Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Cell Death Dis ; 15(6): 387, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38824145

RESUMO

Obesity exacerbates tissue degeneration and compromises the integrity and reparative potential of mesenchymal stem/stromal cells (MSCs), but the underlying mechanisms have not been sufficiently elucidated. Mitochondria modulate the viability, plasticity, proliferative capacity, and differentiation potential of MSCs. We hypothesized that alterations in the 5-hydroxymethylcytosine (5hmC) profile of mitochondria-related genes may mediate obesity-driven dysfunction of human adipose-derived MSCs. MSCs were harvested from abdominal subcutaneous fat of obese and age/sex-matched non-obese subjects (n = 5 each). The 5hmC profile and expression of nuclear-encoded mitochondrial genes were examined by hydroxymethylated DNA immunoprecipitation sequencing (h MeDIP-seq) and mRNA-seq, respectively. MSC mitochondrial structure (electron microscopy) and function, metabolomics, proliferation, and neurogenic differentiation were evaluated in vitro, before and after epigenetic modulation. hMeDIP-seq identified 99 peaks of hyper-hydroxymethylation and 150 peaks of hypo-hydroxymethylation in nuclear-encoded mitochondrial genes from Obese- versus Non-obese-MSCs. Integrated hMeDIP-seq/mRNA-seq analysis identified a select group of overlapping (altered levels of both 5hmC and mRNA) nuclear-encoded mitochondrial genes involved in ATP production, redox activity, cell proliferation, migration, fatty acid metabolism, and neuronal development. Furthermore, Obese-MSCs exhibited decreased mitochondrial matrix density, membrane potential, and levels of fatty acid metabolites, increased superoxide production, and impaired neuronal differentiation, which improved with epigenetic modulation. Obesity elicits epigenetic changes in mitochondria-related genes in human adipose-derived MSCs, accompanied by structural and functional changes in their mitochondria and impaired fatty acid metabolism and neurogenic differentiation capacity. These observations may assist in developing novel therapies to preserve the potential of MSCs for tissue repair and regeneration in obese individuals.


Assuntos
Tecido Adiposo , Diferenciação Celular , Epigênese Genética , Células-Tronco Mesenquimais , Mitocôndrias , Obesidade , Humanos , Células-Tronco Mesenquimais/metabolismo , Obesidade/metabolismo , Obesidade/genética , Obesidade/patologia , Mitocôndrias/metabolismo , Tecido Adiposo/metabolismo , Diferenciação Celular/genética , Feminino , Masculino , 5-Metilcitosina/análogos & derivados , 5-Metilcitosina/metabolismo , Adulto , Pessoa de Meia-Idade , Proliferação de Células
2.
Methods Mol Biol ; 2708: 155-174, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37558970

RESUMO

The injection of therapies into the eye is common practice, both clinically and pre-clinically. The most straightforward delivery route is via an intravitreal injection, which introduces the treatment into the largest cavity at the posterior of the eye. This technique is frequently used to deliver gene therapies, including those containing recombinant adeno-associated viral vectors (AAVs), to the back of the eye to enable inner retinal targeting. This chapter provides detailed methodology on how to successfully perform an intravitreal injection in mice. The chapter covers vector preparation considerations, advice on how to minimize vector loss in the injection device, and ways to reduce vector reflux from the eye when administering a therapy. Finally, a protocol is provided on common retinal histology processing techniques to assess vector-mediated expression in retinal ganglion cells. It is hoped that this chapter will enable researchers to carry out effective and consistent intravitreal injections that transduce the inner retinal surface while avoiding common pitfalls.


Assuntos
Retina , Células Ganglionares da Retina , Camundongos , Animais , Injeções Intravítreas , Transdução Genética , Retina/metabolismo , Terapia Genética/métodos , Dependovirus/genética , Vetores Genéticos/genética
3.
Gene Ther ; 30(9): 723-735, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37386155

RESUMO

Adeno-associated virus serotype 2 (AAV2) is a viral vector that can be used to deliver therapeutic genes to diseased cells in the retina. One strategy for altering AAV2 vectors involves the mutation of phosphodegron residues, which are thought to be phosphorylated/ubiquitinated in the cytosol, facilitating degradation of the vector and the inhibition of transduction. As such, mutation of phosphodegron residues have been correlated with increased transduction of target cells, however, an assessment of the immunobiology of wild-type and phosphodegron mutant AAV2 vectors following intravitreal (IVT) delivery to immunocompetent animals is lacking in the current literature. In this study, we show that IVT of a triple phosphodegron mutant AAV2 capsid is associated with higher levels of humoral immune activation, infiltration of CD4 and CD8 T-cells into the retina, generation of splenic germinal centre reactions, activation of conventional dendritic cell subsets, and elevated retinal gliosis compared to wild-type AAV2 capsids. However, we did not detect significant changes in electroretinography arising after vector administration. We also demonstrate that the triple AAV2 mutant capsid is less susceptible to neutralisation by soluble heparan sulphate and anti-AAV2 neutralising antibodies, highlighting a possible utility for the vector in terms of circumventing pre-existing humoral immunity. In summary, the present study highlights novel aspects of rationally-designed vector immunobiology, which may be relevant to their application in preclinical and clinical settings.


Assuntos
Capsídeo , Parvovirinae , Camundongos , Animais , Capsídeo/metabolismo , Sorogrupo , Transdução Genética , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Parvovirinae/genética , Dependovirus/metabolismo , Vetores Genéticos/genética
4.
Gene Ther ; 30(6): 503-519, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36635457

RESUMO

Recombinant adeno-associated viral vectors (AAVs) are an effective system for gene transfer. AAV serotype 2 (AAV2) is commonly used to deliver transgenes to retinal ganglion cells (RGCs) via intravitreal injection. The AAV serotype however is not the only factor contributing to the effectiveness of gene therapies. Promoters influence the strength and cell-selectivity of transgene expression. This study compares five promoters designed to maximise AAV2 cargo space for gene delivery: chicken ß-actin (CBA), cytomegalovirus (CMV), short CMV early enhancer/chicken ß-actin/short ß-globulin intron (sCAG), mouse phosphoglycerate kinase (PGK), and human synapsin (SYN). The promoters driving enhanced green fluorescent protein (eGFP) were examined in adult C57BL/6J mice eyes and tissues of the visual system. eGFP expression was strongest in the retina, optic nerves and brain when driven by the sCAG and SYN promoters. CBA, CMV, and PGK had moderate expression by comparison. The SYN promoter had almost exclusive transgene expression in RGCs. The PGK promoter had predominant expression in both RGCs and AII amacrine cells. The ubiquitous CBA, CMV, and sCAG promoters expressed eGFP in a variety of cell types across multiple retinal layers including Müller glia and astrocytes. We also found that these promoters could transduce human retina ex vivo, although expression was predominantly in glial cells due to low RGC viability. Taken together, this promoter comparison study contributes to optimising AAV-mediated transduction in the retina, and could be valuable for research in ocular disorders, particularly those with large or complex genetic cargos.


Assuntos
Infecções por Citomegalovirus , Parvovirinae , Camundongos , Animais , Humanos , Células Ganglionares da Retina/metabolismo , Actinas/genética , Actinas/metabolismo , Transdução Genética , Camundongos Endogâmicos C57BL , Transgenes , Dependovirus/genética , Dependovirus/metabolismo , Parvovirinae/genética , Proteínas de Fluorescência Verde/genética , Infecções por Citomegalovirus/genética , Infecções por Citomegalovirus/metabolismo , Vetores Genéticos/genética
5.
Exp Eye Res ; 213: 108793, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34656549

RESUMO

Membrane contact sites (MCS) play crucial roles in cell physiology with dysfunction in several MCS proteins being linked with neurological and optic nerve diseases. Although there have been significant advances in imaging these interactions over the past two decades with advanced electron microscopy techniques, super-resolution imaging and proximity-dependent fluorescent reporters, a technique to observe and quantify MCS in mammalian optic nerve tissues has not yet been reported. We demonstrate for the first time that proximity ligation assay (PLA), a technique already used in mammalian cell lines, can be used as an efficient method of quantifying inter-organelle contact sites, namely mitochondria-endoplasmic reticulum (ER) and mitochondria-late-endosomes, in mammalian optic nerve tissues treated with adeno-associated virus (AAV) gene therapy with wild-type or phosphomimetic (active) protrudin. PLA utilises complementary single-stranded DNA oligomers bound to secondary antibodies that hybridise and complete a circular piece of DNA when the primary antibodies of interest interact. These interactions can be detected by amplifying the circular DNA and adding fluorescent probes. We show that PLA is a useful method that can be used to quantify MCS in optic nerve tissues. We have found that upregulation of protrudin with gene therapy significantly increases the number of mitochondria-ER and mitochondria-Rab7-late endosomes contact sites in optic nerves.


Assuntos
Bioensaio/métodos , Retículo Endoplasmático/metabolismo , Endossomos/metabolismo , Membranas Mitocondriais/metabolismo , Nervo Óptico/metabolismo , Animais , Sítios de Ligação , Dependovirus/genética , Feminino , Expressão Gênica , Terapia Genética , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Organelas , Fixação de Tecidos , Proteínas de Transporte Vesicular/genética
6.
Biol Rev Camb Philos Soc ; 96(4): 1616-1644, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33837614

RESUMO

Viral vectors can be utilised to deliver therapeutic genes to diseased cells. Adeno-associated virus (AAV) is a commonly used viral vector that is favoured for its ability to infect a wide range of tissues whilst displaying limited toxicity and immunogenicity. Most humans harbour anti-AAV neutralising antibodies (NAbs) due to subclinical infections by wild-type virus during infancy and these pre-existing NAbs can limit the efficiency of gene transfer depending on the target cell type, route of administration and choice of serotype. Vector administration can also result in de novo NAb synthesis that could limit the opportunity for repeated gene transfer to diseased sites. A number of strategies have been described in preclinical models that could circumvent NAb responses in humans, however, the successful translation of these innovations into the clinical arena has been limited. Here, we provide a comprehensive review of the humoral immune response to AAV gene therapy in the ocular compartment. We cover basic AAV biology and clinical application, the role of pre-existing and induced NAbs, and possible approaches to overcoming antibody responses. We conclude with a framework for a comprehensive strategy for circumventing humoral immune responses to AAV in the future.


Assuntos
Dependovirus , Imunidade Humoral , Dependovirus/genética , Terapia Genética , Vetores Genéticos , Humanos
7.
J Diabetes Res ; 2019: 5140521, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31485452

RESUMO

Diabetic retinopathy (DR) is the commonest cause of blindness in the working-age population of the developed world. The molecular pathophysiology of DR is complex, and a complete spatiotemporal model of the disease is still being elucidated. Recently, a role for angiopoietin (Ang) proteins in the pathophysiology of DR has been proposed by several research groups, and several aspects of Ang signalling are being explored as novel therapeutic strategies. Here, we review the role of the Ang proteins in two important forms of DR, diabetic macular oedema and proliferative diabetic retinopathy. The function of the Ang proteins in regulating blood vessel permeability and neovascularisation is discussed, and we also evaluate recent preclinical and clinical studies highlighting the potential benefits of modulating Ang signalling as a treatment for DR.


Assuntos
Angiopoietinas/fisiologia , Retinopatia Diabética/etiologia , Angiopoietinas/sangue , Animais , Complicações do Diabetes/sangue , Complicações do Diabetes/tratamento farmacológico , Diabetes Mellitus/sangue , Diabetes Mellitus/tratamento farmacológico , Retinopatia Diabética/sangue , Retinopatia Diabética/prevenção & controle , Humanos , Hipoglicemiantes/uso terapêutico , Edema Macular/sangue , Edema Macular/etiologia , Edema Macular/prevenção & controle , Transdução de Sinais/efeitos dos fármacos
8.
Expert Opin Biol Ther ; 18(12): 1257-1270, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30408422

RESUMO

INTRODUCTION: Diabetic retinopathy (DR) is the leading cause of vision loss in the working age population of the developed world. DR encompasses a complex pathology, and one that is reflected in the variety of currently available treatments, which include laser photocoagulation, glucocorticoids, vitrectomy and agents which neutralize vascular endothelial growth factor (VEGF). Whilst these options demonstrate modest clinical benefits, none is yet to fully attenuate clinical progression or reverse damage to the retina. This has led to an interest in developing novel therapies for the condition, such as mediators of angiopoietin signaling axes, immunosuppressants, nonsteroidal anti-inflammatory drugs (NSAIDs), oxidative stress inhibitors and vitriol viscosity inhibitors. Further, preclinical research suggests that gene therapy treatment for DR could provide significant benefits over existing treatments options. AREAS COVERED: Here we review the pathophysiology of DR and provide an overview of currently available treatments. We then outline recent advances made towards improved patient outcomes and highlight the potential of the gene therapy paradigm to revolutionize DR management. EXPERT OPINION: Whilst significant progress has been made towards our understanding of DR, further research is required to enable the development of a detailed spatiotemporal model of the disease. In addition, we hope that improvements in our knowledge of the condition facilitate therapeutic innovations that continue to address unmet medical need and improve patient outcomes, with a focus on the development of targeted medicines.


Assuntos
Retinopatia Diabética/etiologia , Retinopatia Diabética/terapia , Terapias em Estudo , Inibidores da Angiogênese/uso terapêutico , Animais , Retinopatia Diabética/diagnóstico , Terapia Genética/efeitos adversos , Glucocorticoides/uso terapêutico , Humanos , Terapias em Estudo/métodos , Terapias em Estudo/tendências
9.
Cell Death Dis ; 9(10): 1007, 2018 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-30258047

RESUMO

Previous studies have demonstrated that intravitreal delivery of brain-derived neurotrophic factor (BDNF) by injection of recombinant protein or by gene therapy can alleviate retinal ganglion cell (RGC) loss after optic nerve injury. BDNF gene therapy can improve RGC survival in experimental models of glaucoma, the leading cause of irreversible blindness worldwide. However, the therapeutic efficacy of BDNF supplementation alone is time limited at least in part due to BDNF receptor downregulation. Tropomyosin-related receptor kinase-B (TrkB) downregulation has been reported in many neurological diseases including glaucoma, potentially limiting the effect of sustained or repeated BDNF delivery.Here, we characterize a novel adeno-associated virus (AAV) gene therapy (AAV2 TrkB-2A-mBDNF) that not only increases BDNF production but also improves long-term neuroprotective signaling by increasing expression of the BDNF receptor (TrkB) within the inner retina. This approach leads to significant and sustained elevation of survival signaling pathways ERK and AKT within RGCs over 6 months and avoids the receptor downregulation which we observe with treatment with AAV2 BDNF alone. We validate the neuroprotective efficacy of AAV2 TrkB-2A-mBDNF in a mouse model of optic nerve injury, where it outperforms conventional AAV2 BDNF or AAV2 TrkB therapy, before showing powerful proof of concept neuroprotection of RGCs and axons in a rat model of chronic intraocular pressure (IOP) elevation. We also show that there are no adverse effects of the vector on retinal structure or function as assessed by histology and electroretinography in young or aged animals. Further studies are underway to explore the potential of this vector as a candidate for progression into clinical studies to protect RGCs in patients with glaucoma and progressive visual loss despite conventional IOP-lowering treatment.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/genética , Glicoproteínas de Membrana/genética , Neuroproteção/genética , Receptor trkB/genética , Células Ganglionares da Retina/patologia , Transdução de Sinais/genética , Animais , Axônios/patologia , Dependovirus/genética , Modelos Animais de Doenças , Regulação para Baixo/genética , Terapia Genética/métodos , Glaucoma/genética , Glaucoma/patologia , Células HEK293 , Humanos , Pressão Intraocular/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Traumatismos do Nervo Óptico/genética , Traumatismos do Nervo Óptico/patologia , Ratos , Ratos Sprague-Dawley , Retina/patologia
10.
Hum Gene Ther ; 29(7): 828-841, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29466871

RESUMO

Brain-derived neurotrophic factor (BDNF) acting through the tropomyosin-related receptor-B (TrkB) is an important signaling system for the maintenance and survival of neurons. Gene therapy using either recombinant adeno-associated virus (AAV) or lentiviral vectors can provide sustained delivery of BDNF to tissues where reduced BDNF signaling is hypothesized to contribute to disease pathophysiology. However, elevation in BDNF at target sites has been shown to lead to a downregulation of TrkB receptors, thereby reducing the effect of chronic BDNF delivery over time. A novel gene sequence has been designed coding both the ligand (BDNF) and the TrkB receptor in a single transgene separated by a short viral-2A sequence. The single transgene is efficiently processed intracellularly in vitro and in vivo to yield the two mature proteins, which are then independently transported to their final cellular locations: TrkB receptors to the cell surface, and BDNF contained within secretory vesicles. To accommodate the coding sequences of both BDNF and TrkB receptors within the narrow confines of the AAV vectors (4.7 kb pairs), the coding region for the pro-domain of BDNF was removed and the signal peptide sequence modified to improve production, intracellular transport, and secretion of mature BDNF (mBDNF). Intracellular processing and efficacy was shown in HEK293 cells and SH-SY5Y neuroblastoma cells using plasmid DNA and after incorporating the TrkB-2A-mBDNF into an AAV2 vector. Increased BDNF/TrkB-mediated intracellular signaling pathways were observed after AAV2 vector transfection while increased TrkB phosphorylation could be detected in combination with neuroprotection from hydrogen peroxide-induced oxidative stress. Correct processing was also shown in vivo in mouse retinal ganglion cells after AAV2 vector administration to the eye. This novel construct is currently being investigated for its efficacy in animal models to determine its potential to progress to human clinical studies in the future.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/genética , Terapia Genética , Neurônios/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Receptor trkB/genética , Animais , Fator Neurotrófico Derivado do Encéfalo/administração & dosagem , Dependovirus/genética , Células HEK293 , Humanos , Peróxido de Hidrogênio/toxicidade , Ligantes , Glicoproteínas de Membrana/genética , Camundongos , Neurônios/patologia , Estresse Oxidativo/genética , Fosforilação , Sinais Direcionadores de Proteínas/genética , Receptor trkB/administração & dosagem , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/patologia
11.
Stem Cells ; 36(1): 65-78, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29044808

RESUMO

Optic neuropathies such as glaucoma occur when retinal ganglion cells (RGCs) in the eye are injured. Strong evidence suggests mesenchymal stem cells (MSCs) could be a potential therapy to protect RGCs; however, little is known regarding their effect on the human retina. We, therefore, investigated if human MSCs (hMSCs), or platelet-derived growth factor (PDGF) as produced by hMSC, could delay RGC death in a human retinal explant model of optic nerve injury. Our results showed hMSCs and the secreted growth factor PDGF-AB could substantially reduce human RGC loss and apoptosis following axotomy. The neuroprotective pathways AKT, ERK, and STAT3 were activated in the retina shortly after treatments with labeling seen in the RGC layer. A dose dependent protective effect of PDGF-AB was observed in human retinal explants but protection was not as substantial as that achieved by culturing hMSCs on the retina surface which resulted in RGC cell counts similar to those immediately post dissection. These results demonstrate that hMSCs and PDGF have strong neuroprotective action on human RGCs and may offer a translatable, therapeutic strategy to reduce degenerative visual loss. Stem Cells 2018;36:65-78.


Assuntos
Células-Tronco Mesenquimais/metabolismo , Fármacos Neuroprotetores/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Células Ganglionares da Retina/metabolismo , Humanos , Transdução de Sinais
12.
Stem Cells ; 33(10): 3006-16, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26175331

RESUMO

A variety of diseases lead to degeneration of retinal ganglion cells (RGCs) and their axons within the optic nerve resulting in loss of visual function. Although current therapies may delay RGC loss, they do not restore visual function or completely halt disease progression. Regenerative medicine has recently focused on stem cell therapy for both neuroprotective and regenerative purposes. However, significant problems remain to be addressed, such as the long-term impact of reactive gliosis occurring in the host retina in response to transplanted stem cells. The aim of this work was to investigate retinal glial responses to intravitreally transplanted bone marrow mesenchymal stem cells (BM-MSCs) to help identify factors able to modulate graft-induced reactive gliosis. We found in vivo that intravitreal BM-MSC transplantation is associated with gliosis-mediated retinal folding, upregulation of intermediate filaments, and recruitment of macrophages. These responses were accompanied by significant JAK/STAT3 and MAPK (ERK1/2 and JNK) cascade activation in retinal Muller glia. Lipocalin-2 (Lcn-2) was identified as a potential new indicator of graft-induced reactive gliosis. Pharmacological inhibition of STAT3 in BM-MSC cocultured retinal explants successfully reduced glial fibrillary acidic protein expression in retinal Muller glia and increased BM-MSC retinal engraftment. Inhibition of stem cell-induced reactive gliosis is critical for successful transplantation-based strategies for neuroprotection, replacement, and regeneration of the optic nerve.


Assuntos
Gliose/terapia , Transplante de Células-Tronco Mesenquimais , Neuroglia/patologia , Medicina Regenerativa , Animais , Axônios/patologia , Células da Medula Óssea/citologia , Células Ependimogliais/patologia , Gliose/patologia , Humanos , Células-Tronco Mesenquimais , Camundongos , Nervo Óptico/patologia , Retina/crescimento & desenvolvimento , Retina/patologia , Células Ganglionares da Retina/patologia
13.
PLoS One ; 9(1): e87345, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24475278

RESUMO

Interstitial fibrosis, a histological process common to many kidney diseases, is the precursor state to end stage kidney disease, a devastating and costly outcome for the patient and the health system. Fibrosis is historically associated with chronic kidney disease (CKD) but emerging evidence is now linking many forms of acute kidney disease (AKD) with the development of CKD. Indeed, we and others have observed at least some degree of fibrosis in up to 50% of clinically defined cases of AKD. Epithelial cells of the proximal tubule (PTEC) are central in the development of kidney interstitial fibrosis. We combine the novel techniques of laser capture microdissection and multiplex-tandem PCR to identify and quantitate "real time" gene transcription profiles of purified PTEC isolated from human kidney biopsies that describe signaling pathways associated with this pathological fibrotic process. Our results: (i) confirm previous in-vitro and animal model studies; kidney injury molecule-1 is up-regulated in patients with acute tubular injury, inflammation, neutrophil infiltration and a range of chronic disease diagnoses, (ii) provide data to inform treatment; complement component 3 expression correlates with inflammation and acute tubular injury, (iii) identify potential new biomarkers; proline 4-hydroxylase transcription is down-regulated and vimentin is up-regulated across kidney diseases, (iv) describe previously unrecognized feedback mechanisms within PTEC; Smad-3 is down-regulated in many kidney diseases suggesting a possible negative feedback loop for TGF-ß in the disease state, whilst tight junction protein-1 is up-regulated in many kidney diseases, suggesting feedback interactions with vimentin expression. These data demonstrate that the combined techniques of laser capture microdissection and multiplex-tandem PCR have the power to study molecular signaling within single cell populations derived from clinically sourced tissue.


Assuntos
Injúria Renal Aguda/fisiopatologia , Células Epiteliais/fisiologia , Regulação da Expressão Gênica/fisiologia , Túbulos Renais Proximais/citologia , Microdissecção e Captura a Laser/métodos , Reação em Cadeia da Polimerase Multiplex/métodos , Transdução de Sinais/fisiologia , Análise de Variância , Fibrose , Perfilação da Expressão Gênica , Humanos , Imuno-Histoquímica , Túbulos Renais Proximais/fisiopatologia , Reação em Cadeia da Polimerase em Tempo Real
14.
Brain ; 137(Pt 2): 503-19, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24176979

RESUMO

The development of neuroprotective strategies to attenuate retinal ganglion cell death could lead to novel therapies for chronic optic neuropathies such as glaucoma. Intravitreal transplantation of mesenchymal stem cells slows retinal ganglion cell death in models of optic nerve injury, but the mechanism of action remains unclear. Here we characterized the neuroprotective effects of mesenchymal stem cells and mesenchymal stem cell-derived factors in organotypic retinal explant culture and an in vivo model of ocular hypertensive glaucoma. Co-culture of rat and human bone marrow-derived mesenchymal stem cells with retinal explants increased retinal ganglion cell survival, after 7 days ex vivo, by ∼2-fold and was associated with reduced apoptosis and increased nerve fibre layer and inner plexiform layer thicknesses. These effects were not demonstrated by co-culture with human or mouse fibroblasts. Conditioned media from mesenchymal stem cells conferred neuroprotection, suggesting that the neuroprotection is mediated, at least partly, by secreted factors. We compared the concentrations of 29 factors in human mesenchymal stem cell and fibroblast conditioned media, and identified 11 enriched in the mesenchymal stem cell secretome. Treatment of retinal explants with a cocktail of these factors conferred retinal ganglion cell neuroprotection, with factors from the platelet-derived growth factor family being the most potent. Blockade of platelet-derived growth factor signalling with neutralizing antibody or with small molecule inhibitors of platelet-derived growth factor receptor kinase or downstream phosphatidylinositol 3 kinase eliminated retinal ganglion cell neuroprotection conferred by mesenchymal stem cell co-culture. Intravitreal injection of platelet-derived growth factor -AA or -AB led to profound optic nerve neuroprotection in vivo following experimental induction of elevated intraocular pressure. These data demonstrate that mesenchymal stem cells secrete a number of neuroprotective proteins and suggest that platelet-derived growth factor secretion in particular may play an important role in mesenchymal stem cell-mediated retinal ganglion cell neuroprotection. Furthermore, platelet-derived growth factor may represent an independent target for achieving retinal ganglion cell neuroprotection.


Assuntos
Células-Tronco Mesenquimais/metabolismo , Fármacos Neuroprotetores/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Células Ganglionares da Retina/metabolismo , Animais , Axotomia/efeitos adversos , Técnicas de Cocultura/métodos , Humanos , Células-Tronco Mesenquimais/patologia , Ratos , Ratos Sprague-Dawley , Ratos Transgênicos , Células Ganglionares da Retina/patologia
15.
Nature ; 455(7215): 984-7, 2008 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-18923526

RESUMO

An important step in the biosynthesis of many proteins is their partial or complete translocation across the plasma membrane in prokaryotes or the endoplasmic reticulum membrane in eukaryotes. In bacteria, secretory proteins are generally translocated after completion of their synthesis by the interaction of the cytoplasmic ATPase SecA and a protein-conducting channel formed by the SecY complex. How SecA moves substrates through the SecY channel is unclear. However, a recent structure of a SecA-SecY complex raises the possibility that the polypeptide chain is moved by a two-helix finger domain of SecA that is inserted into the cytoplasmic opening of the SecY channel. Here we have used disulphide-bridge crosslinking to show that the loop at the tip of the two-helix finger of Escherichia coli SecA interacts with a polypeptide chain right at the entrance into the SecY pore. Mutagenesis demonstrates that a tyrosine in the loop is particularly important for translocation, but can be replaced by some other bulky, hydrophobic residues. We propose that the two-helix finger of SecA moves a polypeptide chain into the SecY channel with the tyrosine providing the major contact with the substrate, a mechanism analogous to that suggested for hexameric, protein-translocating ATPases.


Assuntos
Adenosina Trifosfatases/química , Adenosina Trifosfatases/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Escherichia coli/enzimologia , Proteínas de Membrana Transportadoras/química , Proteínas de Membrana Transportadoras/metabolismo , Adenosina Trifosfatases/genética , Motivos de Aminoácidos , Proteínas de Bactérias/genética , Reagentes de Ligações Cruzadas , Dissulfetos/química , Dissulfetos/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Proteínas de Membrana Transportadoras/genética , Modelos Biológicos , Modelos Moleculares , Conformação Proteica , Transporte Proteico , Canais de Translocação SEC , Proteínas SecA , Relação Estrutura-Atividade , Tirosina/metabolismo
16.
J Biol Chem ; 283(23): 15709-15, 2008 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-18359943

RESUMO

In bacteria most secretory proteins are transported across the plasma membrane by the interplay of the ATPase SecA with the translocation channel formed by the SecY complex; SecA uses cycles of ATP hydrolysis to "push" consecutive segments of a polypeptide substrate through the channel. Here we have addressed the mechanism of this process by following the fate of stalled translocation intermediates. These were generated by using a polypeptide substrate containing a bulky disulfide-bonded loop, thus preventing the final residues from passing through the channel. Protease protection experiments showed that the intermediates were stable in the presence of ATP and could complete translocation once the block was removed. The translocation intermediate was also stable when SecA associated with ATPgammaS, a poorly hydrolyzable ATP analog, or ADP plus AlF(4), which mimics the transition state during ATP hydrolysis. In contrast, when SecA was in its ADP-bound state, the translocating polypeptide moved back into the cytosol, as indicated by the disappearance of the protected fragment. Backsliding was not significantly altered by deletion of the plug domain, a short helix in the center of the SecY channel, but it was slowed down when changes were introduced into the pore ring, the constriction of the hourglass-shaped channel. In all cases, backsliding was significantly slower than forward translocation. Together, these data suggest that SecA binds the polypeptide chain in its ATP state and releases it in the ADP state. The channel itself does not bind the polypeptide chain but provides "friction" that minimizes backsliding when ADP-bound SecA resets to "grab" the next segment of the substrate.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Membrana Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Trifosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/metabolismo , Proteínas da Membrana Bacteriana Externa/genética , Membrana Celular/genética , Proteínas de Ligação a DNA/genética , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Hidrólise , Estrutura Secundária de Proteína/fisiologia , Transporte Proteico/fisiologia , Canais de Translocação SEC
17.
Cell ; 129(1): 97-110, 2007 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-17418789

RESUMO

Many proteins are translocated across the bacterial plasma membrane by the interplay of the cytoplasmic ATPase SecA with a protein-conducting channel, formed from the evolutionarily conserved heterotrimeric SecY complex. Here, we have used purified E. coli components to address the mechanism of translocation. Disulfide bridge crosslinking demonstrates that SecA transfers both the signal sequence and the mature region of a secreted substrate into a single SecY molecule. However, protein translocation involves oligomers of the SecY complex, because a SecY molecule defective in translocation can be rescued by linking it covalently with a wild-type SecY copy. SecA interacts through one of its domains with a nontranslocating SecY copy and moves the polypeptide chain through a neighboring SecY copy. Oligomeric channels with only one active pore likely mediate protein translocation in all organisms.


Assuntos
Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Transporte Proteico , Adenosina Trifosfatases/química , Adenosina Trifosfatases/metabolismo , Sequência de Aminoácidos , Proteínas da Membrana Bacteriana Externa/química , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Escherichia coli/enzimologia , Proteínas de Escherichia coli/química , Proteínas de Membrana Transportadoras/química , Proteínas de Membrana Transportadoras/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Sinais Direcionadores de Proteínas , Estrutura Terciária de Proteína , Canais de Translocação SEC , Proteínas SecA
18.
Annu Rev Cell Dev Biol ; 21: 529-50, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16212506

RESUMO

The conserved protein-conducting channel, referred to as the Sec61 channel in eukaryotes or the SecY channel in eubacteria and archaea, translocates proteins across cellular membranes and integrates proteins containing hydrophobic transmembrane segments into lipid bilayers. Structural studies illustrate how the protein-conducting channel accomplishes these tasks. Three different mechanisms, each requiring a different set of channel binding partners, are employed to move polypeptide substrates: The ribosome feeds the polypeptide chain directly into the channel, a ratcheting mechanism is used by the eukaryotic endoplasmic reticulum chaperone BiP, and a pushing mechanism is utilized by the bacterial ATPase SecA. We review these translocation mechanisms, relating biochemical and genetic observations to the structures of the protein-conducting channel and its binding partners.


Assuntos
Proteínas Fúngicas/química , Proteínas Fúngicas/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Transporte Proteico , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Animais , Cristalografia por Raios X , Retículo Endoplasmático/metabolismo , Canais Iônicos/química , Proteínas de Membrana Transportadoras , Modelos Biológicos , Modelos Moleculares , Ribossomos/metabolismo , Canais de Translocação SEC , Relação Estrutura-Atividade
19.
Biochim Biophys Acta ; 1659(1): 1-18, 2004 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-15511523

RESUMO

A large class of ATPases contains a RecA-like structural domain and uses the energy of nucleotide binding and hydrolysis to perform mechanical work, for example, to move polypeptides or nucleic acids. These ATPases include helicases, ABC transporters, clamp loaders, and proteases. The functional units of the ATPases contain different numbers of RecA-like domains, but the nucleotide is always bound at the interface between two adjacent RecA-like folds and the two domains move relative to one another during the ATPase cycle. The structures determined for different RecA-like motor ATPases begin to reveal how they move macromolecules.


Assuntos
Adenosina Trifosfatases/química , Adenosina Trifosfatases/fisiologia , Proteínas Motores Moleculares/química , Proteínas Motores Moleculares/fisiologia , Recombinases Rec A/química , Recombinases Rec A/metabolismo , Animais , Sítios de Ligação , Transporte Biológico Ativo/fisiologia , Ativação Enzimática , Humanos , Modelos Biológicos , Modelos Moleculares , Movimento (Física) , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Relação Estrutura-Atividade , Especificidade por Substrato
20.
Proc Natl Acad Sci U S A ; 101(30): 10937-42, 2004 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-15256599

RESUMO

The ATPase SecA mediates the posttranslational translocation of a wide range of polypeptide substrates through the SecY channel in the cytoplasmic membrane of bacteria. We have determined the crystal structure of a monomeric form of Bacillus subtilis SecA at a 2.2-A resolution. A comparison with the previously determined structures of SecA reveals a nucleotide-independent, large conformational change that opens a deep groove similar to that in other proteins that interact with diverse polypeptides. We propose that the open form of SecA represents an activated state.


Assuntos
Adenosina Trifosfatases/química , Proteínas de Bactérias/química , Proteínas de Membrana Transportadoras/química , Adenosina Trifosfatases/metabolismo , Bacillus subtilis/metabolismo , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Clonagem Molecular , Cristalografia por Raios X , Proteínas de Membrana Transportadoras/metabolismo , Modelos Moleculares , Conformação Proteica , Estrutura Secundária de Proteína , Transporte Proteico , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Canais de Translocação SEC , Proteínas SecA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA