Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Immunother Cancer ; 12(7)2024 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-39043604

RESUMO

BACKGROUND: Advances in pediatric oncology have occurred for some cancers; however, new therapies for sarcoma have been inadequate. Cellular immunotherapy using chimeric antigen receptor (CAR) T cells has shown dramatic benefits in leukemia, lymphoma, and multiple myeloma but has been far less successful in pediatric solid tumors such as rhabdomyosarcoma (RMS) and osteosarcoma (OS). Balancing issues of "on-target, off-tumor toxicity", investigators have identified B7-H3 as a broadly expressed tumor antigen with otherwise restricted expression on normal tissues. We hypothesized that rapid homing via a chemokine receptor and CAR engagement through B7-H3 would enhance CAR T cell efficacy in solid tumors. METHODS: We generated B7-H3 CAR T cells that also express the Interleukin-8 (IL-8) receptor, CXCR2. Cytokine production, flow cytometry, Seahorse assays and RNA sequencing were used to compare the B7-H3 CXCR2 (BC2) CAR T cells with B7-H3 CAR T cells. We developed an IL-8 overexpressing human RMS mouse model to test homing and cytotoxicity in vivo. RESULTS: We demonstrate that IL-8 is expressed by RMS and OS and expression significantly increases after radiation. Overexpression of an IL-8 receptor, CXCR2, on B7-H3 CAR T cells enhances homing into IL-8 expressing tumors, augments T cell metabolism and leads to significant tumor regression. CONCLUSION: These findings warrant further investigation into the use of BC2 CAR T cells as a treatment for patients with RMS, OS and other B7-H3-expressing, IL-8 producing solid tumors.


Assuntos
Antígenos B7 , Interleucina-8 , Receptores de Antígenos Quiméricos , Humanos , Animais , Camundongos , Antígenos B7/metabolismo , Interleucina-8/metabolismo , Receptores de Antígenos Quiméricos/imunologia , Receptores de Antígenos Quiméricos/metabolismo , Imunoterapia Adotiva/métodos , Sarcoma/terapia , Sarcoma/imunologia , Linhagem Celular Tumoral , Criança , Linfócitos T/imunologia , Linfócitos T/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Stem Cells Transl Med ; 11(5): 513-526, 2022 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-35349707

RESUMO

Epigenetic modification is an important process during hematopoietic cell differentiation. Histone deacetylase (HDAC) inhibitors have previously been shown to enhance expansion of umbilical cord blood-derived hematopoietic stem cells (HSCs). However, the effect of HDAC inhibitors on pluripotent stem cells (PSCs) in this context is less understood. For years, investigators have considered PSC-derived natural killer (NK) and T-cell therapies. These "off-the-shelf" cellular therapies are now entering the clinic. However, the in vitro commitment of PSCs to the hematopoietic lineage is inefficient and represents a major bottleneck. We investigated whether HDAC inhibitors (HDACi) influence human PSC differentiation into CD34+CD45+ hematopoietic stem and progenitor cells (HSPCs), focusing on hemogenic endothelium (HE). Pluripotent stem cells cultured in the presence of HDACi showed a 2-5 times increase in HSPCs. Concurrent with this, HDACi-treated PSCs increased expression of 7 transcription factors (HOXA5, HOXA9, HOXA10, RUNX1, ERG, SPI1, and LCOR) recently shown to convert HE to HSPCs. ChIP-qPCR showed that SAHA upregulated acetylated-H3 at the promoter region of the above key genes. SAHA-treated human PSC-derived CD34+CD45+ cells showed primary engraftment in immunodeficient mice, but not serial transplantation. We further demonstrate that SAHA-derived HSPCs could differentiate into functional NK cells in vitro. The addition of SAHA is an easy and effective approach to overcoming the bottleneck in the transition from PSC to HSPCs for "off-the-shelf" cellular immunotherapy.


Assuntos
Hemangioblastos , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Pluripotentes , Animais , Antígenos CD34/metabolismo , Diferenciação Celular , Células Cultivadas , Hemangioblastos/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Camundongos
3.
J Infect Dis ; 220(9): 1453-1461, 2019 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-30835269

RESUMO

BACKGROUND: In temporal arteries (TAs) from patients with giant cell arteritis, varicella zoster virus (VZV) is seen in perineurial cells that surround adventitial nerve bundles and form the peripheral nerve-extrafascicular tissue barrier (perineurium). We hypothesized that during VZV reactivation from ganglia, virus travels transaxonally and disrupts the perineurium to infect surrounding cells. METHODS: Mock- and VZV-infected primary human perineurial cells (HPNCs) were examined for alterations in claudin-1, E-cadherin, and N-cadherin. Conditioned supernatant was analyzed for a soluble factor(s) mediating these alterations and for the ability to increase cell migration. To corroborate in vitro findings, a VZV-infected TA was examined. RESULTS: In VZV-infected HPNCs, claudin-1 redistributed to the nucleus; E-cadherin was lost and N-cadherin gained, with similar changes seen in VZV-infected perineurial cells in a TA. VZV-conditioned supernatant contained increased interleukin 6 (IL-6) that induced E-cadherin loss and N-cadherin gain and increased cell migration when added to uninfected HPNCs; anti-IL-6 receptor antibody prevented these changes. CONCLUSIONS: IL-6 secreted from VZV-infected HPNCs facilitated changes in E- and N-cadherin expression and cell migration, reminiscent of an epithelial-to-mesenchymal cell transition, potentially contributing to loss of perineurial cell barrier integrity and viral spread. Importantly, an anti-IL-6 receptor antibody prevented virus-induced perineurial cell disruption.


Assuntos
Antígenos CD/biossíntese , Caderinas/biossíntese , Herpesvirus Humano 3/crescimento & desenvolvimento , Interações Hospedeiro-Patógeno , Interleucina-6/metabolismo , Miofibroblastos/virologia , Movimento Celular , Células Cultivadas , Claudina-1/biossíntese , Meios de Cultivo Condicionados , Expressão Gênica , Humanos , Miofibroblastos/metabolismo
4.
J Neuroinflammation ; 15(1): 318, 2018 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-30442152

RESUMO

BACKGROUND: Varicella zoster virus (VZV) is a ubiquitous alphaherpesvirus that produces varicella and zoster. VZV can infect multiple cell types in the spinal cord and brain, including astrocytes, producing myelopathy and encephalopathy. While studies of VZV-astrocyte interactions are sparse, a recent report showed that quiescent primary human spinal cord astrocytes (qHA-sps) did not appear activated morphologically during VZV infection. Since astrocytes play a critical role in host defenses during viral infections of the central nervous system, we examined the cytokine responses of qHA-sps and quiescent primary human hippocampal astrocytes (qHA-hps) to VZV infection in vitro, as well as the ability of conditioned supernatant to recruit immune cells. METHODS: At 3 days post-infection, mock- and VZV-infected qHA-sps and qHA-hps were examined for morphological changes by immunofluorescence antibody assay using antibodies directed against glial fibrillary acidic protein and VZV. Conditioned supernatants were analyzed for proinflammatory cytokines [interleukin (IL)-1ß, IL-2, IL-4, IL-6, IL-8, IL-10, IL-12p70, IL-13, interferon-gamma, and tumor necrosis factor-α] using the Meso Scale Discovery multiplex ELISA platform. Finally, the ability of conditioned supernatants to attract peripheral blood mononuclear cells (PBMCs) was determined using a chemotaxis assay. Quiescent primary human perineurial cells (qHPNCs) served as a control for VZV-induced cytokine production and PBMC migration. To confirm that the astrocytes have the ability to increase cytokine secretion, qHA-sps and qHA-hps were treated with IL-1ß and examined for morphological changes and IL-6 secretion. RESULTS: VZV-infected qHA-sps displayed extensive cellular processes, whereas VZV-infected qHA-hps became swollen and clustered together. Astrocytes had the capacity to secrete IL-6 in response to IL-1ß. Compared to mock-infected cells, VZV-infected qHA-sps showed significantly reduced secretion of IL-2, IL-4, IL-6, IL-12p70, and IL-13, while VZV-infected qHA-hps showed significantly reduced IL-8 secretion. In contrast, levels of all 10 cytokines examined were significantly increased in VZV-infected qHPNCs. Consistent with these results, conditioned supernatant from VZV-infected qHPNCs, but not that from VZV-infected qHA-sps and qHA-hps, recruited PBMCs. CONCLUSIONS: VZV-infected qHA-sps and qHA-hps have distinct morphological alterations and patterns of proinflammatory cytokine suppression that could contribute to ineffective viral clearance in VZV myelopathy and encephalopathy, respectively.


Assuntos
Astrócitos/metabolismo , Astrócitos/virologia , Citocinas/metabolismo , Regulação Viral da Expressão Gênica/fisiologia , Hipocampo/patologia , Medula Espinal/patologia , Idoso , Astrócitos/efeitos dos fármacos , Movimento Celular/fisiologia , Citocinas/genética , Citocinas/farmacologia , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/metabolismo , Herpesvirus Humano 3/metabolismo , Humanos , Masculino , RNA Mensageiro/metabolismo , Infecção pelo Vírus da Varicela-Zoster/patologia
5.
J Control Release ; 286: 85-93, 2018 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-30030182

RESUMO

Decades of research into improving drug delivery to tumors has documented uptake of particulate delivery systems by resident macrophages in the lung, liver, and spleen, and correlated short circulation times with reduced tumor accumulation. An implicit assumption in these studies is that nanoparticles present in the blood are available for distribution to the tumor. This study documents significant levels of lipoplex uptake by circulating leukocytes, and its effect on distribution to the tumor and other organs. In agreement with previous studies, PEGylation dramatically extends circulation times and enhances tumor delivery. However, our studies suggest that this relationship is not straightforward, and that particle sequestration by leukocytes can significantly alter biodistribution, especially with non-PEGylated nanoparticle formulations. We conclude that leukocyte uptake should be considered in biodistribution studies, and that delivery to these circulating cells may present opportunities for treating viral infections and leukemia.


Assuntos
Leucócitos/metabolismo , Nanopartículas/metabolismo , Polietilenoglicóis/metabolismo , Animais , DNA/administração & dosagem , DNA/farmacocinética , Feminino , Técnicas de Transferência de Genes , Camundongos Endogâmicos BALB C , Camundongos SCID , Neoplasias/metabolismo , Plasmídeos/administração & dosagem , Plasmídeos/farmacocinética , Distribuição Tecidual
6.
PLoS One ; 12(7): e0180870, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28719615

RESUMO

While the immune system is essential for the maintenance of the homeostasis, health and survival of humans, aberrant immune responses can lead to chronic inflammatory and autoimmune disorders. Pharmacological modulation of drug targets in the immune system to ameliorate disease also carry a risk of immunosuppression that could lead to adverse outcomes. Therefore, it is important to understand the 'immune fingerprint' of novel therapeutics as they relate to current and, clinically used immunological therapies to better understand their potential therapeutic benefit as well as immunosuppressive ability that might lead to adverse events such as infection risks and cancer. Since the mechanistic investigation of pharmacological modulators in a drug discovery setting is largely compound- and mechanism-centric but not comprehensive in terms of immune system impact, we developed a human tissue based functional assay platform to evaluate the impact of pharmacological modulators on a range of innate and adaptive immune functions. Here, we demonstrate that it is possible to generate a qualitative and quantitative immune system impact of pharmacological modulators, which might help better understand and predict the benefit-risk profiles of these compounds in the treatment of immune disorders.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Sistema Imunitário/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Quimiocinas/biossíntese , Humanos , Sistema Imunitário/citologia , Sistema Imunitário/imunologia , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Fagócitos/efeitos dos fármacos , Fagócitos/imunologia , Fagócitos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Receptores Toll-Like/metabolismo , Transcriptoma/efeitos dos fármacos
7.
J Neuroimmunol ; 308: 112-117, 2017 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-28335992

RESUMO

Varicella zoster virus (VZV) is a ubiquitous, human alphaherpesvirus that produces varicella on primary infection then becomes latent in ganglionic neurons along the entire neuraxis. In elderly and immunocompromised individuals, VZV reactivates and travels along nerve fibers peripherally resulting in zoster. However, VZV can also spread centrally and infect cerebral and extracranial arteries (VZV vasculopathy) to produce transient ischemic attacks, stroke, aneurysm, sinus thrombosis and giant cell arteritis, as well as granulomatous aortitis. The mechanisms of virus-induced pathological vascular remodeling are not fully elucidated; however, recent studies suggest that inflammation and dysregulation of programmed death ligand-1 play a significant role.


Assuntos
Varicela , Herpes Zoster , Herpesvirus Humano 3/patogenicidade , Acidente Vascular Cerebral/etiologia , Animais , Varicela/complicações , Varicela/imunologia , Varicela/virologia , Herpes Zoster/complicações , Herpes Zoster/imunologia , Herpes Zoster/virologia , Humanos , Acidente Vascular Cerebral/virologia
8.
J Virol ; 90(23): 10527-10534, 2016 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-27630241

RESUMO

Varicella-zoster virus (VZV) vasculopathy produces stroke, giant cell arteritis, and granulomatous aortitis, and it develops after virus reactivates from ganglia and spreads transaxonally to arterial adventitia, resulting in persistent inflammation and pathological vascular remodeling. The mechanism(s) by which inflammatory cells persist in VZV-infected arteries is unknown; however, virus-induced dysregulation of programmed death ligand 1 (PD-L1) may play a role. Specifically, PD-L1 can be expressed on virtually all nucleated cells and suppresses the immune system by interacting with the programmed cell death protein receptor 1, found exclusively on immune cells; thus, downregulation of PD-L1 may promote inflammation, as seen in some autoimmune diseases. Both flow cytometry and immunofluorescence analyses to test whether VZV infection of adventitial cells downregulates PD-L1 showed decreased PD-L1 expression in VZV-infected compared to mock-infected human brain vascular adventitial fibroblasts (HBVAFs), perineural cells (HPNCs), and fetal lung fibroblasts (HFLs) at 72 h postinfection. Quantitative RT-PCR analyses showed no change in PD-L1 transcript levels between mock- and VZV-infected cells, indicating a posttranscriptional mechanism for VZV-mediated downregulation of PD-L1. Flow cytometry analyses showed decreased major histocompatibility complex class I (MHC-I) expression in VZV-infected cells and adjacent uninfected cells compared to mock-infected cells. These data suggest that reduced PD-L1 expression in VZV-infected adventitial cells contribute to persistent vascular inflammation observed in virus-infected arteries from patients with VZV vasculopathy, while downregulation of MHC-I prevents viral clearance. IMPORTANCE: Here, we provide the first demonstration that VZV downregulates PD-L1 expression in infected HBVAFs, HPNCs, and HFLs, which, together with the noted VZV-mediated downregulation of MHC-I, might foster persistent inflammation in vessels, leading to pathological vascular remodeling during VZV vasculopathy and persistent inflammation in infected lungs to promote subsequent infection of T cells and hematogenous virus spread. Identification of a potential mechanism by which persistent inflammation in the absence of effective viral clearance occurs in VZV vasculopathy and VZV infection of the lung is a step toward targeted therapy of VZV-induced disease.


Assuntos
Antígeno B7-H1/metabolismo , Herpesvirus Humano 3/patogenicidade , Antígenos de Histocompatibilidade Classe I/metabolismo , Túnica Adventícia/irrigação sanguínea , Túnica Adventícia/imunologia , Túnica Adventícia/virologia , Antígeno B7-H1/genética , Encéfalo/irrigação sanguínea , Encéfalo/imunologia , Encéfalo/virologia , Células Cultivadas , Regulação para Baixo , Fibroblastos/imunologia , Fibroblastos/virologia , Infecções por Herpesviridae/etiologia , Infecções por Herpesviridae/imunologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Pulmão/imunologia , Pulmão/virologia , Neuroglia/imunologia , Neuroglia/virologia
9.
Development ; 142(3): 438-43, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25564652

RESUMO

Cell-based bone regeneration strategies offer promise for traumatic bone injuries, congenital defects, non-union fractures and other skeletal pathologies. Postnatal bone remodeling and fracture healing provide evidence that an osteochondroprogenitor cell is present in adult life that can differentiate to remodel or repair the fractured bone. However, cell-based skeletal repair in the clinic is still in its infancy, mostly due to poor characterization of progenitor cells and lack of knowledge about their in vivo behavior. Here, we took a combined approach of high-throughput screening, flow-based cell sorting and in vivo transplantation to isolate markers that identify osteochondroprogenitor cells. We show that the presence of tetraspanin CD9 enriches for osteochondroprogenitors within CD105(+) mesenchymal cells and that these cells readily form bone upon transplantation. In addition, we have used Thy1.2 and the ectonucleotidase CD73 to identify subsets within the CD9(+) population that lead to endochondral or intramembranous-like bone formation. Utilization of this unique cell surface phenotype to enrich for osteochondroprogenitor cells will allow for further characterization of the molecular mechanisms that regulate their osteogenic properties.


Assuntos
Biomarcadores/metabolismo , Regeneração Óssea/fisiologia , Condrócitos/fisiologia , Osteoblastos/fisiologia , Células-Tronco/fisiologia , 5'-Nucleotidase/metabolismo , Animais , Condrócitos/citologia , Condrócitos/metabolismo , Citometria de Fluxo , Ensaios de Triagem em Larga Escala , Processamento de Imagem Assistida por Computador , Rim/diagnóstico por imagem , Camundongos , Camundongos Endogâmicos C57BL , Análise em Microsséries , Osteoblastos/citologia , Osteoblastos/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Tetraspanina 29/metabolismo , Microtomografia por Raio-X
10.
J Exp Med ; 210(9): 1793-806, 2013 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-23918955

RESUMO

Osteoblasts are responsible for the formation and mineralization of the skeleton. To identify novel regulators of osteoblast differentiation, we conducted an unbiased forward genetic screen using a lentiviral-based shRNA library. This functional genomics analysis led to the identification of the microtubule-associated protein DCAMKL1 (Doublecortin-like and CAM kinase-like 1) as a novel regulator of osteogenesis. Mice with a targeted disruption of Dcamkl1 displayed elevated bone mass secondary to increased bone formation by osteoblasts. Molecular experiments demonstrated that DCAMKL1 represses osteoblast activation by antagonizing Runx2, the master transcription factor in osteoblasts. Key elements of the cleidocranial dysplasia phenotype observed in Runx2(+/-) mice are reversed by the introduction of a Dcamkl1-null allele. Our results establish a genetic linkage between these two proteins in vivo and demonstrate that DCAMKL1 is a physiologically relevant regulator of anabolic bone formation.


Assuntos
Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Microtúbulos/metabolismo , Osteoblastos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Sequência de Aminoácidos , Animais , Osso e Ossos/metabolismo , Diferenciação Celular , Células Cultivadas , Subunidade alfa 1 de Fator de Ligação ao Core/química , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Quinases Semelhantes a Duplacortina , Epistasia Genética , Inativação Gênica , Genômica , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Lentivirus/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Mutantes , Dados de Sequência Molecular , Tamanho do Órgão , Osteoblastos/patologia , Osteoclastos/metabolismo , Osteoclastos/patologia , Fosforilação , Fosfosserina/metabolismo , Polimerização , Proteínas Serina-Treonina Quinases/genética , RNA Interferente Pequeno/metabolismo
11.
J Clin Invest ; 123(9): 4010-22, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23945236

RESUMO

Mice deficient in Schnurri-3 (SHN3; also known as HIVEP3) display increased bone formation, but harnessing this observation for therapeutic benefit requires an improved understanding of how SHN3 functions in osteoblasts. Here we identified SHN3 as a dampener of ERK activity that functions in part downstream of WNT signaling in osteoblasts. A D-domain motif within SHN3 mediated the interaction with and inhibition of ERK activity and osteoblast differentiation, and knockin of a mutation in Shn3 that abolishes this interaction resulted in aberrant ERK activation and consequent osteoblast hyperactivity in vivo. Additionally, in vivo genetic interaction studies demonstrated that crossing to Lrp5(-/-) mice partially rescued the osteosclerotic phenotype of Shn3(-/-) mice; mechanistically, this corresponded to the ability of SHN3 to inhibit ERK-mediated suppression of GSK3ß. Inducible knockdown of Shn3 in adult mice resulted in a high-bone mass phenotype, providing evidence that transient blockade of these pathways in adults holds promise as a therapy for osteoporosis.


Assuntos
Proteínas de Ligação a DNA/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Osteoblastos/metabolismo , Via de Sinalização Wnt , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Osso e Ossos/fisiopatologia , Proteínas de Ligação a DNA/química , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Células HEK293 , Humanos , Células-Tronco Mesenquimais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Dados de Sequência Molecular , Osteogênese , Osteoporose/tratamento farmacológico , Osteoporose/metabolismo , Estrutura Terciária de Proteína , beta Catenina/metabolismo
12.
Proc Natl Acad Sci U S A ; 109(21): 8173-8, 2012 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-22573816

RESUMO

Mice lacking the large zinc finger protein Schnurri-3 (Shn3) display increased bone mass, in part, attributable to augmented osteoblastic bone formation. Here, we show that in addition to regulating bone formation, Shn3 indirectly controls bone resorption by osteoclasts in vivo. Although Shn3 plays no cell-intrinsic role in osteoclasts, Shn3-deficient animals show decreased serum markers of bone turnover. Mesenchymal cells lacking Shn3 are defective in promoting osteoclastogenesis in response to selective stimuli, likely attributable to reduced expression of the key osteoclastogenic factor receptor activator of nuclear factor-κB ligand. The bone phenotype of Shn3-deficient mice becomes more pronounced with age, and mice lacking Shn3 are completely resistant to disuse osteopenia, a process that requires functional osteoclasts. Finally, selective deletion of Shn3 in the mesenchymal lineage recapitulates the high bone mass phenotype of global Shn3 KO mice, including reduced osteoclastic bone catabolism in vivo, indicating that Shn3 expression in mesenchymal cells directly controls osteoblastic bone formation and indirectly regulates osteoclastic bone resorption.


Assuntos
Reabsorção Óssea/fisiopatologia , Proteínas de Ligação a DNA/genética , Hiperparatireoidismo Secundário/fisiopatologia , Osteoblastos/fisiologia , Osteoclastos/fisiologia , Envelhecimento/fisiologia , Animais , Reabsorção Óssea/genética , Células Cultivadas , Técnicas de Cocultura , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas de Ligação a DNA/metabolismo , Hiperparatireoidismo Secundário/genética , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteoblastos/citologia , Osteoclastos/citologia , Fenótipo , Ligante RANK/metabolismo , Elementos Reguladores de Transcrição/fisiologia , Crânio/citologia
13.
J Clin Invest ; 121(11): 4383-92, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21965325

RESUMO

Mutations in human FYVE, RhoGEF, and PH domain-containing 1 (FGD1) cause faciogenital dysplasia (FGDY; also known as Aarskog syndrome), an X-linked disorder that affects multiple skeletal structures. FGD1 encodes a guanine nucleotide exchange factor (GEF) that specifically activates the Rho GTPase CDC42. However, the mechanisms by which mutations in FGD1 affect skeletal development are unknown. Here, we describe what we believe to be a novel signaling pathway in osteoblasts initiated by FGD1 that involves the MAP3K mixed-lineage kinase 3 (MLK3). We observed that MLK3 functions downstream of FGD1 to regulate ERK and p38 MAPK, which in turn phosphorylate and activate the master regulator of osteoblast differentiation, Runx2. Mutations in FGD1 found in individuals with FGDY ablated its ability to activate MLK3. Consistent with our description of this pathway and the phenotype of patients with FGD1 mutations, mice with a targeted deletion of Mlk3 displayed multiple skeletal defects, including dental abnormalities, deficient calvarial mineralization, and reduced bone mass. Furthermore, mice with knockin of a mutant Mlk3 allele that is resistant to activation by FGD1/CDC42 displayed similar skeletal defects, demonstrating that activation of MLK3 specifically by FGD1/CDC42 is important for skeletal mineralization. Thus, our results provide a putative biochemical mechanism for the skeletal defects in human FGDY and suggest that modulating MAPK signaling may benefit these patients.


Assuntos
Desenvolvimento Ósseo/genética , Desenvolvimento Ósseo/fisiologia , Nanismo/genética , Doenças Genéticas Ligadas ao Cromossomo X/genética , Deformidades Congênitas da Mão/genética , Cardiopatias Congênitas/genética , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/fisiologia , Mutação , Proteínas/genética , Proteínas/fisiologia , Animais , Modelos Animais de Doenças , Nanismo/patologia , Nanismo/fisiopatologia , Ativação Enzimática , Face/anormalidades , Face/patologia , Face/fisiopatologia , Feminino , Técnicas de Introdução de Genes , Doenças Genéticas Ligadas ao Cromossomo X/patologia , Doenças Genéticas Ligadas ao Cromossomo X/fisiopatologia , Genitália Masculina/anormalidades , Genitália Masculina/patologia , Genitália Masculina/fisiopatologia , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/fisiologia , Deformidades Congênitas da Mão/patologia , Deformidades Congênitas da Mão/fisiopatologia , Cardiopatias Congênitas/patologia , Cardiopatias Congênitas/fisiopatologia , Humanos , MAP Quinase Quinase Quinases/deficiência , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Osteoblastos/patologia , Osteoblastos/fisiologia , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , MAP Quinase Quinase Quinase 11 Ativada por Mitógeno
14.
J Clin Invest ; 121(7): 2534-42, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21737885

RESUMO

Over the past decade and a half, the biomedical community has uncovered a previously unappreciated reciprocal relationship between cells of the immune and skeletal systems. Work in this field, which has been termed "osteoimmunology," has resulted in the development of clinical therapeutics for seemingly disparate diseases linked by the common themes of inflammation and bone remodeling. Here, the important concepts and discoveries in osteoimmunology are discussed in the context of the diseases bridging these two organ systems, including arthritis, osteoporosis, cancer, and infection, and the targeted treatments used by clinicians to combat them.


Assuntos
Artrite/imunologia , Infecções/imunologia , Neoplasias/imunologia , Osteoporose/imunologia , Animais , Artrite/tratamento farmacológico , Humanos , Sistema Imunitário/imunologia , Interleucina-17/imunologia , Neoplasias/tratamento farmacológico , Osteoblastos/imunologia , Osteoclastos/imunologia , Osteoporose/tratamento farmacológico , Ligante RANK/imunologia
15.
Nature ; 472(7341): 105-9, 2011 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-21475200

RESUMO

Generation of a diverse and self-tolerant T-cell repertoire requires appropriate interpretation of T-cell antigen receptor (TCR) signals by CD4(+ ) CD8(+) double-positive thymocytes. Thymocyte cell fate is dictated by the nature of TCR-major-histocompatibility-complex (MHC)-peptide interactions, with signals of higher strength leading to death (negative selection) and signals of intermediate strength leading to differentiation (positive selection). Molecules that regulate T-cell development by modulating TCR signal strength have been described but components that specifically define the boundaries between positive and negative selection remain unknown. Here we show in mice that repression of TCR-induced death pathways is critical for proper interpretation of positive selecting signals in vivo, and identify schnurri-2 (Shn2; also known as Hivep2) as a crucial death dampener. Our results indicate that Shn2(-/-) double-positive thymocytes inappropriately undergo negative selection in response to positive selecting signals, thus leading to disrupted T-cell development. Shn2(-/-) double-positive thymocytes are more sensitive to TCR-induced death in vitro and die in response to positive selection interactions in vivo. However, Shn2-deficient thymocytes can be positively selected when TCR-induced death is genetically ablated. Shn2 levels increase after TCR stimulation, indicating that integration of multiple TCR-MHC-peptide interactions may fine-tune the death threshold. Mechanistically, Shn2 functions downstream of TCR proximal signalling compenents to dampen Bax activation and the mitochondrial death pathway. Our findings uncover a critical regulator of T-cell development that controls the balance between death and differentiation.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Linfócitos T/citologia , Animais , Proteínas Reguladoras de Apoptose/deficiência , Proteínas Reguladoras de Apoptose/genética , Proteína 11 Semelhante a Bcl-2 , Morte Celular , Diferenciação Celular , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais , Linfócitos T/imunologia , Linfócitos T/metabolismo , Timo/citologia , Timo/imunologia
16.
Proc Natl Acad Sci U S A ; 107(18): 8254-8, 2010 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-20404140

RESUMO

Formation and remodeling of the skeleton relies on precise temporal and spatial regulation of genes expressed in cartilage and bone cells. Debilitating diseases of the skeletal system occur when mutations arise that disrupt these intricate genetic regulatory programs. Here, we report that mice bearing parallel null mutations in the adapter proteins Schnurri2 (Shn2) and Schnurri3 (Shn3) exhibit defects in patterning of the axial skeleton during embryogenesis. Postnatally, these compound mutant mice develop a unique osteochondrodysplasia. The deletion of Shn2 and Shn3 impairs growth plate maturation during endochondral ossification but simultaneously results in massively elevated trabecular bone formation. Hence, growth plate maturation and bone formation can be uncoupled under certain circumstances. These unexpected findings demonstrate that both unique and redundant functions reside in the Schnurri protein family that are required for proper skeletal patterning and remodeling.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Lâmina de Crescimento/crescimento & desenvolvimento , Lâmina de Crescimento/metabolismo , Osteogênese , Animais , Densidade Óssea , Proteínas de Ligação a DNA/deficiência , Dosagem de Genes , Regulação da Expressão Gênica no Desenvolvimento , Lâmina de Crescimento/embriologia , Camundongos , Camundongos Knockout , Osteocondrodisplasias/genética , Osteocondrodisplasias/metabolismo , Osteocondrodisplasias/patologia , Fenótipo
17.
J Clin Invest ; 118(2): 491-504, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18219387

RESUMO

Drug targeting of adult stem cells has been proposed as a strategy for regenerative medicine, but very few drugs are known to target stem cell populations in vivo. Mesenchymal stem/progenitor cells (MSCs) are a multipotent population of cells that can differentiate into muscle, bone, fat, and other cell types in context-specific manners. Bortezomib (Bzb) is a clinically available proteasome inhibitor used in the treatment of multiple myeloma. Here, we show that Bzb induces MSCs to preferentially undergo osteoblastic differentiation, in part by modulation of the bone-specifying transcription factor runt-related transcription factor 2 (Runx-2) in mice. Mice implanted with MSCs showed increased ectopic ossicle and bone formation when recipients received low doses of Bzb. Furthermore, this treatment increased bone formation and rescued bone loss in a mouse model of osteoporosis. Thus, we show that a tissue-resident adult stem cell population in vivo can be pharmacologically modified to promote a regenerative function in adult animals.


Assuntos
Regeneração Óssea , Ácidos Borônicos/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Osteoblastos/citologia , Inibidores de Proteases/farmacologia , Pirazinas/farmacologia , Animais , Bortezomib , Diferenciação Celular , Feminino , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos C57BL
18.
Ann N Y Acad Sci ; 1116: 174-81, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18083927

RESUMO

The completed skeleton undergoes continuous remodeling for the duration of adult life. Rates of bone formation by osteoblasts and bone resorption by osteoclasts determine adult bone mass. Abnormalities in either the osteoblast or osteoclast compartment affect bone mass and result in skeletal disorders, the most common of which is osteoporosis, a state of low bone mass. Much is known about the molecular control of bone formation and resorption from rare single gene disorders resulting in elevated or reduced bone mass. Such genetic disorders can be attributed either to osteoclast deficiencies, collectively termed "osteopetrosis," or to intrinsically elevated osteoblast activity, termed "osteosclerosis." However, an increasing need for anabolic therapies to prevent age-induced bone loss has stimulated a search for additional genes that act at the level of the osteoblast to regulate matrix synthesis. Recently, we have discovered a zinc finger adaptor protein called Schnurri-3 (Shn3) that potently regulates adult bone mass. Mice that lack Shn3 have normal skeletal morphogenesis but display profoundly elevated bone mass that increases with age. The molecular mechanism was revealed to be the recruitment of WWP1, a Nedd4 family E3 ubiquitin ligase, by Shn3 to the major transcriptional regulator of the osteoblast, Runx2. In the absence of Shn3, Runx2 degradation by WWP1 is inhibited resulting in increased levels of Runx2 protein and enhanced expression of Runx2 target genes leading to increased osteoblast synthetic activity. Small molecules that inhibit Shn3 or WWP1 may be attractive candidates for the treatment of diseases of low bone mass.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Tamanho do Órgão/fisiologia , Osteogênese/fisiologia , Animais , Subunidade alfa 1 de Fator de Ligação ao Core/fisiologia , Camundongos , Ubiquitina-Proteína Ligases/fisiologia
19.
Ann Rheum Dis ; 66 Suppl 3: iii49-51, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17934096

RESUMO

Skeletal remodelling is a cyclical process where under normal physiological conditions, bone formation occurs at sites where bone resorption has previously taken place. Homeostatic remodelling of the skeleton is mediated by osteoclasts, giant multinucleated cells of haematopoietic origin that are responsible for bone resorption and osteoblasts, which originate from mesenchymal stem cells, and synthesise the matrix constituents on bone-forming surfaces.1 Proliferation, differentiation and bone remodelling activities of these cells involve a complex temporal network of growth factors, signalling proteins and transcription factors. Dysregulation of any one component may disrupt the remodelling process and contribute to the pathogenesis of common skeletal disorders, like osteoporosis and Paget's disease. Rare single gene disorders resulting in elevated bone mass due to osteoclast defects are collectively termed osteopetrosis. Rarer still are single gene disorders, collectively termed osteosclerosis, in which elevated bone mass is due to intrinsically elevated osteoblast activity.2 While we have learned much about the molecular control of skeletal formation and remodelling from these mutations, additional genes that regulate bone mass have yet to be characterised.


Assuntos
Remodelação Óssea/genética , Proteínas de Ligação a DNA/genética , Osteoblastos/fisiologia , Adulto , Animais , Proteínas de Ligação a DNA/antagonistas & inibidores , Regulação da Expressão Gênica/genética , Humanos , Células-Tronco Mesenquimais/fisiologia , Camundongos , Pessoa de Meia-Idade , Osteogênese/fisiologia , Osteoporose/tratamento farmacológico , Ubiquitina-Proteína Ligases/genética , Dedos de Zinco/genética
20.
Adv Exp Med Biol ; 602: 1-13, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17966382

RESUMO

Schnurri-3, a large zinc finger protein distantly related to Drosophila Shn, is a potent and essential regulator of adult bone formation. Mice lacking Shn3 display an osteosclerotic phenotype with profoundly increased bone mass due to augmented osteoblast activity. Shn3 controls protein levels of Runx2, the principal regulator of osteoblast differentiation, by promoting its degradation. In osteoblasts, Shn3 functions as a component of a trimeric complex between Runx2 and the E3 ubiquitin ligase WWP1. This complex inhibits Runx2 function and expression of genes involved in extracellular matrix mineralization due to the ability of WWP1 to promote Runx2 polyubiquitination and proteasome-dependent degradation. Our study reveals an essential role for Shn3 as a regulator of postnatal bone mass. Compounds designed to block Shn3/WWP1 function may be possible therapeutic agents for the treatment of osteoporosis.


Assuntos
Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Proteínas de Ligação a DNA/metabolismo , Osteoblastos/metabolismo , Osteogênese , Ubiquitina-Proteína Ligases/metabolismo , Animais , Densidade Óssea , Osso e Ossos , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica , Camundongos , Osteoblastos/citologia , Osteoclastos/metabolismo , Osteoporose/fisiopatologia , Ubiquitina-Proteína Ligases/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA