Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 142
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Rural Remote Health ; 23(3): 7809, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37429740

RESUMEN

INTRODUCTION: Major inequities exist in levels of health and wellbeing, availability, and access to healthcare services between seniors of Indigenous and non-Indigenous background in Ontario. First Nations elders are 45-55% more frail than the average senior in Ontario. Additionally, needed rehabilitation services are not easily accessible or available in the first language of most First Nations elders within their home communities. A literature review demonstrated community-based rehabilitation assistant models had been successfully developed and implemented in regions facing similar equity and access challenges. Building on these findings, a needs assessment was conducted to capture unique needs and requirements in Northwestern Ontario relating to rehabilitation among First Nations elders. METHODS: The needs assessment resulted in four First Nations, three Indigenous health organizations, three rehabilitation health organizations, and two academic institutions iteratively developing and evaluating curriculum for a Community Rehabilitation Worker (CRW) program in treaty territories 5, 9, and Robinson-Superior. The goal of the program is to train local CRWs, familiar with local languages and cultures, to provide rehabilitative services that support ageing in place, health, wellbeing, and quality of life for First Nations elders. The study employed a community participatory action research approach aligning with the OCAP® (Ownership, Control, Access, and Possession) framework for working with Indigenous populations. Seventeen community partners were active participants in the program development, evaluation, and adaptation of the CRW curriculum. Feedback was received through advisory committee meetings, surveys, and individual and group interviews. RESULTS: All 101 participants agreed, across all curriculum modules, that (1) the time allotment was realistic; (2) instructional materials, activities, and resources were appropriate and easy to understand; (3) evaluation activities accurately measured learning; and (4) participants identifying as Indigenous felt that Indigenous culture was adequately reflected. The qualitative findings highlighted the importance of incorporating culture, spirituality, traditions, local language use, and reintegration of First Nations elders into traditional activities and community activities for both the CRW curriculum and rehabilitation efforts. The need for locally available First Nations, elder-focused mental health support, transportation options, and gathering spaces such as those commonly seen in urban areas was also highlighted. CONCLUSION: The process of iteratively developing and evaluating a CRW program resulted in a Northwestern Ontario college welcoming the first cohort of students to the CRW program in March 2022. The program is co-facilitated with a First Nations Elder and includes components of local culture, language, and the reintegration of First Nations elders into community as part of the rehabilitation efforts. In addition, to appropriately support the quality of life, health, and wellbeing of First Nations elders, the project team called upon provincial and federal governments to work with First Nations to make available dedicated funding to address inequities in resources available to First Nations elders in Northwestern Ontario urban and First Nations remote communities. This included elder-focused transportation options, mental health services, and gathering places. The program implementation will be evaluated with the first cohort of CRWs for further adaptations considering potential scale and spread. As such, the project and findings may also represent a resource for others wishing to pursue similar development using participatory approaches in rural and remote communities both nationally and internationally.


Asunto(s)
Medicina , Calidad de Vida , Anciano , Humanos , Ontario , Vida Independiente , Pueblos Indígenas
2.
Exp Cell Res ; 374(1): 38-45, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30419192

RESUMEN

IGFBP-3 has both stimulatory and inhibitory effects on cancer progression. The growth of EO771 mammary carcinoma cells as syngeneic tumors in C57BL/6 mice is reduced in Igfbp3-null (BP3KO) mice, suggesting that systemic IGFBP-3 enhances tumor progression. In this study we assessed the growth of EO771 cells expressing human IGFBP-3 in BP3KO mice. Cells expressing hIGFBP-3 showed decreased proliferation in vitro and increased levels of IGF-1 receptor (IGF1R) protein but not mRNA, consistent with sequestration of endogenous IGF by IGFBP-3. The growth rate of these cells was restored by exposure to IGF-1 or analogues with reduced affinity for IGFBP-3 (long Arg3-IGF-1) or IGF1R (Leu24-IGF-1). In EO771 cells implanted orthotopically into mice, hIGFBP-3 expression by the cells inhibited tumor establishment in BP3KO but not wild-type mice. For tumors that successfully established, final weight was not affected significantly by hIGFBP-3 expression. However, final tumor weight was inversely related to intratumoral T cell counts, and sera from BP3KO mice with tumors showed low-titer immunoreactivity against IGFBP-3. The contrasting effects on tumor establishment and progression of IGFBP-3 expressed by mammary carcinoma cells, compared to systemic stromal and circulating IGFBP-3, highlights the complexity of growth regulation by IGFBP-3 in mammary tumors.


Asunto(s)
Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/patología , Microambiente Tumoral , Inmunidad Adaptativa , Tejido Adiposo/patología , Animales , Anticuerpos/sangre , Anticuerpos/metabolismo , Línea Celular Tumoral , Proliferación Celular , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias Mamarias Animales/sangre , Neoplasias Mamarias Animales/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Microambiente Tumoral/inmunología
3.
Cell Mol Life Sci ; 76(10): 2015-2030, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30725116

RESUMEN

Women with triple-negative breast cancer (TNBC) are generally treated by chemotherapy but their responsiveness may be blunted by DNA double-strand break (DSB) repair. We previously reported that IGFBP-3 forms nuclear complexes with EGFR and DNA-dependent protein kinase (DNA-PKcs) to modulate DSB repair by non-homologous end-joining (NHEJ) in TNBC cells. To discover IGFBP-3 binding partners involved in chemoresistance through stimulation of DSB repair, we analyzed the IGFBP-3 interactome by LC-MS/MS and confirmed interactions by coimmunoprecipitation and proximity ligation assay. Functional effects were demonstrated by DNA end-joining in vitro and measurement of γH2AX foci. In response to 20 µM etoposide, the DNA/RNA-binding protein, non-POU domain-containing octamer-binding protein (NONO) and its dimerization partner splicing factor, proline/glutamine-rich (SFPQ) formed complexes with IGFBP-3, demonstrated in basal-like TNBC cell lines HCC1806 and MDA-MB-468. NONO binding to IGFBP-3 was also shown in a cell-free biochemical assay. IGFBP-3 complexes with NONO and SFPQ were blocked by inhibiting EGFR with gefitinib or DNA-PKcs with NU7026, and by the PARP inhibitors veliparib and olaparib, which also reduced DNA end-joining activity and delayed the resolution of the γH2AX signal (i.e. inhibited DNA DSB repair). Downregulation of the long noncoding RNA in NHEJ pathway 1 (LINP1) by siRNA also blocked IGFBP-3 interaction with NONO-SFPQ. These findings suggest a PARP-dependent role for NONO and SFPQ in IGFBP-3-dependent DSB repair and the involvement of LINP1 in the complex formation. We propose that targeting of the DNA repair function of IGFBP-3 may enhance chemosensitivity in basal-like TNBC, thus improving patient outcomes.


Asunto(s)
Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Proteínas Asociadas a Matriz Nuclear/metabolismo , Factores de Transcripción de Octámeros/metabolismo , Factor de Empalme Asociado a PTB/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas de Unión al ARN/metabolismo , Bencimidazoles/farmacología , Línea Celular Tumoral , Reparación del ADN por Unión de Extremidades/efectos de los fármacos , Proteínas de Unión al ADN , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Proteínas Asociadas a Matriz Nuclear/genética , Factores de Transcripción de Octámeros/genética , Factor de Empalme Asociado a PTB/genética , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Poli(ADP-Ribosa) Polimerasas/genética , Unión Proteica/efectos de los fármacos , Unión Proteica/genética , Interferencia de ARN , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Proteínas de Unión al ARN/genética , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología
4.
Breast Cancer Res ; 19(1): 90, 2017 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-28778177

RESUMEN

BACKGROUND: New molecular targets are needed for women with triple-negative breast cancer (TNBC). This pre-clinical study investigated the combination of the EGFR inhibitor gefitinib with the sphingosine kinase (SphK) inhibitor FTY720 (Fingolimod), aiming to block tumorigenic signaling downstream of IGFBP-3, which is abundantly expressed in basal-like TNBC. METHODS: In studies of breast cancer cell growth in culture, proliferation was monitored by IncuCyte live-cell imaging, and protein abundance was determined by western blotting. In vivo studies of mammary tumor growth used two models: orthotopic xenograft tumors derived from three basal-like TNBC cell lines, grown in immune-deficient mice, and syngeneic murine 4T1 tumors grown in immune-competent mice. Protein abundance in tumor tissue was assessed by immunohistochemistry. RESULTS: Quantitated by live-cell imaging, the inhibitor combination showed synergistic cytostatic activity in basal-like cell lines across several TNBC molecular subtypes, the synergy being decreased by IGFBP-3 downregulation. Suppression of the tumorigenic mediator CD44 by gefitinib was potentiated by FTY720, consistent with CD44 involvement in the targeted pathway. In MDA-MB-468 and HCC1806 orthotopic TNBC xenograft tumors in nude mice, the drug combination inhibited tumor growth and prolonged mouse survival, although this effect was not significant for the gefitinib-resistant cell line HCC70. Combination treatment of murine 4T1 TNBC tumors in syngeneic BALB/c mice was more effective in immune-competent than immune-deficient (nude) mice, and a relative loss of tumor CD3 (T-cell) immunoreactivity caused by FTY720 treatment alone was alleviated by the drug combination, suggesting that, even at an FTY720 dose causing relative lymphopenia, the combination is still effective in an immune-competent setting. Immunohistochemistry of xenograft tumors showed significant enhancement of caspase-3 cleavage and suppression of Ki67 and phospho-EGFR by the drug combination, but SphK1 downregulation occurred only in MDA-MB-468 tumors, so is unlikely to be integral to treatment efficacy. CONCLUSIONS: Our data indicate that targeting IGFBP-3-dependent signaling pathways through gefitinib-FTY720 co-therapy may be effective in many basal-like breast cancers, and suggest tissue IGFBP-3 and CD44 measurement as potential biomarkers of treatment efficacy.


Asunto(s)
Receptores ErbB/genética , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Animales , Caspasa 3 , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Clorhidrato de Fingolimod/administración & dosificación , Gefitinib , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Receptores de Hialuranos/genética , Ratones , Inhibidores de Proteínas Quinasas , Quinazolinas/administración & dosificación , Transducción de Señal/efectos de los fármacos , Neoplasias de la Mama Triple Negativas/genética , Ensayos Antitumor por Modelo de Xenoinjerto
5.
BMC Cancer ; 17(1): 820, 2017 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-29207959

RESUMEN

BACKGROUND: Targeting the type 1 insulin-like growth factor receptor (IGF1R) in breast cancer remains an ongoing clinical challenge. Oncogenic IGF1R-signaling occurs via activation of PI3K/AKT/MAPK downstream mediators which regulate cell proliferation and protein synthesis. To further understand IGF1R signaling we have investigated the involvement of the oncogenic IGF1R-related sphingosine kinase (SphK) pathway. METHODS: The prognostic (overall survival, OS) and therapeutic (anti-endocrine therapy) co-contribution of IGF1R and SphK1 were investigated using breast cancer patient samples (n = 236) for immunohistochemistry to measure total and phosphorylated IGF1R and SphK1. Kaplan-Meier and correlation analyses were performed to determine the contribution of high versus low IGF1R and/or SphK1 expression to OS in patients treated with anti-endocrine therapy. Cell viability and colony formation in vitro studies were completed using estrogen receptor (ER) positive and negative breast cancer cell-lines to determine the benefit of IGF1R inhibitor (OSI-906) and SphK inhibitor (SKI-II) co-therapy. Repeated measures and 1-way ANOVA were performed to compare drug treatments groups and the Chou-Talalay combination index (CI) was calculated to estimate drug synergism in vitro (CI < 1). RESULTS: High IGF1R and SphK1 protein co-expression in tumor tissue was associated with improved OS specifically in ER-positive disease and stratified for anti-endocrine therapy. A significant synergistic inhibition of cell viability and/or colony formation following OSI-906 and SKI-II co-treatment in vitro was evident (p < 0.05, CI < 1). CONCLUSION: We conclude that high IGF1R and SphK1 co-expression act together as prognostic indicators and are potentially, dual therapeutic targets for the development of a more effective IGF1R-directed combination breast cancer therapy.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama Masculina/metabolismo , Neoplasias de la Mama/metabolismo , Carcinoma Ductal de Mama/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Receptores de Somatomedina/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama Masculina/diagnóstico , Neoplasias de la Mama Masculina/tratamiento farmacológico , Neoplasias de la Mama Masculina/mortalidad , Carcinoma Ductal de Mama/diagnóstico , Carcinoma Ductal de Mama/tratamiento farmacológico , Carcinoma Ductal de Mama/mortalidad , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Femenino , Humanos , Estimación de Kaplan-Meier , Células MCF-7 , Masculino , Persona de Mediana Edad , Análisis Multivariante , Pronóstico , Modelos de Riesgos Proporcionales , Receptor IGF Tipo 1 , Resultado del Tratamiento , Adulto Joven
6.
Breast Cancer Res Treat ; 155(2): 203-13, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26740212

RESUMEN

FXYD3, also known as mammary tumor protein 8, is overexpressed in several common cancers, including in many breast cancers. We examined if such overexpression might protect Na(+)/K(+)-ATPase and cancer cells against the high levels of oxidative stress characteristic of many tumors and often induced by cancer treatments. We measured FXYD3 expression, Na(+)/K(+)-ATPase activity and glutathionylation of the ß1 subunit of Na(+)/K(+)-ATPase, a reversible oxidative modification that inhibits the ATPase, in MCF-7 and MDA-MB-468 cells. Expression of FXYD3 was suppressed by transfection with FXYD3 siRNA. A colorimetric end-point assay was used to estimate cell viability. Apoptosis was estimated by caspase 3/7 (DEVDase) activation using a Caspase fluorogenic substrate kit. Expression of FXYD3 in MCF-7 breast cancer cells was ~eightfold and ~twofold higher than in non-cancer MCF-10A cells and MDA-MB-468 cancer cells, respectively. A ~50 % reduction in FXYD3 expression increased glutathionylation of the ß1 Na(+)/K(+)-ATPase subunit and reduced Na(+)/K(+)-ATPase activity by ~50 %, consistent with the role of FXYD3 to facilitate reversal of glutathionylation of the ß1 subunit of Na(+)/K(+)-ATPase and glutathionylation-induced inhibition of Na(+)/K(+)-ATPase. Treatment of MCF-7 and MDA-MB- 468 cells with doxorubicin or γ-radiation decreased cell viability and induced apoptosis. The treatments upregulated FXYD3 expression in MCF-7 but not in MDA-MB-468 cells and suppression of FXYD3 in MCF-7 but not in MDA-MB-468 cells amplified effects of treatments on Na(+)/K(+)-ATPase activity and treatment-induced cell death and apoptosis. Overexpression of FXYD3 may be a marker of resistance to cancer treatments and a potentially important therapeutic target.


Asunto(s)
Neoplasias de la Mama/genética , Supervivencia Celular/genética , Doxorrubicina/farmacología , Rayos gamma/uso terapéutico , Silenciador del Gen/efectos de los fármacos , Proteínas de la Membrana/genética , Proteínas de Neoplasias/genética , ATPasa Intercambiadora de Sodio-Potasio/genética , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/radioterapia , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Femenino , Expresión Génica/efectos de los fármacos , Expresión Génica/efectos de la radiación , Humanos , Células MCF-7
7.
Glob Chang Biol ; 21(10): 3712-25, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25969925

RESUMEN

Quantifying landscape-scale methane (CH4 ) fluxes from boreal and arctic regions, and determining how they are controlled, is critical for predicting the magnitude of any CH4 emission feedback to climate change. Furthermore, there remains uncertainty regarding the relative importance of small areas of strong methanogenic activity, vs. larger areas with net CH4 uptake, in controlling landscape-level fluxes. We measured CH4 fluxes from multiple microtopographical subunits (sedge-dominated lawns, interhummocks and hummocks) within an aapa mire in subarctic Finland, as well as in drier ecosystems present in the wider landscape, lichen heath and mountain birch forest. An intercomparison was carried out between fluxes measured using static chambers, up-scaled using a high-resolution landcover map derived from aerial photography and eddy covariance. Strong agreement was observed between the two methodologies, with emission rates greatest in lawns. CH4 fluxes from lawns were strongly related to seasonal fluctuations in temperature, but their floating nature meant that water-table depth was not a key factor in controlling CH4 release. In contrast, chamber measurements identified net CH4 uptake in birch forest soils. An intercomparison between the aerial photography and satellite remote sensing demonstrated that quantifying the distribution of the key CH4 emitting and consuming plant communities was possible from satellite, allowing fluxes to be scaled up to a 100 km(2) area. For the full growing season (May to October), ~ 1.1-1.4 g CH4  m(-2) was released across the 100 km(2) area. This was based on up-scaled lawn emissions of 1.2-1.5 g CH4  m(-2) , vs. an up-scaled uptake of 0.07-0.15 g CH4  m(-2) by the wider landscape. Given the strong temperature sensitivity of the dominant lawn fluxes, and the fact that lawns are unlikely to dry out, climate warming may substantially increase CH4 emissions in northern Finland, and in aapa mire regions in general.


Asunto(s)
Bosques , Metano/metabolismo , Humedales , Regiones Árticas , Cambio Climático , Finlandia
8.
J Biol Chem ; 288(1): 59-68, 2013 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-23166326

RESUMEN

Insulin-like growth factor II (IGF-II) is a major embryonic growth factor belonging to the insulin-like growth factor family, which includes insulin and IGF-I. Its expression in humans is tightly controlled by maternal imprinting, a genetic restraint that is lost in many cancers, resulting in up-regulation of both mature IGF-II mRNA and protein expression. Additionally, increased expression of several longer isoforms of IGF-II, termed "pro" and "big" IGF-II, has been observed. To date, it is ambiguous as to what role these IGF-II isoforms have in initiating and sustaining tumorigenesis and whether they are bioavailable. We have expressed each individual IGF-II isoform in their proper O-glycosylated format and established that all bind to the IGF-I receptor and both insulin receptors A and B, resulting in their activation and subsequent stimulation of fibroblast proliferation. We also confirmed that all isoforms are able to be sequestered into binary complexes with several IGF-binding proteins (IGFBP-2, IGFBP-3, and IGFBP-5). In contrast to this, ternary complex formation with IGFBP-3 or IGFBP-5 and the auxillary protein, acid labile subunit, was severely diminished. Furthermore, big-IGF-II isoforms bound much more weakly to purified ectodomain of the natural IGF-II scavenging receptor, IGF-IIR. IGF-II isoforms thus possess unique biological properties that may enable them to escape normal sequestration avenues and remain bioavailable in vivo to sustain oncogenic signaling.


Asunto(s)
Factor II del Crecimiento Similar a la Insulina/química , Neoplasias/metabolismo , Animales , Proliferación Celular , Fibroblastos/citología , Regulación Neoplásica de la Expresión Génica , Glicosilación , Células HEK293 , Humanos , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/química , Factor I del Crecimiento Similar a la Insulina/química , Espectrometría de Masas/métodos , Ratones , Unión Proteica , Isoformas de Proteínas , Estructura Terciaria de Proteína , Proteínas Tirosina Quinasas Receptoras/química , Transducción de Señal
9.
Breast Cancer Res ; 16(3): R63, 2014 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-24935269

RESUMEN

INTRODUCTION: Serum profiling using proteomic techniques has great potential to detect biomarkers that might improve diagnosis and predict outcome for breast cancer patients (BC). This study used surface-enhanced laser desorption/ionization time-of-flight (SELDI-TOF) mass spectrometry (MS) to identify differentially expressed proteins in sera from BC and healthy volunteers (HV), with the goal of developing a new prognostic biomarker panel. METHODS: Training set serum samples from 99 BC and 51 HV subjects were applied to four adsorptive chip surfaces (anion-exchange, cation-exchange, hydrophobic, and metal affinity) and analyzed by time-of-flight MS. For validation, 100 independent BC serum samples and 70 HV samples were analyzed similarly. Cluster analysis of protein spectra was performed to identify protein patterns related to BC and HV groups. Univariate and multivariate statistical analyses were used to develop a protein panel to distinguish breast cancer sera from healthy sera, and its prognostic potential was evaluated. RESULTS: From 51 protein peaks that were significantly up- or downregulated in BC patients by univariate analysis, binary logistic regression yielded five protein peaks that together classified BC and HV with a receiver operating characteristic (ROC) area-under-the-curve value of 0.961. Validation on an independent patient cohort confirmed the five-protein parameter (ROC value 0.939). The five-protein parameter showed positive association with large tumor size (P = 0.018) and lymph node involvement (P = 0.016). By matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) MS, immunoprecipitation and western blotting the proteins were identified as a fragment of apolipoprotein H (ApoH), ApoCI, complement C3a, transthyretin, and ApoAI. Kaplan-Meier analysis on 181 subjects after median follow-up of >5 years demonstrated that the panel significantly predicted disease-free survival (P = 0.005), its efficacy apparently greater in women with estrogen receptor (ER)-negative tumors (n = 50, P = 0.003) compared to ER-positive (n = 131, P = 0.161), although the influence of ER status needs to be confirmed after longer follow-up. CONCLUSIONS: Protein mass profiling by MS has revealed five serum proteins which, in combination, can distinguish between serum from women with breast cancer and healthy control subjects with high sensitivity and specificity. The five-protein panel significantly predicts recurrence-free survival in women with ER-negative tumors and may have value in the management of these patients.


Asunto(s)
Biomarcadores de Tumor/sangre , Proteínas Sanguíneas/análisis , Neoplasias de la Mama/sangre , Apolipoproteína A-I/sangre , Apolipoproteína C-I/sangre , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/mortalidad , Complemento C3a/análisis , Supervivencia sin Enfermedad , Femenino , Humanos , Persona de Mediana Edad , Prealbúmina/análisis , Receptores de Estrógenos/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , beta 2 Glicoproteína I/sangre
10.
Biofouling ; 30(1): 29-39, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24138163

RESUMEN

Dental handpieces (DHPs) become biofouled internally with patient derived material that is difficult to access for removal and inactivation. This study undertook a quantitative and qualitative investigation of protein contamination of internal components from three different types of DHP: the turbine, slow speed contra-angle and surgical. Eluates from the high speed turbine, low speed spray channels and surgical gear were assayed for protein using an orthophthaldehyde assay. Eluates concentrated by Amicon ultrafiltration were also analysed by SDS-PAGE, mass spectroscopy, Western blotting and ELISA. The surfaces of handpiece components were also investigated by SEM, EFSCAN and EDAX microscopy. Surgical gears contained highest levels of protein (403 µg), followed by low speed spray channels (17.7 µg) and the high speed turbine (<5 µg). Mass spectroscopy of surgical gears demonstrated mostly serum derived proteins. Decontamination of the DHPs using an automated washer disinfector and handpiece irrigator showed a significant reduction in residual protein levels.


Asunto(s)
Incrustaciones Biológicas , Descontaminación , Instrumentos Dentales , Contaminación de Equipos , Transmisión de Enfermedad Infecciosa , Desinfección , Ensayo de Inmunoadsorción Enzimática , Humanos , Enfermedades por Prión/prevención & control , Enfermedades por Prión/transmisión , Proteómica , Esterilización
11.
Nat Rev Endocrinol ; 20(7): 414-425, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38514815

RESUMEN

The acid-labile subunit (ALS) of the insulin-like growth factor (IGF) binding protein (IGFBP) complex, encoded in humans by IGFALS, has a vital role in regulating the endocrine transport and bioavailability of IGF-1 and IGF-2. Accordingly, ALS has a considerable influence on postnatal growth and metabolism. ALS is a leucine-rich glycoprotein that forms high-affinity ternary complexes with IGFBP-3 or IGFBP-5 when they are occupied by either IGF-1 or IGF-2. These complexes constitute a stable reservoir of circulating IGFs, blocking the potentially hypoglycaemic activity of unbound IGFs. ALS is primarily synthesized by hepatocytes and its expression is lower in non-hepatic tissues. ALS synthesis is strongly induced by growth hormone and suppressed by IL-1ß, thus potentially serving as a marker of growth hormone secretion and/or activity and of inflammation. IGFALS mutations in humans and Igfals deletion in mice cause modest growth retardation and pubertal delay, accompanied by decreased osteogenesis and enhanced adipogenesis. In hepatocellular carcinoma, IGFALS is described as a tumour suppressor; however, its contribution to other cancers is not well delineated. This Review addresses the endocrine physiology and pathology of ALS, discusses the latest cell and proteomic studies that suggest emerging cellular roles for ALS and outlines its involvement in other disease states.


Asunto(s)
Glicoproteínas , Humanos , Animales , Glicoproteínas/metabolismo , Proteínas Portadoras/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Ratones , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/fisiología , Factor II del Crecimiento Similar a la Insulina/metabolismo , Sistema Endocrino/metabolismo , Péptidos Similares a la Insulina
12.
Cureus ; 16(2): e54907, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38544579

RESUMEN

The ascent of medical technology places augmented reality (AR) at the forefront of potential advancements in interventional radiology (IR) training. This review delves into the symbiotic relationship between AR and conventional IR training, casting light on the opportunities and hurdles intrinsic to this integration. A targeted literature review was conducted using the databases PubMed, Cochrane Library, and Embase. Search terms included ((("Augmented Reality" OR "Virtual Reality")) AND ((Education OR Training))) AND (("Interventional Radiology")). Ten studies identified using the comprehensive inclusion criteria helped scrutinize the use of AR in IR training. Key outcomes include improved procedural accuracy, reduced training duration, and heightened trainee confidence. However, it also identifies limitations such as small sample sizes, reliance on simulation environments, and technological constraints in AR implementation. Despite these challenges, the review underscored the transformative potential of AR in IR education, suggesting its capacity to revolutionize training methodologies. However, it also calls for continued technological development and empirical research to address current challenges and fully leverage AR's capabilities in medical education.

13.
Endocr Rev ; 44(5): 753-778, 2023 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-36974712

RESUMEN

The 6 high-affinity insulin-like growth factor binding proteins (IGFBPs) are multifunctional proteins that modulate cell signaling through multiple pathways. Their canonical function at the cellular level is to impede access of insulin-like growth factor (IGF)-1 and IGF-2 to their principal receptor IGF1R, but IGFBPs can also inhibit, or sometimes enhance, IGF1R signaling either through their own post-translational modifications, such as phosphorylation or limited proteolysis, or by their interactions with other regulatory proteins. Beyond the regulation of IGF1R activity, IGFBPs have been shown to modulate cell survival, migration, metabolism, and other functions through mechanisms that do not appear to involve the IGF-IGF1R system. This is achieved by interacting directly or functionally with integrins, transforming growth factor ß family receptors, and other cell-surface proteins as well as intracellular ligands that are intermediates in a wide range of pathways. Within the nucleus, IGFBPs can regulate the diverse range of functions of class II nuclear hormone receptors and have roles in both cell senescence and DNA damage repair by the nonhomologous end-joining pathway, thus potentially modifying the efficacy of certain cancer therapeutics. They also modulate some immune functions and may have a role in autoimmune conditions such as rheumatoid arthritis. IGFBPs have been proposed as attractive therapeutic targets, but their ubiquity in the circulation and at the cellular level raises many challenges. By understanding the diversity of regulatory pathways with which IGFBPs interact, there may still be therapeutic opportunities based on modulation of IGFBP-dependent signaling.


Asunto(s)
Proteínas de Unión a Factor de Crecimiento Similar a la Insulina , Factor I del Crecimiento Similar a la Insulina , Humanos , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/genética , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Factor II del Crecimiento Similar a la Insulina/metabolismo , Transducción de Señal/fisiología
14.
Neural Netw ; 164: 275-309, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37163846

RESUMEN

Neocortical pyramidal neurons have many dendrites, and such dendrites are capable of, in isolation of one-another, generating a neuronal spike. It is also now understood that there is a large amount of dendritic growth during the first years of a humans life, arguably a period of prodigious learning. These observations inspire the construction of a local, stochastic algorithm based on an earlier stochastic, homeostatic, Hebbian developmental theory. Here we investigate the neurocomputational advantages and limits on this novel algorithm that combines dendritogenesis with supervised adaptive synaptogenesis. Neurons created with this algorithm have enhanced memory capacity, can avoid catastrophic interference (forgetting), and have the ability to unmix mixture distributions. In particular, individual dendrites develop within each class, in an unsupervised manner, to become feature-clusters that correspond to the mixing elements of class-conditional mixture distribution. Error-free classification is demonstrated with input perturbations up to 40%. Although discriminative problems are used to understand the capabilities of the stochastic algorithm and the neuronal connectivity it produces, the algorithm is in the generative class, it thus seems ideal for decisions that require generalization, i.e., extrapolation beyond previous learning.


Asunto(s)
Dendritas , Sinapsis , Humanos , Dendritas/fisiología , Sinapsis/fisiología , Neuronas/fisiología , Células Piramidales/fisiología , Aprendizaje , Modelos Neurológicos
15.
Lancet Digit Health ; 5(7): e446-e457, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37391265

RESUMEN

BACKGROUND: It is unclear what effect the pattern of health-care use before admission to hospital with COVID-19 (index admission) has on the long-term outcomes for patients. We sought to describe mortality and emergency readmission to hospital after discharge following the index admission (index discharge), and to assess associations between these outcomes and patterns of health-care use before such admissions. METHODS: We did a national, retrospective, complete cohort study by extracting data from several national databases and linking the databases for all adult patients admitted to hospital in Scotland with COVID-19. We used latent class trajectory modelling to identify distinct clusters of patients on the basis of their emergency admissions to hospital in the 2 years before the index admission. The primary outcomes were mortality and emergency readmission up to 1 year after index admission. We used multivariable regression models to explore associations between these outcomes and patient demographics, vaccination status, level of care received in hospital, and previous emergency hospital use. FINDINGS: Between March 1, 2020, and Oct 25, 2021, 33 580 patients were admitted to hospital with COVID-19 in Scotland. Overall, the Kaplan-Meier estimate of mortality within 1 year of index admission was 29·6% (95% CI 29·1-30·2). The cumulative incidence of emergency hospital readmission within 30 days of index discharge was 14·4% (95% CI 14·0-14·8), with the number increasing to 35·6% (34·9-36·3) patients at 1 year. Among the 33 580 patients, we identified four distinct patterns of previous emergency hospital use: no admissions (n=18 772 [55·9%]); minimal admissions (n=12 057 [35·9%]); recently high admissions (n=1931 [5·8%]), and persistently high admissions (n=820 [2·4%]). Patients with recently or persistently high admissions were older, more multimorbid, and more likely to have hospital-acquired COVID-19 than patients with no or minimal admissions. People in the minimal, recently high, and persistently high admissions groups had an increased risk of mortality and hospital readmission compared with those in the no admissions group. Compared with the no admissions group, mortality was highest in the recently high admissions group (post-hospital mortality HR 2·70 [95% CI 2·35-2·81]; p<0·0001) and the risk of readmission was highest in the persistently high admissions group (3·23 [2·89-3·61]; p<0·0001). INTERPRETATION: Long-term mortality and readmission rates for patients hospitalised with COVID-19 were high; within 1 year, one in three patients had died and a third had been readmitted as an emergency. Patterns of hospital use before index admission were strongly predictive of mortality and readmission risk, independent of age, pre-existing comorbidities, and COVID-19 vaccination status. This increasingly precise identification of individuals at high risk of poor outcomes from COVID-19 will enable targeted support. FUNDING: Chief Scientist Office Scotland, UK National Institute for Health Research, and UK Research and Innovation.


Asunto(s)
Vacunas contra la COVID-19 , COVID-19 , Adulto , Humanos , Estudios de Cohortes , Estudios Retrospectivos , COVID-19/epidemiología , COVID-19/terapia , Hospitales
16.
J Proteome Res ; 11(2): 1240-50, 2012 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-22133146

RESUMEN

Treatment of breast cancer is complex and challenging due to the heterogeneity of the disease. To avoid significant toxicity and adverse side-effects of chemotherapy in patients who respond poorly, biomarkers predicting therapeutic response are essential. This study has utilized a proteomic approach integrating 2D-DIGE, LC-MS/MS, and bioinformatics to analyze the proteome of breast cancer (ZR-75-1 and MDA-MB-231) and breast epithelial (MCF-10A) cell lines induced to undergo apoptosis using a combination of doxorubicin and TRAIL administered in sequence (Dox-TRAIL). Apoptosis induction was confirmed using a caspase-3 activity assay. Comparative proteomic analysis between whole cell lysates of Dox-TRAIL and control samples revealed 56 differentially expressed spots (≥2-fold change and p < 0.05) common to at least two cell lines. Of these, 19 proteins were identified yielding 11 unique protein identities: CFL1, EIF5A, HNRNPK, KRT8, KRT18, LMNA, MYH9, NACA, RPLP0, RPLP2, and RAD23B. A subset of the identified proteins was validated by selected reaction monitoring (SRM) and Western blotting. Pathway analysis revealed that the differentially abundant proteins were associated with cell death, cellular organization, integrin-linked kinase signaling, and actin cytoskeleton signaling pathways. The 2D-DIGE analysis has yielded candidate biomarkers of response to treatment in breast cancer cell models. Their clinical utility will depend on validation using patient breast biopsies pre- and post-treatment with anticancer drugs.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Doxorrubicina/farmacología , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Biomarcadores de Tumor/análisis , Western Blotting , Neoplasias de la Mama/patología , Caspasa 3/metabolismo , Línea Celular Tumoral , Regulación hacia Abajo , Doxorrubicina/administración & dosificación , Electroforesis en Gel Bidimensional , Femenino , Humanos , Proteínas de Neoplasias/análisis , Proteínas de Neoplasias/metabolismo , Mapas de Interacción de Proteínas , Proteoma/efectos de los fármacos , Proteómica/métodos , Reproducibilidad de los Resultados , Ligando Inductor de Apoptosis Relacionado con TNF/administración & dosificación , Regulación hacia Arriba
17.
J Proteome Res ; 11(7): 3561-72, 2012 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-22587632

RESUMEN

Breast cancer is a molecularly heterogeneous disease, and predicting response to chemotherapy remains a major clinical challenge. To minimize adverse side-effects or cumulative toxicity in patients unlikely to benefit from treatment, biomarkers indicating treatment efficacy are critically needed. iTRAQ labeling coupled with multidimensional LC-MS/MS of the enriched mitochondria and endoplasmic reticulum fraction, key organelles regulating apoptosis, has led to the discovery of several differentially abundant proteins in breast cancer cells treated with the chemotherapeutic agent doxorubicin followed by the death receptor ligand, TRAIL, among 571 and 801 unique proteins identified in ZR-75-1 and MDA-MB-231 breast cancer cell lines, respectively. The differentially abundant proteins represent diverse biological processes associated with cellular assembly and organization, molecular transport, oxidative stress, cell motility, cell death, and cancer. Despite many differences in molecular phenotype between the two breast cancer cell lines, a comparison of their subproteomes following drug treatment revealed three proteins displaying common regulation: PPIB, AHNAK, and SLC1A5. Changes in these proteins, detected by iTRAQ, were confirmed by immunofluorescence, visualized by confocal microscopy. These novel potential biomarkers may have clinical utility for assessing response to cancer treatment and may provide insight into new therapeutic targets for breast cancer.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Doxorrubicina/farmacología , Proteoma/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Sistema de Transporte de Aminoácidos ASC/metabolismo , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral/efectos de los fármacos , Movimiento Celular , Quimioterapia Adyuvante , Ciclofilinas/metabolismo , Retículo Endoplásmico/metabolismo , Femenino , Humanos , Proteínas de la Membrana/metabolismo , Redes y Vías Metabólicas , Antígenos de Histocompatibilidad Menor , Mitocondrias/metabolismo , Terapia Neoadyuvante , Proteínas de Neoplasias/metabolismo , Estrés Oxidativo , Proteómica , Espectrometría de Masas en Tándem
18.
J Biol Chem ; 286(34): 29540-7, 2011 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-21737444

RESUMEN

Insulin-like growth factor-binding protein-3 (IGFBP-3) expression is frequently suppressed in liver cancers and can be reactivated by histone deacetylase (HDAC) inhibition. This study examined the role of IGFBP-3 in mediating the effects of the HDAC inhibitor MS-275 in liver cancer cells and identified IGFBP-3-dependent proteins that regulate proliferation and migration. In HepG2 cells, MS-275 inhibited DNA synthesis, cell cycle activity, and cell viability concomitantly with increased binding of acetylated histone H3 to IGFBP-3 promoter sequences and induction of IGFBP-3 expression. IGFBP-3 down-regulation by siRNA significantly reversed the inhibition of cell viability and DNA synthesis by MS-275, indicating an intermediary role for IGFBP-3. Induction of the cyclin-dependent kinase inhibitor p21 by MS-275 was attenuated by IGFBP-3 down-regulation, providing an explanation for IGFBP-3-dependent effects of MS-275 on cell cycle activity. In contrast, MS-275 stimulated HepG2 cell migration, an effect also inhibited by IGFBP-3 down-regulation. Among genes whose induction by MS-275 was attenuated by IGFBP-3 down-regulation, LYVE1 and THBS2 (thrombospondin-2) were identified as mediators of IGFBP-3-dependent effects of MS-275. Silencing of either protein had no effect on the inhibition of HepG2 viability by MS-275 but reversed its stimulatory effect on cell migration. We conclude that among genes up-regulated by MS-275, IGFBP-3 is a key mediator of effects on hepatoma cell growth and migration, involving IGFBP-3-dependent proteins p21 (proliferation) and LYVE1 and THBS2 (migration). The enhanced cell motility that accompanies reactivation of IGFBP-3 expression in liver cancer by HDAC inhibition suggests the possibility of increased metastatic spread despite inhibited cell proliferation.


Asunto(s)
Benzamidas/farmacología , Carcinoma Hepatocelular/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/biosíntesis , Neoplasias Hepáticas/metabolismo , Proteínas de Neoplasias/biosíntesis , Piridinas/farmacología , Acetilación/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Carcinoma Hepatocelular/genética , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Regulación Neoplásica de la Expresión Génica/genética , Silenciador del Gen/efectos de los fármacos , Células Hep G2 , Histonas/genética , Histonas/metabolismo , Humanos , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/genética , Neoplasias Hepáticas/genética , Proteínas de Neoplasias/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Trombospondinas/genética , Trombospondinas/metabolismo , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
19.
Expert Rev Proteomics ; 9(6): 599-614, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23256671

RESUMEN

Although the molecular classification and prognostic assessment of breast tumors based on gene expression profiling is well established, a number of proteomic studies that propose potential breast cancer biomarkers has not yet led to any new diagnostic, prognostic or predictive test in wide clinical use. This review examines the current status of breast cancer biomarkers, discusses sample types (including plasma, tumor tissue, nipple aspirate and ductal lavage, as well as cell culture models) and different electrophoretic and mass spectrometry methods that have been widely used for the discovery of proteomic biomarkers in breast cancer, and also considers several approaches to biomarker validation. The pathway leading from the initial proteomic discovery and validation process to translation into a clinically useful test is also discussed.


Asunto(s)
Neoplasias de la Mama/diagnóstico , Proteínas de Neoplasias/metabolismo , Proteómica , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Electroforesis en Gel Bidimensional , Femenino , Humanos , Espectrometría de Masas/métodos
20.
Pancreatology ; 12(2): 124-9, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22487522

RESUMEN

MOTIVATION: Reports of serum pancreatic cancer (PC) biomarkers using SELDI-TOF MS have been inconsistent because different chip surfaces and interference with high-abundant proteins. This study examines the influence of these factors on the detection of discriminating diagnostic biomarkers. METHODS: Serum from fourteen from patients with PC, disease controls (DC, n = 14) and healthy volunteers (HV, n = 14) were evaluated by SELDI using H50, IMAC, Q10 and CM10 chips. A further evaluation was undertaken after depletion of seven high-abundant proteins using spin cartridges. RESULTS: More protein peaks were detected in whole serum than in depleted serum for IMAC, H50 and Q10 chips: 60 vs 39, 56 vs 48 and 69 vs 65, respectively, while the CM10 found less peaks in serum (27 vs 47 peaks). However, there were more differentially expressed peaks in the depleted serum samples for PC vs DC and PC vs HV samples using the H50, Q10 and CM10 ProteinChip arrays, whereas for IMAC arrays, more discriminating peaks were seen in non-depleted serum. The highly significant peaks observed on Q10, CM10 and H50 are consistent with the previous finding of ApoA-I (m/z 27,910-28000) and ApoA-II (m/z 8758 and 17,240). In addition, a number of new discriminating protein peaks were found on different ProteinChip arrays, notably peaks at m/z 4280 and 7763 on IMAC arrays. CONCLUSION: This study confirms the diagnostic value of ApoA-I&II and identifies further potential diagnostic biomarkers for pancreatic cancer when multiple chip surfaces are used with depletion of the most highly-abundant proteins.


Asunto(s)
Biomarcadores de Tumor/sangre , Carcinoma Ductal Pancreático/diagnóstico , Neoplasias Pancreáticas/diagnóstico , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma Ductal Pancreático/sangre , Cromatografía de Afinidad/métodos , Cistoadenoma Mucinoso/sangre , Cistoadenoma Mucinoso/diagnóstico , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neoplasias Pancreáticas/sangre , Pancreatitis Crónica/sangre , Pancreatitis Crónica/diagnóstico , Análisis por Matrices de Proteínas , Proteómica , Reproducibilidad de los Resultados , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA