Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
2.
Curr Gene Ther ; 15(1): 3-14, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25545919

RESUMO

Immune responses to expressed foreign transgenes continue to hamper progress of gene therapy development. Translated foreign proteins with intracellular location are generally less accessible to the immune system, nevertheless they can be presented to the immune system through both MHC Class I and Class II pathways. When the foreign protein luciferase was expressed following intramuscular delivery of plasmid DNA in outbred mice, expression rapidly declined over 4 weeks. Through modifications to the expression plasmid and the luciferase transgene we examined the effect of detargeting expression away from antigen-presenting cells (APCs), targeting expression to skeletal muscle and fusion with glycine-alanine repeats (GAr) that block MHC-Class I presentation on the duration of luciferase expression. De-targeting expression from APCs with miR142-3p target sequences incorporated into the luciferase 3'UTR reduced the humoral immune response to both native and luciferase modified with a short GAr sequence but did not prolong the duration of expression. When a skeletal muscle specific promoter was combined with the miR target sequences the humoral immune response was dampened and luciferase expression persisted at higher levels for longer. Interestingly, fusion of luciferase with a longer GAr sequence promoted the decline in luciferase expression and increased the humoral immune response to luciferase. These studies demonstrate that expression elements and transgene modifications can alter the duration of transgene expression but other factors will need to overcome before foreign transgenes expressed in skeletal muscle are immunologically silent.


Assuntos
Células Apresentadoras de Antígenos/citologia , Dipeptídeos/metabolismo , Marcação de Genes , Luciferases/genética , Repetições de Microssatélites , Músculo Esquelético/metabolismo , Animais , Escherichia coli , Feminino , Técnicas de Transferência de Genes , Vetores Genéticos , Células HEK293 , Humanos , Luciferases/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , MicroRNAs/genética , MicroRNAs/metabolismo , Regiões Promotoras Genéticas , Análise de Sequência de DNA , Transgenes , Células U937
3.
J Neuroimmunol ; 276(1-2): 18-23, 2014 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-25175067

RESUMO

GAP-43 has been studied in the rodent and mammalian brain and shown to be present specifically in areas undergoing axonal elongation and synapse formation. GAP-43 was cloned using the baculovirus expression system and purified. A sandwich ELISA was developed using the recombinant GAP-43 as standard and validated. CSF GAP-43 levels were analysed in benign intracranial hypertension, movement disorders, multiple sclerosis, neuropathy, CNS infections, motor neuron disease, and headache (neurological controls). GAP-43 levels were low in all disorders analysed (in particular motor neuron disease; p=0.001, and movement disorders and multiple sclerosis; p<0.0001) compared to controls, aside from CNS infections. GAP-43 is preferentially reduced in the CSF of neurological disorders associated with neurodegeneration.


Assuntos
Clonagem de Organismos , Ensaio de Imunoadsorção Enzimática/métodos , Proteína GAP-43/genética , Proteína GAP-43/metabolismo , Doenças do Sistema Nervoso/diagnóstico , Animais , Humanos , Camundongos , Doenças do Sistema Nervoso/líquido cefalorraquidiano
4.
Ann Rheum Dis ; 73(9): 1728-36, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23813971

RESUMO

BACKGROUND: Latent cytokines are engineered by fusing the latency associated peptide (LAP) derived from transforming growth factor-ß (TGF-ß) with the therapeutic cytokine, in this case interferon-ß (IFN-ß), via an inflammation-specific matrix metalloproteinase (MMP) cleavage site. OBJECTIVES: To demonstrate latency and specific delivery in vivo and to compare therapeutic efficacy of aggrecanase-mediated release of latent IFN-ß in arthritic joints to the original MMP-specific release. METHODS: Recombinant fusion proteins with MMP, aggrecanase or devoid of cleavage site were expressed in CHO cells, purified and characterised in vitro by Western blotting and anti-viral protection assays. Therapeutic efficacy and half-life were assessed in vivo using the mouse collagen-induced arthritis model (CIA) of rheumatoid arthritis and a model of acute paw inflammation, respectively. Transgenic mice with an IFN-regulated luciferase gene were used to assess latency in vivo and targeted delivery to sites of disease. RESULTS: Efficient localised delivery of IFN-ß to inflamed paws, with low levels of systemic delivery, was demonstrated in transgenic mice using latent IFN-ß. Engineering of latent IFN-ß with an aggrecanase-sensitive cleavage site resulted in efficient cleavage by ADAMTS-4, ADAMTS-5 and synovial fluid from arthritic patients, with an extended half-life similar to the MMP-specific molecule and greater therapeutic efficacy in the CIA model. CONCLUSIONS: Latent cytokines require cleavage in vivo for therapeutic efficacy, and they are delivered in a dose dependent fashion only to arthritic joints. The aggrecanase-specific cleavage site is a viable alternative to the MMP cleavage site for the targeting of latent cytokines to arthritic joints.


Assuntos
Antirreumáticos/administração & dosagem , Artrite Experimental/tratamento farmacológico , Artrite Reumatoide/tratamento farmacológico , Interferon beta/administração & dosagem , Animais , Antirreumáticos/farmacocinética , Antirreumáticos/uso terapêutico , Células CHO , Cricetulus , Citocinas , Relação Dose-Resposta a Droga , Sistemas de Liberação de Medicamentos/métodos , Avaliação Pré-Clínica de Medicamentos/métodos , Endopeptidases , Meia-Vida , Interferon beta/farmacocinética , Interferon beta/uso terapêutico , Metaloproteinases da Matriz , Camundongos , Camundongos Transgênicos , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/farmacocinética , Proteínas Recombinantes de Fusão/uso terapêutico
5.
FASEB J ; 28(1): 373-81, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24072781

RESUMO

Gene therapy is a powerful approach to treat disease locally. However, if the therapeutic target is intracellular, the therapeutic will be effective only in the cells where the therapeutic gene is delivered. We have engineered a fusion protein containing an intracellular inhibitor of the transcription factor NF-κB pathway that can be effectively secreted from producing cells. This fusion protein is cleaved extracellularly by metalloproteinases allowing release of a protein transduction domain (PTD) linked to the NF-κB inhibitor for translocation into neighboring cells. We show that engineered molecules can be efficiently secreted (>80%); are cleaved with matrix metalloprotease-1; inhibit NF-κB driven transcription in a biological assay with a human reporter cell line; and display significant inhibition in mouse paw inflammation models when delivered by lentivirus or secreting cells. No inhibition of NF-κB transcription or therapeutic effect was seen using molecules devoid of the PTD and NF-κB inhibitory domains. By creating a fusion protein with an endogenous secretion partner, we demonstrate a novel approach to efficiently secrete PTD-containing protein domains, overcoming previous limitations, and allowing for potent paracrine effects.


Assuntos
NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Linhagem Celular , Terapia Genética/métodos , Humanos , Metaloproteinase 1 da Matriz/genética , Metaloproteinase 1 da Matriz/metabolismo , Proteínas Recombinantes de Fusão/genética
6.
Expert Opin Drug Deliv ; 11(1): 5-16, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24073618

RESUMO

OBJECTIVES: Targeting cytokines to sites of disease has clear advantages because it increases their therapeutic index. We designed fusion proteins of the latent-associated peptide (LAP) derived from TGF-ß with various cytokines via a matrix metalloproteinase (MMP) cleavage site. This design confers latency, increased half-life and targeting to sites of inflammation. The aim of this study is to determine whether this approach can be applied to cytokines of different molecular structures and sizes. METHODS: Mature cytokines cloned downstream of LAP and a MMP cleavage site were expressed in 293T cells and assessed for latency and biological activity by Western blotting and bioassay. RESULTS: We demonstrate here that fusion proteins of TGF-ß, erythropoietin, IL-1ra, IL-10, IL-4, BMP-7, IGF1 and IL-17 were rendered latent by fusion to LAP, requiring cleavage to become active in respective bioassays. As further proof of principle, we also show that delivery of engineered TGF-ß can inhibit experimental autoimmune encephalomyelitis and that this approach can be used to efficiently deliver cytokines to the brain and spinal cord in mice with this disease. CONCLUSIONS: The latent cytokine approach can be successfully applied to a range of molecules, including cytokines of different molecular structure and mass, growth factors and a cytokine antagonist.


Assuntos
Citocinas/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Metaloproteinase 1 da Matriz/metabolismo , Peptídeos/metabolismo , Precursores de Proteínas/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Embrião de Galinha , Citocinas/genética , Encefalomielite Autoimune Experimental/tratamento farmacológico , Fibroblastos , Células HEK293 , Células HeLa , Humanos , Fator de Crescimento Insulin-Like I/genética , Metaloproteinase 1 da Matriz/genética , Camundongos , Camundongos Endogâmicos DBA , Vison , Terapia de Alvo Molecular , Peptídeos/genética , Peptídeos/uso terapêutico , Precursores de Proteínas/genética , Precursores de Proteínas/uso terapêutico , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/uso terapêutico , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/uso terapêutico
7.
Expert Opin Drug Deliv ; 11(1): 101-10, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24294995

RESUMO

INTRODUCTION: The use of cytokines as therapeutic agents is important, given their potent biological effects. However, this very potency, coupled with the pleiotropic nature and short half-life of these molecules, has limited their therapeutic use. Strategies to increase the half-life and to decrease toxicity are necessary to allow effective treatment with these molecules. AREAS COVERED: A number of strategies are used to overcome the natural limitations of cytokines, including PEGylation, encapsulation in liposomes, fusion to targeting peptides or antibodies and latent cytokines. Latent cytokines are engineered using the latency-associated peptide of transforming growth factor-ß to produce therapeutic cytokines/peptides that are released only at the site of disease by cleavage with disease-induced matrix metalloproteinases. The principles underlying the latent cytokine technology are described and are compared to other methods of cytokine delivery. The potential of this technology for developing novel therapeutic strategies for the treatment of diseases with an inflammatory-mediated component is discussed. EXPERT OPINION: Methods of therapeutic cytokine delivery are addressed. The latent cytokine technology holds significant advantages over other methods of drug delivery by providing simultaneously increased half-life and localised drug delivery without systemic effects. Cytokines that failed clinical trials should be reassessed using this delivery system.


Assuntos
Citocinas/administração & dosagem , Sistemas de Liberação de Medicamentos , Peptídeos/administração & dosagem , Precursores de Proteínas/administração & dosagem , Fator de Crescimento Transformador beta/administração & dosagem , Animais , Citocinas/química , Humanos , Inflamação/tratamento farmacológico , Peptídeos/química , Precursores de Proteínas/química , Fator de Crescimento Transformador beta/química
8.
Mol Med ; 19: 223-9, 2013 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-23821361

RESUMO

Erythropoietin (EPO) has protective effects in neurodegenerative and neuroinflammatory diseases, including in animal models of multiple sclerosis, where EPO decreases disease severity. EPO also promotes neurogenesis and is protective in models of toxic demyelination. In this study, we asked whether EPO could promote neurorepair by also inducing remyelination. In addition, we investigated whether the effect of EPO could be mediated by the classical erythropoietic EPO receptor (EPOR), since it is still questioned if EPOR is functional in nonhematopoietic cells. Using CG4 cells, a line of rat oligodendrocyte precursor cells, we found that EPO increases the expression of myelin genes (myelin oligodendrocyte glycoprotein [MOG] and myelin basic protein [MBP]). EPO had no effect in wild-type CG4 cells, which do not express EPOR, whereas it increased MOG and MBP expression in cells engineered to overexpress EPOR (CG4-EPOR). This was reflected in a marked increase in MOG protein levels, as detected by Western blot. In these cells, EPO induced by 10-fold the early growth response gene 2 (Egr2), which is required for peripheral myelination. However, Egr2 silencing with a siRNA did not reverse the effect of EPO, indicating that EPO acts through other pathways. In conclusion, EPO induces the expression of myelin genes in oligodendrocytes and this effect requires the presence of EPOR. This study demonstrates that EPOR can mediate neuroreparative effects.


Assuntos
Eritropoetina/farmacologia , Bainha de Mielina/genética , Oligodendroglia/efeitos dos fármacos , Receptores da Eritropoetina/metabolismo , Animais , Linhagem Celular , Linhagem Celular Tumoral , Regulação da Expressão Gênica/efeitos dos fármacos , Proteína Básica da Mielina/genética , Glicoproteína Mielina-Oligodendrócito/genética , Glicoproteína Mielina-Oligodendrócito/metabolismo , Oligodendroglia/metabolismo , Ratos
9.
J Invest Dermatol ; 133(11): 2576-2584, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23604101

RESUMO

We have previously designed a conditionally replicative oncolytic adenovirus (CRAd) named Ad-F512 that can target both the stromal and the malignant melanoma cell compartments. The replication capacity of this CRAd is driven by a 0.5-Kb SPARC promoter fragment (named F512). To improve CRAd's efficacy, we cloned into F512 motives responsive to hypoxia (hypoxia-responsive element (HRE)) and inflammation (nuclear factor kappa B) to obtain a chimeric promoter named κBF512HRE. Using luciferase as a reporter gene, we observed 10-15-fold increased activity under hypoxia and 10-80-fold induction upon tumor necrosis factor-α addition. We next constructed a CRAd (Ad-κBF512HRE) where E1A activity was under κBF512HRE regulation. Treatment of nude mice harboring established tumors made of a mix of SB2 melanoma cells and WI-38 fibroblasts with Ad-κBF512HRE led to the complete elimination of tumors in 100% of mice (8/8). Moreover, Ad-5/3-κBF512HRE, a viral variant pseudotyped with a chimeric 5/3 fiber, exerted a strong lytic effect on CAR-negative melanoma cells and was highly effective in vivo on established tumors made of melanoma cells and WI-38 fibroblasts, leading to the complete elimination of 4/5 tumors. These results indicate that this improved stroma-targeted oncolytic adenovirus can override the resistance of melanoma tumors and might become of significant importance for melanoma therapeutics.


Assuntos
Adenoviridae/genética , Melanoma/terapia , Terapia Viral Oncolítica/métodos , Neoplasias Cutâneas/terapia , Células Estromais/virologia , Microambiente Tumoral/fisiologia , Animais , Linhagem Celular Tumoral , Fibroblastos/citologia , Células HEK293 , Humanos , Masculino , Melanoma/patologia , Camundongos , Camundongos Nus , Regiões Promotoras Genéticas/genética , Neoplasias Cutâneas/patologia , Células Estromais/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Ann Rheum Dis ; 72(1): 129-35, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22843486

RESUMO

OBJECTIVES: The synovial endothelium targeting peptide (SyETP) CKSTHDRLC has been identified previously and was shown to preferentially localise to synovial xenografts in the human/severe combined immunodeficient (SCID) mouse chimera model of rheumatoid arthritis (RA). The objective of the current work was to generate SyETP-anti-inflammatory-cytokine fusion proteins that would deliver bioactive cytokines specifically to human synovial tissue. METHODS: Fusion proteins consisting of human interleukin (IL)-4 linked via a matrix metalloproteinase (MMP)-cleavable sequence to multiple copies of either SyETP or scrambled control peptide were expressed in insect cells, purified by Ni-chelate chromatography and bioactivity tested in vitro. The ability of SyETP to retain bioactive cytokine in synovial but not control skin xenografts in SCID mice was determined by in vivo imaging using nano-single-photon emission computed tomography-computed tomography (nano-SPECT-CT) and measuring signal transducer and activator of transcription 6 (STAT6) phosphorylation in synovial grafts following intravenous administration of the fusion protein. RESULTS: In vitro assays confirmed that IL-4 and the MMP-cleavable sequence were functional. IL-4-SyETP augmented production of IL-1 receptor antagonist (IL-1ra) by fibroblast-like synoviocytes (FLS) stimulated with IL-1ß  in a dose-dependent manner. In vivo imaging showed that IL-4-SyETP was retained in synovial but not in skin tissue grafts and the period of retention was significantly enhanced through increasing the number of SyETP copies from one to three. Finally, retention correlated with increased bioactivity of the cytokine as quantified by STAT6 phosphorylation in synovial grafts. CONCLUSIONS: The present work demonstrates that SyETP specifically delivers fused IL-4 to human rheumatoid synovium transplanted into SCID mice, thus providing a proof of concept for peptide-targeted tissue-specific immunotherapy in RA. This technology is potentially applicable to other biological treatments providing enhanced potency to inflammatory sites and reducing systemic toxicity.


Assuntos
Artrite Reumatoide , Citocinas/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Imunoterapia/métodos , Interleucina-4/administração & dosagem , Proteínas Recombinantes de Fusão/administração & dosagem , Animais , Artrite Reumatoide/tratamento farmacológico , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos SCID , Imagem Multimodal , Peptídeos/administração & dosagem , Tomografia por Emissão de Pósitrons , Membrana Sinovial/efeitos dos fármacos , Membrana Sinovial/metabolismo , Tomografia Computadorizada por Raios X , Transplante Heterólogo
11.
J Biotechnol ; 161(3): 269-77, 2012 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-22835852

RESUMO

The development of novel protein therapeutics relies on the ability to express appreciable amounts of correctly folded recombinant proteins. Latent IFN-ß is engineered using the latency-associated peptide (LAP) of transforming growth factor ß1 (TGF-ß1) to maintain IFN-ß in a biologically inactive form until such time as it is released at sites of inflammation by matrix metalloproteinase activity (see Adams et al., 2003). CHO cells cultured in suspension were used for expression of latent IFN-ß to allow medium scale transient transfection. However, the recombinant protein expressed in this system consisted of a mixture of properly linked disulphide dimers and monomers. The ratio of dimer:monomer produced could be significantly altered towards increased dimer production by the addition of L-cystine to the CHO culture medium. The total yield of latent IFN-ß was increased by co-transfection of plasmid coding for the simian virus (SV) 40 large T antigen to the plasmid with the SV40 origin of replication expressing latent IFN-ß DNA. These results provide valuable new insights for developing protocols to produce substantial quantities of latent cytokine dimers in CHO cells in suspension.


Assuntos
Cistina/farmacologia , Dissulfetos/metabolismo , Proteínas de Ligação a TGF-beta Latente/metabolismo , Multimerização Proteica/efeitos dos fármacos , Proteínas Recombinantes de Fusão/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Antígenos Transformantes de Poliomavirus/metabolismo , Células CHO , Células Cultivadas , Clonagem Molecular , Cricetinae , Meios de Cultura/química , DNA/metabolismo , Dosagem de Genes/genética , Interferon beta/metabolismo , Espaço Intracelular/metabolismo , Camundongos , Proteínas Mutantes/metabolismo , Plasmídeos/genética , Isomerases de Dissulfetos de Proteínas/metabolismo , Substâncias Redutoras/farmacologia , Temperatura , Transfecção
12.
Proc Natl Acad Sci U S A ; 109(24): 9617-22, 2012 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-22645329

RESUMO

Erythropoietin (EPO) is a neuroprotective cytokine in models of ischemic and nervous system injury, where it reduces neuronal apoptosis and inflammatory cytokines and increases neurogenesis and angiogenesis. EPO also improves cognition in healthy volunteers and schizophrenic patients. We studied the effect of EPO administration on the gene-expression profile in the ischemic cortex of rats after cerebral ischemia at early time points (2 and 6 h). EPO treatment up-regulated genes already increased by ischemia. Hierarchical clustering and analysis of overrepresented functional categories identified genes implicated in synaptic plasticity-Arc, BDNF, Egr1, and Egr2, of which Egr2 was the most significantly regulated. Up-regulation of Arc, BDNF, Dusp5, Egr1, Egr2, Egr4, and Nr4a3 was confirmed by quantitative PCR. We investigated the up-regulation of Egr2/Krox20 further because of its role in neuronal plasticity. Its elevation by EPO was confirmed in an independent in vivo experiment of cerebral ischemia in rats. Using the rat neuroblastoma B104, we found that wild-type cells that do not express EPO receptor (EPOR) do not respond to EPO by inducing Egr2. However, EPOR-expressing B104 cells induce Egr2 early upon incubation with EPO, indicating that Egr2 induction is a direct effect of EPO and that EPOR mediates this effect. Because these changes occur in vivo before decreased inflammatory cytokines or neuronal apoptosis is evident, these findings provide a molecular mechanism for the neuroreparative effects of cytokines and suggest a mechanism of neuroprotection by which promotion of a plastic phenotype results in decreased inflammation and neuronal death.


Assuntos
Encéfalo/metabolismo , Eritropoetina/fisiologia , Perfilação da Expressão Gênica , Plasticidade Neuronal/genética , Acidente Vascular Cerebral/genética , Animais , Reação em Cadeia da Polimerase , Ratos
14.
Ann Rheum Dis ; 71(1): 143-9, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21998117

RESUMO

OBJECTIVE: To facilitate the targeting to inflammation sites of small anti-inflammatory peptides, with short half-lives, by fusion with the latency-associated peptide (LAP) of transforming growth factor ß1 through a cleavable matrix metalloproteinase (MMP) linker. This design improves efficacy, overcoming the limitations to their clinical use. METHODS: We generated latent forms of vasoactive intestinal peptide (VIP), α-melanocyte-stimulating hormone (MSH) and γ(3)MSH by fusion to LAP through an MMP cleavage site using recombinant DNA technology. The biological activities of these latent therapeutics were studied in vivo using monosodium urate (MSU)-induced peritonitis and collagen-induced arthritis (CIA) models. We assessed gene therapy and purified protein therapy. RESULTS: The recruitment of the polymorphonuclear cells induced by MSU injection into mouse peritoneal cavity was reduced by 35% with γ(3)MSH (1 nmol), whereas administration of a much lower dose of purified latent LAP-MMP-γ(3)MSH (0.03 nmol) attenuated leucocyte influx by 50%. Intramuscular gene delivery of plasmids coding LAP-MMP-VIP and LAP-MMP-αMSH at disease onset reduced the development of CIA compared with LAP-MMP, which does not contain any therapeutic moiety. Histological analysis confirmed a significantly lower degree of inflammation, bone and cartilage erosion in groups treated with LAP-MMP-VIP or LAP-MMP-αMSH. Antibody titres to collagen type II and inflammatory cytokine production were also reduced in these two groups. CONCLUSION: Incorporation of small anti-inflammatory peptides within the LAP shell and delivered as recombinant protein or through gene therapy can control inflammatory and arthritic disease. This platform delivery can be developed to control human arthritides and other autoimmune diseases.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Artrite Experimental/terapia , Hormônios Estimuladores de Melanócitos/uso terapêutico , Fragmentos de Peptídeos/uso terapêutico , Peritonite/terapia , Proteínas Recombinantes de Fusão/uso terapêutico , Fator de Crescimento Transformador beta/uso terapêutico , Peptídeo Intestinal Vasoativo/uso terapêutico , Animais , Anti-Inflamatórios não Esteroides/farmacocinética , Artrite Experimental/tratamento farmacológico , Artrite Experimental/metabolismo , Artrite Experimental/patologia , Citocinas/sangue , Sistemas de Liberação de Medicamentos , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos/métodos , Terapia Genética/métodos , Meia-Vida , Masculino , Hormônios Estimuladores de Melanócitos/genética , Hormônios Estimuladores de Melanócitos/farmacocinética , Camundongos , Camundongos Endogâmicos DBA , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/farmacocinética , Peritonite/tratamento farmacológico , Peritonite/metabolismo , Proteínas Recombinantes de Fusão/farmacocinética , Distribuição Tecidual , Resultado do Tratamento , Peptídeo Intestinal Vasoativo/genética , Peptídeo Intestinal Vasoativo/farmacocinética
15.
J Mol Med (Berl) ; 90(4): 401-11, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22038171

RESUMO

The aim of this study was to construct a promoter containing DNA motifs for an endogenous transcription factor associated with inflammation along with motifs for pharmacological regulation factors. We demonstrate in transfected cells that expression of a gene of interest is induced by hypoxic conditions or through pharmacological induction, and also show pharmacological repression. In vivo studies utilised electroporation of plasmid to mouse paws, a delivery method shown to be effective by bioluminescence imaging. For gene therapy, the promoter was used to drive expression of IL-1Ra in a paw inflammation model with therapeutic effect observed which was further enhanced when the promoter was additionally induced with a pharmacological activator. One of the most important observations from this study was that promoter induction by hypoxia or inflammation could be prevented by the pharmacological repressor in the absence of doxycycline. These studies demonstrate that hybrid promoters enable pharmacological adjustment to the pathophysiological level of gene expression and, importantly, that they allow termination of gene expression even in the presence of pathophysiological stimuli.


Assuntos
Terapia Genética/métodos , Inflamação/terapia , Proteína Antagonista do Receptor de Interleucina 1/genética , Regiões Promotoras Genéticas , Fatores de Transcrição/genética , Animais , Hipóxia Celular , Linhagem Celular , Regulação da Expressão Gênica , Humanos , Inflamação/genética , Proteína Antagonista do Receptor de Interleucina 1/uso terapêutico , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Motivos de Nucleotídeos , Plasmídeos/administração & dosagem , Plasmídeos/genética , Regiões Promotoras Genéticas/efeitos dos fármacos , Tetraciclinas/farmacologia , Transfecção
17.
Biochim Biophys Acta ; 1813(8): 1428-37, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21600935

RESUMO

In order to generate neural stem cells with increased ability to survive after transplantation in brain parenchyma we developed a chimeric receptor (ChR) that binds to myelin oligodendrocyte glycoprotein (MOG) via its ectodomain and activates the insulin-like growth factor receptor type 1 (IGF1R) signalling cascade. Activation of this pro-survival pathway in response to ligand broadly available in the brain might increase neuroregenerative potential of transplanted precursors. The ChR was produced by fusing a MOG-specific single chain antibody with the extracellular boundary of the IGF1R transmembrane segment. The ChR is expressed on the cellular surface, predominantly as a monomer, and is not N-glycosylated. To show MOG-dependent functionality of the ChR, neuroblastoma cells B104 expressing this ChR were stimulated with monolayers of cells expressing recombinant MOG. The ChR undergoes MOG-dependent tyrosine phosphorylation and homodimerisation. It promotes insulin and IGF-independent growth of the oligodendrocyte progenitor cell line CG4. The proposed mode of the ChR activation is by MOG-induced dimerisation which promotes kinase domain transphosphorylation, by-passing the requirement of conformation changes known to be important for IGF1R activation. Another ChR, which contains a segment of the ß-chain ectodomain, was produced in an attempt to recapitulate some of these conformational changes, but proved non-functional.


Assuntos
Glicoproteína Associada a Mielina/imunologia , Glicoproteína Associada a Mielina/metabolismo , Células-Tronco Neurais/metabolismo , Oligodendroglia/metabolismo , Receptor IGF Tipo 1/metabolismo , Anticorpos de Cadeia Única/metabolismo , Animais , Especificidade de Anticorpos , Diferenciação Celular , Linhagem Celular , Sobrevivência Celular , Humanos , Insulina/metabolismo , Camundongos , Proteínas da Mielina , Glicoproteína Mielina-Oligodendrócito , Células-Tronco Neurais/citologia , Células-Tronco Neurais/imunologia , Células-Tronco Neurais/transplante , Oligodendroglia/citologia , Oligodendroglia/imunologia , Oligodendroglia/transplante , Fosforilação , Subunidades Proteicas , Ratos , Receptor IGF Tipo 1/química , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais
18.
Arthritis Rheum ; 62(4): 1007-16, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20131274

RESUMO

OBJECTIVE: To demonstrate that posttranslational modification of type II collagen (CII) by reactive oxygen species (ROS), which are known to be present in inflamed arthritic joints, can give rise to epitopes specific to damaged cartilage in rheumatoid arthritis (RA) and osteoarthritis (OA) and to establish a proof of concept that antibodies specific to ROS-modified CII can be used to target therapeutics specifically to inflamed arthritic joints. METHODS: We used a semisynthetic phage display human antibody library to raise single-chain variable fragments (scFv) specific to ROS-modified CII. The specificity of anti-ROS-modified CII scFv to damaged arthritic cartilage was assessed in vitro by immunostaining articular cartilage from RA and OA patients and from normal controls. The in vivo targeting potential was tested using mice with antigen-induced arthritis, in which localization of anti-ROS-modified CII scFv in the joints was determined. The therapeutic effect of anti-ROS-modified CII scFv fused to soluble murine tumor necrosis factor receptor II-Fc fusion protein (mTNFRII-Fc) was also investigated. RESULTS: The anti-ROS-modified CII scFv bound to damaged arthritic cartilage from patients with RA and OA but not to normal preserved cartilage. When systemically administered to arthritic mice, the anti-ROS-modified CII accumulated selectively at the inflamed joints. Importantly, when fused to mTNFRII-Fc, it significantly reduced inflammation in arthritic mice, as compared with the effects of mTNFRII-Fc alone or of mTNFRII-Fc fused to an irrelevant scFv. CONCLUSION: Our findings indicate that biologic therapeutics can be targeted specifically to arthritic joints and suggest a new approach for the development of novel treatments of arthritis.


Assuntos
Artrite Reumatoide/imunologia , Cartilagem/imunologia , Anticorpos de Cadeia Única/genética , Animais , Artrite Reumatoide/genética , Cartilagem/patologia , Cartilagem Articular/imunologia , Cartilagem Articular/patologia , Bovinos , Modelos Animais de Doenças , Epitopos/imunologia , Humanos , Fragmentos Fc das Imunoglobulinas/imunologia , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Osteoartrite/genética , Osteoartrite/imunologia , Osteoprotegerina/imunologia , Espécies Reativas de Oxigênio/imunologia
19.
J Tissue Eng Regen Med ; 4(5): 387-94, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20101620

RESUMO

Anti-inflammatory gene therapy can inhibit inflammation driven by TNFalpha in experimental models of rheumatoid arthritis. However, assessment of the therapeutic effect on cartilage and bone quality is either missing or unsatisfactory. A multimodal imaging approach, using confocal laser scanning microscopy (CLSM) and micro-computed tomography (microCT), was used for gathering 3D quantitative image data on diseased and treated murine joints. As proof of concept, the efficacy of anti-TNF-based gene therapy was assessed, comparing imaging techniques with classical investigations. SCID mice knees were injected with human synoviocytes overexpressing TNFalpha. Two days later, electric pulse-mediated DNA transfer was performed after injection of the pGTRTT-plasmid containing a dimeric soluble-TNF receptor (dsTNFR) under the control of a doxycycline-inducible promoter. After 21 days the mice were sacrificed, TNFalpha levels were measured and the joints assessed for cartilage and bone degradation, using CLSM, microCT and histology. TNFalpha levels were decreased in the joints of mice treated with the plasmid in the presence of doxycycline. Concomitantly, histological analysis showed an increase in cartilage thickness and a decrease in specific synovial hyperplasia and cartilage erosion. Bone morphometry revealed that groups with the plasmid in the presence of doxycycline displayed a higher cortical thickness and decreased porosity. Using an anti-TNF gene therapy approach, known to reduce inflammation, as proof of concept, 3D imaging allowed quantitative evaluation of its benefits to joint architecture. It showed that local delivery of a regulated anti-TNF vector allowed decreasing arthritis severity through TNFalpha inhibition. These tools are valuable for understanding the efficacy of gene therapy on whole-joint morphometry.


Assuntos
Artrite Experimental/diagnóstico por imagem , Osso e Ossos/diagnóstico por imagem , Cartilagem/diagnóstico por imagem , Diagnóstico por Imagem/métodos , Terapia Genética , Animais , Artrite Experimental/patologia , Osso e Ossos/patologia , Cartilagem/patologia , Ensaio de Imunoadsorção Enzimática , Humanos , Articulação do Joelho/diagnóstico por imagem , Articulação do Joelho/patologia , Camundongos , Tíbia/diagnóstico por imagem , Tíbia/patologia , Fator de Necrose Tumoral alfa/metabolismo , Microtomografia por Raio-X
20.
Biomaterials ; 30(28): 4986-95, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19539363

RESUMO

Non-invasive bioluminescence imaging (BLI) to monitor changes in gene expression of cells implanted in live animals should facilitate the development of biomaterial scaffolds for tissue regeneration. We show that, in vitro, induction of chondrogenic differentiation in mouse bone marrow stromal cell line (CL1) and human adipose tissue derived mesenchymal stromal cells (hAMSCs), permanently transduced with a procollagen II (COL2A1) promoter driving a firefly luciferase gene reporter (PLuc) (COL2A1p.PLuc), induces PLuc expression in correlation with increases in COL2A1 and Sox9 mRNA expression and acquisition of chondrocytic phenotype. To be able to simultaneously monitor in vivo cell differentiation and proliferation, COL2A1p.PLuc labelled cells were also genetically labelled with a renilla luciferase (RLuc) gene driven by a constitutively active cytomegalovirus promoter, and then seeded in demineralized bone matrix (DBM) subcutaneously implanted in SCID mice. Non-invasive BLI monitoring of the implanted mice showed that the PLuc/RLuc ratio reports on gene expression changes indicative of cell differentiation. Large (CL1) and moderated (hAMSCs) changes in the PLuc/RLuc ratio over a 6 week period, revealed different patterns of in vivo chondrogenic differentiation for the CL1 cell line and primary MSCs, in agreement with in vitro published data and our results from histological analysis of DBM sections. This double bioluminescence labelling strategy together with BLI imaging to analyze behaviour of cells implanted in live animals should facilitate the development of progenitor cell/scaffold combinations for tissue repair.


Assuntos
Diferenciação Celular , Colágeno Tipo II/genética , Luciferases de Vaga-Lume/análise , Luciferases de Vaga-Lume/metabolismo , Medições Luminescentes/métodos , Células-Tronco Mesenquimais/citologia , Tecido Adiposo/citologia , Animais , Células da Medula Óssea/citologia , Linhagem Celular , Proliferação de Células , Condrócitos/citologia , Colágeno Tipo II/metabolismo , Diagnóstico por Imagem/métodos , Regulação da Expressão Gênica , Genes Reporter , Humanos , Luciferases de Vaga-Lume/genética , Transplante de Células-Tronco Mesenquimais , Camundongos , Camundongos SCID , Osteogênese , Células Estromais/citologia , Alicerces Teciduais , Transdução Genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA