Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Inherit Metab Dis ; 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38487984

RESUMO

Altered activity of specific enzymes in phenylalanine-tyrosine (phe-tyr) metabolism results in incomplete breakdown of various metabolite substrates in this pathway. Increased biofluid concentration and tissue accumulation of the phe-tyr pathway metabolite homogentisic acid (HGA) is central to pathophysiology in the inherited disorder alkaptonuria (AKU). Accumulation of metabolites upstream of HGA, including tyrosine, occurs in patients on nitisinone, a licenced drug for AKU and hereditary tyrosinaemia type 1, which inhibits the enzyme responsible for HGA production. The aim of this study was to investigate the phe-tyr metabolite content of key biofluids and tissues in AKU mice on and off nitisinone to gain new insights into the biodistribution of metabolites in these altered metabolic states. The data show for the first time that HGA is present in bile in AKU (mean [±SD] = 1003[±410] µmol/L; nitisinone-treated AKU mean [±SD] = 45[±23] µmol/L). Biliary tyrosine, 3(4-hydroxyphenyl)pyruvic acid (HPPA) and 3(4-hydroxyphenyl)lactic acid (HPLA) are also increased on nitisinone. Urine was confirmed as the dominant elimination route of HGA in untreated AKU, but with indication of biliary excretion. These data provide new insights into pathways of phe-tyr metabolite biodistribution and metabolism, showing for the first time that hepatobiliary excretion contributes to the total pool of metabolites in this pathway. Our data suggest that biliary elimination of organic acids and other metabolites may play an underappreciated role in disorders of metabolism. We propose that our finding of approximately 3.8 times greater urinary HGA excretion in AKU mice compared with patients is one reason for the lack of extensive tissue ochronosis in the AKU mouse model.

2.
Sci Rep ; 12(1): 16083, 2022 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-36167967

RESUMO

Nitisinone (NIT) produces inevitable but varying degree of tyrosinaemia. However, the understanding of the dynamic adaptive relationships within the tyrosine catabolic pathway has not been investigated fully. The objective of the study was to assess the contribution of protein intake, serum NIT (sNIT) and tyrosine pathway metabolites to nitisinone-induced tyrosinaemia in alkaptonuria (AKU). Samples of serum and 24-h urine collected during SONIA 2 (Suitability Of Nitisinone In Alkaptonuria 2) at months 3 (V2), 12 (V3), 24 (V4), 36 (V5) and 48 (V6) were included in these analyses. Homogentisic acid (HGA), tyrosine (TYR), phenylalanine (PHE), hydroxyphenylpyruvate (HPPA), hydroxyphenyllactate (HPLA) and sNIT were analysed at all time-points in serum and urine. Total body water (TBW) metabolites were derived using 60% body weight. 24-h urine and TBW metabolites were summed to obtain combined values. All statistical analyses were post-hoc. 307 serum and 24-h urine sampling points were analysed. Serum TYR from V2 to V6, ranging from 478 to 1983 µmol/L were stratified (number of sampling points in brackets) into groups < 701 (47), 701-900 (105), 901-1100 (96) and > 1100 (59) µmol/L. The majority of sampling points had values greater than 900 µmol/L. sPHE increased with increasing sTYR (p < 0.001). Tyrosine, HPPA and HPLA in serum and TBW all increased with rising sTYR (p < 0.001), while HPLA/TYR ratio decreased (p < 0.0001). During NIT therapy, adaptive response to minimise TYR formation was demonstrated. Decreased conversion of HPPA to HPLA, relative to TYR, seems to be most influential in determining the degree of tyrosinaemia.


Assuntos
Alcaptonúria , Encefalopatias Metabólicas Congênitas , Tirosinemias , Alcaptonúria/tratamento farmacológico , Cicloexanonas/uso terapêutico , Ácido Homogentísico , Humanos , Nitrobenzoatos/uso terapêutico , Fenilalanina , Fenilpropionatos , Tirosina/metabolismo , Tirosinemias/tratamento farmacológico
3.
Mol Genet Metab ; 2022 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-35680516

RESUMO

BACKGROUND: Adaptations within the phenylalanine (PHE)/tyrosine (TYR) pathway during nitisinone (NIT) are not fully understood. OBJECTIVE: To characterise the temporal changes in metabolic features in NIT-treated patients with alkaptonuria. PATIENTS AND METHODS: Serum (s) and 24-urine (u) homogentisic acid (sHGA, uHGA24), TYR (sTYR, uTYR24), PHE (sPHE, uPHE24), hydroxyphenylpyruvate (sHPPA, uHPPA24), hydroxyphenyllactate (sHPLA, uHPLA24) and sNIT were measured at baseline (V1) and until month 48 (V6) in 69 NIT-treated patients, recommended to reduce protein intake. The 24-h urine urea (uUREA24), creatinine (uCREAT24) and body weight were also measured. Amounts of tyrosine metabolites in total body water (TBW) were derived by multiplying the serum concentrations by 60% body weight, and sum of TBW and urine metabolites resulted in combined values (c). RESULTS: uUREA24 and uCREAT24 decreased between V1 and V6 during NIT, whereas body weight and sNIT increased. Linear regression coefficient between uUREA24 and uCREAT24 was extremely strong (R = 0.84). sPHE, TBWPHE and cPHE24 increased gradually from V1 to V6. A decrease in cTYR24/cPHE24, sTYR/sPHE and TBWTYR/TBWPHE was seen from V2 to V6. Serum, 24-urine and combined TYR, HPPA and HPLA either remained stable or decreased from V2 to V6. DISCUSSION: The gradual increase in PHE suggests adaptation to increasing TYR during NIT therapy. The decrease in protein intake resulted in decreased muscle mass and increased weight gain. CONCLUSION: Progressive adaptation by decreasing PHE conversion to TYR occurs over time during NIT therapy. A low protein diet results in loss of muscle mass but also weight gain suggesting an increase in fat mass.

4.
Mol Genet Metab Rep ; 30: 100846, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35242577

RESUMO

BACKGROUND: Although changes in the tyrosine pathway during nitisinone therapy are known, a complete characterization of the induced tyrosinaemia is lacking to improve disease management. PATIENTS AND METHODS: Our research aims were addressed by 24-h blood sampling. 40 patients with alkaptonuria (AKU), treated with 0, 1, 2, 4 and 8 mg nitisinone daily (n = 8), were studied over four weeks. Serum homogentisic acid (sHGA), tyrosine (sTYR), phenylalanine (sPHE), hydroxyphenylpyruvate (sHPPA), hydroxyphenyllactate (sHPLA) and nitisinone (sNIT) were measured at baseline and after four weeks. RESULTS: sNIT showed a clear dose-proportional response. sTYR increased markedly but with less clear-cut dose responses after nitisinone. Fasting and average 24-h (Cav) sTYR responses were similar. Individual patient sTYR 24-h profiles showed significant fluctuations during nitisinone therapy. At week 4, sTYR, sHPPA and sHPPL all showed dose-related increases compared to V0, with the greatest difference between 1 and 8 mg nitisinone seen for HPLA, while there was no change from V0 in sPHE. sHGA decreased to values around the lower limit of quantitation. DISCUSSION: There was sustained tyrosinaemia after four weeks of nitisinone therapy with significant fluctuations over the day in individual patients. Diet and degree of conversion of HPPA to HPLA may determine extent of nitisinone-induced tyrosinaemia. CONCLUSION: A fasting blood sample is recommended to monitor sTYR during nitisinone therapy Adaptations in HPPA metabolites as well as the inhibition of tyrosine aminotransferase could be contributing factors generating tyrosinaemia during nitisinone therapy.

5.
Osteoarthr Cartil Open ; 2(2): 100044, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32596691

RESUMO

OBJECTIVE: This study investigated the effect of hUC-MSCs on osteoarthritis (OA) progression in a xenogeneic model. DESIGN: Male, 10 week-old C57BL/6 mice underwent sham surgery (n = 15) or partial medial meniscectomy (PMM; n = 76). 5x105 hUC-MSCs (from 3 donors: D1, D2 and D3) were phenotyped via RT-qPCR and immunoprofiling their response to inflammatory stimuli.They were injected into the mouse joints 3 and 6 weeks post-surgery, harvesting joints at 8 and 12 weeks post-surgery, respectively. A no cell 'control' group was also used (n = 29). All knee joints were assessed via micro-computed tomography (µCT) and histology and 10 plasma markers were analysed at 12 weeks. RESULTS: PMM resulted in cartilage loss and osteophyte formation resembling human OA at both time-points. Injection of one donor's hUC-MSCs into the joint significantly reduced the loss of joint space at 12 weeks post-operatively compared with the PMM control.This 'effective' population of MSCs up-regulated the genes, IDO and TSG6, when stimulated with inflammatory cytokines, more than those from the other two donors.No evidence of an inflammatory response to the injected cells in any animals, either histologically or with plasma biomarkers, arose. CONCLUSION: Beneficial change in a PMM joint was seen with only one hUC-MSC population, perhaps indicating that cell therapy is not appropriate for severely osteoarthritic joints. However, none of the implanted cells appeared to elicit an inflammatory response at the time-points studied. The variability of UC donors suggests some populations may be more therapeutic than others and donor characterisation is essential in developing allogeneic cell therapies.

6.
Metabolomics ; 15(5): 81, 2019 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-31104147

RESUMO

The original publication of this article contained an incorrect version that did not include some final reviewers' suggestions, was inadvertently received for production and published. The original article has been corrected.

7.
Metabolomics ; 15(5): 68, 2019 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-31037385

RESUMO

OBJECTIVE: Nitisinone induced hypertyrosinaemia is a concern in patients with Alkaptonuria (AKU). It has been suggested that this may alter neurotransmitter metabolism, specifically dopamine and serotonin. Herein mass spectrometry imaging (MSI) is used for the direct measurement of 2,4-diphenyl-pyranylium tetrafluoroborate (DPP-TFB) derivatives of monoamine neurotransmitters in brain tissue from a murine model of AKU following treatment with nitisinone. METHODS: Metabolite changes were assessed using MSI on DPP-TFB derivatised fresh frozen tissue sections directing analysis towards primary amine neurotransmitters. Matched tail bleed plasma samples were analysed using LC-MS/MS. Eighteen BALB/c mice were included in this study: HGD-/- (n = 6, treated with nitisinone-4 mg/L, in drinking water); HGD-/- (n = 6, no treatment) and HGD+/- (n = 6, no treatment). RESULTS: Ion intensity and distribution of DPP-TFB derivatives in brain tissue for dopamine, 3-methoxytyramine, noradrenaline, tryptophan, serotonin, and glutamate were not significantly different following treatment with nitisinone in HGD -/- mice, and no significant differences were observed between HGD-/- and HGD+/- mice that received no treatment. Tyrosine (10-fold in both comparisons, p = 0.003; [BALB/c HGD-/- (n = 6) and BALB/c HGD+/- (n = 6) (no treatment) vs. BALB/c HGD-/- (n = 6, treated)] and tyramine (25-fold, p = 0.02; 32-fold, p = 0.02) increased significantly following treatment with nitisinone. Plasma tyrosine and homogentisic acid increased (ninefold, p = < 0.0001) and decreased (ninefold, p = 0.004), respectively in HGD-/- mice treated with nitisinone. CONCLUSIONS: Monoamine neurotransmitters in brain tissue from a murine model of AKU did not change following treatment with nitisinone. These findings have significant implications for patients with AKU as they suggest monoamine neurotransmitters are not altered following treatment with nitisinone.


Assuntos
Encéfalo/metabolismo , Modelos Animais de Doenças , Metabolômica , Neurotransmissores/metabolismo , Tirosinemias/metabolismo , Administração Oral , Animais , Encéfalo/diagnóstico por imagem , Cicloexanonas/administração & dosagem , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Nitrobenzoatos/administração & dosagem , Imagem Óptica , Tirosinemias/sangue , Tirosinemias/induzido quimicamente
8.
Osteoarthritis Cartilage ; 24(7): 1263-73, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26853752

RESUMO

OBJECTIVE: Regulation of anabolic and catabolic factors is considered essential in maintaining the homoeostasis of healthy articular cartilage. In this study we investigated the influence of RNA binding proteins (RNABPs) in this process. DESIGN: Using small interfering RNA (siRNA), RNABP expression was knocked down in SW1353 chondrosarcoma cells and human articular chondrocytes. Gene expression and messenger RNA (mRNA) decay of anabolic (SOX9, Aggrecan) and catabolic (matrix metalloproteinase (MMP)13) factors were analysed using reverse transcription quantitative polymerase chain reaction (RT-qPCR). RNA-electromobility shift assays (EMSAs) were used to investigate RNABP interactions with the SOX9 mRNA 3' untranslated region (UTR). Immunohistochemical localisation of MMP13 and the RNABP human antigen R (HuR) was performed in E13.5 and E16.5 mouse embryo sections. RESULTS: SOX9 mRNA, mRNA half-life and protein expression were increased with siRNA targeting the RNABP tristetraprolin (TTP) in both HACs and SW1353s. TTP knockdown also stimulated aggrecan mRNA expression but did not affect its stability. RNA-EMSAs demonstrated that adenine uracil (AU)-rich elements in the SOX9 mRNA 3'UTR interacted with chondrocyte proteins with three specific elements interacting with TTP. HuR knockdown significantly increased MMP13 expression and also regulated the expression of a number of known transcriptional repressors of MMP13. HuR was ubiquitously expressed within mouse embryos yet displayed regional down-regulation within developing skeletal structures. CONCLUSION: This study demonstrates for the first time how RNABPs are able to affect the balance of anabolic and catabolic gene expression in human chondrocytes. The post-transcriptional mechanisms controlled by RNABPs present novel avenues of regulation and potential points of intervention for controlling the expression of SOX9 and MMP13 in chondrocytes.


Assuntos
Condrócitos , Animais , Cartilagem Articular , Células Cultivadas , Expressão Gênica , Regulação da Expressão Gênica , Humanos , Camundongos , Proteínas de Ligação a RNA
9.
Osteoarthritis Cartilage ; 23(4): 616-28, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25545425

RESUMO

OBJECTIVE: Identify gene changes in articular cartilage of the medial tibial plateau (MTP) at 2, 4 and 8 weeks after destabilisation of the medial meniscus (DMM) in mice. Compare our data with previously published datasets to ascertain dysregulated pathways and genes in osteoarthritis (OA). DESIGN: RNA was extracted from the ipsilateral and contralateral MTP cartilage, amplified, labelled and hybridized on Illumina WGv2 microarrays. Results were confirmed by real-time polymerase chain reaction (PCR) for selected genes. RESULTS: Transcriptional analysis and network reconstruction revealed changes in extracellular matrix and cytoskeletal genes induced by DMM. TGFß signalling pathway and complement and coagulation cascade genes were regulated at 2 weeks. Fibronectin (Fn1) is a hub in a reconstructed network at 2 weeks. Regulated genes decrease over time. By 8 weeks fibromodulin (Fmod) and tenascin N (Tnn) are the only dysregulated genes present in the DMM operated knees. Comparison with human and rodent published gene sets identified genes overlapping between our array and eight other studies. CONCLUSIONS: Cartilage contributes a minute percentage to the RNA extracted from the whole joint (<0.2%), yet is sensitive to changes in gene expression post-DMM. The post-DMM transcriptional reprogramming wanes over time dissipating by 8 weeks. Common pathways between published gene sets include focal adhesion, regulation of actin cytoskeleton and TGFß. Common genes include Jagged 1 (Jag1), Tetraspanin 2 (Tspan2), neuroblastoma, suppression of tumourigenicity 1 (Nbl1) and N-myc downstream regulated gene 2 (Ndrg2). The concomitant genes and pathways we identify may warrant further investigation as biomarkers or modulators of OA.


Assuntos
Cartilagem Articular/metabolismo , Meniscos Tibiais/metabolismo , Análise em Microsséries/métodos , Osteoartrite do Joelho/etiologia , Osteoartrite do Joelho/metabolismo , Transdução de Sinais/fisiologia , Transcrição Gênica/fisiologia , Ferimentos e Lesões/complicações , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Cartilagem Articular/patologia , Proteínas de Ciclo Celular , Modelos Animais de Doenças , Proteínas da Matriz Extracelular/metabolismo , Fibromodulina , Fibronectinas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteína Jagged-1 , Masculino , Proteínas de Membrana/metabolismo , Meniscos Tibiais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/metabolismo , Osteoartrite do Joelho/patologia , Proteínas/metabolismo , Proteoglicanas/metabolismo , Proteínas Serrate-Jagged , Transdução de Sinais/genética , Tenascina/metabolismo , Tetraspaninas/metabolismo , Fator de Crescimento Transformador beta/metabolismo
10.
Osteoarthritis Cartilage ; 22(6): 862-8, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24747174

RESUMO

OBJECTIVE: To detect and determine disease severity of osteoarthritis (OA) using a probe activated by matrix metalloproteinase-13 (MMP-13) in vivo in the murine destabilised medial meniscus (DMM) surgical model of OA. DESIGN: We have previously described MMP12ap and MMP13ap, internally quenched fluorescent peptide substrate probes that are activated respectively by MMP-12 and MMP-13. Here we used these probes to follow enzyme activity in vivo in mice knees 4, 6 and 8 weeks following DMM surgery. After in vivo optical imaging, disease severity was determined through traditional histological analysis. The amount of probe activation was analysed for discrimination between DMM, contralateral and sham operated knees, as well as for congruence between activity and histological damage. RESULTS: There was no specific activation of MMP12ap at the time points observed between sham operated and DMM operated, or their respective contralateral joints. The activation of the MMP13ap in the DMM model was highest 6 weeks after surgery, but was only specific compared against sham surgery 8 weeks after surgery (1.5-fold increase). The activation of MMP13ap correlated with histological damage 6 and 8 weeks after surgery, with correlations of 0.484 (P = 0.0032) and 0.478 respectively (P = 0.0049). This correlation dropped to 0.218 (P = 0.011) if all data were considered. CONCLUSION: The current MMP-13 activity probe is suitable for the discrimination between DMM and sham or contralateral knees 8 weeks after surgery, when cartilage loss is typified by the appearance of small fissures up to the tidemark, but not earlier. This activity correlates with the histological damage observed.


Assuntos
Articulação do Joelho/cirurgia , Metaloproteinase 13 da Matriz/metabolismo , Osteoartrite do Joelho/patologia , Animais , Biomarcadores/metabolismo , Biópsia por Agulha , Diagnóstico por Imagem/métodos , Modelos Animais de Doenças , Progressão da Doença , Fluorescência , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Anatômicos , Osteoartrite do Joelho/fisiopatologia , Distribuição Aleatória , Valores de Referência , Sensibilidade e Especificidade , Fatores de Tempo
11.
J Pathol ; 214(5): 627-36, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18266309

RESUMO

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.


Assuntos
Colágeno Tipo I/deficiência , Doenças Fetais/terapia , Células-Tronco Fetais/transplante , Nefropatias/terapia , Glomérulos Renais/ultraestrutura , Transplante de Células-Tronco Mesenquimais/métodos , Animais , Colágeno Tipo I/biossíntese , Colágeno Tipo I/metabolismo , Modelos Animais de Doenças , Feminino , Terapias Fetais/métodos , Sobrevivência de Enxerto , Humanos , Nefropatias/metabolismo , Nefropatias/patologia , Glomérulos Renais/metabolismo , Masculino , Camundongos , Microscopia Eletrônica
12.
J Pathol ; 214(3): 377-86, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18095257

RESUMO

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.


Assuntos
Células da Medula Óssea/patologia , Cicatriz/patologia , Infarto do Miocárdio/patologia , Miocárdio/patologia , Cicatrização , Actinas/análise , Actinas/genética , Animais , Cicatriz/enzimologia , Colágeno/genética , Colágeno/metabolismo , Colágeno Tipo I , Fibroblastos/patologia , Expressão Gênica , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Imuno-Histoquímica , Ligadura , Luciferases/análise , Luciferases/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Animais , Infarto do Miocárdio/enzimologia , Miocárdio/enzimologia , beta-Galactosidase/análise , beta-Galactosidase/genética
13.
Gene Ther ; 10(2): 131-42, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12571642

RESUMO

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.


Assuntos
Terapia Genética/métodos , Músculo Esquelético/metabolismo , Proteínas/genética , Transfecção/métodos , Animais , Biolística , Eletroporação , Expressão Gênica , Engenharia Genética , Vetores Genéticos , Humanos , Lipossomos , Peptídeos , Proteínas/metabolismo , Transcrição Gênica , Transgenes
14.
J Biol Chem ; 276(24): 21754-64, 2001 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-11279244

RESUMO

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.


Assuntos
Colágeno/genética , Elementos Facilitadores Genéticos , Evolução Molecular , Regulação da Expressão Gênica no Desenvolvimento , Regiões Promotoras Genéticas , Animais , Sequência de Bases , Linhagem Celular , Cromatina/genética , Colágeno Tipo I , Sequência Conservada , Cruzamentos Genéticos , DNA/química , Pegada de DNA , Desoxirribonuclease I , Desenvolvimento Embrionário e Fetal , Fibroblastos/metabolismo , Humanos , Pulmão , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Transgênicos , Dados de Sequência Molecular , Mapeamento por Restrição , Alinhamento de Sequência , Homologia de Sequência do Ácido Nucleico
15.
Arthritis Rheum ; 44(3): 712-22, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11263787

RESUMO

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.


Assuntos
Fibroblastos/metabolismo , Pró-Colágeno/genética , Escleroderma Sistêmico/genética , Animais , Modelos Animais de Doenças , Elementos Facilitadores Genéticos/genética , Regulação da Expressão Gênica , Camundongos , Camundongos Transgênicos
16.
J Invest Dermatol ; 116(3): 417-25, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11231316

RESUMO

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.


Assuntos
Tecido Conjuntivo/fisiologia , Endotelina-1/fisiologia , Matriz Extracelular/genética , Fibroblastos/metabolismo , Expressão Gênica/fisiologia , Células Cultivadas , Colágeno/biossíntese , Colágeno/fisiologia , Endotelina-1/farmacologia , Matriz Extracelular/fisiologia , Fibroblastos/fisiologia , Humanos , Metaloproteinase 1 da Matriz/biossíntese , RNA Mensageiro/metabolismo , Receptor de Endotelina A , Receptor de Endotelina B , Receptores de Endotelina/genética , Receptores de Endotelina/fisiologia , Valores de Referência , Escleroderma Sistêmico/metabolismo , Escleroderma Sistêmico/patologia , Transcrição Gênica/efeitos dos fármacos
17.
Lab Invest ; 80(6): 881-91, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10879739

RESUMO

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.


Assuntos
Envelhecimento , Músculo Esquelético/patologia , Distrofia Muscular Animal/imunologia , Distrofia Muscular Animal/patologia , Linfócitos T/imunologia , Animais , Proteínas da Matriz Extracelular/análise , Proteínas da Matriz Extracelular/metabolismo , Fibrose , Hidroxiprolina/análise , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Camundongos Nus , Músculo Esquelético/imunologia , Músculo Esquelético/fisiologia , Especificidade de Órgãos , Regeneração , Linfócitos T/patologia
18.
Cell Tissue Res ; 298(2): 371-5, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10571126

RESUMO

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.


Assuntos
Músculos/efeitos da radiação , Músculos/transplante , Animais , Relação Dose-Resposta à Radiação , Distrofina/análise , Técnicas In Vitro , Camundongos , Camundongos Nus , Músculos/citologia , Regeneração , Transplante de Tecidos/métodos
19.
Gene Ther ; 6(6): 1021-9, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10455404

RESUMO

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.


Assuntos
DNA/fisiologia , Fibras Musculares Esqueléticas/citologia , Transfecção , alfa 1-Antitripsina/metabolismo , Animais , DNA/administração & dosagem , Ensaio de Imunoadsorção Enzimática , Humanos , Injeções , Camundongos , Camundongos Nus , Plasmídeos , Reação em Cadeia da Polimerase/métodos
20.
Biochem Biophys Res Commun ; 250(3): 606-11, 1998 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-9784393

RESUMO

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.


Assuntos
Colágeno/genética , Regulação da Expressão Gênica , Cirrose Hepática Experimental/genética , Regiões Promotoras Genéticas/genética , Precursores de Proteínas/genética , Animais , Colágeno/metabolismo , Camundongos , Camundongos Transgênicos , Precursores de Proteínas/metabolismo , Fator de Crescimento Transformador beta/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...