Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 104
Filtrar
1.
Cell ; 187(6): 1335-1342, 2024 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-38490175

RESUMO

Gender inequality in STEM fields remains pervasive and undermines the ability for talented individuals to excel. Despite advances, women still encounter obstacles in pursuing academic careers and reaching leadership positions. This commentary discusses the "scissor-shaped curve" and examines effective strategies to fix it, including data-driven initiatives that we have implemented at our university.


Assuntos
Academia , Equidade de Gênero , Humanos , Feminino , Liderança , Universidades
2.
Cell ; 187(7): 1589-1616, 2024 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-38552609

RESUMO

The last 50 years have witnessed extraordinary developments in understanding mechanisms of carcinogenesis, synthesized as the hallmarks of cancer. Despite this logical framework, our understanding of the molecular basis of systemic manifestations and the underlying causes of cancer-related death remains incomplete. Looking forward, elucidating how tumors interact with distant organs and how multifaceted environmental and physiological parameters impinge on tumors and their hosts will be crucial for advances in preventing and more effectively treating human cancers. In this perspective, we discuss complexities of cancer as a systemic disease, including tumor initiation and promotion, tumor micro- and immune macro-environments, aging, metabolism and obesity, cancer cachexia, circadian rhythms, nervous system interactions, tumor-related thrombosis, and the microbiome. Model systems incorporating human genetic variation will be essential to decipher the mechanistic basis of these phenomena and unravel gene-environment interactions, providing a modern synthesis of molecular oncology that is primed to prevent cancers and improve patient quality of life and cancer outcomes.


Assuntos
Neoplasias , Humanos , Carcinogênese , Microbiota , Neoplasias/genética , Neoplasias/patologia , Neoplasias/terapia , Obesidade/complicações , Qualidade de Vida
3.
Cell ; 186(8): 1523-1527, 2023 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-37059060

RESUMO

Our understanding of tumorigenesis and cancer progression as well as clinical therapies for different cancer types have evolved dramatically in recent years. However, even with this progress, there are big challenges for scientists and oncologists to tackle, ranging from unpacking the molecular and cellular mechanisms involved to therapeutics and biomarker development to quality of life in the aftermath of therapy. In this article, we asked researchers to comment on the questions that they think are important to address in the coming years.


Assuntos
Neoplasias , Pesquisadores , Humanos , Carcinogênese , Neoplasias/sangue , Neoplasias/patologia , Neoplasias/terapia , Qualidade de Vida , Pesquisa , Biomarcadores Tumorais/sangue
4.
Cell ; 186(21): 4546-4566.e27, 2023 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-37769657

RESUMO

Neutrophils are abundant immune cells in the circulation and frequently infiltrate tumors in substantial numbers. However, their precise functions in different cancer types remain incompletely understood, including in the brain microenvironment. We therefore investigated neutrophils in tumor tissue of glioma and brain metastasis patients, with matched peripheral blood, and herein describe the first in-depth analysis of neutrophil phenotypes and functions in these tissues. Orthogonal profiling strategies in humans and mice revealed that brain tumor-associated neutrophils (TANs) differ significantly from blood neutrophils and have a prolonged lifespan and immune-suppressive and pro-angiogenic capacity. TANs exhibit a distinct inflammatory signature, driven by a combination of soluble inflammatory mediators including tumor necrosis factor alpha (TNF-ɑ) and Ceruloplasmin, which is more pronounced in TANs from brain metastasis versus glioma. Myeloid cells, including tumor-associated macrophages, emerge at the core of this network of pro-inflammatory mediators, supporting the concept of a critical myeloid niche regulating overall immune suppression in human brain tumors.

5.
Cell ; 181(7): 1643-1660.e17, 2020 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-32470396

RESUMO

Brain malignancies encompass a range of primary and metastatic cancers, including low-grade and high-grade gliomas and brain metastases (BrMs) originating from diverse extracranial tumors. Our understanding of the brain tumor microenvironment (TME) remains limited, and it is unknown whether it is sculpted differentially by primary versus metastatic disease. We therefore comprehensively analyzed the brain TME landscape via flow cytometry, RNA sequencing, protein arrays, culture assays, and spatial tissue characterization. This revealed disease-specific enrichment of immune cells with pronounced differences in proportional abundance of tissue-resident microglia, infiltrating monocyte-derived macrophages, neutrophils, and T cells. These integrated analyses also uncovered multifaceted immune cell activation within brain malignancies entailing converging transcriptional trajectories while maintaining disease- and cell-type-specific programs. Given the interest in developing TME-targeted therapies for brain malignancies, this comprehensive resource of the immune landscape offers insights into possible strategies to overcome tumor-supporting TME properties and instead harness the TME to fight cancer.


Assuntos
Neoplasias Encefálicas/imunologia , Glioma/patologia , Microambiente Tumoral/imunologia , Encéfalo/imunologia , Encéfalo/metabolismo , Neoplasias Encefálicas/patologia , Feminino , Glioma/metabolismo , Humanos , Linfócitos do Interstício Tumoral/imunologia , Macrófagos/imunologia , Masculino , Microglia/metabolismo , Neutrófilos/metabolismo , Linfócitos T/metabolismo
6.
Immunity ; 53(5): 985-1000.e11, 2020 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-33128876

RESUMO

Central memory CD8+ T cells (Tcm) control systemic secondary infections and can protect from chronic infection and cancer as a result of their stem-cell-like capacity to expand, differentiate, and self-renew. Central memory is generally thought to emerge following pathogen clearance and to form based on the de-differentiation of cytolytic effector cells. Here, we uncovered rare effector-phase CD8+ T cells expressing high amounts of the transcription factor Tcf7 (Tcf1) that showed no evidence of prior cytolytic differentiation and that displayed key hallmarks of Tcm cells. These effector-phase Tcf7hi cells quantitatively yielded Tcm cells based on lineage tracing. Mechanistically, Tcf1 counteracted the differentiation of Tcf7hi cells and sustained the expression of conserved adult stem-cell genes that were critical for CD8+ T cell stemness. The discovery of stem-cell-like CD8+ T cells during the effector response to acute infection provides an opportunity to optimize Tcm cell formation by vaccination.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Diferenciação Celular/imunologia , Citotoxicidade Imunológica , Fator 1-alfa Nuclear de Hepatócito/metabolismo , Memória Imunológica , Fator 1 de Transcrição de Linfócitos T/metabolismo , Animais , Linfócitos T CD8-Positivos/citologia , Diferenciação Celular/genética , Montagem e Desmontagem da Cromatina , Citotoxicidade Imunológica/genética , Imunofluorescência , Expressão Gênica , Fator 1-alfa Nuclear de Hepatócito/química , Fator 1-alfa Nuclear de Hepatócito/genética , Humanos , Imunização , Memória Imunológica/genética , Imunofenotipagem , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Moleculares , Conformação Proteica , Baço/imunologia , Baço/metabolismo , Relação Estrutura-Atividade , Fator 1 de Transcrição de Linfócitos T/química , Fator 1 de Transcrição de Linfócitos T/genética
7.
Cell ; 153(2): 449-60, 2013 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-23562644

RESUMO

The p53 tumor suppressor can restrict malignant transformation by triggering cell-autonomous programs of cell-cycle arrest or apoptosis. p53 also promotes cellular senescence, a tumor-suppressive program that involves stable cell-cycle arrest and secretion of factors that modify the tissue microenvironment. In the presence of chronic liver damage, we show that ablation of a p53-dependent senescence program in hepatic stellate cells increases liver fibrosis and cirrhosis associated with reduced survival and enhances the transformation of adjacent epithelial cells into hepatocellular carcinoma. p53-expressing senescent stellate cells release factors that skew macrophage polarization toward a tumor-inhibiting M1-state capable of attacking senescent cells in culture, whereas proliferating p53-deficient stellate cells secrete factors that stimulate polarization of macrophages into a tumor-promoting M2-state and enhance the proliferation of premalignant cells. Hence, p53 can act non-cell autonomously to suppress tumorigenesis by promoting an antitumor microenvironment, in part, through secreted factors that modulate macrophage function.


Assuntos
Transformação Celular Neoplásica , Senescência Celular , Células Estreladas do Fígado/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Proteína Supressora de Tumor p53/metabolismo , Animais , Microambiente Celular , Fibrose/patologia , Células Estreladas do Fígado/citologia , Humanos , Inflamação/metabolismo , Células de Kupffer/metabolismo , Células de Kupffer/patologia , Fígado/citologia , Fígado/patologia , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , NF-kappa B
8.
Immunity ; 48(1): 13-16, 2018 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-29343434

RESUMO

Myeloid cells, including neutrophils, are important regulators of tumor growth and metastasis. In Science, Engblom et al. (2017) reveal how lung tumors remotely engage bone-resident cells through a relay mechanism that achieves a sustained supply of tumor-promoting neutrophils.


Assuntos
Osso e Ossos , Neutrófilos , Humanos , Neoplasias Pulmonares , Células Mieloides , Microambiente Tumoral
9.
Immunity ; 56(6): 1153-1154, 2023 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-37315527
10.
11.
EMBO J ; 39(15): e103790, 2020 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-32567735

RESUMO

Tumour-associated microglia/macrophages (TAM) are the most numerous non-neoplastic populations in the tumour microenvironment in glioblastoma multiforme (GBM), the most common malignant brain tumour in adulthood. The mTOR pathway, an important regulator of cell survival/proliferation, is upregulated in GBM, but little is known about the potential role of this pathway in TAM. Here, we show that GBM-initiating cells induce mTOR signalling in the microglia but not bone marrow-derived macrophages in both in vitro and in vivo GBM mouse models. mTOR-dependent regulation of STAT3 and NF-κB activity promotes an immunosuppressive microglial phenotype. This hinders effector T-cell infiltration, proliferation and immune reactivity, thereby contributing to tumour immune evasion and promoting tumour growth in mouse models. The translational value of our results is demonstrated in whole transcriptome datasets of human GBM and in a novel in vitro model, whereby expanded-potential stem cells (EPSC)-derived microglia-like cells are conditioned by syngeneic patient-derived GBM-initiating cells. These results raise the possibility that microglia could be the primary target of mTOR inhibition, rather than the intrinsic tumour cells in GBM.


Assuntos
Neoplasias Encefálicas/imunologia , Glioblastoma/imunologia , Tolerância Imunológica , Microglia/imunologia , Proteínas de Neoplasias/imunologia , Serina-Treonina Quinases TOR/imunologia , Microambiente Tumoral/imunologia , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Camundongos , Camundongos Knockout , Microglia/patologia , Proteínas de Neoplasias/genética , Serina-Treonina Quinases TOR/genética , Microambiente Tumoral/genética
12.
Genes Dev ; 30(2): 220-32, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26773004

RESUMO

Proteases are important for regulating multiple tumorigenic processes, including angiogenesis, tumor growth, and invasion. Elevated protease expression is associated with poor patient prognosis across numerous tumor types. Several multigene protease families have been implicated in cancer, including cysteine cathepsins. However, whether individual family members have unique roles or are functionally redundant remains poorly understood. Here we demonstrate stage-dependent effects of simultaneously deleting cathepsin B (CtsB) and CtsS in a murine pancreatic neuroendocrine tumor model. Early in tumorigenesis, the double knockout results in an additive reduction in angiogenic switching, whereas at late stages, several tumorigenic phenotypes are unexpectedly restored to wild-type levels. We identified CtsZ, which is predominantly supplied by tumor-associated macrophages, as the compensatory protease that regulates the acquired tumor-promoting functions of lesions deficient in both CtsB and CtsS. Thus, deletion of multiple cathepsins can lead to stage-dependent, compensatory mechanisms in the tumor microenvironment, which has potential implications for the clinical consideration of selective versus pan-family cathepsin inhibitors in cancer.


Assuntos
Carcinoma Neuroendócrino/enzimologia , Catepsinas/genética , Catepsinas/metabolismo , Deleção de Genes , Neoplasias Pancreáticas/enzimologia , Animais , Apoptose/genética , Carcinogênese/genética , Carcinoma Neuroendócrino/genética , Carcinoma Neuroendócrino/fisiopatologia , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica , Técnicas de Inativação de Genes , Macrófagos/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Invasividade Neoplásica/genética , Neovascularização Patológica/enzimologia , Neovascularização Patológica/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/fisiopatologia
13.
Genes Dev ; 28(21): 2331-47, 2014 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-25367033

RESUMO

Pericellular proteases have long been associated with cancer invasion and metastasis due to their ability to degrade extracellular matrix components. Recent studies demonstrate that proteases also modulate tumor progression and metastasis through highly regulated and complex processes involving cleavage, processing, or shedding of cell adhesion molecules, growth factors, cytokines, and kinases. In this review, we address how cancer cells, together with their surrounding microenvironment, regulate pericellular proteolysis. We dissect the multitude of mechanisms by which pericellular proteases contribute to cancer progression and discuss how this knowledge can be integrated into therapeutic opportunities.


Assuntos
Neoplasias/enzimologia , Neoplasias/fisiopatologia , Peptídeo Hidrolases/metabolismo , Microambiente Tumoral/fisiologia , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Exossomos/metabolismo , Transporte Proteico , Proteólise
14.
Genes Dev ; 28(19): 2134-50, 2014 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-25274726

RESUMO

During the process of tumor progression, cancer cells can produce the requisite growth- and invasion-promoting factors and can also rely on noncancerous cells in the tumor microenvironment as an alternative, cell-extrinsic source. However, whether the cellular source influences the function of such tumor-promoting factors remains an open question. Here, we examined the roles of the cathepsin Z (CtsZ) protease, which is provided by both cancer cells and macrophages in pancreatic neuroendocrine tumors in humans and mice. We found that tumor proliferation was exclusively regulated by cancer cell-intrinsic functions of CtsZ, whereas tumor invasion required contributions from both macrophages and cancer cells. Interestingly, several of the tumor-promoting functions of CtsZ were not dependent on its described catalytic activity but instead were mediated via the Arg-Gly-Asp (RGD) motif in the enzyme prodomain, which regulated interactions with integrins and the extracellular matrix. Together, these results underscore the complexity of interactions within the tumor microenvironment and indicate that cellular source can indeed impact molecular function.


Assuntos
Catepsina Z/metabolismo , Matriz Extracelular/metabolismo , Macrófagos/enzimologia , Neoplasias/enzimologia , Neoplasias/fisiopatologia , Animais , Linhagem Celular Tumoral , Integrinas/metabolismo , Camundongos Endogâmicos C57BL , Invasividade Neoplásica/fisiopatologia
15.
Int J Geriatr Psychiatry ; 37(1)2021 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-34741340

RESUMO

BACKGROUND: Poor social health is prevalent in older adults and may be associated with worse cognition, and increased dementia risk. The aim of this study was to determine whether social isolation, social support and loneliness are independently associated with cognitive function and incident dementia over 5 years in older adults, and to investigate potential gender differences. METHODS: Participants were 11,498 community-dwelling relatively healthy Australians aged 70-94, in the ASPREE Longitudinal Study of Older Persons (ALSOP). Social isolation, social support, loneliness and cognitive function were assessed through self-report. Outcomes examined were cognitive decline (>1.5 SD decline in cognitive performance since baseline) and incident dementia (adjudicated according to DSM-IV criteria). RESULTS: Most participants self-reported good social health (92%) with very few socially isolated (2%), with low social support (2%) or lonely (5%). Among women, social isolation and low social support were consistently associated with lower cognitive function (e.g., social support and cognition ß = -1.17, p < 0.001). No consistent longitudinal associations were observed between baseline social health and cognitive decline (over median 3.1 years) or incident dementia (over median 4.4 years; social isolation: HR = 1.00, p = 0.99; low social support: HR = 1.79, p = 0.11; loneliness: HR = 0.72, p = 0.34 among women and men). CONCLUSION: Our study provides evidence that social isolation and a low social support are associated with worse cognitive function in women, but not men. Social health did not predict incident cognitive decline or dementia, but we lacked power to stratify dementia analyses by gender.

16.
Int J Geriatr Psychiatry ; 36(11): 1795-1809, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34231940

RESUMO

BACKGROUND: Social health reflects one's ability to form interpersonal relationships. Poor social health is a risk factor for cardiovascular disease (CVD), however an in-depth exploration of the link through CVD risk factors is lacking. AIM: To examine the relationship between social health (social isolation, social support, loneliness) and CVD risk factors among healthy older women and men. METHODS: Data were from 11,498 healthy community-dwelling Australians aged ≥70 years from the ASPirin in Reducing Events in the Elderly (ASPREE) trial and the ASPREE Longitudinal Study of Older Persons sub-study. Ten-year CVD risk was estimated using the Atherosclerotic CVD Risk Scale (ASCVDRS) and the Framingham Risk Score (FRS). RESULTS: Physical inactivity and experiencing depressive symptoms were the only CVD risk factors that consistently differed by all three social health constructs. Loneliness was associated with greater ASCVDRS (women: ß = 0.01, p < 0.05; men: ß = 0.03, p < 0.001), social isolation with greater FRS (women: ß = 0.02, p < 0.01; men: ß = 0.03, p < 0.01) and the social health composite of being lonely (regardless of social isolation and/or social support status) with greater ASCVDRS (women: ß = 0.01, p = 0.02; men: ß = 0.03, p < 0.001). Among men, loneliness was also associated with greater FRS (ß = 0.03, p < 0.001) and social support with greater ASCVDRS (ß = 0.02, p = 0.01). Men were more socially isolated, less socially supported and less lonely than women. CONCLUSION: Social isolation, social support and loneliness displayed diverse relationships with CVD risk factors and risk scores, emphasising the importance of distinguishing between these constructs. These findings inform on potential avenues to manage poor social health and CVD risk among older adults.


Assuntos
Doenças Cardiovasculares , Isolamento Social , Idoso , Idoso de 80 Anos ou mais , Austrália/epidemiologia , Doenças Cardiovasculares/epidemiologia , Estudos Transversais , Feminino , Humanos , Solidão , Estudos Longitudinais , Masculino , Fatores de Risco , Apoio Social
17.
Proc Natl Acad Sci U S A ; 114(11): 2934-2939, 2017 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-28246332

RESUMO

The genetic and phenotypic diversity of cells within tumors is a major obstacle for cancer treatment. Because of the stochastic nature of genetic alterations, this intratumoral heterogeneity is often viewed as chaotic. Here we show that the altered metabolism of cancer cells creates predictable gradients of extracellular metabolites that orchestrate the phenotypic diversity of cells in the tumor microenvironment. Combining experiments and mathematical modeling, we show that metabolites consumed and secreted within the tumor microenvironment induce tumor-associated macrophages (TAMs) to differentiate into distinct subpopulations according to local levels of ischemia and their position relative to the vasculature. TAMs integrate levels of hypoxia and lactate into progressive activation of MAPK signaling that induce predictable spatial patterns of gene expression, such as stripes of macrophages expressing arginase 1 (ARG1) and mannose receptor, C type 1 (MRC1). These phenotypic changes are functionally relevant as ischemic macrophages triggered tube-like morphogenesis in neighboring endothelial cells that could restore blood perfusion in nutrient-deprived regions where angiogenic resources are most needed. We propose that gradients of extracellular metabolites act as tumor morphogens that impose order within the microenvironment, much like signaling molecules convey positional information to organize embryonic tissues. Unearthing embryology-like processes in tumors may allow us to control organ-like tumor features such as tissue repair and revascularization and treat intratumoral heterogeneity.


Assuntos
Neoplasias/metabolismo , Neoplasias/patologia , Microambiente Tumoral , Linhagem Celular Tumoral , Análise por Conglomerados , Metabolismo Energético , Espaço Extracelular/metabolismo , Perfilação da Expressão Gênica , Humanos , Hipóxia/metabolismo , Ácido Láctico/metabolismo , Sistema de Sinalização das MAP Quinases , Macrófagos/metabolismo , Macrófagos/patologia , Neoplasias/genética , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Oxigênio/metabolismo , Fenótipo , Transcriptoma , Microambiente Tumoral/genética
18.
Genes Dev ; 25(23): 2465-79, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-22156207

RESUMO

The microenvironment is known to critically modulate tumor progression, yet its role in regulating treatment response is poorly understood. Here we found increased macrophage infiltration and cathepsin protease levels in mammary tumors following paclitaxel (Taxol) chemotherapy. Cathepsin-expressing macrophages protected against Taxol-induced tumor cell death in coculture, an effect fully reversed by cathepsin inhibition and mediated partially by cathepsins B and S. Macrophages were also found to protect against tumor cell death induced by additional chemotherapeutics, specifically etoposide and doxorubicin. Combining Taxol with cathepsin inhibition in vivo significantly enhanced efficacy against primary and metastatic tumors, supporting the therapeutic relevance of this effect. Additionally incorporating continuous low-dose cyclophosphamide dramatically impaired tumor growth and metastasis and improved survival. This study highlights the importance of integrated targeting of the tumor and its microenvironment and implicates macrophages and cathepsins in blunting chemotherapeutic response.


Assuntos
Antineoplásicos/farmacologia , Catepsinas/metabolismo , Macrófagos/patologia , Neoplasias Mamárias Animais/tratamento farmacológico , Paclitaxel/farmacologia , Animais , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Interações Medicamentosas , Resistencia a Medicamentos Antineoplásicos , Etoposídeo/farmacologia , Etoposídeo/uso terapêutico , Feminino , Neoplasias Pulmonares/prevenção & controle , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Animais/enzimologia , Neoplasias Mamárias Animais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Invasividade Neoplásica , Paclitaxel/uso terapêutico
19.
Genes Dev ; 24(3): 241-55, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-20080943

RESUMO

Innate immune cells can constitute a substantial proportion of the cells within the tumor microenvironment and have been associated with tumor malignancy in patients and animal models of cancer; however, the mechanisms by which they modulate cancer progression are incompletely understood. Here, we show that high levels of cathepsin protease activity are induced in the majority of macrophages in the microenvironment of pancreatic islet cancers, mammary tumors, and lung metastases during malignant progression. We further show that tumor-associated macrophage (TAM)-supplied cathepsins B and S are critical for promoting pancreatic tumor growth, angiogenesis, and invasion in vivo, and markedly enhance the invasiveness of cancer cells in culture. Finally, we demonstrate that interleukin-4 (IL-4) is responsible for inducing cathepsin activity in macrophages in vitro and in vivo. Together, these data establish IL-4 as an important regulator, and cathepsin proteases as critical mediators, of the cancer-promoting functions of TAMs.


Assuntos
Catepsinas/metabolismo , Interleucina-4/metabolismo , Macrófagos/enzimologia , Invasividade Neoplásica , Neoplasias/enzimologia , Animais , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Feminino , Neoplasias Pulmonares/patologia , Macrófagos/metabolismo , Neoplasias Mamárias Experimentais , Camundongos , Camundongos Transgênicos , Neoplasias/patologia , Neovascularização Patológica/enzimologia , Neovascularização Patológica/patologia , Neoplasias Pancreáticas/patologia
20.
PLoS Pathog ; 11(3): e1004704, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25738962

RESUMO

The intracellular bacterial pathogen Legionella pneumophila provokes strong host responses and has proven to be a valuable model for the discovery of novel immunosurveillance pathways. Our previous work revealed that an environmental isolate of L. pneumophila induces a noncanonical form of cell death, leading to restriction of bacterial replication in primary mouse macrophages. Here we show that such restriction also occurs in infections with wild type clinical isolates. Importantly, we found that a lysine to arginine mutation at residue 88 (K88R) in the ribosome protein RpsL that not only confers bacterial resistance to streptomycin, but more importantly, severely attenuated the induction of host cell death and enabled L. pneumophila to replicate in primary mouse macrophages. Although conferring similar resistance to streptomycin, a K43N mutation in RpsL does not allow productive intracellular bacterial replication. Further analysis indicated that RpsL is capable of effectively inducing macrophage death via a pathway involved in lysosomal membrane permeabilization; the K88R mutant elicits similar responses but is less potent. Moreover, cathepsin B, a lysosomal protease that causes cell death after being released into the cytosol upon the loss of membrane integrity, is required for efficient RpsL-induced macrophage death. Furthermore, despite the critical role of cathepsin B in delaying RpsL-induced cell death, macrophages lacking cathepsin B do not support productive intracellular replication of L. pneumophila harboring wild type RpsL. This suggests the involvement of other yet unidentified components in the restriction of bacterial replication. Our results identified RpsL as a regulator in the interactions between bacteria such as L. pneumophila and primary mouse macrophages by triggering unique cellular pathways that restrict intracellular bacterial replication.


Assuntos
Proteínas de Bactérias/imunologia , Doença dos Legionários/imunologia , Macrófagos/imunologia , Proteínas Ribossômicas/imunologia , Animais , Morte Celular , Citosol/imunologia , Citosol/metabolismo , Immunoblotting , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Legionella pneumophila , Lisossomos/imunologia , Lisossomos/metabolismo , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA