RESUMO
Among blood cancers, multiple myeloma (MM) represents the second most common neoplasm and is characterized by the accumulation and proliferation of monoclonal plasma cells within the bone marrow. Despite the last few decades being characterized by the development of different therapeutic strategies against MM, at present such disease is still considered incurable. Although MM is highly heterogeneous in terms of genetic and molecular subtypes, about 67% of MM cases are associated with abnormal activity of the transcription factor c-Myc, which has so far revealed a protein extremely difficult to target. We have recently demonstrated that activation of fibroblast growth factor (FGF) signaling protects MM cells from oxidative stress-induced apoptosis by stabilizing the oncoprotein c-Myc. Accordingly, secretion of FGF ligands and autocrine activation of FGF receptors (FGFR) is observed in MM cells and FGFR3 genomic alterations represent some 15-20% MM cases and are associated with poor outcome. Thus, FGF/FGFR blockade may represent a promising strategy to indirectly target c-Myc in MM. On this basis, the present review aims at providing an overview of recently explored connections between the FGF/FGFR system and c-Myc oncoprotein, sustaining the therapeutic potential of targeting the FGF/FGFR/c-Myc axis in MM by using inhibitors targeting FGF ligands or FGF receptors. Importantly, the provided findings may represent the rationale for using FDA approved FGFR TK inhibitors (i.e. Pemigatinib, Futibatinib, Erdafitinib) for the treatment of MM patients presenting with an aberrant activation of this axis.
Assuntos
Fatores de Crescimento de Fibroblastos , Mieloma Múltiplo , Proteínas Proto-Oncogênicas c-myc , Receptores de Fatores de Crescimento de Fibroblastos , Humanos , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Mieloma Múltiplo/genética , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Fatores de Crescimento de Fibroblastos/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Transdução de Sinais/efeitos dos fármacos , Terapia de Alvo Molecular , AnimaisRESUMO
BACKGROUND: The imbalance between T helper 17 (Th17) and T regulatory (Treg) cells plays a key role in the progression of multiple myeloma (MM). METHODS: The gene expression profiles of MM were acquired and examined from the Gene Expression Omnibus (GEO) database (GSE72213). Our research involved experimental investigations conducted using the MOPC-MM mouse model. Dysregulation of Treg and Th17 cells was evaluated through flow cytometry, while the levels of inflammatory factors were measured using the enzyme-linked immunosorbent assay. Cell proliferation was gauged using the Cell Counting Kit-8 assay, and cell apoptosis was quantified via flow cytometry. Cell metastasis capabilities were determined by conducting transwell assays. To confirm the relationship between miR-27a and PI3K, a dual-luciferase reporter assay was employed. Finally, proteins associated with the PI3K/AKT/mTOR signaling pathway were assessed using western blotting. RESULTS: MiR-27a exhibited reduced expression levels in MM. Moreover, it exerted control over the equilibrium of Th17 and Treg cells while reducing the expression of inflammatory mediators such as TGF-ß1 and IL-10 in an in vivo setting. Elevated miR-27a levels led to the inhibition of cell viability, colony formation capacity, migratory and invasive traits in an in vitro context. The PI3K/AKT/mTOR signaling pathway was identified as a direct target of miR-27a and could reverse the effects induced by miR-27a in MM cells. Notably, PI3K was directly targeted by miR-27a. CONCLUSIONS: Our study revealed that miR-27a inhibited MM evolution by regulating the Th17/Treg balance. Inhibition of the PI3K/AKT/mTOR signaling pathway by miR-27a may play a potential mechanistic role.
Assuntos
Proliferação de Células , MicroRNAs , Mieloma Múltiplo , Fosfatidilinositol 3-Quinases , Transdução de Sinais , Linfócitos T Reguladores , Células Th17 , MicroRNAs/genética , MicroRNAs/metabolismo , Mieloma Múltiplo/genética , Mieloma Múltiplo/patologia , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/metabolismo , Células Th17/imunologia , Células Th17/metabolismo , Animais , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Humanos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Metástase Neoplásica , ApoptoseRESUMO
OBJECTIVES: Patients with multiple myeloma often require multiple treatment lines. The order in which treatments are sequenced has impact on clinical outcomes. This study aimed to estimate progression-free survival (PFS) and overall survival (OS) with common treatment sequences used in Portugal and the incremental benefit of an optimal sequence in transplant-ineligible patients with multiple myeloma. METHODS: A state-transition sequential model with a five-health state conceptual structure was developed to simulate and compare survival outcomes between treatment sequences up to four lines of treatments. Data sources included randomized clinical trials and indirect treatment comparisons. A panel of Portuguese hematologists listed four most common treatment sequences and optimal sequence of choice in transplant-ineligible patients. RESULTS: Our simulation estimated an OS between 6.1 and 7.8 years using the most common sequences, with VMP + DRd + Pd + Kd as the most effective (7.8 years). Optimal sequence of choice (DRd + PVd + Kd + Vd) achieved OS of 9.8 years and may extend OS in 2.0-3.7 years vs. most common sequences (26%-61% increase). This benefit was mostly explained by extended PFS in the first line of treatment. CONCLUSION: Model results demonstrate that choosing the most effective treatment upfront is crucial in delaying disease progression thus yielding better survival outcomes in transplant-ineligible patients. There was a clear survival benefit in using daratumumab-based regimens in first line. This modelling exercise highlights the need to raise awareness around the impact of sequencing strategies to improve patient's outcomes.
Assuntos
Mieloma Múltiplo , Humanos , Mieloma Múltiplo/mortalidade , Mieloma Múltiplo/terapia , Mieloma Múltiplo/genética , Mieloma Múltiplo/tratamento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Intervalo Livre de Progressão , Simulação por Computador , Portugal/epidemiologia , Ensaios Clínicos Controlados Aleatórios como AssuntoRESUMO
Introduction: Multiple myeloma (MM) is an incurable hematological malignancy with high chromosome instability and heavy dependence on the immunosuppressive bone marrow microenvironment. P53 mutations are adverse prognostic factors in MM; however, clinically, some patients without P53 mutations also exhibit aggressive disease progression. DNp73, an inhibitor of TP53 tumor suppressor family members, drives drug resistance and cancer progression in several solid malignancies. Nevertheless, the biological functions of DNp73 and the molecular mechanisms in myelomagenesis remain unclear. Methods: The effects of DNp73 on proliferation and drug sensitivity were assessed using flow cytometry and xenograft models. To investigate the mechanisms of drug resistance, RNA-seq and ChIP-seq analyses were performed in MM cell lines, with validation by Western blot and RT-qPCR. Immunofluorescence and transwell assays were used to assess DNA damage and cell invasion in MM cells. Additionally, in vitro phagocytosis assays were conducted to confirm the role of DNp73 in immune evasion. Results: Our study found that activation of NF-κB-p65 in multiple myeloma cells with different p53 mutation statuses upregulates DNp73 expression at the transcriptional level. Forced expression of DNp73 promoted aggressive proliferation and multidrug resistance in MM cells. Bulk RNA-seq analysis was conducted to assess the levels of MYCN, MYC, and CDK7. A ChIP-qPCR assay was used to reveal that DNp73 acts as a transcription factor regulating MYCN gene expression. Bulk RNA-seq analysis demonstrated increased levels of MYCN, MYC, and CDK7 with forced DNp73 expression in MM cells. A ChIP-qPCR assay revealed that DNp73 upregulates MYCN gene expression as a transcription factor. Additionally, DNp73 promoted immune evasion of MM cells by upregulating MYC target genes CD47 and PD-L1. Blockade of the CD47/SIRPα and PD-1/PD-L1 signaling pathways by the SIRPα-Fc fusion protein IMM01 and monoclonal antibody atezolizumab significantly restored the anti-MM activity of macrophages and T cells in the microenvironment, respectively. Discussion: In summary, our study demonstrated for the first time that the p53 family member DNp73 remarkably induces proliferation, drug resistance, and immune escape of myeloma cells by directly targeting MYCN and regulating the MYC pathway. The oncogenic function of DNp73 is independent of p53 status in MM cells. These data contribute to a better understanding of the function of TP53 and its family members in tumorigenesis. Moreover, our study clarified that DNp73 overexpression not only promotes aggressive growth of tumor cells but, more importantly, promotes immune escape of MM cells through upregulation of immune checkpoints. DNp73 could serve as a biomarker for immunotherapy targeting PD-L1 and CD47 blockade in MM patients.
Assuntos
Mieloma Múltiplo , Proteína Proto-Oncogênica N-Myc , Proteínas Proto-Oncogênicas c-myc , Proteína Tumoral p73 , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/genética , Mieloma Múltiplo/patologia , Humanos , Proteína Proto-Oncogênica N-Myc/genética , Animais , Linhagem Celular Tumoral , Camundongos , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteína Tumoral p73/genética , Proteína Tumoral p73/imunologia , Proliferação de Células , Transdução de Sinais , Regulação Neoplásica da Expressão Gênica , Evasão Tumoral , Progressão da Doença , Resistencia a Medicamentos Antineoplásicos/genética , Microambiente Tumoral/imunologia , Microambiente Tumoral/genéticaRESUMO
BACKGROUND: Angiogenesis is associated with tumour growth, infiltration, and metastasis. This study aimed to detect the mechanisms of angiogenesis-related genes (ARGs) in multiple myeloma (MM) and to construct a new prognostic model. METHODS: MM research foundation (MMRF)-CoMMpass cohort, GSE47552, GSE57317, and ARGs were sourced from public databases. Differentially expressed genes (DEGs) in the tumour and control cohorts in GSE47552 were determined through differential expression analysis and were enriched with Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses. Weighted gene coexpression network analysis (WGCNA) was applied to derive modules linked to the ARG scores and obtain module genes in GSE47552. Differentially expressed ARGs (DE-ARGs) were selected for subsequent analyses by overlapping DEGs and module genes. Furthermore, prognostic genes were selected using univariate Cox and least absolute shrinkage and selection operator (LASSO) regression analyses. Depending on the prognostic genes, a risk model was constructed, and risk scores were determined. Moreover, MM samples from MMRF-CoMMpass were sorted into high- and low-risk teams on account of the median risk score. Additionally, correlations among clinical characteristics, gene set variation analysis (GSVA), gene set enrichment analysis (GSEA), immune analysis, immunotherapy predictions and the mRNAâmiRNAâlncRNA network were carried out. RESULTS: A total of 898 DEGs, 211 module genes, 24 DE-ARGs and three prognostic genes (AKAP12, C11orf80 and EMP1) were selected for this study. Enrichment analysis revealed that the DEGs were related to 86 GO terms, such as 'cytoplasmic translation', and 41 KEGG pathways, such as 'small cell lung cancer'. A prognostic gene-based risk model was created in MMRF-CoMMpass and confirmed with the GSE57317 dataset. Moreover, a nomogram was established on the basis of independent prognostic factors that have proven to be good predictors. In addition, the immune cell infiltration results suggested that memory B cells were enriched in the high-risk group and that immature B cells were enriched in the low-risk group. Finally, the mRNAâmiRNAâlncRNA network demonstrated that hsa-miR-508-5p was tightly associated with EMP1 and AKAP12. RTâqPCR was used to validate the expression of the genes associated with prognosis. CONCLUSION: A new prognostic model of MM associated with ARGs was created and validated, providing a new perspective for exploring the connection between ARGs and MM.
Assuntos
Perfilação da Expressão Gênica , Mieloma Múltiplo , Neovascularização Patológica , Humanos , Mieloma Múltiplo/genética , Mieloma Múltiplo/patologia , Prognóstico , Neovascularização Patológica/genética , Regulação Neoplásica da Expressão Gênica , Biomarcadores Tumorais/genética , Redes Reguladoras de Genes , Bases de Dados Genéticas , Ontologia Genética , AngiogêneseRESUMO
The development of computational tools for the systematic prediction of metabolic vulnerabilities of cancer cells constitutes a central question in systems biology. Here, we present gmctool, a freely accessible online tool that allows us to accomplish this task in a simple, efficient and intuitive environment. gmctool exploits the concept of genetic Minimal Cut Sets (gMCSs), a theoretical approach to synthetic lethality based on genome-scale metabolic networks, including a unique database of synthetic lethals computed from Human1, the most recent metabolic reconstruction of human cells. gmctool introduces qualitative and quantitative improvements over our previously developed algorithms to predict, visualize and analyze metabolic vulnerabilities in cancer, demonstrating a superior performance than competing algorithms. A detailed illustration of gmctool is presented for multiple myeloma (MM), an incurable hematological malignancy. We provide in vitro experimental evidence for the essentiality of CTPS1 (CTPS synthase) and UAP1 (UDP-N-Acetylglucosamine Pyrophosphorylase 1) in specific MM patient subgroups.
Assuntos
Algoritmos , Redes e Vias Metabólicas , Mieloma Múltiplo , Humanos , Redes e Vias Metabólicas/genética , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Biologia Computacional/métodos , Neoplasias/genética , Neoplasias/metabolismo , Software , Biologia de Sistemas/métodos , Mutações Sintéticas Letais/genéticaRESUMO
Lenalidomide (LEN) is widely used immunomodulatory drug (IMiD). Nonetheless, despite its efficacy, over time patients become resistant to LEN and relapse. Due to high clinical relevance, drug resistance in MM is being thoroughly investigated. However, less is known about predictors of good response to LEN-based treatment. The aim of this study was to identify molecular pathways associated with good and long response to LEN. The study included newly diagnosed MM patients (NDMM) and MM patients treated with first-line LEN and dexamethasone (RD) who achieved and least very good partial remission (VGPR). RNA was isolated from MM cells and new-generation sequencing was performed. Obtained results were validated with qRT-PCR. A global increase in gene expression was found in the RD group compared to NDMM, suggesting the involvement of epigenetic mechanisms. Moreover, upregulation of genes controlling the interaction within MM niche was detected. Next, genes controlling immune response were upregulated. In particular, the gene encoding the IL-17 receptor was overexpressed in the RD group which is a novel finding. This should be emphasized because IL-17-related signaling can potentially be targeted, providing the rationale for future research. Establishing the molecular background associated with long-lasting and profound response to LEN may improve LEN-based chemotherapy regimens and facilitate the development of adjuvant therapies to enhance its anti-MM activity.
Assuntos
Dexametasona , Lenalidomida , Mieloma Múltiplo , Receptores de Interleucina-17 , Humanos , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/genética , Dexametasona/farmacologia , Dexametasona/uso terapêutico , Feminino , Masculino , Lenalidomida/farmacologia , Lenalidomida/uso terapêutico , Receptores de Interleucina-17/metabolismo , Receptores de Interleucina-17/genética , Pessoa de Meia-Idade , Idoso , Plasmócitos/metabolismo , Plasmócitos/efeitos dos fármacos , Plasmócitos/imunologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologiaRESUMO
BACKGROUND: In the era of novel agents, the clinical outcomes of induction treatment and the impact of the number of high-risk cytogenetic abnormalities (HRA) in newly diagnosed multiple myeloma (NDMM) need to be explored. OBJECTIVE: Through this study, we aim to analyze the effectiveness of different induction treatments and explore the survival outcomes of patients with varying numbers of HRA. METHODS: A total of 734 patients from seven medical centers were included in our study. RESULTS: Patients in the CD38 monoclonal antibody or IMiDs plus proteasome inhibitors (PI) groups had significantly superior overall survival (OS) and progression-free survival (PFS) compared to those receiving IMiDs or PI alone. Additionally, the CD38 monoclonal antibody conferred a PFS advantage over IMiDs plus PI. Patients with ≥ 2 high-risk cytogenetic abnormalities (HRA) exhibited an extremely poor prognosis and should be considered ultra-high-risk individuals in multiple myeloma (MM). The CD38 monoclonal antibody, transplantation, and achieving minimal residual disease (MRD) negativity only partly mitigated the poor prognosis in patients with HRA. Furthermore, patients with 1q21 gain/amplification (1q21+) only had a significantly worse prognosis compared to patients without HRA, and those with 1q21+ plus del17p or t(4;14) exhibited an inferior prognosis compared to those with 1q21+ alone. CONCLUSION: Our results suggested that double-hit multiple myeloma was associated with extremely poor survival outcomes, and more effective treatments needed to be explored for this particular subtype of MM.
Assuntos
Aberrações Cromossômicas , Mieloma Múltiplo , Humanos , Mieloma Múltiplo/genética , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/mortalidade , Mieloma Múltiplo/terapia , Mieloma Múltiplo/patologia , Masculino , Feminino , Idoso , Pessoa de Meia-Idade , Prognóstico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Adulto , Quimioterapia de Indução , Resultado do Tratamento , Idoso de 80 Anos ou mais , Inibidores de Proteassoma/uso terapêutico , ADP-Ribosil Ciclase 1/metabolismo , Intervalo Livre de Progressão , Estudos RetrospectivosRESUMO
The impact of exosome-mediated crosstalk between multiple myeloma (MM) cells and osteoclasts (OCs) on bone lesions remains to be investigated. Here, we identified NSUN2 and YBX1-mediated m5C modifications upregulated LncRNA MALAT1 expression in MM cells, which could be transported to OCs via exosomes and promote bone lesions. Methodologically, RNA-seq was carried out to detect the cargoes of exosomes. TRAP staining and WB were used to evaluate osteoclastogenesis in vitro. Micro-CT and bone histomorphometric analyses were performed to identify bone destruction in vivo. RNA pull-down, RIP, MeRIP, and luciferase reporter assays were used to test the interactions between molecules. The clinical features of MALAT1, NSUN2 and YBX1 were verified through public datasets and clinicopathological data analyses. Mechanistically, MALAT1 was the highest expressed lncRNA in U266 exosomes and could be transported to RAW264.7 cells. MALAT1 could enhance the differentiation of RAW264.7 cells into OCs by stimulating RANKL expression and its downstream AKT and MAPKs signaling pathways via a ceRNA mechanism. Additionally, MALAT1 could be modified by NSUN2, an m5C methyltransferase, which in turn stabilized MALAT1 through the "reader" YBX1. Clinical studies indicated a notable positive correlation between MALAT1, NSUN2, YBX1 levels and bone destruction features, as well as with RANKL expression.
Assuntos
Exossomos , Mieloma Múltiplo , Osteoclastos , Ligante RANK , RNA Longo não Codificante , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Animais , Camundongos , Humanos , Exossomos/metabolismo , Exossomos/genética , Ligante RANK/metabolismo , Ligante RANK/genética , Osteoclastos/metabolismo , Osteoclastos/patologia , Células RAW 264.7 , Proteína 1 de Ligação a Y-Box/metabolismo , Proteína 1 de Ligação a Y-Box/genética , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , MasculinoRESUMO
Multiple myeloma (MM) is a hematologic malignancy characterized by clonal proliferation of plasma cells. Recent advances in next-generation sequencing technologies have facilitated in-depth genetic exploration of MM, unveiling a more comprehensive genomic landscape that extends beyond classical chromosomal alterations, such as IGH translocations and hyperdiploidy. These studies have elucidated recurrent mutations across various functional pathways including those involving MAPK, NF-κB, cell cycle regulation, and epigenetic modulation. With respect to clinical utility, studies have shown that the number of genetic alterations and biallelic events in TP53 are associated with worse prognosis, and CRBN mutations with resistance to immunomodulatory drugs. We recently analyzed the full landscape of genetic alterations in relapsed and refractory MM using circulating tumor DNA (ctDNA), revealing TP53 mutations as the most frequent driver mutation. Notably, more than half of TP53 mutations were present in only ctDNA, suggesting a subclonal origin. Mutations in six genes, including KRAS and TP53, were associated with poor progression-free survival. In addition, the number of ctDNA mutations was identified as a prognostic factor independent of IGH translocations and clinical factors. Here we summarize recent progress in genetic analysis of MM, focusing on clinical relevance.
Assuntos
Mieloma Múltiplo , Mutação , Mieloma Múltiplo/genética , Mieloma Múltiplo/diagnóstico , Humanos , PrognósticoRESUMO
Tandem autologous stem cell transplantation can improve the prognosis of patients with multiple myeloma. However, the precise role of tandem transplantation remains debatable. We evaluated the clinical benefits of tandem transplantation retrospectively. Of the 655 included patients, 117 underwent tandem transplantation; the remaining were assigned to the control group. After a single transplantation, the tandem group achieved a complete remission (CR) rate of 24.8%, which increased to 46.2% after a second transplantation. The tandem group had a significantly longer median PFS than the control group in patients with International Staging System (ISS) III and high-risk cytogenetics (23.1 vs. 14.7 months, p = 0.007 for ISS III; 21.7 vs. 13.2 months, p = 0.042 for high-risk cytogenetics). The tandem group exhibited significantly superior PFS to the control group (20.3 vs. 12.6 months, p = 0.003) among patients who failed to achieve CR after a single transplantation. Tandem transplantation was associated with significantly improved PFS after adjusting for maintenance therapy in patients with ISS III, those with high-risk cytogenetics, and those who did not achieve CR after a single transplantation. Following propensity score matching, the tandem group exhibited significantly longer PFS than the control group (30.3 vs. 13.5 months, p = 0.028). Tandem transplantation should be considered in high-risk patients.
Assuntos
Transplante de Células-Tronco Hematopoéticas , Mieloma Múltiplo , Transplante Autólogo , Humanos , Mieloma Múltiplo/terapia , Mieloma Múltiplo/mortalidade , Mieloma Múltiplo/genética , Pessoa de Meia-Idade , Masculino , Feminino , República da Coreia/epidemiologia , Transplante de Células-Tronco Hematopoéticas/métodos , Estudos Retrospectivos , Adulto , Idoso , Prognóstico , Resultado do Tratamento , Indução de RemissãoRESUMO
Multiple myeloma (MM) is linked to chronic NF-κB activity in myeloma cells, but this activity is generally considered a cell-autonomous property of the cancer cells. The precise extent of NF-κB activation and the contributions of the physical microenvironment and of cell-to-cell communications remain largely unknown. By quantitative immunofluorescence, we found that NF-κB is mildly and heterogeneously activated in a fraction of MM cells in human BMs, while only a minority of MM cells shows a strong activation. To gain quantitative insights on NF-κB activation in living MM cells, we combined advanced live imaging of endogenous p65 Venus-knocked-in in MM.1S and HS-5 cell lines to model MM and mesenchymal stromal cells (MSCs), cell co-cultures, microfluidics and custom microbioreactors to mimic the 3D-interactions within the bone marrow (BM) microenvironment. We found that i) reciprocal MM-MSC paracrine crosstalk and cell-to-scaffold interactions shape the inflammatory response in the BM; ii) the pro-inflammatory cytokine IL-1ß, abundant in MM patients' plasma, activates MSCs, whose paracrine signals are responsible for strong NF-κB activation in a minority of MM cells; iii) IL-1ß, but not TNF-α, activates NF-κB in vivo in BM-engrafted MM cells, while its receptor inhibitor Anakinra reduces the global NF-κB activation. We propose that NF-κB activation in the BM of MM patients is mild, restricted to a minority of cells and modulated by the interplay of restraining physical microenvironmental cues and activating IL-1ß-dependent stroma-to-MM crosstalk.
Assuntos
Técnicas de Cocultura , Células-Tronco Mesenquimais , Mieloma Múltiplo , NF-kappa B , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Mieloma Múltiplo/genética , Humanos , Células-Tronco Mesenquimais/metabolismo , NF-kappa B/metabolismo , Linhagem Celular Tumoral , Interleucina-1beta/metabolismo , Microambiente Tumoral , Comunicação Celular , Células Estromais/metabolismo , Células Estromais/patologia , Comunicação Parácrina , Transdução de Sinais , Fator de Transcrição RelA/metabolismoRESUMO
Multiple myeloma (MM) is a highly heterogeneous hematological malignancy that is currently incurable. Individualized therapeutic approaches based on accurate risk assessment are essential for improving the prognosis of MM patients. Nevertheless, current prognostic models for MM exhibit certain limitations and prognosis heterogeneity still an unresolved issue. Recent studies have highlighted the pivotal involvement of mitochondrial autophagy in the development and drug sensitivity of MM. This study seeks to conduct an integrative analysis of the prognostic significance and immune microenvironment of mitophagy-related signature in MM, with the aim of constructing a novel predictive risk model. GSE4581 and GSE47552 datasets were acquired from the Gene Expression Omnibus database. MM-differentially expressed genes (DEGs) were identified by limma between MM samples and normal samples in GSE47552. Mitophagy key module genes were obtained by weighted gene co-expression network analysis in the Cancer Genome Atlas (TCGA)-MM dataset. Mitophagy DEGs were identified by the overlap genes between MM-DEGs and mitophagy key module genes. Prognostic genes were selected through univariate Cox regression and least absolute shrinkage and selection operator (LASSO) analysis, and a risk model was subsequently constructed based on these prognostic genes. Subsequently, the MM samples were stratified into high- and low-risk groups based on their median risk scores. The validity of the risk model was further evaluated using the GSE4581 dataset. Moreover, a nomogram was developed using the independent prognostic factors identified from the risk score and various clinical indicators. Additionally, analyses were conducted on immune infiltration, immune scores, immune checkpoint, and chemotherapy drug sensitivity. The 17 mitophagy DEGs were obtained by intersection of 803 MM-DEGs and 1084 mitophagy key module genes. Five prognostic genes (CDC6, PRIM1, SNRPB, TOP2A, and ZNF486) were selected via LASSO and univariate cox regression analyses. The predictive performance of the risk model, which was constructed based on the five prognostic genes, demonstrated favorable results in both TCGA-MM and GSE4581 datasets as indicated by the receiver operating characteristic (ROC) curves. In addition, calibration curve, ROC curve, and decision curve analysis curve corroborated that the nomogram exhibited superior predictive accuracy for MM. Furthermore, immune analysis results indicated a significant difference in stromal scores of two risk groups categorized on median risk scores. And four immune checkpoints (CD274, CTLA4, LAG3, and PDCD1LG2) showed significant differences in different risk groups. The analysis of chemotherapy drug sensitivity revealed that etoposide and doxorubicin, which target TOP2A, exhibited superior treatment outcomes in the high-risk group. A novel prognostic model for MM was developed and validated, demonstrating significant potential in predicting patient outcomes and providing valuable guidance for personalized immunotherapy counseling.
Assuntos
Mitofagia , Mieloma Múltiplo , Humanos , Mieloma Múltiplo/genética , Mieloma Múltiplo/patologia , Prognóstico , Mitofagia/genética , Biomarcadores Tumorais/genética , Perfilação da Expressão Gênica , Medição de Risco , Regulação Neoplásica da Expressão Gênica , Microambiente Tumoral/genéticaRESUMO
Many observational studies and experiments have found a strong association between lipid levels and adipokines and multiple myeloma (MM), but the causal relationship between lipid levels, adipokines and MM remains to be determined. We performed a two-sample and multivariate MR analysis to investigate the causal relationship between lipid levels, adipokines and MM. Total cholesterol(TC), high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), and triglycerides (TG) were used to represent lipid levels, and adiponectin, leptin, and resistin were used to represent adipokines. Genetic data for each index and MM were obtained from the Integrated Epidemiology Unit (IEU) Genome-Wide Association Study (GWAS) database, and two-sample MR analyses were performed, as well as multivariate MR analyses of adipokines for causality of MM using BMI as an adjusting factor. In the analyzed results, no significant causal association was found between adipokines, lipid levels and multiple myeloma, and after adjusting for BMI, an association between adipokines and MM was still not found. The results of this MR study do not support an association between genetically predicted adipokines, lipid levels, and risk of MM, but we cannot rule out the existence of a weak association. The mechanisms need to be further investigated.
Assuntos
Adipocinas , Estudo de Associação Genômica Ampla , Análise da Randomização Mendeliana , Mieloma Múltiplo , Mieloma Múltiplo/genética , Mieloma Múltiplo/sangue , Humanos , Adipocinas/sangue , Adipocinas/genética , Lipídeos/sangue , Polimorfismo de Nucleotídeo Único , Triglicerídeos/sangue , Adiponectina/genética , Adiponectina/sangue , Leptina/sangue , Leptina/genética , HDL-Colesterol/sangueRESUMO
Multiple myeloma (MM) is the second most common hematological cancer all over the world. Long non-coding RNA (lncRNA) nuclear-enriched autosomal transcript-1 (NEAT1) have been reported to play important roles in the development and progression of multiple human malignancies like MM. However, the functional role and molecular mechanism of NEAT1 in MM progression still needs more support to identify potential targets of MM. In the present study, we focused on the clinical and biological significance of NEAT1 in MM. We demonstrated that NEAT1 was up-regulated in MM tissues and cell line. NEAT1 silencing significantly inhibited cell proliferation and promoted cell apoptosis in vitro. And we illustrated that miR-485-5p was a direct target of NEAT1 and the effect of down-regulated NEAT1 on MM cells was partially reversed by the miR-485-5p antisense oligonucleotide (ASO-miR-485-5p). Further investigation revealed that ABCB8 directly interacted with miR-485-5p. Similarly, in vivo experiments confirmed that down-regulated NEAT1 inhibited tumor growth and ABCB8 expression. Taken together, our results demonstrate for the first time that NEAT1/miR-485-5p/ABCB8 axis may be a key pathway for the development and progression of MM, and they may provide a novel avenue for targeted therapy.
The effects of the comprehensive NEAT1miRNA regulatory network in MM needs more support. Our results demonstrate for the first time that NEAT1/miR-485-5p/ABCB8 axis may be a key pathway for the development and progression of MM, which may provide a novel avenue for targeted therapy.
Assuntos
MicroRNAs , Mieloma Múltiplo , RNA Longo não Codificante , Mieloma Múltiplo/genética , Mieloma Múltiplo/patologia , Mieloma Múltiplo/metabolismo , RNA Longo não Codificante/genética , Humanos , MicroRNAs/genética , Camundongos , Transformação Celular Neoplásica/genética , Linhagem Celular Tumoral , Animais , Regulação Neoplásica da Expressão Gênica , Proliferação de Células , Masculino , Feminino , ApoptoseRESUMO
OBJECTIVE: To investigate the effects of ATG5 and ATG7 genes on the sensitivity of multiple myeloma (MM) cell line RPMI-8226 cells to ferroptosis. METHODS: CRISPR/Cas9 technology was used to knock out the autophagy key genes ATG5 and ATG7 in RPMI-8226 cells. Western blot was used to identify gene knockout cells, and detect the expression changes of autophagy-related proteins P62 and LC3B. Flow cytometry was used to detect the change of sensitivity of gene knockout cells to RSL3. The content of intracellular ferrous ions and reactive oxygen species (ROS) level in gene knockout cells were detected. RESULTS: Western blot result confirmed that ATG5 and ATG7 genes were knocked out successfully in RPMI-8226 cells. The result of flow cytometry showed that the cell viability of RPMI-8226 cells was dose-dependent on different concentrations of RSL3 (r =-0.969). RSL3 (10 µmol/L) was used to induce ferroptosis in cells of control group and gene knockout groups, then the cell viability in gene knockout groups were both higher than control group after 48 hours (both P < 0.001). After knocking out the ATG5 and ATG7 genes, the content of intracellular Fe2+ decreased significantly compared with control group (both P < 0.01), and the ROS level also decreased (both P < 0.001). CONCLUSION: Knockout of ATG5 and ATG7 genes can inhibit the ferroptosis of MM cells, and LAP pathway may be involved in the regulation.
Assuntos
Proteína 5 Relacionada à Autofagia , Proteína 7 Relacionada à Autofagia , Ferroptose , Espécies Reativas de Oxigênio , Humanos , Proteína 5 Relacionada à Autofagia/genética , Proteína 7 Relacionada à Autofagia/genética , Linhagem Celular Tumoral , Espécies Reativas de Oxigênio/metabolismo , Técnicas de Inativação de Genes , Autofagia , Sobrevivência Celular , Mieloma Múltiplo/genéticaRESUMO
BACKGROUND: Chromosome 1q21 aberrations are one of the most common cytogenetic abnormalities in patients with multiple myeloma (MM). However, the prognostic value remains controversial. This study aimed to determine the prognostic value of numerical abnormalities of chromosome 1q21 for newly diagnosed patients with MM patients in Chinese population. METHODS: We retrospectively analyzed 629 patients with newly diagnosed MM who received the detection of chromosome 1q21 by fluorescence in situ hybridization in China. RESULTS: Among 629 patients, 309 (49.1%) had 1q21 abnormalities, of which 187 (29.7%) had three copies and 122 (19.4%) had four or more copies. Patients with two copies of 1q21 had a significantly longer median overall survival (OS) than those with three copies or ≥4 copies and also had longer progression-free survival (PFS). However, patients with three or ≥4 copies had similar OS and PFS. Univariate Cox proportional hazards regression analyses determined that 1q21 aberrations are associated with shorter OS and PFS. 1q21 aberrations are also independent poor prognostic factors for OS and PFS in multivariable analyses. Del(17p), t(4;14), and t(14;16) are common high-risk cytogenetic abnormalities (HRCAs) in patients with MM. Patients with 1q21+ alone or 1q21+ combined with HRCAs had shorter OS and PFS than patients without cytogenetic abnormalities. Patients with 1q21+ and t(11;14) also had shorter PFS but had similar OS than patients without cytogenetic abnormalities. CONCLUSION: Our study showed that chromosome 1q21 aberrations are poor prognostic factors for newly diagnosed patients with MM.
Assuntos
Aberrações Cromossômicas , Cromossomos Humanos Par 1 , Mieloma Múltiplo , Humanos , Mieloma Múltiplo/genética , Mieloma Múltiplo/mortalidade , Mieloma Múltiplo/diagnóstico , Cromossomos Humanos Par 1/genética , Masculino , Feminino , Pessoa de Meia-Idade , Idoso , Prognóstico , Adulto , Estudos Retrospectivos , Idoso de 80 Anos ou mais , Hibridização in Situ FluorescenteRESUMO
BACKGROUND: Lipid levels have been suggset to be correlated with multiple myeloma (MM) risk, though causality remains unconfirmed. To explore this further, a detailed study combining meta-analysis and Mendelian randomization (MR) was conducted. METHODS: Literature searches were performed on PubMed and Embase; summary data for plasma lipid traits were extracted from the IEU and MM data from the FinnGen database. Meta-analysis and MR were utilized to analyze the link of lipids with MM risk, including mediator MR to identify potential mediators. The study was conducted in accordance with PRISMA and STROBE-MR guidelines. RESULTS: Observational studies analyzed through meta-analysis showed that elevated levels of LDL, HDL, total cholesterol (TC), and triglycerides correlate with a lower risk of MM, with HRs of 0.73, 0.59, 0.60, and 0.84, respectively. MR analysis confirmed a potential causal link of triglyceride with a reduced MM risk (OR: 0.67, 95% CI: 0.46-0.98), independent of BMI. Mediation analysis pointed to X-11,423-O-sulfo-L-tyrosine and neuropilin-2 as potential mediators. CONCLUSIONS: The findings suggest that higher lipid levels (LDL, HDL, TC, and triglycerides) are linked with a reduced MM risk, and higher triglyceride levels are causally associated with a reduced MM risk. This suggests new avenues for therapeutic interventions targeting MM.
Assuntos
Colesterol , Análise da Randomização Mendeliana , Mieloma Múltiplo , Triglicerídeos , Humanos , Colesterol/sangue , Mieloma Múltiplo/sangue , Mieloma Múltiplo/epidemiologia , Mieloma Múltiplo/genética , Fatores de Risco , Triglicerídeos/sangueRESUMO
The t(11;14) translocation is among the most prevalent cytogenetic abnormalities in multiple myeloma (MM), distinguished by its unique features and biology that have been thoroughly explored for decades. What further sets this MM subtype apart is its oscillating prognostic significance, from initially being considered a favorable alteration to intermediate risk and potential future reclassification as favorable risk. Despite not being inherently a high-risk alteration indicative of an aggressive phenotype, it appears that t(11;14)-MM is less responsive to novel agents like proteasome inhibitors and immunomodulatory drugs which have otherwise transformed the disease's treatment landscape, perhaps partially explained by its reduced propensity for immunoglobulin production and oligosecretory nature. However, its distinct reliance on Bcl-2 has heightened its sensitivity to venetoclax. Further subclassification based on morphological and genomic characteristics could enhance our prediction models of treatment responses and enable more tailored therapeutic strategies for patients. This review aims to encapsulate the existing research evidence in this area.