Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Biochim Biophys Acta ; 1865(2): 237-44, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26957416

RESUMO

Cellular senescence is defined by an irreversible growth arrest and is an important biological mechanism for suppression of tumor formation. Although deletion/mutation to DNA sequences is one mechanism by which cancer cells can escape senescence, little is known about the epigenetic factors contributing to this process. Histone modifications and chromatin remodeling related to the function of a histone demethylase, jumonji domain-containing protein 3 (JMJD3; also known as KDM6B), play an important role in development, tissue regeneration, stem cells, inflammation, and cellular senescence and aging. The role of JMJD3 in cancer is poorly understood and its function may be at the intersection of many pathways promoted in a dysfunctional manner such as activation of the senescence-associated secretory phenotype (SASP) observed in aging.


Assuntos
Senescência Celular , Histona Desmetilases com o Domínio Jumonji/fisiologia , Neoplasias/patologia , Reprogramação Celular , Epigênese Genética , Humanos , Neoplasias/genética
2.
Nat Rev Cancer ; 5(11): 899-904, 2005 11.
Artigo em Inglês | MEDLINE | ID: mdl-16327766

RESUMO

Most tumours are derived from a single cell that is transformed into a cancer-initiating cell (cancer stem cell) that has the capacity to proliferate and form tumours in vivo. However, the origin of the cancer stem cell remains elusive. Interestingly, during development and tissue repair the fusion of genetic and cytoplasmic material between cells of different origins is an important physiological process. Such cell fusion and horizontal gene-transfer events have also been linked to several fundamental features of cancer and could be important in the development of the cancer stem cell.


Assuntos
Transformação Celular Neoplásica/patologia , Neoplasias/patologia , Células-Tronco/patologia , Animais , Fusão Celular , Transformação Celular Neoplásica/genética , Transferência Genética Horizontal/fisiologia , Humanos , Neoplasias/genética
3.
Stem Cells ; 30(2): 314-25, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22084033

RESUMO

Metastasis to multiple organs is the primary cause of mortality in breast cancer patients. The poor prognosis for patients with metastatic breast cancer and toxic side effects of currently available treatments necessitate the development of effective tumor-selective therapies. Neural stem cells (NSCs) possess inherent tumor tropic properties that enable them to overcome many obstacles of drug delivery that limit effective chemotherapy strategies for breast cancer. We report that increased NSC tropism to breast tumor cell lines is strongly correlated with the invasiveness of cancer cells. Interleukin 6 (IL-6) was identified as a major cytokine mediating NSC tropism to invasive breast cancer cells. We show for the first time in a preclinical mouse model of metastatic human breast cancer that NSCs preferentially target tumor metastases in multiple organs, including liver, lung, lymph nodes, and femur, versus the primary intramammary fat pad tumor. For proof-of-concept of stem cell-mediated breast cancer therapy, NSCs were genetically modified to secrete rabbit carboxylesterase (rCE), an enzyme that activates the CPT-11 prodrug to SN-38, a potent topoisomerase I inhibitor, to effect tumor-localized chemotherapy. In vitro data demonstrate that exposure of breast cancer cells to conditioned media from rCE-secreting NSCs (NSC.rCE) increased their sensitivity to CPT-11 by 200-fold. In vivo, treatment of tumor-bearing mice with NSC.rCE cells in combination with CPT-11 resulted in reduction of metastatic tumor burden in lung and lymph nodes. These data suggest that NSC-mediated enzyme/prodrug therapy may be more effective and less toxic than currently available chemotherapy strategies for breast cancer metastases.


Assuntos
Antineoplásicos Fitogênicos/uso terapêutico , Camptotecina/análogos & derivados , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/patologia , Células-Tronco Neurais/transplante , Pró-Fármacos/uso terapêutico , Animais , Antineoplásicos Fitogênicos/administração & dosagem , Antineoplásicos Fitogênicos/farmacocinética , Biotransformação , Camptotecina/administração & dosagem , Camptotecina/farmacocinética , Camptotecina/uso terapêutico , Carboxilesterase/biossíntese , Carboxilesterase/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Sobrevivência Celular/efeitos dos fármacos , Sistemas de Liberação de Medicamentos , Feminino , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Irinotecano , Neoplasias Pulmonares/tratamento farmacológico , Metástase Linfática , Neoplasias Mamárias Experimentais/tratamento farmacológico , Camundongos , Camundongos Nus , Invasividade Neoplásica , Células-Tronco Neurais/enzimologia , Células-Tronco Neurais/metabolismo , Pró-Fármacos/administração & dosagem , Pró-Fármacos/farmacocinética , Coelhos , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Biochim Biophys Acta ; 1816(2): 191-8, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21767610

RESUMO

Antibodies and antibody conjugates have emerged as important tools for cancer therapy. However, a major therapeutic challenge for the use of antibodies is their inability to cross the blood-brain barrier (BBB) to reach tumors localized in the central nervous system (CNS). Multiple methods have been developed to enhance antibody delivery to the CNS, including direct injection, mechanical or biochemical disruption of the BBB, conjugation to a 'molecular Trojan horse', cationization, encapsulation in nanoparticles and liposomes, and more recently, stem cell-mediated antibody delivery. In this review, we discuss each of these approaches, highlighting their successes and the obstacles that remain to be overcome.


Assuntos
Anticorpos/administração & dosagem , Encéfalo/metabolismo , Animais , Anticorpos/metabolismo , Barreira Hematoencefálica , Difusão , Humanos , Lipossomos , Nanopartículas , Células-Tronco/fisiologia , Transcitose
5.
Stem Cells ; 28(11): 2084-7, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21089119

RESUMO

Monoclonal antibodies are important tools for cancer therapy, however, three factors limit their effectiveness: toxicity, poor tumor penetration, and inability to cross the blood-brain barrier. This review discusses the emerging field of stem cell-mediated antibody delivery and how this approach may improve antibody therapy of cancer by overcoming these obstacles.


Assuntos
Anticorpos/imunologia , Neoplasias/terapia , Células-Tronco/metabolismo , Animais , Humanos , Modelos Biológicos , Neoplasias/imunologia , Células-Tronco/citologia
6.
Mol Cancer Res ; 6(12): 1819-29, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19074827

RESUMO

Hypoxia is a critical aspect of the microenvironment in glioma and generally signifies unfavorable clinical outcome. Effective targeting of hypoxic areas in gliomas remains a significant therapeutic challenge. New therapeutic platforms using neural stem cells (NSC) for tumor-targeted drug delivery show promise in treatment of cancers that are refractory to traditional therapies. However, the molecular mechanisms of NSC targeting to hypoxic tumor areas are not well understood. Therefore, we investigated the role of hypoxia in directed migration of NSCs to glioma and identified the specific signaling molecules involved. Our data showed that hypoxia caused increased migration of human HB1.F3 NSCs to U251 human glioma-conditioned medium in vitro. In HB1.F3 NSCs, hypoxia led to up-regulation of CXCR4, urokinase-type plasminogen activator receptor (uPAR), vascular endothelial growth factor receptor 2 (VEGFR2), and c-Met receptors. Function-inhibiting antibodies to these receptors inhibited the migration of HB1.F3 cells to glioma-conditioned medium. Small interfering RNA knockdown of hypoxia-inducible factor-1alpha in glioma cells blocked the hypoxia-induced migration of NSCs, which was due to decreased expression of stromal cell-derived factor-1 (SDF-1), uPA, and VEGF in glioma cells. Our in vivo data provided direct evidence that NSCs preferentially distributed to hypoxic areas inside intracranial glioma xenografts, as detected by pimonidazole hypoxia probe, as well as to the tumor edge, and that both areas displayed high SDF-1 expression. These observations indicate that hypoxia is a key factor in determining NSC tropism to glioma and that SDF-1/CXCR4, uPA/uPAR, VEGF/VEGFR2, and hepatocyte growth factor/c-Met signaling pathways mediate increased NSC-to-glioma tropism under hypoxia. These results have significant implications for development of stem cell-mediated tumor-selective gene therapies.


Assuntos
Neoplasias Encefálicas/terapia , Movimento Celular/fisiologia , Glioma/terapia , Hipóxia/patologia , Células-Tronco/citologia , Animais , Especificidade de Anticorpos , Neoplasias Encefálicas/patologia , Linhagem Celular Transformada , Meios de Cultivo Condicionados , Citocinas/genética , Citocinas/imunologia , Sistemas de Liberação de Medicamentos/métodos , Feminino , Regulação Neoplásica da Expressão Gênica , Terapia Genética/métodos , Glioma/patologia , Humanos , Hipóxia/fisiopatologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Camundongos , Camundongos Nus , Testes de Neutralização , RNA Interferente Pequeno , Transplante de Células-Tronco , Células-Tronco/imunologia , Telencéfalo/citologia , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Stem Cells ; 26(1): 292-8, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17962698

RESUMO

This report presents highlights of discussions that focused on the biology of cancer stem cells as conducted at the fifth Annual Meeting of the International Society for Stem Cell Research, held in Cairns, Australia, June 17-20, 2007. The function of adult stem cells is believed to depend on their niches, that is, the microenvironment in which these stem cells reside. A similar concept applies to understanding the development of cancer, as it is becoming increasingly clear that only a small subset of cancer cell populations is capable of initiating/sustaining tumor formation. These tumorigenic cells, commonly referred to as cancer stem cells, also appear to reside in particular niches, and they bear the known, albeit dysfunctional, stem cell characteristics of self-renewal and differentiation. Dysregulation of stem cell niches is thought to contribute to tumorigenesis by affecting the complex network of signaling interactions that occur between stem cells and their neighboring cells, thus imbalancing the physiological controls on self-renewal and differentiation processes. This hypothesis was widely explored at the conference to shed new light on the mechanisms of tumor origin and progression and to unveil novel antitumor therapeutic approaches.


Assuntos
Células-Tronco Adultas , Células-Tronco Neoplásicas , Animais , Epigênese Genética , Humanos
8.
Stem Cells ; 26(6): 1406-13, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18403751

RESUMO

Human neural and mesenchymal stem cells have been identified for cell-based therapies in regenerative medicine and as vehicles for delivering therapeutic agents to areas of injury and tumors. However, the signals required for homing and recruitment of stem cells to these sites are not well understood. Urokinase plasminogen activator (uPA) and urokinase plasminogen activator receptor (uPAR) are involved in chemotaxis and cell guidance during normal development and are upregulated in invasive tumors. Here we provided evidence that activation of uPA and uPAR in malignant solid tumors (brain, lung, prostate, and breast) augments neural and mesenchymal stem cell tropism. Expression levels of uPAR on human solid tumor cell lines correlated with levels of uPA and soluble uPAR in tumor cell-conditioned media. Cytokine expression profiles of these tumor-conditioned media were determined by protein arrays. Among 79 cytokines investigated, interleukin (IL)-6, IL-8, and monocyte chemoattractant protein-1 were the most highly expressed cytokines in uPAR-positive tumors. We provided evidence that human recombinant uPA induced stem cell migration, whereas depletion of uPA from PC-3 prostate cancer cell-conditioned medium blocked stem cell migration. Furthermore, retrovirus-mediated overexpression of uPA and uPAR in neuroblastoma (NB1691) cells induced robust migration of stem cells toward NB1691 cell-conditioned media, compared with media derived from wild-type NB1691 cells. We conclude that expression of uPA and uPAR in cancer cells underlies a novel mechanism of stem cell tropism to malignant solid tumors, which may be important for development of optimal stem cell-based therapies. Disclosure of potential conflicts of interest is found at the end of this article.


Assuntos
Células-Tronco Mesenquimais/fisiologia , Neoplasias/fisiopatologia , Receptores de Superfície Celular/fisiologia , Células-Tronco/fisiologia , Ativador de Plasminogênio Tipo Uroquinase/fisiologia , Neoplasias Encefálicas/fisiopatologia , Neoplasias da Mama/fisiopatologia , Linhagem Celular Tumoral , Feminino , Humanos , Neoplasias Pulmonares/fisiopatologia , Masculino , Mesencéfalo/embriologia , Mesencéfalo/fisiopatologia , Células-Tronco Mesenquimais/citologia , Neuroblastoma , Reação em Cadeia da Polimerase , Neoplasias da Próstata/fisiopatologia , Receptores de Superfície Celular/genética , Receptores de Ativador de Plasminogênio Tipo Uroquinase , Células-Tronco/citologia , Ativador de Plasminogênio Tipo Uroquinase/genética
9.
Stem Cells ; 26(6): 1575-86, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18339768

RESUMO

The utility of neural stem cells (NSCs) has extended beyond regenerative medicine to targeted gene delivery, as NSCs possess an inherent tropism to solid tumors, including invasive gliomas. However, for optimal clinical implementation, an understanding of the molecular events that regulate NSC tumor tropism is needed to ensure their safety and to maximize therapeutic efficacy. We show that human NSC lines responded to multiple tumor-derived growth factors and that hepatocyte growth factor (HGF) induced the strongest chemotactic response. Gliomatropism was critically dependent on c-Met signaling, as short hairpin RNA-mediated ablation of c-Met significantly attenuated the response. Furthermore, inhibition of Ras-phosphoinositide 3-kinase (PI3K) signaling impaired the migration of human neural stem cells (hNSCs) toward HGF and other growth factors. Migration toward tumor cells is a highly regulated process, in which multiple growth factor signals converge on Ras-PI3K, causing direct modification of the cytoskeleton. The signaling pathways that regulate hNSC migration are similar to those that promote unregulated glioma invasion, suggesting shared cellular mechanisms and responses. Disclosure of potential conflicts of interest is found at the end of this article.


Assuntos
Neoplasias Encefálicas/patologia , Glioma/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatidilinositol 3-Quinases/fisiologia , Neoplasias Encefálicas/enzimologia , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular , Quimiotaxia , Glioma/enzimologia , Humanos , Rim , Neurônios/enzimologia , Neurônios/fisiologia , Células-Tronco/enzimologia , Células-Tronco/fisiologia , Proteínas rac1 de Ligação ao GTP/metabolismo
10.
Cancer Res ; 67(1): 22-5, 2007 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17210679

RESUMO

Neural stem cells and progenitor cells migrate selectively to tumor loci in vivo. We exploited the tumor-tropic properties of HB1.F3.C1 cells, an immortalized cell line derived from human fetal telencephalon, to deliver the cDNA encoding a secreted form of rabbit carboxylesterase (rCE) to disseminated neuroblastoma tumors in mice. This enzyme activates the prodrug CPT-11 more efficiently than do human enzymes. Mice bearing multiple tumors were treated with rCE-expressing HB1.F3.C1 cells and schedules of administration of CPT-11 that produced levels of active drug (SN-38) tolerated by patients. Both HB1.F3.C1 cells and CPT-11 were given i.v. None of the untreated mice and 30% of mice that received only CPT-11 survived long term. In contrast, 90% of mice treated with rCE-expressing HB1.F3.C1 cells and 15 mg/kg CPT-11 survived for 1 year without detectable tumors. Plasma carboxylesterase activity and SN-38 levels in mice receiving both rCE-expressing HB1.F3.C1 cells (HB1.F3.C1/AdCMVrCE) and CPT-11 were comparable with those in mice receiving CPT-11 only. These data support the hypothesis that the antitumor effect of the described neural stem/progenitor cell-directed enzyme prodrug therapy (NDEPT) is mediated by production of high concentrations of active drug selectively at tumor sites, thereby maximizing the antitumor effect of CPT-11. NDEPT approaches merit further investigation as effective, targeted therapy for metastatic tumors. We propose that the described approach may have greatest use for eradicating minimum residual disease.


Assuntos
Camptotecina/análogos & derivados , Carboxilesterase/metabolismo , Terapia Genética/métodos , Neuroblastoma/terapia , Pró-Fármacos/farmacologia , Telencéfalo/fisiologia , Adenoviridae/genética , Animais , Antineoplásicos Fitogênicos/farmacocinética , Antineoplásicos Fitogênicos/farmacologia , Camptotecina/farmacocinética , Camptotecina/farmacologia , Carboxilesterase/biossíntese , Carboxilesterase/genética , Linhagem Celular Tumoral , Terapia Combinada , DNA Complementar/administração & dosagem , DNA Complementar/genética , Intervalo Livre de Doença , Humanos , Irinotecano , Camundongos , Neuroblastoma/tratamento farmacológico , Neuroblastoma/enzimologia , Neuroblastoma/genética , Pró-Fármacos/farmacocinética , Telencéfalo/citologia , Telencéfalo/enzimologia , Transdução Genética , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Clin Cancer Res ; 23(12): 2951-2960, 2017 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-27979915

RESUMO

Purpose: Human neural stem cells (NSC) are inherently tumor tropic, making them attractive drug delivery vehicles. Toward this goal, we retrovirally transduced an immortalized, clonal NSC line to stably express cytosine deaminase (HB1.F3.CD.C21; CD-NSCs), which converts the prodrug 5-fluorocytosine (5-FC) to 5-fluorouracil (5-FU).Experimental Design: Recurrent high-grade glioma patients underwent intracranial administration of CD-NSCs during tumor resection or biopsy. Four days later, patients began taking oral 5-FC every 6 hours for 7 days. Study treatment was given only once. A standard 3 + 3 dose escalation schema was used to increase doses of CD-NSCs from 1 × 107 to 5 × 107 and 5-FC from 75 to 150 mg/kg/day. Intracerebral microdialysis was performed to measure brain levels of 5-FC and 5-FU. Serial blood samples were obtained to assess systemic drug concentrations as well as to perform immunologic correlative studies.Results: Fifteen patients underwent study treatment. We saw no dose-limiting toxicity (DLT) due to the CD-NSCs. There was 1 DLT (grade 3 transaminitis) possibly related to 5-FC. We did not see development of anti-CD-NSC antibodies and did not detect CD-NSCs or replication-competent retrovirus in the systemic circulation. Intracerebral microdialysis revealed that CD-NSCs produced 5-FU locally in the brain in a 5-FC dose-dependent manner. Autopsy data indicate that CD-NSCs migrated to distant tumor sites and were nontumorigenic.Conclusions: Collectively, our results from this first-in-human study demonstrate initial safety and proof of concept regarding the ability of NSCs to target brain tumors and locally produce chemotherapy. Clin Cancer Res; 23(12); 2951-60. ©2016 AACR.


Assuntos
Citosina Desaminase/genética , Terapia Genética , Glioma/tratamento farmacológico , Células-Tronco Neurais/transplante , Adolescente , Adulto , Citosina Desaminase/administração & dosagem , Citosina Desaminase/efeitos adversos , Feminino , Flucitosina/administração & dosagem , Fluoruracila/administração & dosagem , Técnicas de Transferência de Genes , Vetores Genéticos , Glioma/genética , Glioma/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Gradação de Tumores
12.
Neuro Oncol ; 8(2): 119-26, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16524944

RESUMO

Brain metastases are an increasingly frequent and serious clinical problem for cancer patients, especially those with advanced melanoma. Given the extensive tropism of neural stem/progenitor cells (NSPCs) for pathological areas in the central nervous system, we expanded investigations to determine whether NSPCs could also target multiple sites of brain metastases in a syngeneic experimental melanoma model. Using cytosine deaminase-expressing NSPCs (CD-NSPCs) and systemic 5-fluorocytosine (5-FC) pro-drug administration, we explored their potential as a cell-based targeted drug delivery system to disseminated brain metastases. Our results indicate a strong tropism of NSPCs for intracerebral melanoma metastases. Furthermore, in our therapeutic paradigm, animals with established melanoma brain metastasis received intracranial implantation of CD-NSPCs followed by systemic 5-FC treatment, resulting in a significant (71%) reduction in tumor burden. These data provide proof of principle for the use of NSPCs for targeted delivery of therapeutic gene products to melanoma brain metastases.


Assuntos
Neoplasias Encefálicas/secundário , Neoplasias Encefálicas/terapia , Melanoma Experimental/secundário , Melanoma Experimental/terapia , Neurônios/transplante , Transplante de Células-Tronco , Animais , Linhagem Celular Tumoral , Imuno-Histoquímica , Camundongos , Transplante de Neoplasias
13.
Stem Cell Reports ; 7(3): 483-495, 2016 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-27546534

RESUMO

Pre-clinical studies indicate that neural stem cells (NSCs) can limit or reverse CNS damage through direct cell replacement, promotion of regeneration, or delivery of therapeutic agents. Immortalized NSC lines are in growing demand due to the inherent limitations of adult patient-derived NSCs, including availability, expandability, potential for genetic modifications, and costs. Here, we describe the generation and characterization of a new human fetal NSC line, immortalized by transduction with L-MYC (LM-NSC008) that in vitro displays both self-renewal and multipotent differentiation into neurons, oligodendrocytes, and astrocytes. These LM-NSC008 cells were non-tumorigenic in vivo, and migrated to orthotopic glioma xenografts in immunodeficient mice. When administered intranasally, LM-NSC008 distributed specifically to sites of traumatic brain injury (TBI). These data support the therapeutic development of immortalized LM-NSC008 cells for allogeneic use in TBI and other CNS diseases.


Assuntos
Diferenciação Celular/genética , Autorrenovação Celular/genética , Expressão Gênica , Genes myc , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Animais , Lesões Encefálicas Traumáticas/metabolismo , Lesões Encefálicas Traumáticas/patologia , Lesões Encefálicas Traumáticas/terapia , Movimento Celular/genética , Proliferação de Células , Transformação Celular Neoplásica/genética , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Estudo de Associação Genômica Ampla , Xenoenxertos , Humanos , Camundongos , Células-Tronco Neurais/patologia , Transplante de Células-Tronco , Transcriptoma , Transdução Genética , Transgenes
14.
J Neurotrauma ; 22(3): 382-97, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15785233

RESUMO

We have previously described the activation of the classical, alternative, and terminal complement cascade pathways after acute contusion spinal cord injury using the New York University (NYU) weight-drop impactor. In the present study, we examined the induction of protein regulators of the complement cascade, factor H (FH), and clusterin, in the same experimental paradigm. The spinal cord of laminectomized adult rats was subjected to mild or severe injury using impactor weight-drop heights of 12.5 and 50 mm, respectively. The spinal cords of control and injured animals were evaluated at 1, 7, and 42 days after injury. Immunocytochemistry revealed a robust increase in the numbers and intensity of staining of FH, and clusterin-positive cells in the injured cord at all three time points, with the highest increases observed at 1 and 42 days after injury. FH and clusterin-positive cells were observed among neurons as well as oligodendrocytes. The increased expression was detected both rostrally and caudally from the injury site, in the latter case at distances up to 20 mm. The precise biological significance of injury-induced upregulation of these proteins remains to be determined. However, FH and clusterin are potent regulators of complement activity targeting upstream (FH) and downstream (clusterin) molecules of the pro-inflammatory cascade, which could be of vital importance in preventing a "runaway" inflammatory reaction in the injured spinal cord.


Assuntos
Proteínas Inativadoras do Complemento/biossíntese , Neuroglia/metabolismo , Neurônios/metabolismo , Traumatismos da Medula Espinal/metabolismo , Regulação para Cima/fisiologia , Animais , Neuroglia/patologia , Neurônios/patologia , Ratos , Ratos Long-Evans , Traumatismos da Medula Espinal/patologia , Vértebras Torácicas
15.
Artigo em Inglês | MEDLINE | ID: mdl-26654402

RESUMO

The depletion of stem cell pools and the accumulation of senescent cells in animal tissues are linked to aging. Planarians are invertebrate flatworms and are unusual in that their stem cells, called neoblasts, are constantly replacing old and dying cells. By eliminating neoblasts in worms via irradiation, the biological principles of aging are exposed in the absence of wound healing and regeneration, making planaria a powerful tool for aging research.

16.
Mol Cancer Res ; 13(4): 636-50, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25652587

RESUMO

UNLABELLED: Jumonji domain-containing protein 3 (JMJD3/KDM6B) demethylates lysine 27 on histone H3 (H3K27me3), a repressive epigenetic mark controlling chromatin organization and cellular senescence. To better understand the functional consequences of JMJD3 its expression was investigated in brain tumor cells. Querying patient expression profile databases confirmed JMJD3 overexpression in high-grade glioma. Immunochemical staining of two glioma cell lines, U251 and U87, indicated intrinsic differences in JMJD3 expression levels that were reflected in changes in cell phenotype and variations associated with cellular senescence, including senescence-associated ß-galactosidase (SA-ß-gal) activity and the senescence-associated secretory phenotype (SASP). Overexpressing wild-type JMJD3 (JMJD3wt) activated SASP-associated genes, enhanced SA-ß-gal activity, and induced nuclear blebbing. Conversely, overexpression of a catalytically inactive dominant negative mutant JMJD3 (JMJD3mut) increased proliferation. In addition, a large number of transcripts were identified by RNA-seq as altered in JMJD3 overexpressing cells, including cancer- and inflammation-related transcripts as defined by Ingenuity Pathway Analysis. These results suggest that expression of the SASP in the context of cancer undermines normal tissue homeostasis and contributes to tumorigenesis and tumor progression. These studies are therapeutically relevant because inflammatory cytokines have been linked to homing of neural stem cells and other stem cells to tumor loci. IMPLICATIONS: This glioma study brings together actions of a normal epigenetic mechanism (JMJD3 activity) with dysfunctional activation of senescence-related processes, including secretion of SASP proinflammatory cytokines and stem cell tropism toward tumors.


Assuntos
Neoplasias Encefálicas/patologia , Senescência Celular , Glioma/metabolismo , Histona Desmetilases com o Domínio Jumonji/metabolismo , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Citocinas/genética , Citocinas/metabolismo , Epigênese Genética , Glioma/patologia , Humanos , Histona Desmetilases com o Domínio Jumonji/genética , Gradação de Tumores , Células-Tronco Neurais/imunologia , Tropismo
17.
Brain Res ; 926(1-2): 118-25, 2002 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-11814413

RESUMO

DNA fragmentation is a key marker of neuronal death during development, yet little is known about the size, pattern or quantities of fragments generated during normal and sensory-deprived development. Since there are few neurons dying at any particular time, it has not been possible to obtain sufficient quantities of material to make such a determination. By using a highly sensitive Taq polymerase-based technique, we revealed DNA fragments of 180 base pairs and multiples thereof both in bulbs and cortex of young rats (P4-P31). The bulbs subjected to olfactory deprivation at P1 had higher levels of internucleosomal DNA fragmentation at P16 than the contra-lateral, non-deprived bulbs. Interestingly, the DNA fragmentation induced by olfactory deprivation displayed a characteristic internucleosomal fragmentation pattern, suggesting that the cells induced to die may do so by apoptosis. A significant inverse correlation between DNA fragmentation and the natural variation in normal bulb size was found, suggesting that bulb size may be related to cell death.


Assuntos
Fragmentação do DNA , Nucleossomos/patologia , Bulbo Olfatório/patologia , Bulbo Olfatório/fisiologia , Privação Sensorial/fisiologia , Animais , Córtex Cerebral/patologia , Feminino , Masculino , Tamanho do Órgão , Ratos , Ratos Wistar
18.
Pathol Oncol Res ; 20(4): 789-97, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25168767

RESUMO

Malignant gliomas are among the deadliest primary brain tumors. Despite multimodal therapy and advances in chemotherapy, imaging, surgical and radiation techniques, these tumors remain virtually incurable. Glioma stem cells may be responsible for resistance to traditional therapies and tumor recurrence. Therefore, elimination of glioma stem cells may be crucial for achieving therapeutic efficacy. Metformin, a small molecule drug widely used in the therapy of type 2 diabetes, has shown significant anti-tumor effects in patients with breast cancer and prostate cancer. Recent preclinical data suggest that metformin also has therapeutic effects against glioma. Here we review the markers and hallmarks of glioma stem cells, and the molecular mechanisms involved in therapeutic targeting of glioma stem cells by metformin.


Assuntos
Biomarcadores Tumorais/análise , Glioma/tratamento farmacológico , Glioma/patologia , Hipoglicemiantes/uso terapêutico , Metformina/uso terapêutico , Células-Tronco Neoplásicas/efeitos dos fármacos , Humanos , Células-Tronco Neoplásicas/patologia , Prognóstico
19.
Stem Cells Transl Med ; 2(12): 983-92, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24167321

RESUMO

CPT-11 (irinotecan) has been investigated as a treatment for malignant brain tumors. However, limitations of CPT-11 therapy include low levels of the drug entering brain tumor sites and systemic toxicities associated with higher doses. Neural stem cells (NSCs) offer a novel way to overcome these obstacles because of their inherent tumor tropism and ability to cross the blood-brain barrier, which enables them to selectively target brain tumor sites. Carboxylesterases (CEs) are enzymes that can convert the prodrug CPT-11 (irinotecan) to its active metabolite SN-38, a potent topoisomerase I inhibitor. We have adenovirally transduced an established clonal human NSC line (HB1.F3.CD) to express a rabbit carboxylesterase (rCE) or a modified human CE (hCE1m6), which are more effective at converting CPT-11 to SN-38 than endogenous human CE. We hypothesized that NSC-mediated CE/CPT-11 therapy would allow tumor-localized production of SN-38 and significantly increase the therapeutic efficacy of irinotecan. Here, we report that transduced NSCs transiently expressed high levels of active CE enzymes, retained their tumor-tropic properties, and mediated an increase in the cytotoxicity of CPT-11 toward glioma cells. CE-expressing NSCs (NSC.CEs), whether administered intracranially or intravenously, delivered CE to orthotopic human glioma xenografts in mice. NSC-delivered CE catalyzed conversion of CPT-11 to SN-38 locally at tumor sites. These studies demonstrate the feasibility of NSC-mediated delivery of CE to glioma and lay the foundation for translational studies of this therapeutic paradigm to improve clinical outcome and quality of life in patients with malignant brain tumors.


Assuntos
Neoplasias Encefálicas/terapia , Camptotecina/análogos & derivados , Hidrolases de Éster Carboxílico/metabolismo , Glioma/terapia , Células-Tronco Neurais/enzimologia , Células-Tronco Neurais/transplante , Inibidores da Topoisomerase I/farmacologia , Adenoviridae/genética , Animais , Biotransformação , Neoplasias Encefálicas/enzimologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Camptotecina/farmacocinética , Camptotecina/farmacologia , Carboxilesterase/deficiência , Carboxilesterase/genética , Hidrolases de Éster Carboxílico/genética , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Estudos de Viabilidade , Vetores Genéticos , Glioma/enzimologia , Glioma/genética , Glioma/patologia , Humanos , Irinotecano , Camundongos , Camundongos Knockout , Camundongos SCID , Células-Tronco Neurais/efeitos dos fármacos , Coelhos , Fatores de Tempo , Distribuição Tecidual , Inibidores da Topoisomerase I/farmacocinética , Transdução Genética , Transfecção , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Sci Transl Med ; 5(184): 184ra59, 2013 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-23658244

RESUMO

High-grade gliomas are extremely difficult to treat because they are invasive and therefore not curable by surgical resection; the toxicity of current chemo- and radiation therapies limits the doses that can be used. Neural stem cells (NSCs) have inherent tumor-tropic properties that enable their use as delivery vehicles to target enzyme/prodrug therapy selectively to tumors. We used a cytosine deaminase (CD)-expressing clonal human NSC line, HB1.F3.CD, to home to gliomas in mice and locally convert the prodrug 5-fluorocytosine to the active chemotherapeutic 5-fluorouracil. In vitro studies confirmed that the NSCs have normal karyotype, tumor tropism, and CD expression, and are genetically and functionally stable. In vivo biodistribution studies demonstrated NSC retention of tumor tropism, even in mice pretreated with radiation or dexamethasone to mimic clinically relevant adjuvant therapies. We evaluated safety and toxicity after intracerebral administration of the NSCs in non-tumor-bearing and orthotopic glioma-bearing immunocompetent and immunodeficient mice. We detected no difference in toxicity associated with conversion of 5-fluorocytosine to 5-fluorouracil, no NSCs outside the brain, and no histological evidence of pathology or tumorigenesis attributable to the NSCs. The average tumor volume in mice that received HB1.F3.CD NSCs and 5-fluorocytosine was about one-third that of the average volume in control mice. On the basis of these results, we conclude that combination therapy with HB1.F3.CD NSCs and 5-fluorocytosine is safe, nontoxic, and effective in mice. These data have led to approval of a first-in-human study of an allogeneic NSC-mediated enzyme/prodrug-targeted cancer therapy in patients with recurrent high-grade glioma.


Assuntos
Glioma/tratamento farmacológico , Glioma/terapia , Células-Tronco Neurais/citologia , Pró-Fármacos/uso terapêutico , Animais , Linhagem Celular , Citosina Desaminase/metabolismo , Feminino , Citometria de Fluxo , Flucitosina/metabolismo , Flucitosina/uso terapêutico , Fluoruracila/metabolismo , Humanos , Masculino , Camundongos , Camundongos Nus , Células-Tronco Neurais/metabolismo , Pró-Fármacos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA