Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 117
Filtrar
1.
Breast Cancer Res ; 26(1): 34, 2024 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-38409028

RESUMO

The role of parathyroid hormone (PTH)-related protein (PTHrP) in breast cancer remains controversial, with reports of PTHrP inhibiting or promoting primary tumor growth in preclinical studies. Here, we provide insight into these conflicting findings by assessing the role of specific biological domains of PTHrP in tumor progression through stable expression of PTHrP (-36-139aa) or truncated forms with deletion of the nuclear localization sequence (NLS) alone or in combination with the C-terminus. Although the full-length PTHrP molecule (-36-139aa) did not alter tumorigenesis, PTHrP lacking the NLS alone accelerated primary tumor growth by downregulating p27, while PTHrP lacking the NLS and C-terminus repressed tumor growth through p27 induction driven by the tumor suppressor leukemia inhibitory factor receptor (LIFR). Induction of p27 by PTHrP lacking the NLS and C-terminus persisted in bone disseminated cells, but did not prevent metastatic outgrowth, in contrast to the primary tumor site. These data suggest that the PTHrP NLS functions as a tumor suppressor, while the PTHrP C-terminus may act as an oncogenic switch to promote tumor progression through differential regulation of p27 signaling.


Assuntos
Neoplasias da Mama , Proteína Relacionada ao Hormônio Paratireóideo , Humanos , Feminino , Proteína Relacionada ao Hormônio Paratireóideo/genética , Proteína Relacionada ao Hormônio Paratireóideo/metabolismo , Neoplasias da Mama/patologia , Receptores de OSM-LIF , Sinais de Localização Nuclear , Proliferação de Células/genética , Subunidade alfa de Receptor de Fator Inibidor de Leucemia
2.
Annu Rev Physiol ; 82: 507-529, 2020 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-31553686

RESUMO

Bone remodeling is essential for the repair and replacement of damaged and old bone. The major principle underlying this process is that osteoclast-mediated resorption of a quantum of bone is followed by osteoblast precursor recruitment; these cells differentiate to matrix-producing osteoblasts, which form new bone to replace what was resorbed. Evidence from osteopetrotic syndromes indicate that osteoclasts not only resorb bone, but also provide signals to promote bone formation. Osteoclasts act upon osteoblast lineage cells throughout their differentiation by facilitating growth factor release from resorbed matrix, producing secreted proteins and microvesicles, and expressing membrane-bound factors. These multiple mechanisms mediate the coupling of bone formation to resorption in remodeling. Additional interactions of osteoclasts with osteoblast lineage cells, including interactions with canopy and reversal cells, are required to achieve coordination between bone formation and resorption during bone remodeling.


Assuntos
Osteoblastos/fisiologia , Osteoclastos/fisiologia , Transdução de Sinais/fisiologia , Animais , Remodelação Óssea/fisiologia , Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Exossomos/fisiologia , Humanos , Osteogênese
3.
Physiol Rev ; 96(3): 831-71, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27142453

RESUMO

Although parathyroid hormone-related protein (PTHrP) was discovered as a cancer-derived hormone, it has been revealed as an important paracrine/autocrine regulator in many tissues, where its effects are context dependent. Thus its location and action in the vasculature explained decades-long observations that injection of PTH into animals rapidly lowered blood pressure by producing vasodilatation. Its roles have been specified in development and maturity in cartilage and bone as a crucial regulator of endochondral bone formation and bone remodeling, respectively. Although it shares actions with parathyroid hormone (PTH) through the use of their common receptor, PTHR1, PTHrP has other actions mediated by regions within the molecule beyond the amino-terminal sequence that resembles PTH, including the ability to promote placental transfer of calcium from mother to fetus. A striking feature of the physiology of PTHrP is that it possesses structural features that equip it to be transported in and out of the nucleus, and makes use of a specific nuclear import mechanism to do so. Evidence from mouse genetic experiments shows that PTHrP generated locally in bone is essential for normal bone remodeling. Whereas the main physiological function of PTH is the hormonal regulation of calcium metabolism, locally generated PTHrP is the important physiological mediator of bone remodeling postnatally. Thus the use of intermittent injection of PTH as an anabolic therapy for bone appears to be a pharmacological application of the physiological function of PTHrP. There is much current interest in the possibility of developing PTHrP analogs that might enhance the therapeutic anabolic effects.


Assuntos
Desenvolvimento Ósseo/fisiologia , Doenças Ósseas/tratamento farmacológico , Cartilagem/metabolismo , Proteína Relacionada ao Hormônio Paratireóideo/fisiologia , Hormônio Paratireóideo/uso terapêutico , Animais , Cartilagem/crescimento & desenvolvimento , Humanos , Camundongos , Hormônio Paratireóideo/fisiologia
4.
J Biol Chem ; 294(19): 7850-7863, 2019 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-30923130

RESUMO

Interleukin 6 (IL-6) supports development of bone-resorbing osteoclasts by acting early in the osteoblast lineage via membrane-bound (cis) or soluble (trans) receptors. Here, we investigated how IL-6 signals and modifies gene expression in differentiated osteoblasts and osteocytes and determined whether these activities can promote bone formation or support osteoclastogenesis. Moreover, we used a genetically altered mouse with circulating levels of the pharmacological IL-6 trans-signaling inhibitor sgp130-Fc to determine whether IL-6 trans-signaling is required for normal bone growth and remodeling. We found that IL-6 increases suppressor of cytokine signaling 3 (Socs3) and CCAAT enhancer-binding protein δ (Cebpd) mRNA levels and promotes signal transducer and activator of transcription 3 (STAT3) phosphorylation by both cis- and trans-signaling in cultured osteocytes. In contrast, RANKL (Tnfsf11) mRNA levels were elevated only by trans-signaling. Furthermore, we observed soluble IL-6 receptor release and ADAM metallopeptidase domain 17 (ADAM17) sheddase expression by osteocytes. Despite the observation that IL-6 cis-signaling occurs, IL-6 stimulated bone formation in vivo only via trans-signaling. Although IL-6 stimulated RANKL (Tnfsf11) mRNA in osteocytes, these cells did not support osteoclast formation in response to IL-6 alone; binucleated TRAP+ cells formed, and only in response to trans-signaling. Finally, pharmacological, sgp130-Fc-mediated inhibition of IL-6 trans-signaling did not impair bone growth or remodeling unless mice had circulating sgp130-Fc levels > 10 µg/ml. At those levels, osteopenia and impaired bone growth occurred, reducing bone strength. We conclude that high sgp130-Fc levels may have detrimental off-target effects on the skeleton.


Assuntos
Receptor gp130 de Citocina/metabolismo , Interleucina-6/metabolismo , Osteoclastos/metabolismo , Osteócitos/metabolismo , Osteogênese , Transdução de Sinais , Proteína ADAM17/metabolismo , Animais , Proteína delta de Ligação ao Facilitador CCAAT/metabolismo , Camundongos , Ligante RANK/metabolismo , Proteína 3 Supressora da Sinalização de Citocinas/metabolismo
5.
Development ; 143(4): 648-57, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26755702

RESUMO

The majority of the skeleton arises by endochondral ossification, whereby cartilaginous templates expand and are resorbed by osteoclasts then replaced by osteoblastic bone formation. Ephrin B2 is a receptor tyrosine kinase expressed by osteoblasts and growth plate chondrocytes that promotes osteoblast differentiation and inhibits osteoclast formation. We investigated the role of ephrin B2 in endochondral ossification using Osx1Cre-targeted gene deletion. Neonatal Osx1Cre.Efnb2(Δ/Δ) mice exhibited a transient osteopetrosis demonstrated by increased trabecular bone volume with a high content of growth plate cartilage remnants and increased cortical thickness, but normal osteoclast numbers within the primary spongiosa. Osteoclasts at the growth plate had an abnormal morphology and expressed low levels of tartrate-resistant acid phosphatase; this was not observed in more mature bone. Electron microscopy revealed a lack of sealing zones and poor attachment of Osx1Cre.Efnb2(Δ/Δ) osteoclasts to growth plate cartilage. Osteoblasts at the growth plate were also poorly attached and impaired in their ability to deposit osteoid. By 6 months of age, trabecular bone mass, osteoclast morphology and osteoid deposition by Osx1Cre.Efnb2(Δ/Δ) osteoblasts were normal. Cultured chondrocytes from Osx1Cre.Efnb2(Δ/Δ) neonates showed impaired support of osteoclastogenesis but no significant change in Rankl (Tnfsf11) levels, whereas Adamts4 levels were significantly reduced. A population of ADAMTS4(+) early hypertrophic chondrocytes seen in controls was absent from Osx1Cre.Efnb2(Δ/Δ) neonates. This suggests that Osx1Cre-expressing cells, including hypertrophic chondrocytes, are dependent on ephrin B2 for their production of cartilage-degrading enzymes, including ADAMTS4, and this might be required for attachment of osteoclasts and osteoblasts to the cartilage surface during endochondral ossification.


Assuntos
Cartilagem/patologia , Condrócitos/metabolismo , Efrina-B2/metabolismo , Osteoclastos/patologia , Osteogênese , Proteínas ADAM/metabolismo , Proteína ADAMTS4 , Animais , Animais Recém-Nascidos , Cartilagem/metabolismo , Adesão Celular , Diferenciação Celular , Condrócitos/patologia , Feminino , Regulação da Expressão Gênica , Imuno-Histoquímica , Integrases/metabolismo , Camundongos Endogâmicos C57BL , Modelos Biológicos , Tamanho do Órgão , Osteoblastos/patologia , Osteoclastos/metabolismo , Osteoclastos/ultraestrutura , Osteogênese/genética , Osteopetrose/genética , Osteopetrose/patologia , Fenótipo , Pró-Colágeno N-Endopeptidase/metabolismo , Tíbia/metabolismo , Tíbia/patologia
6.
PLoS Genet ; 11(4): e1005160, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25859855

RESUMO

RECQL4 mutations are associated with Rothmund Thomson Syndrome (RTS), RAPADILINO Syndrome and Baller-Gerold Syndrome. These patients display a range of benign skeletal abnormalities such as low bone mass. In addition, RTS patients have a highly increased incidence of osteosarcoma (OS). The role of RECQL4 in normal adult bone development and homeostasis is largely uncharacterized and how mutation of RECQL4 contributes to OS susceptibility is not known. We hypothesised that Recql4 was required for normal skeletal development and both benign and malignant osteoblast function, which we have tested in the mouse. Recql4 deletion in vivo at the osteoblastic progenitor stage of differentiation resulted in mice with shorter bones and reduced bone volume, assessed at 9 weeks of age. This was associated with an osteoblast intrinsic decrease in mineral apposition rate and bone formation rate in the Recql4-deficient cohorts. Deletion of Recql4 in mature osteoblasts/osteocytes in vivo, however, did not cause a detectable phenotype. Acute deletion of Recql4 in primary osteoblasts or shRNA knockdown in an osteoblastic cell line caused failed proliferation, accompanied by cell cycle arrest, induction of apoptosis and impaired differentiation. When cohorts of animals were aged long term, the loss of Recql4 alone was not sufficient to initiate OS. We then crossed the Recql4fl/fl allele to a fully penetrant OS model (Osx-Cre p53fl/fl). Unexpectedly, the Osx-Cre p53fl/flRecql4fl/fl (dKO) animals had a significantly increased OS-free survival compared to Osx-Cre p53fl/fl or Osx-Cre p53fl/flRecql4fl/+ (het) animals. The extended survival was explained when the Recql4 status in the tumors that arose was assessed, and in no case was there complete deletion of Recql4 in the dKO OS. These data provide a mechanism for the benign skeletal phenotypes of RECQL4 mutation syndromes. We propose that tumor suppression and osteosarcoma susceptibility are most likely a function of mutant, not null, alleles of RECQL4.


Assuntos
Neoplasias Ósseas/genética , Osteoblastos/metabolismo , Osteogênese , Osteossarcoma/genética , RecQ Helicases/metabolismo , Animais , Neoplasias Ósseas/metabolismo , Proliferação de Células , Deleção de Genes , Camundongos , Camundongos Endogâmicos C57BL , Osteossarcoma/metabolismo , RecQ Helicases/genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
7.
Nature ; 465(7299): 798-802, 2010 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-20383121

RESUMO

The ovarian hormones oestrogen and progesterone profoundly influence breast cancer risk, underpinning the benefit of endocrine therapies in the treatment of breast cancer. Modulation of their effects through ovarian ablation or chemoprevention strategies also significantly decreases breast cancer incidence. Conversely, there is an increased risk of breast cancer associated with pregnancy in the short term. The cellular mechanisms underlying these observations, however, are poorly defined. Here we demonstrate that mouse mammary stem cells (MaSCs) are highly responsive to steroid hormone signalling, despite lacking the oestrogen and progesterone receptors. Ovariectomy markedly diminished MaSC number and outgrowth potential in vivo, whereas MaSC activity increased in mice treated with oestrogen plus progesterone. Notably, even three weeks of treatment with the aromatase inhibitor letrozole was sufficient to reduce the MaSC pool. In contrast, pregnancy led to a transient 11-fold increase in MaSC numbers, probably mediated through paracrine signalling from RANK ligand. The augmented MaSC pool indicates a cellular basis for the short-term increase in breast cancer incidence that accompanies pregnancy. These findings further indicate that breast cancer chemoprevention may be achieved, in part, through suppression of MaSC function.


Assuntos
Estrogênios/metabolismo , Glândulas Mamárias Animais/citologia , Progesterona/metabolismo , Células-Tronco/citologia , Animais , Aromatase/metabolismo , Inibidores da Aromatase/farmacologia , Antígeno CD24/metabolismo , Contagem de Células , Receptores ErbB/metabolismo , Estrogênios/farmacologia , Feminino , Humanos , Integrina beta1/metabolismo , Integrina beta3/metabolismo , Letrozol , Camundongos , Nitrilas/farmacologia , Ovariectomia , Comunicação Parácrina/efeitos dos fármacos , Gravidez , Prenhez/fisiologia , Progesterona/antagonistas & inibidores , Progesterona/farmacologia , Ligante RANK/metabolismo , Receptores de Estrogênio/deficiência , Receptores de Progesterona/deficiência , Transdução de Sinais/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Triazóis/farmacologia
9.
Rev Endocr Metab Disord ; 16(2): 131-9, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25557611

RESUMO

After it was proposed that the osteoblast lineage controlled the formation of osteoclasts, cell culture methods were developed that established this to be the case. Evidence was obtained that cytokines and hormones that promote osteoclast formation act first on osteoblast lineage cells to promote the production of a membrane-bound regulator of osteoclastogenesis. This proved to be receptor activator of NF-kB ligand (RANKL) a member of the tumor necrosis factor ligand family that acts upon its receptor RANK in the hematopoietic lineage, with interaction restricted by a decoy soluble receptor osteoprotegerin (OPG), also a product of the osteoblast lineage. The physiological roles of these factors were established through genetic and pharmacological studies, have led to a new physiology of bone, with complete revision of older ideas over the last 15 years, ultimately leading to the development of new pharmaceutical agents for bone disease.


Assuntos
Osso e Ossos/fisiologia , Osteoprotegerina/fisiologia , Ligante RANK/fisiologia , Animais , Humanos , Mutação/fisiologia , Osteoblastos/fisiologia , Osteoprotegerina/genética , Comunicação Parácrina/genética , Ligante RANK/genética , Receptor Ativador de Fator Nuclear kappa-B/genética , Receptor Ativador de Fator Nuclear kappa-B/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
10.
FASEB J ; 28(10): 4482-96, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24982128

RESUMO

Cells that form bone (osteoblasts) express both ephrinB2 and EphB4, and previous work has shown that pharmacological inhibition of the ephrinB2/EphB4 interaction impairs osteoblast differentiation in vitro and in vivo. The purpose of this study was to determine the role of ephrinB2 signaling in the osteoblast lineage in the process of bone formation. Cultured osteoblasts from mice with osteoblast-specific ablation of ephrinB2 showed delayed expression of osteoblast differentiation markers, a finding that was reproduced by ephrinB2, but not EphB4, RNA interference. Microcomputed tomography, histomorphometry, and mechanical testing of the mice lacking ephrinB2 in osteoblasts revealed a 2-fold delay in bone mineralization, a significant reduction in bone stiffness, and a 50% reduction in osteoblast differentiation induced by anabolic parathyroid hormone (PTH) treatment, compared to littermate sex- and age-matched controls. These defects were associated with significantly lower mRNA levels of late osteoblast differentiation markers and greater levels of osteoblast and osteocyte apoptosis, indicated by TUNEL staining and transmission electron microscopy of bone samples, and a 2-fold increase in annexin V staining and 7-fold increase in caspase 8 activation in cultured ephrinB2 deficient osteoblasts. We conclude that osteoblast differentiation and bone strength are maintained by antiapoptotic actions of ephrinB2 signaling within the osteoblast lineage.


Assuntos
Apoptose , Calcificação Fisiológica , Osteoblastos/metabolismo , Osteogênese , Receptor EphB2/metabolismo , Animais , Anexina A5/genética , Anexina A5/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Osteoblastos/citologia , Receptor EphB2/genética , Receptor EphB4/genética , Receptor EphB4/metabolismo , Transdução de Sinais
11.
Cytokine ; 68(2): 101-9, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24767864

RESUMO

OBJECTIVE: To identify how the gp130-signaling cytokine oncostatin M (OSM), acting alone or in concert with IL-1ß or TNFα, affects synovial fibroblast expression of genes relevant to inflammation and bone erosion in inflammatory arthritis. METHODS: Synovial fibroblasts (SFs) were isolated from non-arthritic wild type (WT) or OSM receptor deficient (OSMR(-/-)) mice and stimulated with OSM, IL-1ß or TNFα and their combinations. Cytokine gene expression was assessed by quantitative RT-PCR. ELISA, flow cytometry and immunohistochemistry identified protein expression. Gene expression patterns were confirmed in SFs isolated from patients with osteoarthritis (OASFs) and rheumatoid arthritis (RASFs). RESULTS: Expression of OSM and its receptors, gp130, OSMR and LIFR, was increased in synovial tissue from the mouse antigen-induced arthritis model. In isolated WT mouse synovial fibroblasts OSM alone, or in synergy with IL-1ß, or together with TNFα, potently induced expression of the pro-inflammatory cytokine IL-6. OSM also induced a sustained increase in mRNA levels of the pro-osteoclastic cytokine RANKL. Combining OSM with IL-1ß, but not with TNFα, further increased RANKL expression. Importantly these effects of OSM were all dependent on the expression of OSMR. Furthermore, OSM also increased expression of its own receptors, gp130 and OSMR and the IL-1 receptor, IL1-R1; the latter effects were also observed in both human OASFs and RASFs. CONCLUSION: Together our data suggests that OSM signaling via OSMR in SFs has the potential to contribute significantly to joint destruction in inflammatory arthritis. It not only induces expression of pro-inflammatory and pro-osteoclastic cytokines but can also augment its own actions and that of IL-1 by inducing expression of OSMR and IL-1R1.


Assuntos
Fibroblastos/metabolismo , Interleucina-1beta/metabolismo , Oncostatina M/metabolismo , Receptores de Oncostatina M/metabolismo , Membrana Sinovial/patologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Artrite Reumatoide/genética , Artrite Reumatoide/patologia , Regulação da Expressão Gênica , Humanos , Interleucina-1beta/genética , Camundongos Endogâmicos C57BL , Osteoprotegerina/genética , Osteoprotegerina/metabolismo , Ligante RANK/genética , Ligante RANK/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Interleucina-1/metabolismo , Receptores de Oncostatina M/deficiência
12.
Calcif Tissue Int ; 94(1): 88-97, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23700149

RESUMO

In the bone remodeling process that takes place throughout the skeleton at bone multicellular units, intercellular communication processes are crucial. The osteoblast lineage has long been known to program osteoclast formation and hence resorption, but the preservation of bone mass and integrity requires tight control of remodeling. This needs local controls that ensure availability of mesenchymal precursors and the provision of local signals that promote differentiation through the osteoblast lineage. Some signals can come from growth factors released from resorbed bone matrix, and there is increasing evidence that the osteoclast lineage itself produces factors that can either enhance or inhibit osteoblast differentiation and hence bone formation. A number of such factors have been identified from predominantly in vitro experiments. The coupling of bone formation to resorption is increasingly recognized as a complex, dynamic process that results from the input of many local factors of cell and matrix origin that can either promote or inhibit bone formation.


Assuntos
Remodelação Óssea/genética , Osteoclastos/metabolismo , Animais , Remodelação Óssea/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteoclastos/citologia , Osteogênese/fisiologia
13.
Blood ; 117(21): 5631-42, 2011 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-21421837

RESUMO

Erythropoietin (Epo) has been used in the treatment of anemia resulting from numerous etiologies, including renal disease and cancer. However, its effects are controversial and the expression pattern of the Epo receptor (Epo-R) is debated. Using in vivo lineage tracing, we document that within the hematopoietic and mesenchymal lineage, expression of Epo-R is essentially restricted to erythroid lineage cells. As expected, adult mice treated with a clinically relevant dose of Epo had expanded erythropoiesis because of amplification of committed erythroid precursors. Surprisingly, we also found that Epo induced a rapid 26% loss of the trabecular bone volume and impaired B-lymphopoiesis within the bone marrow microenvironment. Despite the loss of trabecular bone, hematopoietic stem cell populations were unaffected. Inhibition of the osteoclast activity with bisphosphonate therapy blocked the Epo-induced bone loss. Intriguingly, bisphosphonate treatment also reduced the magnitude of the erythroid response to Epo. These data demonstrate a previously unrecognized in vivo regulatory network coordinating erythropoiesis, B-lymphopoiesis, and skeletal homeostasis. Importantly, these findings may be relevant to the clinical application of Epo.


Assuntos
Linfócitos B/metabolismo , Medula Óssea/efeitos dos fármacos , Osso e Ossos/metabolismo , Eritropoese/fisiologia , Eritropoetina/farmacologia , Homeostase , Linfopoese/fisiologia , Animais , Medula Óssea/metabolismo , Remodelação Óssea/fisiologia , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Eritroblastos/metabolismo , Citometria de Fluxo , Expressão Gênica , Humanos , Masculino , Mesoderma/citologia , Mesoderma/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Receptores da Eritropoetina/metabolismo , Proteínas Recombinantes , Baço/citologia , Baço/metabolismo
14.
Nature ; 445(7130): E19; discussion E19-20, 2007 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-17314931

RESUMO

The RANK/RANKL signalling mechanism is the final common pathway of osteoclast formation and activity. Inhibitors of RANK ligand (RANKL) that bind to RANK (for 'receptor activator of NF-kappaB'), such as osteoprotegerin (OPG), neutralizing antibodies against RANKL and soluble RANK antagonists, are well described inhibitors of bone metastasis in preclinical and clinical models, presumably because of their effects on osteoclasts. Jones et al. show that OPG inhibits bone metastasis after intracardiac injection of B16F10 murine melanoma cells, but claim that bone metastases are entirely independent of osteoclast formation and bone resorption: rather, they are caused by an effect on cell migration through RANK. However, we question whether these surprising conclusions are rigorously supported by their data.


Assuntos
Neoplasias Ósseas/patologia , Neoplasias Ósseas/secundário , Osteoclastos/metabolismo , Osteoclastos/patologia , Ligante RANK/metabolismo , Animais , Neoplasias Ósseas/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Humanos , Melanoma/patologia , Camundongos , Metástase Neoplásica/tratamento farmacológico , Osteoclastos/efeitos dos fármacos , Osteoprotegerina/farmacologia , Osteoprotegerina/uso terapêutico , Ligante RANK/antagonistas & inibidores , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Reprodutibilidade dos Testes
15.
J Biol Chem ; 286(6): 4186-98, 2011 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-21123171

RESUMO

Osteoblasts and adipocytes are derived from common mesenchymal progenitor cells. The bone loss of osteoporosis is associated with altered progenitor differentiation from an osteoblastic to an adipocytic lineage. cDNA microarrays and quantitative real-time PCR (Q-PCR) were carried out in a differentiating mouse stromal osteoblastic cell line, Kusa 4b10, to identify gene targets of factors that stimulate osteoblast differentiation including parathyroid hormone (PTH) and gp130-binding cytokines, oncostatin M (OSM) and cardiotrophin-1 (CT-1). Zinc finger protein 467 (Zfp467) was rapidly down-regulated by PTH, OSM, and CT-1. Retroviral overexpression and RNA interference for Zfp467 in mouse stromal cells showed that this factor stimulated adipocyte formation and inhibited osteoblast commitment compared with controls. Regulation of adipocyte markers, including peroxisome proliferator-activated receptor (PPAR) γ, C/EBPα, adiponectin, and resistin, and late osteoblast/osteocyte markers (osteocalcin and sclerostin) by Zfp467 was confirmed by Q-PCR. Intra-tibial injection of calvarial cells transduced with retroviral Zfp467 doubled the number of marrow adipocytes in C57Bl/6 mice compared with vector control-transduced cells, providing in vivo confirmation of a pro-adipogenic role of Zfp467. Furthermore, Zfp467 transactivated a PPAR-response element reporter construct and recruited a histone deacetylase complex. Thus Zfp467 is a novel co-factor that promotes adipocyte differentiation and suppresses osteoblast differentiation. This has relevance to therapeutic interventions in osteoporosis, including PTH-based therapies currently available, and may be of relevance for the use of adipose-derived stem cells for tissue engineering.


Assuntos
Adipócitos/metabolismo , Diferenciação Celular , Proteínas Nucleares/metabolismo , Osteoblastos/metabolismo , Elementos de Resposta , Fatores de Transcrição/metabolismo , Ativação Transcricional , Adipócitos/patologia , Animais , Antígenos de Diferenciação/biossíntese , Antígenos de Diferenciação/genética , Linhagem Celular , Proteínas de Ligação a DNA , Camundongos , Proteínas Nucleares/genética , Osteoblastos/patologia , Osteoporose/genética , Osteoporose/metabolismo , Osteoporose/patologia , Retroviridae , Fatores de Transcrição/genética , Transdução Genética
16.
Vitam Horm ; 120: 215-230, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35953110

RESUMO

Parathyroid hormone-related protein (PTHrP) was discovered as the tumor product causing the humoral hypercalcemia of malignancy. Its structural similarity to the hormone, PTH, with 8 of the first 13 amino acids identical, was sufficient to explain the sharing by PTHrP and PTH of a common receptor, PTH1R, although the remainder of the sequences are unique. PTHrP has important roles in development of several organs, including breast and bone, and functions as a paracrine factor postnatally in these and other tissues. In addition to its hormonal role in cancer, PTHrP is produced by two thirds of primary breast cancers and 90% of bone metastases from breast cancer, leading to the concept that its production in bone by breast cancer cells promotes bone resorption, thus favoring tumor establishment and expansion, and an exit from tumor dormancy in bone through downregulation of leukemia inducing factor receptor (LIFR). Cancer production of PTHrP is increased by bone-derived growth factors, with particular attention paid to TGFß, as well as by promoter-driven transcriptional effects, such as the hedgehog signaling factor, GLI2, and microenvironment effects including changes in underlying stiffness of substrates for cells. Although interest has been focused on PTHrP-induced bone resorption in bone metastasis, a mechanistically separate, protective effect against tumor progression has been proposed. Although there is conflicting mouse data, there are clinical studies suggesting that increased production of PTHrP by breast cancers confers upon them a less invasive phenotype, an effect distinct from the bone resorption-stimulating action that favors bone metastasis.


Assuntos
Neoplasias Ósseas , Reabsorção Óssea , Neoplasias da Mama/patologia , Proteína Relacionada ao Hormônio Paratireóideo , Animais , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/secundário , Neoplasias da Mama/metabolismo , Proteínas Hedgehog , Humanos , Camundongos , Hormônio Paratireóideo , Proteína Relacionada ao Hormônio Paratireóideo/genética , Proteína Relacionada ao Hormônio Paratireóideo/metabolismo , Fator de Crescimento Transformador beta , Microambiente Tumoral
17.
J Bone Miner Res ; 37(10): 1876-1890, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35856245

RESUMO

Bone strength is determined by the structure and composition of its thickened outer shell (cortical bone), yet the mechanisms controlling cortical consolidation are poorly understood. Cortical bone maturation depends on SOCS3-mediated suppression of IL-6 cytokine-induced STAT3 phosphorylation in osteocytes, the cellular network embedded in bone matrix. Because SOCS3 also suppresses granulocyte-colony-stimulating factor receptor (G-CSFR) signaling, we here tested whether global G-CSFR (Csf3r) ablation altereed bone structure in male and female mice lacking SOCS3 in osteocytes, (Dmp1Cre :Socs3f/f mice). Dmp1Cre :Socs3f/f :Csf3r-/- mice were generated by crossing Dmp1Cre :Socs3f/f mice with Csf3r-/- mice. Although G-CSFR is not expressed in osteocytes, Csf3r deletion further delayed cortical consolidation in Dmp1Cre :Socs3f/f mice. Micro-CT images revealed extensive, highly porous low-density bone, with little true cortex in the diaphysis, even at 26 weeks of age; including more low-density bone and less high-density bone in Dmp1Cre :Socs3f/f :Csf3r-/- mice than controls. By histology, the area where cortical bone would normally be found contained immature compressed trabecular bone in Dmp1Cre :Socs3f/f :Csf3r-/- mice and greater than normal levels of intracortical osteoclasts, extensive new woven bone formation, and the presence of more intracortical blood vessels than the already high levels observed in Dmp1Cre :Socs3f/f controls. qRT-PCR of cortical bone from Dmp1Cre :Socs3f/f :Csf3r-/- mice also showed more than a doubling of mRNA levels for osteoclasts, osteoblasts, RANKL, and angiogenesis markers. The further delay in cortical bone maturation was associated with significantly more phospho-STAT1 and phospho-STAT3-positive osteocytes, and a threefold increase in STAT1 and STAT3 target gene mRNA levels, suggesting G-CSFR deletion further increases STAT signaling beyond that of Dmp1Cre :Socs3f/f bone. G-CSFR deficiency therefore promotes STAT1/3 signaling in osteocytes, and when SOCS3 negative feedback is absent, elevated local angiogenesis, bone resorption, and bone formation delays cortical bone consolidation. This points to a critical role of G-CSF in replacing condensed trabecular bone with lamellar bone during cortical bone formation. © 2022 American Society for Bone and Mineral Research (ASBMR).


Assuntos
Fator Estimulador de Colônias de Granulócitos , Osteócitos , Receptores de Fator Estimulador de Colônias de Granulócitos , Fator de Transcrição STAT3 , Animais , Feminino , Masculino , Camundongos , Osso Cortical/diagnóstico por imagem , Fator Estimulador de Colônias de Granulócitos/genética , Interleucina-6 , Osteócitos/patologia , RNA Mensageiro , Fator de Transcrição STAT3/metabolismo
18.
Cell Metab ; 4(6): 419-20, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17141624

RESUMO

Bone formation is controlled by a network of transcription factors and signaling molecules. In this issue, , studying the role of the transcription factor ATF4 in a new mouse model of neurofibromatosis type I skeletal defects, demonstrate striking effects of changing dietary protein on bone formation abnormalities.


Assuntos
Fator 4 Ativador da Transcrição/metabolismo , Doenças do Desenvolvimento Ósseo/dietoterapia , Doenças do Desenvolvimento Ósseo/metabolismo , Proteínas Alimentares/uso terapêutico , Neurofibromina 1/metabolismo , Osteoblastos/metabolismo , Aminoácidos/metabolismo , Animais , Transporte Biológico Ativo/efeitos dos fármacos , Transporte Biológico Ativo/genética , Doenças do Desenvolvimento Ósseo/congênito , Doenças do Desenvolvimento Ósseo/patologia , Reabsorção Óssea/dietoterapia , Reabsorção Óssea/genética , Reabsorção Óssea/metabolismo , Reabsorção Óssea/patologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Síndrome de Coffin-Lowry/genética , Síndrome de Coffin-Lowry/metabolismo , Síndrome de Coffin-Lowry/patologia , Colágeno/biossíntese , Proteínas Quinases Dependentes de AMP Cíclico/genética , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Camundongos , Camundongos Knockout , Neurofibromina 1/deficiência , Osteoblastos/patologia , Osteoclastos/metabolismo , Osteoclastos/patologia , Osteogênese/genética , Ligante RANK/biossíntese , Ligante RANK/genética , Proteínas Quinases S6 Ribossômicas 90-kDa/deficiência , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo
19.
Bioorg Med Chem Lett ; 21(23): 7089-93, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-22030030

RESUMO

PDE4 inhibitors have been identified as therapeutic targets for a variety of conditions, particularly inflammatory diseases. We have serendipitously identified a novel class of phosphodiesterase 4 (PDE4) inhibitor during a study to discover antagonists of the parathyroid hormone receptor. X-ray crystallographic studies of PDE4D2 complexed to four potent inhibitors reveal the atomic details of how they inhibit the enzyme and a notable contrast to another recently reported thiophene-based inhibitor.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/química , Modelos Moleculares , Inibidores da Fosfodiesterase 4/química , Tiofenos/síntese química , Sítios de Ligação , Domínio Catalítico , Cristalografia por Raios X , Inibidores da Fosfodiesterase 4/síntese química , Ligação Proteica , Tiofenos/química , Tiofenos/farmacologia
20.
Pediatr Nephrol ; 26(5): 799-803, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21161280

RESUMO

Parathyroid hormone-related protein (PTHrP) mediated hypercalcemia of malignancy is rare in children, and even more so in the setting of a benign tumor. We report two infants with PTHrP-mediated hypercalcemia secondary to congenital mesoblastic nephroma and their outcome after removal of the benign tumor. Pre-operatively hypercalcemia was corrected with saline hydration, furosemide, calcitonin and/ or pamidronate. Following resection of the tumor serum PTHrP normalized. Immunohistochemical staining of tumor cells was positive for PTHrP. Post-operatively the infants developed elevated serum parathyroid hormone with low- normal serum Ca and P, and undetectable urinary Ca and P, probably due to their movement into bone. Children needed treatment with calcitriol, Ca and P supplementation for 6-12 weeks until PTH normalized and urinary Ca and P were detected, suggesting bone replenishment. We conclude that benign congenital mesoblastic nephroma can secrete PTHrP that can cause severe hypercalcemia; and following excision one should anticipate the development of a transient modified "hungry bone"-like condition requiring Ca, P and calcitriol therapy for several weeks accompanied by careful monitoring of mineral homeostasis.


Assuntos
Hipercalcemia/etiologia , Hipercalcemia/metabolismo , Neoplasias Renais/complicações , Nefroma Mesoblástico/complicações , Proteína Relacionada ao Hormônio Paratireóideo/metabolismo , Humanos , Hipercalcemia/patologia , Recém-Nascido , Recém-Nascido Prematuro , Neoplasias Renais/metabolismo , Neoplasias Renais/cirurgia , Masculino , Nefrectomia , Nefroma Mesoblástico/metabolismo , Nefroma Mesoblástico/cirurgia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa