Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
2.
Toxicol Rep ; 8: 277-281, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33552926

RESUMO

OBJECTIVE: This study aimed to describe organs and systems damages in persons after mass poisoning with thallium and show the results of treatment. METHODS: Forty-four persons (12 males and 32 females) with acute oral thallium poisoning were tested for thallium levels in blood and urine and examined by a toxicologist and a neurologist, and in some -cases, by a gynecologist, an ophthalmologist, and a psychiatrist. Persons were divided into the following three groups depending on the severity of the poisoning: I: This group consisted of 9 persons (blood thallium level 8.3-26.7 µg/L) and treatment applied in the hospital included intestinal lavage, antidote therapy with potassium-ferric hexacyanoferrate, sodium dimercaptopropanesulfonate; II: This group consisted of 21 persons (0.3-6.1 µg/L) who received a similar treatment at home; and III: This group consisted of 14 (normal) persons who did not receive any treatment. RESULTS: The most common combination of several symptoms such as alopecia (on day 17-23), muscle pain of different localization in the debut of the disease (>88.9 % of the persons), sudden unexplained general weakness (>76.2 % of the persons), peripheral paraparesis or tetraparesis (including no complaints), polyneuropathy (88.89 % in group I vs. 54.14 % in group II, p < 0.05), static and dynamic ataxia (33.3 % in group I vs. 19.4 % in group II, p < 0.05), vertigo (1/3 of the persons), postural tremor (1/3 of the persons), and cognitive and emotional disorders (2/3 of the persons). Ovarian dysfunction was observed in all women of group I but in 42.9 % of group II, p < 0.05. The treatment was successful. In group I, plasma and urine thallium level significantly decreased by 69.3 % and 84 %, respectively. Pain, movement and coordination disorders regressed first while tremor, sensory, cognitive and emotional disorders lasted longer. Polyneuropathies later became mononeuropathies. Tremor could increase despite a decreased thallium concentration. DISCUSSION: The Sklifosovsky Institute conducted the largest study in Russian Federation investigating disorders in persons with acute thallium poisoning confirmed by laboratory tests. The clinical symptoms were consistent with those reported in the literature. The applied treatment was successful and led to better results compared to our previous approaches of treating mass thallium poisoning. CONCLUSIONS: This study shows a typical combination of thallium poisoning symptoms and allows us to recommend a complex therapy without the use of extracorporeal detoxification methods.

3.
Oncol Ther ; 8(1): 133-145, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32700077

RESUMO

Neuroblastoma represents 8-10% of all childhood cancer cases and is responsible for 15% of all cancer-related deaths in infants. Even though patients with low- and intermediate-risk disease have a good prognosis, the 5-year survival rate of the vast majority of patients with high-risk neuroblastoma is 50%. Despite extensive research efforts to find a cure for neuroblastoma, current treatment options are still limited. The aim of our study was to identify novel therapeutic compounds using high-throughput drug screening of a small molecule kinase inhibitor library containing 960 compounds. This screening resulted in the identification of two compounds, ST013381 and ST022328, that showed pronounced cytotoxic effects in six human neuroblastoma cell lines in vitro while having reduced effects in the BJ-5ta control cell line. These effects were observed in both MYCN-amplified and -non-amplified cells, indicating that these compounds can affect a wide range of neuroblastomas. Our experiments also revealed that several signaling pathways underlie the selective elimination of neuroblastoma cells by the ST013381 and ST022328 compounds. In summary, we have identified two novel compounds with a strong cytotoxic effect in vitro as promising agents for the treatment of neuroblastoma.

4.
Oncotarget ; 11(24): 2357-2371, 2020 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-32595833

RESUMO

Neuroblastoma (NB) is the most common cancer in infants and it accounts for six percent of all pediatric malignancies. There are several hypotheses proposed on the origins of NB. While there is little genetic evidence to support this, the prevailing model is that NB originates from neural crest stem cells (NCSCs). Utilizing in vivo mouse models, we demonstrate that targeting MYCN oncogene to NCSCs causes perinatal lethality. During sympathoadrenal (SA) lineage development, SOX transcriptional factors drive the transition from NCSCs to lineage-specific progenitors, characterized by the sequential activation of Sox9/Sox10/Sox4/Sox11 genes. We find the NCSCs factor SOX10 is not expressed in neuroblasts, but rather restricted to the Schwannian stroma and is associated with a good prognosis. On the other hand, SOX9 expression in NB cells was associated with several key biological processes including migration, invasion and differentiation. Moreover, manipulating SOX9 gene predominantly affects lineage-restricted SA progenitors. Our findings highlight a unique molecular SOX signature associated with NB that is highly reminiscent of SA progenitor transcriptional program during embryonic development, providing novel insights into NB pathobiology. In summary, we provide multiple lines of evidence suggesting that multipotent NCSCs do not contribute to NB initiation and maintenance.

5.
Life Sci Alliance ; 3(6)2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32312912

RESUMO

Cutaneous squamous cell carcinoma (cSCC) is the second most common skin cancer characterized by increased mortality. Here, we show for the first time that anaplastic lymphoma kinase (ALK), a receptor tyrosine kinase of the insulin receptor superfamily, plays a pivotal role in the pathogenesis of cSCC. Our data demonstrate that the overexpression of the constitutively active, mutated ALK, ALK F1174L , is sufficient to initiate the development of cSCC and is 100% penetrant. Moreover, we show that cSCC development upon ALK F1174L overexpression is independent of the cell-of-origin. Molecularly, our data demonstrate that ALK F1174L cooperates with oncogenic Kras G12D and loss of p53, well-established events in the biology of cSCC. This cooperation results in a more aggressive cSCC type associated with a higher grade histological morphology. Finally, we demonstrate that Stat3 is a key downstream effector of ALK F1174L and likely plays a role in ALK F1174L -driven cSCC tumorigenesis. In sum, these findings reveal that ALK can exert its tumorigenic potential via cooperation with multiple pathways crucial in the pathogenesis of cSCC. Finally, we show that human cSCCs contain mutations in the ALK gene. Taken together, our data identify ALK as a new key player in the pathogenesis of cSCC, and this knowledge suggests that oncogenic ALK signaling can be a target for future clinical trials.


Assuntos
Quinase do Linfoma Anaplásico/metabolismo , Carcinogênese/metabolismo , Carcinoma de Células Escamosas/metabolismo , Oncogenes , Transdução de Sinais/genética , Neoplasias Cutâneas/metabolismo , Quinase do Linfoma Anaplásico/genética , Animais , Carcinogênese/genética , Carcinoma de Células Escamosas/genética , Modelos Animais de Doenças , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Neoplasias Cutâneas/genética , Transfecção
6.
Oncogene ; 39(20): 4132-4154, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32238882

RESUMO

Despite advances in the systemic treatment of patients with metastatic melanoma using immune checkpoint and tyrosine kinase inhibitors (TKI), the majority of stage IV melanoma patients eventually succumb to the disease. We have previously identified the transcription factor Sox10 as a crucial player in melanoma, yet the underlying molecular mechanisms mediating Sox10-dependent tumorigenesis remain largely uncharacterized. Here, we show that MEK and RAF inhibitors do not suppress levels of SOX10 protein in patient-derived cells in vitro, as well as in melanoma patients in vivo. In a search for pharmacological inhibitors of SOX10, we performed a mass spectrometry-based screen in human melanoma cells. Subsequent analysis revealed that SOX10 directly interacts with ß-catenin, which is a key mediator of canonical Wnt/ß-catenin signaling. We demonstrate that inhibitors of glycogen synthase kinase 3 alpha/beta (GSK3α/ß) efficiently abrogate SOX10 protein in human melanoma cells in vitro and in melanoma mouse models in vivo. The mechanism of action of GSK3-mediated SOX10 suppression is transcription-independent and relies on the presence of a proteasome degradable form of ß-catenin. Taken together, we provide evidence that activation of canonical Wnt signaling has a profound effect on melanoma growth and is able to counteract Sox10-dependent melanoma maintenance both in vitro and in vivo.


Assuntos
Regulação Neoplásica da Expressão Gênica , Melanoma Experimental/metabolismo , Proteínas de Neoplasias/metabolismo , Fatores de Transcrição SOXE/biossíntese , Via de Sinalização Wnt , Animais , Linhagem Celular Tumoral , Humanos , Melanoma Experimental/genética , Melanoma Experimental/patologia , Camundongos , Camundongos Nus , Proteínas de Neoplasias/genética , Fatores de Transcrição SOXE/genética
7.
Front Oncol ; 9: 275, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31058082

RESUMO

Neuroblastoma (NB) is an embryonal malignancy derived from the abnormal differentiation of the sympathetic nervous system. The Anaplastic Lymphoma Kinase (ALK) gene is frequently altered in NB, through copy number alterations and activating mutations, and represents a predisposition in NB-genesis when mutated. Our previously published data suggested that ALK activating mutations may impair the differentiation potential of neural crest (NC) progenitor cells. Here, we demonstrated that the expression of the endogenous ALK gene starts at E10.5 in the developing sympathetic ganglia (SG). To decipher the impact of deregulated ALK signaling during embryogenesis on the formation and differentiation of sympathetic neuroblasts, Sox10-Cre;LSL-ALK-F1174L embryos were produced to restrict the expression of the human ALK-F1174L transgene to migrating NC cells (NCCs). First, ALK-F1174L mediated an embryonic lethality at mid-gestation and an enlargement of SG with a disorganized architecture in Sox10-Cre;LSL-ALK-F1174L embryos at E10.5 and E11.5. Second, early sympathetic differentiation was severely impaired in Sox10-Cre;LSL-ALK-F1174L embryos. Indeed, their SG displayed a marked increase in the proportion of NCCs and a decrease of sympathetic neuroblasts at both embryonic stages. Third, neuronal and noradrenergic differentiations were blocked in Sox10-Cre;LSL-ALK-F1174L SG, as a reduced proportion of Phox2b+ sympathoblasts expressed ßIII-tubulin and almost none were Tyrosine Hydroxylase (TH) positive. Finally, at E10.5, ALK-F1174L mediated an important increase in the proliferation of Phox2b+ progenitors, affecting the transient cell cycle exit observed in normal SG at this embryonic stage. Altogether, we report for the first time that the expression of the human ALK-F1174L mutation in NCCs during embryonic development profoundly disturbs early sympathetic progenitor differentiation, in addition to increasing their proliferation, both mechanisms being potential crucial events in NB oncogenesis.

8.
Nat Commun ; 9(1): 236, 2018 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-29339718

RESUMO

Cutaneous wound healing is a complex process that aims to re-establish the original structure of the skin and its functions. Among other disorders, peripheral neuropathies are known to severely impair wound healing capabilities of the skin, revealing the importance of skin innervation for proper repair. Here, we report that peripheral glia are crucially involved in this process. Using a mouse model of wound healing, combined with in vivo fate mapping, we show that injury activates peripheral glia by promoting de-differentiation, cell-cycle re-entry and dissemination of the cells into the wound bed. Moreover, injury-activated glia upregulate the expression of many secreted factors previously associated with wound healing and promote myofibroblast differentiation by paracrine modulation of TGF-ß signalling. Accordingly, depletion of these cells impairs epithelial proliferation and wound closure through contraction, while their expansion promotes myofibroblast formation. Thus, injury-activated glia and/or their secretome might have therapeutic potential in human wound healing disorders.


Assuntos
Diferenciação Celular/fisiologia , Neuroglia/fisiologia , Pele/fisiopatologia , Cicatrização/fisiologia , Animais , Ciclo Celular/genética , Ciclo Celular/fisiologia , Diferenciação Celular/genética , Células Cultivadas , Perfilação da Expressão Gênica , Humanos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Knockout , Camundongos Transgênicos , Miofibroblastos/metabolismo , Miofibroblastos/fisiologia , Neuroglia/citologia , Neuroglia/metabolismo , Fatores de Transcrição SOXE/genética , Fatores de Transcrição SOXE/metabolismo , Transdução de Sinais/genética , Pele/lesões , Pele/inervação , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Cicatrização/genética
9.
Nat Commun ; 8(1): 1466, 2017 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-29133867

RESUMO

Renal angiomyolipomas (AML) contain an admixture of clonal tumour cells with features of several different mesenchymal lineages, implying the existence of an unidentified AML neoplastic stem cell. Biallelic inactivation of TSC2 or TSC1 is believed to represent the driving event in these tumours. Here we show that TSC2 knockdown transforms senescence-resistant cultured mouse and human renal epithelial cells into neoplastic stem cells that serially propagate renal AML-like tumours in mice. mTOR inhibitory therapy of mouse AML allografts mimics the clinical responses of human renal AMLs. Deletion of Tsc1 in mouse renal epithelia causes differentiation in vivo into cells expressing characteristic AML markers. Human renal AML and a renal AML cell line express proximal tubule markers. We describe the first mouse models of renal AML and provide evidence that these mesenchymal tumours originate from renal proximal tubule epithelial cells, uncovering an unexpected pathological differentiation plasticity of the proximal tubule.


Assuntos
Angiomiolipoma/patologia , Células Epiteliais/citologia , Neoplasias Renais/patologia , Túbulos Renais Proximais/citologia , Células-Tronco Neoplásicas/citologia , Proteínas Supressoras de Tumor/genética , Animais , Diferenciação Celular/genética , Células Epiteliais/patologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Transplante de Neoplasias , Interferência de RNA , RNA Interferente Pequeno/genética , Esferoides Celulares , Serina-Treonina Quinases TOR/antagonistas & inibidores , Transplante Heterólogo , Proteína 1 do Complexo Esclerose Tuberosa , Proteína 2 do Complexo Esclerose Tuberosa , Células Tumorais Cultivadas , Proteínas Supressoras de Tumor/metabolismo
10.
Methods Mol Biol ; 2015 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-26659800

RESUMO

During development, melanocyte progenitors originate from the neural crest, a transient embryonic structure in vertebrates that gives rise to a variety of cell types including neurons and glia of the peripheral nervous system, smooth muscle cells of the cardiovascular system, chondrocytes and osteoblasts of the craniofacial elements, and pigment cells in the skin. In this chapter, we describe a method for the differentiation of multipotent embryonic neural crest stem cells into differentiated pigmented melanocytes by using in vitro explant culture system. This protocol allows the dissection of genetic and cellular mechanisms regulating neural crest stem cell and melanocyte development. Based on this knowledge it is possible to make predictions about processes that might also be implicated in melanoma initiation and progression.

11.
Genome Biol ; 16: 42, 2015 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-25885555

RESUMO

BACKGROUND: Melanoma is the most fatal skin cancer displaying a high degree of molecular heterogeneity. Phenotype switching is a mechanism that contributes to melanoma heterogeneity by altering transcription profiles for the transition between states of proliferation/differentiation and invasion/stemness. As phenotype switching is reversible, epigenetic mechanisms, like DNA methylation, could contribute to the changes in gene expression. RESULTS: Integrative analysis of methylation and gene expression datasets of five proliferative and five invasion melanoma cell cultures reveal two distinct clusters. SOX9 is methylated and lowly expressed in the highly proliferative group. SOX9 overexpression results in decreased proliferation but increased invasion in vitro. In a B16 mouse model, sox9 overexpression increases the number of lung metastases. Transcriptional analysis of SOX9-overexpressing melanoma cells reveals enrichment in epithelial to mesenchymal transition (EMT) pathways. Survival analysis of The Cancer Genome Atlas melanoma dataset shows that metastatic patients with high expression levels of SOX9 have significantly worse survival rates. Additional survival analysis on the targets of SOX9 reveals that most SOX9 downregulated genes have survival benefit for metastatic patients. CONCLUSIONS: Our genome-wide DNA methylation and gene expression study of 10 early passage melanoma cell cultures reveals two phenotypically distinct groups. One of the genes regulated by DNA methylation between the two groups is SOX9. SOX9 induces melanoma cell invasion and metastasis and decreases patient survival. A number of genes downregulated by SOX9 have a negative impact on patient survival. In conclusion, SOX9 is an important gene involved in melanoma invasion and negatively impacts melanoma patient survival.


Assuntos
Melanoma/genética , Invasividade Neoplásica/genética , Fatores de Transcrição SOX9/biossíntese , Neoplasias Cutâneas/genética , Idoso , Animais , Linhagem Celular Tumoral , Proliferação de Células/genética , Metilação de DNA/genética , Transição Epitelial-Mesenquimal/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Melanoma/patologia , Camundongos , Pessoa de Meia-Idade , Invasividade Neoplásica/patologia , Estadiamento de Neoplasias , Prognóstico , Fatores de Transcrição SOX9/genética , Transdução de Sinais , Neoplasias Cutâneas/patologia , Análise de Sobrevida
12.
Nat Commun ; 6: 6051, 2015 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-25609585

RESUMO

Increased activity of the epigenetic modifier EZH2 has been associated with different cancers. However, evidence for a functional role of EZH2 in tumorigenesis in vivo remains poor, in particular in metastasizing solid cancers. Here we reveal central roles of EZH2 in promoting growth and metastasis of cutaneous melanoma. In a melanoma mouse model, conditional Ezh2 ablation as much as treatment with the preclinical EZH2 inhibitor GSK503 stabilizes the disease through inhibition of growth and virtually abolishes metastases formation without affecting normal melanocyte biology. Comparably, in human melanoma cells, EZH2 inactivation impairs proliferation and invasiveness, accompanied by re-expression of tumour suppressors connected to increased patient survival. These EZH2 target genes suppress either melanoma growth or metastasis in vivo, revealing the dual function of EZH2 in promoting tumour progression. Thus, EZH2-mediated epigenetic repression is highly relevant especially during advanced melanoma progression, which makes EZH2 a promising target for novel melanoma therapies.


Assuntos
Inativação Gênica , Melanoma/metabolismo , Complexo Repressor Polycomb 2/fisiologia , Neoplasias Cutâneas/metabolismo , Adenosilmetionina Descarboxilase/metabolismo , Animais , Proliferação de Células , Proteína Potenciadora do Homólogo 2 de Zeste , Epigênese Genética , Transição Epitelial-Mesenquimal , Feminino , Regulação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Genótipo , Homeostase , Humanos , Melanócitos/metabolismo , Melanoma Experimental/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Invasividade Neoplásica , Metástase Neoplásica , Complexo Repressor Polycomb 2/genética , Resultado do Tratamento , Melanoma Maligno Cutâneo
13.
PLoS Genet ; 11(1): e1004877, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25629959

RESUMO

Melanoma is the most fatal skin cancer, but the etiology of this devastating disease is still poorly understood. Recently, the transcription factor Sox10 has been shown to promote both melanoma initiation and progression. Reducing SOX10 expression levels in human melanoma cells and in a genetic melanoma mouse model, efficiently abolishes tumorigenesis by inducing cell cycle exit and apoptosis. Here, we show that this anti-tumorigenic effect functionally involves SOX9, a factor related to SOX10 and upregulated in melanoma cells upon loss of SOX10. Unlike SOX10, SOX9 is not required for normal melanocyte stem cell function, the formation of hyperplastic lesions, and melanoma initiation. To the contrary, SOX9 overexpression results in cell cycle arrest, apoptosis, and a gene expression profile shared by melanoma cells with reduced SOX10 expression. Moreover, SOX9 binds to the SOX10 promoter and induces downregulation of SOX10 expression, revealing a feedback loop reinforcing the SOX10 low/SOX9 high ant,m/ii-tumorigenic program. Finally, SOX9 is required in vitro and in vivo for the anti-tumorigenic effect achieved by reducing SOX10 expression. Thus, SOX10 and SOX9 are functionally antagonistic regulators of melanoma development.


Assuntos
Carcinogênese/genética , Melanoma/genética , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOXE/genética , Animais , Apoptose/genética , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Folículo Piloso , Humanos , Melanócitos/patologia , Melanoma/patologia , Camundongos , RNA Interferente Pequeno , Fatores de Transcrição SOX9/biossíntese , Fatores de Transcrição SOXE/biossíntese
14.
Cell Stem Cell ; 15(6): 720-34, 2014 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-25479748

RESUMO

The open chromatin of embryonic stem cells (ESCs) condenses into repressive heterochromatin as cells exit the pluripotent state. How the 3D genome organization is orchestrated and implicated in pluripotency and lineage specification is not understood. Here, we find that maturation of the long noncoding RNA (lncRNA) pRNA is required for establishment of heterochromatin at ribosomal RNA genes, the genetic component of nucleoli, and this process is inactivated in pluripotent ESCs. By using mature pRNA to tether heterochromatin at nucleoli of ESCs, we find that localized heterochromatin condensation of ribosomal RNA genes initiates establishment of highly condensed chromatin structures outside of the nucleolus. Moreover, we reveal that formation of such highly condensed, transcriptionally repressed heterochromatin promotes transcriptional activation of differentiation genes and loss of pluripotency. Our findings unravel the nucleolus as an active regulator of chromatin plasticity and pluripotency and challenge current views on heterochromatin regulation and function in ESCs.


Assuntos
Nucléolo Celular/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Células-Tronco Embrionárias/fisiologia , Genes de RNAr , Neurônios/fisiologia , Células-Tronco Pluripotentes/fisiologia , RNA Longo não Codificante/metabolismo , Animais , Diferenciação Celular , Linhagem da Célula , Montagem e Desmontagem da Cromatina , Proteínas Cromossômicas não Histona/genética , Epigênese Genética , Genes de RNAr/genética , Heterocromatina/metabolismo , Humanos , Camundongos , Células NIH 3T3 , Transporte Proteico , Processamento Pós-Transcricional do RNA , RNA Longo não Codificante/genética
15.
Transl Oncol ; 7(2): 206-12, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24746711

RESUMO

Despite existing vaccination strategies targeting TRP-2, its function is not yet fully understood. TRP-2 is an enzyme involved in melanin biosynthesis and therefore discussed as a differentiation antigen. However, in mice Trp-2 was shown to be expressed in melanocyte stem cells of the hair follicle and therefore also considered as an indicator of stemness. A proper understanding of the TRP-2 function is crucial, considering a vaccination targeting cells with stemness properties would be highly effective in contrast to a therapy targeting differentiated melanoma cells. Analysing over 200 melanomas including primaries, partly matched metastases and patients' cell cultures we show that TRP-2 is correlated with Melan A expression and decreases with tumor progression. In mice it is expressed in differentiated melanocytes as well as in stem cells. Furthermore, we identify a TRP-2 negative, proliferative, hypoxia related cell subpopulation which is significantly associated with tumor thickness and diseases progression. Patients with a higher percentage of those cells have a less favourable tumor specific survival. Our findings underline that TRP-2 is a differentiation antigen, highlighting the importance to combine TRP-2 vaccination with other strategies targeting the aggressive undifferentiated hypoxia related subpopulation.

16.
Curr Opin Oncol ; 26(2): 215-21, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24441506

RESUMO

PURPOSE OF REVIEW: Metastatic melanoma is the most aggressive skin cancer and despite tremendous efforts and considerable progress in clinical treatment of melanoma patients within recent years, it remains a deadly disease. Current treatments affect melanoma cells indiscriminately, while accumulating evidence suggests that melanoma might be a disease of stem cells. This review aims to summarize the important accomplishments in the field and to emphasize the common molecular and cellular mechanisms regulating self-renewal of neural crest stem cells (NCSCs) and melanoma cells. RECENT FINDINGS: A growing number of publications highlight the existence of phenotypic and functional similarities between embryonic NCSCs and melanoma cells. These studies provide compelling evidence that the propagation of melanoma cells critically depends on genes instrumental in neural crest development. The example of Sox10 and Rac1 genes provides detailed illustration of how interfering with these important genes for neural crest development can prevent melanoma formation. SUMMARY: The development of new therapies, targeting RAF-MEK-ERK pathway, provided major improvements in outcomes for patients with metastatic melanoma; however, acquired resistance followed by tumor recurrence represents a major clinical challenge. The striking parallels between embryonic NCSCs (eNCSCs) and melanoma cells might lead to the development of new targeted therapeutics selectively eliminating cell populations accountable for tumor initiation, progression and relapse.


Assuntos
Melanoma/genética , Crista Neural/metabolismo , Células-Tronco Neurais/metabolismo , Neoplasias Cutâneas/genética , Antineoplásicos/uso terapêutico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Redes Reguladoras de Genes/efeitos dos fármacos , Redes Reguladoras de Genes/genética , Humanos , Melanoma/tratamento farmacológico , Melanoma/patologia , Fatores de Transcrição SOXE/genética , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/patologia , Proteínas rac1 de Ligação ao GTP/genética
17.
Dermatopathology (Basel) ; 1(1): 35-46, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-27047921

RESUMO

BACKGROUND: Congenital melanocytic nevi (CMNs) are melanocytic neoplasms that can transform into melanoma. However, this development is impeded in the majority of cases and mostly affects patients with large or giant CMNs. METHODS: To elucidate mechanisms that keep CMNs from malignant transformation, CMN tissue biopsies were investigated for p-ERK and senescence markers by immunohistochemistry and for SOX10/CD271 (p75(NTR)) by immunofluorescence. CMN cells were cultivated, and MTT assays were performed for evaluating cell viability. Mutation status for NRAS and BRAF was performed by real-time PCR. RESULTS: 13 CMNs (from patients aged 0.5-11.8 years, mean: 2.7) showed immunoreactivity for SOX10/CD271 (p75(NTR)) in 34.2%. p-ERK was immunoreactive in 80% (4/5); ß-galactosidase was significantly stronger expressed in CMNs compared to melanocytic nevi of patients over 70 years (p = 0.0085). The 5 CMN cultures were immunoreactive for SOX10/CD271 (p75(NTR)) in 36.7%. By silencing SOX10 by siRNA in 2 CMN cell cultures, cell viability decreased significantly. NRAS(Q61K) mutation was found in 91.7% (11/12) and BRAF(V600E) in 6.3% of all analyzable CMNs (1/16). CONCLUSIONS: Oncogene-induced senescence might prevent malignant transformation through activation of the mitogen-activated protein kinase pathway. SOX10 is necessary for the viability of human CMN cell cultures and may be responsible for clinical changes during aging.

18.
PLoS Genet ; 9(7): e1003644, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23935512

RESUMO

During embryogenesis, the transcription factor, Sox10, drives the survival and differentiation of the melanocyte lineage. However, the role that Sox10 plays in postnatal melanocytes is not established. We show in vivo that melanocyte stem cells (McSCs) and more differentiated melanocytes express SOX10 but that McSCs remain undifferentiated. Sox10 knockout (Sox10(fl); Tg(Tyr::CreER)) results in loss of both McSCs and differentiated melanocytes, while overexpression of Sox10 (Tg(DctSox10)) causes premature differentiation and loss of McSCs, leading to hair graying. This suggests that levels of SOX10 are key to normal McSC function and Sox10 must be downregulated for McSC establishment and maintenance. We examined whether the mechanism of Tg(DctSox10) hair graying is through increased expression of Mitf, a target of SOX10, by asking if haploinsufficiency for Mitf (Mitf(vga9) ) can rescue hair graying in Tg(DctSox10) animals. Surprisingly, Mitf(vga9) does not mitigate but exacerbates Tg(DctSox10) hair graying suggesting that MITF participates in the negative regulation of Sox10 in McSCs. These observations demonstrate that while SOX10 is necessary to maintain the postnatal melanocyte lineage it is simultaneously prevented from driving differentiation in the McSCs. This data illustrates how tissue-specific stem cells can arise from lineage-specified precursors through the regulation of the very transcription factors important in defining that lineage.


Assuntos
Desenvolvimento Embrionário/genética , Melanócitos/citologia , Fatores de Transcrição SOXE/genética , Células-Tronco/citologia , Animais , Diferenciação Celular/genética , Linhagem da Célula , Cor de Cabelo/genética , Melanócitos/metabolismo , Camundongos , Camundongos Knockout , Fatores de Transcrição SOXE/metabolismo , Células-Tronco/metabolismo
19.
Cancer Lett ; 338(1): 74-81, 2013 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-23073475

RESUMO

Whether tumorigenic cancer stem cells (CSCs) exist in melanoma has been the focus of much controversy in recent years. A number of studies have pointed to the existence of melanoma cell sub-populations that act as CSCs and can be distinguished from other tumor cells based on specific surface marker expression or specific properties such as the capacity for extensive self-renewal. Other studies failed to identify melanoma stem cells and proposed that the potential to initiate tumors is a wide spread feature in melanoma inherent to most if not all cells of the tumor mass. As with normal stem cells, the term CSC is based on an operational definition, indicating not just a tumor-initiating cell, but also a cell with the capacity to sustain long-term tumor propagation. Therefore, the experimental set-up chosen to identify putative CSCs in melanoma is crucial: Both the method of tumor cell preparation and the procedure used to assess CSC properties in vivo influence the experimental outcome and hence its interpretation. In this review, we summarize our current knowledge on CSCs and the role of stem cell properties in melanoma and discuss recent findings with respect to their clinical relevance.


Assuntos
Biomarcadores Tumorais/genética , Proliferação de Células , Melanoma/genética , Células-Tronco Neoplásicas/metabolismo , Animais , Humanos , Melanoma/patologia , Melanoma/terapia , Camundongos , Camundongos Knockout , Células-Tronco Neoplásicas/patologia , Células-Tronco Neoplásicas/transplante , Interferência de RNA , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Nat Cell Biol ; 14(8): 882-90, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22772081

RESUMO

Giant congenital naevi are pigmented childhood lesions that frequently lead to melanoma, the most aggressive skin cancer. The mechanisms underlying this malignancy are largely unknown, and there are no effective therapies. Here we describe a mouse model for giant congenital naevi and show that naevi and melanoma prominently express Sox10, a transcription factor crucial for the formation of melanocytes from the neural crest. Strikingly, Sox10 haploinsufficiency counteracts Nras(Q61K)-driven congenital naevus and melanoma formation without affecting the physiological functions of neural crest derivatives in the skin. Moreover, Sox10 is also crucial for the maintenance of neoplastic cells in vivo. In human patients, virtually all congenital naevi and melanomas are SOX10 positive. Furthermore, SOX10 silencing in human melanoma cells suppresses neural crest stem cell properties, counteracts proliferation and cell survival, and completely abolishes in vivo tumour formation. Thus, SOX10 represents a promising target for the treatment of congenital naevi and melanoma in human patients.


Assuntos
Melanoma/fisiopatologia , Nevo/patologia , Fatores de Transcrição SOXE/metabolismo , Animais , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Western Blotting , Linhagem Celular , Linhagem Celular Tumoral , Criança , Modelos Animais de Doenças , Feminino , Regulação Neoplásica da Expressão Gênica , Haploinsuficiência , Humanos , Imuno-Histoquímica , Lactente , Masculino , Melanoma/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Análise em Microsséries , Nevo/fisiopatologia , Reação em Cadeia da Polimerase em Tempo Real , Fatores de Transcrição SOXE/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA