Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.962
Filtrar
1.
Sci Immunol ; 9(94): eadg1094, 2024 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-38640253

RESUMO

Chronic antigen stimulation is thought to generate dysfunctional CD8 T cells. Here, we identify a CD8 T cell subset in the bone marrow tumor microenvironment that, despite an apparent terminally exhausted phenotype (TPHEX), expressed granzymes, perforin, and IFN-γ. Concurrent gene expression and DNA accessibility revealed that genes encoding these functional proteins correlated with BATF expression and motif accessibility. IFN-γ+ TPHEX effectively killed myeloma with comparable efficacy to transitory effectors, and disease progression correlated with numerical deficits in IFN-γ+ TPHEX. We also observed IFN-γ+ TPHEX within CD19-targeted chimeric antigen receptor T cells, which killed CD19+ leukemia cells. An IFN-γ+ TPHEX gene signature was recapitulated in TEX cells from human cancers, including myeloma and lymphoma. Here, we characterize a TEX subset in hematological malignancies that paradoxically retains function and is distinct from dysfunctional TEX found in chronic viral infections. Thus, IFN-γ+ TPHEX represent a potential target for immunotherapy of blood cancers.


Assuntos
Neoplasias Hematológicas , Mieloma Múltiplo , Humanos , Receptor Celular 2 do Vírus da Hepatite A , Mieloma Múltiplo/metabolismo , Linfócitos T CD8-Positivos , Fenótipo , Microambiente Tumoral
2.
Ann Clin Lab Sci ; 54(1): 56-65, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38514068

RESUMO

OBJECTIVE: Multiple Myeloma (MM) is a malignant hematological disease. Heterogeneous nuclear ribonucleoprotein C1/C2 (HNRNPC) acts as an oncogene in a variety of cancers. However, the role of HNRNPC in MM has not been reported so far. METHODS: The mRNA and protein expressions of HNRN-PC and FOXM1 were detected by qRT-PCR and western blot. CCK8, EDU staining, flow cytometry and western blot were used to detect cell viability and cell cycle. The extracellular flux analyzer XF96 was used to detect the production of oxygen consumption rate (OCR) and extracellular acidification rate (ECAR). Lactic acid and glucose levels in culture medium were detected by lactic acid assay kits and glucose assay kits, respectively. Then, the binding ability of HNRNPC with FOXM1 was detected by RIP and the stability of FOXM1 mRNA was appraised with qRT-PCR. With the application of qRT-PCR and western blot, the transfection efficacy of si-HNRNPC and Oe-FOXM1 was examined. Western blot was applied for the estimation of GLUT1/LDHA signaling pathway-related proteins. RESULTS: The expression of HNRNPC in MM cell line was abnormally elevated. HNRNPC silence significantly inhibited the proliferation, facilitated the apoptosis, induced cycle arrest, and suppressed aerobic glycolysis in MM cells, which were all reversed by FOXM1 overexpression. It was also found that the regulatory effect of HNRNPC is realized by stabilizing FOXM1 mRNA and regulating GLUT1/LDHA pathway. CONCLUSION: HNRNPC regulated GLUT1/LDHA pathway by stabilizing FOXM1 mRNA to promote the progression and aerobic glycolysis of MM.


Assuntos
Proteína Forkhead Box M1 , Ribonucleoproteínas Nucleares Heterogêneas Grupo C , Mieloma Múltiplo , Humanos , Linhagem Celular Tumoral , Proliferação de Células/genética , Proteína Forkhead Box M1/genética , Proteína Forkhead Box M1/metabolismo , Glucose/metabolismo , Transportador de Glucose Tipo 1/metabolismo , Glicólise/genética , Ribonucleoproteínas Nucleares Heterogêneas Grupo C/metabolismo , Ácido Láctico , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , L-Lactato Desidrogenase/metabolismo
3.
Leuk Res ; 139: 107469, 2024 04.
Artigo em Inglês | MEDLINE | ID: mdl-38479337

RESUMO

BACKGROUND: The proteasome inhibitor bortezomib is one of the primary therapies used for the haematological malignancy multiple myeloma (MM). However, intrinsic or acquired resistance to bortezomib, via mechanisms that are not fully elucidated, is a barrier to successful treatment in many patients. Our previous studies have shown that elevated expression of the chemokine receptor CCR1 in MM plasma cells in newly diagnosed MM patients is associated with poor prognosis. Here, we hypothesised that the poor prognosis conferred by CCR1 expression is, in part, due to a CCR1-mediated decrease in MM plasma cell sensitivity to bortezomib. METHODS: In order to investigate the role of CCR1 in MM cells, CCR1 was knocked out in human myeloma cell lines OPM2 and U266 using CRISPR-Cas9. Additionally, CCR1 was overexpressed in the mouse MM cell line 5TGM1. The effect of bortezomib on CCR1 knockout or CCR1-overexpressing cells was then assessed by WST-1 assay, with or without CCL3 siRNA knockdown or addition of recombinant human CCL3. NSG mice were inoculated intratibially with OPM2-CCR1KO cells and were treated with 0.7 mg/kg bortezomib or vehicle twice per week for 3 weeks and GFP+ tumour cells in the bone marrow were quantitated by flow cytometry. The effect of CCR1 overexpression or knockout on unfolded protein response pathways was assessed using qPCR for ATF4, HSPA5, XBP1, ERN1 and CHOP and Western blot for IRE1α and p-Jnk. RESULTS: Using CCR1 overexpression or CRIPSR-Cas9-mediated CCR1 knockout in MM cell lines, we found that CCR1 expression significantly decreases sensitivity to bortezomib in vitro, independent of the CCR1 ligand CCL3. In addition, CCR1 knockout rendered the human MM cell line OPM2 more sensitive to bortezomib in an intratibial MM model in NSG mice in vivo. Moreover, CCR1 expression negatively regulated the expression of the unfolded protein response receptor IRE1 and downstream target gene XBP1, suggesting this pathway may be responsible for the decreased bortezomib sensitivity of CCR1-expressing cells. CONCLUSIONS: Taken together, these studies suggest that CCR1 expression may be associated with decreased response to bortezomib in MM cell lines.


Assuntos
Mieloma Múltiplo , Humanos , Animais , Camundongos , Bortezomib/farmacologia , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Linhagem Celular Tumoral , Receptores de Quimiocinas , Endorribonucleases , Proteínas Serina-Treonina Quinases , Receptores CCR1/genética , Receptores CCR1/metabolismo
4.
Nat Commun ; 15(1): 2513, 2024 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-38514625

RESUMO

In multiple myeloma, abnormal plasma cells establish oncogenic niches within the bone marrow by engaging the NF-κB pathway to nurture their survival while they accumulate pro-proliferative mutations. Under these conditions, many cases eventually develop genetic abnormalities endowing them with constitutive NF-κB activation. Here, we find that sustained NF-κB/p52 levels resulting from such mutations favours the recruitment of enhancers beyond the normal B-cell repertoire. Furthermore, through targeted disruption of p52, we characterise how such enhancers are complicit in the formation of super-enhancers and the establishment of cis-regulatory interactions with myeloma dependencies during constitutive activation of p52. Finally, we functionally validate the pathological impact of these cis-regulatory modules on cell and tumour phenotypes using in vitro and in vivo models, confirming RGS1 as a p52-dependent myeloma driver. We conclude that the divergent epigenomic reprogramming enforced by aberrant non-canonical NF-κB signalling potentiates transcriptional programs beneficial for multiple myeloma progression.


Assuntos
Mieloma Múltiplo , NF-kappa B , Humanos , NF-kappa B/genética , NF-kappa B/metabolismo , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Transcriptoma , Epigenoma , Transdução de Sinais/genética , Subunidade p52 de NF-kappa B/metabolismo
5.
Aging (Albany NY) ; 16(5): 4169-4190, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38431306

RESUMO

BACKGROUND: Multiple myeloma (MM) is an incurable malignant plasma cell disease. We explored the role of RAB22A in exosome secretion, epithelial-mesenchymal transition (EMT) and immune regulation. METHODS: We obtained MM samples from Gene Expression Omnibus (GEO) data sets. We downloaded the "IOBR" package, and used the "PCA" and "ssGSEA" algorithms to calculate the EMT scores and exosome scores. The "CIBERSORT" package was used to analyze the infiltration of immune cells. We extracted the exosomes of mesenchymal stem cell (MSC) to verify the biological function of RAB22A. RESULTS: The expression level of RAB22A in smoldering multiple myeloma (SMM) and MM patients was significantly higher than that in normal people and monoclonal gammopathy of undetermined significance (MGUS) patients, and the expression level of RAB22A in relapse MM patients was significantly higher than that in newly diagnosed patients. The EMT scores and exosome scores of high RAB22A group were significantly higher than those of low RAB22A group, and the exosome scores of MSC in recurrent patients were significantly higher than those of newly diagnosed patients. In addition, the infiltration levels of monocyte, NK cells resting, eosinophils, T cells regulatory and T cells CD4 memory activated were positively correlated with RAB22A. After down-regulating the expression of RAB22A in MM-MSC, the secretion of exosomes decreased. Compared with the exosomes of MSC in si-RAB22A group, the exosomes in control group significantly promoted the proliferation of MM. CONCLUSIONS: RAB22A is a potential therapeutic target to improve the prognosis of MM, which is closely related to exosome secretion, EMT and immune cell infiltration.


Assuntos
Exossomos , Gamopatia Monoclonal de Significância Indeterminada , Mieloma Múltiplo , Humanos , Mieloma Múltiplo/metabolismo , Exossomos/metabolismo , Prognóstico , Gamopatia Monoclonal de Significância Indeterminada/diagnóstico , Gamopatia Monoclonal de Significância Indeterminada/genética , Gamopatia Monoclonal de Significância Indeterminada/metabolismo , Doença Crônica , Recidiva , Progressão da Doença , Proteínas rab de Ligação ao GTP/genética
6.
Cancer Lett ; 588: 216797, 2024 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-38462032

RESUMO

Induction of pyroptosis is proposed as a promising strategy for the treatment of hematological malignancies, but little is known. In the present study, we find clioquinol (CLQ), an anti-parasitic drug, induces striking myeloma and leukemia cell pyroptosis on a drug screen. RNA sequencing reveals that the interferon-inducible genes IFIT1 and IFIT3 are markedly upregulated and are essential for CLQ-induced GSDME activation and cell pyroptosis. Specifically, IFIT1 and IFIT3 form a complex with BAX and N-GSDME therefore directing N-GSDME translocalization to mitochondria and increasing mitochondrial membrane permeabilization and triggering pyroptosis. Furthermore, venetoclax, an activator of BAX and an inhibitor of Bcl-2, displays strikingly synergistic effects with CLQ against leukemia and myeloma via pyroptosis. This study thus reveals a novel mechanism for mitochondrial GSDME in pyroptosis and it also illustrates that induction of IFIT1/T3 and inhibition of Bcl-2 orchestrate the treatment of leukemia and myeloma via pyroptosis.


Assuntos
Leucemia , Mieloma Múltiplo , Humanos , Piroptose , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Proteína X Associada a bcl-2/metabolismo , Mitocôndrias/metabolismo , Proteínas de Ligação a RNA/metabolismo , Leucemia/metabolismo , Caspase 3/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo
7.
Cancer Lett ; 588: 216802, 2024 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-38467180

RESUMO

Multiple myeloma (MM) is a hematological malignancy that remains incurable, primarily due to the high likelihood of relapse or development of resistance to current treatments. To explore and discover new medications capable of overcoming drug resistance in MM, we conducted cell viability inhibition screens of 1504 FDA-approved drugs. Lomitapide, a cholesterol-lowering agent, was found to exhibit effective inhibition on bortezomib-resistant MM cells in vitro and in vivo. Our data also indicated that lomitapide decreases the permeability of the mitochondrial outer membrane and induces mitochondrial dysfunction in MM cells. Next, lomitapide treatment upregulated DRP1 and PINK1 expression levels, coupled with the mitochondrial translocation of Parkin, leading to MM cell mitophagy. Excessive mitophagy caused mitochondrial damage and dysfunction induced by lomitapide. Meanwhile, PARP14 was identified as a direct target of lomitapide by SPR-HPLC-MS, and we showed that DRP1-induced mitophagy was crucial in the anti-MM activity mediated by PARP14. Furthermore, PARP14 is overexpressed in MM patients, implying that it is a novel therapeutic target in MM. Collectively, our results demonstrate that DRP1-mediated mitophagy induced by PARP14 may be the cause for mitochondrial dysfunction and damage in response to lomitapide treatment.


Assuntos
Benzimidazóis , Doenças Mitocondriais , Mieloma Múltiplo , Humanos , Mitofagia , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Mitocôndrias/metabolismo , Recidiva Local de Neoplasia/patologia , Resistência a Medicamentos , Doenças Mitocondriais/metabolismo , Doenças Mitocondriais/patologia , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo
8.
J Investig Med ; 72(4): 333-340, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38373952

RESUMO

Multiple myeloma (MM) is a bone marrow malignancy characterized by plasma cell proliferation. It was aimed to investigate pentraxin 3 (PTX3) levels, oxidative/antioxidative status, and their correlation in MM. In the study, four groups were established, including newly diagnosed MM (NDMM), MM in remission (Rem-MM), relapsed/refractory MM (RRMM) patients, and a healthy control group. PTX3 levels were measured using enzyme-linked immunosorbent assay, and the total antioxidant status (TAS) and total oxidant status (TOS) were assessed with an autoanalyzer. The oxidative stress index (OSI) was calculated using the formula: OSI (arbitrary unit) = TOS (µmol H2O2 Eq/L)/TAS (mmol Trolox Eq/L) × 100. The study involved comparing PTX3, TAS, TOS, and OSI levels among these four groups. PTX3 levels were significantly elevated in NDMM and RRMM groups compared to controls and the Rem-MM group (NDMM vs control; p < 0.001, NDMM vs Rem-MM; p < 0.001, RRMM vs control; p < 0.001, and RRMM vs Rem-MM; p = 0.006). TAS was higher in NDMM and RRMM groups versus controls (p = 0.009 and p < 0.001, respectively), and TOS was higher in rem-MM group versus NDMM and control groups (p < 0.001 and p = 0.016, respectively). OSI was higher in the Rem-MM group than in NDMM and RRMM groups (p < 0.001 and p = 0.009, respectively). Multivariate analysis confirmed associations between MM groups and PTX3 levels. Receiver operating characteristic analysis revealed high specificity (90%) and sensitivity (79%) for PTX3 in NDMM at a >0.56 ng/mL cut-off value. This study suggests that PTX3 levels may have diagnostic and prognostic potential in MM and its relationship with oxidative stress requires further exploration.


Assuntos
Proteína C-Reativa , Mieloma Múltiplo , Estresse Oxidativo , Componente Amiloide P Sérico , Humanos , Antioxidantes/metabolismo , Proteína C-Reativa/química , Proteína C-Reativa/metabolismo , Peróxido de Hidrogênio , Mieloma Múltiplo/diagnóstico , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Oxidantes , Prognóstico
9.
Leuk Res ; 138: 107455, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38368721

RESUMO

OBJECTIVE: To explore the involvement of TFEB-mediated autophagy-lysosomal mechanisms in multiple myeloma (MM) during bortezomib treatment. METHODS: MM cells were exposed to bortezomib or subjected to TFEB knockdown. CCK assay was used to assess the cell proliferation. Western blotting and fluorescent staining were conducted to examine autophagy and lysosomes. The TFEB expression pattern was analyzed, and whole transcriptome sequencing was carried out. Additionally, TFEB target genes were predicted using the GTRD(http://gtrd.biouml.org/) website, and pathway analysis was performed. RESULTS: Bortezomib demonstrated a dose-dependent and time dependent inhibition of cell proliferation. In MM cells treated with bortezomib, LC3B, Beclin-1, TFEB, and Lamp1 exhibited upregulation in a time- and concentration-dependent manner. LysoTracker dye labeling showed an increase in lysosomes in the bortezomib-treated group. Moreover, bortezomib elevated the expression of lysosome-associated factor Lamp1. Bortezomib promoted the nuclear translocation of TFEB, leading to decreased cytoplasmic TFEB and increased nuclear TFEB. TFEB gene silencing reversed bortezomib's inhibitory effect on MM cell lines, significantly reducing autophagosome expression and lysosome numbers. Furthermore, bioinformatic analysis identified the MAPK pathway as a potential downstream target of TFEB. CONCLUSION: Bortezomib effectively inhibits MM cell proliferation and induces autophagy, partly through TFEB-mediated mechanisms, with potential involvement of the MAPK pathway.


Assuntos
Mieloma Múltiplo , Humanos , Bortezomib/farmacologia , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Autofagia , Autofagossomos/metabolismo , Lisossomos/metabolismo , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética
10.
Neoplasia ; 50: 100974, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38364355

RESUMO

Multiple myeloma is a hematological malignancy that has evolved from antibody-secreting B lymphocytes. Like other types of cancers, myeloma cells have acquired functional capabilities which are referred to as "Hallmarks of Cancer", and one of their most important features is the metabolic disorders. Due to the high secretory load of the MM cells, the first-line medicine proteasome inhibitors have found their pronounced effects in MM cells for blocking the degradation of misfolded proteins, leading to their accumulation in the ER and overwhelming ER stress. Moreover, proteasome inhibitors have been reported to be effective in myeloma by targeting glucose, lipid, amino acid metabolism of MM cells. In this review, we have described the abnormal metabolism of the three major nutrients, such as glucose, lipid and amino acids, which participate in the cellular functions. We have described their roles in myeloma progression, how they could be exploited for therapeutic purposes, and current therapeutic strategies targeting these metabolites, hoping to uncover potential novel therapeutic targets and promote the development of future therapeutic approaches.


Assuntos
Mieloma Múltiplo , Inibidores de Proteassoma , Humanos , Inibidores de Proteassoma/farmacologia , Inibidores de Proteassoma/uso terapêutico , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/metabolismo , Glucose , Lipídeos/uso terapêutico , Complexo de Endopeptidases do Proteassoma/metabolismo
11.
Cytokine Growth Factor Rev ; 76: 1-11, 2024 04.
Artigo em Inglês | MEDLINE | ID: mdl-38378397

RESUMO

Immune effector cells in patients with multiple myeloma (MM) are at the forefront of many immunotherapy treatments, and several methods have been developed to fully utilise the antitumour potential of immune cells. T and NK cell-derived immune lymphocytes both expressed activating NK receptor group 2 member D(NKG2D). This receptor can identify eight distinct NKG2D ligands (NKG2DL), including major histocompatibility complex class I (MHC) chain-related protein A and B (MICA and MICB). Their binding to NKG2D triggers effector roles in T and NK cells. NKG2DL is polymorphic in MM cells. The decreased expression of NKG2DL on the cell surface is explained by multiple mechanisms of tumour immune escape. In this review, we discuss the mechanisms by which the NKG2D/NKG2DL axis regulates immune effector cells and strategies for promoting NKG2DL expression and inhibiting its release in multiple myeloma and propose therapeutic strategies that increase the expression of NKG2DL in MM cells while enhancing the activation and killing function of NK cells.


Assuntos
Mieloma Múltiplo , Humanos , Mieloma Múltiplo/terapia , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK , Células Matadoras Naturais , Antígenos de Histocompatibilidade Classe I/metabolismo , Imunoterapia
12.
Hematol Oncol Clin North Am ; 38(2): 533-546, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38233233

RESUMO

Immunocompetent mouse models of multiple myeloma (MM) are particularly needed in the era of T cell redirected therapy to understand drivers of sensitivity and resistance, optimize responses, and prevent toxicities. Three mouse models have been extensively characterized: the Balb/c plasmacytomas, the 5TMM, and the Vk*MYC. In the last year, additional models have been generated, which, for the first time, capture primary MM initiating events, like MMSET/NSD2 or cyclin D1 dysregulation. However, the long latency needed for tumor development and the lack of transplantable lines limit their utilization. Future studies should focus on modeling hyperdiploid MM.


Assuntos
Mieloma Múltiplo , Camundongos , Animais , Humanos , Mieloma Múltiplo/genética , Mieloma Múltiplo/terapia , Mieloma Múltiplo/metabolismo , Modelos Animais de Doenças
13.
J Clin Invest ; 134(1)2024 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-38165043

RESUMO

The concept of induced protein degradation by small molecules has emerged as a promising therapeutic strategy that is particularly effective in targeting proteins previously considered "undruggable." Thalidomide analogs, employed in the treatment of multiple myeloma, stand as prime examples. These compounds serve as molecular glues, redirecting the CRBN E3 ubiquitin ligase to degrade myeloma-dependency factors, IKZF1 and IKZF3. The clinical success of thalidomide analogs demonstrates the therapeutic potential of induced protein degradation. Beyond molecular glue degraders, several additional modalities to trigger protein degradation have been developed and are currently under clinical evaluation. These include heterobifunctional degraders, polymerization-induced degradation, ligand-dependent degradation of nuclear hormone receptors, disruption of protein interactions, and various other strategies. In this Review, we will provide a concise overview of various degradation modalities, their clinical applications, and potential future directions in the field of protein degradation.


Assuntos
Mieloma Múltiplo , Talidomida , Humanos , Proteólise , Lenalidomida/uso terapêutico , Ubiquitinação , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Mieloma Múltiplo/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
14.
Mol Carcinog ; 63(4): 728-741, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38258917

RESUMO

Multiple myeloma (MM) remains an incurable disease. Identification of meaningful co-expressed gene clusters or representative biomarkers of MM may help to identify new pathological mechanisms and promote the development of new therapies. Here, we performed weighted sgene co-expression network analysis and a series of bioinformatics analysis to identify single stranded DNA binding protein 1 (SSBP1) as novel hub gene associated with MM development and prognosis. In vitro, CRISPR/cas9 mediated knockdown of SSBP1 can significantly inhibit the proliferation of MM cells through inducing apoptosis and cell cycle arrest in G0/G1 phase. We also found that decreased SSBP1 expression significantly increased mitochondrial reactive oxygen species (mtROS) generation and the level of phosphorylated p38MAPK. Furthermore, it was further verified that disruption of SSBP1 expression could inhibit the tumor growth via p38MAPK pathway in a human myeloma xenograft model. In summary, our study is the first to demonstrate that SSBP1 promotes MM development by regulating the p38MAPK pathway.


Assuntos
Mieloma Múltiplo , Humanos , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Prognóstico , Proteínas de Ligação a DNA/genética , Transdução de Sinais , Apoptose , Progressão da Doença , Proliferação de Células , Linhagem Celular Tumoral , Proteínas Mitocondriais/metabolismo
15.
Sci Rep ; 14(1): 1756, 2024 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-38243063

RESUMO

Dissemination of multiple myeloma into the bone marrow proceeds through sequential steps mediated by a variety of adhesion molecules and chemokines that eventually results in the extravasation of malignant plasma cells into this protective niche. Selectins are a class of C-type lectins that recognize carbohydrate structures exposed on blood borne cells and participate in the first step of the extravasation cascade, serving as brakes to slow down circulating cells enabling them to establish firm adhesion onto the endothelium. Myeloma cells enriched for the expression of selectin ligands present an aggressive disease in vivo that is refractory to bortezomib treatment and can be reverted by small molecules targeting E-selectin. In this study, we have defined the molecular determinants of the selectin ligands expressed on myeloma cells. We show that PSGL-1 is the main protein carrier of sialyl Lewisa/x-related structures in myeloma. PSGL-1 decorated with sialyl Lewisa/x is essential for P-selectin binding but dispensable for E-selectin binding. Moreover, sialylation is required for E-selectin engagement whereas high affinity binding to P-selectin occurs even in the absence of sialic acid. This study provides further knowledge on the biology of selectin ligands in myeloma, opening the way to their clinical application as diagnostic tools and therapeutic targets.


Assuntos
Selectina E , Glicoproteínas de Membrana , Mieloma Múltiplo , Selectina-P , Antígeno Sialil Lewis X , Humanos , Adesão Celular , Selectina E/metabolismo , Ligantes , Mieloma Múltiplo/metabolismo , Selectina-P/metabolismo , Glicoproteínas de Membrana/metabolismo , Linhagem Celular Tumoral
16.
Int J Hematol ; 119(3): 303-315, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38245883

RESUMO

Resistance to proteasome inhibitors (PIs) has emerged as an important clinical issue. We investigated the mechanisms underlying multiple myeloma (MM) cell resistance to PIs. To mimic their pharmacokinetic/pharmacodynamic (PK/PD) profiles, MM cells were treated with bortezomib and carfilzomib for 1 h at concentrations up to 400 and 1,000 nM, respectively. Susceptibility to these PIs markedly varied among MM cell lines. Pulsatile treatments with PIs suppressed translation, as demonstrated by incorporation of puromycin at 24 h in PI-susceptible MM.1S cells, but not PI-resistant KMS-11 cells. Inhibition of ß5 subunit activity decreased at 24 h in KMS-11 cells, even with the irreversible PI carfilzomib, but not under suppression of protein synthesis with cycloheximide. Furthermore, the proteasome-degradable pro-survival factors PIM2 and NRF2 acutely accumulated in MM cells subjected to pulsatile PI treatments. Accumulated NRF2 was trans-localized into the nucleus to induce the expression of its target gene, HMOX1, in MM cells. PIM and Akt inhibition restored the anti-MM effects of PIs, even against PI-resistant KMS-11 cells. Collectively, these results suggest that increased synthesis of ß5 proteasome subunit and acute accumulation of PIM2 and NRF2 reduce the anti-MM effects of PIs.


Assuntos
Antineoplásicos , Mieloma Múltiplo , Humanos , Inibidores de Proteassoma/farmacologia , Fator 2 Relacionado a NF-E2/farmacologia , Fator 2 Relacionado a NF-E2/uso terapêutico , Mieloma Múltiplo/metabolismo , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Complexo de Endopeptidases do Proteassoma/farmacologia , Resistencia a Medicamentos Antineoplásicos , Linhagem Celular Tumoral , Bortezomib/farmacologia , Bortezomib/uso terapêutico , Antineoplásicos/uso terapêutico , Proteínas Proto-Oncogênicas , Proteínas Serina-Treonina Quinases
17.
Anal Methods ; 16(7): 1050-1057, 2024 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-38269419

RESUMO

Selinexor is a nuclear exportin-1 (XPO1) inhibitor that has been approved for the treatment of multiple myeloma patients. However, sustained use of selinexor may result in some undesirable consequences. Furthermore, selinexor has moderate inter-patient variability. Herein, we developed an ultrahigh-performance liquid chromatography tandem mass spectrometry method for measuring selinexor levels in human plasma ranging from 1 to 1000 ng mL-1. Furthermore, the developed approach was validated in accordance with FDA criteria. The established approach demonstrated inter-day and intra-day precision, expressed as the relative standard deviation, of less than 8%, with accuracies of less than 6%, expressed as relative error. The results showed that the protein depletion was quite complete for selinexor extraction, with recoveries ranging from 85.89 to 108.38%. The validated method facilitates the quantitation of selinexor in multiple myeloma patients. The selinexor plasma concentration exhibits obvious inter-patient' variability after administration. Thus, it is necessary to make a personalized prescription through therapeutic drug monitoring. Furthermore, the change in platelet counts before and after selinexor treatment was shown to be related to the plasma concentration at 3 h after administration, which provides the basis for therapeutic drug monitoring sampling time points and a method for predicting the occurrence of thrombocytopenia. In conclusion, the developed method can be used for the quantification of the plasma concentration of selinexor, and it is of great significance to conduct therapeutic drug monitoring for patients taking selinexor in order to enhance therapeutic effects and prevent the occurrence of adverse reactions.


Assuntos
Hidrazinas , Mieloma Múltiplo , Triazóis , Humanos , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/metabolismo , Espectrometria de Massas em Tandem/métodos , Cromatografia Líquida/métodos , China
19.
Cell Biol Int ; 48(2): 190-200, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37885161

RESUMO

Multiple myeloma (MM) is a hematologic malignancy associated with malignant plasma cell proliferation in the bone marrow. Despite the available treatments, drug resistance and adverse side effects pose significant challenges, underscoring the need for alternative therapeutic strategies. Natural products, like the fungal metabolite neosetophomone B (NSP-B), have emerged as potential therapeutic agents due to their bioactive properties. Our study investigated NSP-B's antitumor effects on MM cell lines (U266 and RPMI8226) and the involved molecular mechanisms. NSP-B demonstrated significant growth inhibition and apoptotic induction, triggered by reduced AKT activation and downregulation of the inhibitors of apoptotic proteins and S-phase kinase protein. This was accompanied by an upregulation of p21Kip1 and p27Cip1 and an elevated Bax/BCL2 ratio, culminating in caspase-dependent apoptosis. Interestingly, NSP-B also enhanced the cytotoxicity of bortezomib (BTZ), an existing MM treatment. Overall, our findings demonstrated that NSP-B induces caspase-dependent apoptosis, increases cell damage, and suppresses MM cell proliferation while improving the cytotoxic impact of BTZ. These findings suggest that NSP-B can be used alone or in combination with other medicines to treat MM, highlighting its importance as a promising phytoconstituent in cancer therapy.


Assuntos
Antineoplásicos , Mieloma Múltiplo , Humanos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Mieloma Múltiplo/metabolismo , Linhagem Celular Tumoral , Apoptose , Transdução de Sinais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Bortezomib/farmacologia , Proliferação de Células
20.
Tissue Cell ; 86: 102263, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37979396

RESUMO

The identification and investigation of key molecules involved in the pathogenesis of multiple myeloma (MM) hold paramount clinical significance. This study primarily focuses on elucidating the role of DEPDC1B within the context of MM. Our findings robustly affirm the abundant expression of DEPDC1B in MM tissues and cell lines. Notably, DEPDC1B depletion exerted inhibitory effects on MM cell proliferation and migration while concurrently facilitating apoptosis and G2 cell cycle arrest. These outcomes stand in stark contrast to the consequences of DEPDC1B overexpression. Furthermore, we identified CCNB1 as a putative downstream target, characterized by a co-expression pattern with DEPDC1B, mediating DEPDC1B's regulatory influence on MM. Additionally, our results suggest that DEPDC1B knockdown may activate the p53 pathway, thereby impeding MM progression. To corroborate these in vitro findings, we conducted in vivo experiments that further validate the regulatory role of DEPDC1B in MM and its interaction with CCNB1 and the p53 pathway. Collectively, our research underscores DEPDC1B as a potent promoter in the development of MM, representing a promising therapeutic target for MM treatment. This discovery bears significant implications for future investigations in this field.


Assuntos
Mieloma Múltiplo , Proteína Supressora de Tumor p53 , Humanos , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Mieloma Múltiplo/metabolismo , Apoptose/genética , Transdução de Sinais/genética , Proliferação de Células/genética , Linhagem Celular Tumoral , Ciclina B1/genética , Ciclina B1/metabolismo , Ciclina B1/farmacologia , Proteínas Ativadoras de GTPase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...