzanne L Advani
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 6-151 61 Queen Street East, Toronto, ON, M5C 2T2, Canada
| | - Youan Liu
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 6-151 61 Queen Street East, Toronto, ON, M5C 2T2, Canada
| | - Madiha Zahra Syeda
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 6-151 61 Queen Street East, Toronto, ON, M5C 2T2, Canada
| | - Sri Nagarjun Batchu
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 6-151 61 Queen Street East, Toronto, ON, M5C 2T2, Canada
| | - Andrew Advani
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 6-151 61 Queen Street East, Toronto, ON, M5C 2T2, Canada.
| |
Collapse
|
2
|
Lumish HS, Sherrid MV, Janssen PML, Ferrari G, Hasegawa K, Castillero E, Adlestein E, Swistel DG, Topkara VK, Maurer MS, Reilly MP, Shimada YJ. Comprehensive Proteomic Profiling of Human Myocardium Reveals Signaling Pathways Dysregulated in Hypertrophic Cardiomyopathy. J Am Coll Cardiol 2024; 84:1999-2011. [PMID: 39365226 PMCID: PMC11817648 DOI: 10.1016/j.jacc.2024.07.043] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/18/2024] [Accepted: 07/29/2024] [Indexed: 10/05/2024]
Abstract
BACKGROUND Hypertrophic cardiomyopathy (HCM) is the most common genetic cardiac disease. Signaling pathways that link genetic sequence variants to clinically overt HCM and progression to severe forms of HCM remain unknown. OBJECTIVES The purpose of this study was to identify signaling pathways that are differentially regulated in HCM, using proteomic profiling of human myocardium, confirmed with transcriptomic profiling. METHODS In this multicenter case-control study, myocardial samples were obtained from cases with HCM and control subjects with nonfailing hearts. Proteomic profiling of 7,289 proteins from myocardial samples was performed using the SomaScan assay (SomaLogic). Pathway analysis of differentially expressed proteins was performed, using a false discovery rate <0.05. Pathway analysis of proteins whose concentrations correlated with clinical indicators of severe HCM (eg, reduced left ventricular ejection fraction, atrial fibrillation, and ventricular tachyarrhythmias) was also executed. Confirmatory analysis of differentially expressed genes was performed using myocardial transcriptomic profiling. RESULTS The study included 99 HCM cases and 15 control subjects. Pathway analysis of differentially expressed proteins revealed dysregulation of the Ras-mitogen-activated protein kinase, ubiquitin-mediated proteolysis, angiogenesis-related (eg, hypoxia-inducible factor-1, vascular endothelial growth factor), and Hippo pathways. Pathways known to be dysregulated in HCM, including metabolic, inflammatory, and extracellular matrix pathways, were also dysregulated. Pathway analysis of proteins associated with clinical indicators of severe HCM and of differentially expressed genes supported these findings. CONCLUSIONS The present study represents the most comprehensive (>7,000 proteins) and largest-scale (n = 99 HCM cases) proteomic profiling of human HCM myocardium to date. Proteomic profiling and confirmatory transcriptomic profiling elucidate dysregulation of both newly recognized (eg, Ras-mitogen-activated protein kinase) and known pathways associated with pathogenesis and progression to severe forms of HCM.
Collapse
Affiliation(s)
- Heidi S Lumish
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Mark V Sherrid
- Leon Charney Division of Cardiology, New York University Langone Health, New York, New York, USA
| | - Paul M L Janssen
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio, USA
| | - Giovanni Ferrari
- Division of Cardiothoracic and Vascular Surgery, Department of Surgery, Columbia University Irving Medical Center, New York, New York, USA; Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | - Kohei Hasegawa
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Estibaliz Castillero
- Division of Cardiothoracic and Vascular Surgery, Department of Surgery, Columbia University Irving Medical Center, New York, New York, USA
| | - Elizabeth Adlestein
- Leon Charney Division of Cardiology, New York University Langone Health, New York, New York, USA
| | - Daniel G Swistel
- Department of Cardiothoracic Surgery, New York University Langone Health, New York, New York, USA
| | - Veli K Topkara
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Mathew S Maurer
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Muredach P Reilly
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA; Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, New York, New York, USA
| | - Yuichi J Shimada
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA.
| |
Collapse
|
3
|
Lumish HS, Liang LW, Hasegawa K, Maurer MS, Fifer MA, Reilly MP, Shimada YJ. Prediction of worsening heart failure in hypertrophic cardiomyopathy using plasma proteomics. Heart 2023; 109:1837-1843. [PMID: 37451849 PMCID: PMC10843738 DOI: 10.1136/heartjnl-2023-322644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023] Open
Abstract
OBJECTIVE Heart failure (HF) is one of the most common and lifestyle-limiting complications of hypertrophic cardiomyopathy (HCM). Prediction of worsening HF using clinical measures alone remains limited. Moreover, the mechanisms by which patients with HCM develop worsening HF have not been elucidated. Therefore, the aim of this study was to develop a plasma proteomics-based model to predict worsening HF among patients with HCM and to identify signalling pathways that are differentially regulated in those who subsequently develop worsening HF. METHODS In this multi-centre, prospective cohort study of 389 patients with HCM, plasma proteomics profiling of 4986 proteins was performed at enrolment. A proteomics-based random forest model was developed to predict worsening HF using data from one institution (training set, n=268). This model was externally validated in patients from a different institution (test set, n=121). Pathway analysis of proteins significantly dysregulated in patients who subsequently developed worsening HF compared with those who did not was executed, using a false discovery rate (FDR) threshold of <0.001. RESULTS Using the 11-protein proteomics-based model derived from the training set, the area under the receiver-operating characteristic curve to predict worsening HF was 0.87 (95% CI: 0.76 to 0.98) in the test set. Pathway analysis revealed that the Ras-MAPK pathway (FDR<0.00001) and related pathways were dysregulated in patients who subsequently developed worsening HF. CONCLUSIONS The present study with comprehensive plasma proteomics profiling demonstrated a high accuracy to predict worsening HF in patients with HCM and identified the Ras-MAPK and related signalling pathways as potential underlying mechanisms.
Collapse
Affiliation(s)
- Heidi S Lumish
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Lusha W Liang
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Kohei Hasegawa
- Department of Emergency Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Mathew S Maurer
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Michael A Fifer
- Division of Cardiology, Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Muredach P Reilly
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
- Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, New York, New York, USA
| | - Yuichi J Shimada
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| |
Collapse
|
4
|
Singh M, Anvekar P, Baraskar B, Pallipamu N, Gadam S, Cherukuri ASS, Damani DN, Kulkarni K, Arunachalam SP. Prospective of Pancreatic Cancer Diagnosis Using Cardiac Sensing. J Imaging 2023; 9:149. [PMID: 37623681 PMCID: PMC10455647 DOI: 10.3390/jimaging9080149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 08/26/2023] Open
Abstract
Pancreatic carcinoma (Ca Pancreas) is the third leading cause of cancer-related deaths in the world. The malignancies of the pancreas can be diagnosed with the help of various imaging modalities. An endoscopic ultrasound with a tissue biopsy is so far considered to be the gold standard in terms of the detection of Ca Pancreas, especially for lesions <2 mm. However, other methods, like computed tomography (CT), ultrasound, and magnetic resonance imaging (MRI), are also conventionally used. Moreover, newer techniques, like proteomics, radiomics, metabolomics, and artificial intelligence (AI), are slowly being introduced for diagnosing pancreatic cancer. Regardless, it is still a challenge to diagnose pancreatic carcinoma non-invasively at an early stage due to its delayed presentation. Similarly, this also makes it difficult to demonstrate an association between Ca Pancreas and other vital organs of the body, such as the heart. A number of studies have proven a correlation between the heart and pancreatic cancer. The tumor of the pancreas affects the heart at the physiological, as well as the molecular, level. An overexpression of the SMAD4 gene; a disruption in biomolecules, such as IGF, MAPK, and ApoE; and increased CA19-9 markers are a few of the many factors that are noted to affect cardiovascular systems with pancreatic malignancies. A comprehensive review of this correlation will aid researchers in conducting studies to help establish a definite relation between the two organs and discover ways to use it for the early detection of Ca Pancreas.
Collapse
Affiliation(s)
- Mansunderbir Singh
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA; (M.S.); (B.B.); (N.P.)
| | - Priyanka Anvekar
- Department of Medicine, Division of Infectious Diseases, Mayo Clinic, Rochester, MN 55905, USA;
| | - Bhavana Baraskar
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA; (M.S.); (B.B.); (N.P.)
| | - Namratha Pallipamu
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA; (M.S.); (B.B.); (N.P.)
| | - Srikanth Gadam
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA; (M.S.); (B.B.); (N.P.)
| | - Akhila Sai Sree Cherukuri
- GIH Artificial Intelligence Laboratory (GAIL), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Microwave Engineering and Imaging Laboratory (MEIL), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Devanshi N. Damani
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA;
- Department of Internal Medicine, Texas Tech University Health Science Center, El Paso, TX 79995, USA
| | - Kanchan Kulkarni
- Centre de Recherche Cardio-Thoracique de Bordeaux, University of Bordeaux, INSERM, U1045, 33000 Bordeaux, France;
- IHU Liryc, Heart Rhythm Disease Institute, Fondation Bordeaux Université, 33600 Bordeaux, France
| | - Shivaram P. Arunachalam
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA; (M.S.); (B.B.); (N.P.)
- GIH Artificial Intelligence Laboratory (GAIL), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Microwave Engineering and Imaging Laboratory (MEIL), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
5
|
Liang LW, Hasegawa K, Maurer MS, Reilly MP, Fifer MA, Shimada YJ. Comprehensive Transcriptomics Profiling of MicroRNA Reveals Plasma Circulating Biomarkers of Hypertrophic Cardiomyopathy and Dysregulated Signaling Pathways. Circ Heart Fail 2023; 16:e010010. [PMID: 37305994 PMCID: PMC10293060 DOI: 10.1161/circheartfailure.122.010010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 03/20/2023] [Indexed: 06/13/2023]
Abstract
BACKGROUND Hypertrophic cardiomyopathy (HCM) is caused by mutations in genes coding for proteins essential for myocardial contraction. However, it remains unclear through which signaling pathways these gene mutations mediate HCM pathogenesis. Growing evidence indicates that microRNAs (miRNAs) play an important role in the regulation of gene expression. We hypothesized that transcriptomics profiling of plasma miRNAs would reveal circulating biomarkers and dysregulated signaling pathways in HCM. METHODS We conducted a multicenter case-control study of cases with HCM and controls with hypertensive left ventricular hypertrophy. We performed plasma transcriptomics profiling of miRNAs using RNA sequencing. We developed a transcriptomics-based discrimination model using samples retrieved during the first two-thirds of the study period at one institution (training set). We prospectively tested its discriminative ability in samples collected thereafter from the same institution (prospective test set). We also externally validated the model by applying it to samples collected from the other institutions (external test set). We executed pathway analysis of dysregulated miRNAs with univariable P<0.05. RESULTS This study included 555 patients (392 cases and 163 controls). One thousand one hundred forty-one miRNAs passed our quality control filters. The area under the receiver operating characteristic curve of the transcriptomics-based model derived from the training set was 0.86 (95% CI, 0.79-0.93) in the prospective test set and 0.94 (95% CI, 0.90-0.97) in the external test set. Pathway analysis revealed dysregulation of the Ras-MAPK (mitogen-activated protein kinase) pathway and pathways related to inflammation in HCM. CONCLUSIONS This study utilized comprehensive transcriptomics profiling with RNA sequencing in HCM, revealing circulating miRNA biomarkers and dysregulated pathways.
Collapse
Affiliation(s)
- Lusha W. Liang
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Kohei Hasegawa
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Mathew S. Maurer
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Muredach P. Reilly
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, New York, NY, USA
| | - Michael A. Fifer
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yuichi J. Shimada
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
6
|
Shimada YJ, Raita Y, Liang LW, Maurer MS, Hasegawa K, Fifer MA, Reilly MP. Prediction of Major Adverse Cardiovascular Events in Patients With Hypertrophic Cardiomyopathy Using Proteomics Profiling. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2022; 15:e003546. [PMID: 36252118 PMCID: PMC9771902 DOI: 10.1161/circgen.121.003546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 06/24/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND Hypertrophic cardiomyopathy often causes major adverse cardiovascular events (MACE), for example, arrhythmias, stroke, heart failure, and sudden cardiac death. Currently, there are no models available to predict MACE. Furthermore, it remains unclear which signaling pathways mediate MACE. Therefore, we aimed to prospectively determine protein biomarkers that predict MACE in hypertrophic cardiomyopathy and to identify signaling pathways differentially regulated in patients who subsequently develop MACE. METHODS In this multi-centre prospective cohort study of patients with hypertrophic cardiomyopathy, we conducted plasma proteomics profiling of 4979 proteins upon enrollment. We developed a proteomics-based model to predict MACE using data from one institution (training set). We tested the predictive ability in independent samples from the other institution (test set) and performed time-to-event analysis. Additionally, we executed pathway analysis of predictive proteins using a false discovery rate threshold of <0.001. RESULTS The study included 245 patients (n=174 in the training set and n=71 in the test set). Using the proteomics-based model to predict MACE derived from the training set, the area under the receiver-operating-characteristic curve was 0.81 (95% CI, 0.68-0.93) in the test set. In the test set, the high-risk group determined by the proteomics-based predictive model had a significantly higher rate of developing MACE (hazard ratio, 13.6 [95% CI, 1.7-107]; P=0.01). The Ras-MAPK (mitogen-activated protein kinase) pathway was upregulated in patients who subsequently developed MACE (false discovery rate<1.0×10-7). Pathways involved in inflammation and fibrosis-for example, the TGF (transforming growth factor)-β pathway-were also upregulated. CONCLUSIONS This study serves as the first to demonstrate the ability of proteomics profiling to predict MACE in hypertrophic cardiomyopathy, exhibiting both novel (eg, Ras-MAPK) and known (eg, TGF-β) pathways differentially regulated in patients who subsequently experience MACE.
Collapse
Affiliation(s)
- Yuichi J. Shimada
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Yoshihiko Raita
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Lusha W. Liang
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Mathew S. Maurer
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Kohei Hasegawa
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Michael A. Fifer
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Muredach P. Reilly
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
- Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, New York, NY
| |
Collapse
|
7
|
Glen C, Tan YY, Waterston A, Evans TRJ, Jones RJ, Petrie MC, Lang NN. Mechanistic and Clinical Overview Cardiovascular Toxicity of BRAF and MEK Inhibitors: JACC: CardioOncology State-of-the-Art Review. JACC CardioOncol 2022; 4:1-18. [PMID: 35492830 PMCID: PMC9040125 DOI: 10.1016/j.jaccao.2022.01.096] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 01/07/2022] [Indexed: 02/07/2023] Open
Abstract
Rapidly accelerated fibrosarcoma B-type (BRAF) and mitogen-activated extracellular signal-regulated kinase (MEK) inhibitors have revolutionized melanoma treatment. Approximately half of patients with melanoma harbor a BRAF gene mutation with subsequent dysregulation of the RAF-MEK-ERK signaling pathway. Targeting this pathway with BRAF and MEK blockade results in control of cell proliferation and, in most cases, disease control. These pathways also have cardioprotective effects and are necessary for normal vascular and cardiac physiology. BRAF and MEK inhibitors are associated with adverse cardiovascular effects including hypertension, left ventricular dysfunction, venous thromboembolism, atrial arrhythmia, and electrocardiographic QT interval prolongation. These effects may be underestimated in clinical trials. Baseline cardiovascular assessment and follow-up, including serial imaging and blood pressure assessment, are essential to balance optimal anti-cancer therapy while minimizing cardiovascular side effects. In this review, an overview of BRAF/MEK inhibitor-induced cardiovascular toxicity, the mechanisms underlying these, and strategies for surveillance, prevention, and treatment of these effects are provided.
Collapse
Key Words
- ACE, angiotensin-converting enzyme
- AF, atrial fibrillation
- BRAF inhibitor
- BRAF, rapidly accelerated fibrosarcoma B-type
- CVAE, cardiovascular adverse event
- EGFR, epidermal growth factor receptor
- ERK, extracellular signal-regulated kinase
- LVSD, left ventricular systolic dysfunction
- MEK inhibitor
- MEK, mitogen-activated extracellular signal-regulated kinase
- RAF, rapidly accelerated fibrosarcoma
- VEGF, vascular endothelial growth factor
- cardio-oncology
- cardiovascular toxicity
- hypertension
- left ventricular systolic dysfunction
- melanoma
Collapse
Affiliation(s)
- Claire Glen
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Yun Yi Tan
- Beatson West of Scotland Cancer Centre, NHS Greater Glasgow and Clyde, Glasgow, United Kingdom
| | - Ashita Waterston
- Beatson West of Scotland Cancer Centre, NHS Greater Glasgow and Clyde, Glasgow, United Kingdom
| | - Thomas R. Jeffry Evans
- Beatson West of Scotland Cancer Centre, NHS Greater Glasgow and Clyde, Glasgow, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Robert J. Jones
- Beatson West of Scotland Cancer Centre, NHS Greater Glasgow and Clyde, Glasgow, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Mark C. Petrie
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Ninian N. Lang
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
- Beatson West of Scotland Cancer Centre, NHS Greater Glasgow and Clyde, Glasgow, United Kingdom
| |
Collapse
|
8
|
Palioura D, Lazou A, Drosatos K. Krüppel-like factor (KLF)5: An emerging foe of cardiovascular health. J Mol Cell Cardiol 2022; 163:56-66. [PMID: 34653523 PMCID: PMC8816822 DOI: 10.1016/j.yjmcc.2021.10.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/22/2021] [Accepted: 10/07/2021] [Indexed: 02/03/2023]
Abstract
Krüppel-like factors (KLFs) are DNA-binding transcriptional factors, which regulate various pathways that pertain to development, metabolism and other cellular mechanisms. KLF5 was first cloned in 1993 and by 1999, it was reported as the intestinal-enriched KLF. Beyond findings that have associated KLF5 with normal development and cancer, it has been associated with various types of cardiovascular (CV) complications and regulation of metabolic pathways in the liver, heart, adipose tissue and skeletal muscle. Specifically, increased KLF5 expression has been linked with cardiomyopathy in diabetes, end-stage heart failure, and as well as in vascular atherosclerotic lesions. In this review article, we summarize research findings about transcriptional, post-transcriptional and post-translational regulation of KLF5, as well as the role of KLF5 in the biology of cells and organs that affect cardiovascular health either directly or indirectly. Finally, we propose KLF5 inhibition as an emerging approach for cardiovascular therapeutics.
Collapse
Affiliation(s)
- Dimitra Palioura
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA;,School of Biology, Aristotle University of Thessaloniki, GR, Greece
| | - Antigone Lazou
- School of Biology, Aristotle University of Thessaloniki, GR, Greece
| | - Konstantinos Drosatos
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| |
Collapse
|
9
|
Ramos-Kuri M, Meka SH, Salamanca-Buentello F, Hajjar RJ, Lipskaia L, Chemaly ER. Molecules linked to Ras signaling as therapeutic targets in cardiac pathologies. Biol Res 2021; 54:23. [PMID: 34344467 PMCID: PMC8330049 DOI: 10.1186/s40659-021-00342-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 06/26/2021] [Indexed: 12/11/2022] Open
Abstract
Abstract The Ras family of small Guanosine Triphosphate (GTP)-binding proteins (G proteins) represents one of the main components of intracellular signal transduction required for normal cardiac growth, but is also critically involved in the development of cardiac hypertrophy and heart failure. The present review provides an update on the role of the H-, K- and N-Ras genes and their related pathways in cardiac diseases. We focus on cardiac hypertrophy and heart failure, where Ras has been studied the most. We also review other cardiac diseases, like genetic disorders related to Ras. The scope of the review extends from fundamental concepts to therapeutic applications. Although the three Ras genes have a nearly identical primary structure, there are important functional differences between them: H-Ras mainly regulates cardiomyocyte size, whereas K-Ras regulates cardiomyocyte proliferation. N-Ras is the least studied in cardiac cells and is less associated to cardiac defects. Clinically, oncogenic H-Ras causes Costello syndrome and facio-cutaneous-skeletal syndromes with hypertrophic cardiomyopathy and arrhythmias. On the other hand, oncogenic K-Ras and alterations of other genes of the Ras-Mitogen-Activated Protein Kinase (MAPK) pathway, like Raf, cause Noonan syndrome and cardio-facio-cutaneous syndromes characterized by cardiac hypertrophy and septal defects. We further review the modulation by Ras of key signaling pathways in the cardiomyocyte, including: (i) the classical Ras-Raf-MAPK pathway, which leads to a more physiological form of cardiac hypertrophy; as well as other pathways associated with pathological cardiac hypertrophy, like (ii) The SAPK (stress activated protein kinase) pathways p38 and JNK; and (iii) The alternative pathway Raf-Calcineurin-Nuclear Factor of Activated T cells (NFAT). Genetic alterations of Ras isoforms or of genes in the Ras-MAPK pathway result in Ras-opathies, conditions frequently associated with cardiac hypertrophy or septal defects among other cardiac diseases. Several studies underline the potential role of H- and K-Ras as a hinge between physiological and pathological cardiac hypertrophy, and as potential therapeutic targets in cardiac hypertrophy and failure. Graphic abstract ![]()
The Ras (Rat Sarcoma) gene family is a group of small G proteins Ras is regulated by growth factors and neurohormones affecting cardiomyocyte growth and hypertrophy Ras directly affects cardiomyocyte physiological and pathological hypertrophy Genetic alterations of Ras and its pathways result in various cardiac phenotypes Ras and its pathway are differentially regulated in acquired heart disease Ras modulation is a promising therapeutic target in various cardiac conditions.
Collapse
Affiliation(s)
- Manuel Ramos-Kuri
- Instituto Nacional de Cancerología, Unidad de Investigación Biomédica en Cáncer, Secretarìa de Salud/Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, México.,Researcher of the Facultad de Bioética, Cátedra de Infertilidad, Universidad Anáhuac, Mexico City, México.,Centro de Investigación en Bioética y Genética, Querétaro, México
| | - Sri Harika Meka
- Division of Nephrology, Department of Medicine, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Clinical and Translational Research Center, 875 Ellicott Street, Suite 8030B, Buffalo, NY, 14203, USA
| | - Fabio Salamanca-Buentello
- University of Toronto Institute of Medical Science, Medical Sciences Building, 1 King's College Circle, Room 2374, Toronto, ON, M5S 1A8, Canada
| | | | - Larissa Lipskaia
- INSERM U955 and Département de Physiologie, Hôpital Henri Mondor, FHU SENEC, AP-HP, and Université Paris-Est Créteil (UPEC), 94010, Créteil, France
| | - Elie R Chemaly
- Division of Nephrology, Department of Medicine, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Clinical and Translational Research Center, 875 Ellicott Street, Suite 8030B, Buffalo, NY, 14203, USA.
| |
Collapse
|
10
|
Shimada YJ, Raita Y, Liang LW, Maurer MS, Hasegawa K, Fifer MA, Reilly MP. Comprehensive Proteomics Profiling Reveals Circulating Biomarkers of Hypertrophic Cardiomyopathy. Circ Heart Fail 2021; 14:e007849. [PMID: 34192899 DOI: 10.1161/circheartfailure.120.007849] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Hypertrophic cardiomyopathy (HCM) is caused by mutations in the genes coding for proteins essential in normal myocardial contraction. However, it remains unclear through which molecular pathways gene mutations mediate the development of HCM. The objectives were to determine plasma protein biomarkers of HCM and to reveal molecular pathways differentially regulated in HCM. METHODS We conducted a multicenter case-control study of cases with HCM and controls with hypertensive left ventricular hypertrophy. We performed plasma proteomics profiling of 1681 proteins. We performed a sparse partial least squares discriminant analysis to develop a proteomics-based discrimination model with data from 1 institution (ie, the training set). We tested the discriminative ability in independent samples from the other institution (ie, the test set). As an exploratory analysis, we executed pathway analysis of significantly dysregulated proteins. Pathways with false discovery rate <0.05 were declared positive. RESULTS The study included 266 cases and 167 controls (n=308 in the training set; n=125 in the test set). Using the proteomics-based model derived from the training set, the area under the receiver operating characteristic curve was 0.89 (95% CI, 0.83-0.94) in the test set. Pathway analysis revealed that the Ras-MAPK (mitogen-activated protein kinase) pathway, along with its upstream and downstream pathways, was upregulated in HCM. Pathways involved in inflammation and fibrosis-for example, the TGF (transforming growth factor)-β pathway-were also upregulated. CONCLUSIONS This study serves as the largest-scale investigation with the most comprehensive proteomics profiling in HCM, revealing circulating biomarkers and exhibiting both novel (eg, Ras-MAPK) and known (eg, TGF-β) pathways differentially regulated in HCM.
Collapse
Affiliation(s)
- Yuichi J Shimada
- Division of Cardiology, Department of Medicine (Y.J.S., L.W.L., M.S.M., M.P.R.), Columbia University Irving Medical Center, New York, NY.,Cardiology Division, Department of Medicine (Y.J.S., M.A.F.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Yoshihiko Raita
- Department of Emergency Medicine (Y.R., K.H.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Lusha W Liang
- Division of Cardiology, Department of Medicine (Y.J.S., L.W.L., M.S.M., M.P.R.), Columbia University Irving Medical Center, New York, NY
| | - Mathew S Maurer
- Division of Cardiology, Department of Medicine (Y.J.S., L.W.L., M.S.M., M.P.R.), Columbia University Irving Medical Center, New York, NY
| | - Kohei Hasegawa
- Department of Emergency Medicine (Y.R., K.H.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Michael A Fifer
- Cardiology Division, Department of Medicine (Y.J.S., M.A.F.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Muredach P Reilly
- Division of Cardiology, Department of Medicine (Y.J.S., L.W.L., M.S.M., M.P.R.), Columbia University Irving Medical Center, New York, NY.,Irving Institute for Clinical and Translational Research (M.P.R.), Columbia University Irving Medical Center, New York, NY
| |
Collapse
|
11
|
Meagher P, Civitarese R, Lee X, Gordon M, Bugyei-Twum A, Desjardins JF, Kabir G, Zhang Y, Kosanam H, Visram A, Leong-Poi H, Advani A, Connelly KA. The Goto Kakizaki rat: Impact of age upon changes in cardiac and renal structure, function. PLoS One 2021; 16:e0252711. [PMID: 34166385 PMCID: PMC8224913 DOI: 10.1371/journal.pone.0252711] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 05/20/2021] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Patients with diabetes are at a high risk for developing cardiac dysfunction in the absence of coronary artery disease or hypertension, a condition known as diabetic cardiomyopathy. Contributing to heart failure is the presence of diabetic kidney disease. The Goto-Kakizaki (GK) rat is a non-obese, non-hypertensive model of type 2 diabetes that, like humans, shares a susceptibility locus on chromosome 10. Herein, we perform a detailed analysis of cardio-renal remodeling and response to renin angiotensin system blockade in GK rats to ascertain the validity of this model for further insights into disease pathogenesis. METHODS Study 1: Male GK rats along with age matched Wistar control animals underwent longitudinal assessment of cardiac and renal function for 32 weeks (total age 48 weeks). Animals underwent regular echocardiography every 4 weeks and at sacrifice, early (~24 weeks) and late (~48 weeks) timepoints, along with pressure volume loop analysis. Histological and molecular characteristics were determined using standard techniques. Study 2: the effect of renin angiotensin system (RAS) blockade upon cardiac and renal function was assessed in GK rats. Finally, proteomic studies were conducted in vivo and in vitro to identify novel pathways involved in remodeling responses. RESULTS GK rats developed hyperglycaemia by 12 weeks of age (p<0.01 c/w Wistar controls). Echocardiographic assessment of cardiac function demonstrated preserved systolic function by 48 weeks of age. Invasive studies demonstrated left ventricular hypertrophy, pulmonary congestion and impaired diastolic function. Renal function was preserved with evidence of hyperfiltration. Cardiac histological analysis demonstrated myocyte hypertrophy (p<0.05) with evidence of significant interstitial fibrosis (p<0.05). RT qPCR demonstrated activation of the fetal gene program, consistent with cellular hypertrophy. RAS blockade resulted in a reduction blood pressure(P<0.05) cardiac interstitial fibrosis (p<0.05) and activation of fetal gene program. No significant change on either systolic or diastolic function was observed, along with minimal impact upon renal structure or function. Proteomic studies demonstrated significant changes in proteins involved in oxidative phosp4horylation, mitochondrial dysfunction, beta-oxidation, and PI3K/Akt signalling (all p<0.05). Further, similar changes were observed in both LV samples from GK rats and H9C2 cells incubated in high glucose media. CONCLUSION By 48 weeks of age, the diabetic GK rat demonstrates evidence of preserved systolic function and impaired relaxation, along with cardiac hypertrophy, in the presence of hyperfiltration and elevated protein excretion. These findings suggest the GK rat demonstrates some, but not all features of diabetes induced "cardiorenal" syndrome. This has implications for the use of this model to assess preclinical strategies to treat cardiorenal disease.
Collapse
Affiliation(s)
- Patrick Meagher
- St. Michael’s Hospital, Keenan Research Centre, Li Ka Shing Knowledge Institute, Toronto, Canada
- Department of Physiology, University of Toronto, Toronto, Canada
| | - Robert Civitarese
- St. Michael’s Hospital, Keenan Research Centre, Li Ka Shing Knowledge Institute, Toronto, Canada
| | - Xavier Lee
- St. Michael’s Hospital, Keenan Research Centre, Li Ka Shing Knowledge Institute, Toronto, Canada
- Department of Physiology, University of Toronto, Toronto, Canada
| | - Mark Gordon
- St. Michael’s Hospital, Keenan Research Centre, Li Ka Shing Knowledge Institute, Toronto, Canada
| | - Antoinette Bugyei-Twum
- St. Michael’s Hospital, Keenan Research Centre, Li Ka Shing Knowledge Institute, Toronto, Canada
- Department of Physiology, University of Toronto, Toronto, Canada
| | - Jean-Francois Desjardins
- St. Michael’s Hospital, Keenan Research Centre, Li Ka Shing Knowledge Institute, Toronto, Canada
| | - Golam Kabir
- St. Michael’s Hospital, Keenan Research Centre, Li Ka Shing Knowledge Institute, Toronto, Canada
| | - Yanling Zhang
- St. Michael’s Hospital, Keenan Research Centre, Li Ka Shing Knowledge Institute, Toronto, Canada
| | - Hari Kosanam
- St. Michael’s Hospital, Keenan Research Centre, Li Ka Shing Knowledge Institute, Toronto, Canada
| | - Aylin Visram
- St. Michael’s Hospital, Keenan Research Centre, Li Ka Shing Knowledge Institute, Toronto, Canada
- Department of Physiology, University of Toronto, Toronto, Canada
| | - Howard Leong-Poi
- St. Michael’s Hospital, Keenan Research Centre, Li Ka Shing Knowledge Institute, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Andrew Advani
- St. Michael’s Hospital, Keenan Research Centre, Li Ka Shing Knowledge Institute, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Kim A. Connelly
- St. Michael’s Hospital, Keenan Research Centre, Li Ka Shing Knowledge Institute, Toronto, Canada
- Department of Physiology, University of Toronto, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Canada
- * E-mail:
| |
Collapse
|
12
|
Empagliflozin Disrupts a Tnfrsf12a-Mediated Feed Forward Loop That Promotes Left Ventricular Hypertrophy. Cardiovasc Drugs Ther 2021; 36:619-632. [PMID: 33886003 DOI: 10.1007/s10557-021-07190-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/14/2021] [Indexed: 01/10/2023]
Abstract
PURPOSE Although the cardioprotective benefits of sodium-glucose cotransporter 2 (SGLT2) inhibitors are now widely appreciated, the mechanisms underlying these benefits remain unresolved. Tumor necrosis factor receptor superfamily member 12a (Tnfrsf12a) is a receptor for tumor necrosis factor superfamily member 12 (Tnfsf12). Tnfrsf12a is highly inducible and plays a key role in the development of cardiac hypertrophy and heart failure. Here we set out to determine if SGLT2 inhibition affects the Tnfsf12/Tnfrsf12a system in the stressed myocardium. METHODS C57BL/6N mice that had undergone sham or transverse aortic constriction (TAC) surgery were treated with either the SGLT2 inhibitor empagliflozin (400 mg/kg diet; 60-65 mg/kg/day) or standard chow alone and were followed for 8 weeks. Tnfrsf12a expression in mouse hearts was assessed by in situ hybridization, qRT-PCR, and immunoblotting. RESULTS Left ventricular (LV) mass, end-systolic volume, and end-diastolic volume were all increased in TAC mice and were significantly lower with empagliflozin. Myocyte hypertrophy and interstitial fibrosis in TAC hearts were similarly attenuated with empagliflozin. Tnfrsf12a expression was upregulated in mouse hearts following TAC surgery but not in the hearts of empagliflozin-treated mice. In cultured cardiomyocytes, Tnfrsf12a antagonism attenuated the increase in cardiomyocyte size that was induced by phenylephrine. CONCLUSION Empagliflozin attenuates LV enlargement in mice with hypertrophic heart failure. This effect may be mediated, at least in part, by a reduction in loading conditions which limits upregulation of the inducible, proinflammatory, and prohypertrophic TNF superfamily receptor, Tnfrsf12a. Disruption of the Tnfsf12/Tnfrsf12a feed forward system may contribute to the cardioprotective benefits of SGLT2 inhibition.
Collapse
|
13
|
ERK1/2: An Integrator of Signals That Alters Cardiac Homeostasis and Growth. BIOLOGY 2021; 10:biology10040346. [PMID: 33923899 PMCID: PMC8072600 DOI: 10.3390/biology10040346] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 12/24/2022]
Abstract
Integration of cellular responses to extracellular cues is essential for cell survival and adaptation to stress. Extracellular signal-regulated kinase (ERK) 1 and 2 serve an evolutionarily conserved role for intracellular signal transduction that proved critical for cardiomyocyte homeostasis and cardiac stress responses. Considering the importance of ERK1/2 in the heart, understanding how these kinases operate in both normal and disease states is critical. Here, we review the complexity of upstream and downstream signals that govern ERK1/2-dependent regulation of cardiac structure and function. Particular emphasis is given to cardiomyocyte hypertrophy as an outcome of ERK1/2 activation regulation in the heart.
Collapse
|
14
|
Ghosh N, Garg I, Srivastava S, Kumar B. Influence of integrins on thrombus formation: a road leading to the unravelling of DVT. Mol Cell Biochem 2021; 476:1489-1504. [PMID: 33398665 DOI: 10.1007/s11010-020-03961-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 10/23/2020] [Indexed: 01/18/2023]
Abstract
Integrins are a group of transmembrane glycoprotein receptors that are responsible for platelet activation through bidirectional signalling. These receptors have left their footprints in various cellular events and have intrigued many groups of scientists that have led to some significant discoveries. A lot of the recent understanding of haemostasis has been possible due to the integrins filling the gaps in between several cellular mechanism. Apart from this, other important functions carried out by integrins are growth and maturation of cardiomyocytes, mechano-transduction, and interaction with actin cytoskeleton. The signalling cascade for integrin activation involves certain intracellular interacting proteins, which initiates the step-by-step activation procedure through 'inside-out' signalling. The signalling cascade gets activated through 'outside-in' signalling with the involvement of agonists such as ADP, Fibronectin, Vitronectin, and so on. This is a crucial step for the downstream processes of platelet spreading, followed by aggregation, clot progression and finally thrombus formation. Researchers throughout the world have shown direct relation of integrins with CVD and cardiac remodelling. The present review aims to summarize the information available so far on the involvement of integrins in thrombosis and its relationship to DVT. This information could be a bedrock of hidden answers to several questions on pathogenesis of deep vein thrombosis.
Collapse
Affiliation(s)
- Nilanjana Ghosh
- Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India
| | - Iti Garg
- Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India.
| | - Swati Srivastava
- Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India
| | - Bhuvnesh Kumar
- Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India
| |
Collapse
|
15
|
Soppert J, Lehrke M, Marx N, Jankowski J, Noels H. Lipoproteins and lipids in cardiovascular disease: from mechanistic insights to therapeutic targeting. Adv Drug Deliv Rev 2020; 159:4-33. [PMID: 32730849 DOI: 10.1016/j.addr.2020.07.019] [Citation(s) in RCA: 188] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 12/12/2022]
Abstract
With cardiovascular disease being the leading cause of morbidity and mortality worldwide, effective and cost-efficient therapies to reduce cardiovascular risk are highly needed. Lipids and lipoprotein particles crucially contribute to atherosclerosis as underlying pathology of cardiovascular disease and influence inflammatory processes as well as function of leukocytes, vascular and cardiac cells, thereby impacting on vessels and heart. Statins form the first-line therapy with the aim to block cholesterol synthesis, but additional lipid-lowering drugs are sometimes needed to achieve low-density lipoprotein (LDL) cholesterol target values. Furthermore, beyond LDL cholesterol, also other lipid mediators contribute to cardiovascular risk. This review comprehensively discusses low- and high-density lipoprotein cholesterol, lipoprotein (a), triglycerides as well as fatty acids and derivatives in the context of cardiovascular disease, providing mechanistic insights into their role in pathological processes impacting on cardiovascular disease. Also, an overview of applied as well as emerging therapeutic strategies to reduce lipid-induced cardiovascular burden is provided.
Collapse
Affiliation(s)
- Josefin Soppert
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Aachen, Aachen, Germany
| | - Michael Lehrke
- Medical Clinic I, University Hospital Aachen, Aachen, Germany
| | - Nikolaus Marx
- Medical Clinic I, University Hospital Aachen, Aachen, Germany
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Aachen, Aachen, Germany; Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht University, the Netherlands
| | - Heidi Noels
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Aachen, Aachen, Germany; Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands.
| |
Collapse
|
16
|
Batchu SN, Thieme K, Zadeh FH, Alghamdi TA, Yerra VG, Hadden MJ, Majumder S, Kabir MG, Bowskill BB, Ladha D, Gramolini AO, Connelly KA, Advani A. The Dipeptidyl Peptidase 4 Substrate CXCL12 Has Opposing Cardiac Effects in Young Mice and Aged Diabetic Mice Mediated by Ca 2+ Flux and Phosphoinositide 3-Kinase γ. Diabetes 2018; 67:2443-2455. [PMID: 30150305 DOI: 10.2337/db18-0410] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 08/14/2018] [Indexed: 11/13/2022]
Abstract
Blood glucose-lowering therapies can positively or negatively affect heart function in type 2 diabetes, or they can have neutral effects. Dipeptidyl peptidase 4 (DPP-4) inhibitors lower blood glucose by preventing the proteolytic inactivation of glucagon-like peptide 1 (GLP-1). However, GLP-1 is not the only peptide substrate of DPP-4. Here, we investigated the GLP-1-independent cardiac effects of DPP-4 substrates. Pointing to GLP-1 receptor (GLP-1R)-independent actions, DPP-4 inhibition prevented systolic dysfunction equally in pressure-overloaded wild-type and GLP-1R knockout mice. Likewise, DPP-4 inhibition or the DPP-4 substrates substance P or C-X-C motif chemokine ligand 12 (CXCL12) improved contractile recovery after no-flow ischemia in the hearts of otherwise healthy young adult mice. Either DPP-4 inhibition or CXCL12 increased phosphorylation of the Ca2+ regulatory protein phospholamban (PLN), and CXCL12 directly enhanced cardiomyocyte Ca2+ flux. In contrast, hearts of aged obese diabetic mice (which may better mimic the comorbid patient population) had diminished levels of PLN phosphorylation. In this setting, CXCL12 paradoxically impaired cardiac contractility in a phosphoinositide 3-kinase γ-dependent manner. These findings indicate that the cardiac effects of DPP-4 inhibition primarily occur through GLP-1R-independent processes and that ostensibly beneficial DPP-4 substrates can paradoxically worsen heart function in the presence of comorbid diabetes.
Collapse
Affiliation(s)
- Sri N Batchu
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Karina Thieme
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Farigol H Zadeh
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Ted Rogers Centre for Heart Research, University of Toronto, Toronto, Ontario, Canada
| | - Tamadher A Alghamdi
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Veera Ganesh Yerra
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Mitchell J Hadden
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Syamantak Majumder
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | - M Golam Kabir
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Bridgit B Bowskill
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Danyal Ladha
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Anthony O Gramolini
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Ted Rogers Centre for Heart Research, University of Toronto, Toronto, Ontario, Canada
| | - Kim A Connelly
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Andrew Advani
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| |
Collapse
|
17
|
Nishikawa T, Sekiguchi M, Ishibashi-Ueda H. More than 50 Years after Konno's Development of the Endomyocardial Biopsy. Int Heart J 2017; 58:840-846. [PMID: 29118298 DOI: 10.1536/ihj.16-316] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The endomyocardial biopsy (EMB) method was first developed by Japan's Dr. Souji Konno in 1962. Since then, this technique has been used worldwide in clinical cardiology for the recognition and diagnosis of cardiomyopathies, arrhythmias, and other heart conditions. Many studies relating to the EMB have been published at the global level, including a large review by Cooper, et al.,1) wherein a limited selection of Japanese papers were cited despite considerable pioneering work on the EMB having been done in Japan. Following this, the Cardiac Biopsy Conference (CABIC) organization, which was founded in Japan in 1979, conducted a nationwide survey of the English language literature on the EMB. Among the collection of 500 studies compiled, approximately 40 abstracts have been selected by the co-editors in CABIC for further discussion. This report aims to supplement Cooper's work and bring to light other prominent contributions of Japanese researchers on the EMB.
Collapse
Affiliation(s)
- Toshio Nishikawa
- Department of Surgical Pathology, Tokyo Women's Medical University.,Department of Konno Memorial Cardiac Pathology Laboratory, Japan Research Promotion Society for Cardiovascular Diseases
| | - Morie Sekiguchi
- Department of Konno Memorial Cardiac Pathology Laboratory, Japan Research Promotion Society for Cardiovascular Diseases
| | | |
Collapse
|
18
|
Matsuda T, Jeong JI, Ikeda S, Yamamoto T, Gao S, Babu GJ, Zhai P, Del Re DP. H-Ras Isoform Mediates Protection Against Pressure Overload-Induced Cardiac Dysfunction in Part Through Activation of AKT. Circ Heart Fail 2017; 10:CIRCHEARTFAILURE.116.003658. [PMID: 28193718 DOI: 10.1161/circheartfailure.116.003658] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 01/11/2017] [Indexed: 11/16/2022]
Abstract
BACKGROUND In general, Ras proteins are thought to promote cardiac hypertrophy, an important risk factor for cardiovascular disease and heart failure. However, the contribution of different Ras isoforms has not been investigated. The objective of this study was to define the role of H- and K-Ras in modulating stress-induced myocardial hypertrophy and failure. METHODS AND RESULTS We used H- and K-Ras gene knockout mice and subjected them to pressure overload to induce cardiac hypertrophy and dysfunction. We observed a worsened cardiac phenotype in Hras-/- mice, while outcomes were improved in Kras+/- mice. We also used a neonatal rat cardiomyocyte culture system to elucidate the mechanisms underlying these observations. Our findings demonstrate that H-Ras, but not K-Ras, promotes cardiomyocyte hypertrophy both in vivo and in vitro. This response was mediated in part through the phosphoinositide 3-kinase-AKT signaling pathway. Adeno-associated virus-mediated increase in AKT activation improved the cardiac function in pressure overloaded Hras null hearts in vivo. These findings further support engagement of the phosphoinositide 3-kinase-AKT signaling axis by H-Ras. CONCLUSIONS Taken together, these findings indicate that H- and K-Ras have divergent effects on cardiac hypertrophy and heart failure in response to pressure overload stress.
Collapse
Affiliation(s)
- Takahisa Matsuda
- From the Cardiovascular Research Institute and the Department of Cell Biology and Molecular Medicine, Rutgers, New Jersey Medical School, Newark, NJ
| | - Jae Im Jeong
- From the Cardiovascular Research Institute and the Department of Cell Biology and Molecular Medicine, Rutgers, New Jersey Medical School, Newark, NJ
| | - Shohei Ikeda
- From the Cardiovascular Research Institute and the Department of Cell Biology and Molecular Medicine, Rutgers, New Jersey Medical School, Newark, NJ
| | - Takanobu Yamamoto
- From the Cardiovascular Research Institute and the Department of Cell Biology and Molecular Medicine, Rutgers, New Jersey Medical School, Newark, NJ
| | - Shumin Gao
- From the Cardiovascular Research Institute and the Department of Cell Biology and Molecular Medicine, Rutgers, New Jersey Medical School, Newark, NJ
| | - Gopal J Babu
- From the Cardiovascular Research Institute and the Department of Cell Biology and Molecular Medicine, Rutgers, New Jersey Medical School, Newark, NJ
| | - Peiyong Zhai
- From the Cardiovascular Research Institute and the Department of Cell Biology and Molecular Medicine, Rutgers, New Jersey Medical School, Newark, NJ
| | - Dominic P Del Re
- From the Cardiovascular Research Institute and the Department of Cell Biology and Molecular Medicine, Rutgers, New Jersey Medical School, Newark, NJ.
| |
Collapse
|
19
|
Bugyei-Twum A, Abadeh A, Thai K, Zhang Y, Mitchell M, Kabir G, Connelly KA. Suppression of NLRP3 Inflammasome Activation Ameliorates Chronic Kidney Disease-Induced Cardiac Fibrosis and Diastolic Dysfunction. Sci Rep 2016; 6:39551. [PMID: 28000751 PMCID: PMC5175152 DOI: 10.1038/srep39551] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 11/23/2016] [Indexed: 01/28/2023] Open
Abstract
Cardiac fibrosis is a common finding in patients with chronic kidney disease. Here, we investigate the cardio-renal effects of theracurmin, a novel formulation of the polyphenolic compound curcumin, in a rat model of chronic kidney disease. Briefly, Sprague-Dawley rats were randomized to undergo sham or subtotal nephrectomy (SNx) surgery. At 3 weeks post surgery, SNx animals were further randomized to received theracurmin via once daily oral gavage or vehicle for 5 consecutive weeks. At 8 weeks post surgery, cardiac function was assessed via echocardiography and pressure volume loop analysis, followed by LV and renal tissue collection for analysis. SNx animals developed key hallmarks of renal injury including hypertension, proteinuria, elevated blood urea nitrogen, and glomerulosclerosis. Renal injury in SNx animals was also associated with significant diastolic dysfunction, macrophage infiltration, and cardiac NLRP3 inflammasome activation. Treatment of SNx animals with theracurmin improved structural and functional manifestations of cardiac injury associated with renal failure and also attenuated cardiac NLRP3 inflammasome activation and mature IL-1β release. Taken together, our findings suggest a significant role for the NLRP3 inflammasome in renal injury-induced cardiac dysfunction and presents inflammasome attenuation as a unique strategy to prevent adverse cardiac remodeling in the setting of chronic kidney disease.
Collapse
Affiliation(s)
- Antoinette Bugyei-Twum
- Keenan Research Centre for Biomedical Science, St. Michael's hospital, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Armin Abadeh
- Keenan Research Centre for Biomedical Science, St. Michael's hospital, Toronto, Ontario, Canada
| | - Kerri Thai
- Keenan Research Centre for Biomedical Science, St. Michael's hospital, Toronto, Ontario, Canada
| | - Yanling Zhang
- Keenan Research Centre for Biomedical Science, St. Michael's hospital, Toronto, Ontario, Canada
| | - Melissa Mitchell
- Keenan Research Centre for Biomedical Science, St. Michael's hospital, Toronto, Ontario, Canada
| | - Golam Kabir
- Keenan Research Centre for Biomedical Science, St. Michael's hospital, Toronto, Ontario, Canada
| | - Kim A Connelly
- Keenan Research Centre for Biomedical Science, St. Michael's hospital, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.,Division of Cardiology, St. Michael's hospital, Toronto, Ontario, Canada
| |
Collapse
|
20
|
Civitarese RA, Kapus A, McCulloch CA, Connelly KA. Role of integrins in mediating cardiac fibroblast–cardiomyocyte cross talk: a dynamic relationship in cardiac biology and pathophysiology. Basic Res Cardiol 2016; 112:6. [DOI: 10.1007/s00395-016-0598-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 12/14/2016] [Indexed: 12/16/2022]
|
21
|
Dyslipidemia and Diabetes Increase the OPG/TRAIL Ratio in the Cardiovascular System. Mediators Inflamm 2016; 2016:6529728. [PMID: 28070143 PMCID: PMC5192341 DOI: 10.1155/2016/6529728] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 10/31/2016] [Accepted: 11/22/2016] [Indexed: 12/26/2022] Open
Abstract
Background. Dyslipidemia and diabetes are two of the most well established risk factors for the development of cardiovascular disease (CVD). Both of them usually activate a complex range of pathogenic pathways leading to organ damage. Here we hypothesized that dyslipidemia and diabetes could affect osteoprotegerin (OPG) and TNF-related apoptosis-inducing ligand (TRAIL) expression in the vessels and the heart. Materials and Methods. Gene and protein expression of OPG, TRAIL, and OPG/TRAIL ratio were quantified in the aorta and the hearts of control mice, dyslipidemic mice, and diabetic mice. Results. Diabetes significantly increased OPG and the OPG/TRAIL ratio expression in the aorta, while dyslipidemia was the major determinant of the changes observed in the heart, where it significantly increased OPG and reduced TRAIL expression, thus increasing cardiac OPG/TRAIL ratio. Conclusions. This work shows that both dyslipidemia and diabetes affect OPG/TRAIL ratio in the cardiovascular system. This could contribute to the changes in circulating OPG/TRAIL which are observed in patients with diabetes and CVD. Most importantly, these changes could mediate/contribute to atherosclerosis development and cardiac remodeling.
Collapse
|
22
|
Civitarese RA, Talior-Volodarsky I, Desjardins JF, Kabir G, Switzer J, Mitchell M, Kapus A, McCulloch CA, Gullberg D, Connelly KA. The α11 integrin mediates fibroblast–extracellular matrix–cardiomyocyte interactions in health and disease. Am J Physiol Heart Circ Physiol 2016; 311:H96-H106. [DOI: 10.1152/ajpheart.00918.2015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 05/05/2016] [Indexed: 11/22/2022]
Abstract
Excessive cardiac interstitial fibrosis impairs normal cardiac function. We have shown that the α11β1 (α11) integrin mediates fibrotic responses to glycated collagen in rat myocardium by a pathway involving transforming growth factor-β. Little is known of the role of the α11 integrin in the developing mammalian heart. Therefore, we examined the impact of deletion of the α11 integrin in wild-type mice and in mice treated with streptozotocin (STZ) to elucidate the role of the α11 integrin in normal cardiac homeostasis and in the pathogenesis of diabetes-related fibrosis. As anticipated, cardiac fibrosis was reduced in α11 integrin knockout mice (α11−/−; C57BL/6 background) treated with STZ compared with STZ-treated wild-type mice ( P < 0.05). Unexpectedly, diastolic function was impaired in both vehicle and STZ-treated α11−/− mice, as shown by the decreased minimum rate of pressure change and prolonged time constant of relaxation in association with increased end-diastolic pressure (all P < 0.05 compared with wild-type mice). Accordingly, we examined the phenotype of untreated α11−/− mice, which demonstrated a reduced cardiomyocyte cross-sectional cell area and myofibril thickness (all P < 0.05 compared with wild-type mice) and impaired myofibril arrangement. Immunostaining for desmin and connexin 43 showed abnormal intermediate filament organization at intercalated disks and impaired gap-junction development. Overall, deletion of the α11 integrin attenuates cardiac fibrosis in the mammalian mouse heart and reduces ECM formation as a result of diabetes. Furthermore, α11 integrin deletion impairs cardiac function and alters cardiomyocyte morphology. These findings shed further light on the poorly understood interaction between the fibroblast–cardiomyocyte and the ECM.
Collapse
Affiliation(s)
- Robert A. Civitarese
- Keenan Research Center for Biomedical Science, St. Michael's Hospital and University of Toronto, Toronto, Ontario, Canada
| | | | - Jean-Francois Desjardins
- Keenan Research Center for Biomedical Science, St. Michael's Hospital and University of Toronto, Toronto, Ontario, Canada
| | - Golam Kabir
- Keenan Research Center for Biomedical Science, St. Michael's Hospital and University of Toronto, Toronto, Ontario, Canada
| | - Jennifer Switzer
- Keenan Research Center for Biomedical Science, St. Michael's Hospital and University of Toronto, Toronto, Ontario, Canada
| | - Melissa Mitchell
- Keenan Research Center for Biomedical Science, St. Michael's Hospital and University of Toronto, Toronto, Ontario, Canada
| | - Andras Kapus
- Keenan Research Center for Biomedical Science, St. Michael's Hospital and University of Toronto, Toronto, Ontario, Canada
| | | | - Donald Gullberg
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Kim A. Connelly
- Keenan Research Center for Biomedical Science, St. Michael's Hospital and University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
23
|
Hou N, Ye B, Li X, Margulies KB, Xu H, Wang X, Li F. Transcription Factor 7-like 2 Mediates Canonical Wnt/β-Catenin Signaling and c-Myc Upregulation in Heart Failure. Circ Heart Fail 2016; 9. [PMID: 27301468 DOI: 10.1161/circheartfailure.116.003010] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 05/16/2016] [Indexed: 01/17/2023]
Abstract
BACKGROUND How canonical Wnt/β-catenin signals in adult hearts, especially in different diseased states, remains unclear. The proto-oncogene, c-Myc, is a Wnt target and an early response gene during cardiac stress. It is not clear whether c-Myc is activated or how it is regulated during heart failure. METHODS AND RESULTS We investigated canonical Wnt/β-catenin signaling and how it regulated c-Myc expression in failing hearts of human ischemic heart disease, idiopathic dilated cardiomyopathy, and murine desmin-related cardiomyopathy. Our data demonstrated that canonical Wnt/β-catenin signaling was activated through nuclear accumulation of β-catenin in idiopathic dilated cardiomyopathy, ischemic heart disease, and murine desmin-related cardiomyopathy when compared with nonfailing controls and transcription factor 7-like 2 (TCF7L2) was the main β-catenin partner of the T-cell factor (TCF) family in adult hearts. We further revealed that c-Myc mRNA and protein levels were significantly elevated in failing hearts by real-time reverse transcription polymerase chain reaction, Western blotting, and immunohistochemical staining. Immunoprecipitation and confocal microscopy further showed that β-catenin interacted and colocalized with TCF7L2. More importantly, chromatin immunoprecipitation confirmed that β-catenin and TCF7L2 were recruited to the regulatory elements of c-Myc. This recruitment was associated with increased histone H3 acetylation and transcriptional upregulation of c-Myc. With lentiviral infection, TCF7L2 overexpression increased c-Myc expression and cardiomyocyte size, whereas shRNA-mediated knockdown of TCF7L2 suppressed c-Myc expression and cardiomyocyte growth in cultured neonatal rat cardiomyocytes. CONCLUSIONS This study indicates that TCF7L2 mediates canonic Wnt/β-catenin signaling and c-Myc upregulation during abnormal cardiac remodeling in heart failure and suppression of Wnt/β-catenin to c-Myc axis can be explored for preventing and treating heart failure.
Collapse
Affiliation(s)
- Ning Hou
- Department of Pharmacology, Guangzhou Medical University, Guangzhou, PR China.,Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY
| | - Bo Ye
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY.,Department of Laboratory Medicine and Pathology, University of Minnesota, Room 293, Dwan Variety Club Cardiovascular Research Center, 425 E River Pkwy, Minneapolis, MN
| | - Xiang Li
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY
| | - Kenneth B Margulies
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, 3400 Civic Center, Boulevard, Room 11-101, Philadelphia, PA
| | - Haodong Xu
- Department of Pathology and Laboratory Medicine, UCLA Center for the Health Science, Room 13-145E, 10833 Le Conte Ave, Los Angeles, CA
| | - Xuejun Wang
- Division of Basic Biomedical Sciences, University of South Dakota Sanford School of Medicine, Vermillion, SD
| | - Faqian Li
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY.,Department of Laboratory Medicine and Pathology, University of Minnesota, Room 293, Dwan Variety Club Cardiovascular Research Center, 425 E River Pkwy, Minneapolis, MN
| |
Collapse
|
24
|
Connelly KA, Advani A, Advani SL, Zhang Y, Kim YM, Shen V, Thai K, Kelly DJ, Gilbert RE. Impaired cardiac anti-oxidant activity in diabetes: human and correlative experimental studies. Acta Diabetol 2014; 51:771-82. [PMID: 24925443 DOI: 10.1007/s00592-014-0608-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 05/28/2014] [Indexed: 02/06/2023]
Abstract
Increased reactive oxygen species (ROS) are traditionally viewed as arising from the metabolic flux of diabetes, although reduction in the activity of anti-oxidant systems has also been implicated. Among the latter is the major thiol reducing thioredoxin system, the activity of which may be diminished by high glucose-induced expression of its endogenous inhibitor, thioredoxin interacting protein (TxnIP). We assessed TxnIP mRNA/protein expression along with thioredoxin activity in human right atrial biopsy specimens from subjects with and without diabetes undergoing coronary artery grafting. In correlative experimental studies, we examined TxnIP expression in both type 1 and type 2 rodent models of diabetic cardiomyopathy. Finally, we used in vitro gene silencing to determine the contribution of changes in TxnIP abundance to the high glucose-induced reduction in thioredoxin activity. In human right atrial biopsies, diabetes was associated with a >30-fold increase in TxnIP gene expression and a 17 % increase in TxnIP protein expression (both p < 0.05). This was associated with a 21 % reduction in thioredoxin activity when compared to human non-diabetic cardiac biopsy samples (all p < 0.05). In correlative animal studies, both type 1 and type 2 diabetic rats demonstrated a significant increase in TxnIP mRNA and reduction in thioredoxin activity when compared to non-diabetic animals (all p < 0.05). This was associated with a significant increase in ROS (p < 0.05 when compared with control). In cultured cardiac myocytes, high glucose increased ROS and TxnIP mRNA expression, in association with a reduction in thioredoxin activity (p < 0.01). These findings were abrogated by TxnIP small interfering RNA (siRNA). Scrambled siRNA had no effect upon ROS or TxnIP expression. High glucose reduces thioredoxin activity and increases ROS via TxnIP overexpression. These findings suggest that impaired thiol reductive capacity, through altered TxnIP expression, contributes to increased ROS in the diabetic heart.
Collapse
Affiliation(s)
- Kim A Connelly
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 30 Bond St, 209 Victoria Street, Room 7-052, Toronto, ON, M5B 1W8, Canada,
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Javadov S, Jang S, Agostini B. Crosstalk between mitogen-activated protein kinases and mitochondria in cardiac diseases: therapeutic perspectives. Pharmacol Ther 2014; 144:202-25. [PMID: 24924700 DOI: 10.1016/j.pharmthera.2014.05.013] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 05/30/2014] [Indexed: 02/07/2023]
Abstract
Cardiovascular diseases cause more mortality and morbidity worldwide than any other diseases. Although many intracellular signaling pathways influence cardiac physiology and pathology, the mitogen-activated protein kinase (MAPK) family has garnered significant attention because of its vast implications in signaling and crosstalk with other signaling networks. The extensively studied MAPKs ERK1/2, p38, JNK, and ERK5, demonstrate unique intracellular signaling mechanisms, responding to a myriad of mitogens and stressors and influencing the signaling of cardiac development, metabolism, performance, and pathogenesis. Definitive relationships between MAPK signaling and cardiac dysfunction remain elusive, despite 30 years of extensive clinical studies and basic research of various animal/cell models, severities of stress, and types of stimuli. Still, several studies have proven the importance of MAPK crosstalk with mitochondria, powerhouses of the cell that provide over 80% of ATP for normal cardiomyocyte function and play a crucial role in cell death. Although many questions remain unanswered, there exists enough evidence to consider the possibility of targeting MAPK-mitochondria interactions in the prevention and treatment of heart disease. The goal of this review is to integrate previous studies into a discussion of MAPKs and MAPK-mitochondria signaling in cardiac diseases, such as myocardial infarction (ischemia), hypertrophy and heart failure. A comprehensive understanding of relevant molecular mechanisms, as well as challenges for studies in this area, will facilitate the development of new pharmacological agents and genetic manipulations for therapy of cardiovascular diseases.
Collapse
Affiliation(s)
- Sabzali Javadov
- Department of Physiology, School of Medicine, University of Puerto Rico, PR, USA.
| | - Sehwan Jang
- Department of Physiology, School of Medicine, University of Puerto Rico, PR, USA
| | - Bryan Agostini
- Department of Physiology, School of Medicine, University of Puerto Rico, PR, USA
| |
Collapse
|
26
|
Bugyei-Twum A, Advani A, Advani SL, Zhang Y, Thai K, Kelly DJ, Connelly KA. High glucose induces Smad activation via the transcriptional coregulator p300 and contributes to cardiac fibrosis and hypertrophy. Cardiovasc Diabetol 2014; 13:89. [PMID: 24886336 PMCID: PMC4108062 DOI: 10.1186/1475-2840-13-89] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 04/23/2014] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Despite advances in the treatment of heart failure, mortality remains high, particularly in individuals with diabetes. Activated transforming growth factor beta (TGF-β) contributes to the pathogenesis of the fibrotic interstitium observed in diabetic cardiomyopathy. We hypothesized that high glucose enhances the activity of the transcriptional co-activator p300, leading to the activation of TGF-β via acetylation of Smad2; and that by inhibiting p300, TGF-β activity will be reduced and heart failure prevented in a clinically relevant animal model of diabetic cardiomyopathy. METHODS p300 activity was assessed in H9c2 cardiomyoblasts under normal glucose (5.6 mmol/L-NG) and high glucose (25 mmol/L-HG) conditions. 3H-proline incorporation in cardiac fibroblasts was also assessed as a marker of collagen synthesis. The role of p300 activity in modifying TGF-β activity was investigated with a known p300 inhibitor, curcumin or p300 siRNA in vitro, and the functional effects of p300 inhibition were assessed using curcumin in a hemodynamically validated model of diabetic cardiomyopathy - the diabetic TG m(Ren-2)27 rat. RESULTS In vitro, H9c2 cells exposed to HG demonstrated increased p300 activity, Smad2 acetylation and increased TGF-β activity as assessed by Smad7 induction (all p < 0.05 c/w NG). Furthermore, HG induced 3H-proline incorporation as a marker of collagen synthesis (p < 0.05 c/w NG). p300 inhibition, using either siRNA or curcumin reduced p300 activity, Smad acetylation and TGF-β activity (all p < 0.05 c/w vehicle or scrambled siRNA). Furthermore, curcumin therapy reduced 3H-proline incorporation in HG and TGF-β stimulated fibroblasts (p < 0.05 c/w NG). To determine the functional significance of p300 inhibition, diabetic Ren-2 rats were randomized to receive curcumin or vehicle for 6 weeks. Curcumin treatment reduced cardiac hypertrophy, improved diastolic function and reduced extracellular matrix production, without affecting glycemic control, along with a reduction in TGF-β activity as assessed by Smad7 activation (all p < 0.05 c/w vehicle treated diabetic animals). CONCLUSIONS These findings suggest that high glucose increases the activity of the transcriptional co-regulator p300, which increases TGF-β activity via Smad2 acetylation. Modulation of p300 may be a novel strategy to treat diabetes induced heart failure.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Kim A Connelly
- Keenan Research Centre for Biomedical Science, St, Michael's Hospital, 209 Victoria Street, Toronto, ON M5B 1T8, Canada.
| |
Collapse
|
27
|
Loirand G, Sauzeau V, Pacaud P. Small G Proteins in the Cardiovascular System: Physiological and Pathological Aspects. Physiol Rev 2013; 93:1659-720. [DOI: 10.1152/physrev.00021.2012] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Small G proteins exist in eukaryotes from yeast to human and constitute the Ras superfamily comprising more than 100 members. This superfamily is structurally classified into five families: the Ras, Rho, Rab, Arf, and Ran families that control a wide variety of cell and biological functions through highly coordinated regulation processes. Increasing evidence has accumulated to identify small G proteins and their regulators as key players of the cardiovascular physiology that control a large panel of cardiac (heart rhythm, contraction, hypertrophy) and vascular functions (angiogenesis, vascular permeability, vasoconstriction). Indeed, basal Ras protein activity is required for homeostatic functions in physiological conditions, but sustained overactivation of Ras proteins or spatiotemporal dysregulation of Ras signaling pathways has pathological consequences in the cardiovascular system. The primary object of this review is to provide a comprehensive overview of the current progress in our understanding of the role of small G proteins and their regulators in cardiovascular physiology and pathologies.
Collapse
Affiliation(s)
- Gervaise Loirand
- INSERM, UMR S1087; University of Nantes; and CHU Nantes, l'Institut du Thorax, Nantes, France
| | - Vincent Sauzeau
- INSERM, UMR S1087; University of Nantes; and CHU Nantes, l'Institut du Thorax, Nantes, France
| | - Pierre Pacaud
- INSERM, UMR S1087; University of Nantes; and CHU Nantes, l'Institut du Thorax, Nantes, France
| |
Collapse
|
28
|
Sala V, Gallo S, Leo C, Gatti S, Gelb BD, Crepaldi T. Signaling to cardiac hypertrophy: insights from human and mouse RASopathies. Mol Med 2012; 18:938-47. [PMID: 22576369 DOI: 10.2119/molmed.2011.00512] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Accepted: 05/04/2012] [Indexed: 12/19/2022] Open
Abstract
Cardiac hypertrophy is the heart's response to a variety of extrinsic and intrinsic stimuli, some of which might finally lead up to a maladaptive state. An integral part of the pathogenesis of the hypertrophic cardiomyopathy disease (HCM) is the activation of the rat sarcoma (RAS)/RAF/MEK (mitogen-activated protein kinase kinase)/MAPK (mitogen-activated protein kinase) cascade. Therefore, the molecular signaling involving RAS has been the subject of intense research efforts, particularly after the identification of the RASopathies. These constitute a class of developmental disorders caused by germline mutations affecting proteins contributing to the RAS pathway. Among other phenotypic features, a subset of these syndromes is characterized by HCM, prompting researchers and clinicians to delve into the chief signaling constituents of cardiac hypertrophy. In this review, we summarize current advances in the knowledge of the molecular signaling events involved in the pathogenesis of cardiac hypertrophy through work completed on patients and on genetically manipulated animals with HCM and RASopathies. Important insights are drawn from the recognition of parallels between cardiac hypertrophy and cancer. Future research promises to further elucidate the complex molecular interactions responsible for cardiac hypertrophy, possibly pointing the way for the identification of new specific targets for the treatment of HCM.
Collapse
Affiliation(s)
- Valentina Sala
- Department of Anatomy, Pharmacology and Forensic Medicine, University of Turin, Turin, Italy
| | | | | | | | | | | |
Collapse
|
29
|
Wei BR, Simpson RM, Johann DJ, Dwyer JE, Prieto DA, Kumar M, Ye X, Luke B, Shive HR, Webster JD, Hoover SB, Veenstra TD, Blonder J. Proteomic profiling of H-Ras-G12V induced hypertrophic cardiomyopathy in transgenic mice using comparative LC-MS analysis of thin fresh-frozen tissue sections. J Proteome Res 2012; 11:1561-70. [PMID: 22214408 DOI: 10.1021/pr200612y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Determination of disease-relevant proteomic profiles from limited tissue specimens, such as pathological biopsies and tissues from small model organisms, remains an analytical challenge and a much needed clinical goal. In this study, a transgenic mouse disease model of cardiac-specific H-Ras-G12V induced hypertrophic cardiomyopathy provided a system to explore the potential of using mass spectrometry (MS)-based proteomics to obtain a disease-relevant molecular profile from amount-limited specimens that are routinely used in pathological diagnosis. Our method employs a two-stage methanol-assisted solubilization to digest lysates prepared from 8-μm-thick fresh-frozen histological tissue sections of diseased/experimental and normal/control hearts. Coupling this approach with a nanoflow reversed-phase liquid chromatography (LC) and a hybrid linear ion trap/Fourier transform-ion cyclotron resonance MS resulted in the identification of 704 and 752 proteins in hypertrophic and wild-type (control) myocardium, respectively. The disease driving H-Ras protein along with vimentin were unambiguously identified by LC-MS in hypertrophic myocardium and cross-validated by immunohistochemistry and western blotting. The pathway analysis involving proteins identified by MS showed strong association of proteomic data with cardiovascular disease. More importantly, the MS identification and subsequent cross-validation of Wnt3a and β-catenin, in conjunction with IHC identification of phosphorylated GSK-3β and nuclear localization of β-catenin, provided evidence of Wnt/β-catenin canonical pathway activation secondary to Ras activation in the course of pathogenic myocardial hypertrophic transformation. Our method yields results indicating that the described proteomic approach permits molecular discovery and assessment of differentially expressed proteins regulating H-Ras induced hypertrophic cardiomyopathy. Selected proteins and pathways can be further investigated using immunohistochemical techniques applied to serial tissue sections of similar or different origin.
Collapse
Affiliation(s)
- Bih-Rong Wei
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute , Bethesda, Maryland 20892, United States
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Wolfram JA, Lesnefsky EJ, Hoit BD, Smith MA, Lee HG. Therapeutic potential of c-Myc inhibition in the treatment of hypertrophic cardiomyopathy. Ther Adv Chronic Dis 2011; 2:133-44. [PMID: 21858245 DOI: 10.1177/2040622310393059] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Investigating the pathophysiological importance of the molecular and mechanical development of cardiomyopathy is critical to find new and broader means of protection against this disease that is increasing in prevalence and impact. The current available treatment options for cardiomyopathy mainly focus on treating symptoms and strive to make the patient more comfortable while preventing progression of disease and sudden death. The proto-oncogene c-Myc (Myc) has been shown to be increased in many different types of heart disease, including hypertrophic cardiomyopathy, before any signs of the disease are present. As the mechanisms of action and multiple pathways of dependent actions of Myc are being dissected by many research groups, inhibition of Myc is becoming an attractive paradigm for prevention and treatment of cardiomyopathy and heart failure. Elucidating the role Myc plays in the development, propagation and perpetuation of cardiomyopathy and heart failure will one day translate into potential therapeutics for cardiomyopathy.
Collapse
Affiliation(s)
- Julie A Wolfram
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | | | | | | | | |
Collapse
|
31
|
Connelly KA, Advani A, Kim S, Advani SL, Zhang M, White KE, Kim YM, Parker C, Thai K, Krum H, Kelly DJ, Gilbert RE. The cardiac (pro)renin receptor is primarily expressed in myocyte transverse tubules and is increased in experimental diabetic cardiomyopathy. J Hypertens 2011; 29:1175-84. [PMID: 21505358 DOI: 10.1097/hjh.0b013e3283462674] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND The pro(renin) receptor is a 350 amino acid transmembrane protein, that on ligand binding, increases the catalytic efficiency of angiotensinogen cleavage by both prorenin and renin, augmenting angiotensin I formation at the cell surface. While implicated in a broad range of diseases, studies to date have focused on the kidney, particularly in the diabetic context. We sought to examine the site-specific expression of the pro(renin) receptor within the heart. METHODS Using confocal microscopy, site-specific markers and transmission electron microscopy we assessed the location of the pro(renin) receptor in the heart at both cellular/sub-cellular levels. We assessed pro(renin) receptor expression in the setting of disease and blockade of the renin-angiotensin system, using the TGR[m(Ren2)-27] model of diabetic cardiomyopathy and the direct renin inhibitor, aliskiren. RESULTS The pro(renin) receptor was found predominantly at the Z-disc and dyad of cardiac myocytes coinciding closely with the distributions of the vacuolar H⁺-ATPase and ryanodine receptor, known to be located within T-tubules and the sarcoplasmic reticulum's terminal cisternae, respectively. Pro(renin) receptor mRNA/protein abundance were increased ∼3-fold in the hearts of diabetic rats in association with diastolic dysfunction, myocyte hypertrophy and interstitial fibrosis (all P < 0.01). Direct renin inhibition reduced cardiac pro(renin) receptor expression in association with improved cardiac structure/function (all P < 0.05). CONCLUSION Together, these findings are consistent with the notion that the pro(renin) receptor is a component of the vacuolar H⁺-ATPase, and that like the latter, is increased in the setting of cardiac stress and lowered by the administration of an ostensibly cardioprotective agent.
Collapse
Affiliation(s)
- Kim A Connelly
- Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital and University of Toronto, Toronto, Canada
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Connelly KA, Advani A, Advani S, Zhang Y, Thai K, Thomas S, Krum H, Kelly DJ, Gilbert RE. Combination angiotensin converting enzyme and direct renin inhibition in heart failure following experimental myocardial infarction. Cardiovasc Ther 2011; 31:84-91. [PMID: 21884026 DOI: 10.1111/j.1755-5922.2011.00292.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
AIMS Diminishing the activity of the renin-angiotensin system (RAS) plays a pivotal role in the treatment of heart failure. In addition to angiotensin converting enzyme (ACE) inhibitors and angiotensin-receptor blockers, direct renin inhibition has emerged as a potential adjunctive treatment to conventional RAS blockade. We sought to determine the effectiveness of this strategy after myocardial infarction (MI) in the setting of preexisting hypertension, a common premorbid condition in patients with ischemic heart disease. METHODS AND RESULTS Ten-week-old female heterozygous hypertensive (mRen-2)27 transgenic rats (Ren-2), were randomized to one of five groups (n = 8 per group); sham, MI, MI + aliskiren, MI + lisinopril and MI + combination lisinopril and aliskiren. Cardiac function was assessed by echocardiography and in vivo cardiac catheterization. Untreated MI animals developed heart failure with hypotension, dilation, reduced ejection fraction (EF), and raised left ventricular end-diastolic pressure (LVEDP). Treatment with single agent treatment had only modest effect on cardiac function though combination therapy was associated with significant improvements in EF and LVEDP when compared to untreated MI animals (P < 0.05). Histologic analysis demonstrated increase extracellular matrix deposition and cardiomyocyte hypertrophy in the noninfarct region of all MI groups when compared with sham operated animals (P < 0.05) that was reduced by ACE inhibitor monotherapy and combination treatment but not by aliskiren alone. CONCLUSION In a hypertensive rat model that underwent experimental MI, EF, and LVEDP, key functional indices of heart failure, were improved by treatment with combination ACE and direct renin inhibition when compared with either agent used alone.
Collapse
Affiliation(s)
- K A Connelly
- Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital and University of Toronto, Toronto, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
The advent of statins has revolutionised the treatment of patients with raised plasma cholesterol and increased cardiovascular risk. However, the beneficial effects of this class of drugs are far greater than would be expected from lowering of cholesterol alone, and they appear to offer cardiovascular protection at multiple levels, primarily as a result of their pleiotropic activity. Indeed, their favourable effects on the heart seem to be mediated in part through reduced prenylation and subsequent inhibition of small GTPases, particularly those of the Rho family. Such statin-mediated effects are manifested by reduced onset of heart failure and improvements in cardiac dysfunction and remodelling in heart failure patients. Experimental studies have shown that statins mediate their effects on the two major resident cell types of the heart--cardiomyocytes and cardiac fibroblasts--and thus facilitate improvement of adverse remodelling of ischaemic or non-ischaemic aetiology. This review examines evidence for the cellular effects of statins in the heart, and discusses the underlying molecular mechanisms at the level of the cardiomyocyte (hypertrophy, cell death and contractile function) and the cardiac fibroblast (differentiation, proliferation, migration and extracellular matrix synthesis). The prospects for future therapies and ongoing clinical trials are also summarised.
Collapse
|
34
|
Molecular mechanism of size control in development and human diseases. Cell Res 2011; 21:715-29. [PMID: 21483452 DOI: 10.1038/cr.2011.63] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
How multicellular organisms control their size is a fundamental question that fascinated generations of biologists. In the past 10 years, tremendous progress has been made toward our understanding of the molecular mechanism underlying size control. Original studies from Drosophila showed that in addition to extrinsic nutritional and hormonal cues, intrinsic mechanisms also play important roles in the control of organ size during development. Several novel signaling pathways such as insulin and Hippo-LATS signaling pathways have been identified that control organ size by regulating cell size and/or cell number through modulation of cell growth, cell division, and cell death. Later studies using mammalian cell and mouse models also demonstrated that the signaling pathways identified in flies are also conserved in mammals. Significantly, recent studies showed that dysregulation of size control plays important roles in the development of many human diseases such as cancer, diabetes, and hypertrophy.
Collapse
|
35
|
Enhanced cardiac inflammation and fibrosis in ovariectomized hypertensive rats: a possible mechanism of diastolic dysfunction in postmenopausal women. Hypertens Res 2011; 34:496-502. [PMID: 21248760 DOI: 10.1038/hr.2010.261] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Diastolic dysfunction is more prevalent in individuals with hypertension, particularly postmenopausal women; however, the pathogenesis of diastolic dysfunction remains unknown. Pressure overload activates cardiac inflammation, which induces myocardial fibrosis and diastolic dysfunction in rats with a suprarenal aortic constriction (AC). Therefore, we examined the effects of bilateral ovariectomy (OVX) on left ventricle (LV) remodeling, diastolic dysfunction and cardiac inflammation in hypertensive female rats. Rats were randomized to OVX+AC, OVX and AC groups as well as a Control group receiving sham operations for both the procedures. Rats underwent OVX at 6 weeks and AC at 10 weeks (Day 0). At Day 28, OVX did not appear to affect arterial pressure, cardiac hypertrophy or LV fractional shortening in AC rats. However, OVX increased myocardial fibrosis, elevated LV end-diastolic pressure and reduced the transmitral Doppler spectra early to late filling velocity ratio in AC rats. AC-induced transient myocardial monocyte chemoattractant protein-1 expression and macrophage infiltration, both of which peaked at Day 3 and were augmented and prolonged by OVX. At Day 28, dihydroethidium staining revealed superoxide generation in the intramyocardial arterioles in the OVX+AC group but not in the AC group. NOX1, a functional subunit of nicotinamide adenine dinucleotide phosphate oxidase, was upregulated only in the OVX+AC group at Day 28. Chronic 17β-estradiol replacement prevented the increases in macrophage infiltration, NOX1 upregulation, myocardial fibrosis and diastolic dysfunction in OVX+AC rats. In conclusion, we suggest that estrogen deficiency augments cardiac inflammation and oxidative stress and thereby aggravates myocardial fibrosis and diastolic dysfunction in hypertensive female rats. The findings provide insight into the mechanism underlying diastolic dysfunction in hypertensive postmenopausal women.
Collapse
|
36
|
Simvastatin prevents large blood pressure variability induced aggravation of cardiac hypertrophy in hypertensive rats by inhibiting RhoA/Ras–ERK pathways. Hypertens Res 2010; 34:341-7. [DOI: 10.1038/hr.2010.229] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
37
|
Rose BA, Force T, Wang Y. Mitogen-activated protein kinase signaling in the heart: angels versus demons in a heart-breaking tale. Physiol Rev 2010; 90:1507-46. [PMID: 20959622 PMCID: PMC3808831 DOI: 10.1152/physrev.00054.2009] [Citation(s) in RCA: 563] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Among the myriad of intracellular signaling networks that govern the cardiac development and pathogenesis, mitogen-activated protein kinases (MAPKs) are prominent players that have been the focus of extensive investigations in the past decades. The four best characterized MAPK subfamilies, ERK1/2, JNK, p38, and ERK5, are the targets of pharmacological and genetic manipulations to uncover their roles in cardiac development, function, and diseases. However, information reported in the literature from these efforts has not yet resulted in a clear view about the roles of specific MAPK pathways in heart. Rather, controversies from contradictive results have led to a perception that MAPKs are ambiguous characters in heart with both protective and detrimental effects. The primary object of this review is to provide a comprehensive overview of the current progress, in an effort to highlight the areas where consensus is established verses the ones where controversy remains. MAPKs in cardiac development, cardiac hypertrophy, ischemia/reperfusion injury, and pathological remodeling are the main focuses of this review as these represent the most critical issues for evaluating MAPKs as viable targets of therapeutic development. The studies presented in this review will help to reveal the major challenges in the field and the limitations of current approaches and point to a critical need in future studies to gain better understanding of the fundamental mechanisms of MAPK function and regulation in the heart.
Collapse
Affiliation(s)
- Beth A Rose
- Departments of Anesthesiology, Physiology, and Medicine, David Geffen School of Medicine, Molecular Biology, Institute, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | | | | |
Collapse
|
38
|
Achar S, Rostamian A, Narayan SM. Cardiac and metabolic effects of anabolic-androgenic steroid abuse on lipids, blood pressure, left ventricular dimensions, and rhythm. Am J Cardiol 2010; 106:893-901. [PMID: 20816133 DOI: 10.1016/j.amjcard.2010.05.013] [Citation(s) in RCA: 151] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Revised: 05/03/2010] [Accepted: 05/03/2010] [Indexed: 11/25/2022]
Abstract
Recent surveys and reports suggest that many athletes and bodybuilders abuse anabolic-androgenic steroids (AAS). However, scientific data on the cardiac and metabolic complications of AAS abuse are divergent and often conflicting. A total of 49 studies describing 1,467 athletes were reviewed to investigate the cardiovascular effects of the abuse of AAS. Although studies were typically small and retrospective, some associated AAS abuse with unfavorable effects. Otherwise healthy young athletes abusing AAS may show elevated levels of low-density lipoprotein and low levels of high-density lipoprotein. Although data are conflicting, AAS have also been linked with elevated systolic and diastolic blood pressure and with left ventricular hypertrophy that may persist after AAS cessation. Finally, in small case studies, AAS abuse has been linked with acute myocardial infarction and fatal ventricular arrhythmias. In conclusion, recognition of these adverse effects may improve the education of athletes and increase vigilance when evaluating young athletes with cardiovascular abnormalities.
Collapse
|
39
|
McLeod CJ, Bos JM, Theis JL, Edwards WD, Gersh BJ, Ommen SR, Ackerman MJ. Histologic characterization of hypertrophic cardiomyopathy with and without myofilament mutations. Am Heart J 2009; 158:799-805. [PMID: 19853701 DOI: 10.1016/j.ahj.2009.09.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2009] [Accepted: 09/03/2009] [Indexed: 01/19/2023]
Abstract
BACKGROUND Between 30% and 60% of clinical cases of hypertrophic cardiomyopathy (HC) can be attributed to mutations in the genes encoding cardiac myofilament proteins. Interestingly, it appears that the likelihood of an underlying myofilament mutation can be predicted by echocardiographic assessment of left ventricular morphology. However, it is not known whether genotypically characterized HC exists as a separate entity with discrete phenotypic morphology and histology or to what extent recognized polymorphisms of the renin-angiotensin-aldosterone system (RAAS) influence this relationship. The presence of cardiac myofilament and mutations and RAAS polymorphisms will have a strong association with the severity of histologic features of HC and characteristic septal shape. METHODS We conducted a retrospective review of histology specimens, obtained at septal myectomy among 181 patients with medically refractory symptomatic HC. All patients underwent comprehensive genetic analysis for mutations in 8 myofilament-encoding genes; a subset was genotyped for 6 known RAAS-polymorphisms. Patients underwent comprehensive echocardiography by an expert blinded to genotype and microscopic status. RESULTS Microscopically, severity of myocyte hypertrophy appears to be associated with the presence of recognized HC cardiac myofilament mutations (P = .03). Other histologic features characteristic of HC were not consistently associated with myofilament mutation status. A higher burden of pro-LVH RAAS polymorphisms also appeared to predict only myocyte hypertrophy (P = .01). The presence of RAAS polymorphisms was not associated with the development of a specific septal morphology (P = .6). CONCLUSION Myofilament-positive HC does not appear to represent a distinct clinical phenotypic entity as evidenced by specific histologic characteristics and septal shape.
Collapse
|
40
|
Lee HG, Chen Q, Wolfram JA, Richardson SL, Liner A, Siedlak SL, Zhu X, Ziats NP, Fujioka H, Felsher DW, Castellani RJ, Valencik ML, McDonald JA, Hoit BD, Lesnefsky EJ, Smith MA. Cell cycle re-entry and mitochondrial defects in myc-mediated hypertrophic cardiomyopathy and heart failure. PLoS One 2009; 4:e7172. [PMID: 19779629 PMCID: PMC2747003 DOI: 10.1371/journal.pone.0007172] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2009] [Accepted: 08/30/2009] [Indexed: 11/18/2022] Open
Abstract
While considerable evidence supports the causal relationship between increases in c-Myc (Myc) and cardiomyopathy as a part of a "fetal re-expression" pattern, the functional role of Myc in mechanisms of cardiomyopathy remains unclear. To address this, we developed a bitransgenic mouse that inducibly expresses Myc under the control of the cardiomyocyte-specific MHC promoter. In adult mice the induction of Myc expression in cardiomyocytes in the heart led to the development of severe hypertrophic cardiomyopathy followed by ventricular dysfunction and ultimately death from congestive heart failure. Mechanistically, following Myc activation, cell cycle markers and other indices of DNA replication were significantly increased suggesting that cell cycle-related events might be a primary mechanism of cardiac dysfunction. Furthermore, pathological alterations at the cellular level included alterations in mitochondrial function with dysregulation of mitochondrial biogenesis and defects in electron transport chain complexes I and III. These data are consistent with the known role of Myc in several different pathways including cell cycle activation, mitochondrial proliferation, and apoptosis, and indicate that Myc activation in cardiomyocytes is an important regulator of downstream pathological sequelae. Moreover, our findings indicate that the induction of Myc in cardiomyocytes is sufficient to cause cardiomyopathy and heart failure, and that sustained induction of Myc, leading to cell cycle re-entry in adult cardiomyocytes, represents a maladaptive response for the mature heart.
Collapse
Affiliation(s)
- Hyoung-gon Lee
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America
- * E-mail: (HgL); (MAS)
| | - Qun Chen
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Julie A. Wolfram
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Sandy L. Richardson
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Anna Liner
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
- University Hospitals Case Medical Center, Cleveland, Ohio, United States of America
| | - Sandra L. Siedlak
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Xiongwei Zhu
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Nicholas P. Ziats
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Hisashi Fujioka
- Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Dean W. Felsher
- Division of Oncology, Department of Medicine and Pathology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Rudy J. Castellani
- Department of Pathology, University of Maryland, Baltimore, Maryland, United States of America
| | - Maria L. Valencik
- Department of Biochemistry, University of Nevada Reno, Reno, Nevada, United States of America
| | - John A. McDonald
- Department of Biochemistry, University of Nevada Reno, Reno, Nevada, United States of America
| | - Brian D. Hoit
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
- University Hospitals Case Medical Center, Cleveland, Ohio, United States of America
| | - Edward J. Lesnefsky
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
- Louis Stokes Cleveland DVAMC, Cleveland, Ohio, United States of America
| | - Mark A. Smith
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America
- * E-mail: (HgL); (MAS)
| |
Collapse
|
41
|
Cambronero F, Marín F, Roldán V, Hernández-Romero D, Valdés M, Lip GYH. Biomarkers of pathophysiology in hypertrophic cardiomyopathy: implications for clinical management and prognosis. Eur Heart J 2009; 30:139-51. [PMID: 19136482 DOI: 10.1093/eurheartj/ehn538] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The study of biomarkers and their signalling pathways has allowed the development of new therapeutic strategies in a range of disorders. The aim of the present systematic review is to provide an overview of different biomarkers in patients with hypertrophic cardiomyopathy that could give some insight into the pathophysiologic mechanism(s) underlying the typical clinical and histological manifestations of the disease. Several pathophysiological models are presented and discussed, including studies that have investigated these biomarkers for diagnostic and prognostic reasons, in relation to disease progression and/or mortality.
Collapse
Affiliation(s)
- Francisco Cambronero
- Department of Cardiology, Hospital Universitario Virgen de la Arrixaca, Murcia, Spain
| | | | | | | | | | | |
Collapse
|
42
|
Lezoualc'h F, Métrich M, Hmitou I, Duquesnes N, Morel E. Small GTP-binding proteins and their regulators in cardiac hypertrophy. J Mol Cell Cardiol 2008; 44:623-32. [PMID: 18339399 DOI: 10.1016/j.yjmcc.2008.01.011] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2007] [Revised: 01/30/2008] [Accepted: 01/30/2008] [Indexed: 10/22/2022]
Abstract
Small GTP-binding proteins (small G proteins) act as GDP-GTP-regulated molecular switches and are activated by guanine nucleotide exchange factors (GEFs) in response to diverse extracellular stimuli. During this last decade, numerous molecular and cellular studies, as well as genetically-modified animal models, have highlighted the role of small G proteins in the regulation of cardiac hypertrophy. The growing interest in small G protein signalling comes from the fact that chronic hypertrophic response is considered maladaptive and predisposes individuals to heart failure. Although some of the hypertrophic signalling pathways involving small G proteins have now been identified, a central question deals with the identity of the GEFs that modulate small G protein activation in the context of cardiac hypertrophy. Here, we discuss the precise regulation of Ras and Rho subfamilies of GTPases by GEFs and other regulatory proteins during cardiac hypertrophy. In addition, we summarize recent published data, mainly those describing the role of small G proteins in the development of myocardial hypertrophy and we further present the importance of their downstream effectors in myocardial remodelling.
Collapse
Affiliation(s)
- Frank Lezoualc'h
- Inserm, U769, Signalisation et Physiopathologie Cardiaque, Châtenay-Malabry, F-92296, France.
| | | | | | | | | |
Collapse
|
43
|
DWYER JP, RITCHIE ME, SMYTH GK, HARRAP SB, DELBRIDGE LM, DOMENIGHETTI AA, DI NICOLANTONIO R. Myocardial Gene Expression Associated with Genetic Cardiac Hypertrophy in the Absence of Hypertension. Hypertens Res 2008; 31:941-55. [DOI: 10.1291/hypres.31.941] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
44
|
Abstract
Mitogen-activated protein (MAP) kinases belong to a highly conserved family of Ser-Thr protein kinases in the human kinome and have diverse roles in broad physiological functions. The 4 best-characterized MAP kinase pathways, ERK1/2, JNK, p38, and ERK5, have been implicated in different aspects of cardiac regulation, from development to pathological remodeling. Recent advancements in the development of kinase-specific inhibitors and genetically engineered animal models have revealed significant new insights about MAP kinase pathways in the heart. However, this explosive body of new information also has yielded many controversies about the functional role of specific MAP kinases as either detrimental promoters or critical protectors of the heart during cardiac pathological processes. These uncertainties have raised questions on whether/how MAP kinases can be targeted to develop effective therapies against heart diseases. In this review, recent studies examining the role of MAP kinase subfamilies in cardiac development, hypertrophy, and survival are summarized.
Collapse
Affiliation(s)
- Yibin Wang
- Department of Anesthesiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA.
| |
Collapse
|
45
|
Cardiac Hypertrophy. CARDIOVASCULAR MEDICINE 2007. [DOI: 10.1007/978-1-84628-715-2_54] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
46
|
Schulte I, Bektas H, Klempnauer J, Borlak J. Vitamin E in heart transplantation: effects on cardiac gene expression. Transplantation 2006; 81:736-45. [PMID: 16534476 DOI: 10.1097/01.tp.0000191661.90834.b5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Oxidative stress, as induced in organ storage or upon reperfusion, may impact the quality of the transplant. Vitamin E, a radical scavenger, may alleviate stress associated with cardiac surgery. METHODS We investigated the effects of vitamin E treatment in a rat heart transplantation model and link dose to tissue and plasma vitamin E levels. We further studied expression of 39 genes coding for stress markers, proinflammatory cytokines, apoptotic pathways, structural proteins, extracellular matrix, cardiomyocyte specific transcription factors, and metabolic pathways to obtain information on the benefits of vitamin E treatment. RESULTS Treatment of donor and recipient animals significantly increased blood and heart tissue vitamin E levels (P<0.05). We observed a significant reduction (P<0.05) of genes coding for oxygen detoxification (e.g., SOD), as well as expression of the adhesion molecules, ICAM-1 and PECAM-1 (P<0.05). Notably, transcript levels of the stress marker atrial natriuretic peptide (ANP) was reduced (P<0.05), whereas expression of cardiac and skeletal actin was increased. Further, expression of the disease markers beta-MHC and VCAM-1 was repressed (P<0.05), as was expression of the myocyte enhancer factor MEF2c. Noteworthy, vitamin E repressed expression of SP1 and c-fos transcript level. Specifically, their expression is elevated in reperfusion injured tissue. CONCLUSIONS Based on transcript profiling, we propose a novel role of vitamin E in transplant surgery. Our findings warrant further studies to explore its clinical application.
Collapse
Affiliation(s)
- Ingo Schulte
- Fraunhofer Institute of Toxicology and Experimental Medicine, Center for Drug Research and Medical Biotechnology, Hannover, Germany
| | | | | | | |
Collapse
|
47
|
Shimamura S, Endo H, Kutsuna H, Kobayashi M, Hirao H, Shimizu M, Tanaka R, Yamane Y. Effect of intermittent administration of sustained release isosorbide dinitrate (sr-ISDN) in rats with pressure-overload heart. J Vet Med Sci 2006; 68:213-7. [PMID: 16598163 DOI: 10.1292/jvms.68.213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Recent studies have demonstrated the benefits of nitric oxide (NO) on myocardial hypertrophy and myocardial fibrosis. It was suggested that NO has a protective effect on myocardial cell through the neurohormonal system. This effect serves to highlight the important role of NO in maintaining the function and form of heart with chronic heart failure. However, there are no known reports about on the effect of prolonged administration of nitrate on pressure over-load heart. This study was conducted to examine the long-term effect of oral nitrate therapy in rats with pressure-overloaded heart. An abdominal aorta constricted (AC) model of pressure-overloaded heart was created in male Wistar rats. Sustained release isosorbide dinitrate (sr-ISDN) (5 mg/kg once a daily) was administered to the rats once a daily for 12 weeks. The animals were euthanized during the study period, and the heart was collected and weighed. Histopathological examination was performed to evaluate the effect of sr-ISDN on myocardial hypertrophy and fibrosis. The ratio of heart to body weight increased significantly in AC rat and this increase was significantly prevented by sr-ISDN treatment. Histopathological examination showed significant increase in fibrotic area of AC rat compared to sham rat, this increase was inhibited by sr-ISDN treatment. Cardiomyocyte transverse diameter was significantly increased in AC rat compared with sham rat, but this increase tended to decrease by sr-ISDN treatment. In conclusion, intermittent administration with sr-ISDN has mild effect in inhibiting cardiac hypertrophy and marked effect in inhibiting fibrosis due to pressure-overload.
Collapse
Affiliation(s)
- Shunsuke Shimamura
- Department of Veterinary Surgery, Faculth of Agriculture, Tokyo University of Agriculture and Technology, Fuchushi, Tokyo 183-8509, Japan
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Kai H, Mori T, Tokuda K, Takayama N, Tahara N, Takemiya K, Kudo H, Sugi Y, Fukui D, Yasukawa H, Kuwahara F, Imaizumi T. Pressure Overload-Induced Transient Oxidative Stress Mediates Perivascular Inflammation and Cardiac Fibrosis through Angiotensin II. Hypertens Res 2006; 29:711-8. [PMID: 17249527 DOI: 10.1291/hypres.29.711] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Oxidative stress is implicated in the pathogenesis of various cardiovascular diseases. We have shown that in Wistar rats with a suprarenal aortic constriction (AC), pressure overload-induced transient perivascular inflammation (monocyte chemoattractant protein-1 [MCP-1] induction and macrophage accumulation) in the early phase is the determinant of reactive myocardial fibrosis and resultant diastolic dysfunction in the late phase. Thus, we investigated the role of reactive oxygen species production in cardiac remodeling in AC rats. Superoxide production and the footprint of lipid peroxidation were assessed using dihydroethidium staining and immunohistostaining against 4-hydroxy-2-nonenal (4-HNE), respectively. In sham rats, dihydroethidium and 4-HNE signals were scarcely found in the heart. At day 3, AC rats showed dihydroethidium signals mainly in the intramyocardial arterial wall, whereas modest 4-HNE staining was observed diffusely in the myocardium. These signals declined to lower levels by day 14 despite sustained hypertension. Chronic administration of a subdepressor dose of an angiotensin II type 1 receptor blocker candesartan reduced the pressure overload-induced dihydroethidium and 4-HNE signals at day 3. Moreover, candesartan decreased MCP-1 induction and macrophage infiltration at day 3 and prevented myocardial fibrosis at day 14, without affecting left ventricle and myocyte hypertrophy. In conclusion, acute pressure overload induced self-limited superoxide production mainly in the vascular wall. The reactive oxygen species production would contribute to the perivascular inflammation and subsequent myocardial fibrosis. Angiotensin II was suggested to have a pressure-independent effect on the reactive oxygen species production.
Collapse
Affiliation(s)
- Hisashi Kai
- Department of Internal Medicine Division of Cardio-Vascular Medicine, Kurume University School of Medicine, Kurume, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
The challenge of medical practice today is to identify individuals who are at risk of developing disease, determine the severity of the disease and distinguish the responders from the nonresponders to therapy (individualized medicine). Advances in molecular genetics and biology have shifted the paradigm for identification of markers from large-scale epidemiologic studies to studies on genomic- and proteomic-based techniques. Consequently, a large number of biologic markers, referred to as biomarkers, are being identified and validated to serve for risk stratification, prognostication and individualization of therapy. Identification of biomarkers for cardiovascular diseases could also provide insight into the pathogenesis of the phenotype, which is fundamental for the development of specific therapies. The list of biomarkers for cardiovascular disease is expanding rapidly. Nonetheless, the field is in the early stages of evolution and large-scale clinical studies are required to validate the utility of newly identified biomarkers in diagnosis, risk stratification and treatment of cardiovascular diseases. Selected biomarkers for coronary atherosclerosis, acute coronary syndromes and heart failure are discussed in this review.
Collapse
Affiliation(s)
- A J Marian
- Baylor College of Medicine, One Baylor Plaza, 519D, Houston, TX 77030, USA.
| | | |
Collapse
|
50
|
Arendt T, Gärtner U, Seeger G, Barmashenko G, Palm K, Mittmann T, Yan L, Hümmeke M, Behrbohm J, Brückner MK, Holzer M, Wahle P, Heumann R. Neuronal activation of Ras regulates synaptic connectivity. Eur J Neurosci 2004; 19:2953-66. [PMID: 15182302 DOI: 10.1111/j.0953-816x.2004.03409.x] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
A synRas mouse model was used expressing constitutively activated Ha-Ras (Val12 mutation) in neurons to investigate the role of Ras-MAPkinase signalling for neuronal connectivity in adult brain. Expression of the transgene in the cortex of these mice starts after neuronal differentiation is completed and allows to directly investigate the effects of enhanced Ras activity in differentiated neurons. Activation of Ha-Ras induced an increase in soma size which was sensitive to MEK inhibitor in postnatal organotypic cultures. Adult cortical pyramidal neurons showed complex structural rearrangements associated with an increased size and ramification of dendritic arborization. Dendritic spine density was elevated and correlated with a twofold increase in number of synapses. In acute brain slices of the somatosensory and of the visual cortex, extracellular field potentials were recorded from layer II/III neurons. The input-output relation of synaptically evoked field potentials revealed a significantly higher basal excitability of the transgenic mice cortex compared to wild-type animals. In whole cell patch clamp preparations, the frequency of AMPA receptor-mediated spontaneous excitatory postsynaptic currents was increased while the ratio between NMDA and AMPA-receptor mediated signal amplitude was unchanged. A pronounced depression of paired pulse facilitation indicated that Ras contributes to changes at the presynaptic site. Furthermore, synRas mice showed an increased synaptic long-term potentiation, which was sensitive to blockers of NMDA-receptors and of MEK. We conclude that neuronal Ras is a common switch of plasticity in adult mammalian brain sculpturing neuronal architecture and synaptic connectivity in concert with tuning synaptic efficacy.
Collapse
Affiliation(s)
- Thomas Arendt
- Paul Flechsig Institute of Brain Research, Department of Neuroanatomy, University of Leipzig, D-04109 Leipzig, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|