Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
FASEB Bioadv ; 6(4): 103-104, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38585430
2.
Circulation ; 149(22): 1729-1748, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38487879

RESUMO

BACKGROUND: Myocardial infarction (MI) and heart failure are associated with an increased incidence of cancer. However, the mechanism is complex and unclear. Here, we aimed to test our hypothesis that cardiac small extracellular vesicles (sEVs), particularly cardiac mesenchymal stromal cell-derived sEVs (cMSC-sEVs), contribute to the link between post-MI left ventricular dysfunction (LVD) and cancer. METHODS: We purified and characterized sEVs from post-MI hearts and cultured cMSCs. Then, we analyzed cMSC-EV cargo and proneoplastic effects on several lines of cancer cells, macrophages, and endothelial cells. Next, we modeled heterotopic and orthotopic lung and breast cancer tumors in mice with post-MI LVD. We transferred cMSC-sEVs to assess sEV biodistribution and its effect on tumor growth. Finally, we tested the effects of sEV depletion and spironolactone treatment on cMSC-EV release and tumor growth. RESULTS: Post-MI hearts, particularly cMSCs, produced more sEVs with proneoplastic cargo than nonfailing hearts did. Proteomic analysis revealed unique protein profiles and higher quantities of tumor-promoting cytokines, proteins, and microRNAs in cMSC-sEVs from post-MI hearts. The proneoplastic effects of cMSC-sEVs varied with different types of cancer, with lung and colon cancers being more affected than melanoma and breast cancer cell lines. Post-MI cMSC-sEVs also activated resting macrophages into proangiogenic and protumorigenic states in vitro. At 28-day follow-up, mice with post-MI LVD developed larger heterotopic and orthotopic lung tumors than did sham-MI mice. Adoptive transfer of cMSC-sEVs from post-MI hearts accelerated the growth of heterotopic and orthotopic lung tumors, and biodistribution analysis revealed accumulating cMSC-sEVs in tumor cells along with accelerated tumor cell proliferation. sEV depletion reduced the tumor-promoting effects of MI, and adoptive transfer of cMSC-sEVs from post-MI hearts partially restored these effects. Finally, spironolactone treatment reduced the number of cMSC-sEVs and suppressed tumor growth during post-MI LVD. CONCLUSIONS: Cardiac sEVs, specifically cMSC-sEVs from post-MI hearts, carry multiple protumorigenic factors. Uptake of cMSC-sEVs by cancer cells accelerates tumor growth. Treatment with spironolactone significantly reduces accelerated tumor growth after MI. Our results provide new insight into the mechanism connecting post-MI LVD to cancer and propose a translational option to mitigate this deadly association.


Assuntos
Vesículas Extracelulares , Insuficiência Cardíaca , Infarto do Miocárdio , Animais , Vesículas Extracelulares/metabolismo , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/etiologia , Infarto do Miocárdio/patologia , Infarto do Miocárdio/metabolismo , Camundongos , Humanos , Feminino , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Linhagem Celular Tumoral , Células-Tronco Mesenquimais/metabolismo , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças , Masculino , Proliferação de Células/efeitos dos fármacos
3.
Cancers (Basel) ; 15(23)2023 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-38067320

RESUMO

Tumor progression, from early-stage invasion to the formation of distal metastases, relies on the capacity of tumor cells to modify the extracellular matrix (ECM) and communicate with the surrounding stroma. Extracellular vesicles (EVs) provide an important means to regulate cell invasion due to the selective inclusion of cargoes such as proteases and matrix proteins into EVs that can degrade or modify the ECM. EVs have also been shown to facilitate intercellular communication in the tumor microenvironment through paracrine signaling, which can impact ECM invasion by cancer cells. Here, we describe the current knowledge of EVs as facilitators of tumor invasion by virtue of their effects on proteolytic degradation and modification of the ECM, their ability to educate the stromal cells in the tumor microenvironment, and their role as mediators of long-range communication aiding in cell invasion and matrix remodeling at secondary sites.

4.
ACS Nano ; 17(10): 9388-9404, 2023 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-37071723

RESUMO

Extracellular nanocarriers (extracellular vesicles (EVs), lipoproteins, and ribonucleoproteins) of protein and nucleic acids mediate intercellular communication and are clinically adaptable as distinct circulating biomarkers. However, the overlapping size and density of the nanocarriers have so far prevented their efficient physical fractionation, thus impeding independent downstream molecular assays. Here, we report a bias-free high-throughput and high-yield continuous isoelectric fractionation nanocarrier fractionation technique based on their distinct isoelectric points. This nanocarrier fractionation platform is enabled by a robust and tunable linear pH profile provided by water-splitting at a bipolar membrane and stabilized by flow without ampholytes. The linear pH profile that allows easy tuning is a result of rapid equilibration of the water dissociation reaction and stabilization by flow. The platform is automated with a machine learning procedure to allow recalibration for different physiological fluids and nanocarriers. The optimized technique has a resolution of 0.3 ΔpI, sufficient to separate all nanocarriers and even subclasses of nanocarriers. Its performance is then evaluated with several biofluids, including plasma, urine, and saliva samples. Comprehensive, high-purity (plasma: >93%, urine: >95% and saliva: >97%), high-yield (plasma: >78%, urine: >87% and saliva: >96%), and probe-free isolation of ribonucleoproteins in 0.75 mL samples of various biofluids in 30 min is demonstrated, significantly outperforming affinity-based and highly biased gold standards having low yield and day-long protocols. Binary fractionation of EVs and different lipoproteins is also achieved with similar performance.


Assuntos
Líquidos Corporais , Vesículas Extracelulares , Saliva/metabolismo , Ribonucleoproteínas , Líquidos Corporais/química , Vesículas Extracelulares/metabolismo , Lipoproteínas/análise , Lipoproteínas/metabolismo
5.
Annu Rev Pathol ; 18: 205-229, 2023 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-36202098

RESUMO

Tumor cells release extracellular vesicles (EVs) that can function as mediators of intercellular communication in the tumor microenvironment. EVs contain a host of bioactive cargo, including membrane, cytosolic, and nuclear proteins, in addition to noncoding RNAs, other RNA types, and double-stranded DNA fragments. These shed vesicles may deposit paracrine information and can also be taken up by stromal cells, causing the recipient cells to undergo phenotypic changes that profoundly impact diverse facets of cancer progression. For example, this unique form of cellular cross talk helps condition the premetastatic niche, facilitates evasion of the immune response, and promotes invasive and metastatic activity. These findings, coupled with those demonstrating that the number and content of EVs produced by tumors can vary depending on their tumor of origin, disease stage, or response to therapy, have raised the exciting possibility that EVs can be used for risk stratification, diagnostic, and even prognostic purposes. We summarize recent developments and the current knowledge of EV cargoes, their impact on disease progression, and implementation of EV-based liquid biopsies as tumor biomarkers.


Assuntos
Vesículas Extracelulares , Neoplasias , Humanos , Microambiente Tumoral , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/patologia , Neoplasias/patologia , Progressão da Doença
6.
iScience ; 25(8): 104653, 2022 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-35958027

RESUMO

The extracellular RNA communication consortium (ERCC) is an NIH-funded program aiming to promote the development of new technologies, resources, and knowledge about exRNAs and their carriers. After Phase 1 (2013-2018), Phase 2 of the program (ERCC2, 2019-2023) aims to fill critical gaps in knowledge and technology to enable rigorous and reproducible methods for separation and characterization of both bulk populations of exRNA carriers and single EVs. ERCC2 investigators are also developing new bioinformatic pipelines to promote data integration through the exRNA atlas database. ERCC2 has established several Working Groups (Resource Sharing, Reagent Development, Data Analysis and Coordination, Technology Development, nomenclature, and Scientific Outreach) to promote collaboration between ERCC2 members and the broader scientific community. We expect that ERCC2's current and future achievements will significantly improve our understanding of exRNA biology and the development of accurate and efficient exRNA-based diagnostic, prognostic, and theranostic biomarker assays.

8.
Cell Rep ; 38(9): 110443, 2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-35235806

RESUMO

The shedding of extracellular vesicles (EVs) represents an important but understudied means of cell-cell communication in cancer. Among the currently described classes of EVs, tumor-derived microvesicles (TMVs) comprise a class of vesicles released directly from the cell surface. TMVs contain abundant cargo, including functional proteins and miRNA, which can be transferred to and alter the behavior of recipient cells. Here, we document that a fraction of extracellular double-stranded DNA (dsDNA) is enclosed within TMVs and protected from nuclease degradation. dsDNA inclusion in TMVs is regulated by ARF6 cycling and occurs with the cytosolic DNA sensor, cGAS, but independent of amphisome or micronuclei components. Our studies suggest that dsDNA is trafficked to TMVs via a mechanism distinct from the multivesicular body-dependent secretion reported for the extracellular release of cytosolic DNA. Furthermore, TMV dsDNA can be transferred to recipient cells with consequences to recipient cell behavior, reinforcing its relevance in mediating cell-cell communication.


Assuntos
Micropartículas Derivadas de Células , Vesículas Extracelulares , MicroRNAs , Neoplasias , Micropartículas Derivadas de Células/metabolismo , DNA/metabolismo , Vesículas Extracelulares/metabolismo , Humanos , MicroRNAs/metabolismo , Neoplasias/metabolismo
9.
Exp Cell Res ; 415(1): 113106, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35307409

RESUMO

Invasive melanoma is an aggressive form of skin cancer with high incidence of mortality. The process of tumor invasion is a crucial primary step in the metastatic cascade, yet the mechanisms involved are still under investigation. Here we document a critical role for MLK3 (MAP3K11) in the regulation of melanoma cell invasion. We report the unexpected finding that cellular loss of MLK3 in melanoma cells promotes cell invasion. Cellular depletion of MLK3 expression results in the hyperactivation of ERK, which is linked to the formation of a BRAF/Hsp90/Cdc37 protein complex. ERK hyperactivation leads to enhanced phosphorylation and inactivation of GSK3ß and the stabilization of c-Jun and JNK activity. Blocking of ERK and JNK signaling as well as Hsp90 activity downstream of MLK3-silencing significantly reduces melanoma invasion. Furthermore, ERK activation in the aforementioned context is coupled to MT1-MMP transcription as well as the TOM1L1-dependent localization of the membrane protease to invadopodia at the invasive front. These studies provide critical insight into the mechanisms that couple MLK3 loss with BRAF hyperactivation and its consequence on melanoma invasion.


Assuntos
MAP Quinase Quinase Quinases , Melanoma , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Linhagem Celular Tumoral , Proteínas de Choque Térmico HSP90/metabolismo , Humanos , MAP Quinase Quinase Quinases/metabolismo , Sistema de Sinalização das MAP Quinases , Melanoma/genética , Fosforilação , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas B-raf/metabolismo
11.
12.
Front Cell Dev Biol ; 9: 746432, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34692700

RESUMO

Extracellular vesicles (EVs) are a heterogeneous population of membrane-bound parcels of bioactive proteins, nucleic acids, and lipids released from almost all cell types. The diversity of cargo packaged into EVs proffer the induction of an array of effects on recipient cells. EVs released from tumor cells have emerged as a vital means of communication and immune modulation within the tumor microenvironment (TME). Macrophages are an important contributor to the TME with seemingly paradoxical roles promoting either pro- or anti-tumoral immune function depending on their activated phenotypes. Here, we discuss the influence of tumor-derived extracellular vesicles on the functional plasticity of macrophages in tumor progression.

13.
FASEB Bioadv ; 3(6): 399-406, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-34124595

RESUMO

Microvesicles are a heterogeneous group of membrane-enclosed vesicles that are released from cells into the extracellular space by the outward budding and pinching of the plasma membrane. These vesicles are loaded with multiple selectively sorted proteins and nucleic acids. Although interest in the clinical potential of microvesicles is increasing, there is only limited understanding of different types of microvesicles and the mechanisms involved in their formation. Here, we describe what is presently known about this expanding and complex field of research focusing on the mechanism of biogenesis, cargo loading, and release of microvesicles.

14.
Dev Cell ; 55(2): 111-113, 2020 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-33108750

RESUMO

Intercellular communication is vital to tumor progression. In this issue of Developmental Cell, Bertolini et al. (2020) describe how small extracellular vesicles released from hypoxic mammary tumor cells facilitate intercellular communication, leading to alterations in mitochondrial dynamics and acquisition of invasive phenotypes in normal epithelial cells.


Assuntos
Vesículas Extracelulares , Microambiente Tumoral , Comunicação Celular , Humanos , Dinâmica Mitocondrial , Oxigênio
15.
Adv Exp Med Biol ; 1259: 155-170, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32578176

RESUMO

Extracellular vesicle (EV) shedding is a biologically conserved cellular process across virtually every cell type. In cancer, EVs shed from tumor and stromal cells to the tumor microenvironment play a major role in determining tumor fate, which to a large extent is dictated by the biologically active cargo contained in EVs. Current understanding of various cancer-associated EVs has enabled the outlining of mechanistic connections between cargo and tumor-promoting functions. In this chapter, we describe examples of EV-mediated communication between tumor cells and stromal cells, highlighting the molecular constituents responsible for pro-tumorigenic effects. Furthermore, we discuss the roles of matrix-degrading EVs in cell invasion. Finally, we summarize research on the potential use of EVs as a novel approach to cancer therapeutics.


Assuntos
Vesículas Extracelulares , Neoplasias/patologia , Microambiente Tumoral , Humanos , Neoplasias/terapia , Células Estromais
16.
Nat Commun ; 11(1): 3017, 2020 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-32541798

RESUMO

Breast cancer brain metastases (BCBM) have a 5-20 year latency and account for 30% of mortality; however, mechanisms governing adaptation to the brain microenvironment remain poorly defined. We combine time-course RNA-sequencing of BCBM development with a Drosophila melanogaster genetic screen, and identify Rab11b as a functional mediator of metastatic adaptation. Proteomic analysis reveals that Rab11b controls the cell surface proteome, recycling proteins required for successful interaction with the microenvironment, including integrin ß1. Rab11b-mediated control of integrin ß1 surface expression allows efficient engagement with the brain ECM, activating mechanotransduction signaling to promote survival. Lipophilic statins prevent membrane association and activity of Rab11b, and we provide proof-of principle that these drugs prevent breast cancer adaptation to the brain microenvironment. Our results identify Rab11b-mediated recycling of integrin ß1 as regulating BCBM, and suggest that the recycleome, recycling-based control of the cell surface proteome, is a previously unknown driver of metastatic adaptation and outgrowth.


Assuntos
Neoplasias Encefálicas/metabolismo , Neoplasias da Mama/patologia , Integrina beta1/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/fisiopatologia , Neoplasias Encefálicas/secundário , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Proliferação de Células , Modelos Animais de Doenças , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Feminino , Humanos , Integrina beta1/genética , Camundongos , Camundongos Endogâmicos C57BL , Metástase Neoplásica , Transporte Proteico , Transdução de Sinais , Microambiente Tumoral , Proteínas rab de Ligação ao GTP/genética
17.
J Cell Sci ; 132(20)2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-31615844

RESUMO

Extracellular vesicles (EVs) are a heterogeneous collection of membrane-bound vesicles released by cells that contain bioactive cargoes including proteins, lipids and nucleic acids. Multiple subpopulations of EVs have now been recognized and these include exosomes and microvesicles. EVs have been thought to facilitate intercellular and distal communication to bring about various processes that enable tumor progression and metastases. Here, we describe the current knowledge of the functional cargo contained within EVs, with a focus on tumor microvesicles, and review the emerging theory of how EVs support immune suppression in cancer.


Assuntos
Comunicação Celular/imunologia , Micropartículas Derivadas de Células/imunologia , Vesículas Extracelulares/imunologia , Neoplasias/imunologia , Animais , Micropartículas Derivadas de Células/patologia , Vesículas Extracelulares/patologia , Humanos , Metástase Neoplásica , Neoplasias/patologia
18.
Nat Cell Biol ; 21(7): 856-866, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31235936

RESUMO

Tumour-derived microvesicles (TMVs) comprise a class of extracellular vesicles released from tumour cells that are now understood to facilitate communication between the tumour and the surrounding microenvironment. Despite their significance, the regulatory mechanisms governing the trafficking of bioactive cargos to TMVs at the cell surface remain poorly defined. Here we describe a molecular pathway for the delivery of microRNA (miRNA) cargo to nascent TMVs involving the dissociation of a pre-miRNA/Exportin-5 complex from Ran-GTP following nuclear export and its subsequent transfer to a cytoplasmic shuttle comprised of ARF6-GTP and GRP1. As such, ARF6 activation increases the pre-miRNA cargo contained within TMVs through a process that requires the casein kinase 2-mediated phosphorylation of RanGAP1. Furthermore, TMVs were found to contain pre-miRNA processing machinery including Dicer and Argonaute-2, which allow for cell-free pre-miRNA processing within shed vesicles. These findings offer cellular targets to block the loading and processing of pre-miRNAs within TMVs.


Assuntos
Fatores de Ribosilação do ADP/metabolismo , Carioferinas/metabolismo , MicroRNAs/genética , Microambiente Tumoral/fisiologia , Fator 6 de Ribosilação do ADP , Fatores de Ribosilação do ADP/genética , Transporte Ativo do Núcleo Celular/fisiologia , Micropartículas Derivadas de Células/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Humanos , Carioferinas/genética , Neoplasias/metabolismo , Fosforilação , Interferência de RNA/fisiologia
19.
Clin Cancer Res ; 25(9): 2848-2859, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30718355

RESUMO

PURPOSE: Genetic ancestry influences evolutionary pathways of cancers. However, whether ancestry influences cancer-induced field defects is unknown. The goal of this study was to utilize ancestry-mapped true normal breast tissues as controls to identify cancer-induced field defects in normal tissue adjacent to breast tumors (NATs) in women of African American (AA) and European (EA) ancestry. EXPERIMENTAL DESIGN: A tissue microarray comprising breast tissues of ancestry-mapped 100 age-matched healthy women from the Komen Tissue Bank (KTB) at Indiana University (Indianapolis, IN) and tumor-NAT pairs from 100 women (300 samples total) was analyzed for the levels of ZEB1, an oncogenic transcription factor that is central to cell fate, mature luminal cell-enriched estrogen receptor alpha (ERα), GATA3, FOXA1, and for immune cell composition. RESULTS: ZEB1+ cells, which were localized surrounding the ductal structures of the normal breast, were enriched in the KTB-normal of AA compared with KTB-normal of EA women. In contrast, in EA women, both NATs and tumors compared with KTB-normal contained higher levels of ZEB1+ cells. FOXA1 levels were lower in NATs compared with KTB-normal in AA but not in EA women. We also noted variations in the levels of GATA3, CD8+ T cells, PD1+ immune cells, and PDL1+ cell but not CD68+ macrophages in NATs of AA and EA women. ERα levels did not change in any of our analyses, pointing to the specificity of ancestry-dependent variations. CONCLUSIONS: Genetic ancestry-mapped tissues from healthy individuals are required for proper assessment and development of cancer-induced field defects as early cancer detection markers. This finding is significant in light of recent discoveries of influence of genetic ancestry on both normal biology and tumor evolution.


Assuntos
Biomarcadores Tumorais/metabolismo , População Negra/estatística & dados numéricos , Neoplasias da Mama/patologia , Mama/patologia , Predisposição Genética para Doença , Células-Tronco Neoplásicas/patologia , População Branca/estatística & dados numéricos , Antígeno B7-H1/metabolismo , População Negra/genética , Mama/metabolismo , Neoplasias da Mama/etnologia , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Receptor alfa de Estrogênio/metabolismo , Feminino , Fator de Transcrição GATA3/metabolismo , Fator 3-alfa Nuclear de Hepatócito/metabolismo , Humanos , Células-Tronco Neoplásicas/metabolismo , Prognóstico , Receptor de Morte Celular Programada 1/metabolismo , Análise Serial de Tecidos , População Branca/genética , Homeobox 1 de Ligação a E-box em Dedo de Zinco/metabolismo
20.
Mol Cell Biol ; 39(3)2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30397076

RESUMO

Tumor cell invasion is one result of the bidirectional interactions occurring between tumor cells and the surrounding milieu. The ability of tumor cells to invade through the extracellular matrix is in part regulated by the formation of a class of protease-loaded extracellular vesicles, called tumor microvesicles (TMVs), which are released directly from the cell surface. Here we show that the actin bundling protein, fascin, redistributes to the cell periphery in a ternary complex with podocalyxin and ezrin, where it promotes TMV release. The peripheral localization of fascin is prompted by the loss of Rab35 signaling, which in turn unleashes ARF6 activation. The result is a mechanism through which Rab35 and ARF6 cooperatively and simultaneously regulate the distribution and localization of fascin and promote oncogenic signaling, which leads to TMV release while inhibiting invadopodium formation. These studies are clinically significant as fascin-loaded TMVs can be detected in bodily fluids and elevated fascin expression coupled with low Rab35 levels correlates with poor overall survival in some cancers.


Assuntos
Fatores de Ribosilação do ADP/metabolismo , Proteínas de Transporte/metabolismo , Proteínas dos Microfilamentos/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Fator 6 de Ribosilação do ADP , Actinas/metabolismo , Proteínas de Transporte/fisiologia , Linhagem Celular , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Micropartículas Derivadas de Células/metabolismo , Citoplasma/metabolismo , Proteínas do Citoesqueleto/metabolismo , Matriz Extracelular/metabolismo , Humanos , Proteínas dos Microfilamentos/fisiologia , Invasividade Neoplásica/patologia , Células PC-3 , Sialoglicoproteínas/metabolismo , Transdução de Sinais , Microambiente Tumoral/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA