Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.040
Filtrer
1.
Int J Mol Sci ; 25(12)2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38928138

RÉSUMÉ

Based on the lack of differences in progression-free and overall survival after a median follow-up of 93 months in our HOVON-65/GMMG-HD4 trial (German part; n = 395) randomizing VAD induction (vincristin/adriamycin/dexamthasone)/tandem-transplantation/thalidomide-maintenance vs. PAD induction (bortezomib/adriamycin/dexamethasone)/tandem transplantation/bortezomib maintenance, we discern how chromosomal aberrations determine long-term prognosis by different patterns of association with proliferation and treatment-dependent response, whether responses achieved by different regimens are equal regarding prognosis, and whether subpopulations of patients could be defined as treatable without upfront "novel agents" in cases of limited resources, e.g., in low- or middle-income countries. Serum parameters and risk factors were assessed in 395 patients. CD138-purified plasma cells were subjected to fluorescence in situ hybridization (n = 354) and gene expression profiling (n = 204). We found chromosomal aberrations to be associated in four patterns with survival, proliferation, and response: deletion (del) del17p13, del8p21, del13q14, (gain) 1q21+, and translocation t(4;14) (all adverse) associate with higher proliferation. Of these, del17p is associated with an adverse response (pattern 1), and 1q21+, t(4;14), and del13q14 with a treatment-dependent better response (pattern 2). Hyperdiploidy associates with lower proliferation without impacting response or survival (pattern 3). Translocation t(11;14) has no association with survival but a treatment-dependent adverse response (pattern 4). Significantly fewer patients reach a near-complete response or better with "conventional" (VAD) vs. bortezomib-based treatment after induction or high-dose melphalan. These patients, however, show significantly better median progression-free and overall survival. Molecularly, patients responding to the two regimens differ in gene expression, indicating distinct biological properties of the responding myeloma cells. Patients with normal renal function (89.4%), low cytogenetic risk (72.5%), or low proliferation rate (37.9%) neither benefit in progression-free nor overall survival from bortezomib-based upfront treatment. We conclude that response level, the treatment by which it is achieved, and molecular background determine long-term prognosis. Chromosomal aberrations are associated in four patterns with proliferation and treatment-dependent responses. Associations with faster and deeper responses can be deceptive in the case of prognostically adverse aberrations 1q21+ and t(4;14). Far from advocating a return to "outdated" treatments, if resources do not permit state-of-the-art-treatment, normal renal function and/or molecular profiling identifies patient subpopulations doing well without upfront "novel agents".


Sujet(s)
Aberrations des chromosomes , Myélome multiple , Humains , Myélome multiple/génétique , Myélome multiple/traitement médicamenteux , Myélome multiple/mortalité , Myélome multiple/anatomopathologie , Femelle , Mâle , Adulte d'âge moyen , Sujet âgé , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Pronostic , Adulte , Pays en voie de développement , Dexaméthasone/usage thérapeutique , Dexaméthasone/pharmacologie , Bortézomib/usage thérapeutique , Bortézomib/pharmacologie , Thalidomide/usage thérapeutique
2.
Front Immunol ; 15: 1393906, 2024.
Article de Anglais | MEDLINE | ID: mdl-38911853

RÉSUMÉ

Infections are common in plasma cell cancer multiple myeloma (MM) due to disease-related immune deficiencies and cancer treatment. Myeloma cells express Toll-like receptors (TLRs), and TLR activation has been shown to induce proliferative and pro-survival signals in cancer cells. MM is a complex and heterogeneous disease, and expression levels of TLRs as well as downstream signaling components are likely to differ between patients. Here, we show that in a large cohort of patients, TLR1, TLR4, TLR6, TLR9, and TLR10 are the most highly expressed in primary CD138+ cells. Using an MM cell line expressing TLR4 and TLR9 as a model, we demonstrate that TLR4 and TLR9 activation promoted the expression of well-established pro-survival and oncogenes in MM such as MYC, IRF4, NFKB, and BCL2. TLR4 and TLR9 activation inhibited the efficacy of proteasome inhibitors bortezomib and carfilzomib, drugs used in the treatment of MM. Inhibiting the autophagosome-lysosome protein degradation pathway by hydroxychloroquine (HCQ) diminished the protective effect of TLR activation on proteasome inhibitor-induced cytotoxicity. We also found that TLR signaling downregulated the expression of TNFRSF17, the gene encoding for B-cell maturation antigen (BCMA). MYC, BCL2, and BCL2L1 were upregulated in approximately 50% of primary cells, while the response to TLR signaling in terms of TNFRSF17 expression was dichotomous, as an equal fraction of patients showed upregulation and downregulation of the gene. While proteasome inhibitors are part of first-line MM treatment, several of the new anti-MM immune therapeutic drugs target BCMA. Thus, TLR activation may render MM cells less responsive to commonly used anti-myeloma drugs.


Sujet(s)
Antigène de maturation des cellules B , Régulation de l'expression des gènes tumoraux , Myélome multiple , Protéines proto-oncogènes c-myc , Transduction du signal , Récepteurs de type Toll , Humains , Myélome multiple/génétique , Myélome multiple/immunologie , Myélome multiple/métabolisme , Antigène de maturation des cellules B/génétique , Antigène de maturation des cellules B/métabolisme , Antigène de maturation des cellules B/immunologie , Lignée cellulaire tumorale , Récepteurs de type Toll/métabolisme , Protéines proto-oncogènes c-myc/génétique , Protéines proto-oncogènes c-myc/métabolisme , Protéines proto-oncogènes c-bcl-2/génétique , Protéines proto-oncogènes c-bcl-2/métabolisme , Bortézomib/pharmacologie , Bortézomib/usage thérapeutique , Mâle
3.
Phytomedicine ; 131: 155790, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38851099

RÉSUMÉ

BACKGROUND: A balanced protein homeostasis network helps cholangiocarcinoma (CCA) maintain their oncogenic growth, and disrupting proteostasis therapeutically will induce proteotoxic stress. Phosphatase and tensin homolog (PTEN) have been reported to be involved in proteostasis, and PTEN-associated pathways are commonly altered in CCA. Celastrol, a triterpene from plants, exhibits cytotoxic effects in various types of cancer. However, the underlying mechanisms remain unclear. PURPOSE: We investigated the therapeutic effect of celastrol in CCA and identified the molecular characteristics of tumors that were sensitive to celastrol. The target of celastrol was explored. We then evaluated the candidate combination therapeutic strategy to increase the effectiveness of celastrol in celastrol-insensitive CCA tumors. METHODS: Various CCA cells were categorized as either celastrol-sensitive or celastrol-insensitive based on their response to celastrol. The molecular characteristics of cells from different groups were determined by RNA-seq. PTEN status and its role in proteasome activity in CCA cells were investigated. The CMAP analysis, molecular docking, and functional assay were performed to explore the effect of celastrol on proteasome activities. The correlation between PTEN status and clinical outcomes, as well as proteasomal activity, were measured in CCA patients. The synergistic therapeutic effect of autophagy inhibitors on celastrol-insensitive CCA cells were measured. RESULTS: Diverse responses to celastrol were observed in CCA cells. PTEN expression varied among different CCA cells, and its status could impact cell sensitivity to celastrol: PTENhigh tumor cells were resistant to celastrol, while PTENlow cells were more sensitive. Celastrol induced proteasomal dysregulation in CCA cells by directly targeting PSMB5. Cells with low PTEN status transcriptionally promoted proteasome subunit expression in an AKT-dependent manner, making these cells more reliant on proteasomal activities to maintain proteostasis. This caused the PTENlow CCA cells sensitive to celastrol. A negative correlation was found between PTEN levels and the proteasome signature in CCA patients. Moreover, celastrol treatment could induce autophagy in PTENhigh CCA cells. Disrupting the autophagic pathway in PTENhigh CCA cells enhanced the cytotoxic effect of celastrol. CONCLUSION: PTEN status in CCA cells determines their sensitivity to celastrol, and autophagy inhibitors could enhance the anti-tumor effect in PTENhigh CCA.


Sujet(s)
Tumeurs des canaux biliaires , Cholangiocarcinome , Phosphohydrolase PTEN , Triterpènes pentacycliques , Triterpènes , Cholangiocarcinome/traitement médicamenteux , Triterpènes pentacycliques/pharmacologie , Phosphohydrolase PTEN/métabolisme , Humains , Lignée cellulaire tumorale , Tumeurs des canaux biliaires/traitement médicamenteux , Triterpènes/pharmacologie , Simulation de docking moléculaire , Tripterygium/composition chimique , Antinéoplasiques d'origine végétale/pharmacologie , Proteasome endopeptidase complex/métabolisme , Proteasome endopeptidase complex/effets des médicaments et des substances chimiques , Autophagie/effets des médicaments et des substances chimiques , Bortézomib/pharmacologie
4.
Cell Mol Life Sci ; 81(1): 276, 2024 Jun 23.
Article de Anglais | MEDLINE | ID: mdl-38909325

RÉSUMÉ

N6-methyladenosine (m6A) is one of the most prevalent and conserved RNA modifications. It controls several biological processes, including the biogenesis and function of circular RNAs (circRNAs), which are a class of covalently closed-single stranded RNAs. Several studies have revealed that proteotoxic stress response induction could be a relevant anticancer therapy in Acute Myeloid Leukemia (AML). Furthermore, a strong molecular interaction between the m6A mRNA modification factors and the suppression of the proteotoxic stress response has emerged. Since the proteasome inhibition leading to the imbalance in protein homeostasis is strictly linked to the stress response induction, we investigated the role of Bortezomib (Btz) on m6A regulation and in particular its impact on the modulation of m6A-modified circRNAs expression. Here, we show that treating AML cells with Btz downregulated the expression of the m6A regulator WTAP at translational level, mainly because of increased oxidative stress. Indeed, Btz treatment promoted oxidative stress, with ROS generation and HMOX-1 activation and administration of the reducing agent N-acetylcysteine restored WTAP expression. Additionally, we identified m6A-modified circRNAs modulated by Btz treatment, including circHIPK3, which is implicated in protein folding and oxidative stress regulation. These results highlight the intricate molecular networks involved in oxidative and ER stress induction in AML cells following proteotoxic stress response, laying the groundwork for future therapeutic strategies targeting these pathways.


Sujet(s)
Adénosine , Leucémie aigüe myéloïde , Stress oxydatif , ARN circulaire , Humains , ARN circulaire/génétique , ARN circulaire/métabolisme , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/anatomopathologie , Leucémie aigüe myéloïde/métabolisme , Leucémie aigüe myéloïde/traitement médicamenteux , Adénosine/analogues et dérivés , Adénosine/métabolisme , Adénosine/pharmacologie , Stress oxydatif/effets des médicaments et des substances chimiques , Bortézomib/pharmacologie , Lignée cellulaire tumorale , Espèces réactives de l'oxygène/métabolisme , Facteurs d'épissage des ARN/métabolisme , Facteurs d'épissage des ARN/génétique , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/génétique , Cellules souches tumorales/métabolisme , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/anatomopathologie , Heme oxygenase-1/métabolisme , Heme oxygenase-1/génétique , Protein-Serine-Threonine Kinases , Protéines et peptides de signalisation intracellulaire
5.
Asian Pac J Cancer Prev ; 25(6): 1959-1967, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38918657

RÉSUMÉ

BACKGROUND: As one of the main molecules in BCR-ABL signaling, c-Myc acts as a pivotal key in disease progression and disruption of long-term remission in patients with CML. OBJECTIVES: To clarify the effects of c-Myc inhibition in CML, we examined the anti-tumor property of a well-known small molecule inhibitor of c-Myc 10058-F4 on K562 cell line. METHODS: This experimental study was conducted in K562 cell line for evaluation of cytotoxic activity of 10058-F4 using Trypan blue and MTT assays. Flow cytometry and Quantitative RT-PCR analysis were also conducted to determine its mechanism of action. Additionally, Annexin/PI staining was performed for apoptosis assessment. RESULTS: The results of Trypan blue and MTT assay demonstrated that inhibition of c-Myc, as shown by suppression of c-Myc expression and its associated genes PP2A, CIP2A, and hTERT, could decrease viability and metabolic activity of K562 cells, respectively. Moreover, a robust elevation in cell population in G1-phase coupled with up-regulation of p21 and p27 expression shows that 10058-F4 could hamper cell proliferation, at least partly, through induction of G1 arrest. Accordingly, we found that 10058-F4 induced apoptosis via increasing Bax and Bad; In contrast, no significant alterations were observed NF-KB pathway-targeted anti-apoptotic genes in the mRNA levels. Notably, disruption of the NF-κB pathway with bortezomib as a common proteasome inhibitor sensitized K562 cells to the cytotoxic effect of 10058-F4, substantiating the fact that the NF-κB axis functions probably attenuate the K562 cells sensitivity to c-Myc inhibition. CONCLUSIONS: It can be concluded from the results of this study that inhibition of c-Myc induces anti-neoplastic effects on CML-derived K562 cells as well as increases the efficacy of imatinib. For further insight into the safety and effectiveness of 10058-F4 in CML, in vivo studies will be required.


Sujet(s)
Apoptose , Prolifération cellulaire , Leucémie myéloïde chronique BCR-ABL positive , Protéines proto-oncogènes c-myc , Humains , Leucémie myéloïde chronique BCR-ABL positive/traitement médicamenteux , Leucémie myéloïde chronique BCR-ABL positive/anatomopathologie , Protéines proto-oncogènes c-myc/métabolisme , Protéines proto-oncogènes c-myc/génétique , Apoptose/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cellules K562 , Facteur de transcription NF-kappa B/métabolisme , Facteur de transcription NF-kappa B/antagonistes et inhibiteurs , Antinéoplasiques/pharmacologie , Bortézomib/pharmacologie , Cellules cancéreuses en culture , Acides boroniques/pharmacologie , ARN messager/génétique , Pyrazines/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Telomerase/antagonistes et inhibiteurs
6.
J Am Heart Assoc ; 13(10): e030467, 2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38761081

RÉSUMÉ

BACKGROUND: Many cardiomyopathy-associated FLNC pathogenic variants are heterozygous truncations, and FLNC pathogenic variants are associated with arrhythmias. Arrhythmia triggers in filaminopathy are incompletely understood. METHODS AND RESULTS: We describe an individual with biallelic FLNC pathogenic variants, p.Arg650X and c.970-4A>G, with peripartum cardiomyopathy and ventricular arrhythmias. We also describe clinical findings in probands with FLNC variants including Val2715fs87X, Glu2458Serfs71X, Phe106Leu, and c.970-4A>G with hypertrophic and dilated cardiomyopathy, atrial fibrillation, and ventricular tachycardia. Induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) were generated. The FLNC truncation, Arg650X/c.970-4A>G, showed a marked reduction in filamin C protein consistent with biallelic loss of function mutations. To assess loss of filamin C, gene editing of a healthy control iPSC line was used to generate a homozygous FLNC disruption in the actin binding domain. Because filamin C has been linked to protein quality control, we assessed the necessity of filamin C in iPSC-CMs for response to the proteasome inhibitor bortezomib. After exposure to low-dose bortezomib, FLNC-null iPSC-CMs showed an increase in the chaperone proteins BAG3, HSP70 (heat shock protein 70), and HSPB8 (small heat shock protein B8) and in the autophagy marker LC3I/II. FLNC null iPSC-CMs had prolonged electric field potential, which was further prolonged in the presence of low-dose bortezomib. FLNC null engineered heart tissues had impaired function after low-dose bortezomib. CONCLUSIONS: FLNC pathogenic variants associate with a predisposition to arrhythmias, which can be modeled in iPSC-CMs. Reduction of filamin C prolonged field potential, a surrogate for action potential, and with bortezomib-induced proteasome inhibition, reduced filamin C led to greater arrhythmia potential and impaired function.


Sujet(s)
Filamines , Homéostasie protéique , Filamines/génétique , Filamines/métabolisme , Humains , Femelle , Cellules souches pluripotentes induites/métabolisme , Troubles du rythme cardiaque/génétique , Troubles du rythme cardiaque/métabolisme , Troubles du rythme cardiaque/physiopathologie , Troubles du rythme cardiaque/étiologie , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Cardiomyopathies/génétique , Cardiomyopathies/métabolisme , Cardiomyopathies/physiopathologie , Mâle , Adulte , Mutation , Bortézomib/pharmacologie
7.
Mol Med ; 30(1): 70, 2024 May 24.
Article de Anglais | MEDLINE | ID: mdl-38789926

RÉSUMÉ

BACKGROUND: The development of pulmonary fibrosis involves a cascade of events, in which inflammation mediated by immune cells plays a pivotal role. Chemotherapeutic drugs have been shown to have dual effects on fibrosis, with bleomycin exacerbating pulmonary fibrosis and bortezomib alleviating tissue fibrotic processes. Understanding the intricate interplay between chemotherapeutic drugs, immune responses, and pulmonary fibrosis is likely to serve as the foundation for crafting tailored therapeutic strategies. METHODS: A model of bleomycin-induced pulmonary fibrosis was established, followed by treatment with bortezomib. Tissue samples were collected for analysis of immune cell subsets and functional assessment by flow cytometry and in vitro cell experiments. Additionally, multi-omics analysis was conducted to further elucidate the expression of chemokines and chemokine receptors, as well as the characteristics of cell populations. RESULTS: Here, we observed that the expression of CXCL16 and CXCR6 was elevated in the lung tissue of a pulmonary fibrosis model. In the context of pulmonary fibrosis or TGF-ß1 stimulation in vitro, macrophages exhibited an M2-polarized phenotype and secreted more CXCL16 than those of the control group. Moreover, flow cytometry revealed increased expression levels of CD69 and CXCR6 in pulmonary CD4 T cells during fibrosis progression. The administration of bortezomib alleviated bleomycin-induced pulmonary fibrosis, accompanied by reduced ratio of M2-polarized macrophages and decreased accumulation of CD4 T cells expressing CXCR6. CONCLUSIONS: Our findings provide insights into the key immune players involved in bleomycin-induced pulmonary fibrosis and offer preclinical evidence supporting the repurposing strategy and combination approaches to reduce lung fibrosis.


Sujet(s)
Bléomycine , Bortézomib , Lymphocytes T CD4+ , Chimiokine CXCL16 , Fibrose pulmonaire , Récepteurs CXCR6 , Animaux , Mâle , Souris , Antigènes CD , Antigènes de différenciation des lymphocytes T/métabolisme , Bléomycine/effets indésirables , Bortézomib/pharmacologie , Lymphocytes T CD4+/métabolisme , Lymphocytes T CD4+/immunologie , Chimiokine CXCL16/métabolisme , Chimiotaxie/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Lectines de type C , Macrophages/métabolisme , Macrophages/immunologie , Macrophages/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Fibrose pulmonaire/induit chimiquement , Fibrose pulmonaire/métabolisme , Fibrose pulmonaire/traitement médicamenteux , Récepteurs CXCR6/métabolisme
8.
Oncoimmunology ; 13(1): 2360275, 2024.
Article de Anglais | MEDLINE | ID: mdl-38812570

RÉSUMÉ

Recently, it was revealed that the high-risk, poor-prognosis downregulation of GABA type A receptor-associated protein (GABARAP) causes a defect in both autophagy and surface exposure of calreticulin (CALR) in multiple myeloma (MM) cells responding to bortezomib. Hence, GABARAP-defective MM cells fail to undergo immunogenic cell death.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Antinéoplasiques , Protéines régulatrices de l'apoptose , Bortézomib , Mort cellulaire immunogène , Protéines associées aux microtubules , Myélome multiple , Myélome multiple/traitement médicamenteux , Myélome multiple/anatomopathologie , Myélome multiple/immunologie , Myélome multiple/génétique , Myélome multiple/métabolisme , Humains , Bortézomib/pharmacologie , Bortézomib/usage thérapeutique , Protéines adaptatrices de la transduction du signal/génétique , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/déficit , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Protéines associées aux microtubules/génétique , Protéines associées aux microtubules/métabolisme , Protéines régulatrices de l'apoptose/génétique , Protéines régulatrices de l'apoptose/métabolisme , Mort cellulaire immunogène/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Autophagie/effets des médicaments et des substances chimiques , Calréticuline/métabolisme , Calréticuline/génétique
9.
J Am Soc Mass Spectrom ; 35(6): 1063-1068, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38748611

RÉSUMÉ

Bortezomib, a small dipeptide-like molecule, is a proteasome inhibitor used widely in the treatment of myeloma and lymphoma. This molecule reacts with threonine side chains near the center of the 20S proteasome and disrupts proteostasis by blocking enzymatic sites that are responsible for protein degradation. In this work, we use novel mass-spectrometry-based techniques to examine the influence of bortezomib on the structures and stabilities of the 20S core particle. These studies indicate that bortezomib binding dramatically favors compact 20S structures (in which the axial gate is closed) over larger structures (in which the axial gate is open)─suppressing gate opening by factors of at least ∼400 to 1300 over the temperature range that is studied. Thus, bortezomib may also restrict degradation in the 20S proteasome by preventing substrates from entering the catalytic pore. That bortezomib influences structures at the entrance region of the pore at such a long distance (∼65 to 75 Å) from its binding sites raises a number of interesting biophysical issues.


Sujet(s)
Bortézomib , Proteasome endopeptidase complex , Inhibiteurs du protéasome , Bortézomib/pharmacologie , Bortézomib/composition chimique , Proteasome endopeptidase complex/métabolisme , Proteasome endopeptidase complex/composition chimique , Proteasome endopeptidase complex/effets des médicaments et des substances chimiques , Inhibiteurs du protéasome/composition chimique , Inhibiteurs du protéasome/pharmacologie , Modèles moléculaires , Conformation des protéines/effets des médicaments et des substances chimiques , Humains
10.
ACS Appl Mater Interfaces ; 16(19): 24295-24307, 2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38697643

RÉSUMÉ

Pyroptosis has garnered increasing attention because of its ability to trigger robust antitumor immunity. Pyroptosis is initiated by the activation of inflammasomes, which are regulated by various organelles. The collaboration among organelles offers several protective mechanisms to prevent activation of the inflammasome, thereby limiting the induction of efficient pyroptosis. Herein, a multiorganelle homeostasis disruptor (denoted BLL) is constructed by encapsulating liposomes and bortezomib (BTZ) within a layered double hydroxide (LDH) nanocage to continuously activate inflammasomes for inducing efficient pyroptosis. In lysosomes, the negatively charged liposomes are released to recruit the NLRP3 inflammasomes through electrostatic interactions. ER stress is induced by BTZ to enhance the activation of the NLRP3 inflammasome. Meanwhile, the BLL nanocage exhibited H+-scavenging ability due to the weak alkalinity of LDH, thus disrupting the homeostasis of the lysosome and alleviating the degradation of the NLRP3 inflammasome by lysosomal-associated autophagy. Our results suggest that the BLL nanocage induces homeostatic imbalance in various organelles and efficient pyroptosis. We hope this work can provide new insights into the design of an efficient pyroptosis inducer by disrupting the homeostatic balance of multiple organelles and promote the development of novel antineoplastic platforms.


Sujet(s)
Homéostasie , Inflammasomes , Protéine-3 de la famille des NLR contenant un domaine pyrine , Pyroptose , Pyroptose/effets des médicaments et des substances chimiques , Inflammasomes/métabolisme , Inflammasomes/effets des médicaments et des substances chimiques , Homéostasie/effets des médicaments et des substances chimiques , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Humains , Souris , Bortézomib/pharmacologie , Bortézomib/composition chimique , Liposomes/composition chimique , Animaux , Lysosomes/métabolisme , Lysosomes/effets des médicaments et des substances chimiques , Hydroxydes/composition chimique , Hydroxydes/pharmacologie , Nanostructures/composition chimique , Nanoparticules/composition chimique
11.
Clin Transl Med ; 14(5): e1675, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38689424

RÉSUMÉ

INTRODUCTION: Intrahepatic cholangiocarcinoma (ICC) is characterized by a dismal prognosis with limited therapeutic alternatives. To explore phosphatase and tension homolog (PTEN) as a biomarker for proteasome inhibition in ICC, we conducted a phase II trial to assess the second-line efficacy of bortezomib in PTEN-deficient advanced ICC patients. METHODS: A total of 130 patients with advanced ICC in our centre were screened by PTEN immunohistochemical staining between 1 July 2017, and 31 December 2021, and 16 patients were ultimately enrolled and treated with single-agent bortezomib 1.3 mg/m2 on days 1, 4, 8 and 11 of a 21-day cycle. The primary endpoint was the objective response rate (ORR) according to Response Evaluation Criteria in Solid Tumors v1.1. RESULTS: The median follow-up was 6.55 months (95% confidence interval [CI]: 0.7-19.9 months). Among the 16 enrolled patients, the ORR was 18.75% (3/16) and the disease control rate was 43.75% (7/16). The median progress-free survival was 2.95 months (95% CI: 2.1-5.1 months) and the median overall survival (mOS) was 7.2 months (95% CI: 0.7-21.6 months) in the intent-to-treat-patients. Treatment-related adverse events of any grade were reported in 16 patients, with thrombopenia being the most common toxicity. Patients with PTEN staining scores of 0 were more likely to benefit from bortezomib than those with staining scores > 0. CONCLUSIONS: Bortezomib yielded an encouraging objective response and a favourable OS as a second-line agent in PTEN-deficient ICC patients. Our findings suggest bortezomib as a promising therapeutic option for patients with PTEN-deficient ICC. HIGHLIGHTS: There is a limited strategy for the second-line option of intrahepatic cholangiocarcinoma (ICC). This investigator-initiated phase 2 study evaluated bortezomib in ICC patients with phosphatase and tension homology deficiency. The overall response rate was 18.75% and the overall survival was 7.2 months in the intent-to-treat cohort. These results justify further developing bortezomib in ICC patients with PTEN deficiency.


Sujet(s)
Tumeurs des canaux biliaires , Bortézomib , Cholangiocarcinome , Phosphohydrolase PTEN , Humains , Cholangiocarcinome/traitement médicamenteux , Cholangiocarcinome/génétique , Bortézomib/usage thérapeutique , Bortézomib/pharmacologie , Mâle , Femelle , Adulte d'âge moyen , Sujet âgé , Études prospectives , Phosphohydrolase PTEN/génétique , Phosphohydrolase PTEN/métabolisme , Tumeurs des canaux biliaires/traitement médicamenteux , Tumeurs des canaux biliaires/génétique , Adulte , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/pharmacologie
12.
Cell Commun Signal ; 22(1): 258, 2024 May 06.
Article de Anglais | MEDLINE | ID: mdl-38711131

RÉSUMÉ

Although bortezomib (BTZ) is the cornerstone of anti-multiple myeloma (MM) therapy, the inevitable primary and secondary drug resistance still seriously affects the prognosis of patients. New treatment strategies are in need. Sodium-calcium exchanger 1 (NCX1) is a calcium-permeable ion transporter on the membrane, and our previous studies showed that low NCX1 confers inferior viability in MM cells and suppressed osteoclast differentiation. However, the effect of NCX1 on BTZ sensitivity of MM and its possible mechanism remain unclear. In this study, we investigated the effect of NCX1 on BTZ sensitivity in MM, focusing on cellular processes of autophagy and cell viability. Our results provide evidence that NCX1 expression correlates with MM disease progression and low NCX1 expression increases BTZ sensitivity. NCX1/Ca2+ triggered autophagic flux through non-canonical NFκB pathway in MM cells, leading to attenuated the sensitivity of BTZ. Knockdown or inhibition of NCX1 could potentiate the anti-MM activity of BTZ in vitro and vivo, and inhibition of autophagy sensitized NCX1-overexpressing MM cells to BTZ. In general, this work implicates NCX1 as a potential therapeutic target in MM with BTZ resistance and provides novel mechanistic insights into its vital role in combating BTZ resistance.


Sujet(s)
Autophagie , Bortézomib , Myélome multiple , Échangeur sodium-calcium , Échangeur sodium-calcium/métabolisme , Échangeur sodium-calcium/génétique , Humains , Autophagie/effets des médicaments et des substances chimiques , Animaux , Bortézomib/pharmacologie , Myélome multiple/anatomopathologie , Myélome multiple/métabolisme , Myélome multiple/génétique , Lignée cellulaire tumorale , Souris , Calcium/métabolisme , Résistance aux médicaments antinéoplasiques/génétique , Facteur de transcription NF-kappa B/métabolisme , Survie cellulaire/effets des médicaments et des substances chimiques
13.
JCI Insight ; 9(10)2024 May 07.
Article de Anglais | MEDLINE | ID: mdl-38713510

RÉSUMÉ

Multiple myeloma is a largely incurable and life-threatening malignancy of antibody-secreting plasma cells. An effective and widely available animal model that recapitulates human myeloma and related plasma cell disorders is lacking. We show that busulfan-conditioned human IL-6-transgenic (hIL-6-transgenic) NSG (NSG+hIL6) mice reliably support the engraftment of malignant and premalignant human plasma cells, including from patients diagnosed with monoclonal gammopathy of undetermined significance, pre- and postrelapse myeloma, plasma cell leukemia, and amyloid light chain amyloidosis. Consistent with human disease, NSG+hIL6 mice engrafted with patient-derived myeloma cells developed serum M spikes, and a majority developed anemia, hypercalcemia, and/or bone lesions. Single-cell RNA sequencing showed nonmalignant and malignant cell engraftment, the latter expressing a wide array of mRNAs associated with myeloma cell survival and proliferation. Myeloma-engrafted mice given CAR T cells targeting plasma cells or bortezomib experienced reduced tumor burden. Our results establish NSG+hIL6 mice as an effective patient-derived xenograft model for study and preclinical drug development of multiple myeloma and related plasma cell disorders.


Sujet(s)
Modèles animaux de maladie humaine , Interleukine-6 , Myélome multiple , Animaux , Myélome multiple/immunologie , Myélome multiple/anatomopathologie , Humains , Souris , Interleukine-6/métabolisme , Souris transgéniques , Bortézomib/pharmacologie , Bortézomib/usage thérapeutique , Mâle , Femelle , Plasmocytes/immunologie , Gammapathie monoclonale de signification indéterminée/immunologie , Gammapathie monoclonale de signification indéterminée/anatomopathologie
14.
Int J Mol Sci ; 25(9)2024 Apr 30.
Article de Anglais | MEDLINE | ID: mdl-38732105

RÉSUMÉ

Multiple myeloma is an incurable plasma cell malignancy. Most patients end up relapsing and developing resistance to antineoplastic drugs, like bortezomib. Antibiotic tigecycline has activity against myeloma. This study analyzed tigecycline and bortezomib combination on cell lines and plasma cells from myeloma patients. Apoptosis, autophagic vesicles, mitochondrial mass, mitochondrial superoxide, cell cycle, and hydrogen peroxide were studied by flow cytometry. In addition, mitochondrial antioxidants and electron transport chain complexes were quantified by reverse transcription real-time PCR (RT-qPCR) or western blot. Cell metabolism and mitochondrial activity were characterized by Seahorse and RT-qPCR. We found that the addition of tigecycline to bortezomib reduces apoptosis in proportion to tigecycline concentration. Supporting this, the combination of both drugs counteracts bortezomib in vitro individual effects on the cell cycle, reduces autophagy and mitophagy markers, and reverts bortezomib-induced increase in mitochondrial superoxide. Changes in mitochondrial homeostasis and MYC upregulation may account for some of these findings. These data not only advise to avoid considering tigecycline and bortezomib combination for treating myeloma, but caution on the potential adverse impact of treating infections with this antibiotic in myeloma patients under bortezomib treatment.


Sujet(s)
Apoptose , Bortézomib , Mitochondries , Myélome multiple , Espèces réactives de l'oxygène , Tigecycline , Bortézomib/pharmacologie , Humains , Myélome multiple/traitement médicamenteux , Myélome multiple/métabolisme , Myélome multiple/anatomopathologie , Tigecycline/pharmacologie , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Espèces réactives de l'oxygène/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Antinéoplasiques/pharmacologie , Autophagie/effets des médicaments et des substances chimiques , Mitophagie/effets des médicaments et des substances chimiques , Cycle cellulaire/effets des médicaments et des substances chimiques
15.
Sci Rep ; 14(1): 7733, 2024 04 02.
Article de Anglais | MEDLINE | ID: mdl-38565963

RÉSUMÉ

B-Myb has received considerable attention for its critical tumorigenic function of supporting DNA repair. However, its modulatory effects on chemotherapy and immunotherapy have rarely been reported in colorectal cancer. Bortezomib (BTZ) is a novel compound with chemotherapeutic and immunotherapeutic effects, but it fails to work in colorectal cancer with high B-Myb expression. The present study was designed to investigate whether B-Myb deletion in colorectal cancer could potentiate the immune efficacy of BTZ against colorectal cancer and to clarify the underlying mechanism. Stable B-Myb knockdown was induced in colorectal cancer cells, which increased apoptosis of the cancer cells relative to the control group in vitro and in vivo. We found that BTZ exhibited more favourable efficacy in B-Myb-defective colorectal cancer cells and tumor-bearing mice. BTZ treatment led to differential expression of genes enriched in the p53 signaling pathway promoted more powerful downstream DNA damage, and arrested cell cycle in B-Myb-defective colorectal cancer. In contrast, recovery of B-Myb in B-Myb-defective colorectal cancer cells abated BTZ-related DNA damage, cell cycle arrest, and anticancer efficacy. Moreover, BTZ promoted DNA damage-associated enhancement of immunogenicity, as indicated by potentiated expression of HMGB1 and HSP90 in B-Myb-defective cells, thereby driving M1 polarization of macrophages. Collectively, B-Myb deletion in colorectal cancer facilitates the immunogenic death of cancer cells, thereby further promoting the immune efficacy of BTZ by amplifying DNA damage. The present work provides an effective molecular target for colorectal cancer immunotherapy with BTZ.


Sujet(s)
Antinéoplasiques , Tumeurs colorectales , Animaux , Souris , Bortézomib/pharmacologie , Bortézomib/usage thérapeutique , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Mort cellulaire immunogène , Lignée cellulaire tumorale , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/génétique , Apoptose
16.
Kaohsiung J Med Sci ; 40(6): 542-552, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38682650

RÉSUMÉ

Pulmonary vascular remodeling is a key pathological process of pulmonary arterial hypertension (PAH), characterized by uncontrolled proliferation and migration of pulmonary arterial smooth muscle cells (PASMCs). Bortezomib (BTZ) is the first Food and Drug Administration (FDA)-approved proteasome inhibitor for multiple myeloma treatment. Recently, there is emerging evidence showing its effect on reversing PAH, although its mechanisms are not well understood. In this study, anti-proliferative and anti-migratory effects of BTZ on PASMCs were first examined by different inducers such as fetal bovine serum (FBS), angiotensin II (Ang II) and platelet-derived growth factor (PDGF)-BB, while potential mechanisms including cellular reactive oxygen species (ROS) and mitochondrial ROS were then investigated; finally, signal transduction of ERK and Akt was examined. Our results showed that BTZ attenuated FBS-, Ang II- and PDGF-BB-induced proliferation and migration, with associated decreased cellular ROS production and mitochondrial ROS production. In addition, the phosphorylation of ERK and Akt induced by Ang II and PDGF-BB was also inhibited by BTZ treatment. This study indicates that BTZ can prevent proliferation and migration of PASMCs, which are possibly mediated by decreased ROS production and down-regulation of ERK and Akt. Thus, proteasome inhibition can be a novel pharmacological target in the management of PAH.


Sujet(s)
Bortézomib , Mouvement cellulaire , Prolifération cellulaire , Myocytes du muscle lisse , Inhibiteurs du protéasome , Protéines proto-oncogènes c-akt , Artère pulmonaire , Espèces réactives de l'oxygène , Bortézomib/pharmacologie , Mouvement cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Espèces réactives de l'oxygène/métabolisme , Artère pulmonaire/effets des médicaments et des substances chimiques , Artère pulmonaire/cytologie , Artère pulmonaire/métabolisme , Myocytes du muscle lisse/effets des médicaments et des substances chimiques , Myocytes du muscle lisse/métabolisme , Inhibiteurs du protéasome/pharmacologie , Animaux , Protéines proto-oncogènes c-akt/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Angiotensine-II/pharmacologie , Bécaplermine/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Muscles lisses vasculaires/effets des médicaments et des substances chimiques , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/cytologie , Phosphorylation/effets des médicaments et des substances chimiques , Extracellular Signal-Regulated MAP Kinases/métabolisme
17.
Circ Res ; 134(10): 1276-1291, 2024 May 10.
Article de Anglais | MEDLINE | ID: mdl-38623763

RÉSUMÉ

BACKGROUND: Hypertension is characterized by CD8+ (cluster differentiation 8) T cell activation and infiltration into peripheral tissues. CD8+ T cell activation requires proteasomal processing of antigenic proteins. It has become clear that isoLG (isolevuglandin)-adduced peptides are antigenic in hypertension; however, IsoLGs inhibit the constitutive proteasome. We hypothesized that immunoproteasomal processing of isoLG-adducts is essential for CD8+ T cell activation and inflammation in hypertension. METHODS: IsoLG adduct processing was studied in murine dendritic cells (DCs), endothelial cells (ECs), and B8 fibroblasts. The role of the proteasome and the immunoproteasome in Ang II (angiotensin II)-induced hypertension was studied in C57BL/6 mice treated with bortezomib or the immunoproteasome inhibitor PR-957 and by studying mice lacking 3 critical immunoproteasome subunits (triple knockout mouse). We also examined hypertension in mice lacking the critical immunoproteasome subunit LMP7 (large multifunctional peptidase 7) specifically in either DCs or ECs. RESULTS: We found that oxidant stress increases the presence of isoLG adducts within MHC-I (class I major histocompatibility complex), and immunoproteasome overexpression augments this. Pharmacological or genetic inhibition of the immunoproteasome attenuated hypertension and tissue inflammation. Conditional deletion of LMP7 in either DCs or ECs attenuated hypertension and vascular inflammation. Finally, we defined the role of the innate immune receptors STING (stimulator of interferon genes) and TLR7/8 (toll-like receptor 7/8) as drivers of LMP7 expression in ECs. CONCLUSIONS: These studies define a previously unknown role of the immunoproteasome in DCs and ECs in CD8+ T cell activation. The immunoproteasome in DCs and ECs is critical for isoLG-adduct presentation to CD8+ T cells, and in the endothelium, this guides homing and infiltration of T cells to specific tissues.


Sujet(s)
Bortézomib , Lymphocytes T CD8+ , Cellules dendritiques , Hypertension artérielle , Proteasome endopeptidase complex , Animaux , Mâle , Souris , Angiotensine-II , Bortézomib/pharmacologie , Lymphocytes T CD8+/immunologie , Cellules cultivées , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Cellules endothéliales/métabolisme , Cellules endothéliales/immunologie , Fibroblastes/métabolisme , Antigènes d'histocompatibilité de classe I/métabolisme , Antigènes d'histocompatibilité de classe I/génétique , Hypertension artérielle/métabolisme , Hypertension artérielle/immunologie , Activation des lymphocytes , Souris de lignée C57BL , Souris knockout , Oligopeptides , Stress oxydatif , Proteasome endopeptidase complex/métabolisme , Inhibiteurs du protéasome/pharmacologie
18.
J Cell Mol Med ; 28(8): e18333, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38652192

RÉSUMÉ

Acute myelogenous leukaemia (AML) originates and is maintained by leukaemic stem cells (LSCs) that are inherently resistant to antiproliferative therapies, indicating that a critical strategy for overcoming chemoresistance in AML therapy is to eradicate LSCs. In this work, we investigated the anti-AML activity of bortezomib (BTZ), emphasizing its anti-LSC potential, using KG-1a cells, an AML cell line with stem-like properties. BTZ presented potent cytotoxicity to both solid and haematological malignancy cells and reduced the stem-like features of KG-1a cells, as observed by the reduction in CD34- and CD123-positive cells. A reduction in NF-κB p65 nuclear staining was observed in BTZ-treated KG-1a cells, in addition to upregulation of the NF-κB inhibitor gene NFΚBIB. BTZ-induced DNA fragmentation, nuclear condensation, cell shrinkage and loss of transmembrane mitochondrial potential along with an increase in active caspase-3 and cleaved PARP-(Asp 214) level in KG-1a cells. Furthermore, BTZ-induced cell death was partially prevented by pretreatment with the pancaspase inhibitor Z-VAD-(OMe)-FMK, indicating that BTZ induces caspase-mediated apoptosis. BTZ also increased mitochondrial superoxide levels in KG-1a cells, and BTZ-induced apoptosis was partially prevented by pretreatment with the antioxidant N-acetylcysteine, indicating that BTZ induces oxidative stress-mediated apoptosis in KG-1a cells. At a dosage of 0.1 mg/kg every other day for 2 weeks, BTZ significantly reduced the percentage of hCD45-positive cells in the bone marrow and peripheral blood of NSG mice engrafted with KG-1a cells with tolerable toxicity. Taken together, these data indicate that the anti-LSC potential of BTZ appears to be an important strategy for AML treatment.


Sujet(s)
Bortézomib , Leucémie aigüe myéloïde , Facteur de transcription NF-kappa B , Cellules souches tumorales , Stress oxydatif , Bortézomib/pharmacologie , Stress oxydatif/effets des médicaments et des substances chimiques , Humains , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/anatomopathologie , Leucémie aigüe myéloïde/métabolisme , Animaux , Facteur de transcription NF-kappa B/métabolisme , Lignée cellulaire tumorale , Souris , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Apoptose/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Tests d'activité antitumorale sur modèle de xénogreffe , Souris SCID
19.
Int J Mol Sci ; 25(7)2024 Apr 03.
Article de Anglais | MEDLINE | ID: mdl-38612788

RÉSUMÉ

Proteasome inhibitors are used in the therapy of several cancers, and clinical trials are underway for their use in the treatment of glioblastoma (GBM). However, GBM becomes resistant to chemotherapy relatively rapidly. Recently, the overexpression of ribonucleotide reductase (RNR) genes was found to mediate therapy resistance in GBM. The use of combinations of chemotherapeutic agents is considered a promising direction in cancer therapy. The present work aimed to evaluate the efficacy of the combination of proteasome and RNR inhibitors in yeast and GBM cell models. We have shown that impaired proteasome function results in increased levels of RNR subunits and increased enzyme activity in yeast. Co-administration of the proteasome inhibitor bortezomib and the RNR inhibitor hydroxyurea was found to significantly reduce the growth rate of S. cerevisiae yeast. Accordingly, the combination of bortezomib and another RNR inhibitor gemcitabine reduced the survival of DBTRG-05MG compared to the HEK293 cell line. Thus, yeast can be used as a simple model to evaluate the efficacy of combinations of proteasome and RNR inhibitors.


Sujet(s)
Glioblastome , Saccharomyces cerevisiae , Humains , Proteasome endopeptidase complex , Glioblastome/traitement médicamenteux , Bortézomib/pharmacologie , Cellules HEK293
20.
Leuk Res ; 139: 107469, 2024 04.
Article de Anglais | MEDLINE | ID: mdl-38479337

RÉSUMÉ

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.


Sujet(s)
Myélome multiple , Humains , Animaux , Souris , Bortézomib/pharmacologie , Myélome multiple/traitement médicamenteux , Myélome multiple/génétique , Myélome multiple/métabolisme , Lignée cellulaire tumorale , Récepteurs aux chimiokines , Endoribonucleases , Protein-Serine-Threonine Kinases , Récepteurs CCR1/génétique , Récepteurs CCR1/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...