Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
2.
Mol Cancer Res ; 20(6): 841-853, 2022 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-35302608

RESUMO

Inhibiting androgen signaling using androgen signaling inhibitors (ASI) remains the primary treatment for castrate-resistant prostate cancer. Acquired resistance to androgen receptor (AR)-targeted therapy represents a major impediment to durable clinical response. Understanding resistance mechanisms, including the role of AR expressed in other cell types within the tumor microenvironment, will extend the clinical benefit of AR-targeted therapy. Here, we show the ASI enzalutamide induces vascular catastrophe and promotes hypoxia and microenvironment adaptation. We characterize treatment-induced hypoxia, and subsequent induction of angiogenesis, as novel mechanisms of relapse to enzalutamide, highlighting the importance of two hypoxia-regulated cytokines in underpinning relapse. We confirmed AR expression in CD34+ vascular endothelium of biopsy tissue and human vascular endothelial cells (HVEC). Enzalutamide attenuated angiogenic tubule formation and induced cytotoxicity in HVECs in vitro, and rapidly induced sustained hypoxia in LNCaP xenografts. Subsequent reoxygenation, following prolonged enzalutamide treatment, was associated with increased tumor vessel density and accelerated tumor growth. Hypoxia increased AR expression and transcriptional activity in prostate cells in vitro. Coinhibition of IL8 and VEGF-A restored tumor response in the presence of enzalutamide, confirming the functional importance of their elevated expression in enzalutamide-resistant models. Moreover, coinhibition of IL8 and VEGF-A resulted in a durable, effective resolution of enzalutamide-sensitive prostate tumors. We conclude that concurrent inhibition of two hypoxia-induced factors, IL8 and VEGF-A, prolongs tumor sensitivity to enzalutamide in preclinical models and may delay the onset of enzalutamide resistance. IMPLICATIONS: Targeting hypoxia-induced signaling may extend the therapeutic benefit of enzalutamide, providing an improved treatment strategy for patients with resistant disease.


Assuntos
Antagonistas de Receptores de Andrógenos , Neoplasias de Próstata Resistentes à Castração , Antagonistas de Androgênios/farmacologia , Antagonistas de Receptores de Andrógenos/farmacologia , Androgênios/farmacologia , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Células Endoteliais/metabolismo , Humanos , Hipóxia/tratamento farmacológico , Interleucina-8/genética , Masculino , Recidiva Local de Neoplasia/tratamento farmacológico , Nitrilas/farmacologia , Neoplasias de Próstata Resistentes à Castração/patologia , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Microambiente Tumoral , Fator A de Crescimento do Endotélio Vascular/genética
3.
iScience ; 23(12): 101799, 2020 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-33299970

RESUMO

Functional roles of neutrophil elastase (NE) have not been examined in distinct steps of the metastatic cascade. NE, delivered to primary tumors as a purified enzyme or within intact neutrophils or neutrophil granule content, enhanced human tumor cell intravasation and subsequent dissemination via NE-mediated formation of dilated intratumoral vasculature. These effects depended on picomole range of NE activity, sensitive to its natural inhibitor, α1PI. In Elane-negative mice, the lack of NE decreased lung retention of human tumor cells in experimental metastasis. Furthermore, NE was essential for spontaneous metastasis of murine carcinoma cells in a syngeneic orthotopic model of oral cancer. NE also induced tumor cell survival and migration via Src/PI3K-dependent activation of Akt signaling, vital for tumor cell dissemination in vivo. Together, our findings implicate NE, a potent host enzyme specific for first-responding innate immune cells, as directly involved in early metastatic events and a potential target for therapeutic intervention.

4.
Theranostics ; 10(9): 4116-4133, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32226543

RESUMO

Background: CUB domain-containing protein 1 (CDCP1) is a cell surface receptor regulating key signalling pathways in malignant cells. CDCP1 has been proposed as a molecular target to abrogate oncogenic signalling pathways and specifically deliver anti-cancer agents to tumors. However, the development of CDCP1-targeting agents has been questioned by its frequent proteolytic processing which was thought to result in shedding of the CDCP1 extracellular domain limiting its targetability. In this study, we investigated the relevance of targeting CDCP1 in the context of pancreatic ductal adenocarcinoma (PDAC) and assess the impact of CDCP1 proteolysis on the effectiveness of CDCP1 targeting agents. Methods: The involvement of CDCP1 in PDAC progression was assessed by association analysis in several PDAC cohorts and the proteolytic processing of CDCP1 was evaluated in PDAC cell lines and patient-derived cells. The consequences of CDCP1 proteolysis on its targetability in PDAC cells was assessed using immunoprecipitation, immunostaining and biochemical assays. The involvement of CDCP1 in PDAC progression was examined by loss-of-function in vitro and in vivo experiments employing PDAC cells expressing intact or cleaved CDCP1. Finally, we generated antibody-based imaging and therapeutic agents targeting CDCP1 to demonstrate the feasibility of targeting this receptor for detection and treatment of PDAC tumors. Results: High CDCP1 expression in PDAC is significantly associated with poorer patient survival. In PDAC cells proteolysis of CDCP1 does not always result in the shedding of CDCP1-extracellular domain which can interact with membrane-bound CDCP1 allowing signal transduction between the different CDCP1-fragments. Targeting CDCP1 impairs PDAC cell functions and PDAC tumor growth independently of CDCP1 cleavage status. A CDCP1-targeting antibody is highly effective at delivering imaging radionuclides and cytotoxins to PDAC cells allowing specific detection of tumors by PET/CT imaging and superior anti-tumor effects compared to gemcitabine in in vivo models. Conclusion: Independent of its cleavage status, CDCP1 exerts oncogenic functions in PDAC and has significant potential to be targeted for improved radiological staging and treatment of this cancer. Its elevated expression by most PDAC tumors and lack of expression by normal pancreas and other major organs, suggest that targeting CDCP1 could benefit a significant proportion of PDAC patients. These data support the further development of CDCP1-targeting agents as personalizable tools for effective imaging and treatment of PDAC.


Assuntos
Antígenos de Neoplasias/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Moléculas de Adesão Celular/metabolismo , Neoplasias Pancreáticas/metabolismo , Animais , Biomarcadores Tumorais/metabolismo , Carcinoma Ductal Pancreático/terapia , Linhagem Celular Tumoral , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Neoplasias Pancreáticas/terapia , Medicina de Precisão , Proteólise
5.
Oncogene ; 37(14): 1815-1829, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29348457

RESUMO

Latent transforming growth factor ß (TGFß)-binding proteins (LTBPs) are important for the secretion, activation, and function of mature TGFß, especially so in cancer cell physiology. However, specific roles of the LTBPs remain understudied in the context of the primary tumor microenvironment. Herein, we investigated the role of LTBP3 in the distinct processes involved in cancer metastasis. By using three human tumor cell lines of different tissue origin (epidermoid HEp-3 and prostate PC-3 carcinomas and HT-1080 fibrosarcoma) and several metastasis models conducted in both mammalian and avian settings, we show that LTBP3 is involved in the early dissemination of primary cancer cells, namely in the intravasation step of the metastatic cascade. Knockdown of LTBP3 in all tested cell lines led to significant inhibition of tumor cell intravasation, but did not affect primary tumor growth. LTBP3 was dispensable in the late steps of carcinoma cell metastasis that follow tumor cell intravasation, including vascular arrest, extravasation, and tissue colonization. However, LTBP3 depletion diminished the angiogenesis-inducing potential of HEp-3 cells in vivo, which was restorable by exogenous delivery of LTBP3 protein. A similar compensatory approach rescued the dampened intravasation of LTBP3-deficient HEp-3 cells, suggesting that LTBP3 regulates the induction of the intravasation-supporting angiogenic vasculature within developing primary tumors. Using our recently developed microtumor model, we confirmed that LTBP3 loss resulted in the development of intratumoral vessels with an abnormal microarchitecture incompatible with efficient intravasation of HEp-3 carcinoma cells. Collectively, these findings demonstrate that LTBP3 represents a novel oncotarget that has distinctive functions in the regulation of angiogenesis-dependent tumor cell intravasation, a critical process during early cancer dissemination. Our experimental data are also consistent with the survival prognostic value of LTBP3 expression in early-stage head and neck squamous cell carcinomas, further indicating a specific role for LTBP3 in cancer progression toward metastatic disease.


Assuntos
Proteínas de Ligação a TGF-beta Latente/fisiologia , Neoplasias/genética , Neoplasias/patologia , Animais , Linhagem Celular Tumoral , Embrião de Galinha , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas de Ligação a TGF-beta Latente/antagonistas & inibidores , Proteínas de Ligação a TGF-beta Latente/genética , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Invasividade Neoplásica , Metástase Neoplásica , Neoplasias/mortalidade , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , RNA Interferente Pequeno/farmacologia , Análise de Sobrevida
6.
Thromb Res ; 140 Suppl 1: S27-36, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27067975

RESUMO

Metastasis is the main cause of death in cancer patients, and understanding mechanisms that control tumor cell dissemination may lead to improved therapy. Tumor cell adhesion receptors contribute to cancer spreading. We noted earlier that tumor cells can expressing the adhesion receptor integrin αvß3 in distinct states of activation, and found that cells which metastasize from the blood stream express it in a constitutively high affinity form. Here, we analyzed steps of the metastatic cascade in vivo and asked, when and how the affinity state of integrin αvß3 confers a critical advantage to cancer spreading. Following tumor cells by real time PCR, non-invasive bioluminescence imaging, intravital microscopy and histology allowed us to identify tumor cell extravasation from the blood stream as a rate-limiting step supported by high affinity αvß3. Successful transendothelial migration depended on cooperation between tumor cells and platelets involving the high affinity tumor cell integrin and release of platelet granules. Thus, this study identifies the high affinity conformer of integrin αvß3 and its interaction with platelets as critical for early steps during hematogenous metastasis and target for prevention of metastatic disease.


Assuntos
Plaquetas/patologia , Integrina alfaVbeta3/metabolismo , Metástase Neoplásica/patologia , Células Neoplásicas Circulantes/patologia , Animais , Plaquetas/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Humanos , Integrina alfaVbeta3/análise , Camundongos SCID , Células Neoplásicas Circulantes/metabolismo
7.
Vesalius ; 21(1): 10-8, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26592079

RESUMO

The untimely death of a young Princess of Wales reverberated around the world in August 1997. Diana, Princess of Wales, was not, however, the first holder of that title to suffer an early demise. Princess Charlotte of Wales was fifteen years younger and died exactly one hundred and eighty years earlier. A national feeling of grief and desolation consumed the nation in the same way as it did following the death of the "People's Princess" in the twentieth century. Her death during childbirth led to a change in the practice of obstetrics and a succession crisis in the British monarchy. But were the consequences of the fateful night of 6th November 1817 to be even more profound, indirectly contributing to war in Europe a century later?


Assuntos
Parto Obstétrico/história , Pessoas Famosas , Obstetrícia/história , Parto , Parto Obstétrico/mortalidade , Europa (Continente) , História do Século XIX , História do Século XX , Reino Unido
8.
Neoplasia ; 17(8): 634-49, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26408256

RESUMO

Many malignant characteristics of cancer cells are regulated through pathways induced by the tyrosine kinase activity of the epidermal growth factor receptor (EGFR). Herein, we show that besides directly affecting the biology of cancer cells per se, EGFR also regulates the primary tumor microenvironment. Specifically, our findings demonstrate that both the expression and signaling activity of EGFR are required for the induction of a distinct intratumoral vasculature capable of sustaining tumor cell intravasation, a critical rate-limiting step in the metastatic cascade. An intravasation-sustaining mode of intratumoral angiogenic vessels depends on high levels of tumor cell EGFR and the interplay between EGFR-regulated production of interleukin 8 by tumor cells, interleukin-8-induced influx of tumor-infiltrating neutrophils delivering their unique matrix metalloproteinase-9, and neutrophil matrix metalloproteinase-9-dependent release of the vascular permeability and endothelial growth factor, VEGF. Our data indicate that through VEGF-mediated disruption of endothelial layer integrity and increase of intratumoral vasculature permeability, EGFR activity significantly facilitates active intravasation of cancer cells. Therefore, this study unraveled an important but overlooked function of EGFR in cancer, namely, its ability to create an intravasation-sustaining microenvironment within the developing primary tumor by orchestrating several interrelated processes required for the initial steps of cancer metastasis through vascular routes. Our findings also suggest that EGFR-targeted therapies might be more effective when implemented in cancer patients with early-staged primary tumors containing a VEGF-dependent angiogenic vasculature. Accordingly, early EGFR inhibition combined with various anti-VEGF approaches could synergistically suppress tumor cell intravasation through inhibiting the highly permeable angiogenic vasculature induced by EGFR-overexpressing aggressive cancer cells.


Assuntos
Membrana Corioalantoide/irrigação sanguínea , Receptores ErbB/metabolismo , Neoplasias/metabolismo , Neovascularização Patológica/metabolismo , Animais , Western Blotting , Linhagem Celular Tumoral , Embrião de Galinha , Membrana Corioalantoide/metabolismo , Membrana Corioalantoide/patologia , Receptores ErbB/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Metaloproteinase 9 da Matriz/metabolismo , Microscopia de Fluorescência , Invasividade Neoplásica , Metástase Neoplásica , Transplante de Neoplasias , Neoplasias/irrigação sanguínea , Neoplasias/genética , Neovascularização Patológica/genética , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Microambiente Tumoral/genética , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
9.
Matrix Biol ; 44-46: 94-112, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25912949

RESUMO

Metastasis is a distinct stage of cancer progression that requires the development of angiogenic blood vessels serving as conduits for tumor cell dissemination. An accumulated body of evidence indicates that metastasis-supporting neovasculature should possess certain structural characteristics allowing for the process of tumor cell intravasation, an active entry of cancer cells into the vessel interior. It appears that the development of tumor vessels with lumens of a distinctive size and support of these vessels by a discontinuous pericyte coverage constitute critical microarchitectural requirements to: (a) provide accessible points for vessel wall penetration by primary tumor cells; (b) provide enough lumen space for a tumor cell or cell aggregate upon intravasation; and (c) allow for sufficient rate of blood flow to carry away intravasated cells from the primary tumor to the next, proximal or distal site. This review will primarily focus on the functional roles of matrix metalloproteinases (MMPs), which catalytically trigger the development of an intravasation-sustaining neovasculature at the early stages of tumor growth and are also required for the maintenance of a metastasis-supporting state of blood vessels at later stages of cancer progression.


Assuntos
Metaloproteinases da Matriz/metabolismo , Neoplasias/patologia , Neovascularização Patológica/enzimologia , Animais , Humanos , Metástase Neoplásica , Neoplasias/irrigação sanguínea , Neoplasias/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
10.
Biochem J ; 465(2): 259-70, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25360794

RESUMO

Gelatinase B/matrix metalloproteinase-9 (MMP-9) (EC 3.4.24.35) cleaves many substrates and is produced by most cell types as a zymogen, proMMP-9, in complex with the tissue inhibitor of metalloproteinases-1 (TIMP-1). Natural proMMP-9 occurs as monomers, homomultimers and heterocomplexes, but our knowledge about the overall structure of proMMP-9 monomers and multimers is limited. We investigated biochemical, biophysical and functional characteristics of zymogen and activated forms of MMP-9 monomers and multimers. In contrast with a conventional notion of a dimeric nature of MMP-9 homomultimers, we demonstrate that these are reduction-sensitive trimers. Based on the information from electrophoresis, AFM and TEM, we generated a 3D structure model of the proMMP-9 trimer. Remarkably, the proMMP-9 trimers possessed a 50-fold higher affinity for TIMP-1 than the monomers. In vivo, this finding was reflected in a higher extent of TIMP-1 inhibition of angiogenesis induced by trimers compared with monomers. Our results show that proMMP-9 trimers constitute a novel structural and functional entity that is differentially regulated by TIMP-1.


Assuntos
Precursores Enzimáticos/química , Metaloproteinase 9 da Matriz/química , Modelos Moleculares , Complexos Multiproteicos/química , Inibidor Tecidual de Metaloproteinase-1/química , Precursores Enzimáticos/genética , Precursores Enzimáticos/metabolismo , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Microscopia de Força Atômica , Microscopia Eletrônica de Transmissão , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Complexos Multiproteicos/ultraestrutura , Inibidor Tecidual de Metaloproteinase-1/genética , Inibidor Tecidual de Metaloproteinase-1/metabolismo
11.
Neoplasia ; 16(10): 771-88, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25379015

RESUMO

According to established notion, one of the major angiogenesis-inducing factors, pro-matrix metalloproteinase-9 (proMMP-9), is supplied to the tumor microenvironment by tumor-associated macrophages (TAMs). Accumulated evidence, however, indicates that tumor-associated neutrophils (TANs) are also critically important for proMMP-9 delivery, especially at early stages of tumor development. To clarify how much angiogenic proMMP-9 is actually contributed by TAMs and TANs, we quantitatively evaluated TAMs and TANs from different tumor types, including human xenografts and syngeneic murine tumors grown in wild-type and Mmp9-knockout mice. Whereas host MMP-9 competence was required for full angiogenic potential of both normal and tumor-associated leukocytes, direct comparisons of neutrophils versus macrophages and TANs versus TAMs demonstrated that macrophages and TAMs secrete 40- to 50-fold less proMMP-9 than the same numbers of neutrophils or TANs. Correspondingly, the levels of MMP-9-mediated in vivo angiogenesis induced by neutrophils and TANs substantially exceeded those induced by macrophages and TAMs. MMP-9-delivering TANs were also required for development of metastasis-supporting intratumoral vasculature, characterized by ≥ 11-µm size lumens and partial coverage with stabilizing pericytes. Importantly, MMP-9-producing TAMs exhibit M2-skewed phenotype but do not express tissue inhibitor of metalloproteinases-1 (TIMP-1), a novel characteristic allowing them to secrete TIMP-1-free, neutrophil-like MMP-9 zymogen unencumbered by its natural inhibitor. Together, our findings support the notion whereby TANs, capable of immediate release of their pre-stored cargo, are the major contributors of highly angiogenic MMP-9, whereas tumor-influxing precursors of macrophages require time to differentiate, polarize into M2-skewed TAMs, shut down their TIMP-1 expression, and only then, initiate relatively low-level production of TIMP-free MMP-9 zymogen.


Assuntos
Indutores da Angiogênese/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Neutrófilos/patologia , Microambiente Tumoral/fisiologia , Animais , Linhagem Celular Tumoral , Humanos , Macrófagos/enzimologia , Macrófagos/patologia , Masculino , Metaloproteinase 9 da Matriz/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neovascularização Patológica , Neutrófilos/metabolismo , Neoplasias da Próstata/irrigação sanguínea , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/patologia , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
12.
BMJ Case Rep ; 20142014 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-25323281

RESUMO

We describe a rare but sinister presentation of fatigue and dyspnoea in a 39-year-old woman at 16 weeks gestation. Blood tests and bone marrow aspirate confirmed the diagnosis of multiple myeloma. The patient was managed expectantly during pregnancy with plasma exchange and blood transfusion. The pregnancy continued without event; labour was induced at 35 weeks gestation and a healthy female infant weighing 3100 g was delivered vaginally following a 2 h, 5 min labour. The patient subsequently underwent six cycles of cyclophosphamide, thalidomide and dexamethasone (CTD) chemotherapy followed by an autologous stem cell transplant (SCT) and reduced intensity conditioning matched unrelated donor (RIC MUD) transplant.


Assuntos
Dispneia/etiologia , Fadiga/etiologia , Mieloma Múltiplo/diagnóstico , Complicações Neoplásicas na Gravidez/diagnóstico , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Feminino , Transplante de Células-Tronco Hematopoéticas , Humanos , Mieloma Múltiplo/terapia , Gravidez , Complicações Neoplásicas na Gravidez/terapia , Transplante Autólogo
13.
Blood ; 122(25): 4054-67, 2013 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-24174628

RESUMO

A proangiogenic function of tissue-infiltrating monocytes/macrophages has long been attributed to their matrix metalloproteinase-9 zymogen (proMMP-9). Herein, we evaluated the capacity of human monocytes, mature M0 macrophages, and M1- and M2-polarized macrophages to induce proMMP-9-mediated angiogenesis. Only M2 macrophages induced angiogenesis at levels comparable with highly angiogenic neutrophils previously shown to release their proMMP-9 in a unique form, free of tissue inhibitor of metalloproteinases-1 (TIMP-1). Macrophage differentiation was accompanied by induction of low-angiogenic, TIMP-1-encumbered proMMP-9. However, polarization toward the M2, but not the M1 phenotype, caused a substantial downregulation of TIMP-1 expression, resulting in production of angiogenic, TIMP-deficient proMMP-9. Correspondingly, the angiogenic potency of M2 proMMP-9 was lost after its complexing with TIMP-1, whereas TIMP-1 silencing in M0/M1 macrophages rendered them both angiogenic. Similar to human cells, murine bone marrow-derived M2 macrophages also shut down their TIMP-1 expression and produced proMMP-9 unencumbered by TIMP-1. Providing proof that angiogenic capacity of murine M2 macrophages depended on their TIMP-free proMMP-9, Mmp9-null M2 macrophages were nonangiogenic, although their TIMP-1 was severely downregulated. Our study provides a unifying molecular mechanism for high angiogenic capacity of TIMP-free proMMP-9 that would be uniquely produced in a pathophysiological microenvironment by influxing neutrophils and/or M2 polarized macrophages.


Assuntos
Diferenciação Celular/fisiologia , Precursores Enzimáticos/metabolismo , Macrófagos/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Neovascularização Fisiológica/fisiologia , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Animais , Embrião de Galinha , Regulação para Baixo/fisiologia , Precursores Enzimáticos/genética , Humanos , Macrófagos/citologia , Metaloproteinase 9 da Matriz/genética , Camundongos , Camundongos Mutantes , Neutrófilos/citologia , Neutrófilos/metabolismo , Inibidor Tecidual de Metaloproteinase-1/genética
14.
Cancer Res ; 73(14): 4196-211, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23687338

RESUMO

Intravasation, the active entry of primary tumor cells into the vasculature, remains the least studied step in the metastatic cascade. Protease-mediated escape and stromal invasion of tumor cells represent widely accepted processes leading up to the intravasation step. However, molecular factors that contribute directly to tumor cell vascular penetration have not been identified. In this study, the in vivo role of the collagenolytic protease, MMP-1, in cancer cell intravasation and metastasis was analyzed by using a highly disseminating variant of human HEp3 epidermoid carcinoma, HEp3-hi/diss. Although naturally acquired or experimentally induced MMP-1 deficiency substantially suppressed HEp3-hi/diss intravasation, supplementation of recombinant MMP-1 to MMP-1-silenced primary tumors restored their impaired vascular dissemination. Surprisingly, abrogation of MMP-1 production and activity did not significantly affect HEp3-hi/diss migration or matrix invasion, suggesting noncollagenolytic mechanisms underlying MMP-1-dependent cell intravasation. In support of such noncollagenolytic mechanisms, MMP-1 silencing in HEp3-hi/diss cells modulated the microarchitecture and integrity of the angiogenic vasculature in a novel microtumor model. Concomitantly, MMP-1 deficiency led to decreased levels of intratumoral vascular permeability, tumor cell intravasation, and metastatic dissemination. Taking advantage of PAR1 deficiency of HEp3-hi/diss cells, we further show that endothelial PAR1 is a putative nontumor-cell/nonmatrix target, activation of which by carcinoma-produced MMP-1 regulates endothelial permeability and transendothelial migration. The inhibitory effects of specific PAR1 antagonists in live animals have also indicated that the mechanisms of MMP-1-dependent vascular permeability in tumors involve endothelial PAR1 activation. Together, our findings mechanistically underscore the contribution of a tumor MMP-1/endothelial PAR1 axis to actual intravasation events manifested by aggressive carcinoma cells.


Assuntos
Carcinoma de Células Escamosas/enzimologia , Metaloproteinase 1 da Matriz/metabolismo , Receptor PAR-1/metabolismo , Animais , Permeabilidade Capilar , Carcinoma de Células Escamosas/irrigação sanguínea , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Embrião de Galinha , Endotélio/irrigação sanguínea , Endotélio/enzimologia , Endotélio/metabolismo , Endotélio/patologia , Humanos , Metaloproteinase 1 da Matriz/deficiência , Metaloproteinase 1 da Matriz/genética , Invasividade Neoplásica , Metástase Neoplásica , Neovascularização Patológica/enzimologia , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Receptor PAR-1/deficiência , Receptor PAR-1/genética
15.
Cancer Res ; 73(2): 662-71, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23329645

RESUMO

The transcription factor Twist1 induces epithelial-mesenchymal transition and extracellular matrix degradation to promote tumor metastasis. Although Twist1 also plays a role in embryonic vascular development and tumor angiogenesis, the molecular mechanisms that underlie these processes are not as well understood. Here, we report a novel function for Twist1 in modifying the tumor microenvironment to promote progression. We found that expression of Twist1 in human mammary epithelial cells potently promoted angiogenesis. Surprisingly, Twist1 expression did not increase the secretion of the common proangiogenic factors VEGF and basic fibroblast growth factor but rather induced expression of the macrophage chemoattractant CCL2. Attenuation of endogenous Twist1 in vivo blocked macrophage recruitment and angiogenesis, whereas exogenous CCL2 rescued the ability of tumor cells lacking Twist1 to attract macrophages and promote angiogenesis. Macrophage recruitment also was essential for the ability of Twist1-expressing cells to elicit a strong angiogenic response. Together, our findings show that how Twist1 recruits stromal macrophages through CCL2 induction to promote angiogenesis and tumor progression. As Twist1 expression has been associated with poor survival in many human cancers, this finding suggests that anti-CCL2 therapy may offer a rational strategy to treat Twist1-positive metastatic cancers.


Assuntos
Neoplasias da Mama/irrigação sanguínea , Quimiocina CCL2/metabolismo , Macrófagos/fisiologia , Neovascularização Patológica , Proteínas Nucleares/fisiologia , Proteína 1 Relacionada a Twist/fisiologia , Animais , Linhagem Celular Tumoral , Progressão da Doença , Feminino , Humanos , Neoplasias Mamárias Experimentais/irrigação sanguínea , Camundongos
16.
Vesalius ; 19(2): 72-7, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26035929

RESUMO

The name "Ioseb Besarionis dze Dzhugashvili" is as unassuming as it is unknown. It is the birth name of the brutal dictator who changed the face of Europe and whose actions still influence our lives today. Stalin, the man responsible for the slaughter of over twenty million of his own Soviet citizens and yet, the man who transformed the USSR from the feudalistic society of the Tsar's to a twentieth century military powerhouse that was instrumental in the defeat of Nazi Germany. Known as "Koba" to his friends, he cultivated a cult of personality where he was, and to a certain extent still is, admired in Russia and the former Soviet states. This paper will look at the following questions: why when he fell gravely ill did his comrades wait so long before seeking medical assistance? why were there omissions in the final post mortem report?, and why did one his closest lieutenants boast so openly about having murdered him.


Assuntos
Hemorragia Cerebral/história , Pessoas Famosas , Intoxicação/história , Acidente Vascular Cerebral/história , Transtorno da Personalidade Antissocial/história , Autopsia/história , História do Século XX , Humanos , Masculino , U.R.S.S.
17.
BMJ Case Rep ; 20122012 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-23239771

RESUMO

Ascaris lumbricoides is one of the most prevalent parasitic infections, especially in developing countries. Its presence can lead to a multitude of presentations, one of the rarer ones being obstructive jaundice due to migration of the worm in to the biliary tree. We describe a case of a man who presented as an emergency to the general surgeons complaining of abdominal pain, fever, jaundice and vomiting. Ultrasound was used and the diagnosis of biliary ascariasis was made. The patient underwent surgery consisting of a cholecystectomy, common bile duct exploration and T-tube choledochostomy. Our report highlights the varied aetiology of obstructive jaundice and the importance of including biliary ascariasis in the differential diagnosis of the jaundiced patient, especially from endemic areas.


Assuntos
Ascaríase/complicações , Doenças dos Ductos Biliares/complicações , Doenças dos Ductos Biliares/parasitologia , Icterícia Obstrutiva/parasitologia , Adulto , Animais , Ascaris lumbricoides , Humanos , Masculino
18.
Mol Cancer Res ; 10(12): 1532-43, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23038812

RESUMO

Data accumulated over the latest two decades have established that the serine protease urokinase-type plasminogen activator (uPA) is a potential therapeutic target in cancer. When designing inhibitors of the proteolytic activity of serine proteases, obtaining sufficient specificity is problematic, because the topology of the proteases' active sites are highly similar. In an effort to generate highly specific uPA inhibitors with new inhibitory modalities, we isolated uPA-binding RNA aptamers by screening a library of 35 nucleotides long 2'-fluoro-pyrimidine RNA molecules using a version of human pro-uPA lacking the epidermal growth factor-like and kringle domains as bait. One pro-uPA-binding aptamer sequence, referred to as upanap-126, proved to be highly specific for human uPA. Upanap-126 delayed the proteolytic conversion of human pro-uPA to active uPA, but did not inhibit plasminogen activation catalyzed by two-chain uPA. The aptamer also inhibited the binding of pro-uPA to uPAR and the binding of vitronectin to the preformed pro-uPA/uPAR complex, both in cell-free systems and on cell surfaces. Furthermore, upanap-126 inhibited human tumor cell invasion in vitro in the Matrigel assay and in vivo in the chick embryo assay of cell escape from microtumors. Finally, upanap-126 significantly reduced the levels of tumor cell intravasation and dissemination in the chick embryo model of spontaneous metastasis. Together, our findings show that usage of upanap-126 represents a novel multifunctional mechanistic modality for inhibition of uPA-dependent processes involved in tumor cell spread.


Assuntos
Aptâmeros de Nucleotídeos/genética , Aptâmeros de Nucleotídeos/farmacologia , Neoplasias da Próstata/patologia , Neoplasias da Próstata/terapia , Ativador de Plasminogênio Tipo Uroquinase/antagonistas & inibidores , Ativador de Plasminogênio Tipo Uroquinase/genética , Animais , Linhagem Celular Tumoral , Sistema Livre de Células , Embrião de Galinha , Fator de Crescimento Epidérmico/genética , Fator de Crescimento Epidérmico/metabolismo , Células HEK293 , Humanos , Masculino , Terapia de Alvo Molecular , Invasividade Neoplásica , Plasminogênio/genética , Plasminogênio/metabolismo , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Serina Proteases/genética , Serina Proteases/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
19.
J Biomed Biotechnol ; 2012: 564259, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23097597

RESUMO

Plasmin, one of the most potent and reactive serine proteases, is involved in various physiological processes, including embryo development, thrombolysis, wound healing and cancer progression. The proteolytic activity of plasmin is tightly regulated through activation of its precursor, plasminogen, only at specific times and in defined locales as well as through inhibition of active plasmin by its abundant natural inhibitors. By exploiting the plasminogen activating system and overexpressing distinct components of the plasminogen activation cascade, such as pro-uPA, uPAR and plasminogen receptors, malignant cells can enhance the generation of plasmin which in turn, modifies the tumor microenvironment to sustain cancer progression. While plasmin-mediated degradation and modification of extracellular matrix proteins, release of growth factors and cytokines from the stroma as well as activation of several matrix metalloproteinase zymogens, all have been a focus of cancer research studies for decades, the ability of plasmin to cleave transmembrane molecules and thereby to generate functionally important cleaved products which induce outside-in signal transduction, has just begun to receive sufficient attention. Herein, we highlight this relatively understudied, but important function of the plasmin enzyme as it is generated de novo at the interface between cross-talking cancer and host cells.


Assuntos
Comunicação Celular/fisiologia , Membrana Celular/metabolismo , Fibrinolisina/metabolismo , Modelos Biológicos , Animais , Humanos
20.
PLoS One ; 7(10): e46576, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23056350

RESUMO

After neoplastic cells leave the primary tumor and circulate, they may extravasate from the vasculature and colonize tissues to form metastases. ß1 integrins play diverse roles in tumorigenesis and tumor progression, including extravasation. In blood cells, activation of ß1 integrins can be regulated by "inside-out" signals leading to extravasation from the circulation into tissues. However, a role for inside-out ß1 activation in tumor cell metastasis is uncertain. Here we show that ß1 integrin activation promotes tumor metastasis and that activated ß1 integrin may serve as a biomarker of metastatic human melanoma. To determine whether ß1 integrin activation can influence tumor cell metastasis, the ß1 integrin subunit in melanoma and breast cancer cell lines was stably knocked down with shRNA and replaced with wild-type or constitutively-active ß1. When tumor cells expressing constitutively-active ß1 integrins were injected intravenously into chick embryos or mice, they demonstrated increased colonization of the liver when compared to cells expressing wild-type ß1 integrins. Rescue expression with mutant ß1 integrins revealed that tumor cell extravasation and hepatic colonization required extracellular ligand binding to ß1 as well as ß1 interaction with talin, an intracellular mediator of integrin activation by the Rap1 GTPase. Furthermore, shRNA-mediated knock down of talin reduced hepatic colonization by tumor cells expressing wild-type ß1, but not constitutively-active ß1. Overexpression in tumor cells of the tumor suppressor, Rap1GAP, inhibited Rap1 and ß1 integrin activation as well as hepatic colonization. Using an antibody that detects activated ß1 integrin, we found higher levels of activated ß1 integrins in human metastatic melanomas compared to primary melanomas, suggesting that activated ß1 integrin may serve as a biomarker of invasive tumor cells. Altogether, these studies establish that inside-out activation of ß1 integrins promotes tumor cell extravasation and colonization, suggesting diagnostic and therapeutic approaches for targeting of ß1 integrin signaling in neoplasia.


Assuntos
Integrina beta1/metabolismo , Metástase Neoplásica , Neoplasias/patologia , Linhagem Celular Tumoral , Humanos , Neoplasias/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...