25 research outputs found

    The role of thymosin β4 in vascular development

    Get PDF
    Thymosin β4 (Tβ4) is a 43 amino acid peptide encoded by the Tmsb4x gene located on the X-chromosome. It has previously been shown to act as a secreted factor from the myocardium to the overlying epicardium of the developing murine heart, to mediate transformation of epicardial derived progenitor cells (EPDCs) into the coronary vasculature. This PhD project seeks to build on these studies and characterises the function of Tβ4 in the developing systemic vasculature, using the mouse as a model system. Expression analyses demonstrated specific localisation of Tβ4/Tβ4 in the endothelial cells of the embryonic vasculature. In order to ascertain the function of vascular Tβ4, global and endothelial cell specific in vivo Tβ4 loss of function models were examined. Both global and endothelial-specific Tβ4 mutant embryos displayed a reduced recruitment of vascular mural cells to developing blood vessels. Detailed phenotypic examination revealed that the mural cell deficit could be attributed to impaired differentiation of mature mural cells from undifferentiated mesoderm. This process was modelled in vitro, and it was discovered that treatment of the mural progenitor cell lines 10T1/2 and A404 with exogenous Tβ4 could promote their differentiation into mural cells. This process correlated with an increase in Smad phosphorylation and increased activity of the TGF-β pathway. Decreased levels of TGF-β target genes in vivo in Tβ4β null embryos indicated that TGF-β signalling was perturbed in the absence of Tβ4. These findings suggest a model whereby Tβ4 is secreted by the developing endothelium to stimulate the differentiation of uncommitted mesoderm into mature peri-vascular mural cells, via activation of the TGF-β pathway in the target cell population. As a consequence, Tb4 plays an essential role in vascular stability through mural cell support which has implications for vascular dysfunction in disease

    Feasibility of comparing medical management and surgery (with neurosurgery or stereotactic radiosurgery) with medical management alone in people with symptomatic brain cavernoma - protocol for the Cavernomas: A Randomised Effectiveness (CARE) pilot trial.

    Get PDF
    INTRODUCTION: The top research priority for cavernoma, identified by a James Lind Alliance Priority setting partnership was 'Does treatment (with neurosurgery or stereotactic radiosurgery) or no treatment improve outcome for people diagnosed with a cavernoma?' This pilot randomised controlled trial (RCT) aims to determine the feasibility of answering this question in a main phase RCT. METHODS AND ANALYSIS: We will perform a pilot phase, parallel group, pragmatic RCT involving approximately 60 children or adults with mental capacity, resident in the UK or Ireland, with an unresected symptomatic brain cavernoma. Participants will be randomised by web-based randomisation 1:1 to treatment with medical management and with surgery (neurosurgery or stereotactic radiosurgery) versus medical management alone, stratified by prerandomisation preference for type of surgery. In addition to 13 feasibility outcomes, the primary clinical outcome is symptomatic intracranial haemorrhage or new persistent/progressive focal neurological deficit measured at 6 monthly intervals. An integrated QuinteT Recruitment Intervention (QRI) evaluates screening logs, audio recordings of recruitment discussions, and interviews with recruiters and patients/parents/carers to identify and address barriers to participation. A Patient Advisory Group has codesigned the study and will oversee its progress. ETHICS AND DISSEMINATION: This study was approved by the Yorkshire and The Humber-Leeds East Research Ethics Committee (21/YH/0046). We will submit manuscripts to peer-reviewed journals, describing the findings of the QRI and the Cavernomas: A Randomised Evaluation (CARE) pilot trial. We will present at national specialty meetings. We will disseminate a plain English summary of the findings of the CARE pilot trial to participants and public audiences with input from, and acknowledgement of, the Patient Advisory Group. TRIAL REGISTRATION NUMBER: ISRCTN41647111

    Loss of endogenous thymosin β4 accelerates glomerular disease

    Get PDF
    Glomerular disease is characterized by morphologic changes in podocyte cells accompanied by inflammation and fibrosis. Thymosin β4\beta_4 regulates cell morphology, inflammation, and fibrosis in several organs and administration of exogenous thymosin β4\beta_4 improves animal models of unilateral ureteral obstruction and diabetic nephropathy. However, the role of endogenous thymosin β4\beta_4 in the kidney is unknown. We demonstrate that thymosin β4 is expressed prominently in podocytes of developing and adult mouse glomeruli. Global loss of thymosin β4\beta_4 did not affect healthy glomeruli, but accelerated the severity of immune-mediated nephrotoxic nephritis with worse renal function, periglomerular inflammation, and fibrosis. Lack of thymosin β4\beta_4 in nephrotoxic nephritis led to the redistribution of podocytes from the glomerular tuft toward the Bowman capsule suggesting a role for thymosin β4\beta_4 in the migration of these cells. Thymosin β4\beta_4 knockdown in cultured podocytes also increased migration in a wound-healing assay, accompanied by F-actin rearrangement and increased RhoA activity. We propose that endogenous thymosin β4\beta_4 is a modifier of glomerular injury, likely having a protective role acting as a brake to slow disease progression

    Thymosin beta4 and Ac-SDKP: tools to mend a broken heart.

    No full text
    Thymosin beta4 - an endogenously occurring 43 amino acid peptide - has recently been shown to possess cardioprotective properties in the setting of acute myocardial infarction. This review focuses on the reported mechanisms of action through which Thymosin beta4 might accomplish this effect and the clinical prospects for its use as a therapeutic agent

    Essential role for thymosin β4 in regulating vascular smooth muscle cell development and vessel wall stability.

    No full text
    RATIONALE: Compromised development of blood vessel walls leads to vascular instability that may predispose to aneurysm with risk of rupture and lethal hemorrhage. There is currently a lack of insight into developmental insults that may define the molecular and cellular characteristics of initiating and perpetrating factors in adult aneurismal disease. OBJECTIVE: To investigate a role for the actin-binding protein thymosin β4 (Tβ4), previously shown to be proangiogenic, in mural cell development and vascular wall stability. METHODS AND RESULTS: Phenotypic analyses of both global and endothelial-specific loss-of-function Tβ4 mouse models revealed a proportion of Tβ4-null embryos with vascular hemorrhage coincident with a reduction in smooth muscle cell coverage of their developing vessels. Mechanistic studies revealed that extracellular Tβ4 can stimulate differentiation of mesodermal progenitor cells to a mature mural cell phenotype through activation of the transforming growth factor-beta (TGFβ) pathway and that reduced TGFβ signaling correlates with the severity of hemorrhagic phenotype in Tβ4-null vasculature. CONCLUSIONS: Tβ4 is a novel endothelial secreted trophic factor that functions synergistically with TGFβ to regulate mural cell development and vascular wall stability. These findings have important implications for understanding congenital anomalies that may be causative for adult-onset vascular instability

    Thymosin beta4 facilitates epicardial neovascularization of the injured adult heart.

    No full text
    Ischemic heart disease complicated by coronary artery occlusion causes myocardial infarction (MI), which is the major cause of morbidity and mortality in humans (http://www.who.int/cardiovascular_diseases/resources/atlas/en/index.html). After MI the human heart has an impaired capacity to regenerate and, despite the high prevalence of cardiovascular disease worldwide, there is currently only limited insight into how to stimulate repair of the injured adult heart from its component parts. Efficient cardiac regeneration requires the replacement of lost cardiomyocytes, formation of new coronary blood vessels, and appropriate modulation of inflammation to prevent maladaptive remodeling, fibrosis/scarring, and consequent cardiac dysfunction. Here we show that thymosin beta4 (Tbeta4) promotes new vasculature in both the intact and injured mammalian heart. We demonstrate that limited EPDC-derived endothelial-restricted neovascularization constitutes suboptimal "endogenous repair," following injury, which is significantly augmented by Tbeta4 to increase and stabilize the vascular plexus via collateral vessel growth. As such, we identify Tbeta4 as a facilitator of cardiac neovascularization and highlight adult EPDCs as resident progenitors which, when instructed by Tbeta4, have the capacity to sustain the myocardium after ischemic damage
    corecore