Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Artigo em Inglês | MEDLINE | ID: mdl-36178750

RESUMO

The Choctaw Nation Health Care Center established a first responder naloxone program in 2015. Limited data is available on community naloxone programs specific to tribal communities and the opinions of first responders who may utilize naloxone in the field. The purpose of this article is to highlight the model of a tribal first responder naloxone program in Talihina, Oklahoma and present analysis of the impact of program trainings on first responders' understanding and willingness to administer intranasal naloxone through pre- and post-surveys (n = 758) collected from May 2018 to November 2019. Descriptive analyses were conducted to compare first responders' rating of their support, willingness, and confidence in using naloxone. Overall, 95.1% of first responders reported learning something new from the training. However, the most significant changes in pre- to post-test results were among first responders that had never been at the scene of an overdose. Almost 77% of trainees who reported they never were at a scene of an overdose and responded "not very willing" in administering naloxone at pre-test, responded that they were "very willing" to administer naloxone at post-test.


Assuntos
Overdose de Drogas , Socorristas , Indígenas Norte-Americanos , Analgésicos Opioides/uso terapêutico , Overdose de Drogas/tratamento farmacológico , Humanos , Naloxona/uso terapêutico , Antagonistas de Entorpecentes/uso terapêutico
3.
Oncotarget ; 7(27): 41217-41232, 2016 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-27172799

RESUMO

Dissemination of cancer stem cells (CSCs) serves as the basis of metastasis. Recently, we demonstrated that circulating prostate cancer targets the hematopoietic stem cell (HSCs) 'niche' in marrow during dissemination. Once in the niche, disseminated tumor cells (DTCs) may remain dormant for extended periods. As the major function of the HSC niche is to maintain stem cell functions, we hypothesized that the niche regulates CSC activities of DTCs. Here we show that DTCs recovered from marrow were significantly enriched for a CSC phenotype. Critically, the conversion of DTCs to CSCs is regulated by niche-derived GAS6 through the Mer/mTOR; molecules previously shown to regulate dormancy. The data demonstrate that the niche plays a significant role in maintaining tumor-initiating prostate cancer in marrow and suggests a functional relationship between CSCs and dormancy. Understanding how the marrow niche regulates the conversion of DTCs to CSCs is critical for the development of therapeutics specifically targeting skeletal bone metastasis and dormancy.


Assuntos
Adenocarcinoma/patologia , Medula Óssea/patologia , Neoplasias Ósseas/secundário , Células-Tronco Hematopoéticas/patologia , Células-Tronco Neoplásicas/patologia , Neoplasias da Próstata/patologia , Nicho de Células-Tronco/fisiologia , Animais , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Células-Tronco Hematopoéticas/fisiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Metástase Neoplásica , Células-Tronco Neoplásicas/fisiologia , Fenótipo
4.
Mol Cancer Res ; 13(1): 197-207, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25139998

RESUMO

UNLABELLED: Annexin 2 (ANXA2) plays a critical role in hematopoietic stem cell (HSC) localization to the marrow niche. In part, ANXA2 supports HSCs by serving as an anchor for stromal-derived factor-1 (CXCL12/SDF-1). Recently, it was demonstrated that prostate cancer cells, like HSCs, use ANXA2 to establish metastases in marrow. The present study determined the capacity of ANXA2 expression by bone marrow stromal cells (BMSC) to facilitate tumor recruitment and growth through ANXA2-CXCL12 interactions. Significantly more CXCL12 was expressed by BMSC(Anxa2) (+/+) than by BMSC(Anxa2) (-/-) resulting in more prostate cancer cells migrating and binding to BMSC(Anxa2) (+/+) than BMSC(Anxa2) (-/-), and these activities were reduced when CXCL12 interactions were blocked. To further confirm that BMSC signaling through ANXA2-CXCL12 plays a critical role in tumor growth, immunocompromised SCID mice were subcutaneously implanted with human prostate cancer cells mixed with BMSC(Anxa2) (+/+) or BMSC(Anxa2) (-/-). Significantly larger tumors grew in the mice when the tumors were established with BMSC(Anxa2) (+/+) compared with the tumors established with BMSC(Anxa2) (-/-). In addition, fewer prostate cancer cells underwent apoptosis when cocultured with BMSC(Anxa2) (+/+) compared with BMSC(Anxa2) (-/-), and similar results were obtained in tumors grown in vivo. Finally, significantly more vascular structures were observed in the tumors established with the BMSC(Anxa2) (+/+) compared with the tumors established with BMSC(Anxa2) (-/-). Thus, ANXA2-CXCL12 interactions play a crucial role in the recruitment, growth, and survival of prostate cancer cells in the marrow. IMPLICATIONS: The tumor microenvironment interaction between ANXA2-CXCL12 is critical for metastatic phenotypes and may impact chemotherapeutic potential.


Assuntos
Anexina A2/genética , Quimiocina CXCL12/genética , Células-Tronco Mesenquimais/metabolismo , Neoplasias da Próstata/genética , Animais , Anexina A2/biossíntese , Quimiocina CXCL12/biossíntese , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Células-Tronco Mesenquimais/patologia , Camundongos , Metástase Neoplásica , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/patologia , Transdução de Sinais
5.
J Cell Biochem ; 114(11): 2471-8, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23696192

RESUMO

Erythropoietin (Epo) is used in clinical settings to enhance hematopoietic function and to improve the quality of life for patients undergoing chemotherapy by reducing fatigue and the need for transfusions. However, several meta-analyses have revealed that Epo treatments are associated with an increased risk of mortality in cancer patients. In this study, we examined the role of Epo in prostate cancer (PCa) progression, using in vitro cell culture systems and in vivo bone metastatic assays. We found that Epo did not stimulate the proliferation of PCa cell lines, but did protect PCa cells from apoptosis. In animal models of PCa metastasis, no evidence was found to support the hypothesis that Epo enhances metastasis. Together, these findings suggest that Epo may be useful for treating severe anemia in PCa patients without increasing metastatic risk.


Assuntos
Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/secundário , Proliferação de Células/efeitos dos fármacos , Eritropoetina/uso terapêutico , Metástase Neoplásica/prevenção & controle , Neoplasias da Próstata/complicações , Neoplasias da Próstata/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Linhagem Celular Tumoral , Citometria de Fluxo , Humanos , Masculino , Camundongos , Camundongos SCID
6.
Nat Commun ; 4: 1795, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23653207

RESUMO

Tumours recruit mesenchymal stem cells to facilitate healing, which induces their conversion into cancer-associated fibroblasts that facilitate metastasis. However, this process is poorly understood on the molecular level. Here we show that CXCL16, a ligand for CXCR6, facilitates mesenchymal stem cell or very small embryonic-like cells recruitment into prostate tumours. CXCR6 signalling stimulates the conversion of mesenchymal stem cells into cancer-associated fibroblasts, which secrete stromal-derived factor-1, also known as CXCL12. CXCL12 expressed by cancer-associated fibroblasts then binds to CXCR4 on tumour cells and induces an epithelial-to-mesenchymal transition, which ultimately promotes metastasis to secondary tumour sites. Our results provide the molecular basis for mesenchymal stem cell recruitment into tumours and how this process leads to tumour metastasis.


Assuntos
Células-Tronco Mesenquimais/patologia , Neoplasias da Próstata/patologia , Animais , Células da Medula Óssea/patologia , Proliferação de Células , Quimiocina CXCL12/metabolismo , Quimiocina CXCL16 , Quimiocina CXCL6/metabolismo , Quimiocinas CXC/metabolismo , Transição Epitelial-Mesenquimal , Fibroblastos/metabolismo , Fibroblastos/patologia , Técnicas de Silenciamento de Genes , Humanos , Masculino , Células-Tronco Mesenquimais/metabolismo , Camundongos , Modelos Biológicos , Metástase Neoplásica , Neoplasias da Próstata/metabolismo , Receptores CXCR/metabolismo , Receptores CXCR4/metabolismo , Receptores CXCR6 , Receptores de Quimiocinas/metabolismo , Receptores Depuradores/metabolismo , Receptores Virais/metabolismo , Transdução de Sinais
7.
Stem Cells Dev ; 22(4): 622-30, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23020187

RESUMO

Human very small embryonic-like (hVSEL) cells are a resident population of multipotent stem cells in the bone marrow involved in the turnover and regeneration of tissues. The levels of VSEL cells in blood are greatly increased in response to injury, and they have been shown to repair injured tissues. Adult hVSEL cells, SSEA-4(+)/CD133(+)/CXCR4(+)/Lin(-)/CD45(-), express the pluripotency markers (Oct-4 and Nanog) and may be able to differentiate into cells from all 3 germ lineages. hVSEL cells isolated from blood by apheresis following granulocyte-colony-stimulating factor mobilization were fractionated and enriched by elutriation and fluorescence activated cell sorting. Collagen sponge scaffolds containing 2,000-30,000 hVSEL cells were implanted into cranial defects generated in SCID mice. Analysis by microcomputed tomography showed that a cell population containing VSEL cells produced mineralized tissue within the cranial defects compared with controls at 3 months. Histologic studies showed significant bone formation and cellular organization within the defects compared with cellular or scaffold controls alone. Antibodies to human leukocyte antigens demonstrated that the newly generated tissues were of human origin. Moreover, human osteocalcin was identified circulating in the peripheral blood. There was evidence that some level of hVSEL cells migrated away from the defect site, using quantitative real-time polymerase chain reaction to detect for human-specific Alu sequences. This study demonstrates that hVSEL cells are able to generate human bone tissue in a mouse model of skeletal repair. These studies lay the foundation for future cell-based regenerative therapies for osseous and connective tissue disorders, including trauma and degenerative conditions, such as osteoporosis, fracture repair, and neoplastic repair.


Assuntos
Movimento Celular , Células-Tronco Embrionárias , Osteogênese , Células-Tronco Pluripotentes , Crânio/lesões , Transplante de Células-Tronco , Adulto , Animais , Antígenos de Diferenciação/biossíntese , Remoção de Componentes Sanguíneos , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/transplante , Feminino , Citometria de Fluxo , Humanos , Masculino , Camundongos , Camundongos SCID , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/transplante , Crânio/metabolismo , Transplante Heterólogo
8.
Mol Cancer Res ; 10(6): 703-12, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22516347

RESUMO

The receptor tyrosine kinase Axl is overexpressed in a variety of cancers and is known to play a role in proliferation and invasion. Previous data from our laboratory indicate that Axl and its ligand growth arrest-specific 6 (GAS6) may play a role in establishing metastatic dormancy in the bone marrow microenvironment. In the current study, we found that Axl is highly expressed in metastatic prostate cancer cell lines PC3 and DU145 and has negligible levels of expression in a nonmetastatic cancer cell line LNCaP. Knockdown of Axl in PC3 and DU145 cells resulted in decreased expression of several mesenchymal markers including Snail, Slug, and N-cadherin, and enhanced expression of the epithelial marker E-cadherin, suggesting that Axl is involved in the epithelial-mesenchymal transition in prostate cancer cells. The Axl-knockdown PC3 and DU145 cells also displayed decreased in vitro migration and invasion. Interestingly, when PC3 and DU145 cells were treated with GAS6, Axl protein levels were downregulated. Moreover, CoCl(2), a hypoxia mimicking agent, prevented GAS6-mediated downregulation of Axl in these cell lines. Immunochemical staining of human prostate cancer tissue microarrays showed that Axl, GAS6, and hypoxia-inducible factor-1α (Hif-1α; indicator of hypoxia) were all coexpressed in prostate cancer and in bone metastases compared with normal tissues. Together, our studies indicate that Axl plays a crucial role in prostate cancer metastasis and that GAS6 regulates the expression of Axl. Importantly, in a hypoxic tumor microenvironment Axl expression is maintained leading to enhanced signaling.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Neoplasias da Próstata/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Transdução de Sinais , Antimutagênicos/farmacologia , Western Blotting , Caderinas/metabolismo , Hipóxia Celular , Linhagem Celular Tumoral , Cobalto/farmacologia , Regulação para Baixo/efeitos dos fármacos , Transição Epitelial-Mesenquimal , Regulação Neoplásica da Expressão Gênica , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Masculino , Metástase Neoplásica , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas/genética , Interferência de RNA , Receptores Proteína Tirosina Quinases/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise Serial de Tecidos , Receptor Tirosina Quinase Axl
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...