Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Nature ; 616(7955): 168-175, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36949199

RESUMO

The resistance of cancer cells to therapy is responsible for the death of most patients with cancer1. Epithelial-to-mesenchymal transition (EMT) has been associated with resistance to therapy in different cancer cells2,3. However, the mechanisms by which EMT mediates resistance to therapy remain poorly understood. Here, using a mouse model of skin squamous cell carcinoma undergoing spontaneous EMT during tumorigenesis, we found that EMT tumour cells are highly resistant to a wide range of anti-cancer therapies both in vivo and in vitro. Using gain and loss of function studies in vitro and in vivo, we found that RHOJ-a small GTPase that is preferentially expressed in EMT cancer cells-controls resistance to therapy. Using genome-wide transcriptomic and proteomic profiling, we found that RHOJ regulates EMT-associated resistance to chemotherapy by enhancing the response to replicative stress and activating the DNA-damage response, enabling tumour cells to rapidly repair DNA lesions induced by chemotherapy. RHOJ interacts with proteins that regulate nuclear actin, and inhibition of actin polymerization sensitizes EMT tumour cells to chemotherapy-induced cell death in a RHOJ-dependent manner. Together, our study uncovers the role and the mechanisms through which RHOJ acts as a key regulator of EMT-associated resistance to chemotherapy.


Assuntos
Carcinoma de Células Escamosas , Resistencia a Medicamentos Antineoplásicos , Transição Epitelial-Mesenquimal , Neoplasias Cutâneas , Proteínas rho de Ligação ao GTP , Actinas/efeitos dos fármacos , Actinas/metabolismo , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Proteômica , Proteínas rho de Ligação ao GTP/genética , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Camundongos , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Perfilação da Expressão Gênica , Genoma
2.
Nature ; 556(7702): 463-468, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29670281

RESUMO

In cancer, the epithelial-to-mesenchymal transition (EMT) is associated with tumour stemness, metastasis and resistance to therapy. It has recently been proposed that, rather than being a binary process, EMT occurs through distinct intermediate states. However, there is no direct in vivo evidence for this idea. Here we screen a large panel of cell surface markers in skin and mammary primary tumours, and identify the existence of multiple tumour subpopulations associated with different EMT stages: from epithelial to completely mesenchymal states, passing through intermediate hybrid states. Although all EMT subpopulations presented similar tumour-propagating cell capacity, they displayed differences in cellular plasticity, invasiveness and metastatic potential. Their transcriptional and epigenetic landscapes identify the underlying gene regulatory networks, transcription factors and signalling pathways that control these different EMT transition states. Finally, these tumour subpopulations are localized in different niches that differentially regulate EMT transition states.


Assuntos
Transição Epitelial-Mesenquimal , Neoplasias/patologia , Animais , Cromatina/genética , Epigênese Genética , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Transição Epitelial-Mesenquimal/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Masculino , Neoplasias Mamárias Animais/genética , Neoplasias Mamárias Animais/patologia , Mesoderma/metabolismo , Mesoderma/patologia , Camundongos , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Metástase Neoplásica/genética , Metástase Neoplásica/patologia , Neoplasias/genética , Transdução de Sinais , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Transcrição Gênica
3.
Genes Dev ; 27(1): 39-51, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23271346

RESUMO

The accurate maintenance of genomic integrity is essential for tissue homeostasis. Deregulation of this process leads to cancer and aging. BRCA1 is a critical mediator of this process. Here, we performed conditional deletion of Brca1 during epidermal development and found that BRCA1 is specifically required for hair follicle (HF) formation and for development of adult HF stem cells (SCs). Mice deficient for Brca1 in the epidermis are hairless and display a reduced number of HFs that degenerate progressively. Surprisingly, the interfollicular epidermis and the sebaceous glands remain unaffected by Brca1 deletion. Interestingly, HF matrix transient amplifying progenitors present increased DNA damage, p53 stabilization, and caspase-dependent apoptosis compared with the interfollicular and sebaceous progenitors, leading to hyperproliferation, apoptosis, and subsequent depletion of the prospective adult HF SCs. Concomitant deletion of p53 and Brca1 rescues the defect of HF morphogenesis and loss of HF SCs. During adult homeostasis, BRCA1 is dispensable for quiescent bulge SCs, but upon their activation during HF regeneration, Brca1 deletion causes apoptosis and depletion of Brca1-deficient bulge SCs. Our data reveal a major difference in the requirement of BRCA1 between different types of epidermal SCs and progenitors and during the different activation stages of adult HF SCs.


Assuntos
Proteína BRCA1/deficiência , Proteína BRCA1/metabolismo , Epiderme , Folículo Piloso/citologia , Células-Tronco , Animais , Apoptose/genética , Proteína BRCA1/genética , Dano ao DNA/genética , Células Epidérmicas , Epiderme/metabolismo , Deleção de Genes , Folículo Piloso/embriologia , Folículo Piloso/metabolismo , Camundongos , Camundongos Knockout , Células-Tronco/citologia , Células-Tronco/metabolismo , Proteína Supressora de Tumor p53/metabolismo
4.
Stem Cells ; 35(5): 1355-1364, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28100039

RESUMO

The carcinogenic effect of ionizing radiation has been evaluated based on limited populations accidently exposed to high dose radiation. In contrast, insufficient data are available on the effect of low dose radiation (LDR), such as radiation deriving from medical investigations and interventions, as well as occupational exposure that concern a large fraction of western populations. Using mouse skin epidermis as a model, we showed that LDR results in DNA damage in sebaceous gland (SG) and bulge epidermal stem cells (SCs). While the first commit apoptosis upon low dose irradiation, the latter survive. Bulge SC survival coincides with higher HIF-1α expression and a metabolic switch upon LDR. Knocking down HIF-1α sensitizes bulge SCs to LDR-induced apoptosis, while upregulation of HIF-1α in the epidermis, including SG SCs, rescues cell death. Most importantly, we show that LDR results in cancer formation with full penetrance in the radiation-sensitive Patched1 heterozygous mice. Overall, our results demonstrate for the first time that LDR can be a potent carcinogen in individuals predisposed to cancer. Stem Cells 2017;35:1355-1364.


Assuntos
Epiderme/efeitos da radiação , Neoplasias Cutâneas/etiologia , Células-Tronco/efeitos da radiação , Animais , Carcinoma Basocelular/patologia , Sobrevivência Celular/efeitos da radiação , Dano ao DNA , Relação Dose-Resposta à Radiação , Heterozigoto , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Receptor Patched-1/metabolismo , Penetrância , Glândulas Sebáceas/patologia , Neoplasias Cutâneas/patologia , Células-Tronco/metabolismo
5.
Nature ; 489(7415): 257-62, 2012 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-22940863

RESUMO

The skin interfollicular epidermis (IFE) is the first barrier against the external environment and its maintenance is critical for survival. Two seemingly opposite theories have been proposed to explain IFE homeostasis. One posits that IFE is maintained by long-lived slow-cycling stem cells that give rise to transit-amplifying cell progeny, whereas the other suggests that homeostasis is achieved by a single committed progenitor population that balances stochastic fate. Here we probe the cellular heterogeneity within the IFE using two different inducible Cre recombinase­oestrogen receptor constructs targeting IFE progenitors in mice. Quantitative analysis of clonal fate data and proliferation dynamics demonstrate the existence of two distinct proliferative cell compartments arranged in a hierarchy involving slow-cycling stem cells and committed progenitor cells. After wounding, only stem cells contribute substantially to the repair and long-term regeneration of the tissue, whereas committed progenitor cells make a limited contribution.


Assuntos
Células Epidérmicas , Células-Tronco/citologia , Animais , Diferenciação Celular , Divisão Celular , Linhagem da Célula , Sobrevivência Celular , Células Clonais/citologia , Células Clonais/metabolismo , Integrases/genética , Integrases/metabolismo , Queratina-14/genética , Camundongos , Regiões Promotoras Genéticas/genética , Precursores de Proteínas/genética , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Células-Tronco/metabolismo , Cauda/citologia , Cicatrização/fisiologia
6.
Nature ; 478(7369): 399-403, 2011 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-22012397

RESUMO

Angiogenesis is critical during tumour initiation and malignant progression. Different strategies aimed at blocking vascular endothelial growth factor (VEGF) and its receptors have been developed to inhibit angiogenesis in cancer patients. It has become increasingly clear that in addition to its effect on angiogenesis, other mechanisms including a direct effect of VEGF on tumour cells may account for the efficiency of VEGF-blockade therapies. Cancer stem cells (CSCs) have been described in various cancers including squamous tumours of the skin. Here we use a mouse model of skin tumours to investigate the impact of the vascular niche and VEGF signalling on controlling the stemness (the ability to self renew and differentiate) of squamous skin tumours during the early stages of tumour progression. We show that CSCs of skin papillomas are localized in a perivascular niche, in the immediate vicinity of endothelial cells. Furthermore, blocking VEGFR2 caused tumour regression not only by decreasing the microvascular density, but also by reducing CSC pool size and impairing CSC renewal properties. Conditional deletion of Vegfa in tumour epithelial cells caused tumours to regress, whereas VEGF overexpression by tumour epithelial cells accelerated tumour growth. In addition to its well-known effect on angiogenesis, VEGF affected skin tumour growth by promoting cancer stemness and symmetric CSC division, leading to CSC expansion. Moreover, deletion of neuropilin-1 (Nrp1), a VEGF co-receptor expressed in cutaneous CSCs, blocked VEGF's ability to promote cancer stemness and renewal. Our results identify a dual role for tumour-cell-derived VEGF in promoting cancer stemness: by stimulating angiogenesis in a paracrine manner, VEGF creates a perivascular niche for CSCs, and by directly affecting CSCs through Nrp1 in an autocrine loop, VEGF stimulates cancer stemness and renewal. Finally, deletion of Nrp1 in normal epidermis prevents skin tumour initiation. These results may have important implications for the prevention and treatment of skin cancers.


Assuntos
Carcinoma de Células Escamosas/irrigação sanguínea , Carcinoma de Células Escamosas/patologia , Neuropilina-1/metabolismo , Transdução de Sinais , Neoplasias Cutâneas/irrigação sanguínea , Neoplasias Cutâneas/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Células Epiteliais/citologia , Deleção de Genes , Regulação Neoplásica da Expressão Gênica , Camundongos , Células-Tronco Neoplásicas , Neuropilina-1/genética , Fator A de Crescimento do Endotélio Vascular/genética
7.
Proc Natl Acad Sci U S A ; 108(18): 7431-6, 2011 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-21502497

RESUMO

Squamous cell carcinoma (SCC) is the second most frequent skin cancer. The cellular origin of SCC remains controversial. Here, we used mouse genetics to determine the epidermal cell lineages at the origin of SCC. Using mice conditionally expressing a constitutively active KRas mutant (G12D) and an inducible CRE recombinase in different epidermal lineages, we activated Ras signaling in different cellular compartments of the skin epidermis and determined from which epidermal compartments Ras activation induces squamous tumor formation. Expression of mutant KRas in hair follicle bulge stem cells (SCs) and their immediate progeny (hair germ and outer root sheath), but not in their transient amplifying matrix cells, led to benign squamous skin tumor (papilloma). Expression of KRas(G12D) in interfollicular epidermis also led to papilloma formation, demonstrating that squamous tumor initiation is not restricted to the hair follicle lineages. Whereas no malignant tumor was observed after KRas(G12D) expression alone, expression of KRas(G12D) combined with the loss of p53 induced invasive SCC. Our studies demonstrate that different epidermal lineages including bulge SC are competent to initiate papilloma formation and that multiple genetic hits in the context of oncogenic KRas are required for the development of invasive SCC.


Assuntos
Carcinoma de Células Escamosas/fisiopatologia , Linhagem da Célula/fisiologia , Células Epidérmicas , Neoplasias Cutâneas/fisiopatologia , Animais , Bromodesoxiuridina , Carcinoma de Células Escamosas/genética , Primers do DNA/genética , Citometria de Fluxo , Folículo Piloso/citologia , Integrases/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias Cutâneas/genética , Células-Tronco/metabolismo
8.
Cereb Cortex ; 22(7): 1678-89, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21940705

RESUMO

The patterning of cortical areas is controlled by a combination of intrinsic factors that are expressed in the cortex and external signals such as inputs from the thalamus. EphA7 is a guidance receptor that is involved in key aspects of cortical development and is expressed in gradients within developing cortical areas. Here, we identified a regulatory element of the EphA7 promoter, named pA7, that can recapitulate salient features of the pattern of expression of EphA7, including cortical gradients. Using a pA7-Green fluorescent Protein (GFP) mouse reporter line, we isolated cortical neuron populations displaying different levels of EphA7/GFP expression. Transcriptome analysis of these populations enabled to identify many differentially expressed genes, including 26 transcription factors with putative binding sites in the pA7 element. Among these, Pbx1 was found to bind directly to the EphA7 promoter in the developing cortex. All genes validated further were confirmed to be expressed differentially in the developing cortex, similarly to EphA7. Their expression was unchanged in mutant mice defective for thalamocortical projections, indicating a transcriptional control largely intrinsic to the cortex. Our study identifies a novel repertoire of cortical neuron genes that may act upstream of, or together with EphA7, to control the patterning of cortical areas.


Assuntos
Córtex Cerebral/embriologia , Córtex Cerebral/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Receptor EphA7/metabolismo , Fatores de Transcrição/metabolismo , Ativação Transcricional/fisiologia , Transcriptoma/fisiologia , Animais , Camundongos
9.
J Immunother ; 45(3): 150-161, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35191428

RESUMO

Allogeneic chimeric antigen receptor (CAR) T holds the promise of taking this therapeutic approach to broader patient populations while avoiding the intensive manufacturing demands of autologous cell products. One limitation to delivering an allogeneic CAR T is T-cell receptor (TCR) driven toxicity. In this work, the expression of a peptide to interfere with TCR signaling was assessed for the generation of allogeneic CAR T cells. The expression of a truncated CD3ζ peptide was shown to incorporate into the TCR complex and to result in blunted TCR responses. When coexpressed with a natural killer group 2D (NKG2D) CAR, the allogeneic T cells (called CYAD-101) failed to induce graft-versus-host disease in mouse models while maintaining antitumor activity driven by the CAR in vitro and in vivo. Two clinical grade discrete batches of CYAD-101 cells were produced of single donor apheresis resulting in 48 billion CAR T cells sufficient for the entire dose-escalation phase of the proposed clinical trial. The 2 batches showed high consistency producing a predominantly CD4+ T-cell population that displayed an effector/central memory phenotype with no evidence of exhaustion markers expression. These clinical grade CYAD-101 cells secreted cytokines and chemokines in response to ligands expressing target cells in vitro, demonstrating effector function through the CAR. Moreover, CYAD-101 cells failed to respond to TCR stimulation, indicating a lack of allogeneic potential. This bank of clinical grade, non-gene-edited, allogeneic CYAD-101 cells are used in the alloSHRINK clinical trial (NCT03692429).


Assuntos
Transplante de Células-Tronco Hematopoéticas , Receptores de Antígenos Quiméricos , Animais , Humanos , Imunoterapia Adotiva/métodos , Camundongos , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos Quiméricos/metabolismo
10.
Stem Cells ; 26(11): 2964-73, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18772311

RESUMO

The maintenance of genome integrity in stem cells (SCs) is critical for preventing cancer formation and cellular senescence. The immortal strand hypothesis postulates that SCs protect their genome by keeping the same DNA strand throughout life by asymmetrical cell divisions, thus avoiding accumulation of mutations that can arise during DNA replication. The in vivo relevance of this model remains to date a matter of intense debate. In this study, we revisited this long-standing hypothesis, by analyzing how multipotent hair follicle (HF) SCs segregate their DNA strands during morphogenesis, skin homeostasis, and SC activation. We used three different in vivo approaches to determine how HF SCs segregate their DNA strand during cell divisions. Double-labeling studies using pulse-chase experiments during morphogenesis and the first adult hair cycle showed that HF SCs incorporate two different nucleotide analogs, contradictory to the immortal strand hypothesis. The co-segregation of DNA and chromatin labeling during pulse-chase experiments demonstrated that label retention in HF SCs is rather a mark of relative quiescence. Moreover, DNA labeling of adult SCs, similar to labeling during morphogenesis, also resulted in label retention in HF SCs, indicating that chromosome segregation occurs randomly in most of these cells. Altogether, our results demonstrate that DNA strand segregation occurs randomly in the majority of HF SCs during development, tissue homeostasis, and following SC activation. Disclosure of potential conflicts of interest is found at the end of this article.


Assuntos
Divisão Celular/fisiologia , Segregação de Cromossomos , Células Epidérmicas , Células-Tronco Multipotentes/citologia , Animais , Cromatina/fisiologia , Dano ao DNA/fisiologia , Folículo Piloso/citologia , Folículo Piloso/fisiologia , Queratinócitos/citologia , Queratinócitos/fisiologia , Camundongos , Células-Tronco Multipotentes/fisiologia
11.
Cell Rep ; 29(6): 1458-1468.e3, 2019 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-31693888

RESUMO

Epithelial-to-mesenchymal transition (EMT) has been proposed to be important for metastatic dissemination. However, recent studies have challenged the requirement of EMT for metastasis. Here, we assessed in different models of primary skin squamous cell carcinomas (SCCs) whether EMT is associated with metastasis. The incidence of metastasis was much higher in SCCs presenting EMT compared to SCCs without EMT, supporting the notion that a certain degree of EMT is required to initiate the metastatic cascade in primary skin SCCs. Most circulating tumor cells presented EMT, whereas most lung metastasis did not present EMT, showing that mesenchymal-to-epithelial transition is important for metastatic colonization. In contrast, immunodeficient mice transplanted with SCCs, whether displaying EMT or not, presented metastasis. Altogether, our data demonstrate that the association of EMT and metastasis is model dependent, and metastasis of primary skin SCCs is associated with EMT.


Assuntos
Carcinoma de Células Escamosas/secundário , Transição Epitelial-Mesenquimal , Células Neoplásicas Circulantes/metabolismo , Neoplasias Cutâneas/patologia , Animais , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Molécula de Adesão da Célula Epitelial/metabolismo , Feminino , Incidência , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Metástase Linfática , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Transplante de Neoplasias , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo , Transplante Homólogo
12.
iScience ; 15: 243-256, 2019 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-31082735

RESUMO

Epidermal growth factor receptor (EGFR) signaling controls skin development and homeostasis in mice and humans, and its deficiency causes severe skin inflammation, which might affect epidermal stem cell behavior. Here, we describe the inflammation-independent effects of EGFR deficiency during skin morphogenesis and in adult hair follicle stem cells. Expression and alternative splicing analysis of RNA sequencing data from interfollicular epidermis and outer root sheath indicate that EGFR controls genes involved in epidermal differentiation and also in centrosome function, DNA damage, cell cycle, and apoptosis. Genetic experiments employing p53 deletion in EGFR-deficient epidermis reveal that EGFR signaling exhibits p53-dependent functions in proliferative epidermal compartments, as well as p53-independent functions in differentiated hair shaft keratinocytes. Loss of EGFR leads to absence of LEF1 protein specifically in the innermost epithelial hair layers, resulting in disorganization of medulla cells. Thus, our results uncover important spatial and temporal features of cell-autonomous EGFR functions in the epidermis.

13.
Methods Mol Biol ; 407: 225-43, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18453259

RESUMO

Mesenchymal stem cells (MSCs) are multipotent progenitor cells isolated by various relatively easily accessible tissues, such as bone marrow and cord blood. MSCs gained attention because of their ease for in vitro expansion together with their multilineage potential. More recently, in vitro and in vivo immunosuppressive properties have been ascribed to them, as they are able to modulate the function of all major immune cell populations, thus impeding immune responses. The underlying mechanisms of their differentiation and function are not thoroughly understood, but still they represent important candidates for tissue regeneration and manipulation of the immune response in graft rejection, graft versus host disease, and autoimmune disorders. Characteristics and immunogenic profile of MSCs, their interface with immune system and their potential use as immunosuppressive elements in cellular therapeutic protocols are reviewed in this chapter.


Assuntos
Células-Tronco Mesenquimais/imunologia , Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/prevenção & controle , Humanos , Tolerância Imunológica , Terapia de Imunossupressão , Transplante de Células-Tronco Mesenquimais/métodos , Neoplasias/imunologia , Neoplasias/terapia , Imunologia de Transplantes
14.
Methods Mol Biol ; 407: 245-63, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18453260

RESUMO

Mesenchymal stem cells or marrow stromal cells (MSCs) represent a multipotent adult cellular population with immunomodulatory functions. In the adult human body, they are present in various niches, but their main source is bone marrow (BM). The regeneration capability of MSCs, their ease to undergo gene modification, as well as their immunosuppressive capacity render them as popular candidates for tissue engineering, gene therapy, and immunotherapy. They exhibit a unique in vitro expansion capacity, which however does not always compensate for the large number of cells required for cellular therapeutic applications. Unfortunately, to date, a uniform worldwide approach to MSC-culture is not available. Thus, in this chapter, we try to describe the optimal conditions for the successful isolation and ex vivo expansion of human MSCs from BM, to be used in all types of cellular therapeutic approaches. Moreover, we describe the methods for identification of their quality in terms of both multilineage potentiality and immunosuppressive ability. Detailed protocols for fetal calf serum selection and other culture parameters that affect the final outcome are described.


Assuntos
Células da Medula Óssea/fisiologia , Técnicas de Cultura de Células/métodos , Diferenciação Celular , Técnicas Imunoenzimáticas/métodos , Células-Tronco Mesenquimais/citologia , Ensaio de Unidades Formadoras de Colônias , Humanos
15.
ACS Med Chem Lett ; 8(9): 953-957, 2017 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-28947943

RESUMO

The preparation of heteronanoparticles (NPs) with doxorubicin (DOXO) and cyclopamine (CYP) conjugates is presented. Biological evaluation on A431 cell lines confirms the maintenance of the activity of the parental drugs. The in vivo study shows that self-assembled NPs reduce tumor growth and toxicity of chemotherapy.

16.
Biochim Biophys Acta ; 1725(2): 182-9, 2005 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-16126344

RESUMO

The elimination of cancer cells requires strong cellular immune responses, and these responses are induced by the activation of Th1 lymphocytes. In this work, the possibility of inducing a Th1 type of immune response in vivo by mixing a HER-2/neu synthetic CTL (cytotoxic T lymphocyte) peptide [HER-2/neu (789-797)], with poly-lactide (PLA) microspheres was investigated. Various formulations of the peptide were administered to HLA-A2.1 transgenic (HHD) mice. Cellular experiments, assessing proliferation and cytokine determination in splenocyte culture supernatants, were carried out in order to evaluate the type of immune response to the antigen. The in vivo administration of the peptide antigen admixed with the PLA microspheres induced a potent immune response which was comparable to that induced by the combination of the antigen in complete Freund's adjuvant (CFA). Furthermore, the cytokine profile produced by the T lymphocytes of the immunized animals indicated that the combination of the peptide antigen with the PLA microspheres induced a strong Th1 biased immune response to the antigen. The time of peptide incubation with the microspheres prior to administration did not affect the immune response, which further simplifies the preparation of this type of vaccine. The results justify further investigation of the possibility of inducing effective cellular immune responses against cancer cells overexpressing HER-2/neu molecules by simply mixing appropriate HER-2/neu peptide antigens with PLA microspheres.


Assuntos
Portadores de Fármacos/química , Poliésteres/química , Receptor ErbB-2/administração & dosagem , Receptor ErbB-2/imunologia , Células Th1/efeitos dos fármacos , Células Th1/imunologia , Vacinas/imunologia , Animais , Materiais Revestidos Biocompatíveis/administração & dosagem , Materiais Revestidos Biocompatíveis/química , Difusão , Portadores de Fármacos/administração & dosagem , Epitopos/imunologia , Imunização/métodos , Teste de Materiais , Camundongos , Microesferas , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/imunologia , Ligação Proteica , Receptor ErbB-2/química , Vacinas/uso terapêutico
17.
Chempluschem ; 80(6): 938-943, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31973263

RESUMO

The design and the preparation of a small library of 1,4-diphenyl-1,2,3-triazole derivatives is reported, with the aim to obtain a new class of Hedgehog pathway inhibitors. The smoothened protein is part of the hedgehog signaling pathway that is inhibited by the lead compound Vismodegib. Based on molecular modeling simulations, seven triazole derivatives of Vismodegib are synthesized and their biological effect on different endothelial, cancer, and cancer stem cell lines is reported.

18.
Chempluschem ; 80(9): 1380-1383, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31973361

RESUMO

Cyclopamine- and paclitaxel-containing hetero-nanoparticles generated by self-assembly show combined efficacy in the treatment of three different cancer cell lines. The use of ternary combination with the addition of a dye-squalene conjugate secured the obtainment of fluorescent nanoparticles that permitted the observation of the cellular internalization by confocal microscopy and super-resolution dSTORM (direct stochastic optical reconstruction microscopy).

19.
Drug Discov Today ; 19(10): 1547-62, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24819719

RESUMO

Cancer stem cells (CSCs) are a subpopulation of cancer cells with high clonogenic capacity and ability to reform parental tumors upon transplantation. Resistance to therapy has been shown for several types of CSC and, therefore, they have been proposed as the cause of tumor relapse. Consequently, much effort has been made to design molecules that can target CSCs specifically and sensitize them to therapy. In this review, we summarize the mechanisms underlying CSC resistance, the potential biological targets to overcome resistance and the chemical compounds showing activity against different types of CSC. The chemical compounds discussed here have been divided according to their origin: natural, natural-derived and synthetic compounds.


Assuntos
Antineoplásicos/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Células-Tronco Neoplásicas/efeitos dos fármacos , Animais , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Dano ao DNA , Reparo do DNA , Humanos , Neoplasias/tratamento farmacológico
20.
J Cell Biol ; 202(6): 887-900, 2013 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-24019538

RESUMO

The atypical protein kinase C (aPKC) is a key regulator of polarity and cell fate in lower organisms. However, whether mammalian aPKCs control stem cells and fate in vivo is not known. Here we show that loss of aPKCλ in a self-renewing epithelium, the epidermis, disturbed tissue homeostasis, differentiation, and stem cell dynamics, causing progressive changes in this tissue. This was accompanied by a gradual loss of quiescent hair follicle bulge stem cells and a temporary increase in proliferating progenitors. Lineage tracing analysis showed that loss of aPKCλ altered the fate of lower bulge/hair germ stem cells. This ultimately led to loss of proliferative potential, stem cell exhaustion, alopecia, and premature aging. Inactivation of aPKCλ produced more asymmetric divisions in different compartments, including the bulge. Thus, aPKCλ is crucial for homeostasis of self-renewing stratifying epithelia, and for the regulation of cell fate, differentiation, and maintenance of epidermal bulge stem cells likely through its role in balancing symmetric and asymmetric division.


Assuntos
Diferenciação Celular , Proliferação de Células , Células Epidérmicas , Homeostase/fisiologia , Isoenzimas/fisiologia , Proteína Quinase C/fisiologia , Células-Tronco/citologia , Animais , Animais Recém-Nascidos , Apoptose , Western Blotting , Células Cultivadas , Epiderme/metabolismo , Feminino , Técnicas Imunoenzimáticas , Queratinócitos/citologia , Queratinócitos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Células-Tronco/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA