Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Syst ; 15(4): 362-373.e7, 2024 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-38554709

RESUMEN

Predictive modeling of macromolecular recognition and protein-protein complementarity represents one of the cornerstones of biophysical sciences. However, such models are often hindered by the combinatorial complexity of interactions at the molecular interfaces. Exemplary of this problem is peptide presentation by the highly polymorphic major histocompatibility complex class I (MHC-I) molecule, a principal component of immune recognition. We developed human leukocyte antigen (HLA)-Inception, a deep biophysical convolutional neural network, which integrates molecular electrostatics to capture non-bonded interactions for predicting peptide binding motifs across 5,821 MHC-I alleles. These predictions of generated motifs correlate strongly with experimental peptide binding and presentation data. Beyond molecular interactions, the study demonstrates the application of predicted motifs in analyzing MHC-I allele associations with HIV disease progression and patient response to immune checkpoint inhibitors. A record of this paper's transparent peer review process is included in the supplemental information.


Asunto(s)
Antígenos de Histocompatibilidad Clase I , Péptidos , Humanos , Electricidad Estática , Unión Proteica , Péptidos/química , Antígenos HLA/genética , Antígenos HLA/metabolismo
2.
Adv Healthc Mater ; : e2303995, 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38469995

RESUMEN

Rheumatoid arthritis (RA) causes immunological and metabolic imbalances in tissue, exacerbating inflammation in affected joints. Changes in immunological and metabolic tissue homeostasis at different stages of RA are not well understood. Herein, the changes in the immunological and metabolic profiles in different stages in collagen induced arthritis (CIA), namely, early, intermediate, and late stage is examined. Moreover, the efficacy of the inverse-vaccine, paKG(PFK15+bc2) microparticle, to restore tissue homeostasis at different stages is also investigated. Immunological analyses of inverse-vaccine-treated group revealed a significant decrease in the activation of pro-inflammatory immune cells and remarkable increase in regulatory T-cell populations in the intermediate and late stages compared to no treatment. Also, glycolysis in the spleen is normalized in the late stages of CIA in inverse-vaccine-treated mice, which is similar to no-disease tissues. Metabolomics analyses revealed that metabolites UDP-glucuronic acid and L-Glutathione oxidized are significantly altered between treatment groups, and thus might provide new druggable targets for RA treatment. Flux metabolic modeling identified amino acid and carnitine pathways as the central pathways affected in arthritic tissue with CIA progression. Overall, this study shows that the inverse-vaccines initiate early re-establishment of homeostasis, which persists through the disease span.

3.
Nat Commun ; 14(1): 5333, 2023 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-37660049

RESUMEN

Inhibition of glycolysis in immune cells and cancer cells diminishes their activity, and thus combining immunotherapies with glycolytic inhibitors is challenging. Herein, a strategy is presented where glycolysis is inhibited in cancer cells using PFK15 (inhibitor of PFKFB3, rate-limiting step in glycolysis), while simultaneously glycolysis and function is rescued in DCs by delivery of fructose-1,6-biphosphate (F16BP, one-step downstream of PFKFB3). To demonstrate the feasibility of this strategy, vaccine formulations are generated using calcium-phosphate chemistry, that incorporate F16BP, poly(IC) as adjuvant, and phosphorylated-TRP2 peptide antigen and tested in challenging and established YUMM1.1 tumours in immunocompetent female mice. Furthermore, to test the versatility of this strategy, adoptive DC therapy is developed with formulations that incorporate F16BP, poly(IC) as adjuvant and mRNA derived from B16F10 cells as antigens in established B16F10 tumours in immunocompetent female mice. F16BP vaccine formulations rescue DCs in vitro and in vivo, significantly improve the survival of mice, and generate cytotoxic T cell (Tc) responses by elevating Tc1 and Tc17 cells within the tumour. Overall, these results demonstrate that rescuing glycolysis of DCs using metabolite-based formulations can be utilized to generate immunotherapy even in the presence of glycolytic inhibitor.


