Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
iScience ; 26(11): 108130, 2023 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-37876795

RESUMO

Parkinson's disease (PD) is characterized by the irreversible loss of dopaminergic neurons and the accumulation of α-synuclein in Lewy bodies. The oligomeric α-synuclein (O-αS) is the most toxic form of α-synuclein species, and it has been reported to be a robust inflammatory mediator. Mutations in Leucine-Rich Repeat Kinase 2 (LRRK2) are also genetically linked to PD and neuroinflammation. However, how O-αS and LRRK2 interact in glial cells remains unclear. Here, we reported that LRRK2 G2019S mutation, which is one of the most frequent causes of familial PD, enhanced the effects of O-αS on astrocytes both in vivo and in vitro. Meanwhile, inhibition of LRRK2 kinase activity could relieve the inflammatory effects of both LRRK2 G2019S and O-αS. We also demonstrated that nuclear factor κB (NF-κB) pathway might be involved in the neuroinflammatory responses. These findings revealed that inhibition of LRRK2 kinase activity may be a viable strategy for suppressing neuroinflammation in PD.

2.
Nat Cancer ; 4(9): 1309-1325, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37550516

RESUMO

To avoid regulatory T cell promotion and vascular toxicity, the interleukin-2 receptor-ß/interleukin-2 receptor-γ (IL-2Rßγ)-biased approach is used by most IL-2 analogs in immuno-oncology. However, recent clinical disappointments in these IL-2 agonists have questioned this strategy. Here we show that both wild-type (IL-2wt) and IL-2Rßγ-attenuated (IL-2α-bias) agonists that preserve IL-2Rα (CD25) activities can effectively expand tumor-specific CD8+ T cells (TSTs) and exhibit better antitumor efficacy and safety than the 'non-α' counterpart (IL-2nα). Mechanistically, TSTs coexpress elevated CD25 and PD-1 and are more susceptible to stimulation by IL-2Rα-proficient agonists. Moreover, the antitumor efficacy of anti-PD-1 depends on activation of PD-1+CD25+ TSTs through autocrine IL-2-CD25 signaling. In individuals with cancer, a low IL-2 signature correlates with non-responsiveness to anti-PD-1 treatment. In mouse models, IL-2α-bias, but not IL-2nα, restores the IL-2 signature and synergizes with anti-PD-1 to eradicate large established tumors. These findings underscore the indispensable function of CD25 in IL-2-based immunotherapy and provide rationales for evaluating IL-2Rα-biased agonists in individuals with cancer.


Assuntos
Linfócitos T CD8-Positivos , Neoplasias , Camundongos , Animais , Subunidade alfa de Receptor de Interleucina-2 , Linfócitos T CD8-Positivos/patologia , Interleucina-2/farmacologia , Receptor de Morte Celular Programada 1 , Neoplasias/tratamento farmacológico
3.
Cancer Immunol Immunother ; 72(2): 493-507, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35963895

RESUMO

High rates of relapse and poor prognosis confer an urgent need for novel therapeutic agents for B cell non-Hodgkin lymphomas (B-NHLs). Herein, we describe a human IgG-like anti-CD79b/CD3 bispecific antibody (IBI38D9-L) that selectively depletes antigen-positive malignant B cells as an alternative treatment option for relapsed or refractory NHL patients. The antitumor activity and mechanism of action of IBI38D9-L were investigated in vitro using B-NHL cell lines and human primary effector cells and in vivo using xenograft models reconstituted with human PBMCs (peripheral blood mononuclear cells). Pharmacokinetic (PK) properties and preclinical toxicology were evaluated in cynomolgus monkeys and HSC-NPG mice. IBI38D9-L exerted potent B cell killing as well as T cell activation and proliferation in a tumor cell-dependent manner in vitro and was active against B-NHL cell lines with various CD79b expression levels. Subcutaneous xenograft tumors in NOG mice engrafted with human PBMCs were eradicated by IBI38D9-L treatment. Moreover, IBI38D9-L-treated mice showed a strong infiltration of activated T cells. In HSC-NPG mice, IBI38D9-L resulted in potent B cell depletion in peripheral blood and induced only slight body weight loss and cytokine release syndrome without significant toxicological findings. In cynomolgus monkeys, IBI38D9-L was well tolerated with good pharmacokinetic profiles. Collectively, these preclinical efficacy and safety data provide strong scientific rationales for using anti-CD79b/CD3 bispecific antibody as a promising therapeutic agent for B cell malignancies.


Assuntos
Anticorpos Biespecíficos , Neoplasias , Humanos , Camundongos , Animais , Macaca fascicularis , Leucócitos Mononucleares , Anticorpos Biespecíficos/farmacologia , Linfócitos B , Neoplasias/metabolismo , Complexo CD3
4.
Cell Rep Med ; 3(6): 100660, 2022 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-35732156

RESUMO

Glucocorticoid-induced tumor necrosis factor receptor (GITR) is a co-stimulatory receptor and an important target for cancer immunotherapy. We herein present a potent FcγR-independent GITR agonist IBI37G5 that can effectively activate effector T cells and synergize with anti-programmed death 1 (PD1) antibody to eradicate established tumors. IBI37G5 depends on both antibody bivalency and GITR homo-dimerization for efficient receptor cross-linking. Functional analyses reveal bell-shaped dose responses due to the unique 2:2 antibody-receptor stoichiometry required for GITR activation. Antibody self-competition is observed after concentration exceeded that of 100% receptor occupancy (RO), which leads to antibody monovalent binding and loss of activity. Retrospective pharmacokinetics/pharmacodynamics analysis demonstrates that the maximal efficacy is achieved at medium doses with drug exposure near saturating GITR occupancy during the dosing cycle. Finally, we propose an alternative dose-finding strategy that does not rely on the traditional maximal tolerated dose (MTD)-based paradigm but instead on utilizing the RO-function relations as biomarker to guide the clinical translation of GITR and similar co-stimulatory agonists.


Assuntos
Glucocorticoides , Receptores de IgG , Linhagem Celular Tumoral , Proteína Relacionada a TNFR Induzida por Glucocorticoide/agonistas , Ligantes , Receptores do Fator de Necrose Tumoral/agonistas , Estudos Retrospectivos , Fatores de Necrose Tumoral
5.
J Parkinsons Dis ; 12(1): 295-314, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34719508

RESUMO

BACKGROUND: Previous investigations have suggested that decreased expression of glutamate transporter-1 (GLT-1) is involved in glutamate excitotoxicity and contribute to the development of Parkinson's disease (PD), GLT-1 is decreased in animal models of PD. GLT-1 is mainly expressed in astrocytes, and the striatum is a GLT-1-rich brain area. OBJECTIVE: The aim was to explore the function and mechanism of astrocytic GLT-1 in PD-like changes. METHODS: In the study, PD-like changes and their molecular mechanism in rodents were tested by a behavioral assessment, micro-positron emission tomography/computed tomography (PET/CT), western blotting, immunohistochemical and immunofluorescence staining, and high performance liquid chromatography pre-column derivatization with O-pthaldialdehida after downregulating astrocytic GLT-1 in vivo and in vitro. RESULTS: In vivo, after 6 weeks of brain stereotactic injection of adeno-associated virus into the striatum, rats in the astrocytic GLT-1 knockdown group showed poorer motor performance, abnormal gait, and depression-like feature; but no olfactory disorders. The results of micro-PET/CT and western blotting indicated that the dopaminergic system was impaired in astrocytic GLT-1 knockdown rats. Similarly, tyrosine hydroxylase (TH) positive immune-staining in neurons of astrocytic GLT-1 knockdown rats showed deficit in cell count. In vitro, knockdown of astrocytic GLT-1 via RNA interference led to morphological injury of TH-positive neurons, which may be related to the abnormal calcium signal induced by glutamate accumulation after GLT-1 knockdown. Furthermore, the GLT-1 agonist ceftriaxone showed a protective effect on TH-positive neuron impairment. CONCLUSION: The present findings may shed new light in the future prevention and treatment of PD based on blocking glutamate excitotoxicity.


Assuntos
Astrócitos , Transportador 2 de Aminoácido Excitatório/metabolismo , Doença de Parkinson , Animais , Astrócitos/metabolismo , Regulação para Baixo , Transportador 2 de Aminoácido Excitatório/genética , Transportador 2 de Aminoácido Excitatório/farmacologia , Ácido Glutâmico/genética , Ácido Glutâmico/metabolismo , Ácido Glutâmico/farmacologia , Humanos , Doença de Parkinson/metabolismo , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Ratos , Tirosina 3-Mono-Oxigenase/genética , Tirosina 3-Mono-Oxigenase/metabolismo , Tirosina 3-Mono-Oxigenase/farmacologia
6.
J Gerontol A Biol Sci Med Sci ; 77(4): 705-716, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-34448826

RESUMO

Early-life stress (ELS) can cause long-term effects on human health, ranging from adolescence to adulthood, and even to gerontic. Although clinical retrospective data suggest that ELS may be related to senile neurodegenerative diseases such as Parkinson's disease (PD), there are few prospective investigations to explore its real contribution to PD. Here, we investigated the behavioral, histochemical, neuromorphological, and transcriptional changes induced by maternal separation (MS), an ELS model. Without neurotoxin, MS rats showed behavioral alterations in olfaction, locomotion, and gait characters after depression compared with control rats. Based on neuroimaging and histochemistry, although we found that the dopaminergic system in the striatum was impaired after MS, the decrease of striatal dopamine level was ~33%. Consistently, tyrosine hydroxylase immunostaining positive neurons of MS rats in the substantia nigra showed deficit by about 20% in cell counting. Furthermore, using transcriptome sequencing, we discovered many differentially expressed genes (DEGs) of MS rats in the striatum significantly enriched in the pathway of dopaminergic synapse, and the biological process of locomotion and neuromuscular process controlling balance. Encouragingly, some representative DEGs relating to PD were singled out. These results suggest that ELS-depression rats potentially mimic some key features of prodromal stage of PD during natural senescence. In conclusion, our findings provide some novel insights into the future pathogenesis and therapeutic studies for PD related to depression.


Assuntos
Transtornos Parkinsonianos , Sintomas Prodrômicos , Estresse Psicológico , Animais , Ratos , Envelhecimento , Privação Materna , Estudos Prospectivos , Estudos Retrospectivos , Substância Negra/metabolismo
7.
Front Pharmacol ; 12: 713963, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34335276

RESUMO

Neurodegenerative disease (NDD), including Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis, are characterized by the progressive loss of neurons which leads to the decline of motor and/or cognitive function. Currently, the prevalence of NDD is rapidly increasing in the aging population. However, valid drugs or treatment for NDD are still lacking. The clinical heterogeneity and complex pathogenesis of NDD pose a great challenge for the development of disease-modifying therapies. Numerous animal models have been generated to mimic the pathological conditions of these diseases for drug discovery. Among them, zebrafish (Danio rerio) models are progressively emerging and becoming a powerful tool for in vivo study of NDD. Extensive use of zebrafish in pharmacology research or drug screening is due to the high conserved evolution and 87% homology to humans. In this review, we summarize the zebrafish models used in NDD studies, and highlight the recent findings on pharmacological targets for NDD treatment. As high-throughput platforms in zebrafish research have rapidly developed in recent years, we also discuss the application prospects of these new technologies in future NDD research.

8.
Stem Cell Res Ther ; 12(1): 210, 2021 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-33762014

RESUMO

Stem cells are characterized by their self-renewal and multipotency and have great potential in the therapy of various disorders. However, the blood-brain barrier (BBB) limits the application of stem cells in the therapy of neurological disorders, especially in a noninvasive way. It has been shown that small molecular substances, macromolecular proteins, and even stem cells can bypass the BBB and reach the brain parenchyma following intranasal administration. Here, we review the possible brain-entry routes of transnasal treatment, the cell types, and diseases involved in intranasal stem cell therapy, and discuss its advantages and disadvantages in the treatment of central nervous system diseases, to provide a reference for the application of intranasal stem cell therapy.


Assuntos
Doenças do Sistema Nervoso Central , Administração Intranasal , Barreira Hematoencefálica , Encéfalo , Doenças do Sistema Nervoso Central/tratamento farmacológico , Sistemas de Liberação de Medicamentos , Humanos , Células-Tronco
9.
Cancer Immunol Immunother ; 70(2): 365-376, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32761423

RESUMO

CD47, an immune checkpoint receptor frequently unregulated in various blood and solid tumors, interacts with ligand SIPRα on innate immune cells, and conveys a "do not eat me" signal to inhibit macrophage-mediated tumor phagocytosis. This makes CD47 a valuable target for cancer immunotherapy. However, the therapeutic utility of CD47-SIRPα blockade monoclonal antibodies is largely compromised due to significant red blood cell (RBCs) toxicities and fast target-mediated clearance as a result of extensive expression of CD47 on normal cells. To overcome these limitations and further improve therapeutic efficacy, we designed IBI322, a CD47/PD-L1 bispecific antibody which attenuated CD47 activity in monovalent binding and blocked PD-L1 activity in bivalent binding. IBI322 selectively bound to CD47+PD-L1+ tumor cells, effectively inhibited CD47-SIRPα signal and triggered strong tumor cell phagocytosis in vitro, but only with minimal impact on CD47 single positive cells such as human RBCs. In addition, as a dual blocker of innate and adaptive immune checkpoints, IBI322 effectively accumulated in PD-L1-positive tumors and demonstrated synergistic activity in inducing complete tumor regression in vivo. Furthermore, IBI322 showed only marginal RBCs depletion and was well tolerated in non-human primates (NHP) after repeated weekly injections, suggesting a sufficient therapeutic window in future clinical development of IBI322 for cancer treatment.


Assuntos
Anticorpos Biespecíficos/uso terapêutico , Antígeno B7-H1/uso terapêutico , Antígeno CD47/antagonistas & inibidores , Imunoterapia/métodos , Neoplasias/tratamento farmacológico , Animais , Anticorpos Biespecíficos/farmacologia , Antígeno B7-H1/farmacologia , Humanos , Camundongos , Camundongos Endogâmicos NOD , Neoplasias/patologia
10.
Neuromolecular Med ; 22(3): 335-340, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-31933131

RESUMO

As a type of stress, maternal separation (MS) has been one of the most widely used models in neuropsychiatric research. An increasing number of studies has found that MS not only affects the function of the hypothalamic-pituitary-adrenal axis and hippocampal 5-hydroxytryptamine system, but also causes dysfunction of the central dopamine (DA) system and increases the susceptibility of dopaminergic neurons to pathogenic factors of Parkinson's disease (PD), for instance, 6-hydroxydopamine, thus impairing motor function. We reviewed the impact of MS on the DA system and its correlation with PD and found the following: (1) discrepant effects of MS on the DA system have been reported; (2) MS is a good model to study the impact of stress on the occurrence and development of PD, however, unified modeling criteria of MS are required; (3) correlation between MS and PD may involve the impact of MS on the DA system, which however is not the only connection; (4) intervening measures can block pathways between MS and PD, which provides reference for the prevention of PD in specific populations such as left-behind children.


Assuntos
Dopamina/fisiologia , Sistema Hipotálamo-Hipofisário/fisiopatologia , Privação Materna , Doença de Parkinson/etiologia , Sistema Hipófise-Suprarrenal/fisiopatologia , Adulto , Experiências Adversas da Infância , Criança , Criança Abandonada , Corpo Estriado/fisiopatologia , Neurônios Dopaminérgicos/patologia , Feminino , Heterogeneidade Genética , Humanos , Modelos Neurológicos , Doença de Parkinson/fisiopatologia , Doença de Parkinson/prevenção & controle , Substância Negra/fisiopatologia
11.
Mol Cancer Ther ; 18(6): 1104-1114, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30962319

RESUMO

Although inhibiting EGFR-mediated signaling proved to be effective in treating certain types of cancers, a quickly evolved mechanism that either restores the EGFR signaling or activates an alternative pathway for driving the proliferation and survival of malignant cells limits the efficacy and utility of the approach via suppressing the EGFR functionality. Given the fact that overexpression of EGFR is commonly seen in many cancers, an EGFR-targeting antibody-drug conjugate (ADC) can selectively kill cancer cells independently of blocking EGFR-mediated signaling. Herein, we describe SHR-A1307, a novel anti-EGFR ADC, generated from an anti-EGFR antibody with prolonged half-life, and conjugated with a proprietary toxin payload that has increased index of EGFR targeting-dependent versus EGFR targeting-independent cytotoxicity. SHR-A1307 demonstrated strong and sustained antitumor activities in EGFR-positive tumors harboring different oncogenic mutations on EGFR, KRAS, or PIK3CA. Antitumor efficacy of SHR-A1307 correlated with EGFR expression levels in vitro and in vivo, regardless of the mutation status of EGFR signaling mediators and a resultant resistance to EGFR signaling inhibitors. Cynomolgus monkey toxicology study showed that SHR-A1307 is well tolerated with a wide therapeutic index. SHR-A1307 is a promising therapeutic option for EGFR-expressing cancers, including those resistant or refractory to the EGFR pathway inhibitors.


Assuntos
Aminobenzoatos/imunologia , Anticorpos Monoclonais Humanizados/imunologia , Antineoplásicos Imunológicos/imunologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Imunoconjugados/farmacologia , Imunoconjugados/uso terapêutico , Neoplasias/tratamento farmacológico , Oligopeptídeos/imunologia , Animais , Anticorpos Monoclonais Humanizados/administração & dosagem , Antineoplásicos Imunológicos/administração & dosagem , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos/métodos , Receptores ErbB/imunologia , Feminino , Células HEK293 , Humanos , Macaca fascicularis , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transfecção , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Int J Cancer ; 134(1): 43-54, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23784558

RESUMO

Cancer initiation and progression have been attributed to newly discovered subpopulations of self-renewing, highly tumorigenic, drug-resistant tumor cells termed cancer stem cells. Recently, we and others reported a new phenotypic plasticity wherein highly tumorigenic, drug-resistant cell populations could arise not only from pre-existing cancer stem-like populations but also from cancer cells lacking these properties. In the current study, we hypothesized that this newfound phenotypic plasticity may be mediated by PI3K/Akt and Wnt/ß-catenin signaling, pathways previously implicated in carcinogenesis, pluripotency and drug resistance. Using GFP expression, Hoechst dye exclusion and fluorescence activated cell sorting (FACS) of cancer cell lines, we identified and tracked cancer stem-like side populations (SP) of cancer cells characterized by high tumorigenicity and drug resistance. We found that pharmacological inhibition or genetic depletion of PI3K and AKT markedly reduced the spontaneous conversion of nonside population (NSP) cells into cancer stem-like SP cells, whereas PI3K/Akt activation conversely enhanced NSP to SP conversion. PI3K/AKT signaling was mediated through downstream phosphorylation of GSK3ß, which led to activation and accumulation of ß-catenin. Accordingly, pharmacological or genetic perturbation of GSK3ß or ß-catenin dramatically impacted conversion of NSP to SP. Further downstream, ß-catenin's effects on NSP-SP equilibrium were dependent upon its interaction with CBP, a KAT3 family coactivator. These studies provide a mechanistic model wherein PI3K/Akt/ß-catenin/CBP signaling mediates phenotypic plasticity in and out of a drug-resistant, highly tumorigenic state. Therefore, targeting this pathway has unique potential for overcoming the therapy resistance and disease progression attributed to the cancer stem-like phenotype.


Assuntos
Proteína de Ligação a CREB/metabolismo , Resistencia a Medicamentos Antineoplásicos/fisiologia , Células-Tronco Neoplásicas/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/fisiologia , beta Catenina/metabolismo , Western Blotting , Linhagem Celular Tumoral , Separação Celular , Citometria de Fluxo , Imunofluorescência , Humanos , Imunoprecipitação , Células-Tronco Neoplásicas/patologia , Fenótipo , RNA Interferente Pequeno , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
13.
Clin Adv Hematol Oncol ; 9(6): 442-55, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21841744

RESUMO

Telomerase plays a key role in cell fate: loss of telomerase in normal differentiated cells heralds senescence and limits cell division, whereas reactivation of telomerase sustains proliferation and potentiates mutagenesis and transformation. Given this pivotal role, telomerase has been the subject of intense investigation in the field of developmental cancer therapeutics. To date, a broad spectrum of therapeutic strategies has been developed, ranging from direct targeting or reprogramming of the enzyme, to immune or virus-mediated targeting of cells expressing telomerase, to strategies focusing on the telomeres themselves. The recent discovery and growing interest in cancer stem cells has thrust telomerase therapy into new relief as an approach that may be uniquely suited to neutralizing this treatment-resistant subpopulation of cancer cells. Here we will review the mechanistic rationale and preclinical and clinical state of development of the various telomerase-based therapeutic approaches, with emphasis on the role of telomerase in cancer stem cell biology and its implications for therapeutic efforts.


Assuntos
Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Células-Tronco Neoplásicas/enzimologia , Telomerase/fisiologia , Animais , Humanos
14.
Mol Cancer Ther ; 10(6): 938-48, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21518726

RESUMO

Drug resistance and brisk tumor initiation have traditionally been viewed as preexisting phenotypes present in small subpopulations of neoplastic cells sometimes termed cancer stem cells. However, recent work in cancer cell lines has shown that drug-resistant tumor-initiating features can emerge de novo within fractionated subpopulations of cells initially lacking these phenotypes. In the present study, we asked whether such phenotypic plasticity exists broadly in unperturbed cancer cell lines and tumor xenografts growing spontaneously without interventions such as drug selection or fractionation into subpopulations used in prior studies. To address this question, we used side population (SP) analysis combined with fluorescence labeling to identify a drug-resistant highly tumorigenic subpopulation and to track and analyze its interaction with the larger phenotypically negative population over time. Remarkably, we observed that SP size fluctuated in a cyclical manner: first contracting via differentiation into the non-SP (NSP) and then reexpanding via simultaneous direct conversion of numerous NSP cells back to the SP phenotype both in culture and in tumor xenografts. These findings show for the first time that adaptive, cancer-promoting traits such as drug resistance and brisk tumor initiation arise not only as solitary events under selective pressures but also as highly orchestrated transitions occurring concurrently in large numbers of cells even without specifically induced drug selection, ectopic gene expression, or fractionation into subpopulations. This high level of coordinated phenotypic plasticity bears consideration when using cancer cell lines as experimental models and may have significant implications for therapeutic efforts targeting cancer stem cells, which are marked by a drug-resistant tumor-initiating phenotype.


Assuntos
Neoplasias/tratamento farmacológico , Neoplasias/patologia , Células-Tronco Neoplásicas/patologia , Células da Side Population/patologia , Animais , Linhagem Celular Tumoral , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Resistencia a Medicamentos Antineoplásicos , Humanos , Masculino , Camundongos , Camundongos SCID , Neoplasias/metabolismo , Células-Tronco Neoplásicas/metabolismo , Fenótipo , Ratos , Células da Side Population/metabolismo
15.
Prostate ; 71(13): 1390-400, 2011 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-21321978

RESUMO

BACKGROUND: Cancer progenitor cells (CPCs) have been postulated to promote treatment resistance and disease progression in prostate and other malignancies. We investigated whether the enzyme telomerase, which is active in cancer cells and in normal stem cells, plays an important role in CPC which can be exploited to neutralize these cells. METHODS: We used flow cytometry and assays of gene expression, clonogenicity, and invasiveness to isolate and characterize a putative CPC subpopulation from freshly resected human prostatectomy specimens. Telomerase activity was measured by qPCR-based Telomeric Repeat Amplification Protocol (TRAP). Telomerase interference was achieved by ectopic expression of a mutated telomerase RNA construct which reprograms telomerase to generate "toxic" uncapped telomeres. Treated cells were assayed for apoptosis, proliferation in culture, and xenograft tumor formation. RESULTS: CPC in prostate tumors expressed elevated levels of genes associated with a progenitor phenotype and were highly clonogenic and invasive. Significantly, CPC telomerase activity was 20- to 200-fold higher than in non-CPC from the same tumors, and CPC were exquisitely sensitive to telomerase interference which induced rapid apoptosis and growth inhibition. Similarly, induction of telomerase interference in highly tumorigenic CPC isolated from a prostate cancer cell line abrogated their ability to form tumor xenografts. CONCLUSIONS: Human prostate tumors contain a CPC subpopulation with markedly elevated telomerase activity which renders them acutely susceptible to telomerase interference. These findings offer the first tumor-derived and in vivo evidence that telomerase may constitute a CPC "Achilles heel" which may ultimately form the basis for more effective new CPC-targeting therapies.


Assuntos
Células-Tronco Neoplásicas/enzimologia , Neoplasias da Próstata/patologia , Telomerase/fisiologia , Animais , Humanos , Receptores de Hialuronatos/análise , Integrina alfa2beta1/análise , Masculino , Camundongos , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/terapia , RNA Interferente Pequeno/genética , Telomerase/antagonistas & inibidores , Telomerase/genética , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...