Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Cell Mol Life Sci ; 78(9): 4085-4093, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33570672

RESUMEN

Multiple myeloma is a malignancy caused by the clonal expansion of abnormal plasma cells. Myeloma cells have proven to be incredibly successful at manipulating their microenvironment to promote growth and to evade modern therapies. They have evolved to utilise the integral signalling pathways of the bone and bone marrow to drive disease progression. The bone marrow is often described in the context of a single structure that fills the bone cavity and supports normal haematopoiesis. However, within that structure exists two anatomically different niches, the perivascular niche and the endosteal niche. These contain different cell types functioning to support normal immune and blood cell production as well as healthy bone. These cells secrete numerous signalling molecules that can influence myeloma cell biology and behaviour. The endosteal niche is home to specific bone cell lineages and plays a pivotal role in myeloma cell establishment and survival. This review will concentrate on some of the signalling pathways that are hijacked by myeloma cells to shape a favourable environment, and the different influences myeloma cells are exposed to depending on their spatial location within the bone marrow.


Asunto(s)
Médula Ósea/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Mieloma Múltiple/patología , Transducción de Señal , Proteínas Morfogenéticas Óseas/metabolismo , Huesos/metabolismo , Huesos/patología , Exosomas/metabolismo , Hematopoyesis , Humanos , Mieloma Múltiple/metabolismo
2.
J Cell Physiol ; 233(12): 9159-9166, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29943829

RESUMEN

Obesity has become a global epidemic influencing the establishment and progression of a wide range of diseases, such as diabetes, cardiovascular disease, and cancer. In 2016, International Agency for Research on Cancer reported that obesity is now associated with 13 different cancers, one of which is multiple myeloma (MM), a destructive cancer of plasma cells that predominantly reside in the bone marrow. Obesity is the accumulation of excess body fat, which causes metabolic, endocrine, immunologic, and inflammatory-like changes. Obesity is usually associated with an increase in visceral and/or subcutaneous fat; however, an additional fat depot that also responds to diet-induced changes is bone marrow adipose tissue (BMAT). There have been several studies over the past few decades that have identified BMAT as a key driver in MM progression. Adipocytes secrete numerous adipokines, such as leptin, adiponectin, resistin, adipsin, and visfatin, which when secreted at normal controlled levels have protective properties. However, in obesity these levels of secretion change, coupled with an increase in adipocyte number and size causing a profound and lasting effect on the bone microenvironment, contributing to MM cell growth, survival, and migration as well as potentially fueling bone destruction. Obesity is a modifiable risk factor making it an attractive option for targeted therapy. This review discusses the link between obesity, monoclonal gammopathy of undetermined significance (a benign condition that precedes MM), and myeloma, and the contribution of key adipokines to disease establishment and progression.


Asunto(s)
Adipoquinas/metabolismo , Adiposidad/genética , Mieloma Múltiple/genética , Obesidad/genética , Adipocitos/metabolismo , Adipoquinas/genética , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Humanos , Mieloma Múltiple/complicaciones , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Obesidad/complicaciones , Obesidad/metabolismo , Obesidad/patología , Microambiente Tumoral/genética
3.
Transl Oncol ; 15(1): 101301, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34890968

RESUMEN

Multiple myeloma is a haematological malignancy that is dependent upon interactions within the bone microenvironment to drive tumour growth and osteolytic bone disease. Metformin is an anti-diabetic drug that has attracted attention due to its direct antitumor effects, including anti-myeloma properties. However, the impact of the bone microenvironment on the response to metformin in myeloma is unknown. We have employed in vitro and in vivo models to dissect out the direct effects of metformin in bone and the subsequent indirect myeloma response. We demonstrate how metformin treatment of preosteoblasts increases myeloma cell attachment. Metformin-treated preosteoblasts increased osteopontin (OPN) expression that upon silencing, reduced subsequent myeloma cell adherence. Proliferation markers were reduced in myeloma cells cocultured with metformin-treated preosteoblasts. In vivo, mice were treated with metformin for 4 weeks prior to inoculation of 5TGM1 myeloma cells. Metformin-pretreated mice had an increase in tumour burden, associated with an increase in osteolytic bone lesions and elevated OPN expression in the bone marrow. Collectively, we show that metformin increases OPN expression in preosteoblasts, increasing myeloma cell adherence. In vivo, this translates to an unexpected indirect pro-tumourigenic effect of metformin, highlighting the importance of the interdependence between myeloma cells and cells of the bone microenvironment.

4.
Sci Adv ; 8(8): eabf9096, 2022 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-35213227

RESUMEN

The spread of cancer to bone is invariably fatal, with complex cross-talk between tumor cells and the bone microenvironment responsible for driving disease progression. By combining in silico analysis of patient datasets with metabolomic profiling of prostate cancer cells cultured with bone cells, we demonstrate the changing energy requirements of prostate cancer cells in the bone microenvironment, identifying the pentose phosphate pathway (PPP) as elevated in prostate cancer bone metastasis, with increased expression of the PPP rate-limiting enzyme glucose-6-phosphate dehydrogenase (G6PD) associated with a reduction in progression-free survival. Genetic and pharmacologic manipulation demonstrates that G6PD inhibition reduces prostate cancer growth and migration, associated with changes in cellular redox state and increased chemosensitivity. Genetic blockade of G6PD in vivo results in reduction of tumor growth within bone. In summary, we demonstrate the metabolic plasticity of prostate cancer cells in the bone microenvironment, identifying the PPP and G6PD as metabolic targets for the treatment of prostate cancer bone metastasis.


Asunto(s)
Glucosafosfato Deshidrogenasa , Neoplasias de la Próstata , Línea Celular Tumoral , Glucosafosfato Deshidrogenasa/genética , Glucosafosfato Deshidrogenasa/metabolismo , Humanos , Masculino , Metabolómica , Vía de Pentosa Fosfato/fisiología , Neoplasias de la Próstata/genética , Microambiente Tumoral
5.
Nat Commun ; 13(1): 7868, 2022 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-36550101

RESUMEN

Patients with multiple myeloma, an incurable malignancy of plasma cells, frequently develop osteolytic bone lesions that severely impact quality of life and clinical outcomes. Eliglustat, a U.S. Food and Drug Administration-approved glucosylceramide synthase inhibitor, reduced osteoclast-driven bone loss in preclinical in vivo models of myeloma. In combination with zoledronic acid, a bisphosphonate that treats myeloma bone disease, eliglustat provided further protection from bone loss. Autophagic degradation of TRAF3, a key step for osteoclast differentiation, was inhibited by eliglustat as evidenced by TRAF3 lysosomal and cytoplasmic accumulation. Eliglustat blocked autophagy by altering glycosphingolipid composition whilst restoration of missing glycosphingolipids rescued autophagy markers and TRAF3 degradation thus restoring osteoclastogenesis in bone marrow cells from myeloma patients. This work delineates both the mechanism by which glucosylceramide synthase inhibition prevents autophagic degradation of TRAF3 to reduce osteoclastogenesis as well as highlighting the clinical translational potential of eliglustat for the treatment of myeloma bone disease.


Asunto(s)
Enfermedades Óseas , Mieloma Múltiple , Humanos , Mieloma Múltiple/patología , Factor 3 Asociado a Receptor de TNF/metabolismo , Calidad de Vida , Osteoclastos/metabolismo , Enfermedades Óseas/tratamiento farmacológico , Enfermedades Óseas/metabolismo , Autofagia , Glicoesfingolípidos/metabolismo
6.
Best Pract Res Clin Endocrinol Metab ; 35(4): 101541, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34006450

RESUMEN

Multiple myeloma (MM) is a haematological malignancy characterised by the proliferation and accumulation of terminally differentiated abnormal plasma cells in the bone marrow. Patients suffer from bone pain, factures, anaemia, osteolytic lesions and renal failure. Despite recent advancement in therapy MM remains an incurable disease due to the emergences of drug resistance and frequent relapse. For many decades, research has been heavily focused on understanding the relationship between bone cells such as osteoblast, osteocytes and osteoclasts and the infiltrating tumour cells. However, it is now clear that the tumour-supportive bone microenvironment including cellular and non-cellular components play an important role in driving MM progression and bone disease. One of the most abundant cell types in the bone microenvironment is the bone marrow adipocyte (BMAd). Once thought of as inert space filling cells, they have now been recognised as having specialised functions, signalling in an autocrine, paracrine and endocrine manner to support normal systemic homeostasis. BMAds are both an energy store and a source of secreted adipokines and bioactive substances, MM cells are able to hijack this metabolic machinery to fuel migration, growth and survival. With global obesity on the rise, it has never been more important to further understand the contribution these cells have in both normal and disease settings. The aim of this review is to summarise the large body of emerging evidence supporting the interplay between BMAds and MM cells and to delineate how they fit into the vicious cycle of disease.


Asunto(s)
Médula Ósea , Mieloma Múltiple , Adiposidad , Humanos , Recurrencia Local de Neoplasia , Obesidad , Microambiente Tumoral
7.
J Bone Miner Res ; 35(5): 942-955, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31886918

RESUMEN

Multiple myeloma is caused by abnormal plasma cells that accumulate in the bone marrow and interact with resident cells of the bone microenvironment to drive disease progression and development of an osteolytic bone disease. Bone marrow adipocytes (BMAds) are emerging as having important endocrine functions that can support myeloma cell growth and survival. However, how BMAds respond to infiltrating tumor cells remains poorly understood. Using the C57BL/KaLwRij murine model of myeloma, bone marrow adiposity was found to be increased in early stage myeloma with BMAds localizing along the tumor-bone interface at later stages of disease. Myeloma cells were found to uptake BMAd-derived lipids in vitro and in vivo, although lipid uptake was not associated with the ability of BMAds to promote myeloma cell growth and survival. However, BMAd-derived factors were found to increase myeloma cell migration, viability, and the evasion of apoptosis. BMAds are a major source of adiponectin, which is known to be myeloma-suppressive. Myeloma cells were found to downregulate adiponectin specifically in a model of BMAds but not in white adipocytes. The ability of myeloma cells to downregulate adiponectin was dependent at least in part on TNF-α. Collectively our data support the link between increased bone marrow adiposity and myeloma progression. By demonstrating how TNF-α downregulates BMAd-derived adiponectin, we reveal a new mechanism by which myeloma cells alter the bone microenvironment to support disease progression. © 2019 The Authors. Journal of Bone and Mineral Research published by American Society for Bone and Mineral Research.


Asunto(s)
Neoplasias Óseas , Mieloma Múltiple , Adipocitos , Adiponectina , Animales , Médula Ósea , Células de la Médula Ósea , Ratones , Microambiente Tumoral , Factor de Necrosis Tumoral alfa
8.
Bone ; 118: 42-46, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29548987

RESUMEN

Multiple Myeloma (MM) is an incurable haematological malignancy and is the second most common blood cancer in adults; it is caused by the clonal expansion of abnormal plasma cells within the bone marrow and characterized by osteolytic bone lesions, bone pain, renal disease, and immunodeficiency. MM cells infiltrate the bone marrow where they hijack the microenvironment to sustain growth and survival. The contribution to this process by resident bone cells is well defined. However, the role of bone marrow adipocytes is less clear. As one of the most abundant cell types in the bone marrow these cells are surprisingly understudied. However, in the last few decades they have been recognised as having endocrine function. Adipocytes are metabolically active cells that secrete adipokines, growth factors, and inflammatory mediators, they influence the behaviour and function of neighbouring cells; and have the potential to dysregulate normal bone homeostasis. This review discusses how adipocytes contribute to the metastatic niche in multiple myeloma and cancers that metastasise to the bone and how these new discoveries may contribute to further understanding the mechanisms driving the devastating bone disease associated with MM.


Asunto(s)
Adiposidad , Médula Ósea/patología , Mieloma Múltiple/patología , Adipocitos/patología , Animales , Enfermedades Óseas/patología , Humanos , Regeneración
9.
Nat Commun ; 10(1): 4533, 2019 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-31586071

RESUMEN

Multiple myeloma is an incurable, bone marrow-dwelling malignancy that disrupts bone homeostasis causing skeletal damage and pain. Mechanisms underlying myeloma-induced bone destruction are poorly understood and current therapies do not restore lost bone mass. Using transcriptomic profiling of isolated bone lining cell subtypes from a murine myeloma model, we find that bone morphogenetic protein (BMP) signalling is upregulated in stromal progenitor cells. BMP signalling has not previously been reported to be dysregulated in myeloma bone disease. Inhibition of BMP signalling in vivo using either a small molecule BMP receptor antagonist or a solubilized BMPR1a-FC receptor ligand trap prevents trabecular and cortical bone volume loss caused by myeloma, without increasing tumour burden. BMP inhibition directly reduces osteoclastogenesis, increases osteoblasts and bone formation, and suppresses bone marrow sclerostin levels. In summary we describe a novel role for the BMP pathway in myeloma-induced bone disease that can be therapeutically targeted.


Asunto(s)
Enfermedades Óseas/tratamiento farmacológico , Proteínas Morfogenéticas Óseas/metabolismo , Mieloma Múltiple/complicaciones , Pirazoles/farmacología , Pirimidinas/farmacología , Células Madre/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Densidad Ósea/efectos de los fármacos , Enfermedades Óseas/etiología , Enfermedades Óseas/patología , Médula Ósea/patología , Receptores de Proteínas Morfogenéticas Óseas/antagonistas & inhibidores , Receptores de Proteínas Morfogenéticas Óseas/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Fémur/citología , Fémur/efectos de los fármacos , Fémur/patología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Inyecciones Intraperitoneales , Ratones , Ratones Endogámicos , Mieloma Múltiple/patología , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Osteogénesis/efectos de los fármacos , Pirazoles/uso terapéutico , Pirimidinas/uso terapéutico , RNA-Seq , Transducción de Señal/efectos de los fármacos , Células Madre/patología , Tibia/citología , Tibia/efectos de los fármacos , Tibia/patología , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Artículo en Inglés | MEDLINE | ID: mdl-27471491

RESUMEN

The bone marrow is a favored site for a number of cancers, including the hematological malignancy multiple myeloma, and metastasis of breast and prostate cancer. This specialized microenvironment is highly supportive, not only for tumor growth and survival but also for the development of an associated destructive cancer-induced bone disease. The interactions between tumor cells, osteoclasts and osteoblasts are well documented. By contrast, despite occupying a significant proportion of the bone marrow, the importance of bone marrow adipose tissue is only just emerging. The ability of bone marrow adipocytes to regulate skeletal biology and hematopoiesis, combined with their metabolic activity, endocrine functions, and proximity to tumor cells means that they are ideally placed to impact both tumor growth and bone disease. This review discusses the recent advances in our understanding of how marrow adipose tissue contributes to bone metastasis and cancer-induced bone disease.

11.
J Bone Oncol ; 5(3): 121-123, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27761371

RESUMEN

Adipocytes are a significant component of the bone marrow microenvironment. Although bone marrow adipocytes were first identified more than 100 years ago, it is only in recent years that an understanding of their complex physiological role is emerging. Bone marrow adipocytes act as local regulators of skeletal biology and homeostasis, with recent studies suggesting that marrow adipose tissue is metabolically active, and can function as an endocrine organ. As such, bone marrow adipocytes have the potential to interact with tumour cells, influencing both tumour growth and bone disease. This review discusses the current evidence for the role of bone marrow adipocytes in tumour growth within the bone marrow microenvironment and the development of the associated bone disease.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA