hosted by
publicationslist.org
    
george Bou-Gharios

g.gharios@imperial.ac.uk

Journal articles

2008
 
DOI   
PMID 
M J van Amerongen, G Bou-Gharios, Er Popa, J van Ark, A H Petersen, G M van Dam, M J A van Luyn, M C Harmsen (2008)  Bone marrow-derived myofibroblasts contribute functionally to scar formation after myocardial infarction.   J Pathol 214: 3. 377-386 Feb  
Abstract: Myofibroblasts play a major role in scar formation during wound healing after myocardial infarction (MI). Their origin has been thought to be interstitial cardiac fibroblasts. However, the bone marrow (BM) can be a source of myofibroblasts in a number of organs after injury. We have studied the temporal, quantitative and functional role of BM-derived (BMD) myofibroblasts in myocardial scar formation. MI was induced by permanent coronary artery ligation in mice reconstituted with EGFP or pro-Col1A2 transgenic BM. In the latter, luciferase and beta-galactosidase transgene expression mirrors that of the endogenous pro-collagen 1A2 gene, which allows for functional assessment of the recruited cells. After MI, alpha-SMA-positive myofibroblasts and collagen I gradually increased in the infarct area until day 14 and remained constant afterwards. Numerous EGFP-positive BMD cells were present during the first week post-MI, and gradually decreased afterwards until day 28. Peak numbers of BMD myofibroblasts, co-expressing EGFP and alpha-SMA, were found on day 7 post-MI. An average of 21% of the BMD cells in the infarct area were myofibroblasts. These cells constituted up to 24% of all myofibroblasts present. By in vivo IVIS imaging, BMD myofibroblasts were found to be active for collagen I production and their presence was confined to the infarct area. These results show that BMD myofibroblasts participate actively in scar formation after MI.
Notes:
 
DOI   
PMID 
P V Guillot, H T Cook, C D Pusey, N M Fisk, S Harten, J Moss, I Shore, G Bou-Gharios (2008)  Transplantation of human fetal mesenchymal stem cells improves glomerulopathy in a collagen type I alpha 2-deficient mouse.   J Pathol 214: 5. 627-636 Apr  
Abstract: Fetal mesenchymal stem cell (fetal MSC) therapy has potential to treat genetic diseases with early onset, including those affecting the kidney and urinary tract. A collagen type I alpha 2-deficient mouse has a deletion in the alpha2 chain of the procollagen type I gene, resulting in the synthesis of abnormal alpha1(I)(3) homotrimers, which replace normal alpha 1(I)2 alpha 2(I)1 heterotrimers and a glomerulopathy. We first confirmed that col1 alpha 2-deficient homozygous mice show abnormal collagen deposition in the glomeruli, which increases in frequency and severity with postnatal age. Intrauterine transplantation of human MSCs from first trimester fetal blood led postnatally to a reduction of abnormal homotrimeric collagen type I deposition in the glomeruli of 4-12 week-old col1 alpha 2-deficient mice. Using bioluminescence imaging, in situ hybridization and immunohistochemistry in transplanted col1 alpha 2-deficient mice, we showed that the damaged kidneys preferentially recruited donor cells in glomeruli, around mesangial cells. Real-time RT-PCR demonstrated that this effect was seen at an engraftment level of 1% of total cells in the kidney, albeit higher in glomeruli. We conclude that intrauterine transplantation of human fetal MSCs improves renal glomerulopathy in a collagen type I-deficient mouse model. These data support the feasibility of prenatal treatment for hereditary renal diseases.
Notes:
 
DOI   
PMID 
Pascale V Guillot, Oyebode Abass, J H Duncan Bassett, Sandra J Shefelbine, George Bou-Gharios, Jerry Chan, Hitoshi Kurata, Graham R Williams, Julia Polak, Nicholas M Fisk (2008)  Intrauterine transplantation of human fetal mesenchymal stem cells from first-trimester blood repairs bone and reduces fractures in osteogenesis imperfecta mice.   Blood 111: 3. 1717-1725 Feb  
Abstract: The inherited skeletal dysplasia osteogenesis imperfecta (OI) results in multiple fractures and is currently treated empirically. We transplanted human first-trimester fetal blood mesenchymal stem cells (MSCs) into homozygous oim mice in utero. This resulted in a two-thirds reduction in long bone fractures (P < .01), with fewer fractures per mouse (median 1, range 0-2 in mice that received transplants vs median 3, range 1-5 in mice that did not receive transplants by 12 weeks, P < .01). Nearly all mice that did not receive transplants had fractures (47 [97.9%] of 48), in contrast to 17 (58.6%) of 29 4- to 12-week-old mice that received transplants (P < .01). Transplantation was associated with increased bone strength (P < .01), thickness (P < .01), and length (P < .01), and normalization/reduction of growth plate height in 4- to 12-week-old oim was reduced in mice that underwent transplantion (P < .001). More donor cells were found in bone tissues compared with other organs (P < .001), with cells clustered in areas of active bone formation and remodeling, and at sites of fracture healing. Donor cells found in the bone expressed osteoblast lineage genes, and produced the extracellular bone structural protein osteopontin. Finally, MSC transplantation decreased bone hydroxyproline content. In conclusion, intrauterine transplantation of fetal MSCs markedly reduced fracture rates and skeletal abnormalities in a mouse model of the intermediate severity type III OI, suggesting a scientific basis for MSC treatment of affected human fetuses.
Notes:
2007
 
DOI   
PMID 
Xu Shi-wen, Laura Kennedy, Elisabetta A Renzoni, George Bou-Gharios, Roland M du Bois, Carol M Black, Christopher P Denton, David J Abraham, Andrew Leask (2007)  Endothelin is a downstream mediator of profibrotic responses to transforming growth factor beta in human lung fibroblasts.   Arthritis Rheum 56: 12. 4189-4194 Dec  
Abstract: OBJECTIVE: Fibrosis is excessive scarring caused by the accumulation and contraction of extracellular matrix proteins and is a common end pathway in many chronic diseases, including scleroderma (systemic sclerosis [SSc]). Indeed, pulmonary fibrosis is a major cause of death in SSc. Transforming growth factor beta (TGFbeta) induces endothelin 1 (ET-1) in human lung fibroblasts by a Smad-independent, JNK-dependent mechanism. The goal of this study was to assess whether ET-1 is a downstream mediator of the profibrotic effects of TGFbeta in lung fibroblasts. METHODS: We used a specific endothelin receptor antagonist to determine whether ET-1 is a downstream mediator of TGFbeta responses in lung fibroblasts, using microarray technology, real-time polymerase chain reaction, and Western blot analyses. RESULTS: The ability of TGFbeta to induce the expression of a cohort of profibrotic genes, including type I collagen, fibronectin, and CCN2, and to contract a collagen gel matrix, depends on ET-1. CONCLUSION: ET-1 contributes to the ability of TGFbeta to promote a profibrotic phenotype in human lung fibroblasts, consistent with the notion that endothelin receptor antagonism may be beneficial in controlling fibrogenic responses in lung fibroblasts.
Notes:
 
DOI   
PMID 
Xu Shi-Wen, Elisabetta A Renzoni, Laura Kennedy, Sarah Howat, Yunliang Chen, Jeremy D Pearson, George Bou-Gharios, Michael R Dashwood, Roland M du Bois, Carol M Black, Christopher P Denton, David J Abraham, Andrew Leask (2007)  Endogenous endothelin-1 signaling contributes to type I collagen and CCN2 overexpression in fibrotic fibroblasts.   Matrix Biol 26: 8. 625-632 Oct  
Abstract: Fibrosis is excessive scarring caused by the accumulation of extracellular matrix proteins and is a common end pathway in many chronic diseases. Endothelin-1 is a possible contributor to the persistent fibrotic phenotype of fibroblasts isolated from fibrotic lesions. In this report we used a specific dual endothelin A/B receptor antagonist, bosentan, to determine the role of endogenous endothelin signaling in maintaining the profibrotic phenotype of lung fibroblasts from scleroderma patients. Bosentan treatment of lung fibroblasts cultured from normal individuals and individuals with scleroderma was assessed using Affymetrix genome-wide expression profiling, real-time polymerase chain reaction and Western blot analysis and revealed that approximately one-third of the transcripts elevated greater than two-fold in fibrotic fibroblasts were reduced by Bosentan treatment. Genes whose overexpression in fibrotic fibroblasts that were dependent on endogenous endothelin signaling included the matrix or matrix-associated genes type I collagen, fibronectin and CCN2. The elevated adhesive property of fibrotic fibroblasts was also reduced by endothelin receptor antagonism. Basal expression of collagen, fibronectin and CCN2 and adhesion to matrix was not affected. Thus endogenous endothelin signaling contributes to the fibrotic phenotype of fibrotic fibroblasts, suggesting that antagonizing endothelin receptors may be of benefit in combating fibrotic disease.
Notes:
 
DOI   
PMID 
Catherine Alexakis, Terence Partridge, George Bou-Gharios (2007)  Implication of the satellite cell in dystrophic muscle fibrosis: a self-perpetuating mechanism of collagen overproduction.   Am J Physiol Cell Physiol 293: 2. C661-C669 Aug  
Abstract: Because of its mechanical function, skeletal muscle is heavily influenced by the composition of its extracellular matrix (ECM). Fibrosis generated by chronic damage, such as occurs in muscular dystrophies, is thus particularly disastrous in this tissue. Here, we examined the interrelationship between the muscle satellite cell and the production of collagen type I, a major component of fibrotic ECM, by using both C2C12, a satellite cell-derived cell line, and primary muscle satellite cells. In C2C12 cells, we found that expression of collagen type I mRNA decreases substantially during skeletal muscle differentiation. On a single-cell level, collagen type I and myogenin became mutually exclusive after 3 days in differentiation medium, whereas addition of collagen markedly suppressed differentiation of C2C12 cells. Primary cultures of satellite cells associated with isolated single fibers of the young (4 wk old) mdx dystrophic mouse and of C57BL/10ScSn wild-type controls expressed collagen type I and type III mRNA and protein. This pattern persisted in wild-type mice at all ages. But, curiously, in older (18-mo-old) mdx mice, although the myogenic cells continued to express type III collagen, type I expression became restricted to nonmyogenic cells. These cells typically constituted part of a cellular sheet surrounding the old mdx fibers. This combination of features strongly suggests that the progression to fibrosis in dystrophic muscle involves changes in the mechanisms controlling matrix production, which generates positive feedback that results in a reprogramming of myoblasts to a profibrotic function.
Notes:
2006
 
DOI   
PMID 
Candice Roufosse, George Bou-Gharios, Evangelia Prodromidi, Catherine Alexakis, Rosemary Jeffery, Sarah Khan, William R Otto, Julia Alter, Richard Poulsom, H Terence Cook (2006)  Bone marrow-derived cells do not contribute significantly to collagen I synthesis in a murine model of renal fibrosis.   J Am Soc Nephrol 17: 3. 775-782 Mar  
Abstract: Interstitial fibroblasts play a central role in kidney fibrosis. Their origin is debated, with recent data indicating a contribution of bone marrow (BM)-derived cells to the expanded population of interstitial cells after kidney damage in animals and humans. This study investigated whether these BM-derived cells would respond appropriately to a fibrotic drive by producing collagen. A transgenic mouse that expresses both luciferase and beta-galactosidase reporter molecules under the control of a 17-kb promoter and enhancer element of the gene encoding the alpha2 chain of the collagen I was used. Male transgenic BM was transplanted into female wild-type C57BL/6 mice (n=14), and unilateral ureteric obstruction was performed later to induce renal fibrosis. In the obstructed kidney of the BM-chimeric female mice, a mean of 8.6% of smooth muscle actin-positive interstitial cells were Y chromosome positive. Increased collagen I mRNA in the obstructed kidney was detected by in situ hybridization. No luciferase activity was detected by enzyme assays in tissue homogenates of BM recipients, and very few luciferase mRNA transcripts were seen, mainly in tubular cells. beta-Galactosidase activity was not a useful reporter molecule because it could not be distinguished from enhanced endogenous beta-galactosidase activity in the obstructed kidney. These results indicate that BM-derived interstitial cells do not make a significant contribution to collagen I synthesis in the context of renal injury.
Notes:
 
DOI   
PMID 
Catherine Alexakis, Patrick Maxwell, George Bou-Gharios (2006)  Organ-specific collagen expression: implications for renal disease.   Nephron Exp Nephrol 102: 3-4. e71-e75 11  
Abstract: Chronic kidney disease is characterized by progressive accumulation of extracellular matrix and scarring, leading to the loss of kidney function. Excess deposition of the collagen family of proteins is the hallmark of kidney fibrosis. In this review, we survey the collagens that are associated with renal disease and we highlight the use of a transgenic approach to identify cis-acting sequences in the collagen type I promoter which are capable of directing collagen type I expression specifically in the kidney. Ultimately it may be possible to use this approach to halt the accumulation of collagen selectively in this organ.
Notes:
 
DOI   
PMID 
Xu Shi-wen, Lee Anne Stanton, Laura Kennedy, Daphne Pala, Yunliang Chen, Sarah L Howat, Elisabetta A Renzoni, David E Carter, George Bou-Gharios, Richard J Stratton, Jeremy D Pearson, Frank Beier, Karen M Lyons, Carol M Black, David J Abraham, Andrew Leask (2006)  CCN2 is necessary for adhesive responses to transforming growth factor-beta1 in embryonic fibroblasts.   J Biol Chem 281: 16. 10715-10726 Apr  
Abstract: CCN2 is induced by transforming growth factor-beta (TGFbeta) in fibroblasts and is overexpressed in connective tissue disease. CCN2 has been proposed to be a downstream mediator of TGFbeta action in fibroblasts; however, the role of CCN2 in regulating this process unclear. By using embryonic fibroblasts isolated from ccn2-/- mice, we showed that CCN2 is required for a subset of responses to TGFbeta. Affymetrix genome-wide expression profiling revealed that 942 transcripts were induced by TGFbeta greater than 2-fold in ccn2+/+ fibroblasts, of which 345 were not induced in ccn2-/- fibroblasts, including pro-adhesive and matrix remodeling genes. Whereas TGFbeta properly induced a generic Smad3-responsive promoter in ccn2-/- fibroblasts, TGFbeta-induced activation of focal adhesion kinase (FAK) and Akt was reduced in ccn2-/- fibroblasts. Emphasizing the importance of FAK and Akt activation in CCN2-dependent transcriptional responses to TGFbeta in fibroblasts, CCN2-dependent transcripts were not induced by TGFbeta in fak-/- fibroblasts and were reduced by wortmannin in wild-type fibroblasts. Akt1 overexpression in ccn2-/- fibroblasts rescued the TGFbeta-induced transcription of CCN2-dependent mRNA. Finally, induction of TGFbeta-induced fibroblast adhesion to fibronectin and type I collagen was significantly diminished in ccn2-/- fibroblasts. Thus in embryonic fibroblasts, CCN2 is a necessary cofactor required for TGFbeta to activate the adhesive FAK/Akt/phosphatidylinositol 3-kinase cascade, FAK/Akt-dependent genes, and adhesion to matrix.
Notes:
 
DOI   
PMID 
Rajesh P Menon, Malini R Menon, Xu Shi-Wen, Elisabetta Renzoni, George Bou-Gharios, Carol M Black, David J Abraham (2006)  Hammerhead ribozyme-mediated silencing of the mutant fibrillin-1 of tight skin mouse: insight into the functional role of mutant fibrillin-1.   Exp Cell Res 312: 9. 1463-1474 May  
Abstract: The tight skin (Tsk/+) mouse is a model for fibrotic disorders. The genetic defect in the Tsk/+ is an in-frame duplication between exons 17 and 40 of the fibrillin-1 gene which gives rise to a large transcript and protein. Mice homozygous for the mutation die in utero, whereas heterozygotes survive and spontaneously develop connective tissue disease. In this study, we generated hammerhead ribozymes directed against the mutant fibrillin-1 transcript. A partially mispairing ribozyme was the most effective vehicle to cleave the mutant transcript without undesired cleavage of wild type transcripts, as shown by cell-free RNA cleavage and cleavage in cell lines harboring the ribozyme, by RT-PCR, Northern and Western Blotting. Global gene expression profiling using oligonucleotide microarrays showed the expected reduction in fibrillin-1 mRNA, and down-regulation of several gene cohorts in ribozyme harboring TskR1 cells compared to Tsk/+ cells. Two of the functional clusters included genes regulating extracellular matrix such as connective tissue growth factor, serpine-1 (plasminogen activator inhibitor-1) and TIMP-1 and TIMP-3, and those involved in cytoskeletal organization and myofibroblast formation including calponins and transgelin. Ribozyme-mediated inhibition was confirmed by Western Blot and functional analysis using cell-reporter systems and remodeling of three dimensional collagen gels. Our results underline the therapeutic potential of hammerhead ribozymes in dominant negative defects and suggest that changes in microfibril architecture brought about by fibrillin-1 mutation lead to a complex disease phenotype.
Notes:
 
DOI   
PMID 
Francesco P Russo, Malcolm R Alison, Brian W Bigger, Eunice Amofah, Aikaterini Florou, Farhana Amin, George Bou-Gharios, Rosemary Jeffery, John P Iredale, Stuart J Forbes (2006)  The bone marrow functionally contributes to liver fibrosis.   Gastroenterology 130: 6. 1807-1821 May  
Abstract: BACKGROUND & AIMS: Bone marrow (BM) cells may transdifferentiate into or fuse with organ parenchymal cells. BM therapy shows promise in murine models of cirrhosis, and clinical trials of bone marrow stem cell therapy for organ healing are underway. However, the BM may contribute to scar-forming myofibroblasts in various organs including the liver. We have studied this axis of regeneration and scarring in murine models of cirrhosis, including an assessment of the temporal and functional contribution of the BM-derived myofibroblasts. METHODS: Female mice were lethally irradiated and received male BM transplants. Carbon tetrachloride or thioacetamide was used to induce cirrhosis. BM-derived cells were tracked through in situ hybridization for the Y chromosome. BM transplants from 2 strains of transgenic mice were used to detect intrahepatic collagen production. RESULTS: In the cirrhotic liver, the contribution of BM to parenchymal regeneration was minor (0.6%); by contrast, the BM contributed significantly to hepatic stellate cell (68%) and myofibroblast (70%) populations. These BM-derived cells were found to be active for collagen type 1 transcription in 2 independent assays and could influence the fibrotic response to organ injury. These BM-derived myofibroblasts did not occur through cell fusion between BM-derived cells and indigenous hepatic cells but, instead, originated largely from the BM's mesenchymal stem cells. CONCLUSIONS: The BM contributes functionally and significantly to liver fibrosis and is a potential therapeutic target in liver fibrosis. Clinical trials of BM cell therapy for liver regeneration should be vigilant for the possibility of enhanced organ fibrosis.
Notes:
 
PMID 
Vineeth S Rajkumar, Xu Shiwen, Maria Bostrom, Patricia Leoni, John Muddle, Mikael Ivarsson, Bengt Gerdin, Christopher P Denton, George Bou-Gharios, Carol M Black, David J Abraham (2006)  Platelet-derived growth factor-beta receptor activation is essential for fibroblast and pericyte recruitment during cutaneous wound healing.   Am J Pathol 169: 6. 2254-2265 Dec  
Abstract: Connective tissue remodeling provides mammals with a rapid mechanism to repair wounds after injury. Inappropriate activation of this reparative process leads to scarring and fibrosis. Here, we studied the effects of platelet-derived growth factor receptor-beta blockade in vivo using the platelet-derived growth factor receptor (PDGFR)-beta inhibitor imatinib mesylate on tissue repair. After 7 days, healing of wounds was delayed with significantly reduced wound closure and concomitant reduction in myofibroblast frequency, expression of fibronectin ED-A, and collagen type I. Using a collagen type I transgenic reporter mouse, we showed that inhibiting PDGFR-beta activation restricted the distribution of collagen-synthesizing cells to wound margins and dramatically reduced cell proliferation in vivo. By 14 days, treated wounds were fully closed. Blocking PDGFR-beta signaling did not prevent the differentiation of myofibroblasts in vitro but potently inhibited fibroblast proliferation and migration. In addition, PDGFR-beta inhibition in vivo was accompanied by abnormal microvascular morphogenesis reminiscent of that observed in PDGFR-beta-/- mice with significantly reduced immunostaining of the pericyte marker NG2. Imatinib treatment also inhibited pericyte proliferation and migration in vitro. This study highlights the significance of PDGFR-beta signaling for the recruitment, proliferation, and functional activities of fibro-blasts and pericytes during the early phases of wound healing.
Notes:
 
DOI   
PMID 
Francesco Ramirez, Shizuko Tanaka, George Bou-Gharios (2006)  Transcriptional regulation of the human alpha2(I) collagen gene (COL1A2), an informative model system to study fibrotic diseases.   Matrix Biol 25: 6. 365-372 Aug  
Abstract: During the past two decades, the human pro-alpha2(I) collagen gene (COL1A2) has emerged as an informative model in which to study the general principles that govern the transcriptional control of extracellular matrix deposition in normal and fibrotic conditions. Multiple studies have in fact delineated the genomic regions, cis-acting elements and trans-acting factors implicated in constitutive, cytokine-modulated and tissue-specific expression of COL1A2. These functional components are integrated into a regulatory network that consists of the proximal promoter, far-upstream enhancer and downstream repressor, and which operates according to two mechanisms. The first mechanism is one in which combinatorial interactions among promoter-bound proteins determine transcriptional outcome in different cellular and experimental contexts. The other mechanism is one whereby cooperative assembly of protein complexes at distantly located DNA elements directs spatiotemporal specificity. These transcriptional studies have also an additional value in translational research, in that they are providing the conceptual means to develop new animal models of and therapeutic strategies for fibrotic diseases.
Notes:
2005
 
DOI   
PMID 
Jamie Morrison, Terence Partridge, George Bou-Gharios (2005)  Nude mutation influences limb skeletal muscle development.   Matrix Biol 23: 8. 535-542 Jan  
Abstract: The nude mouse lacks functional T cells and other skin defects due to the absence of Foxn1 transcription factor. The mdx mouse bred onto a nude background (Foxn1-/-), to generate mdx nu/nu, serves as a valuable model for the use in muscle cell transplantation or gene therapy studies, to avoid a detrimental immune response. However, it has been shown previously that the mdx nu/nu mice have a skeletal muscle specific alteration in collagen production, which in the case of the limb muscles acts at or before the onset of the disease. This led to the conclusion that the nude mutation has an effect on the collagen deposition in skeletal muscles, which may be acting in a T cell independent manner. In an attempt to elucidate the role of the nude mutation on collagen production in skeletal muscles, we have measured collagen levels in several muscles of C57BL/10 nu/nu mice and compared them with mdx nu/nu and Foxn1+/+ control mice. We report here that the limb muscles of the young C57BL/10 nu/nu and mdx nu/nu mice contained a far higher proportion of collagen at an early age than Foxn1+/- control mice but regained normal levels by 12 weeks of age. This was largely attributable to the fact that 3 weeks old nude mice had considerably smaller limb muscles than Foxn1+/- mice which subsequently grew to normal size. We conclude that the nude mutation retards the early growth of development of muscle but not of the matrix tissue in limb muscles.
Notes:
 
DOI   
PMID 
Qi Long Lu, Adam Rabinowitz, Yun Chao Chen, Toshifumi Yokota, HaiFang Yin, Julia Alter, Atif Jadoon, George Bou-Gharios, Terence Partridge (2005)  Systemic delivery of antisense oligoribonucleotide restores dystrophin expression in body-wide skeletal muscles.   Proc Natl Acad Sci U S A 102: 1. 198-203 Jan  
Abstract: Antisense oligonucleotide-mediated alternative splicing has great potential for treatment of Duchenne muscular dystrophy (DMD) caused by mutations within nonessential regions of the dystrophin gene. We have recently shown in the dystrophic mdx mouse that exon 23, bearing a nonsense mutation, can be skipped after intramuscular injection of a specific 2'-O-methyl phosphorothioate antisense oligoribonucleotide (2OMeAO). This skipping created a shortened, but in-frame, transcript that is translated to produce near-normal levels of dystrophin expression. This expression, in turn, led to improved muscle function. However, because DMD affects muscles body-wide, effective treatment requires dystrophin induction ideally in all muscles. Here, we show that systemic delivery of specific 2OMeAOs, together with the triblock copolymer F127, induced dystrophin expression in all skeletal muscles but not in cardiac muscle of the mdx dystrophic mice. The highest dystrophin expression was detected in diaphragm, gastrocnemius, and intercostal muscles. Large numbers of fibers with near-normal level of dystrophin were observed in focal areas. Three injections of 2OMeAOs at weekly intervals enhanced the levels of dystrophin. Dystrophin mRNA lacking the targeted exon 23 remained detectable 2 weeks after injection. No evidence of tissue damage was detected after 2OMeAO and F127 treatment either by serum analysis or histological examination of liver, kidney, lung, and muscles. The simplicity and safety of the antisense protocol provide a realistic prospect for treatment of the majority of DMD mutations. We conclude that a significant therapeutic effect may be achieved by further optimization in dose and regime of administration of antisense oligonucleotide.
Notes:
 
DOI   
PMID 
Christopher P Denton, Gisela E Lindahl, Korsa Khan, Xu Shiwen, Voon H Ong, Nicholas J Gaspar, Konstantinos Lazaridis, Dylan R Edwards, Andrew Leask, Mark Eastwood, Patricia Leoni, Elisabetta A Renzoni, George Bou Gharios, David J Abraham, Carol M Black (2005)  Activation of key profibrotic mechanisms in transgenic fibroblasts expressing kinase-deficient type II Transforming growth factor-{beta} receptor (T{beta}RII{delta}k).   J Biol Chem 280: 16. 16053-16065 Apr  
Abstract: We have generated transgenic mice expressing a kinase-deficient type II transforming growth factor-beta (TGFbeta) receptor selectively on fibroblasts (TbetaRIIDeltak-fib). These mice develop dermal and pulmonary fibrosis. In the present study we explore activation of TGFbeta signaling pathways in this strain and examine the profibrotic properties of explanted transgenic fibroblasts including myofibroblast differentiation and abnormal metalloproteinase production. Gene expression profiles of littermate wild type or transgenic fibroblasts were compared using high-density gene arrays and validated by Taqman reverse transcriptase-PCR, Northern and Western blotting. Using a specific inhibitor (SD-208) we demonstrate that the abnormal phenotype of these cells is dependent upon TbetaRI kinase (ALK5) activity, and that transgenic fibroblasts show enhanced expression and activation of TGFbeta together with increased levels of wild type TbetaRII. Moreover, we confirm that transgene expression is itself regulated by TGFbeta and that expression at low levels facilitates signaling, whereas high level expression is inhibitory. For a subset of TGFbeta responsive genes basal up-regulation is normalized or suppressed by exogenous recombinant TGFbeta1 at time points coincident with increased transgene expression. These findings explain the profound refractoriness of TbetaRIIDeltak-fib fibroblasts to exogenous TGFbeta1, despite their activated phenotype. Thus, transgenic fibroblasts recapitulate many hallmark biochemical properties of fibrotic cells, including high level CTGF (CCN2) expression and type I collagen overproduction, altered MMP production, and myofibroblast differentiation. These cells also show an enhanced ability to contract collagen gel matrices. Our study demonstrates that altered high affinity TGFbeta receptor function may lead to ligand-dependent activation of downstream signaling, and provides further evidence of a pivotal role for sustained TGFbeta overactivity in fibrosis.
Notes:
 
PMID 
Yunliang Chen, Xu Shi-Wen, Jonathan van Beek, Laura Kennedy, Marilyn McLeod, Elisabetta A Renzoni, George Bou-Gharios, Sarah Wilcox-Adelman, Paul F Goetinck, Mark Eastwood, Carol M Black, David J Abraham, Andrew Leask (2005)  Matrix contraction by dermal fibroblasts requires transforming growth factor-beta/activin-linked kinase 5, heparan sulfate-containing proteoglycans, and MEK/ERK: insights into pathological scarring in chronic fibrotic disease.   Am J Pathol 167: 6. 1699-1711 Dec  
Abstract: Scarring is characterized by excessive synthesis and contraction of extracellular matrix. Here, we show that fibroblasts from scarred (lesional) areas of patients with the chronic fibrotic disorder diffuse scleroderma [diffuse systemic sclerosis (dSSc)] show an enhanced ability to adhere to and contract extracellular matrix, relative to fibroblasts from unscarred (nonlesional) areas of dSSc patients and dermal fibroblasts from normal, healthy individuals. The contractile abilities of normal and dSSc dermal fibroblasts were suppressed by blocking heparin sulfate-containing proteoglycan biosynthesis or antagonizing transforming growth factor-beta receptor type I [activin-linked kinase (ALK5)] or ras/mitogen-activated protein kinase (MEK)/extracellular signal-regulated kinase (ERK). Compared with both normal and nonlesional fibroblasts, lesional dSSc fibroblasts overexpressed the heparin sulfate-containing proteoglycan syndecan 4. We also found that the procontractile signals from transforming growth factor (TGF)-beta were integrated through syndecan 4 and MEK/ERK because the ability of TGFbeta to induce contraction of dermal fibroblasts was prevented by MEK antagonism. TGFbeta could not induce a contractile phenotype or phosphorylate ERK in syndecan 4(-/-) dermal fibroblasts. These results suggest that integrating TGFbeta and ERK signals via syndecan 4 is essential for the contractile ability of dermal fibroblasts. We conclude that antagonizing MEK/ERK, TGFbeta1/ALK5, or syndecan 4 may alleviate scarring in chronic fibrotic disease.
Notes:
 
PMID 
Jamie Morrison, Donald B Palmer, Stephen Cobbold, Terence Partridge, George Bou-Gharios (2005)  Effects of T-lymphocyte depletion on muscle fibrosis in the mdx mouse.   Am J Pathol 166: 6. 1701-1710 Jun  
Abstract: Duchenne muscular dystrophy was initially described as a myosclerosis because of the conspicuous progression of interstitial fibrosis. Using the mdx mouse homologue, we have shown previously that the accumulation of intramuscular collagen is profoundly influenced by the presence or absence of T lymphocytes. Here we have used thymectomy and antibody depletion to examine the effect of ablating CD4 or CD8 or both subsets of T lymphocytes on skeletal muscle fibrosis in mdx and C57BL10 (wild-type) mice. Depletion of either or both subsets at 4 weeks of age did not influence fibrosis in mdx mice, as determined by measuring hydroxyproline levels and collagen deposition in diaphragm. Additionally, expression of transforming growth factor-beta1, which is implicated in collagen deposition, either decreased (mdx mice) or increased (C57BL/10 mice) after double CD4/8 depletion. Our data suggest that depletion of lymphoid cells may affect the tight regulatory control of transforming growth factor-beta1, with possible pleiotropic effects, and more importantly, that the fibrotic process is self-sustaining from a very early stage.
Notes:
 
DOI   
PMID 
Taras T Antoniv, Shizuko Tanaka, Bayan Sudan, Sarah De Val, Ke Liu, Lu Wang, Dominic J Wells, George Bou-Gharios, Francesco Ramirez (2005)  Identification of a repressor in the first intron of the human alpha2(I) collagen gene (COL1A2).   J Biol Chem 280: 42. 35417-35423 Oct  
Abstract: The human and mouse genes that code for the alpha2 chain of collagen I (COL1A2 and Col1a2, respectively) share a common chromatin structure and nearly identical proximal promoter and far upstream enhancer sequences. Despite these homologies, species-specific differences have been reported regarding the function of individual cis-acting elements, such as the first intron sequence. In the present study, we have investigated the transcriptional contribution of the unique open chromatin site in the first intron of COL1A2 using a transgenic mouse model. DNase I footprinting identified a cluster of three distinct areas of nuclease protection (FI1-3) that span from nucleotides +647 to +760, relative to the transcription start site, and which contain consensus sequences for GATA and interferon regulatory factor (IRF) transcription factors. Gel mobility shift and chromatin immunoprecipitation assays corroborated this last finding by documenting binding of GATA-4 and IRF-1 and IRF-2 to the first intron sequence. Moreover, a short sequence encompassing the three footprints was found to inhibit expression of transgenic constructs containing the COL1A2 proximal promoter and far upstream enhancer in a position-independent manner. Mutations inserted into each of the footprints restored transgenic expression to different extents. These results therefore indicated that the unique open chromatin site of COL1A2 corresponds to a repressor, the activity of which seems to be mediated by the concerted action of GATA and IRF proteins. More generally, the study reiterated the existence of species-specific difference in the regulatory networks of the mammalian alpha2(I) collagen coding genes.
Notes:
 
PMID 
Yutaka Inagaki, Miwa Kushida, Kiyoshi Higashi, Johbu Itoh, Reiichi Higashiyama, Yun Yu Hong, Norifumi Kawada, Kazuhiko Namikawa, Hiroshi Kiyama, George Bou-Gharios, Tetsu Watanabe, Isao Okazaki, Kazuo Ikeda (2005)  Cell type-specific intervention of transforming growth factor beta/Smad signaling suppresses collagen gene expression and hepatic fibrosis in mice.   Gastroenterology 129: 1. 259-268 Jul  
Abstract: BACKGROUND & AIMS: Transforming growth factor beta and its intracellular mediators, Smad proteins, play important roles in stimulating collagen gene transcription and, thus, could be the targets for treating hepatic fibrosis. However, intervention of transforming growth factor beta/Smad signaling affects physiological signal transduction as well and may cause serious adverse effects on clinical application. Here we have attempted to suppress hepatic fibrosis by expressing a transforming growth factor beta/Smad antagonist selectively in collagen-producing cells only in the fibrotic liver. METHODS: Recombinant adenoviruses expressing either green fluorescent protein or a transforming growth factor beta/Smad signal repressor, YB-1, were injected into mice untreated or treated with carbon tetrachloride. Green fluorescent protein expression was analyzed under a confocal laser scanning microscope. Antifibrotic effects of YB-1 overexpression were examined by luciferase assays and histological examination with transgenic reporter mice. RESULTS: When the CAG expression unit was used as a control, green fluorescent protein was strongly expressed in a large number of hepatocytes in both normal and carbon tetrachloride-treated liver. In contrast, green fluorescent protein expression driven by a tissue-specific enhancer of the mouse alpha2(I) collagen gene ( COL1A2 ) was detected in activated hepatic stellate cells in carbon tetrachloride-induced fibrotic liver, but not in untreated normal liver. No green fluorescent protein fluorescence was observed in any other organs when the COL1A2 enhancer was used. Adenovirus-mediated YB-1 expression under the control of the COL1A2 enhancer significantly decreased COL1A2 promoter activity after carbon tetrachloride injection and subsequently suppressed the progression of hepatic fibrosis. CONCLUSIONS: These results validate a new concept of the therapy for hepatic fibrosis to achieve cell type-specific gene expression only in the fibrotic liver, with little damage to other organs.
Notes:
2004
 
DOI   
PMID 
Bari Murtuza, Ken Suzuki, George Bou-Gharios, Jonathan R Beauchamp, Ryszard T Smolenski, Terence A Partridge, Magdi H Yacoub (2004)  Transplantation of skeletal myoblasts secreting an IL-1 inhibitor modulates adverse remodeling in infarcted murine myocardium.   Proc Natl Acad Sci U S A 101: 12. 4216-4221 Mar  
Abstract: After myocardial infarction (MI), adverse remodeling with left ventricular (LV) dilatation is a major determinant of poor outcome. Skeletal myoblast (SkM) implantation improves cardiac function post-MI, although the mechanism is unclear. IL-1 influences post-MI hypertrophy and collagen turnover and is implicated in SkM death after grafting. We hypothesized that SkM expressing secretory IL-1 receptor antagonist (sIL-1ra) at MI border zones would specifically attenuate adverse remodeling and exhibit improved graft cell number. Stable murine male SkM lines (5 x 10(5) cells), expressing or nonexpressing (cont) for sIL-1ra, were implanted into infarct border zones of female nude mice immediately after left coronary artery occlusion. LV ejection fraction (LVEF), end-diastolic diameter, and transmitral peak early/late (E/A) flow velocity ratio were determined by echocardiography. Cardiac myocyte hypertrophy and fibrosis were assessed by morphometry, picrosirius red staining, and hydroxyproline assay. At 3 weeks, cont-SkM-engrafted hearts showed reduced hypertrophy, improved LVEF (55.7 +/- 1.2% vs. MI-only: 40.3 +/- 2.9%), and preserved E/A ratios. sIL-1ra-SkM implantation enhanced these effects (LVEF, 67.0 +/- 2.3%) and significantly attenuated LV dilatation (LV end-diastolic diameter, 4.0 +/- 1.1 mm vs. cont-SkM, 4.5 +/- 1.2 mm vs. MI-only, 4.8 +/- 1.8 mm); this was associated with greater graft numbers, as shown by PCR for male-specific smcy gene. Enzyme zymography showed attenuated matrix metalloproteinase-2 and -9 up-regulation post-MI by either donor SkM type, although infarct-remote zone collagen was reduced only with sIL-1ra-SkM. These results suggest that SkM implantation improves cardiac function post-MI by modulation of adverse remodeling, and that this effect can be significantly enhanced by targeting IL-1 as a key upstream regulator of both adverse remodeling and graft cell death.
Notes:
 
DOI   
PMID 
Shi-wen Xu, Sarah L Howat, Elisabetta A Renzoni, Alan Holmes, Jeremy D Pearson, Michael R Dashwood, George Bou-Gharios, Christopher P Denton, Roland M du Bois, Carol M Black, Andrew Leask, David J Abraham (2004)  Endothelin-1 induces expression of matrix-associated genes in lung fibroblasts through MEK/ERK.   J Biol Chem 279: 22. 23098-23103 May  
Abstract: The endothelins are a family of endothelium-derived peptides that possess a variety of biological activities, including potent vasoconstriction. Endothelin-1 (ET-1) is up-regulated during tissue repair and pulmonary fibrosis. Here, we use genome-wide expression array analysis to show that the addition of ET-1 (100 nm, 4 h) to normal lung fibroblasts directly induces expression of matrix and matrix-associated genes, including the profibrotic protein CCN2 (connective tissue growth factor, or CTGF). ET-1 induces the MEK/ERK MAP kinase pathway in fibroblasts. Blockade of the MEK/ERK kinase pathway with U0126 abrogates the ability of ET-1 to induce expression of matrix and matrix-associated mRNAs and the CCN2 protein. The CCN2 promoter possesses an ET-1 response element, which maps to the previously identified basal control element-1 (BCE-1) site. Our results suggest that ET-1 induces a program of matrix synthesis in lung fibroblasts and that ET-1 may play a key role in connective tissue deposition during wound repair and in pulmonary fibrosis.
Notes:
 
DOI   
PMID 
Markella Ponticos, David Abraham, Catherine Alexakis, Qi-Long Lu, Carol Black, Terence Partridge, George Bou-Gharios (2004)  Col1a2 enhancer regulates collagen activity during development and in adult tissue repair.   Matrix Biol 22: 8. 619-628 Feb  
Abstract: An enhancer region in the type I collagen alpha 2 chain (pro-Col1a2) promoter has been previously identified approximately -17 kb away from the transcription start site. This upstream region termed the far-upstream-enhancer contains three DNAse I hypersensitive sites and has been shown to be conserved between mouse and human genes. In this study, we used transgenic mice harbouring the complete promotor sequence of the pro-Col1a2 gene up to -17 kb to examine the role of this enhancer in the expression and regulation of the collagen gene during development and in adult tissues pre and post injury. By careful histological mapping of the collagen type I endogenous gene distribution with that of the transgene driven by the mouse far upstream enhancer, we are able to show that in early days of collagen expression, E8.5-9.5, the endogenous gene preceded transgene expression. However, by E11.5 the overall pattern becomes synchronous with a few exceptions. In adult tissue, both endogenous and transgene expression are attenuated and both are reactivated in parallel in various organs by physical injury or fibrogenic cytokine injection. These findings suggest that the enhancer is central to the activation of the collagen type I and that mice harbouring this enhancer/reporter provide a useful model to follow collagen gene transcription activity and for investigating cellular activity in tissue fibrosis.
Notes:
 
DOI   
PMID 
Shizuko Tanaka, Taras T Antoniv, Ke Liu, Lu Wang, Dominic J Wells, Francesco Ramirez, George Bou-Gharios (2004)  Cooperativity between far upstream enhancer and proximal promoter elements of the human {alpha}2(I) collagen (COL1A2) gene instructs tissue specificity in transgenic mice.   J Biol Chem 279: 53. 56024-56031 Dec  
Abstract: Interaction between the proximal (-378) promoter and the far upstream (-20 kb) enhancer is essential for tissue-specific expression of the human alpha2(I) collagen gene (COL1A2) in transgenic mice. Previous in vitro studies have shown that three Sp1 binding sites (around -300) are part of a cytokine-responsive element and that two TC-rich boxes (around -160 and -125) and a CBF/NFY consensus sequence (around -80) confer optimal promoter activity by interacting among themselves and with the upstream Sp1 sites. Here we report that mutations of the Sp1 binding sites, TC-rich boxes or CBF/NFY consensus sequence lead to reduced transgene activity, thus underscoring the functional interdependence of the proximal promoter elements. Loss of the Sp1 binding sites was associated with loss of transgene expression in osteoblasts, whereas elimination of the CBF/NFY binding site (alone or in combination with the TC-rich boxes) was correlated with a lack of activity in the ventral fascia and head dermis and musculature. Additionally, transgene expression in skin fascia fibroblasts depended on the integrity of the Sp1 binding sites and TC-rich boxes, and on their physical configuration. Evidence is also presented suggesting cooperativity between cis-acting elements of the far upstream enhancer and proximal promoter in assembling tissue-specific protein complexes. This study thus reiterates the complex and highly combinatorial nature of the regulatory network governing COL1A2 transcription in vivo.
Notes:
 
DOI   
PMID 
Xu Shi-Wen, Yunliang Chen, Christopher P Denton, Mark Eastwood, Elisabetta A Renzoni, George Bou-Gharios, Jeremy D Pearson, Michael Dashwood, Roland M du Bois, Carol M Black, Andrew Leask, David J Abraham (2004)  Endothelin-1 promotes myofibroblast induction through the ETA receptor via a rac/phosphoinositide 3-kinase/Akt-dependent pathway and is essential for the enhanced contractile phenotype of fibrotic fibroblasts.   Mol Biol Cell 15: 6. 2707-2719 Jun  
Abstract: The endothelins are a family of endothelium-derived peptides that possess a variety of functions, including vasoconstriction. Endothelin-1 (ET-1) is up-regulated during tissue repair and promotes myofibroblast contraction and migration, hence contributing to matrix remodeling during tissue repair. Here, we show that addition of ET-1 to normal lung fibroblasts induces expression of proteins that contribute to a contractile phenotype, including alpha-smooth muscle actin (alpha-SMA), ezrin, moesin, and paxillin. We confirm that ET-1 enhances the ability of lung fibroblasts to contract extracellular matrix, a function essential for tissue repair, through induction of de novo protein synthesis. Blockade of the Akt/phosphoinositide 3-kinase (PI3-kinase) pathway with LY294002 and wortmannin prevents the ability of ET-1 to induce alpha-SMA, ezrin, paxillin, and moesin and to promote matrix contraction. Dominant negative rac and Akt blocked the ability of ET-1 to promote formation of alpha-SMA stress fibers. Using specific ET-1 receptor inhibitors, we show that ET-1 induces collagen matrix contraction through the ETA, but not the ETB, receptor. Relative to normal pulmonary fibroblasts, fibroblasts cultured from scars of patients with the fibrotic disease systemic sclerosis (scleroderma) show enhanced ET-1 expression and binding. Systemic sclerosis lung fibroblasts show increased ability to contract a collagen matrix and elevated expression of the procontractile proteins alpha-SMA, ezrin, paxillin, and moesin, which are greatly reduced by antagonizing endogenous ET-1 signaling. Thus, blocking ET-1 or the PI3-kinase/Akt cascades might be beneficial in reducing scar formation in pulmonary fibrosis.
Notes:
 
DOI   
PMID 
Elisabetta A Renzoni, David J Abraham, Sarah Howat, Xu Shi-Wen, Piersante Sestini, George Bou-Gharios, Athol U Wells, Srihari Veeraraghavan, Andrew G Nicholson, Christopher P Denton, Andrew Leask, Jeremy D Pearson, Carol M Black, Kenneth I Welsh, Roland M du Bois (2004)  Gene expression profiling reveals novel TGFbeta targets in adult lung fibroblasts.   Respir Res 5: 11  
Abstract: BACKGROUND: Transforming growth factor beta (TGFbeta), a multifunctional cytokine, plays a crucial role in the accumulation of extracellular matrix components in lung fibrosis, where lung fibroblasts are considered to play a major role. Even though the effects of TGFbeta on the gene expression of several proteins have been investigated in several lung fibroblast cell lines, the global pattern of response to this cytokine in adult lung fibroblasts is still unknown. METHODS: We used Affymetrix oligonucleotide microarrays U95v2, containing approximately 12,000 human genes, to study the transcriptional profile in response to a four hour treatment with TGFbeta in control lung fibroblasts and in fibroblasts from patients with idiopathic and scleroderma-associated pulmonary fibrosis. A combination of the Affymetrix change algorithm (Microarray Suite 5) and of analysis of variance models was used to identify TGFbeta-regulated genes. Additional criteria were an average up- or down- regulation of at least two fold. RESULTS: Exposure of fibroblasts to TGFbeta had a profound impact on gene expression, resulting in regulation of 129 transcripts. We focused on genes not previously found to be regulated by TGFbeta in lung fibroblasts or other cell types, including nuclear co-repressor 2, SMAD specific E3 ubiquitin protein ligase 2 (SMURF2), bone morphogenetic protein 4, and angiotensin II receptor type 1 (AGTR1), and confirmed the microarray results by real time-PCR. Western Blotting confirmed induction at the protein level of AGTR1, the most highly induced gene in both control and fibrotic lung fibroblasts among genes encoding for signal transduction molecules.Upregulation of AGTR1 occurred through the MKK1/MKK2 signalling pathway. Immunohistochemical staining showed AGTR1 expression by lung fibroblasts in fibroblastic foci within biopsies of idiopathic pulmonary fibrosis. CONCLUSIONS: This study identifies several novel TGFbeta targets in lung fibroblasts, and confirms with independent methods the induction of angiotensin II receptor type 1, underlining a potential role for angiotensin II receptor 1 antagonism in the treatment of lung fibrosis.
Notes:
 
DOI   
PMID 
George Bou-Gharios, Markella Ponticos, Vineeth Rajkumar, David Abraham (2004)  Extra-cellular matrix in vascular networks.   Cell Prolif 37: 3. 207-220 Jun  
Abstract: The vascular network is a series of linked conduits of blood vessels composed of the endothelium, a monolayer of cells that adorn the vessel lumen and surrounding layer(s) of mesenchymal cells (vascular smooth muscle, pericytes and fibroblasts). In addition to providing structural support, the mesenchymal cells are essential for vessel contractility. The extracellular matrix is a major constituent of blood vessels and provides a framework in which these various cell types are attached and embedded. The composition and organization of vascular extracellular matrix is primarily controlled by the mesenchymal cells, and is also responsible for the mechanical properties of the vessel wall, forming complex networks of structural proteins which are highly regulated. The extracellular matrix also plays a central role in cellular adhesion, differentiation and proliferation. This review examines the cellular and extracellular matrix components of vessels, with specific emphasis on the regulation of collagen type I and implications in vascular disease.
Notes:
 
DOI   
PMID 
Markella Ponticos, Terrence Partridge, Carol M Black, David J Abraham, George Bou-Gharios (2004)  Regulation of collagen type I in vascular smooth muscle cells by competition between Nkx2.5 and deltaEF1/ZEB1.   Mol Cell Biol 24: 14. 6151-6161 Jul  
Abstract: A major component of the vessel wall of large arteries and veins is the extracellular matrix (ECM), which consists of collagens, elastin, and proteoglycans. Collagen type I is one of the most abundant of the ECM proteins. We have previously shown that the pro-collagen type I alpha 2 gene contains an enhancer which confers tissue-specific expression in the majority of collagen-producing cells, including blood vessels. In this paper, we delineate a specific vascular smooth muscle cell (vSMC) element: a 100-bp sequence around -16.6 kb upstream of the transcription start site that regulates collagen expression exclusively in vSMCs. Furthermore, we show that the expression is activated through the binding of the homeodomain protein Nkx2.5, which is further potentiated in the presence of GATA6. In contrast, this element was repressed by the binding of the zinc-finger protein deltaEF1/ZEB1. We propose a model of regulation where the activating transcription factor Nkx2.5 and the repressor deltaEF1/ZEB1 compete for an overlapping DNA binding site. This element is important in understanding the molecular mechanisms of vessel remodeling and is a potential target for intervention in vascular diseases where there is excessive deposition of collagen in the vessel wall.
Notes:
2003
 
DOI   
PMID 
Q L Lu, G Bou-Gharios, T A Partridge (2003)  Non-viral gene delivery in skeletal muscle: a protein factory.   Gene Ther 10: 2. 131-142 Jan  
Abstract: Ever since the publication of the first reports in 1990 using skeletal muscle as a direct target for expressing foreign transgenes, an avalanche of papers has identified a variety of proteins that can be synthesized and correctly processed by skeletal muscle. The impetus to the development of such applications is not only amelioration of muscle diseases, but also a range of therapeutic applications, from immunization to delivery of therapeutic proteins, such as clotting factors and hormones. Although the most efficient way of introducing transgenes into muscle fibres has been by a variety of recombinant viral vectors, there are potential benefits in the use of non-viral vectors. In this review we assess the recent advances in construction and delivery of naked plasmid DNA to skeletal muscle and highlight the options available for further improvements to raise efficiency to therapeutic levels.
Notes:
 
DOI   
PMID 
Qi Long Lu, Christopher J Mann, Fang Lou, George Bou-Gharios, Glenn E Morris, Shao-an Xue, Sue Fletcher, Terence A Partridge, Stephen D Wilton (2003)  Functional amounts of dystrophin produced by skipping the mutated exon in the mdx dystrophic mouse.   Nat Med 9: 8. 1009-1014 Aug  
Abstract: As a target for gene therapy, Duchenne muscular dystrophy (DMD) presents many obstacles but also an unparalleled prospect for correction by alternative splicing. The majority of mutations in the dystrophin gene occur in the region encoding the spectrin-like central rod domain, which is largely dispensable. Thus, splicing around mutations can generate a shortened but in-frame transcript, permitting translation of a partially functional dystrophin protein. We have tested this idea in vivo in the mdx dystrophic mouse (carrying a mutation in exon 23 of the dystrophin gene) by combining a potent transfection protocol with a 2-O-methylated phosphorothioated antisense oligoribonucleotide (2OMeAO) designed to promote skipping of the mutated exon*. The treated mice show persistent production of dystrophin at normal levels in large numbers of muscle fibers and show functional improvement of the treated muscle. Repeated administration enhances dystrophin expression without eliciting immune responses. Our data establishes the realistic practicality of an approach that is applicable, in principle, to a majority of cases of severe dystrophinopathy.
Notes:
2002
 
DOI   
PMID 
Sarah De Val, Markella Ponticos, Taras T Antoniv, Dominic J Wells, David Abraham, Terence Partridge, George Bou-Gharios (2002)  Identification of the key regions within the mouse pro-alpha 2(I) collagen gene far-upstream enhancer.   J Biol Chem 277: 11. 9286-9292 Mar  
Abstract: Studies using transgenic mice have shown that the mouse pro-alpha2(I) collagen gene contains a far-upstream enhancer, which directs expression in the majority of collagen I-producing cells during development and in response to tissue injury. In this study, we have investigated the minimal functional region required for the enhancer effect and studied the role of the three hypersensitive sites (HS3-HS5) that overlap this region. The results of deletion experiments indicate that the minimal functional unit of this enhancer is a 1.5-kb region between -17.0 and -15.45 kb from the transcription start site. This region includes the core sequences of HS3 and HS4 but not HS5. The HS4 sequences are essential for the functional integrity of the enhancer, whereas HS3 represents tissue-specific elements that direct expression in mesenchymal cells of internal tissues and body wall muscles. The HS3 region appears to bind a complex of transcription factors illustrated by large regions of protected sequences. A 400-bp sequence located between -17.0 and -16.6 is also essential for the enhancer because its deletion results in increased susceptibility to the chromatin environment.
Notes:
 
PMID 
Dolores Vernet, Monica G Ferrini, Eliane G Valente, Thomas R Magee, George Bou-Gharios, Jacob Rajfer, Nestor F Gonzalez-Cadavid (2002)  Effect of nitric oxide on the differentiation of fibroblasts into myofibroblasts in the Peyronie's fibrotic plaque and in its rat model.   Nitric Oxide 7: 4. 262-276 Dec  
Abstract: The myofibroblast shares phenotypic features of both fibroblasts and smooth muscle cells. It plays a critical role in collagen deposition and wound healing and disappears by apoptosis when the wound is closed. Its abnormal persistence leads to hypertrophic scar formation and other fibrotic conditions. Myofibroblasts are present in the fibrotic plaque of the tunica albuginea (TA) of the penis in men with Peyronie's disease (PD), a localized fibrosis that is accompanied by a spontaneous induction of the inducible nitric oxide synthase (iNOS), also observed in the TGFbeta1-elicited, PD-like lesion in the rat model. iNOS expression counteracts fibrosis, by producing nitric oxide (NO) that reduces collagen deposition in part by neutralization of profibrotic reactive oxygen species. In this study we investigated whether fibroblast differentiation into myofibroblasts is enhanced in the human and rat PD-like plaque and in cultures of human tissue fibroblasts. We also examined whether NO reduces this cell differentiation and collagen synthesis. The myofibroblast content in the fibroblast population was measured by quantitative immunohistochemistry as the ratio between alpha-smooth muscle actin (ASMA; myofibroblast marker) and vimentin (general fibroblast marker) levels. We found that myofibroblast content was considerably increased in the human and TGFbeta1-induced rat plaques as compared to control TA. Inhibition of iNOS activity by chronic administration of L-iminoethyl-L-lysine to rats with TGFbeta1-induced TA lesion increased myofibroblast abundance and collagen I synthesis measured in plaque and TA homogenates from animals injected with a collagen I promoter construct driving the expression of beta-galactosidase. Fibroblast differentiation into myofibroblasts occurred with passage in the cell cultures from the human PD plaque, but was minimal in cultures from the TA. Induction of iNOS in PD and TA cultures with a cytokine cocktail and a NO donor, S-nitroso-N-acetyl penicillamine (SNAP), was detected by immunohistochemistry. Both treatments reduced the total number of cells and the number of ASMA positive cells, whereas only SNAP decreased collagen I immunostaining. These results support the hypotheses that myofibroblasts play a role in the development of the PD plaque and that the antifibrotic effects of NO may be mediated at least in part by the reduction of myofibroblast abundance and lead to a reduction in collagen I synthesis.
Notes:
2001
 
DOI   
PMID 
T T Antoniv, S De Val, D Wells, C P Denton, C Rabe, B de Crombrugghe, F Ramirez, G Bou-Gharios (2001)  Characterization of an evolutionarily conserved far-upstream enhancer in the human alpha 2(I) collagen (COL1A2) gene.   J Biol Chem 276: 24. 21754-21764 Jun  
Abstract: We have examined the chromatin structure around and upstream of the transcriptional start site of the human alpha2(I) collagen (COL1A2) gene. Four strong DNase I-hypersensitive sites (HS2-5) were only detected in fibroblasts, and a weaker one (HS1) was identified in type I collagen-negative cells. Another hypersensitive site potentially involved in COL1A2 silencing was found in intron 1 (HS(In)). HS1 and HS2 were mapped within conserved promoter sequences and at locations comparable to the mouse gene. HS3, HS4, and HS5 were likewise mapped approximately 20 kilobases upstream of COL1A2 at about the same position as the mouse far-upstream enhancer and within a remarkably homologous genomic segment. DNase I footprinting identified twelve areas of nuclease protection in the far-upstream region (FU1-12) and within stretches nearly identical to the mouse sequence. The region containing HS3-5 was found to confer high and tissue-specific expression in transgenic mice to the otherwise minimally active COL1A2 promoter. Characterization of the human element documented functional differences with the mouse counterpart. Enhancer activity substantially decreased without the segment containing FU1-7 and HS5, and inclusion of AluI repeats located 3' of HS3 augmented position-independent expression of the transgene. Hence, subtle differences may characterize the regulation of mammalian alpha2(I) collagen genes by evolutionarily conserved sequences.
Notes:
 
DOI   
PMID 
C P Denton, B Zheng, X Shiwen, Z Zhang, G Bou-Gharios, H Eberspaecher, C M Black, B de Crombrugghe (2001)  Activation of a fibroblast-specific enhancer of the proalpha2(I) collagen gene in tight-skin mice.   Arthritis Rheum 44: 3. 712-722 Mar  
Abstract: OBJECTIVE: Reporter transgenes were introduced into the type 1 tight-skin (Tsk1/+) mouse model of scleroderma to test the hypothesis that fibroblast-specific genetic programs are activated in fibrosis. METHODS: Transgenes harboring upstream fragments of the 5' flanking region of the mouse proalpha2(I) collagen gene (Col1a2), linked to a 400-bp minimal Col1a2 promoter driving an Escherichia coli beta-galactosidase (LacZ) reporter gene, were introduced into Tsk1/+ mice by breeding. Expression of these transgenes, which function as lineage-specific markers of fibroblast differentiation, was compared between the Tsk-LacZ mice and non-Tsk littermates. Responsiveness of these constructs to the profibrotic cytokine, transforming growth factor beta1 (TGFbeta1), was investigated by transient transfection of reporter constructs in tissue-culture cells. RESULTS: There was significant activation of reporter genes harboring the upstream enhancer in Tsk1/+ mice starting from 1 week of age. This was maximal at 6 weeks old (mean +/- SD 237 +/- 24% of non-Tsk controls; P= 0.001). Recombinant TGFbeta1 significantly activated reporter genes regulated by the upstream enhancer in transient transfection, and Tsk-LacZ fibroblasts showed elevated LacZ expression in tissue culture. CONCLUSION: These data suggest that activating signals in Tsk1/+ mice may act via fibroblast-specific regulatory elements within the murine Col1a2 gene. Although TGFbeta has been implicated in the pathogenesis of fibrosis, and reporter genes regulated by the upstream enhancer appear to be TGFbeta responsive in vitro, our results suggest that fibroblast-specific pathways may also be involved.
Notes:
 
DOI   
PMID 
X Shi-Wen, C P Denton, M R Dashwood, A M Holmes, G Bou-Gharios, J D Pearson, C M Black, D J Abraham (2001)  Fibroblast matrix gene expression and connective tissue remodeling: role of endothelin-1.   J Invest Dermatol 116: 3. 417-425 Mar  
Abstract: This study examines endothelin-induced modulation of extracellular matrix synthesis and remodeling by fibroblasts, and its potential role in the pathogenesis of systemic sclerosis (scleroderma). Endothelin-1 promoted fibroblast synthesis of collagen types I and III, but not fibronectin, by a mechanism dependent upon both ETA and ETB receptors. Conversely, endothelin-1 inhibited both protein expression of matrix metalloproteinase 1 and zymographic activity exclusively via ETA receptors. A dual regulatory role for endothelin-1 in transcriptional regulation was suggested by the ability of endothelin-1 to enhance steady-state levels of collagen mRNA and activate the proalpha2(I) collagen (Col1a2) promoter, but in contrast to reduce matrix metalloproteinase 1 transcript expression and suppress transcription of a human matrix metalloproteinase 1 promoter reporter construct in transient transfection assays. Although endothelin-1 significantly enhanced remodeling of three-dimensional collagen lattices populated by normal fibroblasts, this was not observed for lattices populated by systemic sclerosis fibroblasts. Promotion of matrix remodeling was dependent upon ETA receptor expression and was blocked by specific inhibitors of tyrosine kinases or protein kinase C. Reverse transcriptase polymerase chain reaction, S1 nuclease, and functional cell surface binding studies showed that normal and systemic sclerosis fibroblasts express both ETA and ETB receptors (predominantly ETA), but that ETA receptor mRNA levels and ETA binding sites on fibroblasts cultured from systemic sclerosis skin biopsies are reduced by almost 50%. Endothelin-1 is thus able to induce a fibrogenic phenotype in normal fibroblasts that is similar to that of lesional systemic sclerosis fibroblasts. Moreover, reduced responsiveness to exogenous endothelin-1 in systemic sclerosis suggests that downstream pathways may have already been activated in vivo. These data further implicate dysregulated endothelin-receptor pathways in fibroblasts in the pathogenesis of connective tissue fibrosis.
Notes:
2000
 
PMID 
J Morrison, Q L Lu, C Pastoret, T Partridge, G Bou-Gharios (2000)  T-cell-dependent fibrosis in the mdx dystrophic mouse.   Lab Invest 80: 6. 881-891 Jun  
Abstract: In Duchenne muscular dystrophy patients, the pathological hallmark of the disease, namely, the chronic accumulation of sclerotic scar tissue in the interstitial space of skeletal muscle is attributed to manifestation of secondary pathological processes. Such anomalous generation of matrix protein is thought to be driven by the continuous degeneration and regeneration of muscle both in Duchenne Muscular Dystrophy and in the mdx mouse homolog. We examined mdx and the control strain C57bl/10 mice over a range of ages with respect to the amounts of collagen present in muscles and other organs, finding that the mdx have significantly higher collagen content at later time points in their kidney and lung as well as their muscles. Surprisingly, when we bred the mdx mice on the nu/nu background, the time course of fibrogenesis was modified depending on the tissue and the collagen content was significantly different in age-matched mice. Transplantation of normal thymic tissue into the mdx-nu/nu mice replenished their T-cells and concomitantly altered the collagen content in their tissues to levels comparable with those in immunocompetent mdx mice. This suggests that T-cells play a role in the onset of the fibrotic events that undermines the ability of dystrophic muscle to regenerate.
Notes:
1999
 
DOI   
PMID 
G Bou-Gharios, D J Wells, Q L Lu, J E Morgan, T Partridge (1999)  Differential expression and secretion of alpha 1 anti-trypsin between direct DNA injection and implantation of transfected myoblast.   Gene Ther 6: 6. 1021-1029 Jun  
Abstract: Muscle can be used for systemic delivery of non-muscle proteins. In order to investigate the relative effectiveness of direct DNA plasmid injection versus implantation of genetically modified myogenic cell lines, we have used the human alpha 1 anti-trypsin (alpha1AT) cDNA driven by either cytomegalovirus (CMV) or the muscle creatine kinase 3.3 kb (MCK) promoter in immunodeficient mice. We demonstrate that the implantation of transfected myoblasts stably expressing the human alpha1AT cDNA generates a more persistent production of alpha1AT than does direct intramuscular injection of the same construct as plasmid DNA. Moreover, immunohistological labelling of muscle sections implanted with myoblasts show that the newly formed muscle fibres are those containing the human protein.
Notes:
 
PMID 
J G Gross, G Bou-Gharios, J E Morgan (1999)  Potentiation of myoblast transplantation by host muscle irradiation is dependent on the rate of radiation delivery.   Cell Tissue Res 298: 2. 371-375 Nov  
Abstract: Transplantation of muscle precursor cells (mpc) has been suggested as a treatment for myopathies, such as Duchenne muscular dystrophy. Irradiation of skeletal muscle with 16-20 Gy prevents muscle regeneration and also augments muscle formation from implanted muscle precursor cells (mpc). However, when mdx nu/nu mouse muscles are preirradiated at 0.73 Gy/min rather than at 1.29 Gy/min prior to their injection with normal mpc, significantly more muscle fibres of donor origin are formed. This suggests that the rate at which irradiation is delivered has a physiological effect on the muscle. Although it would not be feasible to irradiate a patient's muscles prior to mpc implantation, once the factor(s) which are altered in irradiated muscle have been identified, it might be possible to use these to increase the success of myoblast transplantation.
Notes:
1998
 
DOI   
PMID 
Y Inagaki, S Truter, G Bou-Gharios, L A Garrett, B de Crombrugghe, T Nemoto, P Greenwel (1998)  Activation of Proalpha2(I) collagen promoter during hepatic fibrogenesis in transgenic mice.   Biochem Biophys Res Commun 250: 3. 606-611 Sep  
Abstract: We previously identified the promoter sequence that is essential for basal and TGF-beta-stimulated transcription of alpha2(I) collagen gene (COL1A2). In the present study, we examined whether the promoter is activated during hepatic fibrogenesis by utilizing transgenic mice harboring the COL1A2 upstream sequence. Intraperitoneal CCl4 administration activated the -17 kb COL1A2 promoter more than 10-fold, whereas partial hepatectomy resulted in no significant change in the promoter activity. The non-parenchymal cell fraction, but not parenchymal hepatocytes, isolated from mice harboring the -313 COL1A2 promoter linked to a beta-galactosidase reporter gene contained large amounts of beta-galactosidase and endogenous COL1A2 mRNAs. beta-galactosidase activity in the cells from CCl4-treated mice was significantly higher than in those from untreated animals. These results indicated that different molecular mechanisms control COL1A2 transcription in CCl4-induced liver injury/fibrosis and physiological regeneration after partial hepatectomy, and that the -313 COL1A2 promoter is activated in a cell type-specific manner during hepatic fibrogenesis.
Notes:
1997
 
DOI   
PMID 
X Shi-Wen, C P Denton, A McWhirter, G Bou-Gharios, D J Abraham, R M du Bois, C M Black (1997)  Scleroderma lung fibroblasts exhibit elevated and dysregulated type I collagen biosynthesis.   Arthritis Rheum 40: 7. 1237-1244 Jul  
Abstract: OBJECTIVE: To examine whether scleroderma lung fibroblasts show a pattern of aberrant type I collagen (CI) biosynthesis similar to that observed previously in studies of dermal fibroblasts in this disease. METHODS: CI secretion and steady-state pro alpha1(I) collagen messenger RNA (mRNA) levels and COL1A2 gene activation were examined in fibroblasts grown from lung biopsy specimens obtained from 16 scleroderma patients with lung fibrosis and from 10 histologically normal lung specimens (controls). The effect of culture in a 3-dimensional (3-D) CI gel matrix culture on CI mRNA levels was also examined. RESULTS: The mean (+/- SEM) collagen secretion in monolayer culture for scleroderma lung fibroblasts was 90.9 +/- 56 ng/ml/10(6) cells, significantly greater (P < 0.05) than controls (40.2 +/- 17.5). Pro alpha1(I) collagen mRNA levels in monolayer cultures were higher in scleroderma (mean +/- SEM collagen:GAPDH ratio 3.7 +/- 0.9) compared with control (1.9 +/- 0.8) lung fibroblasts. Transient expression assays confirmed that genes coding for CI are transcriptionally activated in scleroderma lung fibroblasts compared with control strains. Although all lung fibroblasts induced equivalent contraction of 3-D CI gel matrices, scleroderma strains failed to show a reduction in steady-state pro alpha1(I) collagen mRNA levels in gel culture. CONCLUSION: We have demonstrated elevated CI biosynthesis and impaired mRNA down-regulation for CI by scleroderma lung fibroblasts. These properties are likely to be highly relevant to the pathogenesis of scleroderma-associated lung fibrosis.
Notes:
1996
 
PMID 
G Bou-Gharios, L A Garrett, J Rossert, K Niederreither, H Eberspaecher, C Smith, C Black, B Crombrugghe (1996)  A potent far-upstream enhancer in the mouse pro alpha 2(I) collagen gene regulates expression of reporter genes in transgenic mice.   J Cell Biol 134: 5. 1333-1344 Sep  
Abstract: We have identified three DNase I-hypersensitive sites in chromatin between 15 and 17 kb upstream of the mouse pro alpha 2 (I) collagen gene. These sites were detected in cells that produce type I collagen but not in cells that do not express these genes. A construction containing the sequences from -17 kb to +54 bp of the mouse pro alpha 2 (I) collagen gene, cloned upstream of either the Escherichia coli beta-galactosidase or the firefly luciferase reporter gene, showed strong enhancer activity in transgenic mice when compared with the levels seen previously in animals harboring shorter promoter fragments. Especially high levels of expression of the reporter gene were seen in dermis, fascia, and the fibrous layers of many internal organs. High levels of expression could also be detected in some osteoblastic cells. When various fragments of the 5' flanking sequences were cloned upstream of the 350-bp proximal pro alpha 2(I) collagen promoter linked to the lacZ gene, the cis-acting elements responsible for enhancement were localized in the region between -13.5 and -19.5 kb, the same region that contains the three DNase I-hypersensitive sites. Moreover, the DNA segment from -13.5 to -19.5 kb was also able to drive the cell-specific expression of a 220-bp mouse pro alpha 1(I) collagen promoter, which is silent in transgenic mice. Hence, our data suggest that a far-upstream enhancer element plays a role in regulating high levels of expression of the mouse pro alpha 2(I) collagen gene.
Notes:
1995
 
PMID 
G Bou-Gharios, J Osman, A Atherton, P Monoghan, R Vancheeswaran, C Black, I Olsen (1995)  Expression of ectopeptidases in scleroderma.   Ann Rheum Dis 54: 2. 111-116 Feb  
Abstract: OBJECTIVES--To examine the expression and concentrations of three ectopeptidases likely to be involved in regulating the functional levels of adhesion molecules and the turnover of connective tissue components, in patients with scleroderma (systemic sclerosis) (SSc) and in normal individuals. METHODS--Monoclonal antibodies against these antigens were used for immunoperoxidase staining of cryostat skin sections and for flow cytometric (fluorescence activated cell sorter) analysis of cultured dermal fibroblasts grown from SSc patients and normal controls. RESULTS--Although neutral endopeptidase-24.11 (NEP) (CD10) was not detected in either SSc or normal skin, aminopeptidase N (APN) (CD13) and dipeptidyl peptidase IV (DPPIV) (CD26) were both readily visualised. However, DPPIV appeared to be present in smaller concentrations in the SSc biopsy specimens. Moreover, while fibroblasts grown in vitro from both SSc and normal skin also had similar concentrations of APN, the expression of DPPIV in the cultured SSc cells was found to be very much less than that present in the normal fibroblasts. It is noteworthy that NEP, which was not detected in the tissue sections, was nevertheless readily detected in fibroblasts in culture. CONCLUSIONS--These results show that a number of cell surface proteases are expressed by dermal fibroblasts both in vivo and in vitro, and it is suggested that the marked downregulation of DPPIV in SSc could be at least partly responsible for the increased concentrations of adhesion molecules and matrix proteins associated with the molecular pathology of this disease.
Notes:
 
PMID 
S Xu, R Vancheeswaran, G Bou-Gharios, M J O'Hare, I Olsen, D Abraham, C Black (1995)  Scleroderma-derived human fibroblasts retain abnormal phenotypic and functional characteristics following retroviral transduction with the SV40 tsT antigen.   Exp Cell Res 220: 2. 407-414 Oct  
Abstract: In this study an amphotropic retrovirus has been used to efficiently transduce normal human (NF) and scleroderma (systemic sclerosis; SSc) dermal fibroblasts (SScF) with a sequence encoding a temperature-sensitive mutant of the SV40 large T antigen (tsA58-U19). From the primary outgrowths of skin explants, cultures were generated whose growth was stringently temperature-dependent. When grown at a low, permissive temperature (35 degrees C), both normal and SSc-transduced cells continuously divided with similar doubling times, whereas at a high, nonpermissive temperature (39.5 degrees C), division of both the NF and SScF cells was rapidly arrested. These cells have been passaged more than 50 times, have the typical morphological appearance of fibroblasts, and have retained an anchorage-dependent phenotype. The transduced normal cells (tsT-NF) synthesized the matrix molecules collagen and fibronectin and expressed phenotypic antigens characteristic of their nontransduced counterparts, including MHC Class I, VLA beta 1 (CD29), Hermes 1 (CD44), VLA-4 alpha (CD49d), ICAM-1 (CD54) and LFA-3 (CD58) and the cell surface ectoenzymes neutral endopeptidase (CD10), aminopeptidase N (CD13), and dipeptidyl peptidase IV (CD26). Analysis of the transduced SSc fibroblasts (tsT-SScF) showed that these cells exhibited certain major features of the SSc pathology, notably the abnormally high synthesis of type I collagen, increased expression of ICAM-1, and depressed levels of CD26. Moreover, these phenotypic characteristics were retained even after prolonged culture in vitro. The tsT-SScF cells also retained their responsiveness to cytokines, since interferon-gamma (IFN-gamma) and tumor necrosis factor-alpha (TNF-alpha) both produced a marked increase in ICAM-1 expression. Our findings show that infection of SScF with the SV40 tsT antigen extends the life span of these cells and does not ablate their abnormal phenotypic and functional characteristics.
Notes:
Powered by publicationslist.org.