Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
BMJ Open ; 12(4): e061421, 2022 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-35437256

RESUMO

INTRODUCTION: The human microbiota, the community of micro-organisms in different cavities, has been increasingly linked with inflammatory and neoplastic diseases. While investigation into the gut microbiome has been robust, the urinary microbiome has only recently been described. Investigation into the relationship between bladder cancer (BC) and the bladder and the intestinal microbiome may elucidate a pathophysiological relationship between the two. The bladder or the intestinal microbiome or the interplay between both may also act as a non-invasive biomarker for tumour behaviour. While these associations have not yet been fully investigated, urologists have been manipulating the bladder microbiome for treatment of BC for more than 40 years, treating high grade non-muscle invasive BC (NMIBC) with intravesical BCG immunotherapy. Neither the association between the microbiome sampled directly from bladder tissue and the response to BCG-therapy nor the association between response to BCG-therapy with the faecal microbiome has been studied until now. A prognostic tool prior to initiation of BCG-therapy is still needed. METHODS AND ANALYSIS: In patients with NMIBC bladder samples will be collected during surgery (bladder microbiome assessment), faecal samples (microbiome assessment), instrumented urine and blood samples (biobank) will also be taken. We will analyse the microbial community by 16S rDNA gene amplicon sequencing. The difference in alpha diversity (diversity of species within each sample) and beta diversity (change in species diversity) between BCG-candidates will be assessed. Subgroup analysis will be performed which will lead to the development of a clinical prediction model estimating risk of BCG-response. ETHICS AND DISSEMINATION: The study has been approved by the Cantonal Ethics Committee Zurich (2021-01783) and it is being conducted in accordance with the Declaration of Helsinki and Good Clinical Practice. Study results will be disseminated through peer-reviewed journals and national and international scientific conferences. TRIAL REGISTRATION NUMBER: NCT05204199.


Assuntos
Microbiota , Neoplasias da Bexiga Urinária , Adjuvantes Imunológicos , Administração Intravesical , Vacina BCG/uso terapêutico , Feminino , Humanos , Masculino , Modelos Estatísticos , Estudos Observacionais como Assunto , Prognóstico , Neoplasias da Bexiga Urinária/patologia
2.
Cancer Res ; 82(4): 681-694, 2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-34916221

RESUMO

Blood-borne metastasis of breast cancer involves a series of tightly regulated sequential steps, including the growth of a primary tumor lesion, intravasation of circulating tumor cells (CTC), and adaptation in various distant metastatic sites. The genes orchestrating each of these steps are poorly understood in physiologically relevant contexts, owing to the rarity of experimental models that faithfully recapitulate the biology, growth kinetics, and tropism of human breast cancer. Here, we conducted an in vivo loss-of-function CRISPR screen in newly derived CTC xenografts, unique in their ability to spontaneously mirror the human disease, and identified specific genetic dependencies for each step of the metastatic process. Validation experiments revealed sensitivities to inhibitors that are already available, such as PLK1 inhibitors, to prevent CTC intravasation. Together, these findings present a new tool to reclassify driver genes involved in the spread of human cancer, providing insights into the biology of metastasis and paving the way to test targeted treatment approaches. SIGNIFICANCE: A loss-of-function CRISPR screen in human CTC-derived xenografts identifies genes critical for individual steps of the metastatic cascade, suggesting novel drivers and treatment opportunities for metastatic breast cancers.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias da Mama/genética , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Células Neoplásicas Circulantes/metabolismo , Animais , Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/sangue , Neoplasias da Mama/patologia , Sistemas CRISPR-Cas , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Metástase Neoplásica , Células Neoplásicas Circulantes/patologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , RNA Guia de Cinetoplastídeos/genética , RNA Guia de Cinetoplastídeos/metabolismo , RNA-Seq/métodos , Análise de Sobrevida , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Quinase 1 Polo-Like
3.
Cell Rep ; 36(5): 109484, 2021 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-34348153

RESUMO

We lack a holistic understanding of the genetic programs orchestrating embryonic colon morphogenesis and governing damage response in the adult. A window into these programs is the transcriptomes of the epithelial and mesenchymal cell populations in the colon. Performing unbiased single-cell transcriptomic analyses of the developing mouse colon at different embryonic stages (embryonic day 14.5 [E14.5], E15.5, and E18.5), we capture cellular and molecular profiles of the stages before, during, and after the appearance of crypt structures, as well as in a model of adult colitis. The data suggest most adult lineages are established by E18.5. We find embryonic-specific gene expression profiles and cell populations that reappear in response to tissue damage. Comparison of the datasets from mice and human colitis suggests the processes are conserved. In this study, we provide a comprehensive single-cell atlas of the developing mouse colon and evidence for the reactivation of embryonic genes in disease.


Assuntos
Colo/embriologia , Colo/patologia , Perfilação da Expressão Gênica , Animais , Diferenciação Celular , Colite/genética , Modelos Animais de Doenças , Embrião de Mamíferos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/patologia , Mucosa Intestinal/embriologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Mesoderma/embriologia , Camundongos Endogâmicos C57BL , Análise de Célula Única
4.
Oncogene ; 40(1): 12-27, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33046799

RESUMO

Despite major progress in breast cancer research, the functional contribution of distinct cancer cell clones to malignant tumor progression and metastasis remains largely elusive. We have assessed clonal heterogeneity within individual primary tumors and metastases and also during the distinct stages of malignant tumor progression using clonal tracking of cancer cells in the MMTV-PyMT mouse model of metastatic breast cancer. Comparative gene expression analysis of clonal subpopulations reveals a substantial level of heterogeneity across and also within the various stages of breast carcinogenesis. The intra-stage heterogeneity is primarily manifested by differences in cell proliferation, also found within invasive carcinomas of luminal A-, luminal B-, and HER2-enriched human breast cancer. Surprisingly, in the mouse model of clonal tracing of cancer cells, chemotherapy mainly targets the slow-proliferative clonal populations and fails to efficiently repress the fast-proliferative populations. These insights may have considerable impact on therapy selection and response in breast cancer patients.


Assuntos
Neoplasias da Mama/patologia , Rastreamento de Células/métodos , Perfilação da Expressão Gênica/métodos , Neoplasias Mamárias Experimentais/patologia , Vírus do Tumor Mamário do Camundongo/patogenicidade , Receptor ErbB-2/genética , Animais , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Proliferação de Células , Evolução Clonal , Progressão da Doença , Feminino , Redes Reguladoras de Genes , Humanos , Microdissecção e Captura a Laser , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/virologia , Camundongos , Metástase Neoplásica , Estadiamento de Neoplasias , Análise de Sequência de RNA
5.
Cell Rep ; 32(10): 108105, 2020 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-32905777

RESUMO

Circulating tumor cells (CTCs) are shed from solid cancers in the form of single or clustered cells, and the latter display an extraordinary ability to initiate metastasis. Yet, the biological phenomena that trigger the shedding of CTC clusters from a primary cancerous lesion are poorly understood. Here, when dynamically labeling breast cancer cells along cancer progression, we observe that the majority of CTC clusters are undergoing hypoxia, while single CTCs are largely normoxic. Strikingly, we find that vascular endothelial growth factor (VEGF) targeting leads to primary tumor shrinkage, but it increases intra-tumor hypoxia, resulting in a higher CTC cluster shedding rate and metastasis formation. Conversely, pro-angiogenic treatment increases primary tumor size, yet it dramatically suppresses the formation of CTC clusters and metastasis. Thus, intra-tumor hypoxia leads to the formation of clustered CTCs with high metastatic ability, and a pro-angiogenic therapy suppresses metastasis formation through prevention of CTC cluster generation.


Assuntos
Hipóxia Celular/imunologia , Células Neoplásicas Circulantes/imunologia , Proteômica/métodos , Animais , Feminino , Humanos , Masculino , Camundongos
6.
Adv Sci (Weinh) ; 7(11): 1903237, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32537399

RESUMO

Cancer cells can be released from a cancerous lesion and migrate into the circulatory system, from whereon they may form metastases at distant sites. Today, it is possible to infer cancer progression and treatment efficacy by determining the number of circulating tumor cells (CTCs) in the patient's blood at multiple time points; further valuable information about CTC phenotypes remains inaccessible. In this article, a microfluidic method for integrated capture, isolation, and analysis of membrane markers as well as quantification of proteins secreted by single CTCs and CTC clusters is introduced. CTCs are isolated from whole blood with extraordinary efficiencies above 95% using dedicated trapping structures that allow co-capture of functionalized magnetic beads to assess protein secretion. The patform is tested with multiple breast cancer cell lines spiked into human blood and mouse-model-derived CTCs. In addition to immunostaining, the secretion level of granulocyte growth stimulating factor (G-CSF), which is shown to be involved in neutrophil recruitment, is quantified The bead-based assay provides a limit of detection of 1.5 ng mL-1 or less than 3700 molecules per cell. Employing barcoded magnetic beads, this platform can be adapted for multiplexed analysis and can enable comprehensive functional CTC profiling in the future.

7.
Recent Results Cancer Res ; 215: 347-368, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31605238

RESUMO

Next-generation sequencing of DNA and RNA obtained from liquid biopsies of cancer patients may reveal important insights into disease progression and metastasis formation, and it holds the promise to enable new methods for noninvasive screening and clinical decision support. However, implementing liquid biopsy sequencing protocols is challenged by capturing circulating tumor cells or cell-free tumor DNA from blood samples, by amplifying genomic DNA and RNA in a reliable and unbiased manner, and by extracting biologically meaningful signals from the noisy sequencing data. In this chapter, we discuss computational methods for the analysis of DNA and RNA sequencing data obtained from liquid biopsies, addressing these challenges.


Assuntos
DNA Tumoral Circulante/análise , DNA Tumoral Circulante/genética , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Biópsia Líquida , Neoplasias/diagnóstico , Neoplasias/genética , Análise de Sequência de DNA/métodos , Análise de Sequência de RNA/métodos , DNA Tumoral Circulante/sangue , Humanos
8.
Cancer Res ; 79(24): 6067-6073, 2019 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-31527091

RESUMO

The crosstalk between cancer cells and the immune system is crucial for disease progression and its therapeutic targeting is providing exciting results, in particular with newly developed immune checkpoint inhibitors. Current approaches primarily focus on cellular interactions occurring between tumor cells and T lymphocytes; however, recent data highlight a crucial role of neutrophils in support of tumor progression and suggest yet unexplored treatment opportunities. In this review, we summarize the current understanding of those interactions that occur between neutrophils and cancer cells, focusing on both protumor and antitumor activities of neutrophils at different stages of cancer progression. These include infiltration of neutrophils into the primary tumor, their interactions with circulating tumor cells (CTC) within the bloodstream, and their involvement in the establishment of a metastatic niche. Additionally, we discuss how further investigation of CTCs and their interacting immune cell partners may point towards novel immune checkpoint inhibition strategies and provide new insights on the efficacy of already existing immunotherapies.


Assuntos
Comunicação Celular/imunologia , Metástase Neoplásica/imunologia , Neoplasias/patologia , Células Neoplásicas Circulantes/imunologia , Neutrófilos/imunologia , Animais , Antineoplásicos Imunológicos/farmacologia , Antineoplásicos Imunológicos/uso terapêutico , Comunicação Celular/efeitos dos fármacos , Receptores Coestimuladores e Inibidores de Linfócitos T/antagonistas & inibidores , Receptores Coestimuladores e Inibidores de Linfócitos T/imunologia , Receptores Coestimuladores e Inibidores de Linfócitos T/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Humanos , Metástase Neoplásica/prevenção & controle , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Células Neoplásicas Circulantes/metabolismo , Infiltração de Neutrófilos , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/metabolismo
9.
Nature ; 573(7774): 439-444, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31485072

RESUMO

Metastasis is the major driver of death in patients with cancer. Invasion of surrounding tissues and metastasis have been proposed to initiate following loss of the intercellular adhesion protein, E-cadherin1,2, on the basis of inverse correlations between in vitro migration and E-cadherin levels3. However, this hypothesis is inconsistent with the observation that most breast cancers are invasive ductal carcinomas and express E-cadherin in primary tumours and metastases4. To resolve this discrepancy, we tested the genetic requirement for E-cadherin in metastasis using mouse and human models of both luminal and basal invasive ductal carcinomas. Here we show that E-cadherin promotes metastasis in diverse models of invasive ductal carcinomas. While loss of E-cadherin increased invasion, it also reduced cancer cell proliferation and survival, circulating tumour cell number, seeding of cancer cells in distant organs and metastasis outgrowth. Transcriptionally, loss of E-cadherin was associated with upregulation of genes involved in transforming growth factor-ß (TGFß), reactive oxygen species and apoptosis signalling pathways. At the cellular level, disseminating E-cadherin-negative cells exhibited nuclear enrichment of SMAD2/3, oxidative stress and increased apoptosis. Colony formation of E-cadherin-negative cells was rescued by inhibition of TGFß-receptor signalling, reactive oxygen accumulation or apoptosis. Our results reveal that E-cadherin acts as a survival factor in invasive ductal carcinomas during the detachment, systemic dissemination and seeding phases of metastasis by limiting reactive oxygen-mediated apoptosis. Identifying molecular strategies to inhibit E-cadherin-mediated survival in metastatic breast cancer cells may have potential as a therapeutic approach for breast cancer.


Assuntos
Antígenos CD , Neoplasias da Mama/patologia , Caderinas , Carcinoma Ductal de Mama/patologia , Invasividade Neoplásica , Metástase Neoplásica , Animais , Antígenos CD/metabolismo , Neoplasias da Mama/metabolismo , Caderinas/metabolismo , Carcinoma Ductal de Mama/metabolismo , Feminino , Humanos , Camundongos , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Fator de Crescimento Transformador beta/metabolismo
10.
J Vis Exp ; (147)2019 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-31157780

RESUMO

Blood-borne metastasis accounts for most cancer-related deaths and involves circulating tumor cells (CTCs) that are successful in establishing new tumors at distant sites. CTCs are found in the bloodstream of patients as single cells (single CTCs) or as multicellular aggregates (CTC clusters and CTC-white blood cell clusters), with the latter displaying a higher metastatic ability. Beyond enumeration, phenotypic and molecular analysis is extraordinarily important to dissect CTC biology and to identify actionable vulnerabilities. Here, we provide a detailed description of a workflow that includes CTC immunostaining and micromanipulation, ex vivo culture to assess proliferative and survival capabilities of individual cells, and in vivo metastasis-formation assays. Additionally, we provide a protocol to achieve the dissociation of CTC clusters into individual cells and the investigation of intra-cluster heterogeneity. With these approaches, for instance, we precisely quantify survival and proliferative potential of single CTCs and individual cells within CTC clusters, leading us to the observation that cells within clusters display better survival and proliferation in ex vivo cultures compared to single CTCs. Overall, our workflow offers a platform to dissect the characteristics of CTCs at the single cell level, aiming towards the identification of metastasis-relevant pathways and a better understanding of CTC biology.


Assuntos
Metástase Neoplásica/diagnóstico , Células Neoplásicas Circulantes , Animais , Humanos , Camundongos , Micromanipulação , Metástase Neoplásica/patologia
11.
Nature ; 566(7745): 553-557, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30728496

RESUMO

A better understanding of the features that define the interaction between cancer cells and immune cells is important for the development of new cancer therapies1. However, focus is often given to interactions that occur within the primary tumour and its microenvironment, whereas the role of immune cells during cancer dissemination in patients remains largely uncharacterized2,3. Circulating tumour cells (CTCs) are precursors of metastasis in several types of cancer4-6, and are occasionally found within the bloodstream in association with non-malignant cells such as white blood cells (WBCs)7,8. The identity and function of these CTC-associated WBCs, as well as the molecular features that define the interaction between WBCs and CTCs, are unknown. Here we isolate and characterize individual CTC-associated WBCs, as well as corresponding cancer cells within each CTC-WBC cluster, from patients with breast cancer and from mouse models. We use single-cell RNA sequencing to show that in the majority of these cases, CTCs were associated with neutrophils. When comparing the transcriptome profiles of CTCs associated with neutrophils against those of CTCs alone, we detect a number of differentially expressed genes that outline cell cycle progression, leading to more efficient metastasis formation. Further, we identify cell-cell junction and cytokine-receptor pairs that define CTC-neutrophil clusters, representing key vulnerabilities of the metastatic process. Thus, the association between neutrophils and CTCs drives cell cycle progression within the bloodstream and expands the metastatic potential of CTCs, providing a rationale for targeting this interaction in treatment of breast cancer.


Assuntos
Neoplasias da Mama/patologia , Ciclo Celular , Metástase Neoplásica/patologia , Células Neoplásicas Circulantes/patologia , Neutrófilos/patologia , Animais , Neoplasias da Mama/terapia , Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células , Éxons/genética , Feminino , Perfilação da Expressão Gênica , Humanos , Junções Intercelulares , Camundongos , Mutação/genética , Metástase Neoplásica/genética , Células Neoplásicas Circulantes/metabolismo , Neutrófilos/metabolismo , Análise de Sequência de RNA , Sequenciamento do Exoma
12.
Cell ; 176(1-2): 98-112.e14, 2019 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-30633912

RESUMO

The ability of circulating tumor cells (CTCs) to form clusters has been linked to increased metastatic potential. Yet biological features and vulnerabilities of CTC clusters remain largely unknown. Here, we profile the DNA methylation landscape of single CTCs and CTC clusters from breast cancer patients and mouse models on a genome-wide scale. We find that binding sites for stemness- and proliferation-associated transcription factors are specifically hypomethylated in CTC clusters, including binding sites for OCT4, NANOG, SOX2, and SIN3A, paralleling embryonic stem cell biology. Among 2,486 FDA-approved compounds, we identify Na+/K+ ATPase inhibitors that enable the dissociation of CTC clusters into single cells, leading to DNA methylation remodeling at critical sites and metastasis suppression. Thus, our results link CTC clustering to specific changes in DNA methylation that promote stemness and metastasis and point to cluster-targeting compounds to suppress the spread of cancer.


Assuntos
Neoplasias da Mama/genética , Metástase Neoplásica/genética , Células Neoplásicas Circulantes/patologia , Animais , Neoplasias da Mama/patologia , Diferenciação Celular , Linhagem Celular Tumoral , Proliferação de Células , Metilação de DNA/fisiologia , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Camundongos Endogâmicos NOD , Proteína Homeobox Nanog/metabolismo , Metástase Neoplásica/fisiopatologia , Células Neoplásicas Circulantes/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Complexo Correpressor Histona Desacetilase e Sin3
13.
Breast Cancer Res ; 20(1): 141, 2018 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-30458879

RESUMO

BACKGROUND: The presence of circulating tumor cells (CTCs) in patients with breast cancer correlates to a bad prognosis. Yet, CTCs are detectable in only a minority of patients with progressive breast cancer, and factors that influence the abundance of CTCs remain elusive. METHODS: We conducted CTC isolation and enumeration in a selected group of 73 consecutive patients characterized by progressive invasive breast cancer, high tumor load and treatment discontinuation at the time of CTC isolation. CTCs were quantified with the Parsortix microfluidic device. Clinicopathological variables, blood counts at the time of CTC isolation and detailed treatment history prior to blood sampling were evaluated for each patient. RESULTS: Among 73 patients, we detected at least one CTC per 7.5 ml of blood in 34 (46%). Of these, 22 (65%) had single CTCs only, whereas 12 (35%) featured both single CTCs and CTC clusters. Treatment with the monoclonal antibody denosumab correlated with the absence of CTCs, both when considering all patients and when considering only those with bone metastasis. We also found that low red blood cell count was associated with the presence of CTCs, whereas high CA 15-3 tumor marker, high mean corpuscular volume, high white blood cell count and high mean platelet volume associated specifically with CTC clusters. CONCLUSIONS: In addition to blood count correlatives to single and clustered CTCs, we found that denosumab treatment associates with most patients lacking CTCs from their peripheral circulation. Prospective studies will be needed to validate the involvement of denosumab in the prevention of CTC generation.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Denosumab/farmacologia , Eritrócitos , Células Neoplásicas Circulantes/efeitos dos fármacos , Idoso , Antineoplásicos/uso terapêutico , Mama/patologia , Neoplasias da Mama/sangue , Neoplasias da Mama/patologia , Contagem de Células/métodos , Denosumab/uso terapêutico , Progressão da Doença , Feminino , Humanos , Técnicas Analíticas Microfluídicas/métodos , Pessoa de Meia-Idade , Invasividade Neoplásica/patologia , Prognóstico , Estudos Retrospectivos
14.
Br J Cancer ; 119(4): 487-491, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30065256

RESUMO

Human glioblastoma (GBM) is a highly aggressive, invasive and hypervascularised malignant brain cancer. Individual circulating tumour cells (CTCs) are sporadically found in GBM patients, yet it is unclear whether multicellular CTC clusters are generated in this disease and whether they can bypass the physical hurdle of the blood-brain barrier.  Here, we assessed CTC presence and composition at multiple time points in 13 patients with progressing GBM during an open-label phase 1/2a study with the microtubule inhibitor BAL101553. We observe CTC clusters ranging from 2 to 23 cells and present at multiple sampling time points in a GBM patient with pleomorphism and extensive necrosis, throughout disease progression. Exome sequencing of GBM CTC clusters highlights variants in 58 cancer-associated genes including ATM, PMS2, POLE, APC, XPO1, TFRC, JAK2, ERBB4 and ALK. Together, our findings represent the first evidence of the presence of CTC clusters in GBM.


Assuntos
Benzimidazóis/administração & dosagem , Neoplasias Encefálicas/tratamento farmacológico , Glioblastoma/tratamento farmacológico , Células Neoplásicas Circulantes/patologia , Oxidiazóis/administração & dosagem , Animais , Benzimidazóis/farmacologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Contagem de Células , Análise por Conglomerados , Progressão da Doença , Feminino , Redes Reguladoras de Genes/efeitos dos fármacos , Variação Genética , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Masculino , Camundongos , Mutação , Células Neoplásicas Circulantes/química , Células Neoplásicas Circulantes/efeitos dos fármacos , Oxidiazóis/farmacologia , Sequenciamento do Exoma , Ensaios Antitumorais Modelo de Xenoenxerto
15.
ESMO Open ; 1(4): e000078, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27843628

RESUMO

The development of a metastatic disease is recognised as the cause of death of over 90% of patients diagnosed with cancer. Understanding the biological features of metastasis has been hampered for a long time by the difficulties to study widespread cancerous lesions in patients, and by the absence of reliable methods to isolate viable metastatic cells during disease progression. These difficulties negatively impact on our ability to develop new agents that are tailored to block the spread of cancer. Yet, recent advances in specialised devices for the isolation of circulating tumour cells (CTCs), hand-in-hand with technologies that enable single cell resolution interrogation of their genome and transcriptome, are now paving the way to understanding those molecular mechanisms that drive the formation of metastasis. In this review, we aim to summarise some of the latest discoveries in CTC biology in the context of several types of cancer, and to highlight those findings that have a potential to improve the clinical management of patients with metastatic cancer.

16.
Ann Rheum Dis ; 75(3): 617-22, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25906316

RESUMO

OBJECTIVES: Autoantibodies reactive with Ro52 are often found in sera of patients with Sjögren's syndrome (SS). This study was undertaken to investigate the role of Ro52-induced immune responses in pathogenesis of SS. METHODS: New Zealand Mixed (NZM) 2758 mice were immunised with Ro52 in alum adjuvant. Control mice were immunised either with maltose-binding protein or injected with alum alone. Mice were monitored for anti-Ro52 antibody, sialoadenitis and pilocarpine-induced salivation. Antibody binding to salivary gland (SG) cells was analysed in vivo and in vitro by immunofluorescence. Sera from immunised mice were passively transferred into untreated or alum injected NZM2758 mice. RESULTS: By day 30 post-immunisation, Ro52 immunised mice generated immunoprecipitating anti-Ro52 antibodies and they had the maximum drop in saliva production. Both Ro52 immunised and control mice showed evidence of mild sialoadenitis. However, only Ro52 immunised mice had antibody deposition in their SG. Passive transfer of Ro52-immune sera induced SG dysfunction in recipient mice, only if the recipients were primed with alum. In vitro, antibodies from Ro52-immune sera were internalised by a SG cell line and this uptake was inhibited by cytochalasin D treatment. CONCLUSIONS: Our data show for the first time that antibodies induced by Ro52 are capable of inducing SG dysfunction, and that this phenomenon is dependent on the activation of innate immunity. The mouse model described in this study implies that autoantibody deposition in the SG might be an important step in the induction of xerostomia and pathogenesis of SS.


Assuntos
Autoanticorpos/imunologia , Modelos Animais de Doenças , Imunidade Inata/imunologia , Imunização Passiva , Camundongos , Ribonucleoproteínas/imunologia , Sialadenite/imunologia , Síndrome de Sjogren/imunologia , Glândula Submandibular/imunologia , Animais , Humanos , Imunoglobulina G/imunologia , Sialadenite/patologia , Síndrome de Sjogren/patologia , Glândula Submandibular/patologia
17.
Nephron Exp Nephrol ; 126(3): 141-7, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24854879

RESUMO

BACKGROUND: Lipoprotein abnormalities are associated with a rapid decline in renal function in patients of chronic kidney disease. In addition, hyperlipidemia is associated with an increased risk of developing renal insufficiency. The underlying molecular mechanisms for these clinical findings are unclear. We have previously reported a role for inhibitor of differentiation 3 (ID3), a transcription factor, in regulating kidney disease in hyperlipidemia. Introducing a genetic deficiency of Id3 in spontaneously hyperlipidemic apolipoprotein E knockout (Apoe(-/-)) mice led to accelerated mesangioproliferative glomerulonephritis. The present study was carried out to further investigate the contribution of ID3 in hyperlipidemia-associated kidney disease. METHODS: Female C57BL/6 mice that were ID3-sufficient wild-type (WT) or ID3-deficient (Id3(-/-)) were fed a Western diet and evaluated for proteinuria, glomerular pathology, and immune infiltrating cells. Primary mesangial cell lines were generated from both mouse strains and stimulated with oxidized phospholipids. Cytokines and chemokines produced were measured by multiplex assays, ELISA, and QPCR. Glomerular isolates were studied for CXCL1 expression by QPCR. RESULTS: Id3(-/-) mice on a Western diet developed accelerated proteinuria and mesangioproliferative glomerulonephritis compared to WT controls. In vitro, Id3(-/-) glomerular mesangial cell lines produced higher levels of the monocyte chemoattractant CXCL1 in response to oxidized phospholipids. This was consistent with the rapid increase in glomerular CXCL1 expression followed by macrophage infiltration in Id3(-/-) mice fed a Western diet. CONCLUSIONS: A functional ID3 influences susceptibility to kidney disease and prevents glomerular injury by regulating local chemokine production and inflammatory cell recruitment.


Assuntos
Hiperlipidemias/metabolismo , Hiperlipidemias/patologia , Proteínas Inibidoras de Diferenciação/fisiologia , Nefropatias/metabolismo , Nefropatias/patologia , Animais , Linhagem Celular , Feminino , Ativação de Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fatores de Transcrição
18.
Clin Immunol ; 152(1-2): 1-9, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24576620

RESUMO

This study was undertaken to test the hypothesis that Sjogren's syndrome Antigen A (SSA)/Ro60-reactive T cells are activated by peptides originating from oral and gut bacteria. T cell hybridomas generated from HLA-DR3 transgenic mice recognized 3 regions on Ro60, with core epitopes mapped to amino acids 228-238, 246-256 and 371-381. BLAST analysis identified several mimicry peptides, originating from human oral, intestinal, skin and vaginal bacteria, as well as environmental bacteria. Amongst these, a peptide from the von Willebrand factor type A domain protein (vWFA) from the oral microbe Capnocytophaga ochracea was the most potent activator. Further, Ro60-reactive T cells were activated by recombinant vWFA protein and whole Escherichia coli expressing this protein. These results demonstrate that peptides derived from normal human microbiota can activate Ro60-reactive T cells. Thus, immune responses to commensal microbiota and opportunistic pathogens should be explored as potential triggers for initiating autoimmunity in SLE and Sjögren's syndrome.


Assuntos
Epitopos de Linfócito T/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Mimetismo Molecular/imunologia , Ribonucleoproteínas/imunologia , Síndrome de Sjogren/imunologia , Sequência de Aminoácidos , Animais , Autoimunidade/imunologia , Capnocytophaga/genética , Capnocytophaga/imunologia , Reações Cruzadas/imunologia , Feminino , Antígeno HLA-DR3/imunologia , Humanos , Hibridomas/imunologia , Intestinos/microbiologia , Ativação Linfocitária/imunologia , Camundongos , Boca/microbiologia , Peptídeos/genética , Peptídeos/imunologia , Proteínas Recombinantes/imunologia , Pele/microbiologia , Linfócitos T/imunologia , Vagina/microbiologia , Fator de von Willebrand/genética , Fator de von Willebrand/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...