Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
2.
Nature ; 522(7554): 106-110, 2015 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-26017313

RESUMO

Tumour metastasis is a complex process involving reciprocal interplay between cancer cells and host stroma at both primary and secondary sites, and is strongly influenced by microenvironmental factors such as hypoxia. Tumour-secreted proteins play a crucial role in these interactions and present strategic therapeutic potential. Metastasis of breast cancer to the bone affects approximately 85% of patients with advanced disease and renders them largely untreatable. Specifically, osteolytic bone lesions, where bone is destroyed, lead to debilitating skeletal complications and increased patient morbidity and mortality. The molecular interactions governing the early events of osteolytic lesion formation are currently unclear. Here we show hypoxia to be specifically associated with bone relapse in patients with oestrogen-receptor negative breast cancer. Global quantitative analysis of the hypoxic secretome identified lysyl oxidase (LOX) as significantly associated with bone-tropism and relapse. High expression of LOX in primary breast tumours or systemic delivery of LOX leads to osteolytic lesion formation whereas silencing or inhibition of LOX activity abrogates tumour-driven osteolytic lesion formation. We identify LOX as a novel regulator of NFATc1-driven osteoclastogenesis, independent of RANK ligand, which disrupts normal bone homeostasis leading to the formation of focal pre-metastatic lesions. We show that these lesions subsequently provide a platform for circulating tumour cells to colonize and form bone metastases. Our study identifies a novel mechanism of regulation of bone homeostasis and metastasis, opening up opportunities for novel therapeutic intervention with important clinical implications.


Assuntos
Neoplasias Ósseas/patologia , Neoplasias Ósseas/secundário , Neoplasias da Mama/enzimologia , Neoplasias da Mama/metabolismo , Metástase Neoplásica , Proteína-Lisina 6-Oxidase/metabolismo , Animais , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/prevenção & controle , Neoplasias da Mama/patologia , Hipóxia Celular , Linhagem Celular Tumoral , Movimento Celular , Feminino , Humanos , Camundongos , Fatores de Transcrição NFATC/metabolismo , Metástase Neoplásica/patologia , Células Neoplásicas Circulantes/patologia , Osteoblastos/metabolismo , Osteoblastos/patologia , Osteoclastos/metabolismo , Osteoclastos/patologia , Proteína-Lisina 6-Oxidase/antagonistas & inibidores , Receptores de Estrogênio/deficiência , Receptores de Estrogênio/genética
3.
Cancer Cell ; 9(1): 33-44, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16413470

RESUMO

Inhibition of ERK-MAPK signaling by expression of dominant-negative MEK1 in the tumor vasculature suppresses angiogenesis and tumor growth. In an organotypic tissue culture angiogenesis assay, ERK-MAPK inhibition during the migratory phase results in loss of bipolarity, detachment, and cell death of isolated endothelial cells and retraction of sprouting tubules. These effects are the consequence of upregulated Rho-kinase signaling. Transient inhibition of Rho-kinase rescues the effects of ERK-MAPK inhibition in vitro and in vivo, promotes sprouting, and increases vessel length in tumors. We propose a regulatory role of Rho-kinase by ERK-MAPK during angiogenesis that acts through the control of actomyosin contractility. Our data delineate a mechanism by which ERK-MAPK promotes endothelial cell survival and sprouting by downregulating Rho-kinase signaling.


Assuntos
Células Endoteliais/fisiologia , Endotélio Vascular/patologia , MAP Quinase Quinase 1/metabolismo , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Neovascularização Patológica/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Actomiosina/metabolismo , Animais , Movimento Celular , Polaridade Celular , Sobrevivência Celular , Células Cultivadas , Técnicas de Cocultura , Fibroblastos/citologia , Regulação Neoplásica da Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , MAP Quinase Quinase 1/genética , Masculino , Camundongos , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Neovascularização Patológica/patologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Transdução de Sinais , Veias Umbilicais/citologia , Quinases Associadas a rho
4.
Angiogenesis ; 15(4): 623-41, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22843200

RESUMO

Sunitinib is a potent and clinically approved tyrosine kinase inhibitor that can suppress tumour growth by inhibiting angiogenesis. However, conflicting data exist regarding the effects of this drug on the growth of metastases in preclinical models. Here we use 4T1 and RENCA tumour cells, which both form lung metastases in Balb/c mice, to re-address the effects of sunitinib on the progression of metastatic disease in mice. We show that treatment of mice with sunitinib prior to intravenous injection of tumour cells can promote the seeding and growth of 4T1 lung metastases, but not RENCA lung metastases, showing that this effect is cell line dependent. However, increased metastasis occurred only upon administration of a very high sunitinib dose, but not when lower, clinically relevant doses were used. Mechanistically, high dose sunitinib led to a pericyte depletion effect in the lung vasculature that correlated with increased seeding of metastasis. By administering sunitinib to mice after intravenous injection of tumour cells, we demonstrate that while sunitinib does not inhibit the growth of 4T1 lung tumour nodules, it does block the growth of RENCA lung tumour nodules. This contrasting response was correlated with increased myeloid cell recruitment and persistent vascularisation in 4T1 tumours, whereas RENCA tumours recruited less myeloid cells and were more profoundly devascularised upon sunitinib treatment. Finally, we show that progression of 4T1 tumours in sunitinib treated mice results in increased hypoxia and increased glucose metabolism in these tumours and that this is associated with a poor outcome. Taken together, these data suggest that the effects of sunitinib on tumour progression are dose-dependent and tumour model-dependent. These findings have relevance for understanding how anti-angiogenic agents may influence disease progression when used in the adjuvant or metastatic setting in cancer patients.


Assuntos
Antineoplásicos/uso terapêutico , Indóis/uso terapêutico , Metástase Neoplásica/tratamento farmacológico , Pirróis/uso terapêutico , Animais , Camundongos , Camundongos Endogâmicos BALB C , Tomografia por Emissão de Pósitrons , Sunitinibe , Tomografia Computadorizada por Raios X
7.
Mol Cancer Res ; 11(11): 1425-36, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24008674

RESUMO

UNLABELLED: Cancer-associated fibroblasts enhance cancer progression when activated by tumor cells through mechanisms not yet fully understood. Blocking mammary tumor cell-derived lysyl oxidase-like 2 (LOXL2) significantly inhibited mammary tumor cell invasion and metastasis in transgenic and orthotopic mouse models. Here, we discovered that tumor-derived LOXL2 directly activated stromal fibroblasts in the tumor microenvironment. Genetic manipulation or antibody inhibition of LOXL2 in orthotopically grown mammary tumors reduced the expression of α-smooth muscle actin (α-SMA). Using a marker for reticular fibroblasts, it was determined that expression of α-SMA was localized to fibroblasts recruited from the host tissue. This marker also revealed that the matrix present in tumors with reduced levels of LOXL2 was more scattered compared with control tumors which exhibited matrices with dense, parallel alignments. Importantly, in vitro assays revealed that tumor-derived LOXL2 and a recombinant LOXL2 protein induced fibroblast branching on collagen matrices, as well as increased fibroblast-mediated collagen contraction and invasion of fibroblasts through extracellular matrix. Moreover, LOXL2 induced the expression of α-SMA in fibroblasts grown on collagen matrices. Mechanistically, it was determined that LOXL2 activated fibroblasts through integrin-mediated focal adhesion kinase activation. These results indicate that inhibition of LOXL2 in tumors not only reduces tumor cell invasion but also attenuates the activation of host cells in the tumor microenvironment. IMPLICATIONS: These findings reveal new insight into the mechanisms of fibroblast activation, a novel function of LOXL2, and further highlight the importance of generating LOXL2-targeted therapies for the prevention of tumor progression and metastasis.


Assuntos
Aminoácido Oxirredutases/metabolismo , Fibroblastos/patologia , Neoplasias Mamárias Experimentais/genética , Transdução de Sinais/genética , Células 3T3 , Actinas/genética , Actinas/metabolismo , Aminoácido Oxirredutases/genética , Animais , Adesão Celular , Linhagem Celular Tumoral , Proliferação de Células , Colágeno/metabolismo , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Feminino , Fibroblastos/metabolismo , Quinase 1 de Adesão Focal/genética , Quinase 1 de Adesão Focal/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Invasividade Neoplásica , Microambiente Tumoral
8.
Cancer Res ; 73(6): 1721-32, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23345161

RESUMO

Tumor metastasis is a highly complex, dynamic, and inefficient process involving multiple steps, yet it accounts for more than 90% of cancer-related deaths. Although it has long been known that fibrotic signals enhance tumor progression and metastasis, the underlying molecular mechanisms are still unclear. Identifying events involved in creating environments that promote metastatic colonization and growth are critical for the development of effective cancer therapies. Here, we show a critical role for lysyl oxidase (LOX) in establishing a milieu within fibrosing tissues that is favorable to growth of metastastic tumor cells. We show that LOX-dependent collagen crosslinking is involved in creating a growth-permissive fibrotic microenvironment capable of supporting metastatic growth by enhancing tumor cell persistence and survival. We show that therapeutic targeting of LOX abrogates not only the extent to which fibrosis manifests, but also prevents fibrosis-enhanced metastatic colonization. Finally, we show that the LOX-mediated collagen crosslinking directly increases tumor cell proliferation, enhancing metastatic colonization and growth manifesting in vivo as increased metastasis. This is the first time that crosslinking of collagen I has been shown to enhance metastatic growth. These findings provide an important link between ECM homeostasis, fibrosis, and cancer with important clinical implications for both the treatment of fibrotic disease and cancer.


Assuntos
Colágeno/metabolismo , Fibrose , Metástase Neoplásica , Proteína-Lisina 6-Oxidase/metabolismo , Western Blotting , Linhagem Celular , Proliferação de Células , Imunofluorescência , Humanos , Neoplasias Pulmonares/secundário
9.
Cancer Res ; 73(2): 583-94, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23188504

RESUMO

Identification of key molecules that drive angiogenesis is critical for the development of new modalities for the prevention of solid tumor progression. Using multiple models of colorectal cancer, we show that activity of the extracellular matrix-modifying enzyme lysyl oxidase (LOX) is essential for stimulating endothelial cells in vitro and angiogenesis in vivo. We show that LOX activates Akt through platelet-derived growth factor receptor ß (PDGFRß) stimulation, resulting in increased VEGF expression. LOX-driven angiogenesis can be abrogated through targeting LOX directly or using inhibitors of PDGFRß, Akt, and VEGF signaling. Furthermore, we show that LOX is clinically correlated with VEGF expression and blood vessel formation in 515 colorectal cancer patient samples. Finally, we validate our findings in a breast cancer model, showing the universality of these observations. Taken together, our findings have broad clinical and therapeutic implications for a wide variety of solid tumor types.


Assuntos
Neoplasias Colorretais/irrigação sanguínea , Neoplasias Colorretais/metabolismo , Células Endoteliais/metabolismo , Neovascularização Patológica/metabolismo , Proteína-Lisina 6-Oxidase/metabolismo , Animais , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Células Endoteliais/patologia , Feminino , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Proteína-Lisina 6-Oxidase/antagonistas & inibidores , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo
10.
J Natl Cancer Inst ; 104(12): 941-52, 2012 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-22556269

RESUMO

BACKGROUND: The tripartite motif family protein 27 (TRIM27) is a transcriptional repressor that interacts with, and attenuates senescence induction by, the retinoblastoma-associated protein (RB1). High expression of TRIM27 was noted in several human cancer types including breast and endometrial cancer, where elevated TRIM27 expression predicts poor prognosis. Here, we investigated the role of TRIM27 expression in cancer development. METHODS: We assessed TRIM27 expression in human cancer using cancer profiling arrays containing paired tumor and normal cRNA (n = 261) as well as in murine skin cancer induced by 7, 12-dimethylbenzanthracene (DMBA)/12-O-tetradecanoylphorbol-13-acetate (TPA). We generated mice with disrupted expression of murine TRIM27 (Trim27(-/-)) and assessed their susceptibility to DMBA/TPA-induced skin tumor development compared with isogenic littermates (n = 26 mice per group). We assessed the effect of Trim27 loss on senescence propensity in mouse embryonic fibroblasts (MEFs) by quantifying cell proliferation alongside senescence markers (senescence-associated ß-galactosidase [SA-ß-gal] activity and hypertrophic cell morphology). The contribution of RB1 on senescence and cancer susceptibility (n > 20 mice per group) in Trim27(-/-) backgrounds was also assessed. Data were analyzed using the Student's t, χ(2), or log-rank test as indicated. All statistical tests were two-sided. RESULTS: TRIM27 transcript levels are statistically significantly increased in common human cancers, including colon and lung, vs normal tissues (TRIM27 expression relative to ubiquitin: cancers vs normal tissues, mean = 0.59, 95% confidence interval [CI] = 0.55 to 0.63 vs mean = 0.46, 95% CI =0.43 to 0.49, P < .001) as well as in chemically induced mouse skin cancer compared with matched normal tissue (Trim27 expression relative to Gapdh control: tumor vs normal skin, mean = 4.2, 95% CI = 3.97 to 4.43 vs mean = 0.96, 95% CI = 0.69 to 1.2, P < .001). Trim27(-/-) mice (n = 14) were resistant to chemically induced skin cancer development (eight [57.2%] of 14 mice were tumor free) compared with Trim27(+/+) wild-type littermates (n = 13) (one [7.7%] of 13 mice was tumor free). Trim27(-/-) MEFs show enhanced senescence propensity in response to replicative (percentage of SA-ß-gal-positive cells: Trim27(+/+) MEFs vs Trim27(-/-) MEFs, mean = 14.2%, 95% CI = 11.1% to 17.4% vs mean = 53.3%, 95% CI = 48.7% to 57.9%, P < .001) or oncogenic stress (percentage of SA-ß-gal-positive cells: Trim27(+/+) MEFs + Ras vs Trim27(-/-) MEFs + Ras, mean = 24.0%, 95% CI = 19.9% to 28.1% vs mean = 37.3%, 95% CI = 32.2% to 42.4%, P < .05) compared with Trim27(+/+) MEFs. These responses were alleviated following inactivation of murine RB1 (Rb1). Furthermore, Trim27(-/-) mice are not protected from cancers arising as a consequence of Rb1 deletion (median survival: Trim27(-/-)Rb(+/-) vs Trim27(+/+)Rb(+/-), 14 vs 13 months; difference = 1.0 month, 95% CI = 0.5 to 1.6 months, P = .14). CONCLUSION: TRIM27 expression is a modifier of disease incidence and progression relevant to the development of common human cancers and is a potential target for intervention in cancer.


Assuntos
Proliferação de Células , Senescência Celular , Proteínas de Ligação a DNA/metabolismo , Proteínas Nucleares/metabolismo , Neoplasias Cutâneas/metabolismo , Fatores de Confusão Epidemiológicos , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Progressão da Doença , Fibroblastos/metabolismo , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Prognóstico , RNA Complementar/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Projetos de Pesquisa , Proteína do Retinoblastoma/deficiência , Proteína do Retinoblastoma/metabolismo , Neoplasias Cutâneas/induzido quimicamente , Ubiquitina-Proteína Ligases , Regulação para Cima
11.
J Natl Cancer Inst ; 103(5): 407-24, 2011 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-21282564

RESUMO

BACKGROUND: Emerging evidence implicates lysyl oxidase (LOX), an extracellular matrix-modifying enzyme, in promoting metastasis of solid tumors. We investigated whether LOX plays an important role in the metastasis of colorectal cancer (CRC). METHODS: We analyzed LOX expression in a patient CRC tissue microarray consisting of normal colon mucosa (n = 49), primary (n = 510), and metastatic (n = 198) tissues. LOX was overexpressed in CRC cell line SW480 (SW480+LOX), and the expression was knocked down in CRC cell line SW620 using LOX-specific short hairpin RNA (SW620+shLOX). Effect of LOX manipulation on three-dimensional cell proliferation and invasion was characterized in vitro. Effect of LOX manipulation on tumor proliferation and metastasis was investigated in a subcutaneous tumor mouse model (n = 3 mice per group) and in an intrasplenic metastatic mouse model (n = 3 mice per group). The mechanism of LOX-mediated effects via v-src sarcoma (Schmidt-Ruppin A-2) viral oncogene homolog (avian) (SRC) was investigated using dasatinib, an inhibitor of SRC activation. All statistical tests were two-sided. RESULTS: Compared with normal colon tissue (n = 49), LOX expression was statistically significantly increased in tumor tissues (n = 510) of CRC patients (P < .001), and a greater increase was observed in metastatic tissue (n = 198). SW480+LOX cells showed a statistically significantly increased three-dimensional proliferation (P = .037) and invasion (P = .015), whereas SW620+shLOX cells showed reduced proliferation (P = .011) and invasion (P = .013) compared with controls. Subcutaneous tumor growth in mice was statistically significantly increased in SW480+LOX tumors (P = .036) and decreased in SW620+shLOX tumors (P = .048), and metastasis was statistically significantly increased in SW480+LOX tumors (P = .044) and decreased in SW620+shLOX tumors (SW620 control vs SW620+shLOX, mean = 1.0 luminescent signal, 95% confidence interval = 0.3 to 1.7 luminescent signal, vs mean = 0.3 luminescent signal, 95% confidence interval = 0.1 to 0.5 luminescent signal; P = .035) compared with controls. LOX-mediated effects on tumor progression were associated with SRC activation, and these effects were inhibited by dasatinib. CONCLUSIONS: LOX showed an important role in CRC cell proliferation and metastasis and was dependent on the activation of SRC. These results have the potential to identify patients with high SRC activity, who may benefit from dasatinib treatment.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Colorretais/enzimologia , Neoplasias Colorretais/patologia , Inibidores de Proteínas Quinases/farmacologia , Proteína-Lisina 6-Oxidase/metabolismo , Pirimidinas/farmacologia , Tiazóis/farmacologia , Quinases da Família src/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias Colorretais/tratamento farmacológico , Dasatinibe , Progressão da Doença , Ativação Enzimática , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Vetores Genéticos , Humanos , Immunoblotting , Imuno-Histoquímica , Camundongos , Camundongos Nus , Mutação , Fosforilação , Análise Serial de Proteínas , Proteína-Lisina 6-Oxidase/genética , RNA Interferente Pequeno/metabolismo , Retroviridae , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Regulação para Cima , Quinases da Família src/antagonistas & inibidores
12.
Cancer Res ; 71(5): 1561-72, 2011 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21233336

RESUMO

More than 90% of cancer patient mortality is attributed to metastasis. In this study, we investigated a role for the lysyl oxidase-related enzyme lysyl oxidase-like 2 (LOXL2) in breast cancer metastasis, in both patient samples and in vivo models. Analysis of a published microarray data set revealed that LOXL2 expression is correlated with metastasis and decreased survival in patients with aggressive breast cancer. In immunocompetent or immunocompromised orthotopic and transgenic breast cancer models we showed that genetic, chemical or antibody-mediated inhibition of LOXL2 resulted in decreased metastasis. Mechanistic investigations revealed that LOXL2 promotes invasion by regulating the expression and activity of the extracellular proteins tissue inhibitor of metalloproteinase-1 (TIMP1) and matrix metalloproteinase-9 (MMP9). We found that LOXL2, TIMP1, and MMP9 are coexpressed during mammary gland involution, suggesting they function together in glandular remodeling after weaning. Finally, we found that LOXL2 is highly expressed in the basal/myoepithelial mammary cell lineage, like many other genes that are upregulated in basal-like breast cancers. Our findings highlight the importance of LOXL2 in breast cancer progression and support the development of anti-LOXL2 therapeutics for the treatment of metastatic breast cancer.


Assuntos
Aminoácido Oxirredutases/metabolismo , Neoplasias da Mama/metabolismo , Aminoácido Oxirredutases/genética , Animais , Western Blotting , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Matriz Extracelular/metabolismo , Feminino , Imunofluorescência , Expressão Gênica , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Metaloproteinase 9 da Matriz/biossíntese , Camundongos , Invasividade Neoplásica/genética , Metástase Neoplásica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise Serial de Tecidos , Inibidor Tecidual de Metaloproteinase-1/biossíntese
13.
Cancer Cell ; 15(1): 35-44, 2009 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-19111879

RESUMO

Tumor cell metastasis is facilitated by "premetastatic niches" formed in destination organs by invading bone marrow-derived cells (BMDCs). Lysyl oxidase (LOX) is critical for premetastatic niche formation. LOX secreted by hypoxic breast tumor cells accumulates at premetastatic sites, crosslinks collagen IV in the basement membrane, and is essential for CD11b+ myeloid cell recruitment. CD11b+ cells adhere to crosslinked collagen IV and produce matrix metalloproteinase-2, which cleaves collagen, enhancing the invasion and recruitment of BMDCs and metastasizing tumor cells. LOX inhibition prevents CD11b+ cell recruitment and metastatic growth. CD11b+ cells and LOX also colocalize in biopsies of human metastases. Our findings demonstrate a critical role for LOX in premetastatic niche formation and support targeting LOX for the treatment and prevention of metastatic disease.


Assuntos
Células da Medula Óssea/citologia , Neoplasias da Mama/enzimologia , Neoplasias da Mama/patologia , Diferenciação Celular , Movimento Celular , Hipóxia/enzimologia , Proteína-Lisina 6-Oxidase/metabolismo , Animais , Membrana Basal/citologia , Membrana Basal/enzimologia , Linhagem Celular Tumoral , Separação Celular , Colágeno Tipo IV/metabolismo , Progressão da Doença , Ativação Enzimática , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Hipóxia/patologia , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/patologia , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Camundongos Nus , Células Mieloides/citologia , Metástase Neoplásica/patologia , Proteína-Lisina 6-Oxidase/genética , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA