Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Trevi A. Ramirez is active.

Publication


Featured researches published by Trevi A. Ramirez.


Circulation Research | 2013

Matrix Metalloproteinase-28 Deletion Exacerbates Cardiac Dysfunction and Rupture After Myocardial Infarction in Mice by Inhibiting M2 Macrophage Activation

Yonggang Ma; Ganesh V. Halade; Jianhua Zhang; Trevi A. Ramirez; Daniel L. Levin; Andrew Voorhees; Yu Fang Jin; Hai Chao Han; Anne M. Manicone; Merry L. Lindsey

Rationale: Matrix metalloproteinase (MMP)-28 regulates the inflammatory and extracellular matrix responses in cardiac aging, but the roles of MMP-28 after myocardial infarction (MI) have not been explored. Objective: To determine the impact of MMP-28 deletion on post-MI remodeling of the left ventricle (LV). Methods and Results: Adult C57BL/6J wild-type (n=76) and MMP null (MMP-28−/−, n=86) mice of both sexes were subjected to permanent coronary artery ligation to create MI. MMP-28 expression decreased post-MI, and its cell source shifted from myocytes to macrophages. MMP-28 deletion increased day 7 mortality because of increased cardiac rupture post-MI. MMP-28−/− mice exhibited larger LV volumes, worse LV dysfunction, a worse LV remodeling index, and increased lung edema. Plasma MMP-9 levels were unchanged in the MMP-28−/− mice but increased in wild-type mice at day 7 post-MI. The mRNA levels of inflammatory and extracellular matrix proteins were attenuated in the infarct regions of MMP-28−/− mice, indicating reduced inflammatory and extracellular matrix responses. M2 macrophage activation was impaired when MMP-28 was absent. MMP-28 deletion also led to decreased collagen deposition and fewer myofibroblasts. Collagen cross-linking was impaired as a result of decreased expression and activation of lysyl oxidase in the infarcts of MMP-28−/− mice. The LV tensile strength at day 3 post-MI, however, was similar between the 2 genotypes. Conclusions: MMP-28 deletion aggravated MI-induced LV dysfunction and rupture as a result of defective inflammatory response and scar formation by suppressing M2 macrophage activation.


Cardiovascular Research | 2012

Matrix metalloproteinase-9 deletion attenuates myocardial fibrosis and diastolic dysfunction in ageing mice

Ying Ann Chiao; Trevi A. Ramirez; Rogelio Zamilpa; S. Michelle Okoronkwo; Qiuxia Dai; Jianhua Zhang; Yu Fang Jin; Merry L. Lindsey

AIMS Age-related diastolic dysfunction has been attributed to an increased passive stiffness, which is regulated by extracellular matrix (ECM). We recently showed that matrix metalloproteinase (MMP)-9, an ECM mediator, increases in the left ventricle (LV) with age. The aim of this study, accordingly, was to determine the role of MMP-9 in cardiac ageing. METHODS AND RESULTS We compared LV function in young (6-9 months), middle-aged (12-15 months), old (18-24 months) and senescent (26-34 months) wild-type (WT) and MMP-9 null mice (n ≥ 12/group). All groups had similar fractional shortenings and aortic peak velocities, indicating that systolic function was not altered by ageing or MMP-9 deletion. The mitral ratios of early to late diastolic filling velocities were reduced in old and senescent WT compared with young controls, and this reduction was attenuated in MMP-9 null mice. Concomitantly, the increase in LV collagen content was reduced in MMP-9 null mice (n = 5-6/group). To dissect the mechanisms of these changes, we evaluated the mRNA expression levels of 84 ECM and adhesion molecules by real-time qPCR (n = 6/group). The expression of pro-fibrotic periostin and connective tissue growth factor (CTGF) increased with senescence, as did transforming growth factor-β (TGF-β)-induced protein levels and Smad signalling, and these increases were blunted by MMP-9 deletion. In senescence, MMP-9 deletion also resulted in a compensatory increase in MMP-8. CONCLUSION MMP-9 deletion attenuates the age-related decline in diastolic function, in part by reducing TGF-β signalling-induced periostin and CTGF expression and increasing MMP-8 expression to regulate myocardial collagen turnover and deposition.


Journal of Molecular and Cellular Cardiology | 2012

Transgenic overexpression of matrix metalloproteinase-9 in macrophages attenuates the inflammatory response and improves left ventricular function post-myocardial infarction.

Rogelio Zamilpa; Jessica Ibarra; Lisandra E. de Castro Brás; Trevi A. Ramirez; Nguyen Nguyen; Ganesh V. Halade; Jianhua Zhang; Qiuxia Dai; Tariq Dayah; Ying Ann Chiao; Wesley Lowell; Seema S. Ahuja; Jeanine D'Armiento; Yu Fang Jin; Merry L. Lindsey

Following myocardial infarction (MI), activated macrophages infiltrate into the necrotic myocardium as part of a robust pro-inflammatory response and secrete matrix metalloproteinase-9 (MMP-9). Macrophage activation, in turn, modulates the fibrotic response, in part by stimulating fibroblast extracellular matrix (ECM) synthesis. We hypothesized that overexpression of human MMP-9 in mouse macrophages would amplify the inflammatory and fibrotic responses to exacerbate left ventricular dysfunction. Unexpectedly, at day 5 post-MI, ejection fraction was improved in transgenic (TG) mice (25±2%) compared to the wild type (WT) mice (18±2%; p<0.05). By gene expression profiling, 23 of 84 inflammatory genes were decreased in the left ventricle infarct (LVI) region from the TG compared to WT mice (all p<0.05). Concomitantly, TG macrophages isolated from the LVI, as well as TG peritoneal macrophages stimulated with LPS, showed decreased inflammatory marker expression compared to WT macrophages. In agreement with attenuated inflammation, only 7 of 84 cell adhesion and ECM genes were increased in the TG LVI compared to WT LVI, while 43 genes were decreased (all p<0.05). These results reveal a novel role for macrophage-derived MMP-9 in blunting the inflammatory response and limiting ECM synthesis to improve left ventricular function post-MI.


American Journal of Physiology-heart and Circulatory Physiology | 2011

SPARC mediates early extracellular matrix remodeling following myocardial infarction

Sarah M. McCurdy; Qiuxia Dai; Jianhua Zhang; Rogelio Zamilpa; Trevi A. Ramirez; Tariq Dayah; Nguyen Nguyen; Yu Fang Jin; Amy D. Bradshaw; Merry L. Lindsey

Secreted protein, acidic, and rich in cysteine (SPARC) is a matricellular protein that functions in the extracellular processing of newly synthesized collagen. Collagen deposition to form a scar is a key event following a myocardial infarction (MI). Because the roles of SPARC in the early post-MI setting have not been defined, we examined age-matched wild-type (WT; n=22) and SPARC-deficient (null; n=25) mice at day 3 post-MI. Day 0 WT (n=28) and null (n=20) mice served as controls. Infarct size was 52 ± 2% for WT and 47 ± 2% for SPARC null (P=NS), indicating that the MI injury was comparable in the two groups. By echocardiography, WT mice increased end-diastolic volumes from 45 ± 2 to 83 ± 5 μl (P < 0.05). SPARC null mice also increased end-diastolic volumes but to a lesser extent than WT (39 ± 3 to 63 ± 5 μl; P < 0.05 vs. day 0 controls and vs. WT day 3 MI). Ejection fraction fell post-MI in WT mice from 57 ± 2 to 19 ± 1%. The decrease in ejection fraction was attenuated in the absence of SPARC (65 ± 2 to 28 ± 2%). Fibroblasts isolated from SPARC null left ventricle (LV) showed differences in the expression of 22 genes encoding extracellular matrix and adhesion molecule genes, including fibronectin, connective tissue growth factor (CTGF; CCN2), matrix metalloproteinase-3 (MMP-3), and tissue inhibitor of metalloproteinase-2 (TIMP-2). The change in fibroblast gene expression levels was mirrored in tissue protein extracts for fibronectin, CTGF, and MMP-3 but not TIMP-2. Combined, the results of this study indicate that SPARC deletion preserves LV function at day 3 post-MI but may be detrimental for the long-term response due to impaired fibroblast activation.


Journal of Proteomics | 2013

Texas 3-Step decellularization protocol: Looking at the cardiac extracellular matrix

Lisandra E. de Castro Brás; Trevi A. Ramirez; Kristine Y. DeLeon-Pennell; Ying Ann Chiao; Yonggang Ma; Qiuxia Dai; Ganesh V. Halade; Kevin Hakala; Susan T. Weintraub; Merry L. Lindsey

UNLABELLED The extracellular matrix (ECM) is a critical tissue component, providing structural support as well as important regulatory signaling cues to govern cellular growth, metabolism, and differentiation. The study of ECM proteins, however, is hampered by the low solubility of ECM components in common solubilizing reagents. ECM proteins are often not detected during proteomics analyses using unbiased approaches due to solubility issues and relatively low abundance compared to highly abundant cytoplasmic and mitochondrial proteins. Decellularization has become a common technique for ECM protein-enrichment and is frequently used in engineering studies. Solubilizing the ECM after decellularization for further proteomic examination has not been previously explored in depth. In this study, we describe testing of a series of protocols that enabled us to develop a novel optimized strategy for the enrichment and solubilization of ECM components. Following tissue decellularization, we use acid extraction and enzymatic deglycosylation to facilitate re-solubilization. The end result is the generation of three fractions for each sample: soluble components, cellular components, and an insoluble ECM fraction. These fractions, developed in mass spectrometry-compatible buffers, are amenable to proteomics analysis. The developed protocol allows identification (by mass spectrometry) and quantification (by mass spectrometry or immunoblotting) of ECM components in tissue samples. BIOLOGICAL SIGNIFICANCE The study of extracellular matrix (ECM) proteins in pathological and non-pathological conditions is often hampered by the low solubility of ECM components in common solubilizing reagents. Additionally, ECM proteins are often not detected during global proteomic analyses due to their relatively low abundance compared to highly abundant cytoplasmic and mitochondrial proteins. In this manuscript we describe testing of a series of protocols that enabled us to develop a final novel optimized strategy for the enrichment and solubilization of ECM components. The end result is the generation of three fractions for each sample: soluble components, cellular components, and an insoluble ECM fraction. By analysis of each independent fraction, differences in protein levels can be detected that in normal conditions would be masked. These fractions are amenable to mass spectrometry analysis to identify and quantify ECM components in tissue samples. The manuscript places a strong emphasis on the immediate practical relevance of the method, particularly when using mass spectrometry approaches; additionally, the optimized method was validated and compared to other methodologies described in the literature.


American Journal of Physiology-heart and Circulatory Physiology | 2013

Reduced BDNF attenuates inflammation and angiogenesis to improve survival and cardiac function following myocardial infarction in mice

Ganesh V. Halade; Yonggang Ma; Trevi A. Ramirez; Jianhua Zhang; Qiuxia Dai; Julie G. Hensler; Elizabeth F. Lopez; Omid Ghasemi; Yu Fang Jin; Merry L. Lindsey

Brain-derived neurotrophic factor (BDNF) increases in failing hearts, but BDNF roles in cardiac remodeling following myocardial infarction (MI) are unclear. Male BDNF(+/+) [wild-type (WT)] and BDNF(+/-) heterozygous (HET) mice at 6-9 mo of age were subjected to MI and evaluated at days 1, 3, 5, 7, or 28 post-MI. At day 28 post-MI, 76% of HET versus 40% of WT survived, whereas fractional shortening improved and neovascularization levels were reduced in the HET (all, P < 0.05). At day 1, post-MI, matrix metalloproteinase-9, and myeloperoxidase (MPO) increased in WT, but not in HET. Concomitantly, monocyte chemotactic protein-1 and -5 levels increased and vascular endothelial growth factor (VEGF)-A decreased in HET. Neutrophil infiltration peaked at days 1-3 in WT mice, and this increase was blunted in HET. To determine if MPO administration could rescue the HET phenotype, MPO was injected at 3 h post-MI. MPO restored VEGF-A levels without altering matrix metalloproteinase-9 or neutrophil content. In conclusion, reduced BDNF levels modulated the early inflammatory and neovascularization responses, leading to improved survival and reduced cardiac remodeling at day 28 post-MI. Thus reduced BDNF attenuates early inflammation following MI by modulating MPO and angiogenic response through VEGF-A.


Journal of Molecular and Cellular Cardiology | 2014

Aliskiren and Valsartan Mediate Left Ventricular Remodeling Post-Myocardial Infarction in Mice through MMP-9 Effects

Trevi A. Ramirez; Rugmani Padmanabhan Iyer; Omid Ghasemi; Elizabeth F. Lopez; Daniel B. Levin; Jianhua Zhang; Rogelio Zamilpa; Youn Min Chou; Yu Fang Jin; Merry L. Lindsey

We evaluated whether aliskiren, valsartan, or a combination of both was protective following myocardial infarction (MI) through effects on matrix metalloproteinase (MMP)-9. C57BL/6J wild type (WT, n=94) and MMP-9 null (null, n=85) mice were divided into 4 groups at 3h post-MI: saline (S), aliskiren (A; 50mg/kg/day), valsartan (V; 40mg/kg/day), or A+V and compared to no MI controls at 28days post-MI. All groups had similar infarct areas, and survival rates were higher in the null mice. The treatments influenced systolic function and hypertrophy index, as well as extracellular matrix (ECM) and inflammatory genes in the remote region, indicating that primary effects were on the viable myocardium. Saline treated WT mice showed increased end systolic and diastolic volumes and hypertrophy index, along with reduced ejection fraction. MMP-9 deletion improved LV function post-MI. Aliskiren attenuated the increase in end systolic volume and hypertrophy index, while valsartan improved end diastolic volumes and aliskiren+valsartan improved the hypertrophy index only when MMP-9 was absent. Extracellular matrix and inflammatory gene expression showed distinct patterns among the treatment groups, indicating a divergence in mechanisms of remodeling. This study shows that MMP-9 regulates aliskiren and valsartan effects in mice. These results in mice provide mechanistic insight to help translate these findings to post-MI patients.


Hypertension Research | 2012

Chronic and intermittent hypoxia differentially regulate left ventricular inflammatory and extracellular matrix responses

Trevi A. Ramirez; Claude Jourdan Le Saux; Anne Joy; Jianhua Zhang; Qiuxia Dai; Steve Mifflin; Merry L. Lindsey

We evaluated the left ventricle (LV) response to hypoxia by comparing male Sprague Dawley rats exposed for 7 days to normoxia (control; n=18), chronic sustained hypoxia (CSH; n=12) and chronic intermittent hypoxia (CIH; n=12). Out of the 168 inflammatory, extracellular matrix and adhesion molecule genes evaluated, Ltb, Cdh4, Col5a1, Ecm1, MMP-11 and TIMP-2 increased in the LV (range: 87–138%), whereas Tnfrsf1a decreased 27%, indicating an increase in inflammatory status with CSH (all P<0.05). CIH decreased Ltb, Spp1 and Ccl5 levels, indicating reduced inflammatory status. While Laminin β2 gene levels increased 123%, MMP-9 and fibronectin gene levels both decreased 74% in CIH (all P<0.05). Right ventricle/body weight ratios increased in CSH (1.1±0.1 g g−1) compared with control (0.7±0.1 g g−1) and CIH (0.8±0.1 g g−1; both P<0.05). Lung to body weight increased in CSH, while LV/body weight ratios were similar among all three groups. With CIH, myocyte cross sectional areas increased 25% and perivascular fibrosis increased 100% (both P<0.05). Gene changes were independent of global changes and were validated by protein levels. MMP-9 protein levels decreased 94% and fibronectin protein levels decreased 42% in CIH (both P<0.05). Consistent with a decreased inflammatory status, HIF-2α and eNOS protein levels were 36% and 44% decreased, respectively, in CIH (both P<0.05). In conclusion, our results indicate that following 7 days of hypoxia, inflammation increases in response to CSH and decreases in response to CIH. This report is the first to demonstrate specific and differential changes seen in the LV during chronic sustained and CIH.


Circulation Research | 2012

Matrix Metalloproteinase-28 Deletion Exacerbates Cardiac Dysfunction and Rupture Following Myocardial Infarction in Mice by Inhibiting M2 Macrophage Activation

Yonggang Ma; Ganesh V. Halade; Jianhua Zhang; Trevi A. Ramirez; Daniel L. Levin; Andrew Voorhees; Yu-Fang Jin; Hai Chao Han; Anne M. Manicone; Merry L. Lindsey

Rationale: Matrix metalloproteinase (MMP)-28 regulates the inflammatory and extracellular matrix responses in cardiac aging, but the roles of MMP-28 after myocardial infarction (MI) have not been explored. Objective: To determine the impact of MMP-28 deletion on post-MI remodeling of the left ventricle (LV). Methods and Results: Adult C57BL/6J wild-type (n=76) and MMP null (MMP-28−/−, n=86) mice of both sexes were subjected to permanent coronary artery ligation to create MI. MMP-28 expression decreased post-MI, and its cell source shifted from myocytes to macrophages. MMP-28 deletion increased day 7 mortality because of increased cardiac rupture post-MI. MMP-28−/− mice exhibited larger LV volumes, worse LV dysfunction, a worse LV remodeling index, and increased lung edema. Plasma MMP-9 levels were unchanged in the MMP-28−/− mice but increased in wild-type mice at day 7 post-MI. The mRNA levels of inflammatory and extracellular matrix proteins were attenuated in the infarct regions of MMP-28−/− mice, indicating reduced inflammatory and extracellular matrix responses. M2 macrophage activation was impaired when MMP-28 was absent. MMP-28 deletion also led to decreased collagen deposition and fewer myofibroblasts. Collagen cross-linking was impaired as a result of decreased expression and activation of lysyl oxidase in the infarcts of MMP-28−/− mice. The LV tensile strength at day 3 post-MI, however, was similar between the 2 genotypes. Conclusions: MMP-28 deletion aggravated MI-induced LV dysfunction and rupture as a result of defective inflammatory response and scar formation by suppressing M2 macrophage activation.


bioinformatics and biomedicine | 2012

A biclustering approach to analyze drug effects on extracellular matrix remodeling post-myocardial infarction

Omid Ghasemi; Nguyen Nguyen; Trevi A. Ramirez; Jianhua Zhang; Merry L. Lindsey; Yu Fang Jin

Extracellular matrix (ECM) remodeling is an important process to determine the functional and geometric changes of the left ventricle (LV) post-myocardial infarction (MI). Currently, little research has been performed to determine key factors associated with extracellular matrix remodeling post-ML We have collected the expression levels of 84 genes in LV extracellular matrix from wild type C57BL/6J mice at day 0 (control group), day 28 (MI saline group), and day 28 MI groups treated with Aliskiren, Valsartan, and a combination of these two drugs, given from 3 h post-MI (number=6 each group). Further, we have categorized these genes using sparse singular value decomposition (SSVD) based biclustering algorithm with measurement noises considered. Our results identified the 10 most significant genes in the infarct region, and these genes were cadherin-1, collagen I and IL connective tissue growth factor, matrix metalloproteinase-3, neural cell adhesion molecule-2, osteopontin, thrombospondin-1, Tissue inhibitor of metallopreteinases-1, and tenascin C. We also identified the 15 most significant genes in the non-infarct region, which shared 6 significant genes with the infarct region (collagen IL connective tissue growth factor, matrix metalloproteinase-3, osteopontin, thrombospondin-1, and tenascin C). We then analyzed pathways enriched by the identified significant genes. Interestingly, cell death and adhesion pathways were the most significant functions identified in the infarct region while cell adhesion, cell migration, and inflammatory pathways were enriched in non-infarct region, suggesting their effect on the LV remodeling process. Our results provide a rationale for future research that target these pathways.

Collaboration


Dive into the Trevi A. Ramirez's collaboration.

Top Co-Authors

Avatar

Merry L. Lindsey

University of Mississippi Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jianhua Zhang

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Qiuxia Dai

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Rogelio Zamilpa

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Yu Fang Jin

University of Texas at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Ganesh V. Halade

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Nguyen Nguyen

University of Texas at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Yonggang Ma

University of Mississippi Medical Center

View shared research outputs
Top Co-Authors

Avatar

Yu-Fang Jin

University of Texas at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Tariq Dayah

University of Texas Health Science Center at San Antonio

View shared research outputs
Researchain Logo
Decentralizing Knowledge