Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 49(5): 959-71, 2013 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-23375500

RESUMEN

The proapoptotic Bcl-2 protein Bax is predominantly found in the cytosol of nonapoptotic cells and is commonly thought to translocate to mitochondria following an apoptotic stimulus. The current model for Bax activation is that BH3 proteins bind to cytosolic Bax, initiating mitochondrial targeting and outer-membrane permeabilization. Here, we challenge this and show that Bax is constitutively targeted to mitochondria but in nonapoptotic cells is constantly translocated back to the cytosol. Using live-cell spinning-disk confocal imaging with a combination of FLIP, FRAP, and photoactivatable GFP-Bax, we demonstrate that disrupting adhesion-dependent survival signals slows the rate of Bax's dissociation from mitochondria, leading to its accumulation on the outer mitochondrial membrane. The overall accumulation of mitochondrial Bax following loss of survival signaling sensitizes cells to proapoptotic BH3 proteins. Our findings show that Bax is normally in a dynamic equilibrium between cytosol and mitochondria, enabling fluctuations in survival signals to finely adjust apoptotic sensitivity.


Asunto(s)
Apoptosis , Citosol/metabolismo , Mitocondrias/metabolismo , Proteína X Asociada a bcl-2/genética , Animales , Células Cultivadas , Células HEK293 , Humanos , Ratones , Membranas Mitocondriales/metabolismo , Transfección , Proteína X Asociada a bcl-2/metabolismo
2.
Int J Mol Sci ; 21(12)2020 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-32630604

RESUMEN

Vascular calcification describes the formation of mineralized tissue within the blood vessel wall, and it is highly associated with increased cardiovascular morbidity and mortality in patients with chronic kidney disease, diabetes, and atherosclerosis. In this article, we briefly review different rodent models used to study vascular calcification in vivo, and critically assess the strengths and weaknesses of the current techniques used to analyze and quantify calcification in these models, namely 2-D histology and the o-cresolphthalein assay. In light of this, we examine X-ray micro-computed tomography (µCT) as an emerging complementary tool for the analysis of vascular calcification in animal models. We demonstrate that this non-destructive technique allows us to simultaneously quantify and localize calcification in an intact vessel in 3-D, and we consider recent advances in µCT sample preparation techniques. This review also discusses the potential to combine 3-D µCT analyses with subsequent 2-D histological, immunohistochemical, and proteomic approaches in correlative microscopy workflows to obtain rich, multifaceted information on calcification volume, calcification load, and signaling mechanisms from within the same arterial segment. In conclusion we briefly discuss the potential use of µCT to visualize and measure vascular calcification in vivo in real-time.


Asunto(s)
Calcificación Vascular/patología , Microtomografía por Rayos X/métodos , Microtomografía por Rayos X/tendencias , Animales , Aterosclerosis/patología , Humanos , Imagenología Tridimensional/métodos , Microscopía/métodos , Modelos Animales , Proteómica , Insuficiencia Renal Crónica/patología , Calcificación Vascular/diagnóstico por imagen , Calcificación Vascular/metabolismo
3.
Breast Cancer Res ; 20(1): 128, 2018 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-30348189

RESUMEN

BACKGROUND: Stem cells are precursors for all mammary epithelia, including ductal and alveolar epithelia, and myoepithelial cells. In vivo mammary epithelia reside in a tissue context and interact with their milieu via receptors such as integrins. Extracellular matrix receptors coordinate important cellular signalling platforms, of which integrins are the central architects. We have previously shown that integrins are required for mammary epithelial development and function, including survival, cell cycle, and polarity, as well as for the expression of mammary-specific genes. In the present study we looked at the role of integrins in mammary epithelial stem cell self-renewal. METHODS: We used an in vitro stem cell assay with primary mouse mammary epithelial cells isolated from genetically altered mice. This involved a 3D organoid assay, providing an opportunity to distinguish the stem cell- or luminal progenitor-driven organoids as structures with solid or hollow appearances, respectively. RESULTS: We demonstrate that integrins are essential for the maintenance and self-renewal of mammary epithelial stem cells. Moreover integrins activate the Rac1 signalling pathway in stem cells, which leads to the stimulation of a Wnt pathway, resulting in expression of ß-catenin target genes such as Axin2 and Lef1. CONCLUSIONS: Integrin/Rac signalling has a role in specifying the activation of a canonical Wnt pathway that is required for mammary epithelial stem cell self-renewal.


Asunto(s)
Autorrenovación de las Células/fisiología , Células Epiteliales/fisiología , Integrinas/metabolismo , Neuropéptidos/metabolismo , Células Madre/fisiología , Proteína de Unión al GTP rac1/metabolismo , Animales , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Epitelio/fisiología , Femenino , Integrinas/genética , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/fisiología , Ratones , Ratones Noqueados , Neuropéptidos/genética , Organoides/fisiología , Cultivo Primario de Células/métodos , Transducción de Señal/fisiología , Proteína de Unión al GTP rac1/genética
4.
PLoS Comput Biol ; 13(2): e1005400, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28245235

RESUMEN

The Notch pathway plays a vital role in determining whether cells in the intestinal epithelium adopt a secretory or an absorptive phenotype. Cell fate specification is coordinated via Notch's interaction with the canonical Wnt pathway. Here, we propose a new mathematical model of the Notch and Wnt pathways, in which the Hes1 promoter acts as a hub for pathway crosstalk. Computational simulations of the model can assist in understanding how healthy intestinal tissue is maintained, and predict the likely consequences of biochemical knockouts upon cell fate selection processes. Chemical reaction network theory (CRNT) is a powerful, generalised framework which assesses the capacity of our model for monostability or multistability, by analysing properties of the underlying network structure without recourse to specific parameter values or functional forms for reaction rates. CRNT highlights the role of ß-catenin in stabilising the Notch pathway and damping oscillations, demonstrating that Wnt-mediated actions on the Hes1 promoter can induce dynamic transitions in the Notch system, from multistability to monostability. Time-dependent model simulations of cell pairs reveal the stabilising influence of Wnt upon the Notch pathway, in which ß-catenin- and Dsh-mediated action on the Hes1 promoter are key in shaping the subcellular dynamics. Where Notch-mediated transcription of Hes1 dominates, there is Notch oscillation and maintenance of fate flexibility; Wnt-mediated transcription of Hes1 favours bistability akin to cell fate selection. Cells could therefore regulate the proportion of Wnt- and Notch-mediated control of the Hes1 promoter to coordinate the timing of cell fate selection as they migrate through the intestinal epithelium and are subject to reduced Wnt stimuli. Furthermore, mutant cells characterised by hyperstimulation of the Wnt pathway may, through coupling with Notch, invert cell fate in neighbouring healthy cells, enabling an aberrant cell to maintain its neighbours in mitotically active states.


Asunto(s)
Mucosa Intestinal/metabolismo , Modelos Biológicos , Receptores Notch/metabolismo , Transducción de Señal/fisiología , Factor de Transcripción HES-1/metabolismo , Vía de Señalización Wnt/fisiología , Relojes Biológicos/fisiología , Células Cultivadas , Simulación por Computador , Humanos , Receptor Cross-Talk/fisiología
5.
J Pathol ; 240(3): 315-328, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27512948

RESUMEN

The initiation and progression of breast cancer from the transformation of the normal epithelium to ductal carcinoma in situ (DCIS) and invasive disease is a complex process involving the acquisition of genetic alterations and changes in gene expression, alongside microenvironmental and recognized histological alterations. Here, we sought to comprehensively characterise the genomic and transcriptomic features of the MCF10 isogenic model of breast cancer progression, and to functionally validate potential driver alterations in three-dimensional (3D) spheroids that may provide insights into breast cancer progression, and identify targetable alterations in conditions more similar to those encountered in vivo. We performed whole genome, exome and RNA sequencing of the MCF10 progression series to catalogue the copy number and mutational and transcriptomic landscapes associated with progression. We identified a number of predicted driver mutations (including PIK3CA and TP53) that were acquired during transformation of non-malignant MCF10A cells to their malignant counterparts that are also present in analysed primary breast cancers from The Cancer Genome Atlas (TCGA). Acquisition of genomic alterations identified MYC amplification and previously undescribed RAB3GAP1-HRAS and UBA2-PDCD2L expressed in-frame fusion genes in malignant cells. Comparison of pathway aberrations associated with progression showed that, when cells are grown as 3D spheroids, they show perturbations of cancer-relevant pathways. Functional interrogation of the dependency on predicted driver events identified alterations in HRAS, PIK3CA and TP53 that selectively decreased cell growth and were associated with progression from preinvasive to invasive disease only when cells were grown as spheroids. Our results have identified changes in the genomic repertoire in cell lines representative of the stages of breast cancer progression, and demonstrate that genetic dependencies can be uncovered when cells are grown in conditions more like those in vivo. The MCF10 progression series therefore represents a good model with which to dissect potential biomarkers and to evaluate therapeutic targets involved in the progression of breast cancer. © 2016 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Asunto(s)
Neoplasias de la Mama/genética , Carcinoma Ductal de Mama/genética , Carcinoma Intraductal no Infiltrante/genética , Modelos Biológicos , Fosfatidilinositol 3-Quinasas/genética , Transcriptoma , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/patología , Carcinoma Intraductal no Infiltrante/patología , Línea Celular Tumoral , Transformación Celular Neoplásica , Fosfatidilinositol 3-Quinasa Clase I , ADN de Neoplasias/química , ADN de Neoplasias/genética , Progresión de la Enfermedad , Exoma/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Genoma , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Mutación , Fosfatidilinositol 3-Quinasas/metabolismo , Análisis de Secuencia de ADN , Esferoides Celulares , Proteína p53 Supresora de Tumor/genética
6.
J Cell Physiol ; 231(11): 2408-17, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27019299

RESUMEN

Epithelial cell adhesion to the surrounding extracellular matrix is necessary for their proper behavior and function. During pregnancy and lactation, mammary epithelial cells (MECs) receive signals from their interaction with laminin via ß1-integrin (ß1-itg) to establish apico-basal polarity and to differentiate in response to prolactin. Downstream of ß1-itg, the scaffold protein Integrin Linked Kinase (ILK) has been identified as the key signal transducer that is required for both lactational differentiation and the establishment of apico-basal polarity. ILK is an adaptor protein that forms the IPP complex with PINCH and Parvins, which are central to its adaptor functions. However, it is not known how ILK and its interacting partners control tissue-specific gene expression. Expression of ILK mutants, which weaken the interaction between ILK and Parvin, revealed that Parvins have a role in mammary epithelial differentiation. This conclusion was supported by shRNA-mediated knockdown of the Parvins. In addition, shRNA knockdown of the Parvin-binding guanine nucleotide exchange factor αPix prevented prolactin-induced differentiation. αPix depletion did not disrupt focal adhesions, MEC proliferation, or polarity. This suggests that αPix represents a differentiation-specific bifurcation point in ß1-itg-ILK adhesive signaling. In summary, this study has identified a new role for Parvin and αPix downstream of the integrin-ILK signaling axis for MEC differentiation. J. Cell. Physiol. 231: 2408-2417, 2016. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Diferenciación Celular , Células Epiteliales/citología , Integrina beta1/metabolismo , Glándulas Mamarias Animales/citología , Proteínas de Microfilamentos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Transducción de Señal , Animales , Diferenciación Celular/efectos de los fármacos , Polaridad Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Ratones , Mutación/genética , Prolactina/farmacología , Unión Proteica/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
7.
Breast Cancer Res ; 18(1): 5, 2016 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-26747277

RESUMEN

BACKGROUND: High mammographic density is a therapeutically modifiable risk factor for breast cancer. Although mammographic density is correlated with the relative abundance of collagen-rich fibroglandular tissue, the causative mechanisms, associated structural remodelling and mechanical consequences remain poorly defined. In this study we have developed a new collaborative bedside-to-bench workflow to determine the relationship between mammographic density, collagen abundance and alignment, tissue stiffness and the expression of extracellular matrix organising proteins. METHODS: Mammographic density was assessed in 22 post-menopausal women (aged 54-66 y). A radiologist and a pathologist identified and excised regions of elevated non-cancerous X-ray density prior to laboratory characterization. Collagen abundance was determined by both Masson's trichrome and Picrosirius red staining (which enhances collagen birefringence when viewed under polarised light). The structural specificity of these collagen visualisation methods was determined by comparing the relative birefringence and ultrastructure (visualised by atomic force microscopy) of unaligned collagen I fibrils in reconstituted gels with the highly aligned collagen fibrils in rat tail tendon. Localised collagen fibril organisation and stiffness was also evaluated in tissue sections by atomic force microscopy/spectroscopy and the abundance of key extracellular proteins was assessed using mass spectrometry. RESULTS: Mammographic density was positively correlated with the abundance of aligned periductal fibrils rather than with the abundance of amorphous collagen. Compared with matched tissue resected from the breasts of low mammographic density patients, the highly birefringent tissue in mammographically dense breasts was both significantly stiffer and characterised by large (>80 µm long) fibrillar collagen bundles. Subsequent proteomic analyses not only confirmed the absence of collagen fibrosis in high mammographic density tissue, but additionally identified the up-regulation of periostin and collagen XVI (regulators of collagen fibril structure and architecture) as potential mediators of localised mechanical stiffness. CONCLUSIONS: These preliminary data suggest that remodelling, and hence stiffening, of the existing stromal collagen microarchitecture promotes high mammographic density within the breast. In turn, this aberrant mechanical environment may trigger neoplasia-associated mechanotransduction pathways within the epithelial cell population.


Asunto(s)
Neoplasias de la Mama/genética , Colágeno/metabolismo , Glándulas Mamarias Humanas/anomalías , Mamografía/métodos , Proteómica , Anciano , Animales , Densidad de la Mama , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Moléculas de Adhesión Celular/metabolismo , Colágeno/ultraestructura , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Femenino , Humanos , Microscopía de Fuerza Atómica , Persona de Mediana Edad , Ratas , Factores de Riesgo
8.
Development ; 139(23): 4405-15, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23132247

RESUMEN

Notch and Wnt are highly conserved signalling pathways that are used repeatedly throughout animal development to generate a diverse array of cell types. However, they often have opposing effects on cell-fate decisions with each pathway promoting an alternate outcome. Commonly, a cell receiving both signals exhibits only Wnt pathway activity. This suggests that Wnt inhibits Notch activity to promote a Wnt-ON/Notch-OFF output; but what might underpin this Notch regulation is not understood. Here, we show that Wnt acts via Dishevelled to inhibit Notch signalling, and that this crosstalk regulates cell-fate specification in vivo during Xenopus development. Mechanistically, Dishevelled binds and directly inhibits CSL transcription factors downstream of Notch receptors, reducing their activity. Furthermore, our data suggest that this crosstalk mechanism is conserved between vertebrate and invertebrate homologues. Thus, we identify a dual function for Dishevelled as an inhibitor of Notch signalling and an activator of the Wnt pathway that sharpens the distinction between opposing Wnt and Notch responses, allowing for robust cell-fate decisions.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Fosfoproteínas/metabolismo , Receptores Notch/metabolismo , Proteínas Wnt/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus/embriología , Animales , Células CHO , Línea Celular , Cricetinae , Proteínas Dishevelled , Epidermis/embriología , Células HEK293 , Humanos , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/antagonistas & inhibidores , Receptores Notch/antagonistas & inhibidores , Vía de Señalización Wnt , Proteínas de Xenopus/antagonistas & inhibidores
9.
Cell Mol Life Sci ; 71(18): 3553-67, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24942883

RESUMEN

The Notch and Wnt pathways are two of only a handful of highly conserved signalling pathways that control cell-fate decisions during animal development (Pires-daSilva and Sommer in Nat Rev Genet 4: 39-49, 2003). These two pathways are required together to regulate many aspects of metazoan development, ranging from germ layer patterning in sea urchins (Peter and Davidson in Nature 474: 635-639, 2011) to the formation and patterning of the fly wing (Axelrod et al in Science 271:1826-1832, 1996; Micchelli et al in Development 124:1485-1495, 1997; Rulifson et al in Nature 384:72-74, 1996), the spacing of the ciliated cells in the epidermis of frog embryos (Collu et al in Development 139:4405-4415, 2012) and the maintenance and turnover of the skin, gut lining and mammary gland in mammals (Clayton et al in Nature 446:185-189, 2007; Clevers in Cell 154:274-284, 2013; Doupe et al in Dev Cell 18:317-323, 2010; Lim et al in Science 342:1226-1230, 2013; Lowell et al in Curr Biol 10:491-500, 2000; van et al in Nature 435:959-963, 2005; Yin et al in Nat Methods 11:106-112, 2013). In addition, many diseases, including several cancers, are caused by aberrant signalling through the two pathways (Bolós et al in Endocr Rev 28: 339-363, 2007; Clevers in Cell 127: 469-480, 2006). In this review, we will outline the two signalling pathways, describe the different points of interaction between them, and cover how these interactions influence development and disease.


Asunto(s)
Modelos Biológicos , Receptores Notch/fisiología , Vía de Señalización Wnt , Animales , Linaje de la Célula , Drosophila/crecimiento & desarrollo , Drosophila/metabolismo , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Mamíferos/crecimiento & desarrollo , Mamíferos/metabolismo , Receptores Notch/metabolismo , Transducción de Señal
10.
J Biol Chem ; 287(29): 24103-12, 2012 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-22511753

RESUMEN

Understanding how cell cycle is regulated in normal mammary epithelia is essential for deciphering defects of breast cancer and therefore for developing new therapies. Signals provided by both the extracellular matrix and growth factors are essential for epithelial cell proliferation. However, the mechanisms by which adhesion controls cell cycle in normal epithelia are poorly established. In this study, we describe the consequences of removing the ß1-integrin gene from primary cultures of mammary epithelial cells in situ, using CreER. Upon ß1-integrin gene deletion, the cells were unable to progress efficiently through S-phase, but were still able to undergo collective two-dimensional migration. These responses are explained by the presence of ß3-integrin in ß1-integrin-null cells, indicating that integrins containing different ß-subunits exert differential control on mammary epithelial proliferation and migration. ß1-Integrin deletion did not inhibit growth factor signaling to Erk or prevent the recruitment of core adhesome components to focal adhesions. Instead the S-phase arrest resulted from defective Rac activation and Erk translocation to the nucleus. Rac inhibition prevented Erk translocation and blocked proliferation. Activated Rac1 rescued the proliferation defect in ß1-integrin-depleted cells, indicating that this GTPase is essential in propagating proliferative ß1-integrin signals. These results show that ß1-integrins promote cell cycle in mammary epithelial cells, whereas ß3-integrins are involved in migration.


Asunto(s)
Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Integrina beta1/metabolismo , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/metabolismo , Animales , Diferenciación Celular/genética , Línea Celular , Movimiento Celular/genética , Proliferación Celular , Células Cultivadas , Femenino , Citometría de Flujo , Immunoblotting , Integrina beta1/genética , Masculino , Ratones , Embarazo , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/genética , Transducción de Señal/fisiología
11.
Breast Cancer Res ; 15(5): R92, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-24286369

RESUMEN

INTRODUCTION: Breast cancer remains a significant scientific, clinical and societal challenge. This gap analysis has reviewed and critically assessed enduring issues and new challenges emerging from recent research, and proposes strategies for translating solutions into practice. METHODS: More than 100 internationally recognised specialist breast cancer scientists, clinicians and healthcare professionals collaborated to address nine thematic areas: genetics, epigenetics and epidemiology; molecular pathology and cell biology; hormonal influences and endocrine therapy; imaging, detection and screening; current/novel therapies and biomarkers; drug resistance; metastasis, angiogenesis, circulating tumour cells, cancer 'stem' cells; risk and prevention; living with and managing breast cancer and its treatment. The groups developed summary papers through an iterative process which, following further appraisal from experts and patients, were melded into this summary account. RESULTS: The 10 major gaps identified were: (1) understanding the functions and contextual interactions of genetic and epigenetic changes in normal breast development and during malignant transformation; (2) how to implement sustainable lifestyle changes (diet, exercise and weight) and chemopreventive strategies; (3) the need for tailored screening approaches including clinically actionable tests; (4) enhancing knowledge of molecular drivers behind breast cancer subtypes, progression and metastasis; (5) understanding the molecular mechanisms of tumour heterogeneity, dormancy, de novo or acquired resistance and how to target key nodes in these dynamic processes; (6) developing validated markers for chemosensitivity and radiosensitivity; (7) understanding the optimal duration, sequencing and rational combinations of treatment for improved personalised therapy; (8) validating multimodality imaging biomarkers for minimally invasive diagnosis and monitoring of responses in primary and metastatic disease; (9) developing interventions and support to improve the survivorship experience; (10) a continuing need for clinical material for translational research derived from normal breast, blood, primary, relapsed, metastatic and drug-resistant cancers with expert bioinformatics support to maximise its utility. The proposed infrastructural enablers include enhanced resources to support clinically relevant in vitro and in vivo tumour models; improved access to appropriate, fully annotated clinical samples; extended biomarker discovery, validation and standardisation; and facilitated cross-discipline working. CONCLUSIONS: With resources to conduct further high-quality targeted research focusing on the gaps identified, increased knowledge translating into improved clinical care should be achievable within five years.


Asunto(s)
Neoplasias de la Mama , Investigación , Investigación Biomédica Traslacional , Animales , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/epidemiología , Neoplasias de la Mama/etiología , Neoplasias de la Mama/terapia , Femenino , Humanos
12.
PLoS One ; 18(12): e0294202, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38134188

RESUMEN

We performed two cross-sectional surveys across three informal settlements in Kenya (within Kisii county, Nairobi, and Nakuru county) to study the effectiveness of public health interventions during the COVID-19 pandemic. A total of 720 participants were surveyed from 120 randomly selected geographical locations (240 participants/settlement/survey), and a coordinated health promotion campaign was delivered between the two surveys by trained staff. Information relating to knowledge, attitudes, and practices (KAP) were collected by trained field workers using a validated questionnaire. The main outcomes showed improvements in: (i) mask-wearing (% of participants 'Always' using their mask increased from 71 to 74%, and the percentage using their masks 'Sometimes' decreased from 15% to 6%; p<0.001); (ii) practices related to face mask usage (% of subjects covering the mouth and nose increased from 91 to 95%, and those covering only part of their face decreased from around 2.5% to <1%; p<0.001). Significant improvements were also seen in the attitudes and expectations relating to mask wearing, and in the understanding of government directives. Over 50% of subjects in the post-campaign survey reported that social distancing was not possible in their communities and fears associated with COVID-19 testing were resistant to change (unchanged at 10%). Access to COVID-19 testing facilities was limited, leaving a large proportion of people unable to test. As willingness to take a COVID-19 test did not change between surveys (69 vs 70%; p = 0.57), despite increased availability, we recommend that policy level interventions are needed, aimed at mitigating adverse consequences of a positive test. Improvements of KAPs in the more crowded urban environment (Nairobi) were less than at settlements in rural or semi-urban settings (Nakuru and Kisii). We conclude that coordinated public health campaigns are effective in facilitating the change of KAPs amongst people living amidst challenging socio-economic conditions in informal settlements.


Asunto(s)
COVID-19 , Humanos , COVID-19/epidemiología , COVID-19/prevención & control , Kenia/epidemiología , Estudios Transversales , Pandemias/prevención & control , Prueba de COVID-19 , Conocimientos, Actitudes y Práctica en Salud , Promoción de la Salud
13.
Oncogene ; 42(9): 679-692, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36599922

RESUMEN

Breast cancer stem cells (BCSC) are presumed to be responsible for treatment resistance, tumor recurrence and metastasis of breast tumors. However, development of BCSC-targeting therapies has been held back by their heterogeneity and the lack of BCSC-selective molecular targets. Here, we demonstrate that RAC1B, the only known alternatively spliced variant of the small GTPase RAC1, is expressed in a subset of BCSCs in vivo and its function is required for the maintenance of BCSCs and their chemoresistance to doxorubicin. In human breast cancer cell line MCF7, RAC1B is required for BCSC plasticity and chemoresistance to doxorubicin in vitro and for tumor-initiating abilities in vivo. Unlike Rac1, Rac1b function is dispensable for normal mammary gland development and mammary epithelial stem cell (MaSC) activity. In contrast, loss of Rac1b function in a mouse model of breast cancer hampers the BCSC activity and increases their chemosensitivity to doxorubicin treatment. Collectively, our data suggest that RAC1B is a clinically relevant molecular target for the development of BCSC-targeting therapies that may improve the effectiveness of doxorubicin-mediated chemotherapy.


Asunto(s)
Neoplasias de la Mama , Neoplasias Mamarias Animales , Animales , Femenino , Humanos , Ratones , Neoplasias de la Mama/patología , Línea Celular Tumoral , Doxorrubicina/uso terapéutico , Resistencia a Antineoplásicos , Neoplasias Mamarias Animales/patología , Recurrencia Local de Neoplasia/patología , Células Madre Neoplásicas/patología
14.
Am J Hematol ; 87(7): 729-31, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22566293

RESUMEN

Plasma cell myeloma (PCM) is a lymphoproliferative disorder characterized by the malignant growth of monoclonal plasma cells within the bone marrow. Although risk factors for the development of PCM have been identified, the etiology on the majority of patients with PCM remains unclear. Cigarette smoking has been postulated as a potential risk factor for lymphoid malignancies; however, the association with PCM is inconclusive. We have carried out a meta-analysis of observational studies to assess the relationship, if any, between cigarette smoking and PCM. A literature search through December 2011 rendered 4 prospective cohort and 13 case­control studies evaluating such association. Our categorical meta-analysis showed that there is no association between ever, current, and former smokers and PCM.This lack of association was maintained when analyzing by study design, study quality, and geographical area of report. Similarly, meta regression analysis showed no association with the number of cigarettes smoked per day. In conclusion, our meta-analysis shows that there is no relationship between cigarette smoking and an increased incidence of PCM. Future studies should focus on other potential risk factors for PCM.


Asunto(s)
Mieloma Múltiple/etiología , Fumar/efectos adversos , Humanos , Incidencia , Mieloma Múltiple/epidemiología , Factores de Riesgo
15.
Cell Death Differ ; 29(11): 2262-2274, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35585181

RESUMEN

Apoptosis is regulated by interactions between the BH3-only and multi-domain Bcl-2 family proteins. These interactions are integrated on the outer mitochondrial membrane (OMM) where they set the threshold for apoptosis, known as mitochondrial priming. However, how mitochondrial priming is controlled at the level of single cells remains unclear. Retrotranslocation of Bcl-XL has been proposed as one mechanism, removing pro-apoptotic Bcl-2 proteins from the OMM, thus reducing priming. Contrary to this view, we now show that Bcl-XL retrotranslocation is inhibited by binding to its BH3-only partners, resulting in accumulation of these protein complexes on mitochondria. We find that Bcl-XL retrotranslocation dynamics are tightly coupled to mitochondrial priming. Quantifying these dynamics indicates the heterogeneity in priming between cells within a population and predicts how they subsequently respond to a pro-apoptotic signal.


Asunto(s)
Mitocondrias , Proteínas Proto-Oncogénicas c-bcl-2 , Citosol/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Mitocondrias/metabolismo , Membranas Mitocondriales/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis , Proteína bcl-X/metabolismo , Proteína X Asociada a bcl-2/metabolismo
16.
Oncogene ; 41(7): 1040-1049, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34916592

RESUMEN

Ectodysplasin A receptor (EDAR) is a death receptor in the Tumour Necrosis Factor Receptor (TNFR) superfamily with roles in the development of hair follicles, teeth and cutaneous glands. Here we report that human Oestrogen Receptor (ER) negative breast carcinomas which display squamous differentiation express EDAR strongly. Using a mouse model with a high Edar copy number, we show that elevated EDAR signalling results in a high incidence of mammary tumours in breeding female mice. These tumours resemble the EDAR-high human tumours in that they are characterised by a lack of oestrogen receptor expression, contain extensive squamous metaplasia, and display strong ß-catenin transcriptional activity. In the mouse model, all of the tumours carry somatic deletions of the third exon of the CTNNB1 gene that encodes ß-catenin. Deletion of this exon yields unconstrained ß-catenin signalling activity. We also demonstrate that ß-catenin activity is required for transformed cell growth, showing that increased EDAR signalling creates an environment in which ß-catenin activity can readily promote tumourigenesis. Together, this work identifies a novel death receptor oncogene in breast cancer, whose mechanism of transformation is based on the interaction between the WNT and Ectodysplasin A (EDA) pathways.


Asunto(s)
Receptores de la Ectodisplasina
17.
Front Cell Dev Biol ; 9: 692173, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34295896

RESUMEN

The Notch signalling pathway is a highly conserved developmental signalling pathway, with vital roles in determining cell fate during embryonic development and tissue homeostasis. Aberrant Notch signalling has been implicated in many disease pathologies, including cancer. In this review, we will outline the mechanism and regulation of the Notch signalling pathway. We will also outline the role Notch signalling plays in normal mammary gland development and how Notch signalling is implicated in breast cancer tumorigenesis and progression. We will cover how Notch signalling controls several different hallmarks of cancer within epithelial cells with sections focussed on its roles in proliferation, apoptosis, invasion, and metastasis. We will provide evidence for Notch signalling in the breast cancer stem cell phenotype, which also has implications for therapy resistance and disease relapse in breast cancer patients. Finally, we will summarise the developments in therapeutic targeting of Notch signalling, and the pros and cons of this approach for the treatment of breast cancer.

18.
Curr Biol ; 31(15): 3409-3418.e6, 2021 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-34111402

RESUMEN

Epithelial tissues are highly sensitive to anisotropies in mechanical force, with cells altering fundamental behaviors, such as cell adhesion, migration, and cell division.1-5 It is well known that, in the later stages of carcinoma (epithelial cancer), the presence of tumors alters the mechanical properties of a host tissue and that these changes contribute to disease progression.6-9 However, in the earliest stages of carcinoma, when a clonal cluster of oncogene-expressing cells first establishes in the epithelium, the extent to which mechanical changes alter cell behavior in the tissue as a whole remains unclear. This is despite knowledge that many common oncogenes, such as oncogenic Ras, alter cell stiffness and contractility.10-13 Here, we investigate how mechanical changes at the cellular level of an oncogenic cluster can translate into the generation of anisotropic strain across an epithelium, altering cell behavior in neighboring host tissue. We generated clusters of oncogene-expressing cells within otherwise normal in vivo epithelium, using Xenopus laevis embryos. We find that cells in kRasV12, but not cMYC, clusters have increased contractility, which introduces radial stress in the tissue and deforms surrounding host cells. The strain imposed by kRasV12 clusters leads to increased cell division and altered division orientation in neighboring host tissue, effects that can be rescued by reducing actomyosin contractility specifically in the kRasV12 cells. Our findings indicate that some oncogenes can alter the mechanical and proliferative properties of host tissue from the earliest stages of cancer development, changes that have the potential to contribute to tumorigenesis.


Asunto(s)
División Celular , Neoplasias , Oncogenes , Proteínas Proto-Oncogénicas p21(ras) , Animales , Anisotropía , Carcinogénesis/genética , Neoplasias/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Xenopus laevis
19.
Sci Rep ; 11(1): 9096, 2021 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-33907274

RESUMEN

Notch and Wnt are two essential signalling pathways that help to shape animals during development and to sustain adult tissue homeostasis. Although they are often active at the same time within a tissue, they typically have opposing effects on cell fate decisions. In fact, crosstalk between the two pathways is important in generating the great diversity of cell types that we find in metazoans. Several different mechanisms have been proposed that allow Notch to limit Wnt signalling, driving a Notch-ON/Wnt-OFF state. Here we explore these different mechanisms in human cells and demonstrate two distinct mechanisms by which Notch itself, can limit the transcriptional activity of ß-catenin. At the membrane, independently of DSL ligands, Notch1 can antagonise ß-catenin activity through an endocytic mechanism that requires its interaction with Deltex and sequesters ß-catenin into the membrane fraction. Within the nucleus, the intracellular domain of Notch1 can also limit ß-catenin induced transcription through the formation of a complex that requires its interaction with RBPjκ. We believe these mechanisms contribute to the robustness of cell-fate decisions by sharpening the distinction between opposing Notch/Wnt responses.


Asunto(s)
Receptores Notch/metabolismo , Vía de Señalización Wnt , beta Catenina/metabolismo , Núcleo Celular/metabolismo , Células HCT116 , Células HEK293 , Humanos , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Receptor Notch1/genética , Receptor Notch1/metabolismo , Receptores Notch/genética , Transcripción Genética , beta Catenina/genética , Proteínas de Unión al GTP rab5/metabolismo
20.
Cancer Res ; 81(4): 847-859, 2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33509944

RESUMEN

Triple-negative breast cancers (TNBC) are resistant to standard-of-care chemotherapy and lack known targetable driver gene alterations. Identification of novel drivers could aid the discovery of new treatment strategies for this hard-to-treat patient population, yet studies using high-throughput and accurate models to define the functions of driver genes in TNBC to date have been limited. Here, we employed unbiased functional genomics screening of the 200 most frequently mutated genes in breast cancer, using spheroid cultures to model in vivo-like conditions, and identified the histone acetyltransferase CREBBP as a novel tumor suppressor in TNBC. CREBBP protein expression in patient tumor samples was absent in 8% of TNBCs and at a high frequency in other tumors, including squamous lung cancer, where CREBBP-inactivating mutations are common. In TNBC, CREBBP alterations were associated with higher genomic heterogeneity and poorer patient survival and resulted in upregulation and dependency on a FOXM1 proliferative program. Targeting FOXM1-driven proliferation indirectly with clinical CDK4/6 inhibitors (CDK4/6i) selectively impaired growth in spheroids, cell line xenografts, and patient-derived models from multiple tumor types with CREBBP mutations or loss of protein expression. In conclusion, we have identified CREBBP as a novel driver in aggressive TNBC and identified an associated genetic vulnerability in tumor cells with alterations in CREBBP and provide a preclinical rationale for assessing CREBBP alterations as a biomarker of CDK4/6i response in a new patient population. SIGNIFICANCE: This study demonstrates that CREBBP genomic alterations drive aggressive TNBC, lung cancer, and lymphomas and may be selectively treated with clinical CDK4/6 inhibitors.


Asunto(s)
Proteína de Unión a CREB/fisiología , Carcinogénesis/genética , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología , Animales , Proteína de Unión a CREB/genética , Proliferación Celular/genética , Células Cultivadas , Ensayos de Selección de Medicamentos Antitumorales/métodos , Femenino , Genómica/métodos , Células HCT116 , Células HEK293 , Humanos , Ratones , Ratones Endogámicos NOD , Ratones Desnudos , Terapia Molecular Dirigida , Mutación , Invasividad Neoplásica , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA