Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Toxicol Mech Methods ; 29(8): 604-615, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31237465

RESUMO

Phosgene is classified as a chemical warfare agent, yet data on its short-duration high concentration toxicity in a nose-only exposure rat model is sparse and inconsistent. Hence, an exposure system for short-term/high concentration exposure was developed and characterized. Herein, we report the median lethal concentration (LC50) for a 10-min nasal exposure of phosgene in a 24-h rat survival model. Male Wistar rats (Envigo) weighing 180-210 g on the day of exposure, were exposed to phosgene gas via nose-only inhalation using a system specifically designed to allow the simultaneous exposure and quantification of phosgene. After 24 h, the surviving rats were euthanized, the lung/body mass ratio determined, and lung tissues analyzed for histopathology. Increased terminal airway edema in the lungs located primarily at the alveoli (resulting in an increased lung/body mass ratio) coincided with the observed mortality. An LC50 value of 129.2 mg/m3 for a 10-min exposure was determined. Furthermore, in agreement with other highly toxic compounds, this study reveals a LC50 concentration value supportive of a nonlinear toxic load model, where the toxic load exponent is >1 (ne = 1.17). Thus, in line with other chemical warfare agents, phosgene toxicity is predicted to be more severe with short-duration, high-concentration exposures than long-duration, low-concentration exposures. This model is anticipated to be refined and developed to screen novel therapeutics against relevant short-term high concentration phosgene exposures expected from a terrorist attack, battlefield deployment, or industrial accident.


Assuntos
Substâncias para a Guerra Química/toxicidade , Exposição por Inalação/efeitos adversos , Pulmão/efeitos dos fármacos , Fosgênio/toxicidade , Edema Pulmonar/induzido quimicamente , Animais , Relação Dose-Resposta a Droga , Exposição por Inalação/análise , Dose Letal Mediana , Pulmão/patologia , Masculino , Edema Pulmonar/patologia , Ratos Wistar , Análise de Sobrevida , Fatores de Tempo
2.
Int J Cancer ; 138(1): 182-6, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26174762

RESUMO

TP53 is a tumor suppressor gene that is mutated in 50% of cancers, and its function is tightly regulated by the E3 ligase, Mdm2. Both p53 and Mdm2 are localized in the cell nucleus, a site that is impervious to therapeutic regulation by most antibodies. We identified a cell-penetrating lupus monoclonal anti-DNA antibody, mAb 3E10, that targets the nucleus, and we engineered mAb 3E10 to function as an intranuclear transport system to deliver therapeutic antibodies into the nucleus as bispecific single chain Fv (scFv) fragments. Bispecific scFvs composed of 3E10 include PAb421 (3E10-PAb421) that binds p53 and restores the function of mutated p53, and 3G5 (3E10-3G5) that binds Mdm2 and prevents destruction of p53 by Mdm2. We documented the therapeutic efficacy of these bispecific scFvs separately in previous studies. In this study, we show that combination therapy with 3E10-PAb421 and 3E10-3G5 augments growth inhibition of cells with p53 mutations compared to the effect of either antibody alone. By contrast, no enhanced response was observed in cells with wild-type p53 or in cells homozygous null for p53.


Assuntos
Anticorpos Monoclonais/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Anticorpos Biespecíficos/metabolismo , Anticorpos Biespecíficos/farmacologia , Anticorpos Monoclonais/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células HT29 , Humanos , Espaço Intracelular/metabolismo , Neoplasias/patologia , Transporte Proteico , Transdução de Sinais/efeitos dos fármacos
3.
Cell Stress Chaperones ; 28(4): 429-439, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37171750

RESUMO

Heat shock proteins (HSPs), especially Hsp70 (HSPA1), have been associated with cellular protection from various cellular stresses including heat, hypoxia-ischemia, neurodegeneration, toxins, and trauma. Endogenous HSPs are often synthesized in direct response to these stresses but in many situations are inadequate in protecting cells. The present study addresses the transduction of Hsp70 into cells providing protection from acute oxidative stress by H2O2. The recombinant Fv-Hsp70 protein and two mutant Fv-Hsp70 proteins minus the ATPase domain and minus the ATPase and terminal lid domains were tested at 0.5 and 1.0 µM concentrations after two different concentrations of H2O2 treatment. All three recombinant proteins protected SH-SY5Y cells from acute H2O2 toxicity. This data indicated that the protein binding domain was responsible for cellular protection. In addition, experiments pretreating cells with inhibitors of antioxidant proteins catalase and gamma-glutamylcysteine synthase (GGCS) before H2O2 resulted in cell death despite treatment with Fv-Hsp70, implying that both enzymes were protected from acute oxidative stress after treatment with Fv-Hsp70. This study demonstrates that Fv-Hsp70 is protective in our experiments primarily by the protein-binding domain. The Hsp70 terminal lid domain was also not necessary for protection.


Assuntos
Peróxido de Hidrogênio , Neuroblastoma , Humanos , Peróxido de Hidrogênio/toxicidade , Cisteína Sintase , Catalase , Proteínas de Choque Térmico HSP70/metabolismo , Proteínas de Choque Térmico/metabolismo , Proteínas Recombinantes , Adenosina Trifosfatases
4.
J Biol Chem ; 285(45): 34299-303, 2010 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-20843808

RESUMO

Synovial fibroblasts destroy articular cartilage and bone in rheumatoid arthritis, but the mechanism of fibroblast transformation remains elusive. Because gain-of-function mutations of BRAF can transform fibroblasts, we examined BRAF in rheumatoid synovial fibroblasts. The strong gain-of-function mutation, V600R, of BRAF found in melanomas and other cancers was identified in first passage synovial fibroblasts from two of nine rheumatoid arthritis patients and confirmed by restriction site mapping. BRAF-specific siRNA inhibited proliferation of synovial fibroblasts with V600R mutations. A BRAF aberrant splice variant with an intact kinase domain and partial loss of the N-terminal autoinhibitory domain was identified in fibroblasts from an additional patient, and fibroblast proliferation was inhibited by BRAF-specific siRNA. Our finding is the first to establish mechanisms for fibroblast transformation responsible for destruction of articular cartilage and bone in rheumatoid arthritis and establishes a new target for therapeutic intervention.


Assuntos
Artrite Reumatoide/enzimologia , Proliferação de Células , Mutação , Proteínas Proto-Oncogênicas B-raf/metabolismo , Sítios de Splice de RNA , Adulto , Idoso , Idoso de 80 Anos ou mais , Artrite Reumatoide/genética , Artrite Reumatoide/patologia , Artrite Reumatoide/terapia , Osso e Ossos/enzimologia , Osso e Ossos/patologia , Cartilagem Articular/enzimologia , Cartilagem Articular/patologia , Feminino , Fibroblastos , Humanos , Masculino , Pessoa de Meia-Idade , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas B-raf/genética , RNA Interferente Pequeno/genética , Membrana Sinovial
5.
Stroke ; 41(3): 538-43, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20075343

RESUMO

BACKGROUND AND PURPOSE: This study investigated the effects of intravenous recombinant Fv-Hsp70 protein on infarction volume and behavior after experimental ischemic stroke. METHODS: Focal cerebral ischemia was produced by occluding the middle cerebral artery using the intraluminal suture technique. Rats subjected to 2 hours of focal ischemia were allowed to survive 24 hours. At 2(1/4) hours and 3 hours after onset of ischemia, Fv-Hsp70 recombinant protein (0.5 mg/kg) or saline was injected through the tail vein. Sensorimotor function and infarction volume were assessed at 24 hours after ischemia. RESULTS: Administration of Fv-Hsp70 after focal cerebral ischemia significantly decreased infarct volume by 68% and significantly improved sensorimotor function compared with the saline-treated control group. Western blots showed Fv-Hsp70 in ischemic but not in control brain; and Fv-Hsp70 suppressed endogenous Hsp70. CONCLUSIONS: Fv-Hsp70 protected the ischemic brain in this experimental stroke model.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Proteínas de Choque Térmico HSP70/administração & dosagem , Fragmentos de Imunoglobulinas/administração & dosagem , Linfocinas/administração & dosagem , Fármacos Neuroprotetores/administração & dosagem , Proteínas Recombinantes/administração & dosagem , Sialoglicoproteínas/administração & dosagem , Animais , Isquemia Encefálica/patologia , Proteínas de Choque Térmico HSP70/fisiologia , Humanos , Fragmentos de Imunoglobulinas/fisiologia , Injeções Intraventriculares , Linfocinas/fisiologia , Masculino , Ratos , Ratos Sprague-Dawley , Sialoglicoproteínas/fisiologia
6.
Nat Neurosci ; 9(2): 234-42, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16415866

RESUMO

Defects in dendritic spines are common to several forms of cognitive deficits, including mental retardation and Alzheimer disease. Because mutation of p21-activated kinase (PAK) can lead to mental retardation and because PAK-cofilin signaling is critical in dendritic spine morphogenesis and actin dynamics, we hypothesized that the PAK pathway is involved in synaptic and cognitive deficits in Alzheimer disease. Here, we show that PAK and its activity are markedly reduced in Alzheimer disease and that this is accompanied by reduced and redistributed phosphoPAK, prominent cofilin pathology and downstream loss of the spine actin-regulatory protein drebrin, which cofilin removes from actin. We found that beta-amyloid (Abeta) was directly involved in PAK signaling deficits and drebrin loss in Abeta oligomer-treated hippocampal neurons and in the Appswe transgenic mouse model bearing a double mutation leading to higher Abeta production. In addition, pharmacological PAK inhibition in adult mice was sufficient to cause similar cofilin pathology, drebrin loss and memory impairment, consistent with a potential causal role of PAK defects in cognitive deficits in Alzheimer disease.


Assuntos
Doença de Alzheimer/enzimologia , Peptídeos beta-Amiloides/metabolismo , Transtornos Cognitivos/enzimologia , Proteínas Serina-Treonina Quinases/metabolismo , Fatores de Despolimerização de Actina/metabolismo , Doença de Alzheimer/complicações , Doença de Alzheimer/patologia , Animais , Células Cultivadas , Transtornos Cognitivos/etiologia , Transtornos Cognitivos/patologia , Espinhas Dendríticas/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Neuropeptídeos/metabolismo , Ratos , Quinases Ativadas por p21
7.
Cancer Res ; 67(4): 1769-74, 2007 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-17308119

RESUMO

To evaluate the clinical efficacy of monoclonal antibody (mAb) 3E10 Fv antibody-mediated p53 protein therapy, an Fv-p53 fusion protein produced in Pichia pastoris was tested on CT26.CL25 colon cancer cells in vitro and in vivo in a mouse model of colon cancer metastasis to the liver. In vitro experiments showed killing of CT26.CL25 cells by Fv-p53 but not Fv or p53 alone, and immunohistochemical staining confirmed that Fv was required for transport of p53 into cells. Prevention of liver metastasis in vivo was tested by splenic injection of 100 nmol/L Fv-p53 10 min and 1 week after injection of CT26.CL25 cancer cells into the portal vein of BALB/c mice. Mice were sacrificed 1 week after the second injection of Fv-p53 and assigned a quantitative metastasis score. Control mice had an average metastasis score of 3.3 +/- 1.3, whereas mice treated with Fv-p53 had an average metastasis score of 0.8 +/- 0.4 (P = 0.004). These results indicate that Fv-p53 treatment had a profound effect on liver metastasis and represent the first demonstration of effective full-length p53 protein therapy in vivo. mAb 3E10 Fv has significant clinical potential as a mediator of intracellular and intranuclear delivery of p53 for prevention and treatment of cancer metastasis.


Assuntos
Fragmentos de Imunoglobulinas/imunologia , Fragmentos de Imunoglobulinas/farmacologia , Neoplasias Hepáticas Experimentais/prevenção & controle , Neoplasias Hepáticas Experimentais/secundário , Proteína Supressora de Tumor p53/imunologia , Proteína Supressora de Tumor p53/farmacologia , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Linhagem Celular Tumoral , Neoplasias do Colo/imunologia , Neoplasias do Colo/terapia , Feminino , Humanos , Fragmentos de Imunoglobulinas/genética , Neoplasias Hepáticas Experimentais/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/terapia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/farmacologia , Proteína Supressora de Tumor p53/genética
8.
J Am Coll Cardiol ; 70(12): 1479-1492, 2017 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-28911512

RESUMO

BACKGROUND: Although early reperfusion is the most desirable intervention after ischemic myocardial insult, it may add to damage through oxidative stress. OBJECTIVES: This study investigated the cardioprotective effects of a single intravenous dose of heat shock protein-72 (HSP72) coupled to a single-chain variable fragment (Fv) of monoclonal antibody 3E10 (3E10Fv) in a rabbit ischemia-reperfusion model. The Fv facilitates rapid transport of HSP72 into cells, even with intact membranes. METHODS: A left coronary artery occlusion (40 min) reperfusion (3 h) model was used in 31 rabbits. Of these, 12 rabbits received the fusion protein (Fv-HSP72) intravenously. The remaining 19 control rabbits received a molar equivalent of 3E10Fv alone (n = 6), HSP72 alone (n = 6), or phosphate-buffered saline (n = 7). Serial echocardiographic examinations were performed to assess left ventricular function before and after reperfusion. Micro-single-photon emission computed tomography imaging of 99mTc-labeled annexin-V was performed with micro-computed tomography scanning to characterize apoptotic damage in vivo, followed by gamma counting of the excised myocardial specimens to quantify cell death. Histopathological characterization of the myocardial tissue and sequential cardiac troponin I measurements were also undertaken. RESULTS: Myocardial annexin-V uptake was 43% lower in the area at risk (p = 0.0003) in Fv-HSP72-treated rabbits compared with control animals receiving HSP72 or 3E10Fv alone. During reperfusion, troponin I release was 42% lower and the echocardiographic left ventricular ejection fraction 27% higher in the Fv-HSP72-treated group compared with control animals. Histopathological analyses confirmed penetration of 3E10Fv-containing molecules into cardiomyocytes in vivo, and treatment with Fv-HSP72 showed fewer apoptotic nuclei compared with control rabbits. CONCLUSIONS: Single-dose administration of Fv-HSP72 fusion protein at the time of reperfusion reduced myocardial apoptosis by almost one-half and improved left ventricular functional recovery after myocardial ischemia-reperfusion injury in rabbits. It might have potential to serve as an adjunct to early reperfusion in the management of myocardial infarction.


Assuntos
Proteínas de Choque Térmico HSP72/administração & dosagem , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Anticorpos de Cadeia Única/administração & dosagem , Animais , Modelos Animais de Doenças , Ecocardiografia , Masculino , Traumatismo por Reperfusão Miocárdica/sangue , Traumatismo por Reperfusão Miocárdica/diagnóstico por imagem , Traumatismo por Reperfusão Miocárdica/patologia , Coelhos
9.
Brain Res ; 1088(1): 187-96, 2006 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-16630585

RESUMO

Intracellular Hsp70 provides cytoprotection against a variety of stressful stimuli, and an effective means of increasing intracellular Hsp70 levels could prove beneficial in the prevention and treatment of a variety of human diseases. A novel protein transduction domain consisting of the single chain Fv fragment of an anti-DNA antibody known to penetrate into living cells and tissues, mAb 3E10, has recently been used to deliver functional proteins to cells. The ability of the single chain Fv fragment to deliver Hsp70 into living cells was tested by generating an Fv-Hsp70 fusion protein. Fv-Hsp70 was produced as a secreted protein in both COS-7 cells and the methylotropic yeast strain Pichia pastoris and was shown capable of penetrating into COS-7 cells and primary rat cortical neurons. Pre-treatment with Fv-Hsp70 protected both COS-7 cells and primary rat cortical neurons against subsequent exposure to hydrogen peroxide. These results provide the first evidence that the Fv fragment of mAb 3E10 is capable of delivering proteins to neurons and indicate its potential in the development of Hsp70 protein therapy.


Assuntos
Proteínas de Choque Térmico HSP70/fisiologia , Região Variável de Imunoglobulina/fisiologia , Neurônios/metabolismo , Proteínas Recombinantes de Fusão/fisiologia , Animais , Animais Recém-Nascidos , Western Blotting/métodos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Córtex Cerebral/citologia , Chlorocebus aethiops , Relação Dose-Resposta a Droga , Proteínas de Choque Térmico HSP70/biossíntese , Proteínas de Choque Térmico HSP70/farmacologia , Humanos , Peróxido de Hidrogênio/toxicidade , Região Variável de Imunoglobulina/imunologia , Neurônios/efeitos dos fármacos , Oxidantes/toxicidade , Transporte Proteico/fisiologia , Ratos , Ratos Wistar , Proteínas Recombinantes de Fusão/biossíntese , Transdução Genética/métodos , Transfecção/métodos
10.
J Drug Target ; 13(2): 81-7, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15823959

RESUMO

The Fv fragment of an antibody that selectively targets and penetrates skeletal muscle in vivo was produced as a fusion protein with a micro-dystrophin for use as a delivery vehicle to transport micro-dystrophin into muscle cells. Fv-micro-dystrophin was produced as a secreted protein by transient transfection of Fv-micro-dystrophin cDNA in COS-7 cells and as a non-secreted protein by permanent transfection in Pichia pastoris. Isolated Fv-micro-dystrophin was shown to be full-length by Western blot analysis. Fv-micro-dystrophin penetrated multiple cell lines in vitro, and it localized to the plasma membrane of a cell line with membrane beta-dystroglycan. In the absence of membrane beta-dystroglycan, it localized to the cytoplasm. Antibody-mediated transduction of micro-dystrophin into muscle cells is a potential therapy for dystrophin-deficient muscular dystrophies.


Assuntos
Membrana Celular/metabolismo , Distrofina/metabolismo , Fragmentos de Imunoglobulinas , Linfocinas , Sinais Direcionadores de Proteínas , Proteínas Recombinantes de Fusão/metabolismo , Sialoglicoproteínas , Animais , Western Blotting , Linhagem Celular , Chlorocebus aethiops , Clonagem Molecular , Citoplasma/metabolismo , Distroglicanas/metabolismo , Distrofina/genética , Humanos , Mioblastos/metabolismo , Pichia/metabolismo , Transporte Proteico , Proteínas Recombinantes de Fusão/genética , Transfecção
11.
ScientificWorldJournal ; 5: 782-8, 2005 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-16170440

RESUMO

Protein therapy refers to the direct delivery of therapeutic proteins to cells and tissues with the goal of ameliorating or modifying a disease process. Current techniques for delivering proteins across cell membranes include taking advantage of receptor-mediated endocytosis or using protein transduction domains that penetrate directly into cells. The most commonly used protein transduction domains are small cell-penetrating peptides derived from such proteins as the HIV-1 Tat protein. A novel protein transduction domain developed as the single chain fragment (Fv) of a murine anti-DNA autoantibody, mAb 3E10, has recently been developed and used to deliver biologically active proteins to living cells in vitro. This review will provide a brief overview of the development of the Fv fragment and provide a summary of recent studies using Fv to deliver therapeutic peptides and proteins (such as a C-terminal p53 peptide, C-terminal p53 antibody fragment, full-length p53, and micro-dystrophin) to cells.


Assuntos
Autoanticorpos/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Produtos do Gene tat/metabolismo , Proteínas/metabolismo , Transdução de Sinais/fisiologia , Animais , Transporte Biológico Ativo/fisiologia , Células Cultivadas , Endocitose/fisiologia , Produtos do Gene tat/imunologia , HIV-1/metabolismo , Humanos , Fragmentos de Imunoglobulinas , Linfocinas/imunologia , Linfocinas/metabolismo , Camundongos , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/metabolismo , Transporte Proteico/fisiologia , Sialoglicoproteínas/imunologia , Sialoglicoproteínas/metabolismo , Produtos do Gene tat do Vírus da Imunodeficiência Humana
12.
Mol Immunol ; 39(13): 783-9, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12617993

RESUMO

Methods for cell type specific targeted intracellular delivery of proteins in vivo remain limited. A murine monoclonal anti-dsDNA antibody, mAb 3E10, was selectively transported into skeletal muscle cells in vivo. The antibody bound a 200 kDa protein only found in lysates of skeletal muscle by Western blotting. The 200 kDa protein was purified from muscle lysate by antibody affinity chromatography and identified as the skeletal muscle specific heavy chain of myosin IIb by electrospray mass spectrometry. Antibody binding specificity for myosin IIb was demonstrated in Western blots by binding myosin in skeletal muscle lysates from mice null for myosin IId but not in mice null for myosin IIb. Myosin IIb is implicated in the specific targeting of mAb 3E10 to skeletal muscle.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Músculo Esquelético/imunologia , Miosina não Muscular Tipo IIB/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Antinucleares/administração & dosagem , Anticorpos Antinucleares/metabolismo , Anticorpos Monoclonais/farmacocinética , Especificidade de Anticorpos , Feminino , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Músculo Esquelético/metabolismo , Miosina não Muscular Tipo IIB/deficiência , Miosina não Muscular Tipo IIB/genética , Miosina não Muscular Tipo IIB/metabolismo , Ratos , Distribuição Tecidual
13.
Cancer Res ; 75(11): 2285-91, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25832653

RESUMO

The specificity of binding by antibodies to target antigens is a compelling advantage to antibody-based cancer therapy, but most antibodies cannot penetrate cells to affect intracellular processes. Select lupus autoantibodies penetrate into cell nuclei, and the potential for application of these antibodies in cancer therapy is an emerging concept. Here, we show that a divalent lupus anti-DNA autoantibody fragment with enhancing mutations that increase its ability to penetrate cell nuclei and bind DNA causes accumulation of DNA double-strand breaks in and is highly and selectively toxic to cancer cells and tumors with defective homology-directed repair of DNA double-strand breaks. These findings provide proof of principle for the use of optimized lupus autoantibodies in targeted cancer therapy.


Assuntos
Anticorpos Antinucleares/imunologia , Quebras de DNA de Cadeia Dupla , Lúpus Eritematoso Sistêmico/imunologia , Neoplasias/imunologia , Anticorpos Antinucleares/uso terapêutico , Células HCT116 , Humanos , Lúpus Eritematoso Sistêmico/patologia , Neoplasias/patologia , Neoplasias/terapia , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/uso terapêutico
14.
Sci Rep ; 5: 12022, 2015 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-26156563

RESUMO

A nuclear-penetrating lupus anti-DNA autoantibody, 3E10, has been found to inhibit DNA repair and selectively kill certain cancer cells that are highly vulnerable to DNA damage. In addition, a 3E10 single chain variable fragment (scFv) has been developed for use as a delivery vehicle to carry therapeutic cargo proteins into cell nuclei. A greater understanding of the mechanism by which 3E10 penetrates cell nuclei is needed to help determine the scope of its potential therapeutic applications. Here we show that the presence of extracellular DNA significantly enhances the nuclear uptake of 3E10 scFv. In addition, we find that 3E10 scFv preferentially localizes into tumor cell nuclei in vivo, likely due to increased DNA in the local environment released from ischemic and necrotic regions of tumor. These data provide insight into the mechanism of nuclear penetration by 3E10 and demonstrate the potential for use of 3E10 in therapeutic approaches to diseases ranging from malignancy to ischemic conditions such as stroke.


Assuntos
Autoanticorpos/imunologia , Núcleo Celular/imunologia , Núcleo Celular/metabolismo , DNA/imunologia , DNA/metabolismo , Anticorpos Antinucleares/imunologia , Anticorpos Antinucleares/metabolismo , Autoanticorpos/metabolismo , Linhagem Celular Tumoral , Humanos , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/metabolismo , Transporte Proteico , Anticorpos de Cadeia Única/imunologia , Anticorpos de Cadeia Única/metabolismo
15.
Cancer Lett ; 195(2): 211-9, 2003 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-12767530

RESUMO

scFv fragments of a monoclonal antibody that penetrates living cells and localizes in nuclei were designed as fusion proteins with C-terminal p53 peptides and tested for restoring p53 function in p53 mutant cancer cells. scFv fragments transported a 30-mer C-terminal peptide of p53 into cancer cells and induced cellular cytotoxicity in contrast to scFv fragments alone and other scFv-p53 fusion peptides. Cellular toxicity was not observed with scFv fragments containing a single mutation in VH that prevented antibody penetration. Our results demonstrate the potential efficacy of antibody scFv fragments as a nuclear delivery system in cancer cells.


Assuntos
Apoptose/efeitos dos fármacos , Núcleo Celular/metabolismo , Imunoconjugados/administração & dosagem , Região Variável de Imunoglobulina/administração & dosagem , Fragmentos de Peptídeos/administração & dosagem , Ativação Transcricional/efeitos dos fármacos , Proteína Supressora de Tumor p53/administração & dosagem , Transporte Ativo do Núcleo Celular , Adenocarcinoma/patologia , Animais , Anticorpos Monoclonais/administração & dosagem , Neoplasias Ósseas/patologia , Células CHO , Células COS , Permeabilidade da Membrana Celular , Chlorocebus aethiops , Neoplasias Colorretais/patologia , Cricetinae , Sistemas de Liberação de Medicamentos , Genes Reporter , Humanos , Imunoconjugados/farmacologia , Imunoconjugados/toxicidade , Região Variável de Imunoglobulina/farmacologia , Camundongos , Camundongos Endogâmicos MRL lpr , Osteossarcoma/patologia , Fragmentos de Peptídeos/farmacologia , Fragmentos de Peptídeos/toxicidade , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/farmacologia , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/química , Proteína Supressora de Tumor p53/fisiologia , Proteína Supressora de Tumor p53/toxicidade
16.
Int J Oncol ; 25(4): 1113-8, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15375563

RESUMO

A bispecific, single-chain antibody Fv fragment (Bs-scFv) was constructed from a single-chain Fv fragment of mAb 3E10 that penetrates living cells and localizes in the nucleus, and a single-chain Fv fragment of a non-penetrating antibody, mAb PAb421 that binds the C-terminal of p53. PAb421 binding restores wild-type functions of some p53 mutants, including those of SW480 human colon cancer cells. The Bs-scFv penetrated SW480 cells and was cytotoxic, suggesting an ability to restore activity to mutant p53. COS-7 cells (monkey kidney cells with wild-type p53) served as a control since they are unresponsive to PAb421 due to the presence of SV40 large T antigen that inhibits binding of PAb421 to p53. Bs-scFv penetrated COS-7 cells but was not cytotoxic, thereby eliminating non-specific toxicity of Bs-scFv unrelated to binding p53. A single mutation in CDR1 of PAb421 VH eliminated binding of the Bs-scFv to p53 and abrogated cytotoxicity for SW480 cells without altering cellular penetration, further supporting the requirement of PAb421 binding to p53 for cytotoxicity. Our study demonstrates the use of an antibody that penetrates living cells in the design of a bispecific single chain antibody to target and restore the function of an intracellular protein.


Assuntos
Anticorpos Biespecíficos/farmacologia , Anticorpos Monoclonais/farmacologia , Neoplasias do Colo/terapia , Genes p53 , Fragmentos de Imunoglobulinas/farmacologia , Mutação , Proteína Supressora de Tumor p53/metabolismo , Animais , Anticorpos Biespecíficos/genética , Anticorpos Biespecíficos/metabolismo , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/metabolismo , Células COS , Neoplasias do Colo/patologia , Humanos , Fragmentos de Imunoglobulinas/metabolismo , Transfecção
17.
Int J Oncol ; 25(6): 1867-73, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15547728

RESUMO

Some human cancers are caused by functional defects in p53 that are restored by gene therapy with wild-type p53. To circumvent the use of viral vectors, we reconstituted cancer cell lines with p53 by protein transduction. A fusion protein was produced from cDNA constructed from the Fv fragment of an antibody that penetrates living cells and wild-type p53 (Fv-p53). Fv-p53 penetrated and killed cancer cells that do not express p53. Additionally, Fv-p53 killed cancer cells that were malignant as a result of mutations within p53, nuclear exclusion of p53 and over-expression of MDM2. Non-specific toxicity was excluded by showing that Fv-p53 penetrated but did not kill primary cells and cancer cells unresponsive to p53. Fv fragments alone were not cytotoxic, indicating that killing was due to transduction of p53. Fv-p53 was shown to penetrate cancer cells engrafted in vivo. These results support continued efforts to evaluate the potential efficacy of Fv-p53 for the treatment of certain cancers in vivo.


Assuntos
Morte Celular , Neoplasias/genética , Neoplasias/patologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/farmacologia , Formação de Anticorpos , DNA Complementar , Terapia Genética , Humanos , Fragmentos de Imunoglobulinas/imunologia , Linfocinas , Sialoglicoproteínas , Transdução Genética , Proteína Supressora de Tumor p53/imunologia
18.
Sci Rep ; 4: 5958, 2014 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-25091037

RESUMO

Cancer cells with defects in DNA repair are highly susceptible to DNA-damaging agents, but delivery of therapeutic agents into cell nuclei can be challenging. A subset of lupus autoantibodies is associated with nucleolytic activity, and some of these antibodies are capable of nuclear penetration. We hypothesized that such antibodies might have potential as therapeutic agents targeted towards DNA repair-deficient malignancies. We identified the lupus autoantibody 5C6 as a cell-penetrating nucleolytic antibody and found that 5C6 has a differential effect on a matched pair of BRCA2-proficient and deficient DLD1 colon cancer cells. 5C6 selectively induced γH2AX in, and suppressed the growth of, the BRCA2-deficient cells. These findings demonstrate the potential utility of 5C6 in targeted therapy for DNA repair-deficient malignancies and strengthen the rationale for studies of additional lupus autoantibodies in order to identify the best candidates for development as therapeutic agents. In addition, the toxic effect of 5C6 on BRCA2-deficient cells provides further support for the hypothesis that some lupus autoantibodies contribute to the lower risk of specific cancers associated with systemic lupus erythematosus.


Assuntos
Anticorpos Antinucleares/farmacologia , Antineoplásicos/farmacologia , Proteína BRCA2/deficiência , Núcleo Celular/efeitos dos fármacos , Peptídeos Penetradores de Células/farmacologia , Células Epiteliais/efeitos dos fármacos , Animais , Anticorpos Antinucleares/isolamento & purificação , Anticorpos Antinucleares/metabolismo , Antineoplásicos/isolamento & purificação , Antineoplásicos/metabolismo , Proteína BRCA2/genética , Transporte Biológico , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Núcleo Celular/patologia , Sobrevivência Celular/efeitos dos fármacos , Peptídeos Penetradores de Células/isolamento & purificação , Peptídeos Penetradores de Células/metabolismo , Colo/efeitos dos fármacos , Colo/metabolismo , Colo/patologia , Dano ao DNA , Reparo do DNA/genética , DNA de Neoplasias/genética , DNA de Neoplasias/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Regulação Neoplásica da Expressão Gênica , Histonas/agonistas , Histonas/genética , Histonas/metabolismo , Humanos , Hibridomas/química , Hibridomas/imunologia , Camundongos
19.
Mol Immunol ; 55(3-4): 247-52, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23517740

RESUMO

Rheumatoid arthritis (RA) is a destructive polyarthritis in which synovial-like fibroblasts (SFs) invade and erode cartilage by expressing membrane-anchored type 1 matrix metalloproteinase (MT1-MMP). The mitogen activated protein kinase (MAPK) pathway is activated in RA SFs, but the mechanism of activation is unknown. Here we identify aberrant BRAF splice variants with deletions in both the kinase domain and RAS-binding domain (RBD) in SFs from the majority of RA patients and show that these BRAF splice variants constitutively activate MAPK through CRAF, increase expression of MT1-MMP, and enhance fibroblast invasion of collagen.


Assuntos
Artrite Reumatoide/genética , Artrite Reumatoide/metabolismo , Sistema de Sinalização das MAP Quinases , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas B-raf/metabolismo , Proteínas Proto-Oncogênicas c-raf/metabolismo , Adulto , Idoso , Processamento Alternativo/imunologia , Animais , Artrite Reumatoide/imunologia , Feminino , Fibroblastos/imunologia , Fibroblastos/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases/imunologia , Masculino , Metaloproteinase 14 da Matriz/metabolismo , Camundongos , Pessoa de Meia-Idade , Células NIH 3T3 , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogênicas B-raf/química , Proteínas Proto-Oncogênicas c-raf/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Membrana Sinovial/imunologia , Membrana Sinovial/metabolismo
20.
Mol Cancer Ther ; 11(10): 2169-73, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22863609

RESUMO

The therapeutic use of antibodies is restricted by the limited access of antibodies to intracellular compartments. To overcome this limitation, we developed a cell-penetrating monoclonal antibody, mAb 3E10, as an intracellular delivery vehicle for the intracellular and intranuclear delivery of antibodies constructed as bispecific single-chain Fv fragments. Because MDM2 is an important target in cancer therapy, we selected monoclonal antibody (mAb) 3G5 for intracellular transport. mAb 3G5 binds MDM2 and blocks binding of MDM2 to p53. Here, we show that the resulting 3E10-3G5 bispecific antibody retains cell-penetrating and MDM2-binding activity, increases tumor p53 levels, and inhibits growth of MDM2-addicted tumors. The use of cell-penetrating bispecific antibodies in targeted molecular therapy will significantly broaden the spectrum of accessible intracellular targets and may have a profound impact in cancer therapy.


Assuntos
Anticorpos Biespecíficos/farmacologia , Peptídeos Penetradores de Células/farmacologia , Espaço Intracelular/metabolismo , Terapia de Alvo Molecular , Animais , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Humanos , Espaço Intracelular/efeitos dos fármacos , Melanoma/patologia , Camundongos , Camundongos Nus , Ligação Proteica/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA