Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 143
Filtrar
1.
Curr Osteoporos Rep ; 22(2): 266-272, 2024 04.
Artigo em Inglês | MEDLINE | ID: mdl-38457001

RESUMO

PURPOSE OF REVIEW: To describe the contributions of osteocytes to the lesions in Paget's disease, which are characterized by locally overactive bone resorption and formation. RECENT FINDINGS: Osteocytes, the most abundant cells in bone, are altered in Paget's disease lesions, displaying increased size, decreased canalicular length, incomplete differentiation, and less sclerostin expression compared to controls in both patients and mouse models. Pagetic lesions show increased senescent osteocytes that express RANK ligand, which drives osteoclastic bone resorption. Abnormal osteoclasts in Paget's disease secrete abundant IGF1, which enhances osteocyte senescence, contributing to lesion formation. Recent data suggest that osteocytes contribute to lesion formation in Paget's disease by responding to high local IGF1 released from abnormal osteoclasts. Here we describe the characteristics of osteocytes in Paget's disease and their role in bone lesion formation based on recent results with mouse models and supported by patient data.


Assuntos
Osteíte Deformante , Osteoclastos , Osteócitos , Osteíte Deformante/metabolismo , Osteíte Deformante/patologia , Osteócitos/metabolismo , Osteócitos/patologia , Humanos , Animais , Osteoclastos/metabolismo , Ligante RANK/metabolismo , Reabsorção Óssea/metabolismo , Camundongos , Fator de Crescimento Insulin-Like I/metabolismo , Modelos Animais de Doenças , Senescência Celular
2.
Haematologica ; 2023 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-37981834

RESUMO

Multiple myeloma (MM) is a malignancy of plasma cells whose antibody secretion creates proteotoxic stress relieved by the N-end rule pathway, a proteolytic system that degrades Narginylated proteins in the proteasome. When the proteasome is inhibited, protein cargo is alternatively targeted for autophagic degradation by binding to the ZZ-domain of p62/sequestosome-1. Here, we demonstrate that XRK3F2, a selective ligand for the ZZ-domain, dramatically improved two major responses to the proteasome inhibitor bortezomib by increasing: 1) killing of human MM cells by stimulating both bortezomib mediated apoptosis and necroptosis, a process regulated by p62; and 2) preservation of bone mass by stimulating osteoblasts differentiation and inhibiting osteoclastic bone destruction. Co-administration of bortezomib and XRK3F2 inhibited both branches of the bimodal N-end rule pathway exhibited synergistic anti-MM effects on MM cell lines and CD138+ cells from MM patients, and prevented stromal-mediated MM cell survival. In mice with established human MM, coadministration of bortezomib and XRK3F2 decreased tumor burden and prevented the progression of MM-induced osteolytic disease by inducing new bone formation more effectively than either single agent alone. The results suggest that p62-ZZ ligands enhance the anti-MM efficacy of proteasome inhibitors and can reduce MM morbidity and mortality by improving bone health.

3.
FASEB J ; 36(3): e22196, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35137455

RESUMO

Parathyroid hormone (PTH) signaling downstream of the PTH 1 receptor (Pth1r) results in both bone anabolic and catabolic actions by mechanisms not yet fully understood. In this study, we show that Pth1r signaling upregulates the expression of several components of the Notch pathway and that Notch signals contribute to the catabolic actions of PTH in bone. We found that constitutive genetic activation of PTH receptor signaling in osteocytes (caPth1rOt ) or treatment with PTH daily increased the expression of several Notch ligands/receptors in bone. In contrast, sustained elevation of endogenous PTH did not change Notch components expression. Deletion of the PTH receptor or sclerostin overexpression in osteocytes abolished Notch increases by PTH. Further, deleting the canonical Notch transcription factor Rbpjk in osteocytes decreased bone mass and increased resorption and Rankl expression in caPth1rOt mice. Moreover, pharmacological bone-targeted Notch inhibition potentiated the bone mass gain induced by intermittent PTH by reducing bone resorption and preserving bone formation. Thus, Notch activation lies downstream of anabolic signaling driven by PTH actions in osteocytes, and Notch pharmacological inhibition maximizes the bone anabolic effects of PTH.


Assuntos
Reabsorção Óssea/metabolismo , Osteogênese , Hormônio Paratireóideo/metabolismo , Receptores Notch/metabolismo , Animais , Reabsorção Óssea/genética , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Osteócitos/metabolismo , Receptor Tipo 1 de Hormônio Paratireóideo/metabolismo , Receptores Notch/genética , Transdução de Sinais
4.
Biol Blood Marrow Transplant ; 26(10): e247-e255, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32589921

RESUMO

The Blood and Marrow Transplant Clinical Trials Network (BMT CTN) Myeloma Intergroup has organized an annual workshop focused on minimal residual disease (MRD) testing and immune profiling (IP) in multiple myeloma since 2016. In 2019, the workshop took place as an American Society of Hematology (ASH) Friday Scientific Workshop titled "Immune Profiling and Minimal Residual Disease Testing in Multiple Myeloma." This workshop focused on 4 main topics: the molecular and immunologic evolution of plasma cell disorders, development of new laboratory- and imaging-based MRD assessment approaches, chimeric antigen receptor T cell therapy research, and statistical and regulatory issues associated with novel clinical endpoints. In this report, we provide a summary of the workshop and discuss future directions.


Assuntos
Mieloma Múltiplo , Medula Óssea , Humanos , Mieloma Múltiplo/terapia , Neoplasia Residual
5.
Lancet Oncol ; 19(3): 370-381, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29429912

RESUMO

BACKGROUND: Multiple myeloma is characterised by monoclonal paraprotein production and osteolytic lesions, commonly leading to skeletal-related events (spinal cord compression, pathological fracture, or surgery or radiotherapy to affected bone). Denosumab, a monoclonal antibody targeting RANKL, reduces skeletal-related events associated with bone lesions or metastases in patients with advanced solid tumours. This study aimed to assess the efficacy and safety of denosumab compared with zoledronic acid for the prevention of skeletal-related events in patients with newly diagnosed multiple myeloma. METHODS: In this international, double-blind, double-dummy, randomised, active-controlled, phase 3 study, patients in 259 centres and 29 countries aged 18 years or older with symptomatic newly diagnosed multiple myeloma who had at least one documented lytic bone lesion were randomly assigned (1:1; centrally, by interactive voice response system using a fixed stratified permuted block randomisation list with a block size of four) to subcutaneous denosumab 120 mg plus intravenous placebo every 4 weeks or intravenous zoledronic acid 4 mg plus subcutaneous placebo every 4 weeks (both groups also received investigators' choice of first-line antimyeloma therapy). Stratification was by intent to undergo autologous transplantation, antimyeloma therapy, International Staging System stage, previous skeletal-related events, and region. The clinical study team and patients were masked to treatment assignments. The primary endpoint was non-inferiority of denosumab to zoledronic acid with respect to time to first skeletal-related event in the full analysis set (all randomly assigned patients). All safety endpoints were analysed in the safety analysis set, which includes all randomly assigned patients who received at least one dose of active study drug. This study is registered with ClinicalTrials.gov, number NCT01345019. FINDINGS: From May 17, 2012, to March 29, 2016, we enrolled 1718 patients and randomly assigned 859 to each treatment group. The study met the primary endpoint; denosumab was non-inferior to zoledronic acid for time to first skeletal-related event (hazard ratio 0·98, 95% CI 0·85-1·14; pnon-inferiority=0·010). 1702 patients received at least one dose of the investigational drug and were included in the safety analysis (850 patients receiving denosumab and 852 receiving zoledronic acid). The most common grade 3 or worse treatment-emergent adverse events for denosumab and zoledronic acid were neutropenia (126 [15%] vs 125 [15%]), thrombocytopenia (120 [14%] vs 103 [12%]), anaemia (100 [12%] vs 85 [10%]), febrile neutropenia (96 [11%] vs 87 [10%]), and pneumonia (65 [8%] vs 70 [8%]). Renal toxicity was reported in 85 (10%) patients in the denosumab group versus 146 (17%) in the zoledronic acid group; hypocalcaemia adverse events were reported in 144 (17%) versus 106 (12%). Incidence of osteonecrosis of the jaw was not significantly different between the denosumab and zoledronic acid groups (35 [4%] vs 24 [3%]; p=0·147). The most common serious adverse event for both treatment groups was pneumonia (71 [8%] vs 69 [8%]). One patient in the zoledronic acid group died of cardiac arrest that was deemed treatment-related. INTERPRETATION: In patients with newly diagnosed multiple myeloma, denosumab was non-inferior to zoledronic acid for time to skeletal-related events. The results from this study suggest denosumab could be an additional option for the standard of care for patients with multiple myeloma with bone disease. FUNDING: Amgen.


Assuntos
Antineoplásicos/uso terapêutico , Conservadores da Densidade Óssea/uso terapêutico , Neoplasias Ósseas/prevenção & controle , Denosumab/uso terapêutico , Fraturas Espontâneas/prevenção & controle , Mieloma Múltiplo/tratamento farmacológico , Compressão da Medula Espinal/prevenção & controle , Ácido Zoledrônico/uso terapêutico , Idoso , Antineoplásicos/efeitos adversos , Conservadores da Densidade Óssea/efeitos adversos , Neoplasias Ósseas/complicações , Neoplasias Ósseas/mortalidade , Neoplasias Ósseas/secundário , Denosumab/efeitos adversos , Método Duplo-Cego , Feminino , Fraturas Espontâneas/etiologia , Fraturas Espontâneas/mortalidade , Fraturas Espontâneas/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Mieloma Múltiplo/complicações , Mieloma Múltiplo/mortalidade , Mieloma Múltiplo/patologia , Intervalo Livre de Progressão , Fatores de Risco , Compressão da Medula Espinal/etiologia , Compressão da Medula Espinal/mortalidade , Compressão da Medula Espinal/patologia , Fatores de Tempo , Resultado do Tratamento , Ácido Zoledrônico/efeitos adversos
6.
Mol Carcinog ; 54(12): 1796-806, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25640641

RESUMO

Cannabinoid receptor-2 (CB2) is expressed dominantly in the immune system, especially on plasma cells. Cannabinergic ligands with CB2 selectivity emerge as a class of promising agents to treat CB2-expressing malignancies without psychotropic concerns. In this study, we found that CB2 but not CB1 was highly expressed in human multiple myeloma (MM) and primary CD138+ cells. A novel inverse agonist of CB2, phenylacetylamide but not CB1 inverse agonist SR141716, inhibited the proliferation of human MM cells (IC50 : 0.62 ∼ 2.5 µM) mediated by apoptosis induction, but exhibited minor cytotoxic effects on human normal mononuclear cells. CB2 gene silencing or pharmacological antagonism markedly attenuated phenylacetylamide's anti-MM effects. Phenylacetylamide triggered the expression of C/EBP homologous protein at the early treatment stage, followed by death receptor-5 upregulation, caspase activation, and ß-actin/tubulin degradation. Cell cycle related protein cdc25C and mitotic regulator Aurora A kinase were inactivated by phenylacetylamide treatment, leading to an increase in the ratio inactive/active cdc2 kinase. As a result, phosphorylation of CDK substrates was decreased, and the MM cell mitotic division was largely blocked by treatment. Importantly, phenylacetylamide could overcome the chemoresistance of MM cells against dexamethasone or melphalan. Thus, targeting CB2 may represent an attractive approach to treat cancers of immune origin.


Assuntos
Agonistas de Receptores de Canabinoides/farmacologia , Proliferação de Células/efeitos dos fármacos , Citoesqueleto/efeitos dos fármacos , Mitose/efeitos dos fármacos , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/genética , Receptor CB2 de Canabinoide/genética , Actinas/genética , Apoptose/efeitos dos fármacos , Apoptose/genética , Proteína Quinase CDC2/genética , Caspases/genética , Proteínas de Ciclo Celular/genética , Linhagem Celular Transformada , Proliferação de Células/genética , Citoesqueleto/genética , Inativação Gênica/efeitos dos fármacos , Humanos , Mitose/genética , Receptor CB2 de Canabinoide/agonistas , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Sindecana-1/genética , Tubulina (Proteína)/genética , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
7.
J Biol Chem ; 288(42): 30399-30410, 2013 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-24005670

RESUMO

Abnormal osteoclast formation and osteolysis are the hallmarks of multiple myeloma (MM) bone disease, yet the underlying molecular mechanisms are incompletely understood. Here, we show that the AKT pathway was up-regulated in primary bone marrow monocytes (BMM) from patients with MM, which resulted in sustained high expression of the receptor activator of NF-κB (RANK) in osteoclast precursors. The up-regulation of RANK expression and osteoclast formation in the MM BMM cultures was blocked by AKT inhibition. Conditioned media from MM cell cultures activated AKT and increased RANK expression and osteoclast formation in BMM cultures. Inhibiting AKT in cultured MM cells decreased their growth and ability to promote osteoclast formation. Of clinical significance, systemic administration of the AKT inhibitor LY294002 blocked the formation of tumor tissues in the bone marrow cavity and essentially abolished the MM-induced osteoclast formation and osteolysis in SCID mice. The level of activating transcription factor 4 (ATF4) protein was up-regulated in the BMM cultures from multiple myeloma patients. Adenoviral overexpression of ATF4 activated RANK expression in osteoclast precursors. These results demonstrate a new role of AKT in the MM promotion of osteoclast formation and bone osteolysis through, at least in part, the ATF4-dependent up-regulation of RANK expression in osteoclast precursors.


Assuntos
Regulação Neoplásica da Expressão Gênica , Mieloma Múltiplo/enzimologia , Osteoclastos/enzimologia , Osteólise/enzimologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Regulação para Cima , Fator 4 Ativador da Transcrição/metabolismo , Animais , Cromonas/farmacologia , Inibidores Enzimáticos/farmacologia , Feminino , Xenoenxertos , Humanos , Masculino , Camundongos , Camundongos SCID , Morfolinas/farmacologia , Mieloma Múltiplo/patologia , Transplante de Neoplasias , Osteoclastos/patologia , Osteólise/patologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Células Tumorais Cultivadas
8.
Blood ; 119(18): 4205-14, 2012 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-22427205

RESUMO

BM stromal cells (BMSCs) are key players in the microenvironmental support of multiple myeloma (MM) cell growth and bone destruction. A spliced form of the X-box-binding protein-1 (XBP1s), a major proximal effector of unfolded protein response signaling, is highly expressed in MM cells and plays an indispensable role in MM pathogenesis. In the present study, we found that XBP1s is induced in the BMSCs of the MM microenvironment. XBP1s overexpression in healthy human BMSCs enhanced gene and/or protein expression of VCAM-1, IL-6, and receptor activator of NF-κB ligand (RANKL), enhancing BMSC support of MM cell growth and osteoclast formation in vitro and in vivo. Conversely, deficiency of XBP1 in healthy donor BMSCs displayed a range of effects on BMSCs that were opposite to those cells with overexpression of XBP1s. Knock-down of XBP1 in MM patient BMSCs greatly compromised their increased VCAM-1 protein expression and IL-6 and RANKL secretion in response to TNFα and reversed their enhanced support of MM-cell growth and osteoclast formation. Our results demonstrate that XBP1s is a pathogenic factor underlying BMSC support of MM cell growth and osteoclast formation and therefore represents a therapeutic target for MM bone disease.


Assuntos
Células da Medula Óssea/metabolismo , Proteínas de Ligação a DNA/fisiologia , Mieloma Múltiplo/patologia , Proteínas de Neoplasias/fisiologia , Osteoclastos/patologia , Células Estromais/metabolismo , Fatores de Transcrição/fisiologia , Animais , Células da Medula Óssea/patologia , Diferenciação Celular , Divisão Celular , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , Técnicas de Cocultura , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Endorribonucleases/deficiência , Endorribonucleases/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Inflamação , Interleucina-6/biossíntese , Interleucina-6/genética , Interleucina-6/metabolismo , Camundongos , Camundongos SCID , Mieloma Múltiplo/metabolismo , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Ligante RANK/biossíntese , Ligante RANK/genética , Ligante RANK/metabolismo , Interferência de RNA , Proteínas Recombinantes de Fusão/fisiologia , Fatores de Transcrição de Fator Regulador X , Células Estromais/patologia , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Microambiente Tumoral , Fator de Necrose Tumoral alfa/farmacologia , Molécula 1 de Adesão de Célula Vascular/biossíntese , Molécula 1 de Adesão de Célula Vascular/genética , Proteína 1 de Ligação a X-Box
9.
Blood ; 119(8): 1888-96, 2012 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-22223826

RESUMO

Multiple myeloma (MM) is an incurable B-cell malignancy in which the marrow microenvironment plays a critical role in our inability to cure MM. Marrow stromal cells in the microenvironment support homing, lodging, and growth of MM cells through activation of multiple signaling pathways in both MM and stromal cells. Recently, we identified annexin II (AXII) as a previously unknown factor produced by stromal cells and osteoclasts (OCL) that is involved in OCL formation, HSC and prostate cancer (PCa) homing to the BM as well as mobilization of HSC and PCa cells. AXII expressed on stromal cells supports PCa cell lodgment via the AXII receptor (AXIIR) on PCa cells, but the role of AXII and AXIIR in MM is unknown. In this study, we show that MM cells express AXIIR, that stromal/osteoblast-derived AXII facilitates adhesion of MM cells to stromal cells via AXIIR, and OCL-derived AXII enhances MM cell growth. Finally, we demonstrate that AXII activates the ERK1/2 and AKT pathways in MM cells to enhance MM cell growth. These results demonstrate that AXII and AXIIR play important roles in MM and that targeting the AXII/AXIIR axis may be a novel therapeutic approach for MM.


Assuntos
Anexina A2/metabolismo , Medula Óssea/metabolismo , Proliferação de Células , Mieloma Múltiplo/metabolismo , Receptores de Peptídeos/metabolismo , Animais , Anexina A2/genética , Anexina A2/farmacologia , Western Blotting , Células da Medula Óssea/metabolismo , Adesão Celular , Linhagem Celular Tumoral , Células Cultivadas , Microambiente Celular , Técnicas de Cocultura , Humanos , Camundongos , Camundongos Knockout , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Mieloma Múltiplo/genética , Mieloma Múltiplo/patologia , Osteoclastos/metabolismo , Fosforilação/efeitos dos fármacos , Ligação Proteica , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , Receptores de Peptídeos/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais
10.
Blood ; 119(20): 4608-13, 2012 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-22451423

RESUMO

This multicenter phase 1/2 trial investigated the combination of bendamustine, lenalidomide, and dexamethasone in repeating 4-week cycles as treatment for relapsed refractory multiple myeloma (MM). Phase 1 established maximum tolerated dose (MTD). Phase 2 assessed overall response rate at the MTD. Secondary endpoints included progression-free survival (PFS) and overall survival (OS). A total of 29 evaluable patients were enrolled. Median age was 63 years (range, 38-80 years). Median number of prior therapies was 3 (range, 1-6). MTD was bendamustine 75 mg/m(2) (days 1 and 2), lenalidomide 10 mg (days 1-21), and dexamethasone 40 mg (weekly) of a 28-day cycle. Partial response rate was 52%, with very good partial response achieved in 24%, and minimal response in an additional 24% of patients. Median follow-up was 13 months; median OS has not been reached. One-year OS is 93% (95% confidence interval [CI], 59%-99%). Median PFS is 6.1 months (95% CI, 3.7-9.4 months) with one-year PFS of 20% (95% CI, 6%-41%). Grade 3/4 adverse events included neutropenia, thrombocytopenia, anemia, hyperglycemia, and fatigue. This first phase 1/2 trial testing bendamustine, lenalidomide, and dexamethasone as treatment of relapsed refractory MM was feasible and highly active. This study is registered at www.clinicaltrials.gov as #NCT01042704.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Dexametasona/administração & dosagem , Mieloma Múltiplo/tratamento farmacológico , Compostos de Mostarda Nitrogenada/administração & dosagem , Talidomida/análogos & derivados , Adulto , Idoso , Idoso de 80 Anos ou mais , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Cloridrato de Bendamustina , Dexametasona/efeitos adversos , Relação Dose-Resposta a Droga , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Estudos de Viabilidade , Feminino , Humanos , Lenalidomida , Masculino , Pessoa de Meia-Idade , Mieloma Múltiplo/diagnóstico , Mieloma Múltiplo/mortalidade , Mieloma Múltiplo/patologia , Compostos de Mostarda Nitrogenada/efeitos adversos , Recidiva , Talidomida/administração & dosagem , Talidomida/efeitos adversos , Resultado do Tratamento
11.
J Natl Compr Canc Netw ; 12(4): 502-11, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24717569

RESUMO

As patients with multiple myeloma live longer, helping them live better through improvements in supportive care is equally as vital. Intrinsic to the disease are bone-related complications at presentation and over the course of illness. Bisphosphonates have been an important therapy for ameliorating the risk of skeletal-related events, and work is ongoing to determine their optimal schedule. Patients with multiple myeloma also encounter several challenges related to their treatment, including peripheral neuropathy, thrombotic complications, and infections. These challenges are being addressed through a better understanding of the risk factors for developing these complications and by mitigating these risks through better dosing schemas and prophylaxis.


Assuntos
Mieloma Múltiplo/complicações , Mieloma Múltiplo/terapia , Cuidados Paliativos/métodos , Humanos
12.
Cancer Metastasis Rev ; 31(3-4): 569-78, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22706844

RESUMO

Bone is one of the most frequent sites of metastasis in patients with malignancies. Up to 90 % of patients with multiple myeloma, and 60 % to 75 % patients with prostate cancer and breast cancer develop bone metastasis at the later stages of their diseases. Bone metastases are responsible for tremendous morbidity in patients with cancer, including severe bone pain, pathologic fractures, spinal cord and nerve compression syndromes, life-threatening hypercalcemia, and increased mortality. Multiple factors produced by tumor cells or produced by the bone marrow microenvironment in response to tumor cells play important roles in activation of osteoclastic bone resorption and modulation of osteoblastic activity in patients with bone metastasis. In this chapter, we will review the genes that play important roles in bone destruction, tumor growth, and osteoblast activity in bone metastasis and discuss the potential therapies targeting the products of these genes to block both bone destruction and tumor growth.


Assuntos
Neoplasias Ósseas/secundário , Ativinas/fisiologia , Animais , Anexina A2/fisiologia , Neoplasias Ósseas/terapia , Quimiocina CCL3/fisiologia , Humanos , Interleucina-6/fisiologia , Osteoblastos/fisiologia , Osteólise , Osteoprotegerina/fisiologia , Proteína Relacionada ao Hormônio Paratireóideo/fisiologia , Ligante RANK/fisiologia
13.
Blood ; 118(26): 6871-80, 2011 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-22042697

RESUMO

Protracted inhibition of osteoblast (OB) differentiation characterizes multiple myeloma (MM) bone disease and persists even when patients are in long-term remission. However, the underlying pathophysiology for this prolonged OB suppression is unknown. Therefore, we developed a mouse MM model in which the bone marrow stromal cells (BMSCs) remained unresponsive to OB differentiation signals after removal of MM cells. We found that BMSCs from both MM-bearing mice and MM patients had increased levels of the transcriptional repressor Gfi1 compared with controls and that Gfi1 was a novel transcriptional repressor of the critical OB transcription factor Runx2. Trichostatin-A blocked the effects of Gfi1, suggesting that it induces epigenetic changes in the Runx2 promoter. MM-BMSC cell-cell contact was not required for MM cells to increase Gfi1 and repress Runx2 levels in MC-4 before OBs or naive primary BMSCs, and Gfi1 induction was blocked by anti-TNF-α and anti-IL-7 antibodies. Importantly, BMSCs isolated from Gfi1(-/-) mice were significantly resistant to MM-induced OB suppression. Strikingly, siRNA knockdown of Gfi1 in BMSCs from MM patients significantly restored expression of Runx2 and OB differentiation markers. Thus, Gfi1 may have an important role in prolonged MM-induced OB suppression and provide a new therapeutic target for MM bone disease.


Assuntos
Neoplasias Ósseas/metabolismo , Proteínas de Ligação a DNA/metabolismo , Mieloma Múltiplo/metabolismo , Osteoblastos/metabolismo , Células Estromais/metabolismo , Fatores de Transcrição/metabolismo , Células 3T3 , Animais , Western Blotting , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Neoplasias Ósseas/genética , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Proteínas de Ligação a DNA/genética , Feminino , Expressão Gênica , Humanos , Interleucina-7/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Mieloma Múltiplo/genética , Mieloma Múltiplo/patologia , Osteoblastos/patologia , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Estromais/patologia , Fatores de Transcrição/genética , Fator de Necrose Tumoral alfa/metabolismo
14.
Res Sq ; 2023 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-37461623

RESUMO

Cancers showing excessive innervation of sensory neurons (SN) in their microenvironments are associated with poor outcomes due to promoted growth, increased tumor recurrence, metastasis, and cancer pain, suggesting SNs play a regulatory role in cancer aggressiveness. Using a preclinical model in which mouse 4T1 breast cancer (BC) cells were injected into the bone marrow of tibiae, we found 4T1 BC cells aggressively colonized bone with bone destruction and subsequently spread to the lung. Of note, 4T1 BC colonization induced the acidic tumor microenvironment in bone in which SNs showed increased innervation and excitation with elevated expression of the acid-sensing nociceptor transient receptor potential vanilloid-1 (TRPV1), eliciting bone pain (BP) assessed by mechanical hypersensitivity. Further, these excited SNs produced increased hepatocyte growth factor (HGF). Importantly, the administration of synthetic and natural TRPV1 antagonists and genetic deletion of TRPV1 decreased HGF production in SNs and inhibited 4T1 BC colonization in bone, pulmonary metastasis from bone, and BP induction. Our results suggest the TRPV1 of SNs promotes BC colonization in bone and lung metastasis via up-regulating HGF production in SNs. The SN TRPV1 may be a novel therapeutic target for BC growing in the acidic bone microenvironment and for BP.

15.
JCI Insight ; 8(14)2023 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-37338990

RESUMO

We previously reported that measles virus nucleocapsid protein (MVNP) expression in osteoclasts (OCLs) of patients with Paget disease (PD) or targeted to the OCL lineage in MVNP-transgenic mice (MVNP mice) increases IGF1 production in osteoclasts (OCL-IGF1) and leads to development of PD OCLs and pagetic bone lesions (PDLs). Conditional deletion of Igf1 in OCLs of MVNP mice fully blocked development of PDLs. In this study, we examined whether osteocytes (OCys), key regulators of normal bone remodeling, contribute to PD. OCys in PDLs of patients and of MVNP mice expressed less sclerostin, and had increased RANKL expression compared with OCys in bones from WT mice or normal patients. To test whether increased OCL-IGF1 is sufficient to induce PDLs and PD phenotypes, we generated TRAP-Igf1 (T-Igf1) transgenic mice to determine whether increased IGF1 expression in the absence of MVNP in OCLs is sufficient to induce PDLs and pagetic OCLs. We found that T-Igf1 mice at 16 months of age developed PD OCLs, PDLs, and OCys, with decreased sclerostin and increased RANKL, similar to MVNP mice. Thus, pagetic phenotypes could be induced by OCLs expressing increased IGF1. OCL-IGF1 in turn increased RANKL production in OCys to induce PD OCLs and PDLs.


Assuntos
Osteíte Deformante , Osteoclastos , Animais , Camundongos , Osso e Ossos/metabolismo , Expressão Gênica , Camundongos Transgênicos , Osteíte Deformante/metabolismo , Osteoclastos/metabolismo , Osteócitos/metabolismo
16.
Blood ; 116(18): 3554-63, 2010 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-20664052

RESUMO

Osteoclast (OC)-mediated lytic bone disease remains a cause of major morbidity in multiple myeloma. Here we demonstrate the critical role of interleukin-17-producing marrow infiltrating lymphocytes (MILs) in OC activation and development of bone lesions in myeloma patients. Unlike MILs from normal bone marrow, myeloma MILs possess few regulatory T cells (Tregs) and demonstrate an interleukin-17 phenotype that enhances OC activation. In univariate analyses of factors mediating bone destruction, levels of cytokines that selectively induce and maintain the Th17 phenotype tightly correlated with the extent of bone disease in myeloma. In contrast, MILs activated under conditions that skew toward a Th1 phenotype significantly reduced formation of mature OC. These findings demonstrate that interleukin-17 T cells are critical to the genesis of myeloma bone disease and that immunologic manipulations shifting MILs from a Th17 to a Th1 phenotype may profoundly diminish lytic bone lesions in multiple myeloma.


Assuntos
Doenças da Medula Óssea/etiologia , Medula Óssea/patologia , Interleucina-17/imunologia , Linfócitos/patologia , Mieloma Múltiplo/complicações , Osteoclastos/patologia , Medula Óssea/imunologia , Células Cultivadas , Citocinas/imunologia , Humanos , Linfócitos/imunologia , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/patologia , Osteoclastos/imunologia , Linfócitos T/patologia , Células Th17/imunologia , Células Tumorais Cultivadas
17.
Blood ; 116(18): 3635-44, 2010 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-20679527

RESUMO

Hepcidin is the principal iron-regulatory hormone and a pathogenic factor in anemia of inflammation. Patients with multiple myeloma (MM) frequently present with anemia. We showed that MM patients had increased serum hepcidin, which inversely correlated with hemoglobin, suggesting that hepcidin contributes to MM-related anemia. Searching for hepcidin-inducing cytokines in MM, we quantified the stimulation of hepcidin promoter-luciferase activity in HuH7 cells by MM sera. MM sera activated the hepcidin promoter significantly more than did normal sera. We then examined the role of bone morphogenetic proteins (BMPs) and interleukin-6 (IL-6), the major transcriptional regulators of hepcidin. Mutations in both BMP-responsive elements abrogated the activation dramatically, while mutations in the IL-6-responsive signal transducer and activator of transcription 3-binding site (STAT3-BS) had only a minor effect. Cotreatment with anti-BMP-2/4 or noggin-Fc blocked the promoter induction with all MM sera, anti-IL-6 blocked it with a minority of sera, whereas anti-BMP-4, -6, or -9 antibodies had no effect. BMP-2-immunodepleted MM sera had decreased promoter stimulatory capacity, and BMP-2 concentrations in MM sera were significantly higher than in normal sera. Our results demonstrate that BMP-2 is a major mediator of the hepcidin stimulatory activity of MM sera.


Assuntos
Anemia/complicações , Anemia/imunologia , Peptídeos Catiônicos Antimicrobianos/sangue , Proteína Morfogenética Óssea 2/imunologia , Mieloma Múltiplo/complicações , Mieloma Múltiplo/imunologia , Peptídeos Catiônicos Antimicrobianos/genética , Peptídeos Catiônicos Antimicrobianos/imunologia , Proteína Morfogenética Óssea 2/sangue , Proteínas Morfogenéticas Ósseas/imunologia , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Hemoglobinas/metabolismo , Hepcidinas , Humanos , Interleucina-6/imunologia , Mutação , Regiões Promotoras Genéticas , Fator de Transcrição STAT3/genética
18.
Blood ; 115(3): 605-14, 2010 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-19965623

RESUMO

The immunomodulatory drugs (IMiDs) lenalidomide and pomalidomide yield high response rates in patients with multiple myeloma, but the use of IMiDs in multiple myeloma is associated with neutropenia and increased risk for venous thromboembolism (VTE) by mechanisms that are unknown. We show that IMiDs down-regulate PU.1, a key transcription factor involved in granulocyte differentiation in vitro and in patients treated with lenalidomide. Loss of PU.1 results in transient maturation arrest with medullary accumulation of immature myeloid precursors and subsequent neutropenia. Accumulation of promyelocytes leads to high levels of the platelet aggregation agonist, cathepsin G stored in the azurophilic granules of promyelocytes. High levels of cathepsin G subsequently may increase the risk of VTE. To our knowledge, this is the first report investigating the underlying mechanism of IMiD-induced neutropenia and increased risk of VTE in multiple myeloma.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Fatores Imunológicos/farmacologia , Células Progenitoras Mieloides/efeitos dos fármacos , Neutropenia/induzido quimicamente , Proteínas Proto-Oncogênicas/genética , Transativadores/genética , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Diferenciação Celular/imunologia , Células Cultivadas , Regulação para Baixo , Fator Estimulador de Colônias de Granulócitos/farmacologia , Células Precursoras de Granulócitos/efeitos dos fármacos , Células Precursoras de Granulócitos/metabolismo , Células Precursoras de Granulócitos/fisiologia , Granulócitos/efeitos dos fármacos , Granulócitos/fisiologia , Humanos , Fatores Imunológicos/efeitos adversos , Fatores Imunológicos/uso terapêutico , Lenalidomida , Modelos Biológicos , Mieloma Múltiplo/sangue , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/patologia , Células Progenitoras Mieloides/metabolismo , Células Progenitoras Mieloides/fisiologia , Neutropenia/imunologia , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Neutrófilos/fisiologia , Agregação Plaquetária/efeitos dos fármacos , Proteínas Proto-Oncogênicas/metabolismo , Fatores de Risco , Talidomida/análogos & derivados , Talidomida/farmacologia , Transativadores/metabolismo , Tromboembolia Venosa/etiologia
19.
Blood ; 115(1): 140-9, 2010 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-19887675

RESUMO

Increased osteoclastogenesis and angiogenesis occur in physiologic and pathologic conditions. However, it is unclear if or how these processes are linked. To test the hypothesis that osteoclasts stimulate angiogenesis, we modulated osteoclast formation in fetal mouse metatarsal explants or in adult mice and determined the effect on angiogenesis. Suppression of osteoclast formation with osteoprotegerin dose-dependently inhibited angiogenesis and osteoclastogenesis in metatarsal explants. Conversely, treatment with parathyroid hormone related protein (PTHrP) increased explant angiogenesis, which was completely blocked by osteoprotegerin. Further, treatment of mice with receptor activator of nuclear factor-kappaB ligand (RANKL) or PTHrP in vivo increased calvarial vessel density and osteoclast number. We next determined whether matrix metalloproteinase-9 (MMP-9), an angiogenic factor predominantly produced by osteoclasts in bone, was important for osteoclast-stimulated angiogenesis. The pro-angiogenic effects of PTHrP or RANKL were absent in metatarsal explants or calvaria in vivo, respectively, from Mmp9(-/-) mice, demonstrating the importance of MMP-9 for osteoclast-stimulated angiogenesis. Lack of MMP-9 decreased osteoclast numbers and abrogated angiogenesis in response to PTHrP or RANKL in explants and in vivo but did not decrease osteoclast differentiation in vitro. Thus, MMP-9 modulates osteoclast-stimulated angiogenesis primarily by affecting osteoclasts, most probably by previously reported migratory effects on osteoclasts. These results clearly demonstrate that osteoclasts stimulate angiogenesis in vivo through MMP-9.


Assuntos
Ossos do Metatarso/irrigação sanguínea , Neovascularização Fisiológica , Osteoclastos/fisiologia , Indutores da Angiogênese/metabolismo , Animais , Feminino , Feto/irrigação sanguínea , Feto/efeitos dos fármacos , Humanos , Masculino , Metaloproteinase 9 da Matriz/deficiência , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Ossos do Metatarso/efeitos dos fármacos , Ossos do Metatarso/embriologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Neovascularização Fisiológica/efeitos dos fármacos , Osteoclastos/efeitos dos fármacos , Osteoclastos/enzimologia , Proteína Relacionada ao Hormônio Paratireóideo/farmacologia , Ligante RANK/farmacologia , Crânio/citologia , Crânio/efeitos dos fármacos , Crânio/enzimologia , Regulação para Cima/efeitos dos fármacos
20.
Cancers (Basel) ; 14(3)2022 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-35159039

RESUMO

Multiple myeloma (MM) remains incurable for most patients due to the emergence of drug resistant clones. Here we report a p53-independent mechanism responsible for Growth Factor Independence-1 (GFI1) support of MM cell survival by its modulation of sphingolipid metabolism to increase the sphingosine-1-phosphate (S1P) level regardless of the p53 status. We found that expression of enzymes that control S1P biosynthesis, SphK1, dephosphorylation, and SGPP1 were differentially correlated with GFI1 levels in MM cells. We detected GFI1 occupancy on the SGGP1 gene in MM cells in a predicted enhancer region at the 5' end of intron 1, which correlated with decreased SGGP1 expression and increased S1P levels in GFI1 overexpressing cells, regardless of their p53 status. The high S1P:Ceramide intracellular ratio in MM cells protected c-Myc protein stability in a PP2A-dependent manner. The decreased MM viability by SphK1 inhibition was dependent on the induction of autophagy in both p53WT and p53mut MM. An autophagic blockade prevented GFI1 support for viability only in p53mut MM, demonstrating that GFI1 increases MM cell survival via both p53WT inhibition and upregulation of S1P independently. Therefore, GFI1 may be a key therapeutic target for all types of MM that may significantly benefit patients that are highly resistant to current therapies.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA