Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Cell Chem Biol ; 28(10): 1394-1406.e10, 2021 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-33979648

RESUMO

Natural products (NPs) encompass a rich source of bioactive chemical entities. Here, we used human cancer stem cells (CSCs) in a chemical genomics campaign with NP chemical space to interrogate extracts from diverse strains of actinomycete for anti-cancer properties. We identified a compound (McM25044) capable of selectively inhibiting human CSC function versus normal stem cell counterparts. Biochemical and molecular studies revealed that McM025044 exerts inhibition on human CSCs through the small ubiquitin-like modifier (SUMO) cascade, found to be hyperactive in a variety of human cancers. McM025044 impedes the SUMOylation pathway via direct targeting of the SAE1/2 complex. Treatment of patient-derived CSCs resulted in reduced levels of SUMOylated proteins and suppression of progenitor and stem cell capacity measured in vitro and in vivo. Our study overcomes a barrier in chemically inhibiting oncogenic SUMOylation activity and uncovers a unique role for SAE2 in the biology of human cancers.


Assuntos
Células-Tronco Neoplásicas/metabolismo , Enzimas Ativadoras de Ubiquitina/metabolismo , Animais , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Sítios de Ligação , Produtos Biológicos/química , Produtos Biológicos/metabolismo , Produtos Biológicos/farmacologia , Produtos Biológicos/uso terapêutico , Linhagem Celular Tumoral , Autorrenovação Celular , Sobrevivência Celular/efeitos dos fármacos , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/patologia , Camundongos , Simulação de Acoplamento Molecular , Células-Tronco Neoplásicas/citologia , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Sumoilação/efeitos dos fármacos , Enzimas Ativadoras de Ubiquitina/química , Enzimas Ativadoras de Ubiquitina/genética
2.
Cell Rep ; 34(10): 108818, 2021 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-33691101

RESUMO

Histone variants (HVs) are a subfamily of epigenetic regulators implicated in embryonic development, but their role in human stem cell fate remains unclear. Here, we reveal that the phosphorylation state of the HV H2A.X (γH2A.X) regulates self-renewal and differentiation of human pluripotent stem cells (hPSCs) and leukemic progenitors. As demonstrated by CRISPR-Cas deletion, H2A.X is essential in maintaining normal hPSC behavior. However, reduced levels of γH2A.X enhances hPSC differentiation toward the hematopoietic lineage with concomitant inhibition of neural development. In contrast, activation and sustained levels of phosphorylated H2A.X enhance hPSC neural fate while suppressing hematopoiesis. This controlled lineage bias correlates to occupancy of γH2A.X at genomic loci associated with ectoderm versus mesoderm specification. Finally, drug modulation of H2A.X phosphorylation overcomes differentiation block of patient-derived leukemic progenitors. Our study demonstrates HVs may serve to regulate pluripotent cell fate and that this biology could be extended to somatic cancer stem cell control.


Assuntos
Autorrenovação Celular/fisiologia , Histonas/metabolismo , Células-Tronco Neoplásicas/citologia , Células-Tronco Pluripotentes/citologia , Sistemas CRISPR-Cas/genética , Diferenciação Celular , Linhagem da Célula , Ectoderma/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Histonas/deficiência , Histonas/genética , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Mesoderma/metabolismo , Células-Tronco Neoplásicas/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Nucleossomos/metabolismo , Fosforilação , Células-Tronco Pluripotentes/metabolismo
3.
Cancer Cell ; 34(3): 483-498.e5, 2018 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-30205048

RESUMO

Despite successful remission induction, recurrence of acute myeloid leukemia (AML) remains a clinical obstacle thought to be caused by the retention of dormant leukemic stem cells (LSCs). Using chemotherapy-treated AML xenografts and patient samples, we have modeled patient remission and relapse kinetics to reveal that LSCs are effectively depleted via cell-cycle recruitment, leaving the origins of relapse unclear. Post-chemotherapy, in vivo characterization at the onset of disease relapse revealed a unique molecular state of leukemic-regenerating cells (LRCs) responsible for disease re-growth. LRCs are transient, can only be detected in vivo, and are molecularly distinct from therapy-naive LSCs. We demonstrate that LRC features can be used as markers of relapse and are therapeutically targetable to prevent disease recurrence.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Leucemia Mieloide Aguda/tratamento farmacológico , Células Progenitoras Mieloides/efeitos dos fármacos , Recidiva Local de Neoplasia/prevenção & controle , Regeneração/efeitos dos fármacos , Adulto , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Ciclo Celular/efeitos dos fármacos , Feminino , Humanos , Leucemia Mieloide Aguda/patologia , Masculino , Camundongos , Células Progenitoras Mieloides/patologia , Recidiva Local de Neoplasia/diagnóstico , Cultura Primária de Células , Prognóstico , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Nat Cell Biol ; 19(11): 1336-1347, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29035359

RESUMO

Acute myeloid leukaemia (AML) is distinguished by the generation of dysfunctional leukaemic blasts, and patients characteristically suffer from fatal infections and anaemia due to insufficient normal myelo-erythropoiesis. Direct physical crowding of bone marrow (BM) by accumulating leukaemic cells does not fully account for this haematopoietic failure. Here, analyses from AML patients were applied to both in vitro co-culture platforms and in vivo xenograft modelling, revealing that human AML disease specifically disrupts the adipocytic niche in BM. Leukaemic suppression of BM adipocytes led to imbalanced regulation of endogenous haematopoietic stem and progenitor cells, resulting in impaired myelo-erythroid maturation. In vivo administration of PPARγ agonists induced BM adipogenesis, which rescued healthy haematopoietic maturation while repressing leukaemic growth. Our study identifies a previously unappreciated axis between BM adipogenesis and normal myelo-erythroid maturation that is therapeutically accessible to improve symptoms of BM failure in AML via non-cell autonomous targeting of the niche.


Assuntos
Adipócitos/patologia , Medula Óssea/patologia , Eritropoese/fisiologia , Leucemia Mieloide Aguda/patologia , Adipogenia/fisiologia , Adulto , Idoso , Animais , Medula Óssea/metabolismo , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Técnicas de Cocultura/métodos , Feminino , Células-Tronco Hematopoéticas , Humanos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Pessoa de Meia-Idade , PPAR gama/metabolismo , Células-Tronco/patologia , Adulto Jovem
5.
Stem Cells ; 35(9): 2095-2102, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28758276

RESUMO

Induced pluripotent stem cell reprogramming has provided critical insights into disease processes by modeling the genetics and related clinical pathophysiology. Human cancer represents highly diverse genetics, as well as inter- and intra-patient heterogeneity, where cellular model systems capable of capturing this disease complexity would be invaluable. Acute myeloid leukemia (AML) represents one of most heterogeneous cancers and has been divided into genetic subtypes correlated with unique risk stratification over the decades. Here, we report our efforts to induce pluripotency from the heterogeneous population of human patients that represents this disease in the clinic. Using robust optimized reprogramming methods, we demonstrate that reprogramming of AML cells harboring leukemic genomic aberrations is a rare event with the exception of those with de novo mixed-lineage leukemia (MLL) mutations that can be reprogrammed and model drug responses in vitro. Our findings indicate that unlike hematopoietic cells devoid of genomic aberrations, AML cells harboring driver mutations are refractory to reprogramming. Expression of MLL fusion proteins in AML cells did not contribute to induced reprogramming success, which continued to select for patient derived cells devoid of AML patient-specific aberrations. Our study reveals that unanticipated blockades to achieving pluripotency reside within the majority of transformed AML patient cells. Stem Cells 2017;35:2095-2102.


Assuntos
Reprogramação Celular , Hematopoese , Células-Tronco Pluripotentes Induzidas/patologia , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Mutação/genética , Células da Medula Óssea/patologia , Humanos , Proteína de Leucina Linfoide-Mieloide/metabolismo , Proteínas de Fusão Oncogênica/metabolismo
6.
Cell Chem Biol ; 24(7): 833-844.e9, 2017 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-28648376

RESUMO

Targeting of human cancer stem cells (CSCs) requires the identification of vulnerabilities unique to CSCs versus healthy resident stem cells (SCs). Unfortunately, dysregulated pathways that support transformed CSCs, such as Wnt/ß-catenin signaling, are also critical regulators of healthy SCs. Using the ICG-001 and CWP family of small molecules, we reveal Sam68 as a previously unappreciated modulator of Wnt/ß-catenin signaling within CSCs. Disruption of CBP-ß-catenin interaction via ICG-001/CWP induces the formation of a Sam68-CBP complex in CSCs that alters Wnt signaling toward apoptosis and differentiation induction. Our study identifies Sam68 as a regulator of human CSC vulnerability.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Ligação a DNA/metabolismo , Células-Tronco Neoplásicas/metabolismo , Fragmentos de Peptídeos/metabolismo , Proteínas de Ligação a RNA/metabolismo , Sialoglicoproteínas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/genética , Adulto , Idoso , Animais , Apoptose/efeitos dos fármacos , Compostos Azabicíclicos/farmacologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , Feminino , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Masculino , Camundongos , Camundongos Endogâmicos NOD , Pessoa de Meia-Idade , Células-Tronco Neoplásicas/citologia , Células-Tronco Neoplásicas/transplante , Organofosfatos/farmacologia , Fragmentos de Peptídeos/antagonistas & inibidores , Fragmentos de Peptídeos/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Pirimidinonas/farmacologia , Interferência de RNA , Proteínas de Ligação a RNA/antagonistas & inibidores , Proteínas de Ligação a RNA/genética , Sialoglicoproteínas/antagonistas & inibidores , Sialoglicoproteínas/genética , Sumoilação/efeitos dos fármacos , Transcriptoma/efeitos dos fármacos , Via de Sinalização Wnt/efeitos dos fármacos , beta Catenina/metabolismo
7.
Cell Rep ; 19(1): 20-35, 2017 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-28380358

RESUMO

Human pluripotent stem cells (hPSCs) have been reported in naive and primed states. However, the ability to generate mature cell types remains the imperative property for utility of hPSCs. Here, we reveal that the naive state enhances self-renewal while restricting lineage differentiation in vitro to neural default fate. Molecular analyses indicate expression of multiple lineage-associated transcripts in naive hPSCs that failed to predict biased functional differentiation capacity. Naive hPSCs can be converted to primed state over long-term serial passage that permits recovery of multi-germ layer differentiation. Suppression of OCT4 but not NANOG allows immediate recovery directly from naive state. To this end, we identified chemical inhibitors of OCT4 that restore naive hPSC differentiation. Our study reveals unique cell-fate restrictions in human pluripotent states and provides an approach to overcome these barriers that harness both efficient naive hPSC growth while maintaining in vitro differentiation essential for hPSC applications.


Assuntos
Diferenciação Celular/genética , Linhagem da Célula/genética , Reprogramação Celular/genética , Camadas Germinativas/citologia , Células-Tronco Pluripotentes/citologia , Animais , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Hepatócitos/metabolismo , Humanos , Camundongos , Miócitos Cardíacos/metabolismo , Proteína Homeobox Nanog/metabolismo , Nistatina/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo , RNA/genética , Teratoma/metabolismo
8.
Cancer Cell ; 29(1): 61-74, 2016 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-26766591

RESUMO

Initial pathway alternations required for pathogenesis of human acute myeloid leukemia (AML) are poorly understood. Here we reveal that removal of glycogen synthase kinase-3α (GSK-3α) and GSK-3ß dependency leads to aggressive AML. Although GSK-3α deletion alone has no effect, GSK-3ß deletion in hematopoietic stem cells (HSCs) resulted in a pre-neoplastic state consistent with human myelodysplastic syndromes (MDSs). Transcriptome and functional studies reveal that each GSK-3ß and GSK-3α uniquely contributes to AML by affecting Wnt/Akt/mTOR signaling and metabolism, respectively. The molecular signature of HSCs deleted for GSK-3ß provided a prognostic tool for disease progression and survival of MDS patients. Our study reveals that GSK-3α- and GSK-3ß-regulated pathways can be responsible for stepwise transition to MDS and subsequent AML, thereby providing potential therapeutic targets of disease evolution.


Assuntos
Quinase 3 da Glicogênio Sintase/metabolismo , Células-Tronco Hematopoéticas/enzimologia , Leucemia Mieloide Aguda/enzimologia , Animais , Modelos Animais de Doenças , Quinase 3 da Glicogênio Sintase/deficiência , Glicogênio Sintase Quinase 3 beta , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/terapia , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/fisiologia
9.
Stem Cell Res ; 15(1): 240-2, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26141785

RESUMO

The combination of OCT4 expression and short-term exposure to reprogramming media induces a state of transcriptional plasticity in human fibroblasts, capable of responding to changes in the extracellular environment. Here we provide characterization of iPSCs established through continued culture of OCT4-induced plastic human fibroblasts in pluripotent-supportive reprogramming media. Human iPSC(OCT4) are morphologically indistinguishable from conventionally derived iPSCs and express core proteins involved in maintenance of pluripotency. iPSC(OCT4) display bona fide functional pluripotency as measured by in vivo teratoma formation consisting of the three germ layers.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Fator 3 de Transcrição de Octâmero/farmacologia , Adulto , Animais , Reprogramação Celular/efeitos dos fármacos , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Camundongos Endogâmicos NOD , Camundongos SCID
10.
Stem Cell Res ; 15(1): 221-30, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26117529

RESUMO

The combination of OCT4 expression and short-term exposure to reprogramming media induces a state of transcriptional plasticity in human fibroblasts, capable of responding to changes in the extracellular environment that facilitate direct cell fate conversion toward lineage specific progenitors. Here we reveal that continued exposure of OCT4-induced plastic human fibroblasts to reprogramming media (RM) is sufficient to induce pluripotency. OCT4-derived induced pluripotent stem cell (iPSC(OCT4)) colonies emerged after prolonged culture in RM, and formed independently of lineage specific progenitors. Human iPSC(OCT4) are morphologically indistinguishable from conventionally derived iPSCs and express core proteins involved in maintenance of pluripotency. iPSC(OCT4) display in vivo functional pluripotency as measured by teratoma formation consisting of the three germ layers, and are capable of targeted in vitro differentiation. Our study indicates that acquisition of pluripotency is one of multiple cell fate choices that can be facilitated through environmental stimulation of OCT4-induced plasticity, and suggests the role of other reprogramming factors to induce pluripotency can be substituted by prolonged culture of plastic fibroblasts.


Assuntos
Fibroblastos/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Fator 3 de Transcrição de Octâmero/farmacologia , Adulto , Animais , Linhagem da Célula/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Humanos , Imunofenotipagem , Camundongos Endogâmicos NOD , Camundongos SCID , Modelos Biológicos
11.
Stem Cells ; 33(4): 1142-52, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25639500

RESUMO

The clinical use of human embryonic stem cells (hESCs) requires efficient cellular expansion that must be paired with an ability to generate specialized progeny through differentiation. Self-renewal and differentiation are deemed inherent hallmarks of hESCs and a growing body of evidence suggests that initial culture conditions dictate these two aspects of hESC behavior. Here, we reveal that defined culture conditions using commercial mTeSR1 media augment the expansion of hESCs and enhance their capacity for neural differentiation at the expense of hematopoietic lineage competency without affecting pluripotency. This culture-induced modification was shown to be reversible, as culture in mouse embryonic fibroblast-conditioned media (MEF-CM) in subsequent passages allowed mTeSR1-expanded hESCs to re-establish hematopoietic differentiation potential. Optimal yield of hematopoietic cells can be achieved by expansion in mTeSR1 followed by a recovery period in MEF-CM. Furthermore, the lineage propensity to hematopoietic and neural cell types could be predicted via analysis of surrogate markers expressed by hESCs cultured in mTeSR1 versus MEF-CM, thereby circumventing laborious in vitro differentiation assays. Our study reveals that hESCs exist in a range of functional states and balance expansion with differentiation potential, which can be modulated by culture conditions in a predictive and quantitative manner.


Assuntos
Técnicas de Cultura de Células/métodos , Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Células-Tronco Embrionárias Humanas/fisiologia , Animais , Proliferação de Células/fisiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID
12.
Innate Immun ; 21(5): 504-11, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25261966

RESUMO

The acquisition of innate immune response is requisite to having bona fide differentiation of airway epithelium. Procedures developed to differentiate lung airway from human pluripotent stem cells (hPSCs) have demonstrated anecdotal evidence for innate immune response, but an in-depth exploration of response levels is lacking. Herein, using an established method of airway epithelial generation from hPSCs, we show that hPSC-derived epithelial cells are able to up-regulate expression of TNFα, IL8 and IL1ß in response to challenge with bacterial endotoxin LPS, but lack response from genes associated with innate immune response in other cell types. Further, stimulation of cells with TNF-α resulted in auto-induction of TNFα transcript, as well as cytokine responses of IL8 and IL1ß. The demonstration of innate immune induction in hPSC-derived airway epithelia gives further strength to the functionality of in vitro protocols aimed at generating differentiated airway cells that can potentially be used in a translational setting. Finally, we propose that innate immune challenge of airway epithelium from human pluripotent stem cell sources be used as a robust validation of functional in vitro differentiation.


Assuntos
Imunidade Inata/imunologia , Células-Tronco Pluripotentes/imunologia , Mucosa Respiratória/imunologia , Diferenciação Celular , Células Cultivadas , Humanos , Interleucina-1beta/biossíntese , Interleucina-8/biossíntese , Lipopolissacarídeos/farmacologia , Mucosa Respiratória/citologia , Fator de Necrose Tumoral alfa/biossíntese , Regulação para Cima
13.
Stem Cells ; 32(8): 2178-87, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24740884

RESUMO

Here we characterize the molecular and biological requirements for OCT4 plasticity induction in human skin derived fibroblasts (hFibs) that allows direct conversion of cell fate without iPSC formation. Our results indicate that adult hFibs not only require OCT4 but also short-term exposure to reprogramming media (RM) to successfully undergo direct conversion to early hematopoietic and neural progenitor fates. RM was found to be essential in this process and allowed for unique changes in global gene expression specific to the combined effects of OCT4 and treatment with reprogramming media to establish a plastic state. This molecular state of hFib plasticity was distinct from transient expression of a full complement of iPSC reprogramming factors consistent with a lack in molecular hallmarks of iPSC formation. Human Fib-derived OCT4 plastic cells display elevated levels of developmentally related genes associated with multiple lineages, but not those associated with pluripotency. In response to changes in the extracellular environment, plastic OCT4-expressing hFibs further activate genes involved in hematopoietic as well as tripotent neural progenitor biology that allow cell fate conversion. Our study provides a working definition of hFib-induced plasticity using OCT4 and a deconvoluted system to elucidate the process of direct cell fate reprogramming.


Assuntos
Linhagem da Célula , Transdiferenciação Celular/fisiologia , Reprogramação Celular/fisiologia , Fibroblastos/citologia , Fator 3 de Transcrição de Octâmero/metabolismo , Células-Tronco/citologia , Envelhecimento , Meios de Cultivo Condicionados/farmacologia , Fibroblastos/metabolismo , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase
14.
Cell Stem Cell ; 13(2): 175-89, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23910084

RESUMO

Numerous studies have shown that the bone marrow (BM) niche plays a key role in mouse hematopoietic stem cell (HSC) function and involves contributions from a broad array of cell types. However, the composition and role of the human BM HSC niche have not been investigated. Here, using human bone biopsy specimens, we provide evidence of HSC propensity to localize to endosteal regions of the trabecular bone area (TBA). Through functional xenograft transplantation, we found that human HSCs localizing to the TBA have superior regenerative and self-renewal capacity and are molecularly distinct from those localizing to the long bone area (LBA). In addition, osteoblasts in the TBA possess unique characteristics and express a key network of factors that regulate TBA- versus LBA-localized human HSCs in vivo. Our study reveals that BM localization and architecture play a critical role in defining the functional and molecular properties of human HSCs.


Assuntos
Células da Medula Óssea/metabolismo , Osso e Ossos/patologia , Células-Tronco Hematopoéticas/metabolismo , Animais , Biópsia , Células da Medula Óssea/patologia , Proliferação de Células , Células-Tronco Hematopoéticas/patologia , Humanos , Ligantes , Camundongos , Osteoblastos/metabolismo , Osteoblastos/patologia , Receptores Notch/metabolismo , Nicho de Células-Tronco , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Exp Hematol ; 41(10): 858-869.e4, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23747997

RESUMO

Recent work has shown that leukemic stem cell self-renewal in chronic myeloid leukemia is dependent on cell-intrinsic hedgehog (Hh) signaling, and early clinical trials suggest that targeting this pathway is also therapeutic in patients with acute myeloid leukemia (AML). In this study, we aimed to better understand Hh signaling in normal hematopoiesis and AML by molecularly and functionally analyzing more than 200 primary human AML patient samples compared with nonleukemic controls. Gene expression analysis indicated that Hh pathway transcripts were similarly regulated in AML and nonleukemic controls, regardless of whether samples were purified based on primitive phenotypes. Consistent with these results, pharmacologic inhibition of Smoothened (SMO) did not preferentially reduce in vitro colony formation of AML versus normal progenitors. Using a unique analytic approach, messenger RNA expression of membrane receptor SMO was found to be unexpectedly rare within all hematopoietic samples analyzed, which is indicative of heterogeneity at the level of Hh signaling machinery. In contrast, abundant SMO expression could be readily detected in the nonhematopoietic fraction of human and murine bone marrow (BM) cells. Our predictions of increased SMO(+) cell frequencies within nonhematopoietic BM fractions were further supported by single-cell protein analyses. Although we did not find support for cell-autonomous sensitivity of AML cells to Hh pathway inhibition, we alternatively suggest that nonhematopoietic BM cells represent an indirect target through which primitive normal and leukemic cells can be modulated. These findings suggest current approaches to applying Hh inhibition should be carefully reevaluated to account for BM niche cell regulation that might be selectively Hh responsive.


Assuntos
Proteínas Hedgehog/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Leucemia Mieloide Aguda/fisiopatologia , Transdução de Sinais , Animais , Células da Medula Óssea/metabolismo , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Leucócitos Mononucleares/metabolismo , Masculino , Camundongos
16.
Cell ; 149(6): 1284-97, 2012 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-22632761

RESUMO

Selective targeting of cancer stem cells (CSCs) offers promise for a new generation of therapeutics. However, assays for both human CSCs and normal stem cells that are amenable to robust biological screens are limited. Using a discovery platform that reveals differences between neoplastic and normal human pluripotent stem cells (hPSC), we identify small molecules from libraries of known compounds that induce differentiation to overcome neoplastic self-renewal. Surprisingly, thioridazine, an antipsychotic drug, selectively targets the neoplastic cells, and impairs human somatic CSCs capable of in vivo leukemic disease initiation while having no effect on normal blood SCs. The drug antagonizes dopamine receptors that are expressed on CSCs and on breast cancer cells as well. These results suggest that dopamine receptors may serve as a biomarker for diverse malignancies, demonstrate the utility of using neoplastic hPSCs for identifying CSC-targeting drugs, and provide support for the use of differentiation as a therapeutic strategy.


Assuntos
Antineoplásicos/farmacologia , Antagonistas de Dopamina/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Células-Tronco Neoplásicas/efeitos dos fármacos , Tioridazina/farmacologia , Animais , Citarabina/farmacologia , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/patologia , Mefloquina/farmacologia , Camundongos , Células-Tronco Pluripotentes/efeitos dos fármacos , Piranos/farmacologia
17.
Mol Biol Cell ; 19(8): 3488-500, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18524849

RESUMO

Despite extensive work on ADP-ribosylation factor (Arf) 1 at the Golgi complex, the functions of Arf2-5 in the secretory pathway, or for that of any Arf at the ER-Golgi intermediate compartment (ERGIC) remain uncharacterized. Here, we examined the recruitment of fluorescently tagged Arf1, -3, -4, and -5 onto peripheral ERGIC. Live cell imaging detected Arfs on peripheral puncta that also contained Golgi-specific brefeldin A (BFA) resistance factor (GBF) 1 and the ERGIC marker p58. Unexpectedly, BFA did not promote corecruitment of Arfs with GBF1 either at the Golgi complex or the ERGIC, but it uncovered striking differences between Arf1,3 and Arf4,5. Although Arf1,3 quickly dissociated from all endomembranes after BFA addition, Arf4,5 persisted on ERGIC structures, even after redistribution of GBF1 to separate compartments. The GDP-arrested Arf4(T31N) mutant localized to the ERGIC, even with BFA and Exo1 present. In addition, loss of Arf x GTP after treatment with Exo1 caused rapid release of all Arfs from the Golgi complex and led to GBF1 accumulation on both Golgi and ERGIC membranes. Our results demonstrate that GDP-bound Arf4,5 associate with ERGIC membranes through binding sites distinct from those responsible for GBF1 recruitment. Furthermore, they provide the first evidence that GBF1 accumulation on membranes may be caused by loss of Arf x GTP, rather than the formation of an Arf x GDP x BFA x GBF1 complex.


Assuntos
Fatores de Ribosilação do ADP/metabolismo , Retículo Endoplasmático/metabolismo , Complexo de Golgi/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Guanosina Difosfato/química , Difosfato de Adenosina/química , Animais , Sítios de Ligação , Brefeldina A/farmacologia , Células COS , Chlorocebus aethiops , Células HeLa , Humanos , Modelos Biológicos , Mutação
18.
BMC Dev Biol ; 7: 133, 2007 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-18053124

RESUMO

BACKGROUND: Fps/Fes and Fer are the only two members of a distinct subclass of cytoplasmic protein tyrosine kinases. Fps/Fes was previously implicated in Semaphorin 3A (Sema3A)-induced growth cone collapse signaling in neurons from the dorsal root ganglion (DRG) through interaction with and phosphorylation of the Sema3A receptor component PlexinA1, and members of the collapsin response mediator protein (CRMP) family of microtubule regulators. However, the potential role of the closely related Fer kinase has not been examined. RESULTS: Here we provide novel biochemical and genetic evidence that Fer plays a prominent role in microtubule regulation in DRG neurons in response to Sema3A. Although Fps/Fes and Fer were both expressed in neonatal brains and isolated DRGs, Fer was expressed at higher levels; and Fer, but not Fps/Fes kinase activity was detected in vivo. Fer also showed higher in vitro kinase activity toward tubulin, as an exogenous substrate; and this activity was higher when the kinases were isolated from perinatal relative to adult brain stages. CRMP2 was a substrate for both kinases in vitro, but both CRMP2 and PlexinA1 inhibited their autophosphorylation activities. Cultured mouse DRG neurons retracted their axons upon exposure to Sema3A, and this response was significantly diminished in Fer-deficient, but only slightly attenuated in Fps/Fes-deficient DRG neurons. CONCLUSION: Fps/Fes and Fer are both capable of phosphorylating tubulin and the microtubule regulator CRMP2 in vitro; and their in vitro kinase activities were both inhibited by CRMP2 or PlexinA1, suggesting a possible regulatory interaction. Furthermore, Fer plays a more prominent role than Fps/Fes in regulating the axon retraction response to Sema3A in DRG neurons. Therefore, Fps/Fes and Fer may play important roles in developmental or regenerative axon pathfinding through signaling from Sema3A to the microtubule cytoskeleton.


Assuntos
Axônios , Gânglios Espinais/citologia , Neurônios/fisiologia , Proteínas Tirosina Quinases/metabolismo , Semaforina-3A/fisiologia , Animais , Animais Recém-Nascidos , Encéfalo/enzimologia , Encéfalo/crescimento & desenvolvimento , Linhagem Celular , Humanos , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/fisiologia , Fosforilação , Receptores de Superfície Celular/fisiologia , Semaforina-3A/metabolismo , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA