Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 134(14): 1176-1189, 2019 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-31383640

RESUMEN

Dysregulation of polycomb repressive complex 2 (PRC2) promotes oncogenesis partly through its enzymatic function for inducing trimethylation of histone H3 lysine 27 (H3K27me3). However, it remains to be determined how PRC2 activity is regulated in normal and diseased settings. We here report a PRC2-associated cofactor, PHD finger protein 19 (PHF19; also known as polycomb-like 3), as a crucial mediator of tumorigenicity in multiple myeloma (MM). Overexpression and/or genomic amplification of PHF19 is found associated with malignant progression of MM and plasma cell leukemia, correlating to worse treatment outcomes. Using various MM models, we demonstrated a critical requirement of PHF19 for tumor growth in vitro and in vivo. Mechanistically, PHF19-mediated oncogenic effect relies on its PRC2-interacting and chromatin-binding functions. Chromatin immunoprecipitation followed by sequencing profiling showed a critical role for PHF19 in maintaining the H3K27me3 landscape. PHF19 depletion led to loss of broad H3K27me3 domains, possibly due to impaired H3K27me3 spreading from cytosine guanine dinucleotide islands, which is reminiscent to the reported effect of an "onco"-histone mutation, H3K27 to methionine (H3K27M). RNA-sequencing-based transcriptome profiling in MM lines also demonstrated a requirement of PHF19 for optimal silencing of PRC2 targets, which include cell cycle inhibitors and interferon-JAK-STAT signaling genes critically involved in tumor suppression. Correlation studies using patient sample data sets further support a clinical relevance of the PHF19-regulated pathways. Lastly, we show that MM cells are generally sensitive to PRC2 inhibitors. Collectively, this study demonstrates that PHF19 promotes MM tumorigenesis through enhancing H3K27me3 deposition and PRC2's gene-regulatory functions, lending support for PRC2 blockade as a means for MM therapeutics.


Asunto(s)
Carcinogénesis/metabolismo , Proteínas de Unión al ADN/metabolismo , Histonas/metabolismo , Mieloma Múltiple/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Factores de Transcripción/metabolismo , Animales , Carcinogénesis/patología , Línea Celular Tumoral , Humanos , Metilación , Ratones , Mieloma Múltiple/patología
2.
Blood ; 128(25): 2949-2959, 2016 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-27733356

RESUMEN

To begin to understand the mechanisms that regulate self-renewal, differentiation, and transformation of human hematopoietic stem cells or to evaluate the efficacy of novel treatment modalities, stem cells need to be studied in their own species-specific microenvironment. By implanting ceramic scaffolds coated with human mesenchymal stromal cells into immune-deficient mice, we were able to mimic the human bone marrow niche. Thus, we have established a human leukemia xenograft mouse model in which a large cohort of patient samples successfully engrafted, which covered all of the important genetic and risk subgroups. We found that by providing a humanized environment, stem cell self-renewal properties were better maintained as determined by serial transplantation assays and genome-wide transcriptome studies, and less clonal drift was observed as determined by exome sequencing. The human leukemia xenograft mouse models that we have established here will serve as an excellent resource for future studies aimed at exploring novel therapeutic approaches.


Asunto(s)
Médula Ósea/patología , Leucemia Mieloide Aguda/patología , Nicho de Células Madre , Andamios del Tejido/química , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Autorrenovación de las Células , Separación Celular , Células Clonales , Femenino , Perfilación de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Células Madre Hematopoyéticas/citología , Humanos , Leucemia Mieloide Aguda/genética , Células Madre Mesenquimatosas/citología , Ratones , Fenotipo , Células del Estroma/patología
3.
Mol Ther ; 25(8): 1946-1958, 2017 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-28506593

RESUMEN

Chimeric antigen receptors (CARs) can effectively redirect cytotoxic T cells toward highly expressed surface antigens on tumor cells. The low expression of several tumor-associated antigens (TAAs) on normal tissues, however, hinders their safe targeting by CAR T cells due to on-target/off-tumor effects. Using the multiple myeloma (MM)-associated CD38 antigen as a model system, here, we present a rational approach for effective and tumor-selective targeting of such TAAs. Using "light-chain exchange" technology, we combined the heavy chains of two high-affinity CD38 antibodies with 176 germline light chains and generated ∼124 new antibodies with 10- to >1,000-fold lower affinities to CD38. After categorizing them into three distinct affinity classes, we incorporated the single-chain variable fragments of eight antibodies from each class into new CARs. T cells carrying these CD38-CARs were extensively evaluated for their on-tumor/off-tumor cytotoxicity as well as CD38-dependent proliferation and cytokine production. We identified CD38-CAR T cells of ∼1,000- fold reduced affinity, which optimally proliferated, produced Th1-like cytokines, and effectively lysed CD382+ MM cells, but spared CD38+ healthy hematopoietic cells in vitro and in vivo. Thus, this systematic approach is highly suitable for the generation of optimal CARs for effective and selective targeting of TAAs.


Asunto(s)
ADP-Ribosil Ciclasa 1/química , ADP-Ribosil Ciclasa 1/inmunología , Receptores de Antígenos de Linfocitos T/química , Receptores de Antígenos de Linfocitos T/metabolismo , Proteínas Recombinantes de Fusión , ADP-Ribosil Ciclasa 1/metabolismo , Animales , Afinidad de Anticuerpos/inmunología , Citocinas/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Inmunoterapia Adoptiva , Activación de Linfocitos/inmunología , Ratones , Mieloma Múltiple/inmunología , Mieloma Múltiple/metabolismo , Mieloma Múltiple/terapia , Unión Proteica/inmunología , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/inmunología , Anticuerpos de Cadena Única/metabolismo , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
4.
J Immunol ; 194(3): 911-20, 2015 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-25520398

RESUMEN

The effectiveness of chimeric Ag receptor (CAR)-transduced T (CAR-T) cells has been attributed to supraphysiological signaling through CARs. Second- and later-generation CARs simultaneously transmit costimulatory signals with CD3ζ signals upon ligation, but may lead to severe adverse effects owing to the recognition of minimal Ag expression outside the target tumor. Currently, the threshold target Ag density for CAR-T cell lysis and further activation, including cytokine production, has not yet been investigated in detail. Therefore, we determined the threshold target Ag density required to induce CAR-T cell responses using novel anti-CD20 CAR-T cells with a CD28 intracellular domain and a CD20-transduced CEM cell model. The newly developed CD20CAR-T cells demonstrated Ag-specific lysis and cytokine secretion, which was a reasonable level as a second-generation CAR. For lytic activity, the threshold Ag density was determined to be ∼200 molecules per target cell, whereas the Ag density required for cytokine production of CAR-T cells was ∼10-fold higher, at a few thousand per target cell. CD20CAR-T cells responded efficiently to CD20-downregulated lymphoma and leukemia targets, including rituximab- or ofatumumab-refractory primary chronic lymphocytic leukemia cells. Despite the potential influence of the structure, localization, and binding affinity of the CAR/Ag, the threshold determined may be used for target Ag selection. An Ag density below the threshold may not result in adverse effects, whereas that above the threshold may be sufficient for practical effectiveness. CD20CAR-T cells also demonstrated significant lytic activity against CD20-downregulated tumor cells and may exhibit effectiveness for CD20-positive lymphoid malignancies.


Asunto(s)
Antígenos de Superficie/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T , Anticuerpos Monoclonales de Origen Murino/uso terapéutico , Antígenos CD20/inmunología , Antígenos CD20/metabolismo , Antígenos de Superficie/metabolismo , Antígenos CD28/inmunología , Antígenos CD28/metabolismo , Complejo CD3/inmunología , Complejo CD3/metabolismo , Línea Celular Tumoral , Citocinas/biosíntesis , Citotoxicidad Inmunológica , Expresión Génica , Orden Génico , Genes Reporteros , Humanos , Inmunofenotipificación , Espacio Intracelular , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Leucemia Linfocítica Crónica de Células B/genética , Leucemia Linfocítica Crónica de Células B/inmunología , Leucemia Linfocítica Crónica de Células B/metabolismo , Activación de Linfocitos , Linfoma de Células B/tratamiento farmacológico , Linfoma de Células B/genética , Linfoma de Células B/inmunología , Linfoma de Células B/metabolismo , Unión Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Rituximab , Transducción de Señal , Especificidad del Receptor de Antígeno de Linfocitos T/genética , Especificidad del Receptor de Antígeno de Linfocitos T/inmunología , Transducción Genética
5.
Haematologica ; 101(5): 616-25, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26858358

RESUMEN

Adoptive transfer of chimeric antigen receptor-transduced T cells is a promising strategy for cancer immunotherapy. The CD38 molecule, with its high expression on multiple myeloma cells, appears a suitable target for antibody therapy. Prompted by this, we used three different CD38 antibody sequences to generate second-generation retroviral CD38-chimeric antigen receptor constructs with which we transduced T cells from healthy donors and multiple myeloma patients. We then evaluated the preclinical efficacy and safety of the transduced T cells. Irrespective of the donor and antibody sequence, CD38-chimeric antigen receptor-transduced T cells proliferated, produced inflammatory cytokines and effectively lysed malignant cell lines and primary malignant cells from patients with acute myeloid leukemia and multi-drug resistant multiple myeloma in a cell-dose, and CD38-dependent manner, despite becoming CD38-negative during culture. CD38-chimeric antigen receptor-transduced T cells also displayed significant anti-tumor effects in a xenotransplant model, in which multiple myeloma tumors were grown in a human bone marrow-like microenvironment. CD38-chimeric antigen receptor-transduced T cells also appeared to lyse the CD38(+) fractions of CD34(+) hematopoietic progenitor cells, monocytes, natural killer cells, and to a lesser extent T and B cells but did not inhibit the outgrowth of progenitor cells into various myeloid lineages and, furthermore, were effectively controllable with a caspase-9-based suicide gene. These results signify the potential importance of CD38-chimeric antigen receptor-transduced T cells as therapeutic tools for CD38(+) malignancies and warrant further efforts to diminish the undesired effects of this immunotherapy using appropriate strategies.


Asunto(s)
ADP-Ribosil Ciclasa 1/metabolismo , Inmunoterapia , Mieloma Múltiple/inmunología , Mieloma Múltiple/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Proteínas Recombinantes de Fusión , Linfocitos T/inmunología , Linfocitos T/metabolismo , ADP-Ribosil Ciclasa 1/genética , ADP-Ribosil Ciclasa 1/inmunología , Animales , Citocinas/biosíntesis , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Citometría de Flujo , Expresión Génica , Técnicas de Transferencia de Gen , Genes Transgénicos Suicidas , Células Madre Hematopoyéticas/metabolismo , Humanos , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados , Mieloma Múltiple/patología , Mieloma Múltiple/terapia , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T/trasplante , Transducción Genética , Carga Tumoral/genética , Carga Tumoral/inmunología
6.
Cancer Immunol Immunother ; 64(8): 951-63, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25920521

RESUMEN

Immunotherapy with allogeneic natural killer (NK) cells offers therapeutic perspectives for multiple myeloma patients. Here, we aimed to refine NK cell therapy by evaluation of the relevance of HLA-class I and HLA-E for NK anti-myeloma reactivity. We show that HLA-class I was strongly expressed on the surface of patient-derived myeloma cells and on myeloma cell lines. HLA-E was highly expressed by primary myeloma cells but only marginally by cell lines. HLA-E(low) expression on U266 cells observed in vitro was strongly upregulated after in vivo (bone marrow) growth in RAG-2(-/-) γc(-/-) mice, suggesting that in vitro HLA-E levels poorly predict the in vivo situation. Concurrent analysis of inhibitory receptors (KIR2DL1, KIR2DL2/3, KIR3DL1 and NKG2A) and NK cell degranulation upon co-culture with myeloma cells revealed that KIR-ligand-mismatched NK cells degranulate more than matched subsets and that HLA-E abrogates degranulation of NKG2A+ subsets. Inhibition by HLA-class I and HLA-E was also observed with IL-2-activated NK cells and at low oxygen levels (0.6 %) mimicking hypoxic bone marrow niches where myeloma cells preferentially reside. Our study demonstrates that NKG2A-negative, KIR-ligand-mismatched NK cells are the most potent subset for clinical application. We envision that infusion of high numbers of this subclass will enhance clinical efficacy.


Asunto(s)
Separación Celular/métodos , Antígenos de Histocompatibilidad Clase I/inmunología , Inmunoterapia/métodos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/trasplante , Mieloma Múltiple/terapia , Subfamília C de Receptores Similares a Lectina de Células NK/inmunología , Animales , Degranulación de la Célula , Línea Celular Tumoral , Técnicas de Cocultivo , Citotoxicidad Inmunológica , Proteínas de Unión al ADN/genética , Citometría de Flujo , Humanos , Interleucina-2/inmunología , Ratones , Ratones Noqueados , Mieloma Múltiple/inmunología , Trasplante de Neoplasias , Oxígeno/metabolismo , Antígenos HLA-E
7.
Blood ; 120(26): 5153-62, 2012 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-23018643

RESUMEN

Immunotherapy with innate immune cells has recently evoked broad interest as a novel treatment option for cancer patients. γ9δ2T cells in particular are emerging as an innate cell population with high frequency and strong antitumor reactivity, which makes them and their receptors promising candidates for immune interventions. However, clinical trials have so far reported only limited tumor control by adoptively transferred γ9δ2T cells. As a potential explanation for this lack of efficacy, we found unexpectedly high variability in tumor recognition within the physiologic human γ9δ2T-cell repertoire, which is substantially regulated by the CDR3 domains of individual γ9δ2TCRs. In the present study, we demonstrate that the reported molecular requirements of CDR3 domains to interact with target cells shape the physiologic γ9δ2T-cell repertoire and, most likely, limit the protective and therapeutic antitumor efficacy of γ9δ2T cells. Based on these findings, we propose combinatorial-γδTCR-chain exchange as an efficient method for designing high-affinity γ9δ2TCRs that mediate improved antitumor responses when expressed in αßT cells both in vitro and in vivo in a humanized mouse model.


Asunto(s)
Genes Codificadores de la Cadena gamma de los Receptores de Linfocito T/fisiología , Cadenas gamma de Inmunoglobulina/fisiología , Especificidad del Receptor de Antígeno de Linfocitos T , Traslado Adoptivo , Animales , Epítopos de Linfocito T/genética , Epítopos de Linfocito T/fisiología , Genes Codificadores de la Cadena gamma de los Receptores de Linfocito T/genética , Humanos , Cadenas gamma de Inmunoglobulina/química , Cadenas gamma de Inmunoglobulina/genética , Inmunoterapia Adoptiva/métodos , Células K562 , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Estructura Terciaria de Proteína/fisiología , Especificidad del Receptor de Antígeno de Linfocitos T/genética , Linfocitos T/inmunología , Linfocitos T/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Blood ; 120(3): e9-e16, 2012 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-22653974

RESUMEN

Interactions within the hematopoietic niche in the BM microenvironment are essential for maintenance of the stem cell pool. In addition, this niche is thought to serve as a sanctuary site for malignant progenitors during chemotherapy. Therapy resistance induced by interactions with the BM microenvironment is a major drawback in the treatment of hematologic malignancies and bone-metastasizing solid tumors. To date, studying these interactions was hampered by the lack of adequate in vivo models that simulate the human situation. In the present study, we describe a unique human-mouse hybrid model that allows engraftment and outgrowth of normal and malignant hematopoietic progenitors by implementing a technology for generating a human bone environment. Using luciferase gene marking of patient-derived multiple myeloma cells and bioluminescent imaging, we were able to follow pMM cells outgrowth and to visualize the effect of treatment. Therapeutic interventions in this model resulted in equivalent drug responses as observed in the corresponding patients. This novel human-mouse hybrid model creates unprecedented opportunities to investigate species-specific microenvironmental influences on normal and malignant hematopoietic development, and to develop and personalize cancer treatment strategies.


Asunto(s)
Células Madre Hematopoyéticas/citología , Mieloma Múltiple/inmunología , Mieloma Múltiple/patología , Nicho de Células Madre/inmunología , Quimera por Trasplante/inmunología , Microambiente Tumoral/inmunología , Animales , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Osículos del Oído/citología , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/inmunología , Ratones , Ratones Mutantes , Trasplante de Neoplasias , Osteólisis/inmunología , Andamios del Tejido , Trasplante Heterólogo
10.
Blood ; 118(1): 50-9, 2011 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-21566093

RESUMEN

Major limitations of currently investigated αßT cells redirected against cancer by transfer of tumor-specific αßTCR arise from their low affinity, MHC restriction, and risk to mediate self-reactivity after pairing with endogenous α or ßTCR chains. Therefore, the ability of a defined γ9δ2TCR to redirect αßT cells selectively against tumor cells was tested and its molecular interaction with a variety of targets investigated. Functional analysis revealed that a γ9δ2TCR efficiently reprograms both CD4(+) and CD8(+) αßT cells against a broad panel of cancer cells while ignoring normal cells, and substantially reduces but does not completely abrogate alloreactivity. γ9δ2TCR-transduced αßT cells reduced colony formation of progenitor cells of primary acute myeloid leukemia blasts and inhibited leukemia growth in a humanized mouse model. Thereby, metabolites of a dysregulated mevalonate pathway are targeted and the additional application of widely used biphosphonates is crucial for in vivo efficacy most likely because of its modulating effect on cytokine secretion of γ9δ2TCR-transduced αßT cells. Expression of NKG2D ligands and F1-ATPase contributed to the activity of γ9δ2TCR-transduced αßT cells but were not mandatory. In summary, γ9δ2 TCRs are an attractive alternative to broadly redirect αßT cells against cancer cells with both an improved efficacy and safety profile compared with currently used αßTCRs.


Asunto(s)
Traslado Adoptivo/métodos , Terapia Genética/métodos , Leucemia Mieloide Aguda/terapia , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/trasplante , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/trasplante , Comunicación Celular/inmunología , Línea Celular Tumoral , Células Dendríticas/citología , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Humanos , Leucemia Mieloide Aguda/inmunología , Ratones , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología
11.
Biol Blood Marrow Transplant ; 18(7): 1023-35, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22579931

RESUMEN

Graft-versus-host disease (GVHD) remains a frequently occurring and difficult-to-treat complication in human allogeneic stem cell transplantation. Murine transplantation models are often used to study and understand the complex pathogenesis of GVHD and to explore new treatment strategies. Although GVHD kinetics may differ in murine and human models, adequate models are essential for identification of the crucial factors responsible for the major pathology in GVHD. We present a detailed description of the specific histological features of a graft-versus-host-induced fibrotic response in xenogeneic RAG2(-/-)γc(-/-) mice after total body irradiation and injection with human peripheral blood mononuclear cells. We describe the full morphological features of this reaction, including a detailed analysis of the specific tissue infiltration patterns of the human peripheral blood mononuclear cells. Our data show the development of fibrosis, predominantly near blood vessels, and reveal different cell populations and specific cell migration patterns in the affected organs. The combination of immunohistochemical cell characterization and mRNA expression analysis of both human (donor)- and murine (host)-derived cytokines reveals an interaction between host tissues and donor-derived cells in an entangled cytokine profile, in which both donor- and host-derived cytokines contribute to the formation of fibrosis.


Asunto(s)
Vasos Sanguíneos/patología , Enfermedad Injerto contra Huésped/patología , Trasplante de Células Madre Hematopoyéticas , Leucocitos Mononucleares/trasplante , Animales , Vasos Sanguíneos/inmunología , Movimiento Celular/inmunología , Citocinas/biosíntesis , Citocinas/inmunología , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Modelos Animales de Enfermedad , Femenino , Enfermedad Injerto contra Huésped/inmunología , Humanos , Inmunohistoquímica , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Subunidad gamma Común de Receptores de Interleucina/genética , Subunidad gamma Común de Receptores de Interleucina/inmunología , Leucocitos Mononucleares/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Esclerosis , Trasplante Heterólogo , Irradiación Corporal Total
12.
Blood ; 115(3): 601-4, 2010 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-19965677

RESUMEN

Expression of the heparan sulfate proteoglycan syndecan-1 is a hallmark of both normal and multiple myeloma (MM) plasma cells. Syndecan-1 could affect plasma cell fate by strengthening integrin-mediated adhesion via its core protein and/or by accommodating and presenting soluble factors via its HS side chains. Here, we show that inducible RNAi-mediated knockdown of syndecan-1 in human MM cells leads to reduced growth rates and a strong increase of apoptosis. Importantly, knockdown of EXT1, a copolymerase critical for HS chain biosynthesis, had similar effects. Using an innovative myeloma xenotransplantation model in Rag-2(-/-)gamma(c)(-/-) mice, we demonstrate that induction of EXT1 knockdown in vivo dramatically suppresses the growth of bone marrow localized myeloma. Our findings provide direct evidence that the HS chains of syndecan-1 are crucial for the growth and survival of MM cells within the bone marrow environment, and indicate the HS biosynthesis machinery as a potential treatment target in MM.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Heparitina Sulfato/fisiología , Mieloma Múltiple/patología , N-Acetilglucosaminiltransferasas/genética , ARN Interferente Pequeño/farmacología , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Doxiciclina/administración & dosificación , Sistemas de Liberación de Medicamentos , Marcación de Gen , Heparitina Sulfato/metabolismo , Humanos , Cadenas gamma de Inmunoglobulina/genética , Ratones , Ratones Noqueados , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/metabolismo , N-Acetilglucosaminiltransferasas/antagonistas & inhibidores , N-Acetilglucosaminiltransferasas/fisiología , Sindecano-1/genética , Sindecano-1/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Haematologica ; 96(11): 1653-61, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21828122

RESUMEN

BACKGROUND: Multiple myeloma is a hematologic malignancy characterized by a clonal expansion of malignant plasma cells in the bone marrow, which is accompanied by the development of osteolytic lesions and/or diffuse osteopenia. The intricate bi-directional interaction with the bone marrow microenvironment plays a critical role in sustaining the growth and survival of myeloma cells during tumor progression. Identification and functional analysis of the (adhesion) molecules involved in this interaction will provide important insights into the pathogenesis of multiple myeloma. DESIGN AND METHODS: Multiple myeloma cell lines and patients' samples were analyzed for expression of the adhesion molecule N-cadherin by immunoblotting, flow cytometry, immunofluorescence microscopy, immunohistochemistry and expression microarray. In addition, by means of blocking antibodies and inducible RNA interference we studied the functional consequence of N-cadherin expression for the myeloma cells, by analysis of adhesion, migration and growth, and for the bone marrow microenvironment, by analysis of osteogenic differentiation. RESULTS: The malignant plasma cells in approximately half of the multiple myeloma patients, belonging to specific genetic subgroups, aberrantly expressed the homophilic adhesion molecule N-cad-herin. N-cadherin-mediated cell-substrate or homotypic cell-cell adhesion did not contribute to myeloma cell growth in vitro. However, N-cadherin directly mediated the bone marrow localization/retention of myeloma cells in vivo, and facilitated a close interaction between myeloma cells and N-cadherin-positive osteoblasts. Furthermore, this N-cadherin-mediated interaction contributed to the ability of myeloma cells to inhibit osteoblastogenesis. CONCLUSIONS: Taken together, our data show that myeloma cells frequently display aberrant expression of N-cadherin and that N-cadherin mediates the interaction of myeloma cells with the bone marrow microenvironment, in particular the osteoblasts. This N-cadherin-mediated interaction inhibits osteoblast differentiation and may play an important role in the pathogenesis of myeloma bone disease.


Asunto(s)
Cadherinas/metabolismo , Comunicación Celular , Diferenciación Celular , Mieloma Múltiple/metabolismo , Proteínas de Neoplasias/metabolismo , Osteoblastos/metabolismo , Microambiente Tumoral , Médula Ósea/metabolismo , Médula Ósea/patología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Cadherinas/genética , Adhesión Celular/genética , Línea Celular Tumoral , Humanos , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Proteínas de Neoplasias/genética , Osteoblastos/patología
15.
Proc Natl Acad Sci U S A ; 105(20): 7281-6, 2008 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-18490653

RESUMEN

Tissue engineering of large bone defects is approached through implantation of autologous osteogenic cells, generally referred to as multipotent stromal cells or mesenchymal stem cells (MSCs). Animal-derived MSCs successfully bridge large bone defects, but models for ectopic bone formation as well as recent clinical trials demonstrate that bone formation by human MSCs (hMSCs) is inadequate. The expansion phase presents an attractive window to direct hMSCs by pharmacological manipulation, even though no profound effect on bone formation in vivo has been described so far using this approach. We report that activation of protein kinase A elicits an immediate response through induction of genes such as ID2 and FosB, followed by sustained secretion of bone-related cytokines such as BMP-2, IGF-1, and IL-11. As a consequence, PKA activation results in robust in vivo bone formation by hMSCs derived from orthopedic patients.


Asunto(s)
Huesos/metabolismo , AMP Cíclico/metabolismo , Células Madre Mesenquimatosas/citología , Ingeniería de Tejidos/métodos , Proteína Morfogenética Ósea 2 , Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Citocinas/metabolismo , Humanos , Proteína 2 Inhibidora de la Diferenciación/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Interleucina-11/metabolismo , Modelos Biológicos , Osteogénesis , Proteínas Proto-Oncogénicas c-fos/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
16.
PLoS One ; 16(10): e0258653, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34669746

RESUMEN

BACKGROUND AND AIM: A fraction of children with obesity have increased serum cortisol levels. In this study, we describe the clinical characteristics of obese children and adolescents with elevated morning serum cortisol levels and the relationship between the cortisol levels and components of the metabolic syndrome. METHODS: Retrospective medical record review study of children aged 4 to 18 years with overweight or obesity seen for obesity management in the Pediatric Obesity Clinic of the UZ Brussel between 2013 and 2015. RESULTS: A total of 234 children (99 boys and 135 girls) with overweight (BMI z-score > 1.3) without underlying endocrine or genetic conditions were included. Mean (SD) age was 10.1 (2.8) years, BMI SD-score 2.5 (0.6), and body fat percentage 37% (7.9). Serum fasting cortisol levels were elevated (>180 µg/L) in 49 children, normal (62-180 µg/L) in 168, and decreased (<62 µg/L) in 12. Serum fasting cortisol was not significantly correlated with gender, age, or degree of adiposity. But correlated significantly with fasting glucose (Rs = 0.193; p < 0.005), triglycerides (Rs = 0. 143; p < 0.05), fibrinogen (Rs = 0.144; p < 0.05) and leptin levels (Rs = 0.145; p < 0.05). After adjustment for serum insulin and leptin, the correlation between serum cortisol and fasting glucose remained significant. CONCLUSION: Elevated morning serum cortisol levels were found in 20% of overweight or obese children and adolescents, irrespective of the degree of adiposity, and were associated with higher fasting glucose, irrespective of underlying insulin resistance. The long-term cardiometabolic consequences of hypercortisolemia in childhood obesity needs further study.


Asunto(s)
Glucemia/análisis , Ayuno/sangre , Hidrocortisona/sangre , Síndrome Metabólico/diagnóstico , Sobrepeso/metabolismo , Obesidad Infantil/metabolismo , Adolescente , Bélgica , Niño , Preescolar , Femenino , Humanos , Leptina/sangre , Masculino , Síndrome Metabólico/sangre , Sobrepeso/sangre , Obesidad Infantil/sangre , Estudios Retrospectivos
17.
Blood ; 111(10): 5233-41, 2008 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-18256318

RESUMEN

Despite the earlier use of potent immunosuppressive or cytostatic drugs and the recent emergence of biologicals as treatment for human autoimmune diseases (AIDs), some patients still remain unresponsive to treatment. To those severely ill patients, autologous bone marrow transplantation (aBMT) is applied as a last resource, leading to disease remission in a majority of patients. The underlying mechanism of action of aBMT is still largely unknown. Here, we showed that regulatory T cells (Tregs) play a role in the natural disease course of proteoglycan-induced arthritis (PGIA) and in disease remission by aBMT. aBMT led to an initial phase of rapid disease improvement corresponding with a relative increase in CD4(+)CD25(+) T cells. At this time, the CD4(+)CD25(+) cells did not yet show an increase in Foxp3 expression and showed less potent suppression. After this initial improvement, disease relapsed but stabilized at a level below the severity before aBMT. This second phase was actively regulated by potently suppressive CD4(+)CD25(+)Foxp3(+) Tregs. This work provided further insight into the role of Tregs in restoration of the immune balance by aBMT and can open the way to explore therapeutic interventions to further improve treatment of AID and disease relapses.


Asunto(s)
Artritis/terapia , Trasplante de Médula Ósea/métodos , Hemostasis , Sistema Inmunológico/fisiología , Linfocitos T Reguladores/fisiología , Animales , Enfermedades Autoinmunes , Recuento de Linfocito CD4 , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Proteoglicanos , Inducción de Remisión , Trasplante Autólogo , Resultado del Tratamiento
18.
Haematologica ; 95(12): 2063-71, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20851867

RESUMEN

BACKGROUND: Incorporation of the chimeric CD20 monoclonal antibody rituximab in the treatment schedule of patients with non-Hodgkin's lymphoma has significantly improved outcome. Despite this success, about half of the patients do not respond to treatment or suffer from a relapse and additional therapy is required. A low CD20-expression level may in part be responsible for resistance against rituximab. We therefore investigated whether the CD20-expression level related resistance to rituximab could be overcome by a new group of CD20 mAbs (HuMab-7D8 and ofatumumab) targeting a unique membrane-proximal epitope on the CD20 molecule. DESIGN AND METHODS: By retroviral transduction of the CD20 gene into CD20-negative cells and clonal selection of transduced cells a system was developed in which the CD20-expression level is the only variable. These CD20 transduced cells were used to study the impact of rituximab and HuMab-7D8 mediated complement-dependent cytotoxicity. To study the in vivo efficacy of these mAbs an in vivo imaging system was generated by retroviral expression of the luciferase gene in the CD20-positive cells. RESULTS: We show that HuMab-7D8 efficiently killed CD20(low) cells that are not susceptible to rituximab-induced killing in vitro. In a mouse xenograft model, we observed a comparable increase in survival time between HuMab-7D8 and rituximab-treated mice. Most significantly, however, HuMab-7D8 eradicated all CD20-expressing cells both in the periphery as well as in the bone marrow whereas after rituximab treatment CD20(low) cells survived. CONCLUSIONS: Cells that are insensitive to in vitro and in vivo killing by rituximab as the result of their low CD20-expression profile may be efficiently killed by an antibody against the membrane-proximal epitope on CD20. Such antibodies should, therefore, be explored to overcome rituximab resistance in the clinic.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/farmacología , Anticuerpos Monoclonales/farmacología , Antígenos CD20/metabolismo , Citotoxicidad Inmunológica/efectos de los fármacos , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales de Origen Murino/inmunología , Antígenos CD20/genética , Antígenos CD20/inmunología , Línea Celular Tumoral , Citotoxicidad Inmunológica/inmunología , Epítopos/inmunología , Citometría de Flujo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Leucemia Experimental/tratamiento farmacológico , Leucemia Experimental/metabolismo , Leucemia Experimental/patología , Luciferasas/genética , Luciferasas/metabolismo , Mediciones Luminiscentes/métodos , Microdominios de Membrana/metabolismo , Ratones , Transporte de Proteínas , Rituximab , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Cell Microbiol ; 11(5): 825-41, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19215224

RESUMEN

Bioluminescence imaging (BLI) is a powerful new method to study virus dissemination in the live animal. Here we used this method to monitor the spatial and temporal progression of mouse hepatitis coronavirus (MHV) infection in mice using luciferase-expressing viruses. Upon intranasal inoculation, virus replication could initially be observed in the nasal cavity and the cervical lymph nodes, after which the infection spread to the brain and frequently to the eyes. The kinetics of virus spread to and clearance from the brain appeared to depend on the inoculation dose. After intraperitoneal inoculation, virus replication was predominantly observed in the liver and occasionally in the intestines, but interestingly also in the tail and paws. BLI thus elucidated new anatomic locations of virus replication. Furthermore, MHV dissemination was shown to be critically depended on the viral spike protein, but also on the mouse strain used. Widespread dissemination was observed in mice lacking a functional type I interferon response. The importance of the type I interferon system in limiting viral spread was also demonstrated by the administration of type I interferons to mice. Our results provide new insights in coronavirus pathogenesis and demonstrate the potential of BLI to study coronavirus-host interactions in vivo.


Asunto(s)
Infecciones por Coronavirus/virología , Virus de la Hepatitis Murina/fisiología , Replicación Viral , Animales , Infecciones por Coronavirus/genética , Infecciones por Coronavirus/patología , Diagnóstico por Imagen/métodos , Heparitina Sulfato/farmacología , Interferón Tipo I/farmacología , Proteínas Luminiscentes/análisis , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Virus de la Hepatitis Murina/efectos de los fármacos , Virus de la Hepatitis Murina/patogenicidad , Proteínas Recombinantes
20.
JBMR Plus ; 4(1): e10247, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31956851

RESUMEN

Multiple myeloma is characterized by accumulation of malignant plasma cells in the bone marrow. Most patients suffer from an osteolytic bone disease, caused by increased bone degradation and reduced bone formation. Bone morphogenetic protein 4 (BMP4) is important for both pre- and postnatal bone formation and induces growth arrest and apoptosis of myeloma cells. BMP4-treatment of myeloma patients could have the potential to reduce tumor growth and restore bone formation. We therefore explored BMP4 gene therapy in a human-mouse model of multiple myeloma where humanized bone scaffolds were implanted subcutaneously in RAG2-/- γC-/-mice. Mice were treated with adeno-associated virus serotype 8 BMP4 vectors (AAV8-BMP4) to express BMP4 in the liver. When mature BMP4 was detectable in the circulation, myeloma cells were injected into the scaffolds and tumor growth was examined by weekly imaging. Strikingly, the tumor burden was reduced in AAV8-BMP4 mice compared with the AAV8-CTRL mice, suggesting that increased circulating BMP4 reduced tumor growth. BMP4-treatment also prevented bone loss in the scaffolds, most likely due to reduced tumor load. To delineate the effects of BMP4 overexpression on bone per se, without direct influence from cancer cells, we examined the unaffected, non-myeloma femurs by µCT. Surprisingly, the AAV8-BMP4 mice had significantly reduced trabecular bone volume, trabecular numbers, as well as significantly increased trabecular separation compared with the AAV8-CTRL mice. There was no difference in cortical bone parameters between the two groups. Taken together, BMP4 gene therapy inhibited myeloma tumor growth, but also reduced the amount of trabecular bone in mice. Our data suggest that care should be taken when considering using BMP4 as a therapeutic agent. © 2019 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA