Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neuron ; 109(14): 2275-2291.e8, 2021 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-34133945

RESUMO

C9orf72 repeat expansions cause inherited amyotrophic lateral sclerosis (ALS)/frontotemporal dementia (FTD) and result in both loss of C9orf72 protein expression and production of potentially toxic RNA and dipeptide repeat proteins. In addition to ALS/FTD, C9orf72 repeat expansions have been reported in a broad array of neurodegenerative syndromes, including Alzheimer's disease. Here we show that C9orf72 deficiency promotes a change in the homeostatic signature in microglia and a transition to an inflammatory state characterized by an enhanced type I IFN signature. Furthermore, C9orf72-depleted microglia trigger age-dependent neuronal defects, in particular enhanced cortical synaptic pruning, leading to altered learning and memory behaviors in mice. Interestingly, C9orf72-deficient microglia promote enhanced synapse loss and neuronal deficits in a mouse model of amyloid accumulation while paradoxically improving plaque clearance. These findings suggest that altered microglial function due to decreased C9orf72 expression directly contributes to neurodegeneration in repeat expansion carriers independent of gain-of-function toxicities.


Assuntos
Envelhecimento/metabolismo , Amiloide/metabolismo , Proteína C9orf72/metabolismo , Microglia/metabolismo , Sinapses/metabolismo , Envelhecimento/genética , Envelhecimento/patologia , Amiloide/genética , Animais , Proteína C9orf72/genética , Expansão das Repetições de DNA , Modelos Animais de Doenças , Lisossomos/metabolismo , Camundongos , Camundongos Knockout , Sinapses/patologia
2.
J Neuroeng Rehabil ; 18(1): 89, 2021 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-34039346

RESUMO

BACKGROUND: Cerebellar electrical stimulation has shown promise in improving motor recovery post-stroke in both rodent and human studies. Past studies have used motor evoked potentials (MEPs) to evaluate how cerebellar stimulation modulates ongoing activity in the cortex, but the underlying mechanisms are incompletely understood. Here we used invasive electrophysiological recordings from the intact and stroke-injured rodent primary motor cortex (M1) to assess how epidural cerebellar stimulation modulates neural dynamics at the level of single neurons as well as at the level of mesoscale dynamics. METHODS: We recorded single unit spiking and local field potentials (LFPs) in both the intact and acutely stroke-injured M1 contralateral to the stimulated cerebellum in adult Long-Evans rats under anesthesia. We analyzed changes in the firing rates of single units, the extent of synchronous spiking and power spectral density (PSD) changes in LFPs during and post-stimulation. RESULTS: Our results show that post-stimulation, the firing rates of a majority of M1 neurons changed significantly with respect to their baseline rates. These firing rate changes were diverse in character, as the firing rate of some neurons increased while others decreased. Additionally, these changes started to set in during stimulation. Furthermore, cross-correlation analysis showed a significant increase in coincident firing amongst neuronal pairs. Interestingly, this increase in synchrony was unrelated to the direction of firing rate change. We also found that neuronal ensembles derived through principal component analysis were more active post-stimulation. Lastly, these changes occurred without a significant change in the overall spectral power of LFPs post-stimulation. CONCLUSIONS: Our results show that cerebellar stimulation caused significant, long-lasting changes in the activity patterns of M1 neurons by altering firing rates, boosting neural synchrony and increasing neuronal assemblies' activation strength. Our study provides evidence that cerebellar stimulation can directly modulate cortical dynamics. Since these results are present in the perilesional cortex, our data might also help explain the facilitatory effects of cerebellar stimulation post-stroke.


Assuntos
Cerebelo/fisiopatologia , Estimulação Elétrica/métodos , Neurônios/fisiologia , Acidente Vascular Cerebral/fisiopatologia , Animais , Eletrocorticografia , Potencial Evocado Motor/fisiologia , Masculino , Ratos , Ratos Long-Evans
4.
Nature ; 585(7823): 96-101, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32814898

RESUMO

Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are neurodegenerative disorders that overlap in their clinical presentation, pathology and genetic origin. Autoimmune disorders are also overrepresented in both ALS and FTD, but this remains an unexplained epidemiologic observation1-3. Expansions of a hexanucleotide repeat (GGGGCC) in the C9orf72 gene are the most common cause of familial ALS and FTD (C9-ALS/FTD), and lead to both repeat-containing RNA and dipeptide accumulation, coupled with decreased C9orf72 protein expression in brain and peripheral blood cells4-6. Here we show in mice that loss of C9orf72 from myeloid cells alone is sufficient to recapitulate the age-dependent lymphoid hypertrophy and autoinflammation seen in animals with a complete knockout of C9orf72. Dendritic cells isolated from C9orf72-/- mice show marked early activation of the type I interferon response, and C9orf72-/- myeloid cells are selectively hyperresponsive to activators of the stimulator of interferon genes (STING) protein-a key regulator of the innate immune response to cytosolic DNA. Degradation of STING through the autolysosomal pathway is diminished in C9orf72-/- myeloid cells, and blocking STING suppresses hyperactive type I interferon responses in C9orf72-/- immune cells as well as splenomegaly and inflammation in C9orf72-/- mice. Moreover, mice lacking one or both copies of C9orf72 are more susceptible to experimental autoimmune encephalitis, mirroring the susceptibility to autoimmune diseases seen in people with C9-ALS/FTD. Finally, blood-derived macrophages, whole blood and brain tissue from patients with C9-ALS/FTD all show an elevated type I interferon signature compared with samples from people with sporadic ALS/FTD; this increased interferon response can be suppressed with a STING inhibitor. Collectively, our results suggest that patients with C9-ALS/FTD have an altered immunophenotype because their reduced levels of C9orf72 cannot suppress the inflammation mediated by the induction of type I interferons by STING.


Assuntos
Proteína C9orf72/genética , Proteína C9orf72/metabolismo , Inflamação/metabolismo , Inflamação/prevenção & controle , Proteínas de Membrana/metabolismo , Células Mieloides/metabolismo , Envelhecimento/imunologia , Esclerose Lateral Amiotrófica/genética , Animais , Proteína C9orf72/deficiência , Células Dendríticas/citologia , Células Dendríticas/imunologia , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/imunologia , Feminino , Humanos , Inflamação/genética , Inflamação/imunologia , Interferon Tipo I/biossíntese , Interferon Tipo I/imunologia , Proteínas de Membrana/antagonistas & inibidores , Camundongos , Células Mieloides/imunologia , Neoplasias/imunologia , Linfócitos T/citologia , Linfócitos T/imunologia
5.
J Clin Invest ; 129(4): 1756-1771, 2019 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-30882371

RESUMO

Mitofusin-2 (MFN2) is a mitochondrial outer-membrane protein that plays a pivotal role in mitochondrial dynamics in most tissues, yet mutations in MFN2, which cause Charcot-Marie-Tooth disease type 2A (CMT2A), primarily affect the nervous system. We generated a transgenic mouse model of CMT2A that developed severe early onset vision loss and neurological deficits, axonal degeneration without cell body loss, and cytoplasmic and axonal accumulations of fragmented mitochondria. While mitochondrial aggregates were labeled for mitophagy, mutant MFN2 did not inhibit Parkin-mediated degradation, but instead had a dominant negative effect on mitochondrial fusion only when MFN1 was at low levels, as occurs in neurons. Finally, using a transgenic approach, we found that augmenting the level of MFN1 in the nervous system in vivo rescued all phenotypes in mutant MFN2R94Q-expressing mice. These data demonstrate that the MFN1/MFN2 ratio is a key determinant of tissue specificity in CMT2A and indicate that augmentation of MFN1 in the nervous system is a viable therapeutic strategy for the disease.


Assuntos
Axônios/metabolismo , Doença de Charcot-Marie-Tooth/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Animais , Axônios/patologia , Doença de Charcot-Marie-Tooth/genética , Doença de Charcot-Marie-Tooth/patologia , Doença de Charcot-Marie-Tooth/prevenção & controle , Modelos Animais de Doenças , GTP Fosfo-Hidrolases/genética , Camundongos , Camundongos Transgênicos , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
6.
Ann Clin Transl Neurol ; 5(2): 186-200, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29468179

RESUMO

Objective: To investigate transplantation of rat Schwann cells or human iPSC-derived neural crest cells and derivatives into models of acquired and inherited peripheral myelin damage. Methods: Primary cultured rat Schwann cells labeled with a fluorescent protein for monitoring at various times after transplantation. Human-induced pluripotent stem cells (iPSCs) were differentiated into neural crest stem cells, and subsequently toward a Schwann cell lineage via two different protocols. Cell types were characterized using flow cytometry, immunocytochemistry, and transcriptomics. Rat Schwann cells and human iPSC derivatives were transplanted into (1) nude rats pretreated with lysolecithin to induce demyelination or (2) a transgenic rat model of dysmyelination due to PMP22 overexpression. Results: Rat Schwann cells transplanted into sciatic nerves with either toxic demyelination or genetic dysmyelination engrafted successfully, and migrated longitudinally for relatively long distances, with more limited axial migration. Transplanted Schwann cells engaged existing axons and displaced dysfunctional Schwann cells to form normal-appearing myelin. Human iPSC-derived neural crest stem cells and their derivatives shared similar engraftment and migration characteristics to rat Schwann cells after transplantation, but did not further differentiate into Schwann cells or form myelin. Interpretation: These results indicate that cultured Schwann cells surgically delivered to peripheral nerve can engraft and form myelin in either acquired or inherited myelin injury, as proof of concept for pursuing cell therapy for diseases of peripheral nerve. However, lack of reliable technology for generating human iPSC-derived Schwann cells for transplantation therapy remains a barrier in the field.

7.
Science ; 351(6279): 1324-9, 2016 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-26989253

RESUMO

Expansions of a hexanucleotide repeat (GGGGCC) in the noncoding region of the C9orf72 gene are the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia. Decreased expression of C9orf72 is seen in expansion carriers, suggesting that loss of function may play a role in disease. We found that two independent mouse lines lacking the C9orf72 ortholog (3110043O21Rik) in all tissues developed normally and aged without motor neuron disease. Instead, C9orf72 null mice developed progressive splenomegaly and lymphadenopathy with accumulation of engorged macrophage-like cells. C9orf72 expression was highest in myeloid cells, and the loss of C9orf72 led to lysosomal accumulation and altered immune responses in macrophages and microglia, with age-related neuroinflammation similar to C9orf72 ALS but not sporadic ALS human patient tissue. Thus, C9orf72 is required for the normal function of myeloid cells, and altered microglial function may contribute to neurodegeneration in C9orf72 expansion carriers.


Assuntos
Esclerose Lateral Amiotrófica/imunologia , Demência Frontotemporal/imunologia , Fatores de Troca do Nucleotídeo Guanina/fisiologia , Macrófagos/imunologia , Microglia/imunologia , Células Mieloides/imunologia , Proteínas/fisiologia , Envelhecimento/imunologia , Esclerose Lateral Amiotrófica/genética , Animais , Proteína C9orf72 , Demência Frontotemporal/genética , Técnicas de Silenciamento de Genes , Fatores de Troca do Nucleotídeo Guanina/genética , Heterozigoto , Humanos , Doenças Linfáticas/genética , Doenças Linfáticas/imunologia , Camundongos , Camundongos Knockout , Proteínas/genética , Ratos , Esplenomegalia/genética , Esplenomegalia/imunologia
8.
Neuron ; 88(5): 892-901, 2015 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-26637796

RESUMO

Noncoding expansions of a hexanucleotide repeat (GGGGCC) in the C9orf72 gene are the most common cause of familial amyotrophic lateral sclerosis and frontotemporal dementia. Here we report transgenic mice carrying a bacterial artificial chromosome (BAC) containing the full human C9orf72 gene with either a normal allele (15 repeats) or disease-associated expansion (∼100-1,000 repeats; C9-BACexp). C9-BACexp mice displayed pathologic features seen in C9orf72 expansion patients, including widespread RNA foci and repeat-associated non-ATG (RAN) translated dipeptides, which were suppressed by antisense oligonucleotides targeting human C9orf72. Nucleolin distribution was altered, supporting that either C9orf72 transcripts or RAN dipeptides promote nucleolar dysfunction. Despite early and widespread production of RNA foci and RAN dipeptides in C9-BACexp mice, behavioral abnormalities and neurodegeneration were not observed even at advanced ages, supporting the hypothesis that RNA foci and RAN dipeptides occur presymptomatically and are not sufficient to drive neurodegeneration in mice at levels seen in patients.


Assuntos
Esclerose Lateral Amiotrófica/patologia , Encéfalo/patologia , Expansão das Repetições de DNA/genética , Demência Frontotemporal/patologia , Proteínas/genética , Medula Espinal/patologia , Fatores Etários , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/fisiopatologia , Animais , Encéfalo/metabolismo , Proteína C9orf72 , Células Cultivadas , Cromossomos Artificiais Bacterianos/genética , Cromossomos Artificiais Bacterianos/metabolismo , Modelos Animais de Doenças , Demência Frontotemporal/genética , Demência Frontotemporal/fisiopatologia , Ácido Glutâmico/farmacologia , Humanos , Camundongos , Camundongos Transgênicos , Atividade Motora/genética , Força Muscular/genética , Junção Neuromuscular/genética , Junção Neuromuscular/patologia , Neurônios/efeitos dos fármacos , Desempenho Psicomotor/fisiologia , Medula Espinal/metabolismo
9.
Neurotherapeutics ; 11(3): 623-35, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24752661

RESUMO

In human gliomas, the RTK/RAS/PI(3)K signaling pathway is nearly always altered. We present a model of experimental gliomagenesis that elucidates the contributions of genes involved in this pathway (PDGF-B ligand, HRAS-G12V, and AKT). We also examine the effect on gliomagenesis by the potential modifier gene, IDH1-R132H. Injections of lentiviral-encoded oncogenes induce de novo gliomas of varying penetrance, tumor progression, and histological grade depending on the specific oncogenes used. Our model mimics hallmark histological structures of high-grade glioma, such as pseudopalisades, glomeruloid microvascular proliferation, and diffuse tumor invasion. We use our model of gliomagenesis to test the efficacy of an experimental brain tumor gene therapy. Our model allowed us to test the contributions of oncogenes in the RTK/RAS/PI(3)K pathway, and their potential modification by over-expression of mutated IDH1, in glioma development and progression in rats. Our model constitutes a clinically relevant system to study gliomagenesis, the effects of modifier genes, and the efficacy of experimental therapeutics.


Assuntos
Neoplasias Encefálicas/genética , Glioma/genética , Isocitrato Desidrogenase/genética , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-sis/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Animais , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/terapia , Modelos Animais de Doenças , Terapia Genética , Glioma/mortalidade , Glioma/terapia , Humanos , Lentivirus/genética , Mutação , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Análise de Sobrevida
10.
Sci Transl Med ; 5(208): 208ra149, 2013 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-24154603

RESUMO

Amyotrophic lateral sclerosis (ALS) is a severe neurodegenerative condition characterized by loss of motor neurons in the brain and spinal cord. Expansions of a hexanucleotide repeat (GGGGCC) in the noncoding region of the C9ORF72 gene are the most common cause of the familial form of ALS (C9-ALS), as well as frontotemporal lobar degeneration and other neurological diseases. How the repeat expansion causes disease remains unclear, with both loss of function (haploinsufficiency) and gain of function (either toxic RNA or protein products) proposed. We report a cellular model of C9-ALS with motor neurons differentiated from induced pluripotent stem cells (iPSCs) derived from ALS patients carrying the C9ORF72 repeat expansion. No significant loss of C9ORF72 expression was observed, and knockdown of the transcript was not toxic to cultured human motor neurons. Transcription of the repeat was increased, leading to accumulation of GGGGCC repeat-containing RNA foci selectively in C9-ALS iPSC-derived motor neurons. Repeat-containing RNA foci colocalized with hnRNPA1 and Pur-α, suggesting that they may be able to alter RNA metabolism. C9-ALS motor neurons showed altered expression of genes involved in membrane excitability including DPP6, and demonstrated a diminished capacity to fire continuous spikes upon depolarization compared to control motor neurons. Antisense oligonucleotides targeting the C9ORF72 transcript suppressed RNA foci formation and reversed gene expression alterations in C9-ALS motor neurons. These data show that patient-derived motor neurons can be used to delineate pathogenic events in ALS.


Assuntos
Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/patologia , Expansão das Repetições de DNA/genética , Células-Tronco Pluripotentes Induzidas/patologia , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Proteínas/genética , RNA/metabolismo , Proteína C9orf72 , Éxons/genética , Técnicas de Silenciamento de Genes , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Neurônios Motores/efeitos dos fármacos , Oligonucleotídeos Antissenso/farmacologia , RNA/biossíntese , RNA/genética , Transcrição Gênica/efeitos dos fármacos
11.
Hum Gene Ther Methods ; 23(4): 271-84, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22950971

RESUMO

Adenoviral vectors (Ads) have been evaluated in clinical trials for glioma. However, systemic immunity against the vectors can hamper therapeutic efficacy. We demonstrated that combined immunostimulation and cytotoxic gene therapy provides long-term survival in preclinical glioma models. Because helper-dependent high-capacity Ads (HC-Ads) elicit sustained transgene expression, in the presence of antiadenoviral immunity, we engineered HC-Ads encoding conditional cytotoxic herpes simplex type 1 thymidine kinase and immunostimulatory cytokine Fms-like tyrosine kinase ligand-3 under the control of the TetOn system. Escalating doses of combined HC-Ads (1×10(8), 1×10(9), and 1×10(10) viral particles [VP]) were delivered into the rat brain. We assessed neuropathology, biodistribution, transgene expression, systemic toxicity, and behavioral impact at acute and chronic time points after vector delivery. Histopathological analysis did not reveal any evidence of toxicity or long-term inflammation at the lower doses tested. Vector genomes were restricted to the injection site. Serum chemistry did not uncover adverse systemic side effects at any of the doses tested. Taken together, our data indicate that doses of up to 1×10(9) VP of each HC-Ad can be safely administered into the normal brain. This comprehensive toxicity and biodistribution study will lay the foundations for implementation of a phase 1 clinical trial for GBM using HC-Ads.


Assuntos
Adenoviridae/genética , Encéfalo/metabolismo , Vetores Genéticos/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Análise Química do Sangue , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Ensaios Clínicos Fase I como Assunto , Modelos Animais de Doenças , Vetores Genéticos/genética , Vetores Genéticos/toxicidade , Glioma/terapia , Herpesvirus Humano 1/enzimologia , Humanos , Masculino , Ratos , Ratos Endogâmicos Lew , Timidina Quinase/genética , Distribuição Tecidual , Transdução Genética , Tirosina Quinase 3 Semelhante a fms/genética
12.
Neoplasia ; 14(8): 757-70, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22952428

RESUMO

Adenovirus-mediated delivery of the immune-stimulatory cytokine Flt3L and the conditionally cytotoxic thymidine kinase (TK) induces tumor regression and long-term survival in preclinical glioma (glioblastoma multiforme [GBM]) models. Flt3L induces expansion and recruitment of plasmacytoid dendritic cells (pDCs) into the brain. Although pDCs can present antigen and produce powerful inflammatory cytokines, that is, interferon α (IFN-α), their role in tumor immunology remains debated. Thus, we studied the role of pDCs and IFN-α in Ad.TK/GCV+ Ad.Flt3L-mediated anti-GBM therapeutic efficacy. Our data indicate that the combined gene therapy induced recruitment of plasmacytoid DCs (pDCs) into the tumor mass; which were capable of in vivo phagocytosis, IFN-α release, and T-cell priming. Thus, we next used either pDCs or an Ad vector encoding IFN-α delivered within the tumor microenvironment. When rats were treated with Ad.TK/GCV in combination with pDCs or Ad-IFN-α, they exhibited 35% and 50% survival, respectively. However, whereas intracranial administration of Ad.TK/GCV + Ad.Flt3L exhibited a high safety profile, Ad-IFN-α led to severe local inflammation, with neurologic and systemic adverse effects. To elucidate whether the efficacy of the immunotherapy was dependent on IFN-α-secreting pDCs, we administered an Ad vector encoding B18R, an IFN-α antagonist, which abrogated the antitumoral effect of Ad.TK/GCV + Ad.Flt3L. Our data suggest that IFN-α release by activated pDCs plays a critical role in the antitumor effect mediated by Ad.TK/GCV + Ad.Flt3L. In summary, taken together, our results demonstrate that pDCs mediate anti-GBM therapeutic efficacy through the production of IFN-α, thus manipulation of pDCs constitutes an attractive new therapeutic target for the treatment of GBM.


Assuntos
Neoplasias Encefálicas/imunologia , Células Dendríticas/imunologia , Glioblastoma/imunologia , Interferon-alfa/imunologia , Microambiente Tumoral , Adenoviridae/genética , Animais , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Terapia Genética , Glioblastoma/patologia , Glioblastoma/terapia , Imunoterapia , Ativação Linfocitária , Teste de Cultura Mista de Linfócitos , Linfócitos do Interstício Tumoral/imunologia , Proteínas de Membrana/genética , Ratos , Linfócitos T/imunologia , Timidina Quinase/genética
13.
Neurotherapeutics ; 9(4): 827-43, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22996231

RESUMO

Immune-mediated gene therapy using adenovirus expressing Flt3 ligand and thymidine kinase followed by ganciclovir administration (Flt3/TK) effectively elicits tumor regression in preclinical glioma models. Herein, we assessed new strategies to optimize Flt3L/TK therapeutic efficacy in a refractory RG2 orthotopic glioblastoma model. Specifically, we aimed to optimize the therapeutic efficacy of Flt3L/TK treatment in the RG2 model by overexpressing the following genes within the brain tumor microenvironment: 1) a TK mutant with enhanced cytotoxicity (SR39 mutant TK), 2) Flt3L-IgG fusion protein that has a longer half-life, 3) CD40L to stimulate DC maturation, 4) T helper cell type 1 polarizing dendritic cell cytokines interleukin-12 or C-X-C motif ligand 10 chemokine (CXCL)-10, 5) C-C motif ligand 2 chemokine (CCL2) or C-C motif ligand 3 chemokine (CCL3) to enhance dendritic cell recruitment into the tumor microenvironment, 6) T helper cell type 1 cytokines interferon-γ or interleukin-2 to enhance effector T-cell functions, and 7) IκBα or p65RHD (nuclear factor kappa-B [NF-κB] inhibitors) to suppress the function of Foxp3+ Tregs and enhanced effector T-cell functions. Anti-tumor immunity and tumor specific effector T-cell functions were assessed by cytotoxic T lymphocyte assay and intracellular IFN-γ staining. Our data showed that overexpression of interferon-γ or interleukin-2, or inhibition of the nuclear factor kappa-B within the tumor microenvironment, enhanced cytotoxic T lymphocyte-mediated immune responses and successfully extended the median survival of rats bearing intracranial RG2 when combined with Flt3L/TK. These findings indicate that enhancement of T-cell functions constitutes a critical therapeutic target to overcome immune evasion and enhance therapeutic efficacy for brain cancer. In addition, our study provides novel targets to be used in combination with immune-therapeutic strategies for glioblastoma, which are currently being tested in the clinic.


Assuntos
Neoplasias Encefálicas/terapia , Terapia Genética/métodos , Glioblastoma/terapia , Imunoterapia/métodos , Transdução de Sinais , Linfócitos T/imunologia , Adenoviridae/genética , Animais , Antivirais/uso terapêutico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/imunologia , Células Dendríticas/imunologia , Modelos Animais de Doenças , Ganciclovir/uso terapêutico , Vetores Genéticos , Glioblastoma/genética , Glioblastoma/imunologia , Humanos , Interleucina-2/imunologia , Linfócitos do Interstício Tumoral/imunologia , Proteínas de Membrana/uso terapêutico , NF-kappa B/imunologia , Ratos , Proteínas Recombinantes/uso terapêutico , Timidina Quinase/uso terapêutico , Microambiente Tumoral/imunologia
14.
Neoplasia ; 13(10): 947-60, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22028620

RESUMO

We have demonstrated that modifying the tumor microenvironment through intratumoral administration of adenoviral vectors (Ad) encoding the conditional cytotoxic molecule, i.e., HSV1-TK and the immune-stimulatory cytokine, i.e., fms-like tyrosine kinase 3 ligand (Flt3L) leads to T-cell-dependent tumor regression in rodent models of glioblastoma. We investigated the role of B cells during immune-mediated glioblastoma multiforme regression. Although treatment with Ad-TK+Ad-Flt3L induced tumor regression in 60% of wild-type (WT) mice, it completely failed in B-cell-deficient Igh6(-/-) mice. Tumor-specific T-cell precursors were detected in Ad-TK+Ad-Flt3L-treated WT mice but not in Igh6(-/-) mice. The treatment also failed in WT mice depleted of total B cells or marginal zone B cells. Because we could not detect circulating antibodies against tumor cells and the treatment was equally efficient in WT mice and in mice with B-cell-specific deletion of Prdm 1 (encoding Blimp-1), in which B cells are present but unable to fully differentiate into antibody-secreting plasma cells, tumor regression in this model is not dependent on B cells' production of tumor antigen-specific immunoglobulins. Instead, B cells seem to play a role as antigen-presenting cells (APCs). Treatment with Ad-TK+Ad-Flt3L led to an increase in the number of B cells in the cervical lymph nodes, which stimulated the proliferation of syngeneic T cells and induced clonal expansion of antitumor T cells. Our data show that B cells act as APCs, playing a critical role in clonal expansion of tumor antigen-specific T cells and brain tumor regression.


Assuntos
Linfócitos B/imunologia , Neoplasias Encefálicas/terapia , Terapia Genética/métodos , Glioblastoma/terapia , Linfócitos T/imunologia , Animais , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Linfócitos B/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Citotoxicidade Imunológica/imunologia , Feminino , Glioblastoma/genética , Glioblastoma/patologia , Herpesvirus Humano 1/enzimologia , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microscopia Confocal , Fator 1 de Ligação ao Domínio I Regulador Positivo , Linfócitos T/metabolismo , Timidina Quinase/genética , Timidina Quinase/imunologia , Timidina Quinase/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/imunologia , Fatores de Transcrição/metabolismo
15.
Clin Cancer Res ; 17(14): 4705-18, 2011 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21632862

RESUMO

PURPOSE: Glioblastoma multiforme (GBM) is a deadly primary brain tumor. Clinical trials for GBM using dendritic cell (DC) vaccination resulted in antitumor immune responses. Herein, we tested the hypothesis that combining in situ (intratumoral) Ad-Flt3L/Ad-TK-mediated gene therapy with DC vaccination would increase therapeutic efficacy and antitumor immunity. EXPERIMENTAL DESIGN: We first assessed the immunogenicity of tumor lysates generated by Ad-TK (+GCV), temozolomide (TMZ), or freeze/thawing cycles (FTC) in a syngeneic brain tumor model. We also assessed phenotypic markers, cytokine release, and phagocytosis of bone marrow-derived DCs generated by fms-like tyrosine kinase 3 ligand (Flt3L) + IL-6 or by granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin (IL) 4. Inhibition of tumor progression and production of anti-GBM antibodies was assessed following vaccination with (i) tumor cell lysates, (ii) DCs generated with either Flt3L/IL-6 or GM-CSF/IL-4 loaded with either Ad-TK/GCV-, TMZ-, or FTC-generated tumor lysates, or (iii) DCs in combination with in situ Ad-Flt3L/Ad-TK gene therapy. RESULTS: DCs loaded with tumor cell lysates generated with either Ad-TK/GCV or TMZ led to increased levels of phagocytosis, therapeutic efficacy, and humoral immune response. In situ immunogene therapy in combination with DC vaccination led to brain tumor regression and long-term survival in about 90% of animals, a significant increase when compared with either therapy alone. CONCLUSIONS: Our results indicate that modifying the tumor microenvironment using intratumoral Ad-Flt3L/Ad-TK-mediated gene therapy potentiates therapeutic efficacy and antitumor immunity induced by DC vaccination. These data support novel phase I clinical trials to assess the safety and efficacy of this combined approach.


Assuntos
Neoplasias Encefálicas/terapia , Vacinas Anticâncer/uso terapêutico , Células Dendríticas/imunologia , Glioblastoma/terapia , Microambiente Tumoral/imunologia , Adenoviridae/genética , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/imunologia , Vacinas Anticâncer/imunologia , Morte Celular/imunologia , Linhagem Celular Tumoral , Ensaios Clínicos como Assunto , Citocinas/metabolismo , Células Dendríticas/transplante , Modelos Animais de Doenças , Regulação Viral da Expressão Gênica , Terapia Genética , Vetores Genéticos/genética , Glioblastoma/genética , Glioblastoma/imunologia , Ratos , Timidina Quinase/genética , Timidina Quinase/metabolismo , Microambiente Tumoral/genética
16.
Curr Gene Ther ; 11(3): 155-80, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21453286

RESUMO

The most common primary brain tumor in adults is glioblastoma. These tumors are highly invasive and aggressive with a mean survival time of 15-18 months from diagnosis to death. Current treatment modalities are unable to significantly prolong survival in patients diagnosed with glioblastoma. As such, glioma is an attractive target for developing novel therapeutic approaches utilizing gene therapy. This review will examine the available preclinical models for glioma including xenographs, syngeneic and genetic models. Several promising therapeutic targets are currently being pursued in pre-clinical investigations. These targets will be reviewed by mechanism of action, i.e., conditional cytotoxic, targeted toxins, oncolytic viruses, tumor suppressors/oncogenes, and immune stimulatory approaches. Preclinical gene therapy paradigms aim to determine which strategies will provide rapid tumor regression and long-term protection from recurrence. While a wide range of potential targets are being investigated preclinically, only the most efficacious are further transitioned into clinical trial paradigms. Clinical trials reported to date are summarized including results from conditionally cytotoxic, targeted toxins, oncolytic viruses and oncogene targeting approaches. Clinical trial results have not been as robust as preclinical models predicted; this could be due to the limitations of the GBM models employed. Once this is addressed, and we develop effective gene therapies in models that better replicate the clinical scenario, gene therapy will provide a powerful approach to treat and manage brain tumors.


Assuntos
Neoplasias Encefálicas/terapia , Terapia Genética/métodos , Glioma/terapia , Toxinas Biológicas/uso terapêutico , Animais , Neoplasias Encefálicas/genética , Terapia Combinada , Marcação de Genes , Glioma/genética , Humanos , Imunoterapia , Interferons/uso terapêutico , Interleucinas/uso terapêutico , Modelos Biológicos
17.
Mol Ther ; 19(10): 1793-801, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21505426

RESUMO

Glioblastoma multiforme (GBM) is a primary brain tumor with a median survival of 14.6 months postdiagnosis. The infiltrative nature of GBM prevents complete resection and residual brain tumor cells give rise to recurrent GBM, a hallmark of this disease. Recurrent GBMs are known to harbor numerous mutations/gene rearrangements when compared to the primary tumor, which leads to the potential expression of novel proteins that could serve as tumor neoantigens. We have developed a combined immune-based gene therapeutic approach for GBM using adenoviral (Ads) mediated gene delivery of Herpes Simplex Virus Type 1-thymidine kinase (TK) into the tumor mass to induce tumor cells' death combined with an adenovirus expressing fms-like tyrosine kinase 3 ligand (Flt3L) to recruit dendritic cells (DCs) into the tumor microenvironment. This leads to the induction of specific anti-brain tumor immunity and immunological memory. In a model of GBM recurrence, we demonstrate that Flt3L/TK mediated immunological memory is capable of recognizing brain tumor neoantigens absent from the original treated tumor. These data demonstrate that the Flt3L/TK gene therapeutic approach can induce systemic immunological memory capable of recognizing a brain tumor neoantigen in a model of recurrent GBM.


Assuntos
Antígenos de Neoplasias/imunologia , Neoplasias Encefálicas/terapia , Terapia Genética , Glioblastoma/terapia , Timidina Quinase/genética , Tirosina Quinase 3 Semelhante a fms/genética , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Glioblastoma/imunologia , Glioblastoma/patologia , Humanos , Memória Imunológica , Interferon gama/metabolismo , Linfócitos T/imunologia
18.
Discov Med ; 10(53): 293-304, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21034670

RESUMO

Glioblastoma multiforme (GBM) is a deadly primary brain tumor in adults, with a median survival of ~12-18 months post-diagnosis. Despite recent advances in conventional therapeutic approaches, only modest improvements in median survival have been achieved; GBM usually recurs within 12 months post-resection, with poor prognosis. Thus, novel therapeutic strategies to target and kill GBM cells are desperately needed. Our group and others are pursuing virotherapy and gene therapy strategies for the treatment of GBM. In this review, we will discuss various virotherapy and gene therapy approaches for GBM currently under pre-clinical and clinical evaluation including direct or conditional cytotoxic, and/or immunostimulatory approaches. We also discuss cutting-edge technologies for drug/gene delivery and targeting brain tumors, including the use of stem cells as delivery platforms, the use of targeted immunotoxins, and the therapeutic potential of using GBM microvesicles to deliver therapeutic siRNAs or virotherapies. Finally, various animal models available to test novel GBM therapies are discussed.


Assuntos
Neoplasias Encefálicas/terapia , Terapia Genética/métodos , Glioblastoma/terapia , Terapia Viral Oncolítica/métodos , Terapias em Estudo/métodos , Adulto , Animais , Terapia Genética/tendências , Humanos , Modelos Biológicos , Terapia Viral Oncolítica/tendências , Terapias em Estudo/tendências
19.
Proc Natl Acad Sci U S A ; 107(46): 20021-6, 2010 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-21030678

RESUMO

Restricting the cytotoxicity of anticancer agents by targeting receptors exclusively expressed on tumor cells is critical when treating infiltrative brain tumors such as glioblastoma multiforme (GBM). GBMs express an IL-13 receptor (IL13Rα2) that differs from the physiological IL4R/IL13R receptor. We developed a regulatable adenoviral vector (Ad.mhIL-4.TRE.mhIL-13-PE) encoding a mutated human IL-13 fused to Pseudomonas exotoxin (mhIL-13-PE) that specifically binds to IL13Rα2 to provide sustained expression, effective anti-GBM cytotoxicity, and minimal neurotoxicity. The therapeutic Ad also encodes mutated human IL-4 that binds to the physiological IL4R/IL13R without interacting with IL13Rα2, thus inhibiting potential binding of mhIL-13-PE to normal brain cells. Using intracranial GBM xenografts and syngeneic mouse models, we tested the Ad.mhIL-4.TRE.mhIL-13-PE and two protein formulations, hIL-13-PE used in clinical trials (Cintredekin Besudotox) and a second-generation mhIL-13-PE. Cintredekin Besudotox doubled median survival without eliciting long-term survival and caused severe neurotoxicity; mhIL-13-PE led to ∼40% long-term survival, eliciting severe neurological toxicity at the high dose tested. In contrast, Ad-mediated delivery of mhIL-13-PE led to tumor regression and long-term survival in over 70% of the animals, without causing apparent neurotoxicity. Although Cintredekin Besudotox was originally developed to target GBM, when tested in a phase III trial it failed to achieve clinical endpoints and revealed neurotoxicity. Limitations of Cintredekin Besudotox include its short half-life, which demanded frequent or continued administration, and binding to IL4R/IL13R, present in normal brain cells. These shortcomings were overcome by our therapeutic Ad, thus representing a significant advance in the development of targeted therapeutics for GBM.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Citotoxinas/genética , Citotoxinas/uso terapêutico , Técnicas de Transferência de Genes , Terapia Genética , Glioma/tratamento farmacológico , Adenoviridae/genética , Animais , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Exotoxinas/genética , Exotoxinas/uso terapêutico , Vetores Genéticos/genética , Glioma/patologia , Humanos , Imunocompetência/imunologia , Interleucina-13/genética , Interleucina-13/uso terapêutico , Camundongos , Camundongos Nus , Mutação/genética , Neurotoxinas/toxicidade , Pseudomonas/metabolismo , Transgenes/genética , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
20.
PLoS One ; 5(6): e11074, 2010 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-20552015

RESUMO

BACKGROUND: Glioblastoma multiforme (GBM) is the most common primary brain tumor in adults and carries a dismal prognosis. We have developed a conditional cytotoxic/immunotherapeutic approach using adenoviral vectors (Ads) encoding the immunostimulatory cytokine, human soluble fms-like tyrosine kinase 3 ligand (hsFlt3L) and the conditional cytotoxic molecule, i.e., Herpes Simplex Type 1- thymide kinase (TK). This therapy triggers an anti-tumor immune response that leads to tumor regression and anti-tumor immunological memory in intracranial rodent cancer models. We aim to test the efficacy of this immunotherapy in dogs bearing spontaneous GBM. In view of the controversy regarding the effect of human cytokines on dog immune cells, and considering that the efficacy of this treatment depends on hsFlt3L-stimulated dendritic cells (DCs), in the present work we tested the ability of Ad-encoded hsFlt3L to generate DCs from dog peripheral blood and compared its effects with canine IL-4 and GM-CSF. METHODOLOGY/PRINCIPAL FINDINGS: Our results demonstrate that hsFlT3L expressed form an Ad vector, generated DCs from peripheral blood cultures with very similar morphological and phenotypic characteristics to canine IL-4 and GM-CSF-cultured DCs. These include phagocytic activity and expression of CD11c, MHCII, CD80 and CD14. Maturation of DCs cultured under both conditions resulted in increased secretion of IL-6, TNF-alpha and IFN-gamma. Importantly, hsFlt3L-derived antigen presenting cells showed allostimulatory potential highlighting their ability to present antigen to T cells and elicit their proliferation. CONCLUSIONS/SIGNIFICANCE: These results demonstrate that hsFlt3L induces the proliferation of canine DCs and support its use in upcoming clinical trials for canine GBM. Our data further support the translation of hsFlt3L to be used for dendritic cells' vaccination and gene therapeutic approaches from rodent models to canine patients and its future implementation in human clinical trials.


Assuntos
Neoplasias Encefálicas/veterinária , Células Dendríticas/imunologia , Doenças do Cão/terapia , Glioma/veterinária , Imunoterapia , Proteínas de Membrana/imunologia , Animais , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/terapia , Proliferação de Células , Células Cultivadas , Doenças do Cão/imunologia , Cães , Vetores Genéticos , Glioma/imunologia , Glioma/terapia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/administração & dosagem , Herpesvirus Humano 1/genética , Interleucina-4/administração & dosagem , Ativação Linfocitária , Proteínas de Membrana/genética , Fagocitose
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...