Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cancer Res ; 81(9): 2415-2428, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33526510

RESUMEN

Multiple myeloma promotes systemic skeletal bone disease that greatly contributes to patient morbidity. Resorption of type I collagen-rich bone matrix by activated osteoclasts results in the release of sequestered growth factors that can drive progression of the disease. Matrix metalloproteinase-13 (MMP13) is a collagenase expressed predominantly in the skeleton by mesenchymal stromal cells (MSC) and MSC-derived osteoblasts. Histochemical analysis of human multiple myeloma specimens also demonstrated that MMP13 largely localizes to the stromal compartment compared with CD138+ myeloma cells. In this study, we further identified that multiple myeloma induces MMP13 expression in bone stromal cells. Because of its ability to degrade type I collagen, we examined whether bone stromal-derived MMP13 contributed to myeloma progression. Multiple myeloma cells were inoculated into wild-type or MMP13-null mice. In independent in vivo studies, MMP13-null mice demonstrated significantly higher overall survival rates and lower levels of bone destruction compared with wild-type controls. Unexpectedly, no differences in type I collagen processing between the groups were observed. Ex vivo stromal coculture assays showed reduced formation and activity in MMP13-null osteoclasts. Analysis of soluble factors from wild-type and MMP13-null MSCs revealed decreased bioavailability of various osteoclastogenic factors including CXCL7. CXCL7 was identified as a novel MMP13 substrate and regulator of osteoclastogenesis. Underscoring the importance of host MMP13 catalytic activity in multiple myeloma progression, we demonstrate the in vivo efficacy of a novel and highly selective MMP13 inhibitor that provides a translational opportunity for the treatment of this incurable disease. SIGNIFICANCE: Genetic and pharmacologic approaches show that bone stromal-derived MMP13 catalytic activity is critical for osteoclastogenesis, bone destruction, and disease progression. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/9/2415/F1.large.jpg.


Asunto(s)
Metaloproteinasa 13 de la Matriz/metabolismo , Mieloma Múltiple/metabolismo , Mieloma Múltiple/mortalidad , Osteólisis/genética , Transducción de Señal/genética , Animales , Diferenciación Celular/genética , Línea Celular Tumoral , Quimiocinas CXC/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Humanos , Masculino , Metaloproteinasa 13 de la Matriz/genética , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteogénesis/genética , Tasa de Supervivencia
2.
Oncotarget ; 8(26): 41827-41840, 2017 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-28611279

RESUMEN

Multiple myeloma is a plasma cell malignancy that homes aberrantly to bone causing extensive skeletal destruction. Despite the development of novel therapeutic agents that have significantly improved overall survival, multiple myeloma remains an incurable disease. Matrix metalloproteinase-2 (MMP-2) is associated with cancer and is significantly overexpressed in the bone marrow of myeloma patients. These data provide rationale for selectively inhibiting MMP-2 activity as a multiple myeloma treatment strategy. Given that MMP-2 is systemically expressed, we used novel "bone-seeking" bisphosphonate based MMP-2 specific inhibitors (BMMPIs) to target the skeletal tissue thereby circumventing potential off-target effects of MMP-2 inhibition outside the bone marrow-tumor microenvironment. Using in vivo models of multiple myeloma (5TGM1, U266), we examined the impact of MMP-2 inhibition on disease progression using BMMPIs. Our data demonstrate that BMMPIs can decrease multiple myeloma burden and protect against cancer-induced osteolysis. Additionally, we have shown that MMP-2 can be specifically inhibited in the multiple myeloma-bone microenvironment, underscoring the feasibility of developing targeted and tissue selective MMP inhibitors. Given the well-tolerated nature of bisphosphonates in humans, we anticipate that BMMPIs could be rapidly translated to the clinical setting for the treatment of multiple myeloma.


Asunto(s)
Huesos/patología , Microambiente Celular/efectos de los fármacos , Metaloproteinasa 2 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Animales , Biopsia , Médula Ósea/metabolismo , Médula Ósea/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Microambiente Celular/genética , Modelos Animales de Enfermedad , Activación Enzimática/efectos de los fármacos , Femenino , Expresión Génica , Humanos , Masculino , Metaloproteinasa 2 de la Matriz/genética , Ratones , Ratones Noqueados , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/genética , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Osteólisis/tratamiento farmacológico , Osteólisis/metabolismo , Osteólisis/patología , Transporte de Proteínas
3.
Mol Cancer Ther ; 16(3): 494-505, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28069877

RESUMEN

Bone metastasis is common during breast cancer progression. Matrix metalloproteinase-2 (MMP-2) is significantly associated with aggressive breast cancer and poorer overall survival. In bone, tumor- or host-derived MMP-2 contributes to breast cancer growth and does so by processing substrates, including type I collagen and TGFß latency proteins. These data provide strong rationale for the application of MMP-2 inhibitors to treat the disease. However, in vivo, MMP-2 is systemically expressed. Therefore, to overcome potential toxicities noted with previous broad-spectrum MMP inhibitors (MMPIs), we used highly selective bisphosphonic-based MMP-2 inhibitors (BMMPIs) that allowed for specific bone targeting. In vitro, BMMPIs affected the viability of breast cancer cell lines and osteoclast precursors, but not osteoblasts. In vivo, we demonstrated using two bone metastatic models (PyMT-R221A and 4T1) that BMMPI treatment significantly reduced tumor growth and tumor-associated bone destruction. In addition, BMMPIs are superior in promoting tumor apoptosis compared with the standard-of-care bisphosphonate, zoledronate. We demonstrated MMP-2-selective inhibition in the bone microenvironment using specific and broad-spectrum MMP probes. Furthermore, compared with zoledronate, BMMPI-treated mice had significantly lower levels of TGFß signaling and MMP-generated type I collagen carboxy-terminal fragments. Taken together, our data show the feasibility of selective inhibition of MMPs in the bone metastatic breast cancer microenvironment. We posit that BMMPIs could be easily translated to the clinical setting for the treatment of bone metastases given the well-tolerated nature of bisphosphonates. Mol Cancer Ther; 16(3); 494-505. ©2017 AACR.


Asunto(s)
Neoplasias Óseas/secundario , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Metaloproteinasa 2 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Animales , Conservadores de la Densidad Ósea/farmacología , Neoplasias Óseas/diagnóstico , Neoplasias Óseas/mortalidad , Neoplasias de la Mama/genética , Neoplasias de la Mama/mortalidad , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Difosfonatos/farmacología , Progresión de la Enfermedad , Activación Enzimática/efectos de los fármacos , Femenino , Humanos , Imidazoles/farmacología , Estimación de Kaplan-Meier , Metaloproteinasa 2 de la Matriz/genética , Ratones , Modelos Biológicos , Imagen Multimodal , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Carga Tumoral/efectos de los fármacos , Microambiente Tumoral , Ácido Zoledrónico
4.
Small GTPases ; 6(4): 202-11, 2015 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-26399387

RESUMEN

Bisphosphonate drugs such as zoledronic acid (ZOL), used for the treatment of common bone disorders, target the skeleton and inhibit bone resorption by preventing the prenylation of small GTPases in bone-destroying osteoclasts. Increasing evidence indicates that bisphosphonates also have pleiotropic effects outside the skeleton, most likely via cells of the monocyte/macrophage lineage exposed to nanomolar circulating drug concentrations. However, no effects of such low concentrations of ZOL have been reported using existing approaches. We have optimized a highly sensitive in vitro prenylation assay utilizing recombinant geranylgeranyltransferases to enable the detection of subtle effects of ZOL on the prenylation of Rab- and Rho-family GTPases. Using this assay, we found for the first time that concentrations of ZOL as low as 10nM caused inhibition of Rab prenylation in J774 macrophages following prolonged cell culture. By combining the assay with quantitative mass spectrometry we identified an accumulation of 18 different unprenylated Rab proteins in J774 cells after nanomolar ZOL treatment, with a >7-fold increase in the unprenylated form of Rab proteins associated with the endophagosome pathway (Rab1, Rab5, Rab6, Rab7, Rab11, Rab14 and Rab21). Finally, we also detected a clear effect of subcutaneous ZOL administration in vivo on the prenylation of Rab1A, Rab5B, Rab7A and Rab14 in mouse peritoneal macrophages, confirming that systemic treatment with bisphosphonate drug can inhibit prenylation in myeloid cells in vivo outside the skeleton. These observations begin a new era in defining the precise pharmacological actions of bisphosphonate drugs on the prenylation of small GTPases in vivo.


Asunto(s)
Difosfonatos/farmacología , Imidazoles/farmacología , Macrófagos Peritoneales/metabolismo , Prenilación de Proteína/efectos de los fármacos , Proteínas de Unión al GTP rab/metabolismo , Animales , Línea Celular , Ratones , Ácido Zoledrónico
5.
Matrix Biol ; 44-46: 200-6, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25652204

RESUMEN

Matrix metalloproteinases have long been associated with cancer. Clinical trials of small molecule inhibitors for this family of enzymes however, were spectacularly unsuccessful in a variety of tumor types. Here, we discuss some of the newer roles that have been uncovered for MMPs in cancer that would not have been targeted with those initial inhibitors or in the patient populations analyzed. We also consider novel ways of using cancer-associated MMP activity for clinical benefit.


Asunto(s)
Metaloproteinasas de la Matriz/metabolismo , Neoplasias/patología , Ensayos Clínicos como Asunto , Humanos , Metaloproteinasas de la Matriz/efectos de los fármacos , Metástasis de la Neoplasia , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología
6.
Cancer Discov ; 5(1): 35-42, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25312016

RESUMEN

UNLABELLED: Recent clinical trials have shown that bisphosphonate drugs improve breast cancer patient survival independent of their antiresorptive effects on the skeleton. However, because bisphosphonates bind rapidly to bone mineral, the exact mechanisms of their antitumor action, particularly on cells outside of bone, remain unknown. Here, we used real-time intravital two-photon microscopy to show extensive leakage of fluorescent bisphosphonate from the vasculature in 4T1 mouse mammary tumors, where it initially binds to areas of small, granular microcalcifications that are engulfed by tumor-associated macrophages (TAM), but not tumor cells. Importantly, we also observed uptake of radiolabeled bisphosphonate in the primary breast tumor of a patient and showed the resected tumor to be infiltrated with TAMs and to contain similar granular microcalcifications. These data represent the first compelling in vivo evidence that bisphosphonates can target cells in tumors outside the skeleton and that their antitumor activity is likely to be mediated via TAMs. SIGNIFICANCE: Bisphosphonates are assumed to act solely in bone. However, mouse models and clinical trials show that they have surprising antitumor effects outside bone. We provide unequivocal evidence that bisphosphonates target TAMs, but not tumor cells, to exert their extraskeletal effects, offering a rationale for use in patients with early disease.


Asunto(s)
Conservadores de la Densidad Ósea/metabolismo , Difosfonatos/metabolismo , Macrófagos/metabolismo , Neoplasias/diagnóstico , Tomografía Computarizada de Emisión de Fotón Único , Tomografía Computarizada por Rayos X , Animales , Conservadores de la Densidad Ósea/uso terapéutico , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/inmunología , Calcinosis , Carbocianinas , Difosfonatos/uso terapéutico , Modelos Animales de Enfermedad , Femenino , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Persona de Mediana Edad , Clasificación del Tumor , Invasividad Neoplásica , Neoplasias/tratamiento farmacológico , Fagocitosis/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Cancer Metastasis Rev ; 33(2-3): 511-25, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24414228

RESUMEN

In prostate to bone metastases, the "vicious cycle" paradigm has been traditionally used to illustrate how metastases manipulate the bone forming osteoblasts and resorbing osteoclasts in order to yield factors that facilitate growth and establishment. However, recent advances have illustrated that the cycle is far more complex than this simple interpretation. In this review, we will discuss the role of exosomes and hematopoietic/mesenchymal stem/stromal cells (MSC) that facilitate the establishment and activation of prostate metastases and how cells including myeloid-derived suppressor cells, macrophages, T cells, and nerve cells contribute to the momentum of the vicious cycle. The increased complexity of the tumor-bone microenvironment requires a system level approach. The evolution of computational models to interrogate the tumor-bone microenvironment is also discussed, and the application of this integrated approach should allow for the development of effective therapies to treat and cure prostate to bone metastases.


Asunto(s)
Neoplasias Óseas/secundario , Neoplasias de la Próstata/etiología , Neoplasias de la Próstata/patología , Inmunidad Adaptativa , Animales , Neoplasias Óseas/complicaciones , Humanos , Inmunidad Innata , Masculino , Modelos Biológicos , Células Madre Neoplásicas/metabolismo , Dolor/etiología , Neoplasias de la Próstata/metabolismo , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...