Asunto(s)
Inmunoterapia , Neoplasias , Femenino , Animales , Ratones , Glucólisis , Adyuvantes Inmunológicos/farmacología , Fructosa , Poli I-C , Células Dendríticas
4.
Biomaterials ; 301: 122292, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37643489

RESUMEN

Succinate is an important metabolite that modulates metabolism of immune cells and cancer cells in the tumor microenvironment (TME). Herein, we report that polyethylene succinate (PES) microparticles (MPs) biomaterial mediated controlled delivery of succinate in the TME modulates macrophage responses. Administering PES MPs locally with or without a BRAF inhibitor systemically in an immune-defective aging mice with clinically relevant BRAFV600E mutated YUMM1.1 melanoma decreased tumor volume three-fold. PES MPs in the TME also led to maintenance of M1 macrophages with up-regulation of TSLP and type 1 interferon pathway. Impressively, this led to generation of pro-inflammatory adaptive immune responses in the form of increased T helper type 1 and T helper type 17 cells in the TME. Overall, our findings from this challenging tumor model suggest that immunometabolism-modifying PES MP strategies provide an approach for developing robust cancer immunotherapies.


Asunto(s)
Melanoma , Ácido Succínico , Animales , Ratones , Macrófagos Asociados a Tumores , Microambiente Tumoral , Proteínas Proto-Oncogénicas B-raf , Succinatos
5.
J Control Release ; 358: 541-554, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37182805

RESUMEN

Boosting the metabolism of immune cells while restricting cancer cell metabolism is challenging. Herein, we report that using biomaterials for the controlled delivery of succinate metabolite to phagocytic immune cells activates them and modulates their metabolism in the presence of metabolic inhibitors. In young immunocompetent mice, polymeric microparticles, with succinate incorporated in the backbone, induced strong pro-inflammatory anti-melanoma responses. Administration of poly(ethylene succinate) (PES MP)-based vaccines and glutaminase inhibitor to young immunocompetent mice with aggressive and large, established B16F10 melanoma tumors increased their survival three-fold, a result of increased cytotoxic T cells expressing RORγT (Tc17). Mechanistically, PES MPs directly modulate glutamine and glutamate metabolism, upregulate succinate receptor SUCNR1, activate antigen presenting cells through and HIF-1alpha, TNFa and TSLP-signaling pathways, and are dependent on alpha-ketoglutarate dehydrogenase for their activity, which demonstrates correlation of succinate delivery and these pathways. Overall, our findings suggest that immunometabolism-modifying PES MP strategies provide an approach for developing robust cancer immunotherapies.


Asunto(s)
Vacunas contra el Cáncer , Melanoma , Animales , Ratones , Polímeros , Ácido Succínico/metabolismo , Inmunoterapia , Transducción de Señal , Células Dendríticas
6.
J Immunother Cancer ; 10(12)2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36564125

RESUMEN

BACKGROUND: Increased infiltration of T cells into ovarian tumors has been repeatedly shown to be predictive of enhanced patient survival. However, despite the evidence of an active immune response in ovarian cancer (OC), the frequency of responses to immune checkpoint blockade (ICB) therapy in OC is much lower than other cancer types. Recent studies have highlighted that deficiencies in the DNA damage response (DDR) can drive increased genomic instability and tumor immunogenicity, which leads to enhanced responses to ICB. Protein phosphatase 4 (PP4) is a critical regulator of the DDR; however, its potential role in antitumor immunity is currently unknown. RESULTS: Our results show that the PP4 inhibitor, fostriecin, combined with carboplatin leads to increased carboplatin sensitivity, DNA damage, and micronuclei formation. Using multiple OC cell lines, we show that PP4 inhibition or PPP4C knockdown combined with carboplatin triggers inflammatory signaling via Nuclear factor kappa B (NF-κB) and signal transducer and activator of transcription 1 (STAT1) activation. This resulted in increased expression of the pro-inflammatory cytokines and chemokines: CCL5, CXCL10, and IL-6. In addition, IFNB1 expression was increased suggesting activation of the type I interferon response. Conditioned media from OC cells treated with the combination of PP4 inhibitor and carboplatin significantly increased migration of both CD8 T cell and natural killer (NK) cells over carboplatin treatment alone. Knockdown of stimulator of interferon genes (STING) in OC cells significantly abrogated the increase in CD8 T-cell migration induced by PP4 inhibition. Co-culture of NK-92 cells and OC cells with PPP4C or PPP4R3B knockdown resulted in strong induction of NK cell interferon-γ, increased degranulation, and increased NK cell-mediated cytotoxicity against OC cells. Stable knockdown of PP4C in a syngeneic, immunocompetent mouse model of OC resulted in significantly reduced tumor growth in vivo. Tumors with PP4C knockdown had increased infiltration of NK cells, NK T cells, and CD4+ T cells. Addition of low dose carboplatin treatment led to increased CD8+ T-cell infiltration in PP4C knockdown tumors as compared with the untreated PP4C knockdown tumors. CONCLUSIONS: Our work has identified a role for PP4 inhibition in promoting inflammatory signaling and enhanced immune cell effector function. These findings support the further investigation of PP4 inhibitors to enhance chemo-immunotherapy for OC treatment.


Asunto(s)
Neoplasias Ováricas , Transducción de Señal , Humanos , Ratones , Animales , Femenino , Carboplatino/farmacología , Carboplatino/uso terapéutico , Neoplasias Ováricas/tratamiento farmacológico , Células Asesinas Naturales , Factor de Transcripción STAT1
7.
Biomaterials ; 277: 121079, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34454372

RESUMEN

Dendritic cells (DCs) rely on glycolysis for their energy needs to induce pro-inflammatory antigen-specific immune responses. Therefore, inhibiting DC glycolysis, while presenting the self-antigen, may prevent pro-inflammatory antigen-specific immune responses. Previously we demonstrated that microparticles with alpha-ketoglutarate (aKG) in the polymer backbone (paKG MPs) were able to generate anti-inflammatory DCs by sustained delivery of the aKG metabolite, and by modulating energy metabolism of DCs. Herein, we demonstrate that paKG MPs-based delivery of a glycolytic inhibitor, PFK15, using paKG MPs induces anti-inflammatory DCs (CD86LoMHCII+) by down-regulating glycolysis, CD86, tnf and IL-6 genes, while upregulating oxidative phosphorylation (OXPHOS) and mitochondrial genes. Furthermore, paKG MPs delivering PFK15 and a self-antigen, collagen type II (bc2), in vivo, in a collagen-induced autoimmune arthritis (CIA) mouse model, normalized paw inflammation and arthritis score, by generating antigen-specific immune responses. Specifically, these formulations were able to reduce activation of DCs in draining lymph nodes and impressively generated proliferating bc2-specific anti-inflammatory regulatory T cells in joint-associated popliteal lymph nodes. These data strongly suggest that sustained glycolytic inhibition of DCs in the presence of an antigen can induce antigen-specific immunosuppressive responses, therefore, generating a technology that can be applicable for treating autoimmune diseases.


Asunto(s)
Artritis Experimental , Artritis Reumatoide , Animales , Artritis Reumatoide/tratamiento farmacológico , Células Dendríticas , Glucólisis , Ratones , Ratones Endogámicos DBA , Piridinas , Quinolinas
8.
Cancers (Basel) ; 13(13)2021 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-34283042

RESUMEN

Disruption of metabolic homeostasis at the organismal level can cause metabolic syndrome associated with obesity. The role of adipose tissue in cancer has been investigated over the last several decades with many studies implicating obesity as a risk factor for the development of cancer. Adipose tissue contains a diverse array of immune cell populations that promote metabolic homeostasis through a tightly controlled balance of pro- and anti-inflammatory signals. During obesity, pro-inflammatory cell types infiltrate and expand within the adipose tissue, exacerbating metabolic dysfunction. Some studies have now shown that the intracellular metabolism of immune cells is also deregulated by the lipid-rich environment in obesity. What is not fully understood, is how this may influence cancer progression, metastasis, and anti-tumor immunity. This review seeks to highlight our current understanding of the effect of adipose tissue on immune cell function and discuss how recent results offer new insight into the role that adipose tissue plays in cancer progression and anti-tumor immunity.

9.
J Mater Chem B ; 8(24): 5195-5203, 2020 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-32427266

RESUMEN

Metabolites control immune cell functions, and delivery of these metabolites in a sustained manner may be able to modulate function of the immune cells. In this study, alpha-ketoglutarate (aKG) and diol based polymeric-microparticles (termed paKG MPs) were synthesized to provide sustained release of aKG and promote an immunosuppressive cellular phenotype. Notably, after association with dendritic cells (DCs), paKG MPs modulated the intracellular metabolic-profile/pathways, and decreased glycolysis and mitochondrial respiration in vitro. These metabolic changes resulted in modulation of MHC-II, CD86 expression in DCs, and altered the frequency of regulatory T cells (Tregs), and T-helper type-1/2/17 cells in vitro. This unique strategy of intracellular delivery of key-metabolites in a sustained manner provides a new direction in immunometabolism field-based immunotherapy with potential applications in different diseases associated with immune disorders.


Asunto(s)
Células Dendríticas/metabolismo , Metabolismo Energético , Polímeros/metabolismo , Células Dendríticas/química , Humanos , Tamaño de la Partícula , Polímeros/química , Propiedades de Superficie , Linfocitos T Reguladores/metabolismo , Células TH1/metabolismo
10.
Nature ; 569(7758): 723-728, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31043742

RESUMEN

High-grade serous carcinoma has a poor prognosis, owing primarily to its early dissemination throughout the abdominal cavity. Genomic and proteomic approaches have provided snapshots of the proteogenomics of ovarian cancer1,2, but a systematic examination of both the tumour and stromal compartments is critical in understanding ovarian cancer metastasis. Here we develop a label-free proteomic workflow to analyse as few as 5,000 formalin-fixed, paraffin-embedded cells microdissected from each compartment. The tumour proteome was stable during progression from in situ lesions to metastatic disease; however, the metastasis-associated stroma was characterized by a highly conserved proteomic signature, prominently including the methyltransferase nicotinamide N-methyltransferase (NNMT) and several of the proteins that it regulates. Stromal NNMT expression was necessary and sufficient for functional aspects of the cancer-associated fibroblast (CAF) phenotype, including the expression of CAF markers and the secretion of cytokines and oncogenic extracellular matrix. Stromal NNMT expression supported ovarian cancer migration, proliferation and in vivo growth and metastasis. Expression of NNMT in CAFs led to depletion of S-adenosyl methionine and reduction in histone methylation associated with widespread gene expression changes in the tumour stroma. This work supports the use of ultra-low-input proteomics to identify candidate drivers of disease phenotypes. NNMT is a central, metabolic regulator of CAF differentiation and cancer progression in the stroma that may be therapeutically targeted.


Asunto(s)
Fibroblastos Asociados al Cáncer/metabolismo , Nicotinamida N-Metiltransferasa/metabolismo , Proteómica , Fibroblastos Asociados al Cáncer/enzimología , Línea Celular Tumoral , Células Cultivadas , Metilación de ADN , Progresión de la Enfermedad , Femenino , Histonas/química , Histonas/metabolismo , Humanos , Metástasis de la Neoplasia , Niacinamida/análogos & derivados , Niacinamida/metabolismo , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Fenotipo , Pronóstico , S-Adenosilhomocisteína/metabolismo , S-Adenosilmetionina/metabolismo
11.
Mol Cancer Res ; 17(4): 870-881, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30655321

RESUMEN

The role of phospholipid signaling in ovarian cancer is poorly understood. Sphingosine-1-phosphate (S1P) is a bioactive metabolite of sphingosine that has been associated with tumor progression through enhanced cell proliferation and motility. Similarly, sphingosine kinases (SPHK), which catalyze the formation of S1P and thus regulate the sphingolipid rheostat, have been reported to promote tumor growth in a variety of cancers. The findings reported here show that exogenous S1P or overexpression of SPHK1 increased proliferation, migration, invasion, and stem-like phenotypes in ovarian cancer cell lines. Likewise, overexpression of SPHK1 markedly enhanced tumor growth in a xenograft model of ovarian cancer, which was associated with elevation of key markers of proliferation and stemness. The diabetes drug, metformin, has been shown to have anticancer effects. Here, we found that ovarian cancer patients taking metformin had significantly reduced serum S1P levels, a finding that was recapitulated when ovarian cancer cells were treated with metformin and analyzed by lipidomics. These findings suggested that in cancer the sphingolipid rheostat may be a novel metabolic target of metformin. In support of this, metformin blocked hypoxia-induced SPHK1, which was associated with inhibited nuclear translocation and transcriptional activity of hypoxia-inducible factors (HIF1α and HIF2α). Further, ovarian cancer cells with high SPHK1 were found to be highly sensitive to the cytotoxic effects of metformin, whereas ovarian cancer cells with low SPHK1 were resistant. Together, the findings reported here show that hypoxia-induced SPHK1 expression and downstream S1P signaling promote ovarian cancer progression and that tumors with high expression of SPHK1 or S1P levels might have increased sensitivity to the cytotoxic effects of metformin. IMPLICATIONS: Metformin targets sphingolipid metabolism through inhibiting SPHK1, thereby impeding ovarian cancer cell migration, proliferation, and self-renewal.


Asunto(s)
Metformina/farmacología , Neoplasias Ováricas/tratamiento farmacológico , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Movimiento Celular/efectos de los fármacos , Femenino , Humanos , Hipoglucemiantes/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia , Lisofosfolípidos/metabolismo , Ratones , Ratones Desnudos , Terapia Molecular Dirigida , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Cell Metab ; 29(1): 141-155.e9, 2019 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-30174305

RESUMEN

Successful metastasis requires the co-evolution of stromal and cancer cells. We used stable isotope labeling of amino acids in cell culture coupled with quantitative, label-free phosphoproteomics to study the bidirectional signaling in ovarian cancer cells and human-derived, cancer-associated fibroblasts (CAFs) after co-culture. In cancer cells, the interaction with CAFs supported glycogenolysis under normoxic conditions and induced phosphorylation and activation of phosphoglucomutase 1, an enzyme involved in glycogen metabolism. Glycogen was funneled into glycolysis, leading to increased proliferation, invasion, and metastasis of cancer cells co-cultured with human CAFs. Glycogen mobilization in cancer cells was dependent on p38α MAPK activation in CAFs. In vivo, deletion of p38α in CAFs and glycogen phosphorylase inhibition in cancer cells reduced metastasis, suggesting that glycogen is an energy source used by cancer cells to facilitate metastatic tumor growth.


Asunto(s)
Fibroblastos Asociados al Cáncer/metabolismo , Glucógeno/metabolismo , Neoplasias Ováricas/metabolismo , Animales , Fibroblastos Asociados al Cáncer/patología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Técnicas de Cocultivo/métodos , Femenino , Glucólisis , Humanos , Sistema de Señalización de MAP Quinasas , Ratones Desnudos , Microambiente Tumoral
13.
Cell ; 175(1): 159-170.e16, 2018 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-30241606

RESUMEN

Most high-grade serous ovarian cancer (HGSOC) patients develop resistance to platinum-based chemotherapy and recur, but 15% remain disease free over a decade. To discover drivers of long-term survival, we quantitatively analyzed the proteomes of platinum-resistant and -sensitive HGSOC patients from minute amounts of formalin-fixed, paraffin-embedded tumors. This revealed cancer/testis antigen 45 (CT45) as an independent prognostic factor associated with a doubling of disease-free survival in advanced-stage HGSOC. Phospho- and interaction proteomics tied CT45 to DNA damage pathways through direct interaction with the PP4 phosphatase complex. In vitro, CT45 regulated PP4 activity, and its high expression led to increased DNA damage and platinum sensitivity. CT45-derived HLA class I peptides, identified by immunopeptidomics, activate patient-derived cytotoxic T cells and promote tumor cell killing. This study highlights the power of clinical cancer proteomics to identify targets for chemo- and immunotherapy and illuminate their biological roles.


Asunto(s)
Antígenos de Neoplasias/fisiología , Resistencia a Antineoplásicos/genética , Proteómica/métodos , Anciano , Secuencia de Aminoácidos/genética , Antineoplásicos/uso terapéutico , Metilación de ADN/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Inmunoterapia/métodos , Estimación de Kaplan-Meier , Persona de Mediana Edad , Recurrencia Local de Neoplasia/tratamiento farmacológico , Neoplasias Ováricas/tratamiento farmacológico , Fosfoproteínas Fosfatasas/metabolismo , Fosfoproteínas Fosfatasas/fisiología , Pronóstico
14.
Trends Cancer ; 4(8): 517-519, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30064659

RESUMEN

Historically, cancer has been studied at the genomic level with a focus on malignant cells. However, tumors are comprised of various host cells, which are co-opted by cancer cells to promote tumor growth and metastasis. Using multilevel analysis of human tumor biopsies, recent work identifies a reproducible 22-gene expression signature in the ovarian tumor microenvironment (TME) that correlates with disease progression and survival. These findings suggest that the TME is a promising therapeutic target that could be effective across a wide patient population, regardless of the genetic changes within their tumor cells.


Asunto(s)
Carcinoma Epitelial de Ovario , Neoplasias Ováricas , Progresión de la Enfermedad , Femenino , Humanos , Microambiente Tumoral
16.
Nat Commun ; 6: 6220, 2015 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-25653139

RESUMEN

The tumour microenvironment contributes to cancer metastasis and drug resistance. However, most high throughput screening (HTS) assays for drug discovery use cancer cells grown in monolayers. Here we show that a multilayered culture containing primary human fibroblasts, mesothelial cells and extracellular matrix can be adapted into a reliable 384- and 1,536-multi-well HTS assay that reproduces the human ovarian cancer (OvCa) metastatic microenvironment. We validate the identified inhibitors in secondary in vitro and in vivo biological assays using three OvCa cell lines: HeyA8, SKOV3ip1 and Tyk-nu. The active compounds directly inhibit at least two of the three OvCa functions: adhesion, invasion and growth. In vivo, these compounds prevent OvCa adhesion, invasion and metastasis, and improve survival in mouse models. Collectively, these data indicate that a complex three-dimensional culture of the tumour microenvironment can be adapted for quantitative HTS and may improve the disease relevance of assays used for drug screening.


Asunto(s)
Antineoplásicos/farmacología , Matriz Extracelular/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento/métodos , Neoplasias Ováricas/tratamiento farmacológico , Microambiente Tumoral/efectos de los fármacos , Animales , Antineoplásicos/química , Benzofenantridinas/química , Benzofenantridinas/farmacología , Biguanidas/química , Biguanidas/farmacología , Cantaridina/química , Cantaridina/farmacología , Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Técnicas de Cocultivo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Escina/química , Escina/farmacología , Matriz Extracelular/metabolismo , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Ensayos Analíticos de Alto Rendimiento/instrumentación , Humanos , Concentración 50 Inhibidora , Isoquinolinas/química , Isoquinolinas/farmacología , Ratones , Ratones Desnudos , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Cultivo Primario de Células , Proclorperazina/química , Proclorperazina/farmacología , Tomatina/química , Tomatina/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Public Health Nurs ; 25(4): 383-6, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18666945

RESUMEN

An historical reprint, this account of a public health nurse's work with village residents during an outbreak of pneumonia related to tuberculosis was originally published in Public Health Nursing in December 1948. The author, Marion Curtis, was a public health nurse in the Territorial Health Department of Alaska called upon to respond to the rapid death of several victims of an unknown disease affecting a small outpost on Kodiak Island in the Territory of Alaska. Her narrative tells us about the settlement village and what happened from the time of the wire for help to the time she left the community for home. Her original spelling of names has been retained. Curtis's response to the emergency was quick and demonstrated experience. Her application of infection control principles helped quickly control the outbreak. At the end of her story, Curtis reflected on the impact of disease on communities and civilizations, predicting that villages like the one she had described might vanish within 20 years due to tuberculosis. Readers may reflect whether Curtis's view presents an etic or an emic perspective. Old Harbor, however, remains a small but vital community on Kodiak Island's southwestern shore.


Asunto(s)
Brotes de Enfermedades , Control de Infecciones/métodos , Neumonía/epidemiología , Enfermería en Salud Pública , Población Rural , Alaska/epidemiología , Anécdotas como Asunto , Humanos , Neumonía/etiología , Tuberculosis/complicaciones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...