Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 545
Filtrer
1.
Turk J Med Sci ; 54(4): 804-810, 2024.
Article de Anglais | MEDLINE | ID: mdl-39295619

RÉSUMÉ

Background/aim: Uterine leiomyosarcomas (uLMS) are extremely rare high-grade tumors with a poor prognosis. Their etiopathogenesis remains largely unknown. The uterus is the most frequent site for LMS. uLMS and uterine leiomyoma (uLM) must frequently be differentiated in patients with a uterine mass. Nicotinamide N-methyltransferase (NNMT), a cytoplasmic protein, is involved in the progression and spread of a variety of cancer types. The expression of NNMT in a mesenchymal malignancy was not examined previously. This study represents the first investigation into NNMT expression in uLMS, uLM and benign uterine myometrium and correlates NNMT overexpression with worse prognosis in uLMS. Materials and methods: The expression of NNMT was investigated by immunohistochemistry on formalin-fixed paraffin-embedded tissue of uLMS in 31 patients, uLM in seven patients and benign myometrial in 31 patients. Results: The expression of NNMT in uLMS was markedly higher than in uLM and normal myometrial tissue (p < 0.001). The expression of NNMT in early stage uLMS was lower than in advanced stage disease (p = 0.034). NNMT expression was an independent prognostic factor in predicting recurrence-free survival in uLMS (p = 0.037). Conclusion: NNMT can aid in the preoperative differentiation of uLMS and uLM. The consequences of NNMT overexpression, such as the activation and inactivation of oncoproteins and tumor suppressor proteins, respectively, as well as the enrichment of the cancer stem cell population, overlap with the major mechanisms responsible for poor prognosis in mesenchymal tumors. NNMT may be investigated further in the context of antitumor treatment in patients with mesenchymal malignancies.


Sujet(s)
Léiomyosarcome , Nicotinamide N-methyltransferase , Tumeurs de l'utérus , Humains , Femelle , Léiomyosarcome/métabolisme , Léiomyosarcome/mortalité , Léiomyosarcome/anatomopathologie , Tumeurs de l'utérus/métabolisme , Tumeurs de l'utérus/mortalité , Tumeurs de l'utérus/génétique , Tumeurs de l'utérus/anatomopathologie , Adulte d'âge moyen , Pronostic , Adulte , Nicotinamide N-methyltransferase/métabolisme , Nicotinamide N-methyltransferase/génétique , Marqueurs biologiques tumoraux/métabolisme , Sujet âgé , Léiomyome/métabolisme , Léiomyome/anatomopathologie , Léiomyome/génétique , Immunohistochimie
2.
Cells ; 13(17)2024 Aug 28.
Article de Anglais | MEDLINE | ID: mdl-39273015

RÉSUMÉ

Uterine leiomyosarcoma (uLMS) is the most common type of uterine sarcoma, associated with poor prognosis, high rates of recurrence, and metastasis. Currently, the molecular mechanism of the origin and development of uLMS is limited. Bromodomain and extra-terminal (BET) proteins are involved in both physiological and pathological events. However, the role of BET proteins in the pathogenesis of uLMS is unknown. Here, we show for the first time that BET protein family members, BRD2, BRD3, and BRD4, are aberrantly overexpressed in uLMS tissues compared to the myometrium, with a significant change by histochemical scoring assessment. Furthermore, inhibiting BET proteins with their small, potent inhibitors (JQ1 and I-BET 762) significantly inhibited the uLMS proliferation dose-dependently via cell cycle arrest. Notably, RNA-sequencing analysis revealed that the inhibition of BET proteins with JQ1 and I-BET 762 altered several critical pathways, including the hedgehog pathway, EMT, and transcription factor-driven pathways in uLMS. In addition, the targeted inhibition of BET proteins altered several other epigenetic regulators, including DNA methylases, histone modification, and m6A regulators. The connections between BET proteins and crucial biological pathways provide a fundamental structure to better understand uterine diseases, particularly uLMS pathogenesis. Accordingly, targeting the vulnerable epigenome may provide an additional regulatory mechanism for uterine cancer treatment.


Sujet(s)
Léiomyosarcome , Facteurs de transcription , Tumeurs de l'utérus , Humains , Femelle , Léiomyosarcome/métabolisme , Léiomyosarcome/anatomopathologie , Léiomyosarcome/génétique , Tumeurs de l'utérus/métabolisme , Tumeurs de l'utérus/anatomopathologie , Tumeurs de l'utérus/génétique , Facteurs de transcription/métabolisme , Prolifération cellulaire , Azépines/pharmacologie , Régulation de l'expression des gènes tumoraux , Triazoles/pharmacologie , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/génétique , Lignée cellulaire tumorale , Épigenèse génétique , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Points de contrôle du cycle cellulaire/génétique , Adulte d'âge moyen , Protéines contenant un bromodomaine , Benzodiazépines , Protéines
3.
Georgian Med News ; (350): 42-48, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-39089269

RÉSUMÉ

Most mesenchymal tumors found in the uterine corpus are benign tumors; however, uterine leiomyosarcoma is a malignant tumor with unknown risk factors that repeatedly recurs and metastasizes. In some cases, the histopathologic findings of uterine leiomyoma and uterine leiomyosarcoma are similar and surgical pathological diagnosis using excised tissue samples is difficult. It is necessary to analyze the risk factors for human uterine leiomyosarcoma and establish diagnostic biomarkers and treatments. Female mice deficient in the proteasome subunit low molecular mass peptide 2 (LMP2)/ß1i develop uterine leiomyosarcoma spontaneously. MATERIAL AND METHODS: Out of 334 patients with suspected uterine mesenchymal tumors, patients diagnosed with smooth muscle tumors of the uterus were selected from the pathological file. To investigate the expression status of biomarker candidate factors, immunohistochemical staining was performed with antibodies of biomarker candidate factors on thin-cut slides of human uterine leiomyosarcoma, uterine leiomyoma, and other uterine mesenchymal tumors. RESULTS AND DISCUSSION: In human uterine leiomyosarcoma, there was a loss of LMP2/ß1i expression and enhanced cyclin E1 and Ki-67/MIB1 expression. In human uterine leiomyomas and normal uterine smooth muscle layers, enhanced LMP2/ß1i expression and the disappearance of the expression of E1 and Ki-67/MIB1 were noted. The pattern of expression of each factor in other uterine mesenchymal tumors was different from that of uterine leiomyosarcoma. CONCLUSIONS: LMP2/ß1i, cyclin E1, and Ki-67/MIB1 may be candidate factors for biomarkers of human uterine leiomyosarcoma. Further large-cohort clinical trials should be conducted to establish treatments and diagnostics for uterine mesenchymal tumors.


Sujet(s)
Marqueurs biologiques tumoraux , Cycline E , Léiomyome , Léiomyosarcome , Protéines oncogènes , Tumeurs de l'utérus , Humains , Femelle , Tumeurs de l'utérus/anatomopathologie , Tumeurs de l'utérus/métabolisme , Tumeurs de l'utérus/diagnostic , Tumeurs de l'utérus/génétique , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Léiomyosarcome/génétique , Léiomyosarcome/métabolisme , Léiomyosarcome/anatomopathologie , Léiomyosarcome/diagnostic , Léiomyome/métabolisme , Léiomyome/anatomopathologie , Léiomyome/diagnostic , Léiomyome/génétique , Protéines oncogènes/génétique , Protéines oncogènes/métabolisme , Cycline E/métabolisme , Cycline E/génétique , Proteasome endopeptidase complex/métabolisme , Proteasome endopeptidase complex/génétique , Antigène KI-67/génétique , Antigène KI-67/métabolisme , Adulte d'âge moyen , Adulte , Cysteine endopeptidases
4.
Genomics ; 116(5): 110909, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39103003

RÉSUMÉ

Transposable elements (TEs) are of interest as immunomodulators for cancer therapies. TEs can fold into dsRNAs that trigger the interferon response. Here, we investigated the effect of different HDAC inhibitors (HDACIs) on the expression of TEs in leiomyosarcoma cells. Our data show that endogenous retroviruses (ERVs), especially ERV1 elements, are upregulated after treatment with HDAC1/2/3-specific inhibitors. Surprisingly, the interferon response was not activated. We observed an increase in A-to-I editing of upregulated ERV1. This could have an impact on the stability of dsRNAs and the activation of the interferon response. We also found that H3K27ac levels are increased in the LTR12 subfamilies, which could be regulatory elements controlling the expression of proapoptotic genes such as TNFRSF10B. In summary, we provide a detailed characterization of TEs modulation in response to HDACIs and suggest the use of HDACIs in combination with ADAR inhibitors to induce cell death and support immunotherapy in cancer.


Sujet(s)
Éléments transposables d'ADN , Rétrovirus endogènes , Inhibiteurs de désacétylase d'histone , Interférons , Léiomyosarcome , Régulation positive , Rétrovirus endogènes/génétique , Humains , Inhibiteurs de désacétylase d'histone/pharmacologie , Léiomyosarcome/génétique , Léiomyosarcome/virologie , Léiomyosarcome/traitement médicamenteux , Léiomyosarcome/métabolisme , Lignée cellulaire tumorale , Interférons/génétique , Interférons/métabolisme , Histone/métabolisme
5.
Anticancer Res ; 44(8): 3331-3336, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39060085

RÉSUMÉ

BACKGROUND/AIM: Uterine leiomyosarcomas (uLMS) are the most common mesenchymal tumors of the female genital tract. uLMS genetics encompass complex karyotypes with no specific molecular alterations. The Hippo pathway has been implicated in the pathogenesis of epithelioid hemangio-endotheliomas and endometrial sarcomas. Hippo pathway effectors are YAP1 and TAZ co-transcriptional factors. PATIENTS AND METHODS: We studied Hippo pathway in a series of 32 uLMS patients and its association with clinicopathological parameters. MATERIALS AND METHODS: Immunohistochemical analysis of YAP1 and TAZ proteins accompanied with fluorescent in situ hybridization study of YAP1 gene was performed in patient samples. Age, sex, tumor size, stage at the time of diagnosis and treatment have been analyzed. Overall survival (OS) was calculated from the time of diagnosis until death, loss of follow up or data cut-off. RESULTS: Hippo signaling was found to be dysregulated in 20 (62.5%) patients with uLMS. Regarding OS we detected a trend of Hippo deregulation, designating it as a positive prognostic factor. CONCLUSION: The Hippo pathway is implicated in uLMS oncogenesis, since nuclear expression of YAP1 was detected in 17 (53.1%) of the 32 patients with immunohistochemistry and YAP1 amplification was found in 8 (25%) patients.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Voie de signalisation Hippo , Léiomyosarcome , Protein-Serine-Threonine Kinases , Transduction du signal , Facteurs de transcription , Tumeurs de l'utérus , Protéines de signalisation YAP , Humains , Femelle , Léiomyosarcome/génétique , Léiomyosarcome/anatomopathologie , Léiomyosarcome/métabolisme , Tumeurs de l'utérus/anatomopathologie , Tumeurs de l'utérus/métabolisme , Tumeurs de l'utérus/génétique , Adulte d'âge moyen , Protein-Serine-Threonine Kinases/génétique , Protein-Serine-Threonine Kinases/métabolisme , Protéines de signalisation YAP/métabolisme , Protéines de signalisation YAP/génétique , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Sujet âgé , Protéines adaptatrices de la transduction du signal/génétique , Protéines adaptatrices de la transduction du signal/métabolisme , Adulte , Pronostic , Immunohistochimie , Hybridation fluorescente in situ , Régulation de l'expression des gènes tumoraux , Transcriptional coactivator with PDZ-binding motif proteins
6.
Oncol Rep ; 52(1)2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38874019

RÉSUMÉ

2',3',4'­trihydroxyflavone (2­D08), a SUMO E2 inhibitor, has several biological functions, including anticancer activity, but its effects on uterine leiomyosarcoma (Ut­LMS) are unknown. The anticancer activity of 2­D08 was explored in an in vitro model using SK­LMS­1 and SK­UT­1B cells (human Ut­LMS cells). Treatment with 2­D08 inhibited cell viability in a dose­ and time­dependent manner and significantly inhibited the colony­forming ability of Ut­LMS cells. In SK­UT­1B cells treated with 2­D08, flow cytometric analysis revealed a slight increase in apoptotic rates, while cell cycle progression remained unaffected. Western blotting revealed elevated levels of RIP1, indicating induction of necrosis, but LC3B levels remained unchanged, suggesting no effect on autophagy. A lactate dehydrogenase (LDH) assay confirmed increased LDH release, further supporting the induction of apoptosis and necrosis by 2­D08 in SK­UT­1B cells. 2­D08­induced production of reactive oxygen species and apoptosis progression were observed in SK­LMS­1 cells. Using Ki67 staining and bromodeoxyuridine assays, it was found that 2­D08 suppressed proliferation in SK­LMS­1 cells, while treatment for 48 h led to cell­cycle arrest. 2­D08 upregulated p21 protein expression in SK­LMS­1 cells and promoted apoptosis through caspase­3. Evaluation of α­SM­actin, calponin 1 and TAGLN expression indicated that 2­D08 did not directly initiate smooth muscle phenotypic switching in SK­LMS­1 cells. Transcriptome analysis on 2­D08­treated SK­LMS­1 cells identified significant differences in gene expression and suggested that 2­D08 modulates cell­cycle­ and apoptosis­related pathways. The analysis identified several differentially expressed genes and significant enrichment for biological processes related to DNA replication and molecular functions associated with the apoptotic process. It was concluded that 2­D08 exerts antitumor effects in Ut­LMS cells by modulating multiple signaling pathways and that 2­D08 may be a promising candidate for the treatment of human Ut­LMS. The present study expanded and developed knowledge regarding Ut­LMS management and indicated that 2­D08 represents a notable finding in the exploration of fresh treatment options for such cancerous tumors.


Sujet(s)
Apoptose , Prolifération cellulaire , Léiomyosarcome , Tumeurs de l'utérus , Humains , Léiomyosarcome/traitement médicamenteux , Léiomyosarcome/anatomopathologie , Léiomyosarcome/métabolisme , Femelle , Tumeurs de l'utérus/traitement médicamenteux , Tumeurs de l'utérus/anatomopathologie , Tumeurs de l'utérus/métabolisme , Lignée cellulaire tumorale , Apoptose/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Espèces réactives de l'oxygène/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Flavones/pharmacologie , Antinéoplasiques/pharmacologie , Cycle cellulaire/effets des médicaments et des substances chimiques , Autophagie/effets des médicaments et des substances chimiques
7.
Cell Mol Life Sci ; 81(1): 219, 2024 May 17.
Article de Anglais | MEDLINE | ID: mdl-38758230

RÉSUMÉ

HMGA1 is a structural epigenetic chromatin factor that has been associated with tumor progression and drug resistance. Here, we reported the prognostic/predictive value of HMGA1 for trabectedin in advanced soft-tissue sarcoma (STS) and the effect of inhibiting HMGA1 or the mTOR downstream pathway in trabectedin activity. The prognostic/predictive value of HMGA1 expression was assessed in a cohort of 301 STS patients at mRNA (n = 133) and protein level (n = 272), by HTG EdgeSeq transcriptomics and immunohistochemistry, respectively. The effect of HMGA1 silencing on trabectedin activity and gene expression profiling was measured in leiomyosarcoma cells. The effect of combining mTOR inhibitors with trabectedin was assessed on cell viability in vitro studies, whereas in vivo studies tested the activity of this combination. HMGA1 mRNA and protein expression were significantly associated with worse progression-free survival of trabectedin and worse overall survival in STS. HMGA1 silencing sensitized leiomyosarcoma cells for trabectedin treatment, reducing the spheroid area and increasing cell death. The downregulation of HGMA1 significantly decreased the enrichment of some specific gene sets, including the PI3K/AKT/mTOR pathway. The inhibition of mTOR, sensitized leiomyosarcoma cultures for trabectedin treatment, increasing cell death. In in vivo studies, the combination of rapamycin with trabectedin downregulated HMGA1 expression and stabilized tumor growth of 3-methylcholantrene-induced sarcoma-like models. HMGA1 is an adverse prognostic factor for trabectedin treatment in advanced STS. HMGA1 silencing increases trabectedin efficacy, in part by modulating the mTOR signaling pathway. Trabectedin plus mTOR inhibitors are active in preclinical models of sarcoma, downregulating HMGA1 expression levels and stabilizing tumor growth.


Sujet(s)
Protéine HMGA1a , Sarcomes , Trabectédine , Trabectédine/pharmacologie , Humains , Sarcomes/traitement médicamenteux , Sarcomes/anatomopathologie , Sarcomes/génétique , Sarcomes/métabolisme , Protéine HMGA1a/métabolisme , Protéine HMGA1a/génétique , Animaux , Lignée cellulaire tumorale , Souris , Antinéoplasiques alcoylants/pharmacologie , Antinéoplasiques alcoylants/usage thérapeutique , Résistance aux médicaments antinéoplasiques/génétique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Sérine-thréonine kinases TOR/métabolisme , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Pronostic , Femelle , Léiomyosarcome/traitement médicamenteux , Léiomyosarcome/anatomopathologie , Léiomyosarcome/génétique , Léiomyosarcome/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe
8.
Reprod Biomed Online ; 48(6): 103816, 2024 06.
Article de Anglais | MEDLINE | ID: mdl-38608337

RÉSUMÉ

RESEARCH QUESTION: What is the expression pattern of Raf kinase inhibitory protein (RKIP) in different subtypes of leiomyoma (usual type, cellular, apoplectic or haemorrhagic leiomyoma, leiomyoma with bizarre nuclei and lipoleiomyoma) and leiomyosarcoma specimens, and what is its biological role in leiomyosarcoma cells? DESIGN: Leiomyoma and leiomyosarcoma specimens underwent immunohistochemistry staining. Leiomyosarcoma SK-LMS-1 cell line was RKIP knocked down and RKIP overexpressed, and cell viability, wound healing migration and clonogenicity assays were carried out. RESULTS: A higher immunohistochemical expression of RKIP was observed in bizarre leiomyomas, than in usual-type leiomyomas. Decreased expression was also found in cellular leiomyoma, with generally absent staining in leiomyosarcomas. Upon RKIP expression manipulation in SK-LMS-1 cell line, no major differences were observed in cell viability and migration capacity over time. RKIP knockout, however, resulted in a significant increase in the cell's ability to form colonies (P = 0.011). CONCLUSION: RKIP distinct expression pattern among leiomyoma histotype and leiomyosarcoma, and its effect on leiomyosarcoma cells on colony formation, encourages further studies of RKIP in uterine smooth muscle disorders.


Sujet(s)
Marqueurs biologiques tumoraux , Léiomyome , Léiomyosarcome , Protéine de liaison de phosphatidyl-éthanolamine , Tumeurs de l'utérus , Humains , Léiomyosarcome/métabolisme , Léiomyosarcome/anatomopathologie , Léiomyosarcome/diagnostic , Femelle , Tumeurs de l'utérus/métabolisme , Tumeurs de l'utérus/anatomopathologie , Tumeurs de l'utérus/génétique , Protéine de liaison de phosphatidyl-éthanolamine/métabolisme , Protéine de liaison de phosphatidyl-éthanolamine/génétique , Léiomyome/métabolisme , Léiomyome/anatomopathologie , Léiomyome/diagnostic , Marqueurs biologiques tumoraux/métabolisme , Tumeur du muscle lisse/métabolisme , Tumeur du muscle lisse/anatomopathologie , Tumeur du muscle lisse/diagnostic , Lignée cellulaire tumorale , Adulte d'âge moyen , Mouvement cellulaire , Adulte , Immunohistochimie
9.
Clinics (Sao Paulo) ; 79: 100350, 2024.
Article de Anglais | MEDLINE | ID: mdl-38636197

RÉSUMÉ

OBJECTIVE: The present study aimed to investigate FOXO3a deregulation in Uterine Smooth Muscle Tumors (USMT) and its potential association with cancer development and prognosis. METHODS: The authors analyzed gene and protein expression profiles of FOXO3a in 56 uterine Leiomyosarcomas (LMS), 119 leiomyomas (comprising conventional and unusual leiomyomas), and 20 Myometrium (MM) samples. The authors used techniques such as Immunohistochemistry (IHC), FISH/CISH, and qRT-PCR for the present analyses. Additionally, the authors conducted an in-silico analysis to understand the interaction network involving FOXO3a and its correlated genes. RESULTS: This investigation revealed distinct expression patterns of the FOXO3a gene and protein, including both normal and phosphorylated forms. Expression levels were notably elevated in LMS, and Unusual Leiomyomas (ULM) compared to conventional Leiomyomas (LM) and Myometrium (MM) samples. This upregulation was significantly associated with metastasis and Overall Survival (OS) in LMS patients. Intriguingly, FOXO3a deregulation did not seem to be influenced by EGF/HER-2 signaling, as there were minimal levels of EGF and VEGF expression detected, and HER-2 and EGFR were negative in the analyzed samples. In the examination of miRNAs, the authors observed upregulation of miR-96-5p and miR-155-5p, which are known negative regulators of FOXO3a, in LMS samples. Conversely, the tumor suppressor miR-let7c-5p was downregulated. CONCLUSIONS: In summary, the outcomes of the present study suggest that the imbalance in FOXO3a within Uterine Smooth Muscle Tumors might arise from both protein phosphorylation and miRNA activity. FOXO3a could emerge as a promising therapeutic target for individuals with Unusual Leiomyomas and Leiomyosarcomas (ULM and LMS), offering novel directions for treatment strategies.


Sujet(s)
Protéine O3 à motif en tête de fourche , Léiomyome , Tumeurs de l'utérus , Humains , Femelle , Protéine O3 à motif en tête de fourche/métabolisme , Protéine O3 à motif en tête de fourche/génétique , Tumeurs de l'utérus/génétique , Tumeurs de l'utérus/anatomopathologie , Tumeurs de l'utérus/métabolisme , Adulte d'âge moyen , Léiomyome/génétique , Léiomyome/anatomopathologie , Léiomyome/métabolisme , Adulte , Immunohistochimie , Régulation de l'expression des gènes tumoraux/génétique , Léiomyosarcome/génétique , Léiomyosarcome/anatomopathologie , Léiomyosarcome/métabolisme , Tumeur du muscle lisse/génétique , Tumeur du muscle lisse/anatomopathologie , Tumeur du muscle lisse/métabolisme , Régulation positive , microARN/génétique , microARN/métabolisme , Pronostic , Sujet âgé , Myomètre/métabolisme , Myomètre/anatomopathologie
10.
Biochim Biophys Acta Mol Basis Dis ; 1870(4): 167103, 2024 04.
Article de Anglais | MEDLINE | ID: mdl-38417460

RÉSUMÉ

OBJECTIVE: Uterine leiomyosarcoma (ULMS) is a rare malignant tumor, which is aggressive, and has a poor prognosis even during its early stages. Extracellular vesicles (EVs) carry cargo, such as microRNAs (miRNAs), which are involved in intercellular communication in the tumor microenvironment and other processes. Because there are no studies on EV-related miRNAs in ULMS, we identified EV-related miRNAs in ULMS and examined their function. METHODS: Small EVs (sEVs) and medium/large EVs (m/lEVs) were extracted from ULMS cells by ultracentrifugation and their basic characteristics were evaluated. Then, small RNA sequencing was done to obtain EV-related miRNA profiles. Next, miRNA expression levels in sera and tissues of ULMS patients were compared with those of myoma patients. RESULTS: miR-654-3p and miR-369-3p were indicated to be highly expressed in both sera and tissues of ULMS patients. These two miRNAs are also highly expressed in ULMS cell lines and ULMS-derived EVs. Some cancer-associated fibroblast (CAF) markers were increased when fibroblasts were treated with ULMS-derived EVs. Furthermore, fibroblasts took up EVs derived from ULMS as determined by confocal laser microscopy. In addition, the transfection of the two candidate miRNAs into fibroblasts significantly increased some CAF markers, particularly ACTA2. CONCLUSION: miR-654-3p and miR-369-3p are highly expressed in ULMS-derived EVs, indicating that these EV-related miRNAs induce the formation of cancer-associated fibroblasts.


