Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Trends Immunol ; 43(6): 438-448, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35550875

RESUMEN

α-Fetoprotein (AFP) is a fetal glycoprotein produced by most human hepatocellular carcinoma tumors. Research has focused on its immunosuppressive properties in pregnancy, autoimmunity, and cancer, and human AFP directly limits the viability and functionality of human natural killer (NK) cells, monocytes, and dendritic cells (DCs). AFP-altered DCs can promote the differentiation of naïve T cells into regulatory T cells. These properties may work to shield tumors from the immune system. Recent efforts to define the molecular characteristics of AFP identified key structural immunoregulatory domains and bioactive roles of AFP-bound ligands in immunomodulation. We propose that a key mechanism of AFP immunomodulation skews DC function through cellular metabolism. Delineating differences between fetal 'normal' AFP (nAFP) and tumor-derived AFP (tAFP) has uncovered a novel role for tAFP in altering metabolism via lipid-binding partners.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Carcinoma Hepatocelular/metabolismo , Células Dendríticas , Femenino , Humanos , Inmunomodulación , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Embarazo , alfa-Fetoproteínas/metabolismo
2.
J Biol Chem ; 293(52): 19942-19956, 2018 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-30355733

RESUMEN

We previously reported that transcription of the human IL1B gene, encoding the proinflammatory cytokine interleukin 1ß, depends on long-distance chromatin looping that is stabilized by a mutual interaction between the DNA-binding domains (DBDs) of two transcription factors: Spi1 proto-oncogene at the promoter and CCAAT enhancer-binding protein (C/EBPß) at a far-upstream enhancer. We have also reported that the C-terminal tail sequence beyond the C/EBPß leucine zipper is critical for its association with Spi1 via an exposed residue (Arg-232) located within a pocket at one end of the Spi1 DNA-recognition helix. Here, combining in vitro interaction studies with computational docking and molecular dynamics of existing X-ray structures for the Spi1 and C/EBPß DBDs, along with the C/EBPß C-terminal tail sequence, we found that the tail sequence is intimately associated with Arg-232 of Spi1. The Arg-232 pocket was computationally screened for small-molecule binding aimed at IL1B transcription inhibition, yielding l-arginine, a known anti-inflammatory amino acid, revealing a potential for disrupting the C/EBPß-Spi1 interaction. As evaluated by ChIP, cultured lipopolysaccharide (LPS)-activated THP-1 cells incubated with l-arginine had significantly decreased IL1B transcription and reduced C/EBPß's association with Spi1 on the IL1B promoter. No significant change was observed in direct binding of either Spi1 or C/EBPß to cognate DNA and in transcription of the C/EBPß-dependent IL6 gene in the same cells. These results support the notion that disordered sequences extending from a leucine zipper can mediate protein-protein interactions and can serve as druggable targets for regulating gene promoter activity.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Interleucina-1beta/genética , Mapas de Interacción de Proteínas , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo , Activación Transcripcional , Animales , Sitios de Unión , Proteína beta Potenciadora de Unión a CCAAT/química , Línea Celular , Cristalografía por Rayos X , Humanos , Ratones , Simulación del Acoplamiento Molecular , Regiones Promotoras Genéticas , Conformación Proteica , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/química , Transactivadores/química
3.
Cytokine ; 111: 373-381, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30300855

RESUMEN

Interleukin 1ß is a pro-inflammatory cytokine important for both normal immune responses and chronic inflammatory diseases. The regulation of the 31 kDa proIL-1ß precursor coded by the IL1B gene has been extensively studied in myeloid cells, but not in lymphoid-derived CD4 T cells. Surprisingly, we found that some CD4 T cell subsets express higher levels of proIL-1ß than unstimulated monocytes, despite relatively low IL1B mRNA levels. We observed a significant increase in IL1B transcription and translation in CD4 T cells upon ex vivo CD3/CD28 activation, and a similar elevation in the CCR5+ effector memory population compared to CCR5- T cells in vivo. The rapid and vigorous increase in IL1B gene transcription for stimulated monocytes has previously been associated with the presence of Spi-1/PU.1 (Spi1), a myeloid-lineage transcription factor, pre-bound to the promoter. In the case of CD4 T cells, this increase occurred despite the lack of detectable Spi1 at the IL1B promoter. Additionally, we found altered epigenetic regulation of the IL1B locus in CD3/CD28-activated CD4 T cells. Unlike monocytes, activated CD4 T cells possess bivalent H3K4me3+/H3K27me3+ nucleosome marks at the IL1B promoter, reflecting low transcriptional activity. These results support a model in which the IL1B gene in CD4 T cells is transcribed from a low-activity bivalent promoter independent of Spi1. Accumulated cytoplasmic proIL-1ß may ultimately be cleaved to mature 17 kDa bioactive IL-1ß, regulating T cell polarization and pathogenic chronic inflammation.


Asunto(s)
Linfocitos T CD4-Positivos/fisiología , Interleucina-1beta/genética , Monocitos/fisiología , Transcripción Genética/genética , Biomarcadores/metabolismo , Antígenos CD28/genética , Complejo CD3/genética , Epigénesis Genética/genética , Regulación de la Expresión Génica/genética , Humanos , Nucleosomas/genética , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas/genética , ARN Mensajero/genética , Receptores CCR5/genética , Activación Transcripcional/genética
4.
Cytokine ; 64(1): 404-12, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23800789

RESUMEN

Prostaglandin E2 (PGE2), IL-23 and IL-1ß are implicated in inflammatory bowel disease susceptibility, likely in part by modulating IL-17 producing CD4(+) T helper (Th17) cells. To better understand how these three mediators affect Th17 cell memory responses, we characterized the gene expression profiles of activated human peripheral CD4(+) effector memory T cells and sorted Th17 memory cells from healthy donors concurrent with IL17A mRNA induction mediated by PGE2 and/or IL-23 plus IL-1ß. We discovered that PGE2 and IL-23 plus IL-1ß differentially regulate Th17 cytokine expression and synergize to induce IL-17A, but not IL-17F. IL-23 plus IL-1ß preferentially induce IL-17F expression. The addition of PGE2 to IL-23 plus IL-1ß only enhances IL-17A expression as mediated by the PGE2 EP4 receptor, and promotes a switch from an IL-17F to an IL-17A predominant immune response. The human Th17 HuT-102 cell line was also found to constitutively express IL-17A, but not IL-17F. We went on to show that the IL17A and IL17F loci have divergent epigenetic architectures in unstimulated HuT-102 and primary Th17 cells and are poised for preferential expression of IL17A. We conclude that the chromatin for IL17A and IL17F are distinctly regulated, which may play an important role in mucosal health and disease.


Asunto(s)
Histonas/metabolismo , Interleucina-17/genética , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Células Th17/metabolismo , Línea Celular , Cromatina/genética , Cromatina/metabolismo , AMP Cíclico/metabolismo , Histonas/genética , Humanos , Memoria Inmunológica/inmunología , Interleucina-17/metabolismo , Interleucina-1beta/metabolismo , Interleucina-23/metabolismo , Prostaglandinas E/metabolismo , ARN Mensajero/biosíntesis , Transducción de Señal/inmunología , Células Th17/inmunología
5.
Nat Commun ; 14(1): 7211, 2023 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-37938561

RESUMEN

Efficacy of cancer vaccines remains low and mechanistic understanding of antigen presenting cell function in cancer may improve vaccine design and outcomes. Here, we analyze the transcriptomic and immune-metabolic profiles of Dendritic Cells (DCs) from 35 subjects enrolled in a trial of DC vaccines in late-stage melanoma (NCT01622933). Multiple platforms identify metabolism as an important biomarker of DC function and patient overall survival (OS). We demonstrate multiple immune and metabolic gene expression pathway alterations, a functional decrease in OCR/OXPHOS and increase in ECAR/glycolysis in patient vaccines. To dissect molecular mechanisms, we utilize single cell SCENITH functional profiling and show patient clinical outcomes (OS) correlate with DC metabolic profile, and that metabolism is linked to immune phenotype. With single cell metabolic regulome profiling, we show that MCT1 (monocarboxylate transporter-1), a lactate transporter, is increased in patient DCs, as is glucose uptake and lactate secretion. Importantly, pre-vaccination circulating myeloid cells in patients used as precursors for DC vaccine generation are significantly skewed metabolically as are several DC subsets. Together, we demonstrate that the metabolic profile of DC is tightly associated with the immunostimulatory potential of DC vaccines from cancer patients. We link phenotypic and functional metabolic changes to immune signatures that correspond to suppressed DC differentiation.


Asunto(s)
Vacunas contra el Cáncer , Melanoma , Humanos , Melanoma/terapia , Metabolómica , Investigadores , Células Dendríticas
6.
Cancer Res ; 83(9): 1543-1557, 2023 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-36847613

RESUMEN

α-Fetoprotein (AFP) is expressed by stem-like and poor outcome hepatocellular cancer tumors and is a clinical tumor biomarker. AFP has been demonstrated to inhibit dendritic cell (DC) differentiation and maturation and to block oxidative phosphorylation. To identify the critical metabolic pathways leading to human DC functional suppression, here, we used two recently described single-cell profiling methods, scMEP (single-cell metabolic profiling) and SCENITH (single-cell energetic metabolism by profiling translation inhibition). Glycolytic capacity and glucose dependence of DCs were significantly increased by tumor-derived, but not normal cord blood-derived, AFP, leading to increased glucose uptake and lactate secretion. Key molecules in the electron transport chain in particular were regulated by tumor-derived AFP. These metabolic changes occurred at mRNA and protein levels, with negative impact on DC stimulatory capacity. Tumor-derived AFP bound significantly more polyunsaturated fatty acids (PUFA) than cord blood-derived AFP. PUFAs bound to AFP increased metabolic skewing and promoted DC functional suppression. PUFAs inhibited DC differentiation in vitro, and ω-6 PUFAs conferred potent immunoregulation when bound to tumor-derived AFP. Together, these findings provide mechanistic insights into how AFP antagonizes the innate immune response to limit antitumor immunity. SIGNIFICANCE: α-Fetoprotein (AFP) is a secreted tumor protein and biomarker with impact on immunity. Fatty acid-bound AFP promotes immune suppression by skewing human dendritic cell metabolism toward glycolysis and reduced immune stimulation.


Asunto(s)
Neoplasias Hepáticas , alfa-Fetoproteínas , Humanos , alfa-Fetoproteínas/genética , alfa-Fetoproteínas/metabolismo , Neoplasias Hepáticas/patología , Ácidos Grasos Insaturados/metabolismo , Ácidos Grasos/metabolismo , Biomarcadores/metabolismo , Células Dendríticas
7.
Sci Transl Med ; 14(670): eabo4632, 2022 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-36350992

RESUMEN

Cancer vaccines have been shown clinically to drive tumor-reactive cell activation, proliferation, and effector function. Unfortunately, tumor eradication by treatment with cancer vaccines has been unsuccessful in many patients. Critical steps are under way to improve vaccine efficacy and combine them with immunotherapy and standard-of-care treatments.


Asunto(s)
Vacunas contra el Cáncer , Neoplasias , Humanos , Vacunas contra el Cáncer/uso terapéutico , Inmunoterapia , Neoplasias/terapia
8.
Nat Commun ; 13(1): 5184, 2022 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-36056019

RESUMEN

Cellular metabolism underpins immune cell functionality, yet our understanding of metabolic influences in human dendritic cell biology and their ability to orchestrate immune responses is poorly developed. Here, we map single-cell metabolic states and immune profiles of inflammatory and tolerogenic monocytic dendritic cells using recently developed multiparametric approaches. Single-cell metabolic pathway activation scores reveal simultaneous engagement of multiple metabolic pathways in distinct monocytic dendritic cell differentiation stages. GM-CSF/IL4-induce rapid reprogramming of glycolytic monocytes and transient co-activation of mitochondrial pathways followed by TLR4-dependent maturation of dendritic cells. Skewing of the mTOR:AMPK phosphorylation balance and upregulation of OXPHOS, glycolytic and fatty acid oxidation metabolism underpin metabolic hyperactivity and an immunosuppressive phenotype of tolerogenic dendritic cells, which exhibit maturation-resistance and a de-differentiated immune phenotype marked by unique immunoregulatory receptor signatures. This single-cell dataset provides important insights into metabolic pathways impacting the immune profiles of human dendritic cells.


Asunto(s)
Células Dendríticas , Monocitos , Diferenciación Celular , Glucólisis , Humanos , Monocitos/metabolismo , Fosforilación Oxidativa
9.
J Extracell Vesicles ; 10(12): e12155, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34669267

RESUMEN

Extracellular vesicles (EVs) are characterized by complex cargo composition and carry a wide array of signalling cargo, including growth factors (GFs). Beyond surface-associated GFs, it is unclear if EV intralumenal growth factors are biologically active. Here, bone morphogenetic protein-2 (BMP2), loaded directly into the lumen of EVs designated engineered BMP2-EVs (eBMP2-EVs), was comprehensively characterized including its regulation of osteoblastogenesis. eBMP2-EVs and non-EV 'free' BMP2 were observed to similarly regulate osteoblastogenesis. Furthermore, cell trafficking experiments suggest rapid BMP2 recycling and its extracellular release as 'free' BMP2 and natural occurring BMP2-EVs (nBMP2-EVs), with both being osteogenic. Interestingly, BMP2 occurs on the EV surface of nBMP2-EVs and is susceptible to proteolysis, inhibition by noggin and complete dissociation from nBMP2-EVs over 3 days. Whereas, within the eBMP2-EVs, BMP2 is protected from proteolysis, inhibition by noggin and is retained in EV lumen at 100% for the first 24 h and ∼80% after 10 days. Similar to 'free' BMP2, bioprinted eBMP2-EV microenvironments induced osteogenesis in vitro and in vivo in spatial registration to the printed patterns. Taken together, BMP2 signalling involves dynamic BMP2 cell trafficking in and out of the cell involving EVs, with distinct differences between these nBMP2-EVs and eBMP2-EVs attributable to the BMP2 cargo location with EVs. Lastly, eBMP2-EVs appear to deliver BMP2 directly into the cytoplasm, initiating BMP2 signalling within the cell, bypassing its cell surface receptors.


Asunto(s)
Proteína Morfogenética Ósea 2/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Vesículas Extracelulares/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Humanos , Masculino , Ratones , Transducción de Señal
10.
J Bone Miner Res ; 35(1): 181-195, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31487061

RESUMEN

Key osteoclast (OCL) regulatory gene promoters in bone marrow-derived monocytes harbor bivalent histone modifications that combine activating Histone 3 lysine 4 tri-methyl (H3K4me3) and repressive H3K27me3 marks, which upon RANKL stimulation resolve into repressive or activating architecture. Enhancer of zeste homologue 2 (EZH2) is the histone methyltransferase component of the polycomb repressive complex 2, which catalyzes H3K27me3 modifications. Immunofluorescence microscopy reveals that EZH2 localization during murine osteoclastogenesis is dynamically regulated. Using EZH2 knockdown and small molecule EZH2 inhibitor GSK126, we show that EZH2 plays a critical epigenetic role in OCL precursors (OCLp) during the first 24 hours of RANKL activation. RANKL triggers EZH2 translocation into the nucleus where it represses OCL-negative regulators MafB, Irf8, and Arg1. Consistent with its cytoplasmic localization in OCLp, EZH2 methyltransferase activity is required during early RANKL signaling for phosphorylation of AKT, resulting in downstream activation of the mTOR complex, which is essential for induction of OCL differentiation. Inhibition of RANKL-induced pmTOR-pS6RP signaling by GSK126 altered the translation ratio of the C/EBPß-LAP and C/EBPß-LIP isoforms and reduced nuclear translocation of the inhibitory C/EBPß-LIP, which is necessary for transcriptional repression of the OCL negative-regulatory transcription factor MafB. EZH2 in multinucleated OCL is primarily cytoplasmic and mature OCL cultured on bone segments in the presence of GSK126 exhibit defective cytoskeletal architecture and reduced resorptive activity. Here we present new evidence that EZH2 plays epigenetic and cytoplasmic roles during OCL differentiation by suppressing MafB transcription and regulating early phases of PI3K-AKT-mTOR-mediated RANKL signaling, respectively. Consistent with its cytoplasmic localization, EZH2 is required for cytoskeletal dynamics during resorption by mature OCL. Thus, EZH2 exhibits complex roles in supporting osteoclast differentiation and function. © 2019 American Society for Bone and Mineral Research.


Asunto(s)
Resorción Ósea , Osteoclastos , Animales , Resorción Ósea/genética , Diferenciación Celular , Proteína Potenciadora del Homólogo Zeste 2 , Epigénesis Genética , Ratones , Fosfatidilinositol 3-Quinasas
11.
Cancer Immunol Res ; 8(12): 1554-1567, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33051240

RESUMEN

Therapeutic cancer vaccines targeting melanoma-associated antigens are commonly immunogenic but are rarely effective in promoting objective clinical responses. To identify critical molecules for activation of effective antitumor immunity, we have profiled autologous dendritic cell (DC) vaccines used to treat 35 patients with melanoma. We showed that checkpoint molecules induced by ex vivo maturation correlated with in vivo DC vaccine activity. Melanoma patient DCs had reduced expression of cell surface inducible T-cell costimulator ligand (ICOSL) and had defective intrinsic NF-κB signaling. Chromatin immunoprecipitation assays revealed NF-κB-dependent transcriptional regulation of ICOSL expression by DCs. Blockade of ICOSL on DCs reduced priming of antigen-specific CD8+ and CD4+ T cells from naïve donors in vitro Concentration of extracellular/soluble ICOSL released from vaccine DCs positively correlated with patient clinical outcomes, which we showed to be partially regulated by ADAM10/17 sheddase activity. These data point to the critical role of canonical NF-κB signaling, the regulation of matrix metalloproteinases, and DC-derived ICOSL in the specific priming of cognate T-cell responses in the cancer setting. This study supports the implementation of targeted strategies to augment these pathways for improved immunotherapeutic outcomes in patients with cancer.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Células Dendríticas/trasplante , Ligando Coestimulador de Linfocitos T Inducibles/metabolismo , Melanoma/terapia , Subunidad p50 de NF-kappa B/metabolismo , Neoplasias Cutáneas/terapia , Adulto , Anciano , Anciano de 80 o más Años , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/genética , Terapia Combinada/métodos , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Femenino , Humanos , Inmunogenicidad Vacunal , Ligando Coestimulador de Linfocitos T Inducibles/genética , Interferón-alfa/administración & dosificación , Masculino , Melanoma/inmunología , Melanoma/mortalidad , Persona de Mediana Edad , Subunidad p50 de NF-kappa B/genética , Pronóstico , Supervivencia sin Progresión , Transducción de Señal/efectos de los fármacos , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/mortalidad , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología
12.
J Exp Med ; 217(7)2020 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-32369107

RESUMEN

Immune and molecular profiling of CD8 T cells of patients receiving DC vaccines expressing three full-length melanoma antigens (MAs) was performed. Antigen expression levels in DCs had no significant impact on T cell or clinical responses. Patients who received checkpoint blockade before DC vaccination had higher baseline MA-specific CD8 T cell responses but no evidence for improved functional responses to the vaccine. Patients who showed the best clinical responses had low PD-1 expression on MA-specific T cells before and after DC vaccination; however, blockade of PD-1 during antigen presentation by DC had minimal functional impact on PD-1high MA-specific T cells. Gene and protein expression analyses in lymphocytes and tumor samples identified critical immunoregulatory pathways, including CTLA-4 and PD-1. High immune checkpoint gene expression networks correlated with inferior clinical outcomes. Soluble serum PD-L2 showed suggestive positive association with improved outcome. These findings show that checkpoint molecular pathways are critical for vaccine outcomes and suggest specific sequencing of vaccine combinations.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Inhibidores de Puntos de Control Inmunológico/farmacología , Activación de Linfocitos/inmunología , Antígenos de Neoplasias/metabolismo , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Antígeno CTLA-4/metabolismo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Supervivencia sin Enfermedad , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Interferón gamma/metabolismo , Activación de Linfocitos/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Antígeno MART-1/metabolismo , Melanoma/sangre , Melanoma/genética , Melanoma/inmunología , Melanoma/patología , Receptor de Muerte Celular Programada 1/metabolismo , Vacunación
13.
Mol Cancer Ther ; 19(2): 420-431, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31784454

RESUMEN

Bone is the most preferred site for colonization of metastatic breast cancer cells for each subtype of the disease. The standard of therapeutic care for breast cancer patients with bone metastasis includes bisphosphonates (e.g., zoledronic acid), which have poor oral bioavailability, and a humanized antibody (denosumab). However, these therapies are palliative, and a subset of patients still develop new bone lesions and/or experience serious adverse effects. Therefore, a safe and orally bioavailable intervention for therapy of osteolytic bone resorption is still a clinically unmet need. This study demonstrates suppression of breast cancer-induced bone resorption by a small molecule (sulforaphane, SFN) that is safe clinically and orally bioavailable. In vitro osteoclast differentiation was inhibited in a dose-dependent manner upon addition of conditioned media from SFN-treated breast cancer cells representative of different subtypes. Targeted microarrays coupled with interrogation of The Cancer Genome Atlas data set revealed a novel SFN-regulated gene signature involving cross-regulation of runt-related transcription factor 2 (RUNX2) and nuclear factor-κB and their downstream effectors. Both RUNX2 and p65/p50 expression were higher in human breast cancer tissues compared with normal mammary tissues. RUNX2 was recruited at the promotor of NFKB1 Inhibition of osteoclast differentiation by SFN was augmented by doxycycline-inducible stable knockdown of RUNX2. Oral SFN administration significantly increased the percentage of bone volume/total volume of affected bones in the intracardiac MDA-MB-231-Luc model indicating in vivo suppression of osteolytic bone resorption by SFN. These results indicate that SFN is a novel inhibitor of breast cancer-induced osteolytic bone resorption in vitro and in vivo.


Asunto(s)
Neoplasias Óseas/secundario , Resorción Ósea/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Redes Reguladoras de Genes/genética , Isotiocianatos/uso terapéutico , Animales , Femenino , Humanos , Isotiocianatos/farmacología , Ratones , Sulfóxidos
14.
JBMR Plus ; 3(3): e10183, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30918921

RESUMEN

Multiple myeloma (MM) bone disease is characterized by the development of osteolytic lesions, which cause severe complications affecting the morbidity, mortality, and treatment of myeloma patients. Myeloma tumors seeded within the bone microenvironment promote hyperactivation of osteoclasts and suppression of osteoblast differentiation. Because of this prolonged suppression of bone marrow stromal cells' (BMSCs) differentiation into functioning osteoblasts, bone lesions in patients persist even in the absence of active disease. Current antiresorptive therapy provides insufficient bone anabolic effects to reliably repair MM lesions. It has become widely accepted that myeloma-exposed BMSCs have an altered phenotype with pro-inflammatory, immune-modulatory, anti-osteogenic, and pro-adipogenic properties. In this review, we focus on the role of epigenetic-based modalities in the establishment and maintenance of myeloma-induced suppression of osteogenic commitment of BMSCs. We will focus on recent studies demonstrating the involvement of chromatin-modifying enzymes in transcriptional repression of osteogenic genes in MM-BMSCs. We will further address the epigenetic plasticity in the differentiation commitment of osteoprogenitor cells and assess the involvement of chromatin modifiers in MSC-lineage switching from osteogenic to adipogenic in the context of the inflammatory myeloma microenvironment. Lastly, we will discuss the potential of employing small molecule epigenetic inhibitors currently used in the MM research as therapeutics and bone anabolic agents in the prevention or repair of osteolytic lesions in MM. © 2019 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.

15.
J Bone Oncol ; 13: 62-70, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30591859

RESUMEN

Multiple myeloma (MM) is the most frequent cancer to involve the skeleton with patients developing osteolytic bone lesions due to hyperactivation of osteoclasts and suppression of BMSCs differentiation into functional osteoblasts. Although new therapies for MM have greatly improved survival, MM remains incurable for most patients. Despite the major advances in current anti-MM and anti-resorptive treatments that can significantly improve osteolytic bone lysis, many bone lesions can persist even after therapeutic remission of active disease. Bone marrow mesenchymal stem cells (BMSCs) from MM patients are phenotypically distinct from their healthy counterparts and the mechanisms associated with the long-term osteogenic suppression are largely unknown. In this review we will highlight recent results of transcriptomic profiling studies that provide new insights into the establishment and maintenance of the persistent pathological alterations in MM-BMSCs that occur in MM. We will we discuss the role of genomic instabilities and senescence in propagating the chronically suppressed state and pro-inflammatory phenotype associated with MM-BMSCs. Lastly we describe the role of epigenetic-based mechanisms in regulating osteogenic gene expression to establish and maintain the pro-longed suppression of MM-BMSC differentiation into functional OBs.

16.
Artículo en Inglés | MEDLINE | ID: mdl-29971044

RESUMEN

Semaphorin 4D (Sema4D; CD100) is a transmembrane homodimer 150-kDa glycoprotein member of the Semaphorin family. Semaphorins were first identified as chemorepellants that guide neural axon growth. Sema4D also possesses immune regulatory activity. Recent data suggest other Sema4D functions: inactivation of platelets, stimulation of angiogenesis, and regulation of bone formation. Sema4D is a coupling factor expressed on osteoclasts that inhibits osteoblast differentiation. Blocking Sema4D may, therefore, be anabolic for bone. Sema4D and its receptor Plexin-B1 are commonly dysregulated in cancers, suggesting roles in cancer progression, invasion, tumor angiogenesis, and skeletal metastasis. This review focuses on Sema4D in bone and cancer biology and the molecular pathways involved, particularly Sema4D-Plexin-B1 signaling crosstalk between cancer cells and the bone marrow microenvironment-pertinent areas since a humanized Sema4D-neutralizing antibody is now in early phase clinical trials in cancers and neurological disorders.

17.
Artículo en Inglés | MEDLINE | ID: mdl-30008697

RESUMEN

Multiple myeloma bone disease (MMBD) is characterized by non-healing lytic bone lesions that persist even after a patient has achieved a hematologic remission. We previously reported that p62 (sequestosome-1) in bone marrow stromal cells (BMSC) is critical for the formation of MM-induced signaling complexes that mediate OB suppression. Importantly, XRK3F2, an inhibitor of the p62-ZZ domain, blunted MM-induced Runx2 suppression in vitro, and induced new bone formation and remodeling in the presence of tumor in vivo. Additionally, we reported that MM cells induce the formation of repressive chromatin on the Runx2 gene in BMSC via direct binding of the transcriptional repressor GFI1, which recruits the histone modifiers, histone deacetylase 1 (HDAC1) and Enhancer of zeste homolog 2 (EZH2). In this study we investigated the mechanism by which blocking p62-ZZ domain-dependent signaling prevents MM-induced suppression of Runx2 in BMSC. XRK3F2 prevented MM-induced upregulation of Gfi1 and repression of the Runx2 gene when present in MM-preOB co-cultures. We also show that p62-ZZ-domain blocking by XRK3F2 also prevented MM conditioned media and TNF plus IL7-mediated Gfi1 mRNA upregulation and the concomitant Runx2 repression, indicating that XRK3F2's prevention of p62-ZZ domain signaling within preOB is involved in the response. Chromatin immunoprecipitation (ChIP) analyses revealed that XRK3F2 decreased MM-induced GFI1 occupancy at the Runx2-P1 promoter and prevented recruitment of HDAC1, thus preserving the transcriptionally permissive chromatin mark H3K9ac on Runx2 and allowing osteogenic differentiation. Furthermore, treatment of MM-exposed preOB with XRK3F2 after MM removal decreased GFI1 enrichment at Runx2-P1 and rescued MM-induced suppression of Runx2 mRNA and its downstream osteogenic gene targets together with increased osteogenic differentiation. Further, primary BMSC (hBMSC) from MM patients (MM-hBMSC) had little ability to increase H3K9ac on the Runx2 promoter in osteogenic conditions when compared to hBMSC from healthy donors (HD). XRK3F2 treatment enriched Runx2 gene H3K9ac levels in MM-hBMSC to the level observed in HD-hBMSC, but did not alter HD-hBMSC H3K9ac. Importantly, XRK3F2 treatment of long-term MM-hBMSC cultures rescued osteogenic differentiation and mineralization. Our data show that blocking p62-ZZ domain-dependent signaling with XRK3F2 can reverse epigenetic-based mechanisms of MM-induced Runx2 suppression and promote osteogenic differentiation.

18.
Mol Cancer Res ; 15(4): 405-417, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28119431

RESUMEN

In multiple myeloma, osteolytic lesions rarely heal because of persistent suppressed osteoblast differentiation resulting in a high fracture risk. Herein, chromatin immunoprecipitation analyses reveal that multiple myeloma cells induce repressive epigenetic histone changes at the Runx2 locus that prevent osteoblast differentiation. The most pronounced multiple myeloma-induced changes were at the Runx2-P1 promoter, converting it from a poised bivalent state to a repressed state. Previously, it was observed that multiple myeloma induces the transcription repressor GFI1 in osteoblast precursors, which correlates with decreased Runx2 expression, thus prompting detailed characterization of the multiple myeloma and TNFα-dependent GFI1 response element within the Runx2-P1 promoter. Further analyses reveal that multiple myeloma-induced GFI1 binding to Runx2 in osteoblast precursors and recruitment of the histone modifiers HDAC1, LSD1, and EZH2 is required to establish and maintain Runx2 repression in osteogenic conditions. These GFI1-mediated repressive chromatin changes persist even after removal of multiple myeloma. Ectopic GFI1 is sufficient to bind to Runx2, recruit HDAC1 and EZH2, increase H3K27me3 on the gene, and prevent osteogenic induction of endogenous Runx2 expression. Gfi1 knockdown in MC4 cells blocked multiple myeloma-induced recruitment of HDAC1 and EZH2 to Runx2, acquisition of repressive chromatin architecture, and suppression of osteoblast differentiation. Importantly, inhibition of EZH2 or HDAC1 activity in pre-osteoblasts after multiple myeloma exposure in vitro or in osteoblast precursors from patients with multiple myeloma reversed the repressive chromatin architecture at Runx2 and rescued osteoblast differentiation.Implications: This study suggests that therapeutically targeting EZH2 or HDAC1 activity may reverse the profound multiple myeloma-induced osteoblast suppression and allow repair of the lytic lesions. Mol Cancer Res; 15(4); 405-17. ©2017 AACR.


Asunto(s)
Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Proteínas de Unión al ADN/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Histona Desacetilasa 1/metabolismo , Mieloma Múltiple/genética , Osteoblastos/citología , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Técnicas de Cocultivo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Proteínas de Unión al ADN/genética , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Epigénesis Genética , Histona Desacetilasa 1/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas/administración & dosificación , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Indoles/administración & dosificación , Indoles/farmacología , Ratones , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/metabolismo , Osteoblastos/efectos de los fármacos , Regiones Promotoras Genéticas , Piridonas/administración & dosificación , Piridonas/farmacología , Factores de Transcripción/genética
19.
PLoS One ; 8(8): e70622, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23936458

RESUMEN

Interleukin-1ß and Tumor Necrosis Factor α play related, but distinct, roles in immunity and disease. Our study revealed major mechanistic distinctions in the Toll-like receptor (TLR) signaling-dependent induction for the rapidly expressed genes (IL1B and TNF) coding for these two cytokines. Prior to induction, TNF exhibited pre-bound TATA Binding Protein (TBP) and paused RNA Polymerase II (Pol II), hallmarks of poised immediate-early (IE) genes. In contrast, unstimulated IL1B displayed very low levels of both TBP and paused Pol II, requiring the lineage-specific Spi-1/PU.1 (Spi1) transcription factor as an anchor for induction-dependent interaction with two TLR-activated transcription factors, C/EBPß and NF-κB. Activation and DNA binding of these two pre-expressed factors resulted in de novo recruitment of TBP and Pol II to IL1B in concert with a permissive state for elongation mediated by the recruitment of elongation factor P-TEFb. This Spi1-dependent mechanism for IL1B transcription, which is unique for a rapidly-induced/poised IE gene, was more dependent upon P-TEFb than was the case for the TNF gene. Furthermore, the dependence on phosphoinositide 3-kinase for P-TEFb recruitment to IL1B paralleled a greater sensitivity to the metabolic state of the cell and a lower sensitivity to the phenomenon of endotoxin tolerance than was evident for TNF. Such differences in induction mechanisms argue against the prevailing paradigm that all IE genes possess paused Pol II and may further delineate the specific roles played by each of these rapidly expressed immune modulators.


Asunto(s)
Genes Inmediatos-Precoces/genética , Interleucina-1beta/genética , Activación Transcripcional , Factor de Necrosis Tumoral alfa/genética , Animales , Transporte Biológico/efectos de los fármacos , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Ontología de Genes , Sitios Genéticos/genética , Glucosa/metabolismo , Células HEK293 , Humanos , Lipopolisacáridos/farmacología , Ratones , Monocitos/metabolismo , Nucleosomas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Factor B de Elongación Transcripcional Positiva/metabolismo , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas/metabolismo , ARN Polimerasa II/química , ARN Polimerasa II/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA