Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(2)2024 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-38255842

RESUMEN

NOTCH1 PEST domain mutations are often seen in hematopoietic malignancies, including T-cell acute lymphoblastic leukemia (T-ALL), chronic lymphocytic leukemia (CLL), splenic marginal zone lymphoma (SMZL), mantle cell lymphoma (MCL), and diffuse large B-cell lymphoma (DLBCL). These mutations play a key role in the development and progression of lymphoproliferative tumors by increasing the Notch signaling and, consequently, promoting cell proliferation, survival, migration, and suppressing apoptosis. There is currently no specific treatment available for cancers caused by NOTCH1 PEST domain mutations. However, several NOTCH1 inhibitors are in development. Among these, inhibition of the Sarco-endoplasmic Ca2+-ATPase (SERCA) showed a greater effect in NOTCH1-mutated tumors compared to the wild-type ones. One example is CAD204520, a benzimidazole derivative active in T-ALL cells harboring NOTCH1 mutations. In this study, we preclinically assessed the effect of CAD204520 in CLL and MCL models and showed that NOTCH1 PEST domain mutations sensitize cells to the anti-leukemic activity mediated by CAD204520. Additionally, we tested the potential of CAD204520 in combination with the current first-line treatment of CLL, venetoclax, and ibrutinib. CAD204520 enhanced the synergistic effect of this treatment regimen only in samples harboring the NOTCH1 PEST domain mutations, thus supporting a role for Notch inhibition in these tumors. In summary, our work provides strong support for the development of CAD204520 as a novel therapeutic approach also in chronic lymphoproliferative disorders carrying NOTCH1 PEST domain mutations, emerging as a promising molecule for combination treatment in this aggressive subset of patients.


Asunto(s)
Neoplasias Hematológicas , Leucemia Linfocítica Crónica de Células B , Trastornos Linfoproliferativos , Leucemia-Linfoma Linfoblástico de Células T Precursoras , Humanos , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Leucemia Linfocítica Crónica de Células B/genética , Trastornos Linfoproliferativos/tratamiento farmacológico , Trastornos Linfoproliferativos/genética , Mutación , Receptor Notch1/genética
5.
Haematologica ; 102(4): 773-784, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28057743

RESUMEN

Bone marrow monocytes are primarily committed to osteoclast formation. It is, however, unknown whether potential primary alterations are specifically present in bone marrow monocytes from patients with multiple myeloma, smoldering myeloma or monoclonal gammopathy of undetermined significance. We analyzed the immunophenotypic and transcriptional profiles of bone marrow CD14+ monocytes in a cohort of patients with different types of monoclonal gammopathies to identify alterations involved in myeloma-enhanced osteoclastogenesis. The number of bone marrow CD14+CD16+ cells was higher in patients with active myeloma than in those with smoldering myeloma or monoclonal gammopathy of undetermined significance. Interestingly, sorted bone marrow CD14+CD16+ cells from myeloma patients were more pro-osteoclastogenic than CD14+CD16-cells in cultures ex vivo Moreover, transcriptional analysis demonstrated that bone marrow CD14+ cells from patients with multiple myeloma (but neither monoclonal gammopathy of undetermined significance nor smoldering myeloma) significantly upregulated genes involved in osteoclast formation, including IL21RIL21R mRNA over-expression by bone marrow CD14+ cells was independent of the presence of interleukin-21. Consistently, interleukin-21 production by T cells as well as levels of interleukin-21 in the bone marrow were not significantly different among monoclonal gammopathies. Thereafter, we showed that IL21R over-expression in CD14+ cells increased osteoclast formation. Consistently, interleukin-21 receptor signaling inhibition by Janex 1 suppressed osteoclast differentiation from bone marrow CD14+ cells of myeloma patients. Our results indicate that bone marrow monocytes from multiple myeloma patients show distinct features compared to those from patients with indolent monoclonal gammopathies, supporting the role of IL21R over-expression by bone marrow CD14+ cells in enhanced osteoclast formation.


Asunto(s)
Expresión Génica , Monocitos/metabolismo , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Osteoclastos/metabolismo , Receptores de Interleucina-21/genética , Biomarcadores , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Análisis por Conglomerados , Citocinas/metabolismo , Femenino , Perfilación de la Expresión Génica , Humanos , Inmunofenotipificación , Receptores de Lipopolisacáridos/metabolismo , Masculino , Gammopatía Monoclonal de Relevancia Indeterminada/genética , Gammopatía Monoclonal de Relevancia Indeterminada/metabolismo , Gammopatía Monoclonal de Relevancia Indeterminada/patología , Mieloma Múltiple/metabolismo , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de IgG/metabolismo , Receptores de Interleucina-21/metabolismo
6.
Cytogenet Genome Res ; 147(1): 24-30, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26645620

RESUMEN

Variations of DNA sequences in the human genome range from large, microscopically visible chromosome anomalies to single nucleotide changes. Submicroscopic genomic copy number variations, i.e. chromosomal imbalances which are undetectable by conventional cytogenetic analysis, play an intriguing clinical role. In this study, we describe the clinical consequences of the concurrent presence of an interstitial deletion in 13q34 and a terminal deletion in 4q35.2 in an Italian family. The index patient, a 19-year-old male, as well as his 12-year-old sister are carriers of both deletions, one of maternal and the other of paternal origin. The phenotype includes language delay, multiorgan involvement and bleeding diathesis with mild deficiency of factors X and VII. In the sister, the concomitant presence of Noonan syndrome may partly explain the clinical symptoms. The deleted region on chromosome 13 involves several genes (ATP11A, MCF2L, F7, F10, PROZ, PCID2, CUL4A, and LAMP1); some of these seem to play a role in the proband's phenotype. The terminal deletion in 4q35.2 contains other OMIM genes (FRG1, FRG2 and DBET); moreover, the 4q region is reported as a susceptibility locus for Crohn's disease, diagnosed in the proband's father. To our knowledge, this is the first report of a family with these 2 submicroscopic copy number changes. We tried to relate the clinical phenotype of the proband and his family to the molecular function of the involved genes.


Asunto(s)
Deleción Cromosómica , Cromosomas Humanos Par 13 , Cromosomas Humanos Par 4 , Deficiencia del Factor VII/genética , Deficiencia del Factor X/genética , Trastornos Hemorrágicos/genética , Síndrome de Noonan/genética , Niño , Bandeo Cromosómico , Variaciones en el Número de Copia de ADN , Deficiencia del Factor VII/patología , Deficiencia del Factor X/patología , Femenino , Trastornos Hemorrágicos/patología , Humanos , Hibridación Fluorescente in Situ , Patrón de Herencia , Italia , Masculino , Síndrome de Noonan/patología , Linaje , Fenotipo , Adulto Joven
8.
Nat Commun ; 15(1): 4739, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38834613

RESUMEN

The overexpression of the ecotropic viral integration site-1 gene (EVI1/MECOM) marks the most lethal acute myeloid leukemia (AML) subgroup carrying chromosome 3q26 abnormalities. By taking advantage of the intersectionality of high-throughput cell-based and gene expression screens selective and pan-histone deacetylase inhibitors (HDACis) emerge as potent repressors of EVI1. To understand the mechanism driving on-target anti-leukemia activity of this compound class, here we dissect the expression dynamics of the bone marrow leukemia cells of patients treated with HDACi and reconstitute the EVI1 chromatin-associated co-transcriptional complex merging on the role of proliferation-associated 2G4 (PA2G4) protein. PA2G4 overexpression rescues AML cells from the inhibitory effects of HDACis, while genetic and small molecule inhibition of PA2G4 abrogates EVI1 in 3q26 AML cells, including in patient-derived leukemia xenografts. This study positions PA2G4 at the crosstalk of the EVI1 leukemogenic signal for developing new therapeutics and urges the use of HDACis-based combination therapies in patients with 3q26 AML.


Asunto(s)
Cromosomas Humanos Par 3 , Inhibidores de Histona Desacetilasas , Leucemia Mieloide Aguda , Proteína del Locus del Complejo MDS1 y EV11 , Proteogenómica , Animales , Femenino , Humanos , Ratones , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Cromosomas Humanos Par 3/genética , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Proteína del Locus del Complejo MDS1 y EV11/metabolismo , Proteína del Locus del Complejo MDS1 y EV11/genética , Proteogenómica/métodos , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Nat Commun ; 15(1): 1203, 2024 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-38331987

RESUMEN

DNA damage resistance is a major barrier to effective DNA-damaging therapy in multiple myeloma (MM). To discover mechanisms through which MM cells overcome DNA damage, we investigate how MM cells become resistant to antisense oligonucleotide (ASO) therapy targeting Interleukin enhancer binding factor 2 (ILF2), a DNA damage regulator that is overexpressed in 70% of MM patients whose disease has progressed after standard therapies have failed. Here, we show that MM cells undergo adaptive metabolic rewiring to restore energy balance and promote survival in response to DNA damage activation. Using a CRISPR/Cas9 screening strategy, we identify the mitochondrial DNA repair protein DNA2, whose loss of function suppresses MM cells' ability to overcome ILF2 ASO-induced DNA damage, as being essential to counteracting oxidative DNA damage. Our study reveals a mechanism of vulnerability of MM cells that have an increased demand for mitochondrial metabolism upon DNA damage activation.


Asunto(s)
Mieloma Múltiple , Humanos , Mieloma Múltiple/genética , ADN Helicasas/metabolismo , Reprogramación Metabólica , Reparación del ADN , Daño del ADN
11.
Oncoimmunology ; 11(1): 2120275, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36105747

RESUMEN

The humoral and cellular response to SARS-CoV-2 mRNA full vaccination and booster dose as well as the impact of the spike variants, including Omicron, are still unclear in patients with multiple myeloma (MM) and those with pre-malignant monoclonal gammopathies. In this study, involving 40 patients, we found that MM patients with relapsed-refractory disease (MMR) had reduced spike-specific antibody levels and neutralizing titers after SARS-CoV-2 vaccination. The five analyzed variants, remarkably Omicron, had a significant negative impact on the neutralizing ability of the vaccine-induced antibodies in all patients with MM and smoldering MM. Moreover, lower spike-specific IL-2-producing CD4+ T cells and reduced cytotoxic spike-specific IFN-γ and TNF-α-producing CD8+ T cells were found in MM patients as compared to patients with monoclonal gammopathy of undetermined significance. We found that a heterologous booster immunization improved SARS-CoV-2 spike humoral and cellular responses in newly diagnosed MM (MMD) patients and in most, but not all, MMR patients. After the booster dose, a significant increase of the neutralizing antibody titers against almost all the analyzed variants was achieved in MMD. However, in MMR patients, Omicron retained a negative impact on neutralizing ability, suggesting further approaches to potentiating the effectiveness of SARS-CoV-2 vaccination in these patients.


Asunto(s)
COVID-19 , Mieloma Múltiple , Vacunas Virales , Anticuerpos Antivirales , Linfocitos T CD8-positivos , COVID-19/prevención & control , Vacunas contra la COVID-19 , Humanos , Inmunidad , ARN Mensajero , SARS-CoV-2 , Vacunación , Vacunas Virales/genética
12.
Am J Hematol ; 86(12): E64-6, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21953534

RESUMEN

Homeobox (HOX) gene transcription factors are frequently deregulated in hematologic malignancies and involved in leukemogenic transformation [1]. Moreover, their overexpression has been associated with tumoral-induced neoangiogenesis in solid cancer [2]. The expression and the role of these genes have not yet been completely elucidated in multiple myeloma (MM). Recently, we reported that a small fraction of MM patients shows a HOXB7 overexpression as compared with normal samples and that HOXB7 expression correlates with bone marrow angiogenesis and the production of the proangiogenic factors by MM cells [3]. Other authors previously reported that HOXA cluster genes are expressed in a small fraction of MM patients [4]. Herein, we extended our previous evidences with the evaluation of the expression level of HOXB7 and the other gene family members in a large number of primary MM cells in relationship with the different molecular subgroups of MM and the presence of specific chromosome translocations. We found that HOXB7 and other genes of HOX family have a preferential distribution based on the characteristics of molecular MM subtypes based on the translocations/cyclins (TC) classification, suggesting a potential relationship between HOX genes expression, angiogenesis, and molecular features of MM patients.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Genes Homeobox , Proteínas de Homeodominio/genética , Mieloma Múltiple/genética , Mieloma Múltiple/metabolismo , Células Plasmáticas/metabolismo , Anciano , Médula Ósea/irrigación sanguínea , Estudios de Cohortes , Bases de Datos Factuales , Perfilación de la Expresión Génica , Genes Relacionados con las Neoplasias , Proteínas de Homeodominio/metabolismo , Humanos , Hibridación Fluorescente in Situ , Persona de Mediana Edad , Familia de Multigenes , Mieloma Múltiple/patología , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neovascularización Patológica , Células Plasmáticas/patología , Células Tumorales Cultivadas
13.
Cells ; 10(6)2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34205916

RESUMEN

Multiple myeloma (MM) is characterized by an accumulation of malignant plasma cells (PCs) in the bone marrow (BM). The amplification of 1q21 is one of the most common cytogenetic abnormalities occurring in around 40% of de novo patients and 70% of relapsed/refractory MM. Patients with this unfavorable cytogenetic abnormality are considered to be high risk with a poor response to standard therapies. The gene(s) driving amplification of the 1q21 amplicon has not been fully studied. A number of clear candidates are under investigation, and some of them (IL6R, ILF2, MCL-1, CKS1B and BCL9) have been recently proposed to be potential drivers of this region. However, much remains to be learned about the biology of the genes driving the disease progression in MM patients with 1q21 amp. Understanding the mechanisms of these genes is important for the development of effective targeted therapeutic approaches to treat these patients for whom effective therapies are currently lacking. In this paper, we review the current knowledge about the pathological features, the mechanism of 1q21 amplification, and the signal pathway of the most relevant candidate genes that have been suggested as possible therapeutic targets for the 1q21 amplicon.


Asunto(s)
Cromosomas Humanos Par 1 , Amplificación de Genes , Mieloma Múltiple , Proteínas de Neoplasias , Transducción de Señal , Cromosomas Humanos Par 1/genética , Cromosomas Humanos Par 1/metabolismo , Humanos , Mieloma Múltiple/genética , Mieloma Múltiple/metabolismo , Mieloma Múltiple/terapia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo
14.
Front Immunol ; 11: 613007, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33488620

RESUMEN

Background: The PD-1/PD-L1 axis has recently emerged as an immune checkpoint that controls antitumor immune responses also in hematological malignancies. However, the use of anti-PD-L1/PD-1 antibodies in multiple myeloma (MM) patients still remains debated, at least in part because of discordant literature data on PD-L1/PD-1 expression by MM cells and bone marrow (BM) microenvironment cells. The unmet need to identify patients which could benefit from this therapeutic approach prompts us to evaluate the BM expression profile of PD-L1/PD-1 axis across the different stages of the monoclonal gammopathies. Methods: The PD-L1/PD-1 axis was evaluated by flow cytometry in the BM samples of a total cohort of 141 patients with monoclonal gammopathies including 24 patients with Monoclonal Gammopathy of Undetermined Significance (MGUS), 38 patients with smoldering MM (SMM), and 79 patients with active MM, including either newly diagnosed or relapsed-refractory patients. Then, data were correlated with the main immunological and clinical features of the patients. Results: First, we did not find any significant difference between MM and SMM patients in terms of PD-L1/PD-1 expression, on both BM myeloid (CD14+) and lymphoid subsets. On the other hand, PD-L1 expression by CD138+ MM cells was higher in both SMM and MM as compared to MGUS patients. Second, the analysis on the total cohort of MM and SMM patients revealed that PD-L1 is expressed at higher level in CD14+CD16+ non-classical monocytes compared with classical CD14+CD16- cells, independently from the stage of disease. Moreover, PD-L1 expression on CD14+ cells was inversely correlated with BM serum levels of the anti-tumoral cytokine, IL-27. Interestingly, relapsed MM patients showed an inverted CD4+/CD8+ ratio along with high levels of pro-tumoral IL-6 and a positive correlation between %CD14+PD-L1+ and %CD8+PD-1+ cells as compared to both SMM and newly diagnosed MM patients suggesting a highly compromised immune-compartment with low amount of CD4+ effector cells. Conclusions: Our data indicate that SMM and active MM patients share a similar PD-L1/PD-1 BM immune profile, suggesting that SMM patients could be an interesting target for PD-L1/PD-1 inhibition therapy, in light of their less compromised and more responsive immune-compartment.


Asunto(s)
Antígeno B7-H1/inmunología , Médula Ósea/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Mieloma Múltiple Quiescente/inmunología , Microambiente Tumoral/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Linfocitos T CD8-positivos/inmunología , Femenino , Humanos , Interleucina-27/inmunología , Interleucina-6/inmunología , Receptores de Lipopolisacáridos/inmunología , Masculino , Persona de Mediana Edad , Gammopatía Monoclonal de Relevancia Indeterminada/inmunología , Monocitos/inmunología , Paraproteinemias/inmunología , Receptores de IgG/inmunología
15.
Cancers (Basel) ; 12(5)2020 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-32456143

RESUMEN

Genomic analysis could contribute to a better understanding of the biological determinants of the evolution of multiple myeloma (MM) precursor disease and an improved definition of high-risk patients. To assess the feasibility and value of next-generation sequencing approaches in an asymptomatic setting, we performed a targeted gene mutation analysis and a genome-wide assessment of copy number alterations (CNAs) by ultra-low-pass whole genome sequencing (ULP-WGS) in six patients with monoclonal gammopathy of undetermined significance and 25 patients with smoldering MM (SMM). Our comprehensive genomic characterization highlighted heterogeneous but substantial values of the tumor fraction, especially in SMM; a rather high degree of genomic complexity, in terms of both mutations and CNAs, and inter-patient variability; a higher incidence of gene mutations and CNAs in SMM, confirming ongoing evolution; intraclonal heterogeneity; and instances of convergent evolution. ULP-WGS of these patients proved effective in revealing the marked genome-wide level of their CNAs, most of which are not routinely investigated. Finally, the analysis of our small SMM cohort suggested that chr(8p) deletions, the DNA tumor fraction, and the number of alterations may have clinical relevance in the progression to overt MM. Although validation in larger series is mandatory, these findings highlight the promising impact of genomic approaches in the clinical management of SMM.

16.
J Mol Diagn ; 22(5): 629-639, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32142900

RESUMEN

T-cell acute lymphoblastic leukemia (T-ALL) results from deregulation of a number of genes via multiple genomic mechanisms. We designed a comprehensive fluorescence in situ hybridization (CI-FISH) assay that consists of genomic probes to simultaneously investigate oncogenes and oncosuppressors recurrently involved in chromosome rearrangements in T-ALL, which was applied to 338 T-ALL cases. CI-FISH provided genetic classification into one of the well-defined genetic subgroups (ie, TAL/LMO, HOXA, TLX3, TLX1, NKX2-1/2-2, or MEF2C) in 80% of cases. Two patients with translocations of the LMO3 transcription factor were identified, suggesting that LMO3 activation may serve as an alternative to LMO1/LMO2 activation in the pathogenesis of this disease. Moreover, intrachromosomal rearrangements that involved the 10q24 locus were found as a new mechanism of TLX1 activation. An unequal distribution of cooperating genetic defects was found among the six genetic subgroups. Interestingly, deletions that targeted TCF7 or TP53 were exclusively found in HOXA T-ALL, LEF1 defects were prevalent in NKX2-1 rearranged patients, CASP8AP2 and PTEN alterations were significantly enriched in TAL/LMO leukemias, and PTPN2 and NUP214-ABL1 abnormalities occurred in TLX1/TLX3. This work convincingly shows that CI-FISH is a powerful tool to define genetic heterogeneity of T-ALL, which may be applied as a rapid and accurate diagnostic test.


Asunto(s)
Biomarcadores de Tumor , Hibridación Fluorescente in Situ/métodos , Leucemia-Linfoma Linfoblástico de Células T Precursoras/diagnóstico , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Adolescente , Adulto , Anciano , Niño , Preescolar , Evolución Clonal/genética , Femenino , Reordenamiento Génico , Heterogeneidad Genética , Pruebas Genéticas , Estudio de Asociación del Genoma Completo , Humanos , Hibridación Fluorescente in Situ/normas , Masculino , Persona de Mediana Edad , Translocación Genética , Adulto Joven
17.
Bone Marrow Transplant ; 54(Suppl 2): 698-702, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31431717

RESUMEN

In patients with hematological malignancies at high risk for relapse, a mismatched hematopoietic stem cells transplants can be offered with no undue delay between decision-making and transplantation as virtually all patients have a full-haplotype mismatched member who could serve immediately as a donor. Using a T-cell depletion approach, these patients can benefit from a graft-vs-leukemia effect in the absence of both acute and chronic graft-vs-host disease. Over the past decade, efforts have concentrated on developing new conditioning regimens, optimizing the graft processing and improving the posttransplant immunological recovery. The innovative strategy based on the selective depletion of alpha/beta-positive T lymphocytes from G-CSF-mobilized peripheral blood precursor cells has shown very promising results in the setting of the pediatric transplantation. This paper reports the outcome in adult patients with hematological malignancies.


Asunto(s)
Antígenos CD19/metabolismo , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre de Sangre Periférica/inmunología , Acondicionamiento Pretrasplante/métodos , Trasplante Haploidéntico/métodos , Adulto , Anciano , Femenino , Factor Estimulante de Colonias de Granulocitos , Humanos , Masculino , Persona de Mediana Edad , Células Madre , Adulto Joven
19.
Tumori ; 93(1): 97-9, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17455879

RESUMEN

Most of the information about the genetic composition of parathyroid tumors has been obtained by comparative genomic hybridization (CGH) and loss of heterozygosity (LOH) studies, whereas only few conventional cytogenetic investigation results are available. We have performed cytogenetic analysis of short-term cultures from 3 parathyroid adenoma tissue samples. Two cases showed a normal karyotype in all the metaphases obtained from independent primary cultures. In one case 5 metaphases (in a total of 25) from 2 independent cultures showed a nonrandom translocation t(4;13)(q21;q14), which was therefore accepted as clonal. To our knowledge this is the second clonal translocation described in this tumor type. Further conventional cytogenetic analysis of more parathyroid tumor specimens would be necessary to identify other specific abnormalities and the involved genes with a potential important role in the diagnosis, prognosis and pathogenesis of parathyroid tumors.


Asunto(s)
Adenoma/genética , Aberraciones Cromosómicas , Cromosomas Humanos Par 13/genética , Cromosomas Humanos Par 4/genética , Neoplasias de las Paratiroides/genética , Translocación Genética , Análisis Citogenético , Humanos , Cariotipificación
20.
Int J Hematol ; 105(1): 104-108, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27699576

RESUMEN

The skin is a possible site of extramedullary localization in multiple myeloma (MM) patients; however, the mechanisms involved in this process are poorly understood. We describe the case of a refractory MM patient who developed a cutaneous localization under bortezomib treatment and we further expanded observations in other eight MM patients. We focused on the expression of genes involved in plasma cell skin homing, including CCR10, which was highly expressed. Moreover, we observed a lack of CXCR4 surface expression and the down-regulation of ICAM1/CD54 throughout the progression of the disease, suggesting a possible mechanism driving the escape of MM cells from the bone marrow into the skin.


Asunto(s)
Antineoplásicos/uso terapéutico , Médula Ósea/patología , Bortezomib/uso terapéutico , Mieloma Múltiple/patología , Células Plasmáticas/patología , Plasmacitoma/secundario , Neoplasias Cutáneas/secundario , Piel/patología , Médula Ósea/efectos de los fármacos , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Humanos , Molécula 1 de Adhesión Intercelular/genética , Masculino , Persona de Mediana Edad , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/genética , Células Plasmáticas/efectos de los fármacos , Plasmacitoma/genética , Plasmacitoma/patología , Receptores CXCR4/genética , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA