Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Int J Mol Sci ; 22(15)2021 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-34361081

RESUMO

Cancer cachexia is a common deleterious paraneoplastic syndrome that represents an area of unmet clinical need, partly due to its poorly understood aetiology and complex multifactorial nature. We have interrogated multiple genetically defined larval Drosophila models of tumourigenesis against key features of human cancer cachexia. Our results indicate that cachectic tissue wasting is dependent on the genetic characteristics of the tumour and demonstrate that host malnutrition or tumour burden are not sufficient to drive wasting. We show that JAK/STAT and TNF-α/Egr signalling are elevated in cachectic muscle and promote tissue wasting. Furthermore, we introduce a dual driver system that allows independent genetic manipulation of tumour and host skeletal muscle. Overall, we present a novel Drosophila larval paradigm to study tumour/host tissue crosstalk in vivo, which may contribute to future research in cancer cachexia and impact the design of therapeutic approaches for this pathology.


Assuntos
Caquexia/patologia , Carcinogênese/patologia , Modelos Animais de Doenças , Larva/crescimento & desenvolvimento , Neoplasias/complicações , Animais , Caquexia/etiologia , Caquexia/metabolismo , Carcinogênese/genética , Carcinogênese/metabolismo , Drosophila , Perfilação da Expressão Gênica , Humanos , Janus Quinases/genética , Janus Quinases/metabolismo , Larva/genética , Larva/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais
2.
Rev. Esc. Enferm. USP ; 55: e03670, 2021. tab
Artigo em Inglês | BDENF - Enfermagem, LILACS | ID: biblio-1287962

RESUMO

ABSTRACT Objective: To assess the distribution of citations of nursing authors in Spanish in Google Scholar as well as to compare the possible differences between this source and Web of Science and Scopus. Method: This is a descriptive cross-sectional study based on the citation systems offered by Google Scholar, Web of Science, and Scopus. Results: Nursing researchers present a verified mean h-index of 7.82 in Academic Google. 74% of researchers belong to the academic field, compared to 26%, who are in health services. Most of them live in Spain (83%), followed by Colombia (12%), Mexico (4%), and Chile (1%). In Spain, the community with the largest number of researchers is Andalusia (41.5%), followed by Valencia (14.6%), and Madrid (7.3%). Conclusion: The Google Scholar citation system requires adjustments in its algorithm for selecting works and citations, and it should also allow some system of confirmation by authors. Nursing can have relatively low h-index values compared to other courses due to short research development.


RESUMO Objetivo: Avaliar a distribuição de citações de autores de enfermagem em espanhol no Google Scholar, bem como comparar as possíveis diferenças entre esta fonte e Web of Science e Scopus. Método: Estudo descritivo transversal baseado nos sistemas de citação oferecidos pelo Google Scholar, Web of Science e Scopus. Resultados: Pesquisadores da área de enfermagem apresentam índice h médio verificado no Google Acadêmico de 7,82. 74% dos pesquisadores pertencem à área acadêmica, contra 26% que estão agrupados nos serviços de saúde. A maioria deles está localizada na Espanha (83%), seguida pela Colômbia (12%), México (4%) e Chile (1%). Na Espanha, a comunidade com maior número de pesquisadores é a Andaluzia (41,5%), seguida da Comunidade Valenciana (14,6%) e Madrid (7,3%). Conclusão: O sistema de citações do Google Scholar requer ajustes em seu algoritmo de seleção de obras e citações, devendo também permitir algum sistema de confirmação por parte dos autores. A enfermagem pode ter valores relativamente baixos do índice h em comparação com outras disciplinas devido ao curto desenvolvimento da pesquisa.


RESUMEN Objetivo: Evaluar la distribución de citas de autores enfermeros en español en Google Académico, así como comparar las posibles diferencias entre esta fuente y Web of Science y Scopus. Método: Estudio descriptivo transversal basado en los sistemas de citas ofrecidos por Google Académico, Web of Science y Scopus. Resultados: Los investigadores del área de enfermería presentan un índice h verificado medio de 7.82 en Google Académico. El 74% de los investigadores pertenece al ámbito académico, frente a un 26% que se aglutina en los servicios de salud. La mayoría de ellos se ubican en España (83%), seguido de Colombia (12%), Méjico (4%) y Chile (1%). En España, la comunidad que mayor número de investigadores aglutina es Andalucía (41,5%), seguida de la Comunidad Valencia (14,6%) y Madrid (7,3%). Conclusión: El sistema de citación de Google Académico precisa de ajustes en su algoritmo de selección de trabajos y citas, además debería permitir algún sistema de confirmación por parte de los autores. Enfermería puede tener valores relativamente bajos del índice h frente a otras disciplinas debido al breve desarrollo investigador.


Assuntos
Pesquisa em Enfermagem , Bibliometria , Comunicação e Divulgação Científica , Comunicação Acadêmica
3.
Elife ; 82019 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-31358113

RESUMO

Antimicrobial peptides (AMPs) are small cationic molecules best known as mediators of the innate defence against microbial infection. While in vitro and ex vivo evidence suggest AMPs' capacity to kill cancer cells, in vivo demonstration of an anti-tumour role of endogenous AMPs is lacking. Using a Drosophila model of tumourigenesis, we demonstrate a role for the AMP Defensin in the control of tumour progression. Our results reveal that Tumour Necrosis Factor mediates exposure of phosphatidylserine (PS), which makes tumour cells selectively sensitive to the action of Defensin remotely secreted from tracheal and fat tissues. Defensin binds tumour cells in PS-enriched areas, provoking cell death and tumour regression. Altogether, our results provide the first in vivo demonstration for a role of an endogenous AMP as an anti-cancer agent, as well as a mechanism that explains tumour cell sensitivity to the action of AMPs.


Assuntos
Morte Celular , Defensinas/metabolismo , Fatores Imunológicos/metabolismo , Neoplasias/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Modelos Animais de Doenças , Drosophila , Análise de Sobrevida
4.
Cell Cycle ; 15(12): 1538-44, 2016 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-27191973

RESUMO

Bursicon is the main regulator of post molting and post eclosion processes during arthropod development. The active Bursicon hormone is a heterodimer of Burs-α and Burs-ß. However, adult midguts express Burs-α to regulate the intestinal stem cell niche. Here, we examined the potential expression and function of its heterodimeric partner, Burs-ß in the adult midgut. Unexpectedly, our evidence suggests that Burs-ß is not significantly expressed in the adult midgut. burs-ß mutants displayed the characteristic developmental defects but showed wild type-like adult midguts, thus uncoupling the developmental and adult phenotypes seen in burs-α mutants. Gain of function data and ex vivo experiments using a cAMP biosensor, demonstrated that Burs-α is sufficient to drive stem cell quiescence and to activate dLGR2 in the adult midgut. Our evidence suggests that the post developmental transactivation of dLGR2 in the adult midgut is mediated by Burs-α and that the ß subunit of Bursicon is dispensable for these activities.


Assuntos
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Trato Gastrointestinal/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Hormônios de Invertebrado/genética , Subunidades Proteicas/genética , Receptores Acoplados a Proteínas G/genética , Animais , AMP Cíclico/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/metabolismo , Trato Gastrointestinal/crescimento & desenvolvimento , Hormônios de Invertebrado/metabolismo , Muda/genética , Fenótipo , Multimerização Proteica , Subunidades Proteicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Nicho de Células-Tronco/genética , Ativação Transcricional
5.
Nature ; 517(7535): 497-500, 2015 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-25383520

RESUMO

Inactivation of APC is a strongly predisposing event in the development of colorectal cancer, prompting the search for vulnerabilities specific to cells that have lost APC function. Signalling through the mTOR pathway is known to be required for epithelial cell proliferation and tumour growth, and the current paradigm suggests that a critical function of mTOR activity is to upregulate translational initiation through phosphorylation of 4EBP1 (refs 6, 7). This model predicts that the mTOR inhibitor rapamycin, which does not efficiently inhibit 4EBP1 (ref. 8), would be ineffective in limiting cancer progression in APC-deficient lesions. Here we show in mice that mTOR complex 1 (mTORC1) activity is absolutely required for the proliferation of Apc-deficient (but not wild-type) enterocytes, revealing an unexpected opportunity for therapeutic intervention. Although APC-deficient cells show the expected increases in protein synthesis, our study reveals that it is translation elongation, and not initiation, which is the rate-limiting component. Mechanistically, mTORC1-mediated inhibition of eEF2 kinase is required for the proliferation of APC-deficient cells. Importantly, treatment of established APC-deficient adenomas with rapamycin (which can target eEF2 through the mTORC1-S6K-eEF2K axis) causes tumour cells to undergo growth arrest and differentiation. Taken together, our data suggest that inhibition of translation elongation using existing, clinically approved drugs, such as the rapalogs, would provide clear therapeutic benefit for patients at high risk of developing colorectal cancer.


Assuntos
Transformação Celular Neoplásica/patologia , Neoplasias Intestinais/metabolismo , Neoplasias Intestinais/patologia , Complexos Multiproteicos/metabolismo , Elongação Traducional da Cadeia Peptídica , Serina-Treonina Quinases TOR/metabolismo , Proteína da Polipose Adenomatosa do Colo/deficiência , Proteína da Polipose Adenomatosa do Colo/genética , Animais , Proliferação de Células , Transformação Celular Neoplásica/metabolismo , Quinase do Fator 2 de Elongação/deficiência , Quinase do Fator 2 de Elongação/genética , Quinase do Fator 2 de Elongação/metabolismo , Ativação Enzimática , Genes APC , Neoplasias Intestinais/genética , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Endogâmicos C57BL , Proteína Oncogênica p55(v-myc)/metabolismo , Fator 2 de Elongação de Peptídeos/metabolismo , Proteínas Quinases S6 Ribossômicas/metabolismo , Transdução de Sinais , Proteínas Wnt/metabolismo
6.
Curr Biol ; 24(11): 1199-211, 2014 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-24814146

RESUMO

BACKGROUND: Enteroendocrine cells populate gastrointestinal tissues and are known to translate local cues into systemic responses through the release of hormones into the bloodstream. RESULTS: Here we report a novel function of enteroendocrine cells acting as local regulators of intestinal stem cell (ISC) proliferation through modulation of the mesenchymal stem cell niche in the Drosophila midgut. This paracrine signaling acts to constrain ISC proliferation within the epithelial compartment. Mechanistically, midgut enteroendocrine cells secrete the neuroendocrine hormone Bursicon, which acts-beyond its known roles in development-as a paracrine factor on the visceral muscle (VM). Bursicon binding to its receptor, DLGR2, the ortholog of mammalian leucine-rich repeat-containing G protein-coupled receptors (LGR4-6), represses the production of the VM-derived EGF-like growth factor Vein through activation of cAMP. CONCLUSIONS: We therefore identify a novel paradigm in the regulation of ISC quiescence involving the conserved ligand/receptor Bursicon/DLGR2 and a previously unrecognized tissue-intrinsic role of enteroendocrine cells.


Assuntos
Drosophila melanogaster/fisiologia , Células Enteroendócrinas/fisiologia , Células-Tronco Mesenquimais/metabolismo , Comunicação Parácrina , Animais , Diferenciação Celular , Proliferação de Células , AMP Cíclico/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/genética , Feminino , Regulação da Expressão Gênica , Homeostase , Intestinos/citologia , Intestinos/fisiologia , Hormônios de Invertebrado/genética , Hormônios de Invertebrado/metabolismo , Músculos/metabolismo , Neurregulinas/genética , Neurregulinas/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
7.
EMBO J ; 33(13): 1474-91, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24788409

RESUMO

The non-receptor tyrosine kinase c-Src, hereafter referred to as Src, is overexpressed or activated in multiple human malignancies. There has been much speculation about the functional role of Src in colorectal cancer (CRC), with Src amplification and potential activating mutations in up to 20% of the human tumours, although this has never been addressed due to multiple redundant family members. Here, we have used the adult Drosophila and mouse intestinal epithelium as paradigms to define a role for Src during tissue homeostasis, damage-induced regeneration and hyperplasia. Through genetic gain and loss of function experiments, we demonstrate that Src is necessary and sufficient to drive intestinal stem cell (ISC) proliferation during tissue self-renewal, regeneration and tumourigenesis. Surprisingly, Src plays a non-redundant role in the mouse intestine, which cannot be substituted by the other family kinases Fyn and Yes. Mechanistically, we show that Src drives ISC proliferation through upregulation of EGFR and activation of Ras/MAPK and Stat3 signalling. Therefore, we demonstrate a novel essential role for Src in intestinal stem/progenitor cell proliferation and tumourigenesis initiation in vivo.


Assuntos
Transformação Celular Neoplásica/metabolismo , Neoplasias Colorretais/enzimologia , Proteínas de Drosophila/metabolismo , Mucosa Intestinal/enzimologia , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Regeneração , Células-Tronco/enzimologia , Quinases da Família src/metabolismo , Animais , Proteína Tirosina Quinase CSK , Proliferação de Células , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Proteínas de Drosophila/genética , Drosophila melanogaster , Receptores ErbB/genética , Receptores ErbB/metabolismo , Amplificação de Genes , Humanos , Mucosa Intestinal/patologia , Sistema de Sinalização das MAP Quinases/genética , Camundongos , Camundongos Transgênicos , Proteínas Tirosina Quinases/genética , Proteínas Proto-Oncogênicas/genética , Receptores de Peptídeos de Invertebrados/genética , Receptores de Peptídeos de Invertebrados/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Células-Tronco/patologia , Quinases da Família src/genética
8.
Cell Rep ; 6(5): 855-67, 2014 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-24582964

RESUMO

High tumor burden is associated with increased levels of circulating inflammatory cytokines that influence the pathophysiology of the tumor and its environment. The cellular and molecular events mediating the organismal response to a growing tumor are poorly understood. Here, we report a bidirectional crosstalk between epithelial tumors and the fat body-a peripheral immune tissue-in Drosophila. Tumors trigger a systemic immune response through activation of Eiger/TNF signaling, which leads to Toll pathway upregulation in adipocytes. Reciprocally, Toll elicits a non-tissue-autonomous program in adipocytes, which drives tumor cell death. Hemocytes play a critical role in this system by producing the ligands Spätzle and Eiger, which are required for Toll activation in the fat body and tumor cell death. Altogether, our results provide a paradigm for a long-range tumor suppression function of adipocytes in Drosophila, which may represent an evolutionarily conserved mechanism in the organismal response to solid tumors.


Assuntos
Adipócitos/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Membrana/metabolismo , Receptores Toll-Like/metabolismo , Animais , Apoptose/fisiologia , Carcinogênese/metabolismo , Processos de Crescimento Celular/fisiologia , Drosophila melanogaster , Feminino , Hemócitos/citologia , Hemócitos/metabolismo , Masculino , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo
9.
PLoS Genet ; 10(3): e1004262, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24676055

RESUMO

Receptor Tyrosine Kinases (RTKs) and Focal Adhesion Kinase (FAK) regulate multiple signalling pathways, including mitogen-activated protein (MAP) kinase pathway. FAK interacts with several RTKs but little is known about how FAK regulates their downstream signalling. Here we investigated how FAK regulates signalling resulting from the overexpression of the RTKs RET and EGFR. FAK suppressed RTKs signalling in Drosophila melanogaster epithelia by impairing MAPK pathway. This regulation was also observed in MDA-MB-231 human breast cancer cells, suggesting it is a conserved phenomenon in humans. Mechanistically, FAK reduced receptor recycling into the plasma membrane, which resulted in lower MAPK activation. Conversely, increasing the membrane pool of the receptor increased MAPK pathway signalling. FAK is widely considered as a therapeutic target in cancer biology; however, it also has tumour suppressor properties in some contexts. Therefore, the FAK-mediated negative regulation of RTK/MAPK signalling described here may have potential implications in the designing of therapy strategies for RTK-driven tumours.


Assuntos
Neoplasias da Mama/genética , Quinase 1 de Adesão Focal/genética , Sistema de Sinalização das MAP Quinases/genética , Receptores Proteína Tirosina Quinases/genética , Animais , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células , Drosophila melanogaster/genética , Células Epiteliais/metabolismo , Feminino , Quinase 1 de Adesão Focal/metabolismo , Humanos , Fosforilação , Receptores Proteína Tirosina Quinases/metabolismo
10.
Gut ; 63(3): 480-93, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23585469

RESUMO

OBJECTIVE: Colorectal cancer (CRC) is a major contributor to cancer mortality and morbidity. LIM kinase 2 (LIMK2) promotes tumour cell invasion and metastasis. The objectives of this study were to determine how LIMK2 expression is associated with CRC progression and patient outcome, and to use genetically modified Drosophila and mice to determine how LIMK2 deletion affects gastrointestinal stem cell regulation and tumour development. DESIGN: LIMK2 expression and activity were measured by immunostaining tumours from CRC-prone mice, human CRC cell lines and 650 human tumours. LIMK knockdown in Drosophila or Limk2 deletion in mice allowed for assessment of their contributions to gastrointestinal stem cell homeostasis and tumour development. RESULTS: LIMK2 expression was reduced in intestinal tumours of cancer-prone mice, as well as in human CRC cell lines and tumours. Reduced LIMK2 expression and substrate phosphorylation were associated with shorter patient survival. Genetic analysis in Drosophila midgut and intestinal epithelial cells isolated from genetically modified mice revealed a conserved role for LIMK2 in constraining gastrointestinal stem cell proliferation. Limk2 deletion increased colon tumour size in a colitis-associated colorectal mouse cancer model. CONCLUSIONS: This study revealed that LIMK2 expression and activity progressively decrease with advancing stage, and supports the hypothesis that there is selective pressure for reduced LIMK2 expression in CRC to relieve negative constraints imposed upon gastrointestinal stem cells.


Assuntos
Biomarcadores Tumorais/metabolismo , Colo/enzimologia , Neoplasias Colorretais/enzimologia , Mucosa Intestinal/enzimologia , Quinases Lim/metabolismo , Células-Tronco Neoplásicas/enzimologia , Animais , Biomarcadores Tumorais/deficiência , Linhagem Celular Tumoral , Proliferação de Células , Colo/patologia , Colo/fisiopatologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Neoplasias Colorretais/fisiopatologia , Metilação de DNA , Progressão da Doença , Regulação para Baixo , Drosophila melanogaster , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Mucosa Intestinal/patologia , Mucosa Intestinal/fisiopatologia , Quinases Lim/deficiência , Camundongos , Camundongos Knockout , Células-Tronco Neoplásicas/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise Serial de Tecidos
11.
Cell Cycle ; 12(18): 2973-7, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-23974108

RESUMO

Adult stem cells are responsible for maintaining the balance between cell proliferation and differentiation within self-renewing tissues. The molecular and cellular mechanisms mediating such balance are poorly understood. The production of reactive oxygen species (ROS) has emerged as an important mediator of stem cell homeostasis in various systems. Our recent work demonstrates that Rac1-dependent ROS production mediates intestinal stem cell (ISC) proliferation in mouse models of colorectal cancer (CRC). Here, we use the adult Drosophila midgut and the mouse small intestine to directly address the role of Rac1 in ISC proliferation and tissue regeneration in response to damage. Our results demonstrate that Rac1 is necessary and sufficient to drive ISC proliferation and regeneration in an ROS-dependent manner. Our data point to an evolutionarily conserved role of Rac1 in intestinal homeostasis and highlight the value of combining work in the mammalian and Drosophila intestine as paradigms to study stem cell biology.


Assuntos
Proteínas de Drosophila/metabolismo , Intestinos/fisiologia , Regeneração , Células-Tronco/citologia , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Proliferação de Células , Drosophila , Concentração de Íons de Hidrogênio , Camundongos , Camundongos Endogâmicos C57BL , Estresse Oxidativo , Interferência de RNA , Espécies Reativas de Oxigênio/metabolismo , Células-Tronco/metabolismo , Proteínas rac1 de Ligação ao GTP/antagonistas & inibidores , Proteínas rac1 de Ligação ao GTP/genética
12.
Cell Stem Cell ; 12(6): 761-73, 2013 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-23665120

RESUMO

The Adenomatous Polyposis Coli (APC) gene is mutated in the majority of colorectal cancers (CRCs). Loss of APC leads to constitutively active WNT signaling, hyperproliferation, and tumorigenesis. Identification of pathways that facilitate tumorigenesis after APC loss is important for therapeutic development. Here, we show that RAC1 is a critical mediator of tumorigenesis after APC loss. We find that RAC1 is required for expansion of the LGR5 intestinal stem cell (ISC) signature, progenitor hyperproliferation, and transformation. Mechanistically, RAC1-driven ROS and NF-κB signaling mediate these processes. Together, these data highlight that ROS production and NF-κB activation triggered by RAC1 are critical events in CRC initiation.


Assuntos
Neoplasias Colorretais/patologia , Intestino Delgado/citologia , NF-kappa B/metabolismo , Neuropeptídeos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Células-Tronco/citologia , Proteínas Wnt/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Proliferação de Células , Neoplasias Colorretais/metabolismo , Intestino Delgado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais , Células-Tronco/metabolismo
13.
Development ; 139(24): 4524-35, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23172913

RESUMO

Inactivating mutations within adenomatous polyposis coli (APC), a negative regulator of Wnt signaling, are responsible for most sporadic and hereditary forms of colorectal cancer (CRC). Here, we use the adult Drosophila midgut as a model system to investigate the molecular events that mediate intestinal hyperplasia following loss of Apc in the intestine. Our results indicate that the conserved Wnt target Myc and its binding partner Max are required for the initiation and maintenance of intestinal stem cell (ISC) hyperproliferation following Apc1 loss. Importantly, we find that loss of Apc1 leads to the production of the interleukin-like ligands Upd2/3 and the EGF-like Spitz in a Myc-dependent manner. Loss of Apc1 or high Wg in ISCs results in non-cell-autonomous upregulation of upd3 in enterocytes and subsequent activation of Jak/Stat signaling in ISCs. Crucially, knocking down Jak/Stat or Spitz/Egfr signaling suppresses Apc1-dependent ISC hyperproliferation. In summary, our results uncover a novel non-cell-autonomous interplay between Wnt/Myc, Egfr and Jak/Stat signaling in the regulation of intestinal hyperproliferation. Furthermore, we present evidence suggesting potential conservation in mouse models and human CRC. Therefore, the Drosophila adult midgut proves to be a powerful genetic system to identify novel mediators of APC phenotypes in the intestine.


Assuntos
Proteínas de Drosophila/fisiologia , Drosophila , Receptores ErbB/fisiologia , Intestinos/patologia , Janus Quinases/fisiologia , Receptores de Peptídeos de Invertebrados/fisiologia , Fatores de Transcrição STAT/fisiologia , Fatores de Transcrição/fisiologia , Células-Tronco Adultas/metabolismo , Células-Tronco Adultas/patologia , Células-Tronco Adultas/fisiologia , Fatores Etários , Animais , Animais Geneticamente Modificados , Subunidade Apc1 do Ciclossomo-Complexo Promotor de Anáfase , Replicação do DNA/genética , Replicação do DNA/fisiologia , Drosophila/genética , Drosophila/crescimento & desenvolvimento , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Enterócitos/metabolismo , Enterócitos/patologia , Enterócitos/fisiologia , Receptores ErbB/genética , Receptores ErbB/metabolismo , Hiperplasia/genética , Mucosa Intestinal/metabolismo , Janus Quinases/genética , Janus Quinases/metabolismo , Receptor Cross-Talk/fisiologia , Receptores de Peptídeos de Invertebrados/genética , Receptores de Peptídeos de Invertebrados/metabolismo , Fatores de Transcrição STAT/genética , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
14.
EMBO J ; 31(19): 3901-17, 2012 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-22948071

RESUMO

The ability to regenerate following stress is a hallmark of self-renewing tissues. However, little is known about how regeneration differs from homeostatic tissue maintenance. Here, we study the role and regulation of Wingless (Wg)/Wnt signalling during intestinal regeneration using the Drosophila adult midgut. We show that Wg is produced by the intestinal epithelial compartment upon damage or stress and it is exclusively required for intestinal stem cell (ISC) proliferation during tissue regeneration. Reducing Wg or downstream signalling components from the intestinal epithelium blocked tissue regeneration. Importantly, we demonstrate that Wg from the undifferentiated progenitor cell, the enteroblast, is required for Myc-dependent ISC proliferation during regeneration. Similar to young regenerating tissues, ageing intestines required Wg and Myc for ISC hyperproliferation. Unexpectedly, our results demonstrate that epithelial but not mesenchymal Wg is essential for ISC proliferation in response to damage, while neither source of the ligand is solely responsible for ISC maintenance and tissue self-renewal in unchallenged tissues. Therefore, fine-tuning Wnt results in optimal balance between the ability to respond to stress without negatively affecting organismal viability.


Assuntos
Proteínas de Drosophila/fisiologia , Drosophila melanogaster/fisiologia , Intestinos/fisiologia , Regeneração/fisiologia , Células-Tronco/fisiologia , Proteína Wnt1/fisiologia , Animais , Proliferação de Células , Feminino , Transdução de Sinais/fisiologia
15.
Int J Oral Maxillofac Implants ; 27(2): 375-82, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22442778

RESUMO

PURPOSE: This research sought to compare two different systems to monitor sedated patients undergoing implant surgery in the dental office: the bispectral index (BIS) and the Ramsay scale. This information was used to establish an optimal BIS range for surgery in these patients and to calculate differences in drug consumption in both groups. MATERIALS AND METHODS: Consecutive patients undergoing implant surgery were studied and randomly assigned to two groups. Patients were sedated using intravenous propofol, fentanyl, and midazolam. The sedation level in group A was measured using the Ramsay scale. In group B, the Ramsay scale and the BIS were used together. Heart rate, blood pressure, and peripheral oxygen saturation were monitored in all patients. The levels of anxiety, satisfaction, and drug consumption were compared between groups. RESULTS: Forty-three patients were included; 20 were placed in group A and 23 were included in group B. There were no differences in the hemodynamic and respiratory parameters monitored or in anxiety or satisfaction levels in both groups. In group B patients, the BIS values stabilized around 85; the Ramsay scale stabilized around 3 in both groups and remained at these levels until the end of the procedure. Drug consumption was significantly lower in the BIS group. CONCLUSION: The optimal BIS value during intravenous sedation in sedated ambulatory patients in dental surgery should be within the 80 to 85 range. BIS monitoring allows for reduced consumption of propofol, fentanyl, and midazolam.


Assuntos
Sedação Profunda/métodos , Eletroencefalografia/métodos , Monitorização Intraoperatória/métodos , Procedimentos Cirúrgicos Bucais/métodos , Adulto , Idoso , Aumento do Rebordo Alveolar/métodos , Anestesia Dentária/métodos , Anestésicos Intravenosos/administração & dosagem , Ansiedade/classificação , Pressão Sanguínea/efeitos dos fármacos , Estado de Consciência/efeitos dos fármacos , Monitores de Consciência , Sedação Profunda/classificação , Implantação Dentária Endóssea/métodos , Feminino , Fentanila/administração & dosagem , Frequência Cardíaca/efeitos dos fármacos , Humanos , Hipnóticos e Sedativos/administração & dosagem , Masculino , Midazolam/administração & dosagem , Pessoa de Meia-Idade , Oxigênio/sangue , Satisfação do Paciente , Propofol/administração & dosagem , Estudos Prospectivos
16.
Cancer Res ; 72(4): 897-907, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22186138

RESUMO

Hedgehog (Hh) signaling is implicated in bone development and cellular transformation. Here we show that inhibition of Hh pathway activity inhibits tumor growth through effects on the microenvironment. Pharmacologic inhibition of the Hh effector Smoothened (Smo) increased trabecular bone in vivo and inhibited osteoclastogenesis in vitro. In addition, enhanced Hh signaling due to heterozygosity of the Hh inhibitory receptor Patched (Ptch1(+/-)) increased bone resorption, suggesting direct regulation of osteoclast (OC) activity by the Hh pathway. Ptch1(+/-) mice had increased bone metastatic and subcutaneous tumor growth, suggesting that increased Hh activation in host cells promoted tumor growth. Subcutaneous growth of Hh-resistant tumor cells was inhibited by LDE225, a novel orally bioavailable SMO antagonist, consistent with effects on tumor microenvironment. Knockdown of the Hh ligand Sonic Hh (SHH) in these cells decreased subcutaneous tumor growth and decreased stromal cell production of interleukin-6, indicating that tumor-derived Hh ligands stimulated tumor growth in a paracrine fashion. Together our findings show that inhibition of the Hh pathway can reduce tumor burden, regardless of tumor Hh responsiveness, through effects on tumor cells, OCs, and stromal cells within the tumor microenvironment. Hh may be a promising therapeutic target for solid cancers and bone metastases.


Assuntos
Neoplasias Ósseas/prevenção & controle , Osso e Ossos/efeitos dos fármacos , Proteínas Hedgehog/metabolismo , Neoplasias Experimentais/metabolismo , Microambiente Tumoral , Alcaloides de Veratrum/farmacologia , Animais , Neoplasias Ósseas/secundário , Linhagem Celular , Linhagem Celular Tumoral , Feminino , Proteínas Hedgehog/antagonistas & inibidores , Heterozigoto , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Experimentais/patologia , Osteoclastos/efeitos dos fármacos , Receptores Patched , Receptor Patched-1 , Receptores de Superfície Celular/genética , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Transdução de Sinais , Receptor Smoothened
17.
Nat Cell Biol ; 14(1): 51-60, 2011 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-22138575

RESUMO

Here we describe a mechanism that cancer cells use to survive when flux through the Src/FAK pathway is severely perturbed. Depletion of FAK, detachment of FAK-proficient cells or expression of non-phosphorylatable FAK proteins causes sequestration of active Src away from focal adhesions into intracellular puncta that co-stain with several autophagy regulators. Inhibition of autophagy results in restoration of active Src at peripheral adhesions, and this leads to cancer cell death. Autophagic targeting of active Src is associated with a Src-LC3B complex, and is mediated by c-Cbl. However, this is independent of c-Cbl E3 ligase activity, but is mediated by an LC3-interacting region. Thus, c-Cbl-mediated autophagic targeting of active Src can occur in cancer cells to maintain viability when flux through the integrin/Src/FAK pathway is disrupted. This exposes a previously unrecognized cancer cell vulnerability that may provide a new therapeutic opportunity.


Assuntos
Autofagia/fisiologia , Quinase 1 de Adesão Focal/metabolismo , Quinases da Família src/metabolismo , Animais , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Adesão Celular/fisiologia , Sobrevivência Celular/fisiologia , Quinase 1 de Adesão Focal/biossíntese , Quinase 1 de Adesão Focal/genética , Imunoprecipitação , Integrinas/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Proto-Oncogênicas c-cbl/metabolismo , Transdução de Sinais , Transfecção , Células Tumorais Cultivadas
18.
J Genet Genomics ; 38(10): 431-8, 2011 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-22035864

RESUMO

Invasion and metastasis are the most deadly hallmarks of cancer. Once a cancer has acquired the ability to colonize new sites in the body it becomes dramatically more difficult to treat. This has made it a focus of much of cancer research. The humble fruit fly, Drosophila melanogaster, has despite its relative simplicity, made significant contributions to the understanding of tumor progression. In this review we outline and highlight those with an emphasis on modeling the genetic and epigenetic changes required for invasion and metastasis. We will revisit the early years of cancer modeling in Drosophila where the first parallels were drawn between Drosophila and vertebrate neoplasms and highlight recent advances using genetic screens and interactions with the epithelial microenvironment and innate immune system. We focus on the power and limitations of current fly models of metastasis.


Assuntos
Drosophila/genética , Regulação Neoplásica da Expressão Gênica , Imunidade Inata/genética , Invasividade Neoplásica/genética , Metástase Neoplásica/genética , Microambiente Tumoral/genética , Animais , Metilação de DNA , Modelos Animais de Doenças , Epigênese Genética/genética , Humanos
19.
Cell Adh Migr ; 5(4): 366-72, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21836393

RESUMO

Metastasis is the most deadly phase of cancer progression, during which cells detach from their original niche to invade distant tissues, yet the biological processes underlying the spread of cancer are still poorly understood. The fruit fly Drosophila melanogaster provides important insights in our understanding of how epithelial cells migrate from their original location and find their way into surrounding and distant tissues in the metastatic process. Here we review recent studies on the mechanisms of migration of embryonic haemocytes, the macrophage-like immuno-surveillance cells, during normal development and wound healing. We highlight the interesting finding that hydrogen peroxide (H2O2) has been identified as the driving force for haemocyte chemotaxis. We also give a special emphasis to studies suggesting the concept that haemocytes, together with the tumor microenvironment, act as potential inducers of the epithelial de-lamination required for tumor invasion. We propose that cell delamination and migration could be uncoupled from loss of cell polarity via a tumor-related inflammatory response.


Assuntos
Movimento Celular , Drosophila melanogaster/metabolismo , Células Epiteliais/metabolismo , Metástase Neoplásica/patologia , Animais , Moléculas de Adesão Celular/metabolismo , Polaridade Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/embriologia , Desenvolvimento Embrionário , Células Epiteliais/citologia , Transição Epitelial-Mesenquimal , Hemócitos/metabolismo , Peróxido de Hidrogênio/metabolismo , Invasividade Neoplásica/patologia , Neoplasias/patologia , Transdução de Sinais , Cicatrização
20.
Cell Cycle ; 9(19): 3851-6, 2010 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-20935490

RESUMO

The fruit fly Drosophila is an important model for biological research; however, due to its relatively short lifespan its relevance in cancer research is often questioned. Nevertheless, among many other intriguing Drosophila models, scribble group mutants provided early evidence for the existence of tumor suppressor genes and their importance in mammalian systems is beginning to emerge. In this review, I discuss recent advances in our understanding of the phenotypes of scrib group mutants, in which the activation of JNK signaling plays a crucial role. Several mechanisms can account for the activation of JNK within scrib group mutant cells, including a mechanical stress triggered by the loss of polarity, cell competition, intrinsic tumor suppression by autonomous production of Eiger, and an inflammatory response mediated by Eiger-producing haemocytes. Eiger, the sole Drosophila homolog of tumor necrosis factor, is emerging as a 'danger signal' initiated upon the presence of external pathogens, damaged tissues and the appearance of pre-malignant cells. Remarkably, in the presence of the Ras oncoprotein Eiger can act as a tumor promoter by stimulating invasive migration and delaying the onset of metamorphosis.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Neoplasias/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/fisiologia , Ativação Enzimática , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Neoplasias/genética , Neoplasias/patologia , Transdução de Sinais/fisiologia , Fator de Necrose Tumoral alfa/genética , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA