• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 7
  • 1
  • Tagged with
  • 12
  • 12
  • 6
  • 4
  • 3
  • 3
  • 3
  • 3
  • 3
  • 3
  • 3
  • 3
  • 2
  • 2
  • 2
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
1

Rac1 and RhoA Differentially Regulate Angiotensinogen Gene Expression in Stretched Cardiac Fibroblasts

Verma, Suresh K., Lal, Hind, Golden, Honey B., Gerilechaogetu, Fnu, Smith, Manuela, Guleria, Rakeshwar S., Foster, Donald M., Lu, Guangrong, Dostal, David E. 01 April 2011 (has links)
Aims Angiotensin II (Ang II) stimulates cardiac remodelling and fibrosis in the mechanically overloaded myocardium. Although Rho GTPases regulate several cellular processes, including myocardial remodelling, involvement in mediating mechanical stretch-induced regulation of angiotensinogen (Ao), the precursor to Ang II, remains to be determined. We, therefore, examined the role and associated signalling mechanisms of Rho GTPases (Rac1 and RhoA) in regulation of Ao gene expression in a stretch model of neonatal rat cardiac fibroblasts (CFs). Methods and resultsCFs were plated on deformable stretch membranes. Equiaxial mechanical stretch caused significant activation of both Rac1 and RhoA within 25 min. Rac1 activity returned to control levels after 4 h, whereas RhoA remained at a high level of activity until the end of the stretch period (24 h). Mechanical stretch initially caused a moderate decrease in Ao gene expression, but was significantly increased at 824 h. RhoA had a major role in mediating both the stretch-induced inhibition of Ao at 4 h and the subsequent upregulation of Ao expression at 24 h. β1 integrin receptor blockade by Tac β1 expression impaired acute (2 and 15 min) stretch-induced Rac1 activation, but increased RhoA activity. Molecular experiments revealed that Ao gene expression was inhibited by Rac1 through both JNK-dependent and independent mechanisms, and stimulated by RhoA through a p38-dependent mechanism. Conclusion These results indicate that stretch-induced activation of Rac1 and RhoA differentially regulates Ao gene expression by modulating p38 and JNK activation.
2

The TIR/BB-loop mimetic AS-1 Mimetic as-1 Attenuates Mechanical Stress-Induced Cardiac Fibroblast Activation and Paracrine Secretion via Modulation of Large Tumor Suppressor kinase 1

Fan, Min, Song, Juan, He, Yijie, Shen, Xin, Li, Jiantao, Que, Linli, Zhu, Guoqing, Zhu, Quan, Cai, Xin, Ha, Tuanzhu, Chen, Qi, Xu, Yong, Li, Chuanfu, Li, Yuehua 01 June 2016 (has links)
The TIR/BB-loop mimetic AS-1 has been reported to prevent cardiac hypertrophy by inhibiting interleukin-1 receptor (IL-1R)-mediated myeloid differentiation primary response gene 88 (MyD88)-dependent signaling. To date, it remains unknown whether and if so how AS-1 contributes to mechanical stress (MS)-induced cardiac fibroblast activation, a key process in pressure overload-induced cardiac remodeling and heart failure. Here, we show that phosphorylation and expression of large tumor suppressor kinase 1 (LATS1), a key molecule in the Hippo-Yes associated protein (YAP) signaling pathway, were down-regulated in primary neonatal rat cardiac fibroblasts (NRCFs) in response to MS and in the hearts of mice subjected to transverse aortic constriction (TAC) procedure; AS-1 treatment was able to restore LATS1 phosphorylation and expression both in vitro and in vivo. AS-1 treatment suppressed the induction of proliferation, differentiation and collagen synthesis in response to MS in NRCFs. AS-1 also ameliorated cardiomyocyte hypertrophy and apoptosis through dampening paracrine secretion of stretched cardiac fibroblasts. In mice, AS-1 treatment could protect against TAC-induced cardiac hypertrophy, myocardial fibrosis and heart failure. Of note, LATS1 depletion using siRNA completely abrogated the inhibitory effects of AS-1 on NRCFs under MS including accelerated proliferation, differentiation, enhanced ability to produce collagen and augmented paracrine secretion of tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) to induce cardiomyocyte hypertrophy. Therefore, our results delineate a previously unrecognized role for LATS1 in cardiac fibroblast to mediate the beneficial effects of AS-1 in preventing pressure overload-induced cardiac remodeling and heart failure.
3

Pellino1-Mediated TGF-β1 Synthesis Contributes to Mechanical Stress Induced Cardiac Fibroblast Activation

Song, Juan, Zhu, Yun, Li, Jiantao, Liu, Jiahao, Gao, Yun, Ha, Tuanzhu, Que, Linli, Liu, Li, Zhu, Guoqing, Chen, Qi, Xu, Yong, Li, Chuanfu, Li, Yuehua 01 February 2015 (has links)
Activation of cardiac fibroblasts is a key event in the progression of cardiac fibrosis that leads to heart failure. However, the molecular mechanisms underlying mechanical stress-induced cardiac fibroblast activation are complex and poorly understood. This study demonstrates that Pellino1, an E3 ubiquitin ligase, was activated in vivo in pressure overloaded rat hearts and in cultured neonatal rat cardiac fibroblasts (NRCFs) exposed to mechanical stretch in vitro. Suppression of the expression and activity of Pellino1 by adenovirus-mediated delivery of shPellino1 (adv-shpeli1) attenuated pressure overload-induced cardiac dysfunction and cardiac hypertrophy and decreased cardiac fibrosis in rat hearts. Transfection of adv-shpeli1 also significantly attenuated mechanical stress-induced proliferation, differentiation and collagen synthesis in NRCFs. Pellino1 silencing also abrogated mechanical stretch-induced polyubiquitination of tumor necrosis factor-alpha receptor association factor-6 (TRAF6) and receptor-interacting protein 1 (RIP1) and consequently decreased the DNA binding activity of nuclear factor-kappa B (NF-κB) in NRCFs. In addition, Pellino1 silencing prevented stretch-induced activation of p38 and activator protein 1 (AP-1) binding activity in NRCFs. Chromatin Immunoprecipitation (ChIP) and luciferase reporter assays showed that Pellino1 silencing prevented the binding of NF-κB and AP-1 to the promoter region of transforming growth factor-β1 (TGF-β1) thus dampening TGF-β1 transactivation. Our data reveal a previously unrecognized role of Pellino1 in extracellular matrix deposition and cardiac fibroblast activation in response to mechanical stress and provides a novel target for treatment of cardiac fibrosis and heart failure.
4

Investigating inherent functional differences between human cardiac fibroblasts cultured from non-diabetic and type 2 diabetic donors

Sedgwick, B., Riches-Suman, Kirsten, Bageghni, S.A., O'Regan, D.J., Porter, K.E., Turner, N.A. 26 March 2014 (has links)
yes / Introduction Type 2 diabetes mellitus (T2DM) promotes adverse myocardial remodeling and increased risk of heart failure; effects that can occur independently of hypertension or coronary artery disease. As cardiac fibroblasts (CFs) are key effectors of myocardial remodeling, we investigated whether inherent phenotypic differences exist in CF derived from T2DM donors compared with cells from nondiabetic (ND) donors. Methods Cell morphology (cell area), proliferation (cell counting over 7-day period), insulin signaling [phospho-Akt and phospho-extracellular signal-regulated kinase (ERK) Western blotting], and mRNA expression of key remodeling genes [real-time reverse transcription-polymerase chain reaction (RT-PCR)] were compared in CF cultured from atrial tissue from 14 ND and 12 T2DM donors undergoing elective coronary artery bypass surgery. Results The major finding was that Type I collagen (COL1A1) mRNA levels were significantly elevated by twofold in cells derived from T2DM donors compared with those from ND donors; changes reflected at the protein level. T2DM cells had similar proliferation rates but a greater variation in cell size and a trend towards increased cell area compared with ND cells. Insulin-induced Akt and ERK phosphorylation were similar in the two cohorts of cells. Conclusion CF from T2DM individuals possess an inherent profibrotic phenotype that may help to explain the augmented cardiac fibrosis observed in diabetic patients.
5

Role of Non-myocytes in Engineering of Highly Functional Pluripotent Stem Cell-derived Cardiac Tissues

Liau, Brian January 2013 (has links)
<p>Massive loss of cardiac tissue as a result of myocardial infarction can create a poorly-conducting substrate with impaired contractility, ultimately leading to heart failure and lethal arrhythmias. Recent advances in pluripotent stem cell research have provided investigators with potent sources of cardiogenic cells that may be transplanted into failing hearts to provide electrical and mechanical support. Experiments in both small and large animal models have shown that standard cell delivery techniques suffer from poor retention and engraftment of cells. In contrast, the transplantation of engineered cardiac tissues may provide improved cell retention at the injury site, creating a more localized paracrine effect and yielding more efficient structural and functional repair. However, tissue engineering methodologies to assemble cardiomyocytes or cardiac progenitors into aligned, 3-dimensional (3D) myocardial tissues capable of physiologically relevant electrical conduction and force generation are lacking. The objective of this thesis was thus to develop a methodology to generate highly functional engineered cardiac tissues starting from pluripotent stem cells.</p><p>To accomplish this goal, we first derived purified populations of cardiac myocytes from mouse embryonic stem cells (mESC-CMs) by antibiotic selection driven by an &#945;-myosin heavy-chain promoter. Culture conditions that yielded robust mESC-CM electrical coupling and fast action potential propagation were optimized in confluent cell monolayers. We then developed a microfabrication-based tissue engineering approach to create engineered cardiac tissues ("patches") with uniform 3D cell alignment. We found that, unlike in monolayers, mESC-CMs required a population of supporting cardiac fibroblasts to enable the formation of 3D engineered tissues. Detailed structural, electrical and mechanical characterization demonstrated that engineered cardiac patches consisted of dense, uniformly aligned, highly differentiated and electromechanically coupled mESC-CMs and supported rapid action potential conduction velocities between 22 - 25cm/s and contractile force amplitudes of up to 2mN. </p><p>Next, we sought to circumvent the use of primary cardiac fibroblasts by utilizing a single pluripotent stem cell-derived source, multipotent cardiovascular progenitors (CVPs) capable of differentiating into vascular smooth muscle and endothelial cells in addition to cardiomyocytes. CVPs were derived from mouse embryonic stem cells and induced pluripotent stem (iPS) cells by antibiotic selection driven by an Nkx2-5 enhancer element. Similar to mESC-CMs, CVPs formed highly differentiated cell monolayers with electrophysiological properties that improved with time in culture to levels achieved with pure mESC-CMs. However, unlike mESC-CMs, CVPs formed highly functional 3D engineered cardiac tissues without the addition of cardiac fibroblasts, enabling engineered cardiac tissues to be formed from a single, entirely stem cell-derived source.</p><p>Finally, we explored mechanisms of synergistic cardiac fibroblast/myocyte signaling in 3D engineered tissues by using cardiac fibroblasts of different developmental stages in the settings of direct 3D co-culture as well as in conditioned media studies. When co-cultured with fetal cardiac fibroblasts, mESC-CMs were capable of two-fold faster action potential propagation and 1.5-fold higher maximum contractile force generation than when co-cultured with adult cardiac fibroblasts. These functional improvements were associated with enhanced mESC-CM spreading and upregulation of important ion channel, coupling, and contractile proteins. Conditioned medium studies revealed that compared to adult fibroblasts, fetal cardiac fibroblasts secreted distinct paracrine factors that promoted mESC-CM spreading and spontaneous contractility in 3D engineered tissues and acted via the MEK-ERK pathway. Quantitative gene expression analysis revealed paracrine factor candidates that may mediate this action.</p><p>In summary, this thesis presents methods and underlying mechanisms for generation of highly functional cardiac tissues from pluripotent stem cell sources. These techniques and findings provide foundation for future engineering of human ES and iPS cell-based cardiac tissues for therapeutic and drug screening applications.</p> / Dissertation
6

Regulation of cardiac fibroblast function via cyclic AMP, collagen I, III, and VI: implications for post-infarction remodeling

Naugle, Jennifer Elaine 01 August 2006 (has links)
No description available.
7

ROLE OF MECHANOSENSITIVE ION CHANNEL TRPV4 IN CARDIAC REMODELING

Adapala, Ravi kumar 28 March 2018 (has links)
No description available.
8

The role of tubulin acetylation in cardiac fibroblasts

Mügge, Felicitas 27 September 2018 (has links)
No description available.
9

RhoGTPases and their relevance for the afterload-dependent myocardial fibrosis

Ongherth, Anita 11 November 2016 (has links)
No description available.
10

Effects of Methylglyoxal on the Extracellular Matrix and its Interaction with Cardiac Cells

Sheppard-Perkins, Eva 03 January 2023 (has links)
Cardiovascular disease (CVD) is ranked the second leading cause of death in Canada, with 53,704 heart disease-related deaths documented in 2020 alone. After a patient sustains cardiac injury, such as a myocardial infarction (MI), the heart is often unable to undergo sufficient self-recovery for healthy cardiac regeneration and repair; this is largely attributed to fibrotic tissue development at the injury site and subsequent pathological ventricular remodeling. The prevalence of MI events has created a considerable demand to develop novel strategies for effective and safe post-MI therapies. Research has indicated that post-MI modifications interfere with endogenous cardiac repair mechanisms, resulting in a pathological state. After an infarction, there is an accumulation of methylglyoxal (MG) at the site of injury. It has been suggested that MG contributes to ventricular fibrotic development, however its underlying mechanism remains unclear. Additionally, the effects that the post-MI cardiac environment, specifically MG accumulation, has on post-MI therapies and biomaterials has not been sufficiently established. Accordingly, the primary focus of this research project is to elucidate the effects of MG on the collagen-rich extracellular matrix (ECM) of the heart and key cardiac cells involved in the repair process. Further, the interaction between MG and a promising collagen-based hydrogel therapy is investigated, exploring the effects of MG on the hydrogel’s degradative process. It was found that the MG modification of hydrogels did not alter the degradation rate. Additionally, the degradation products of hydrogels, and MG-modified substrates did not affect the properties and formation of myofibroblasts.

Page generated in 0.054 seconds