Sujet(s)
Fibroblastes associés au cancer , Vésicules extracellulaires , Léiomyosarcome , microARN , Humains , Fibroblastes associés au cancer/métabolisme , Léiomyosarcome/génétique , Léiomyosarcome/métabolisme , microARN/génétique , microARN/métabolisme , Vésicules extracellulaires/génétique , Vésicules extracellulaires/métabolisme , Lignée cellulaire , Microenvironnement tumoral/génétique
11.
Georgian Med News ; (343): 119-126, 2023 Oct.
Article de Anglais | MEDLINE | ID: mdl-38096528

RÉSUMÉ

Benign uterine leiomyoma (U.LMA) and malignant uterine leiomyosarcoma (U.LMS), both uterine mesenchymal tumors, are distinguished by the number of cells exhibiting mitotic activity. However, uterine mesenchymal tumors contain tumor cells with various cell morphologies; therefore, making a diagnosis, including differentiating between benign and malignant tumors, is difficult. For example, cotyledonoid dissecting leiomyoma (CDL) or uterine smooth muscle tumors of uncertain malignant potential (STUMPs) are a group of uterine mesenchymal tumors for which a differential diagnosis is challenging. To date, a standardized classification system for uterine mesenchymal tumors has not yet been established. Furthermore, definitive preoperative imaging techniques or hematological examinations for the potential inclusion of CDL or STUMP in the differential diagnosis have not been defined. Several clinical studies have reported that there is no correlation between biomarker expression and mitotic rate or tumor recurrence. The immunohistochemical biomarkers reported so far cannot effectively help determine the malignant potential of CDL or STUMPs in patients who wish to become pregnant in the future. The establishment of gene expression profiles or detection of pathogenic variants by using next-generation molecular techniques can facilitate disease prediction, diagnosis, treatment, and prognosis. We examined the oncological properties of STUMP in adults using molecular pathological techniques on tissue excised from patients with uterine mesenchymal tumor. In a clinical study conducted by our medical team, the results of gene expression profiling indicated factors that may be associated with malignancy of uterine mesenchymal tumors. We herein describe the problems in diagnosing uterine mesenchymal tumors along with the results of the latest clinical studies. It is expected that the establishment of a diagnostic method targeting the characteristics of mesenchymal tumor cells will lead to the treatment of malignant tumors with a low risk of recurrence and metastasis.


Sujet(s)
Léiomyome , Léiomyosarcome , Tumeur du muscle lisse , Tumeurs de l'utérus , Adulte , Femelle , Humains , Léiomyosarcome/diagnostic , Léiomyosarcome/métabolisme , Léiomyosarcome/anatomopathologie , Pronostic , Immunohistochimie , Récidive tumorale locale/diagnostic , Tumeurs de l'utérus/diagnostic , Tumeurs de l'utérus/anatomopathologie , Léiomyome/diagnostic , Marqueurs biologiques tumoraux , Tumeur du muscle lisse/diagnostic , Tumeur du muscle lisse/métabolisme , Tumeur du muscle lisse/anatomopathologie
12.
Clin Cancer Res ; 29(22): 4679-4684, 2023 11 14.
Article de Anglais | MEDLINE | ID: mdl-37699075

RÉSUMÉ

PURPOSE: According to the World Health Organization classification system, uterine leiomyosarcomas (ULMS) are high-grade. A diagnosis of smooth-muscle tumors of uncertain malignant potential (STUMP) is made when Stanford Criteria for ULMS are not met. When a STUMP recurs, the tumor is diagnosed as ULMS and medical treatment is the same as for ULMS. In recent years, some sarcoma centers valued the less aggressive clinical behavior of several recurring STUMP and, given their expression of estrogen and progesterone receptors, started to treat them with hormonal therapy. EXPERIMENTAL DESIGN: This was a retrospective cohort analysis conducted at three referral centers joining the Leiomyosarcoma Foundation Roundtable. We selected all cases of uterine smooth muscle tumors consistent with STUMP and treated with hormonal therapy. RESULTS: 27 consecutive patients were identified. Median age at diagnosis was 43 years. Stage was IA-IB in more than 70% of patients. In these patients, median time to relapse was 62 months. Sites of first relapses were mostly pelvis and peritoneum (76%). After a median follow-up of 49 months, 14 patients (52%) had a partial response while 10 (37%) had a minor response or stable disease. Median time to progression was not reached. CONCLUSIONS: We observed a response or long-term stability rate on hormonal therapy in the 90% range; in all cases the time to relapse was significantly longer than in ULMS and in most cases the relapse was abdominal. On the basis of these findings, we conclude that a proportion of patients with uterine smooth muscle neoplasms actually present with a "low-grade ULMS."


Sujet(s)
Léiomyosarcome , Tumeurs du bassin , Tumeurs de l'utérus , Femelle , Humains , Adulte , Léiomyosarcome/métabolisme , Études rétrospectives , Récidive tumorale locale/traitement médicamenteux , Récidive tumorale locale/anatomopathologie , Tumeurs de l'utérus/diagnostic , Récidive
13.
Nutrients ; 15(11)2023 May 30.
Article de Anglais | MEDLINE | ID: mdl-37299521

RÉSUMÉ

Leiomyosarcoma is an aggressive soft tissue sarcoma derived from the smooth muscle cells of the uterus. We tested the effect of Romina strawberry extract treatment on three-dimensional cultured uterine leiomyosarcoma cells. We established 3D cultures in agarose gel, where the cells seeded were able to form spheroids. We performed the observation and counting of the spheroids with a phase-contrast optical microscope, finding a decrease in the number of spheroids formed in the plates after 24 and 48 h treatment with 250 µg/mL of cultivar Romina strawberry extract. We also characterized the spheroids morphology by DNA binding fluorescent-stain observation, hematoxylin and eosin stain, and Masson's trichrome stain. Finally, the real-time PCR showed a reduced expression of extracellular matrix genes after strawberry treatment. Overall, our data suggest that the fruit extract of this strawberry cultivar may be a useful therapeutic adjuvant for the management of uterine leiomyosarcoma.


Sujet(s)
Fragaria , Léiomyosarcome , Sarcomes , Tumeurs de l'utérus , Humains , Femelle , Léiomyosarcome/traitement médicamenteux , Léiomyosarcome/métabolisme , Fragaria/composition chimique , Tumeurs de l'utérus/traitement médicamenteux , Tumeurs de l'utérus/métabolisme , Myocytes du muscle lisse/métabolisme
14.
Reprod Biomed Online ; 47(1): 15-25, 2023 07.
Article de Anglais | MEDLINE | ID: mdl-37137790

RÉSUMÉ

RESEARCH QUESTION: Is the hypusinated form of the eukaryotic translation initiation factor 5A (EIF5A) present in human myometrium, leiomyoma and leiomyosarcoma, and does it regulate cell proliferation and fibrosis? DESIGN: The hypusination status of eIF5A in myometrial and leiomyoma patient-matched tissues was evaluated by immunohistochemistry and Western blotting as well as in leiomyosarcoma tissues by immunohistochemistry. Myometrial, leiomyoma and leiomyosarcoma cell lines were treated with N1-guanyl-1,7-diaminoheptane (GC-7), responsible for the inhibition of the first step of eIF5A hypunization, and the proliferation rate was determined by MTT assay; fibronectin expression was analysed by Western blotting. Finally, expression of fibronectin in leiomyosarcoma tissues was detected by immunohistochemistry. RESULTS: The hypusinated form of eIF5A was present in all tissues examined, with an increasing trend of hypusinated eIF5A levels from normal myometrium to neoplastic benign leiomyoma up to neoplastic malignant leiomyosarcoma. The higher levels in leiomyoma compared with myometrium were confirmed by Western blotting (P = 0.0046). The inhibition of eIF5A hypusination, with GC-7 treatment at 100 nM, reduced the cell proliferation in myometrium (P = 0.0429), leiomyoma (P = 0.0030) and leiomyosarcoma (P = 0.0044) cell lines and reduced the expression of fibronectin in leiomyoma (P = 0.0077) and leiomyosarcoma (P = 0.0280) cells. The immunohistochemical staining of leiomyosarcoma tissue revealed that fibronectin was highly expressed in the malignant aggressive (central) part of the leiomyosarcoma lesion, where hypusinated eIF5A was also highly represented. CONCLUSIONS: These data support the hypothesis that eIF5A may be involved in the pathogenesis of myometrial benign and malignant pathologies.


Sujet(s)
Léiomyome , Léiomyosarcome , Tumeurs de l'utérus , Femelle , Humains , Fibronectines/métabolisme , Léiomyosarcome/métabolisme , Léiomyosarcome/anatomopathologie , Léiomyome/anatomopathologie , Prolifération cellulaire , Myomètre/métabolisme , Tumeurs de l'utérus/anatomopathologie , Eukaryotic Translation Initiation Factor 5A
15.
J Pharmacol Sci ; 150(4): 259-266, 2022 Dec.
Article de Anglais | MEDLINE | ID: mdl-36344048

RÉSUMÉ

Uterine leiomyosarcoma is an aggressive soft tissue tumor. Stathmin, a phosphoprotein that modulates microtubule dynamics, is highly expressed in many malignancies including leiomyosarcoma. The microtubule-depolymerizing agent eribulin has been recently approved for treating malignant soft tissue tumors. Although eribulin inhibits microtubule polymerization, little is known about the relationship between eribulin treatment and stathmin dynamics. In this study, we explored the role of stathmin expression in the action of eribulin in leiomyosarcoma cells. Eribulin induced phosphorylation of stathmin and reduced expression of subunits A and C of protein phosphatase 2A (PP2A) in a leiomyosarcoma cell line. The PP2A activator FTY720 reduced levels of phosphorylated stathmin. Eribulin decreased stathmin protein levels without affecting stathmin mRNA expression. Furthermore, stathmin knockdown attenuated the inhibitory effects of eribulin on cell viability, whereas stathmin overexpression enhanced the anti-proliferative effect of eribulin. Eribulin-resistant leiomyosarcoma cell lines had enhanced expression of the class Ⅰ ß-tubulin TUBB1, multi-drug resistance 1 protein MDR1 and breast cancer-resistance protein BCRP, and decreased expression of stathmin. Taken together, these results suggest that stathmin expression modulates the pharmacological efficacy of eribulin in uterine leiomyosarcoma cells.


Sujet(s)
Léiomyosarcome , Stathmine , Humains , Stathmine/génétique , Stathmine/métabolisme , Stathmine/pharmacologie , Léiomyosarcome/traitement médicamenteux , Léiomyosarcome/génétique , Léiomyosarcome/métabolisme , Membre-2 de la sous-famille G des transporteurs à cassette liant l'ATP/métabolisme , Protéines tumorales/métabolisme , Microtubules/métabolisme , Microtubules/anatomopathologie
16.
Int J Mol Sci ; 23(11)2022 May 26.
Article de Anglais | MEDLINE | ID: mdl-35682660

RÉSUMÉ

Uterine leiomyosarcoma (uLMS) is a rare and aggressive cancer with few effective therapeutics. The Notch signaling pathway is evolutionarily conserved with oncogenic properties, but it has not been well studied in uLMS. The purpose of our study was to determine expression of Notch family genes and proteins and to investigate the therapeutic effect of γ-secretase inhibitors (GSIs), indirect inhibitors of Notch signaling, in uLMS. We determined expression of Notch genes and proteins in benign uterine smooth muscle tissue, fibroids, and uLMS samples by immunostaining and in two uLMS cell lines, SK-UT-1B (uterine primary) and SK-LMS-1 (vulvar metastasis) by RT-PCR, Western blot and immunostaining. We exposed our cell lines to GSIs, DAPT and MK-0752, and measured expression of HES1, a downstream effector of Notch. Notch proteins were differentially expressed in uLMS. Expression of NOTCH3 and NOTCH4 was higher in uLMS samples than in benign uterine smooth muscle and fibroids. Expression of NOTCH4 was higher in SK-LMS-1 compared to SK-UT-1B. Exposure of SK-UT-1B and SK-LMS-1 to DAPT and MK-0752 decreased expression of HES1 and decreased uLMS cell viability in a dose- and time-dependent manner that was unique to each GSI. Our findings suggest that GSIs are potential therapeutics for uLMS, albeit with limited efficacy.


Sujet(s)
Léiomyome , Léiomyosarcome , Tumeurs du bassin , Tumeurs de l'utérus , Femelle , Inhibiteurs et modulateurs de la gamma-secrétase , Humains , Léiomyosarcome/traitement médicamenteux , Léiomyosarcome/génétique , Léiomyosarcome/métabolisme , Antiagrégants plaquettaires/usage thérapeutique , Récepteurs Notch , Transduction du signal , Tumeurs de l'utérus/anatomopathologie
17.
Clin Cancer Res ; 28(17): 3850-3861, 2022 09 01.
Article de Anglais | MEDLINE | ID: mdl-35727598

RÉSUMÉ

PURPOSE: To evaluate the anticancer effects of cabozantinib, temozolomide, and their combination in uterine sarcoma cell lines and mouse xenograft models. EXPERIMENTAL DESIGN: Human uterine sarcoma cell lines (SK-LMS-1, SK-UT-1, MES-SA, and SKN) were used to evaluate the anticancer activity of cabozantinib, temozolomide, and their combination. The optimal dose of each drug was determined by MTT assay. Cell proliferation and apoptosis were assessed 48 and 72 hours after the drug treatments. The tumor weights were measured in an SK-LMS-1 xenograft mouse model and a patient-derived xenograft (PDX) model of leiomyosarcoma treated with cabozantinib, temozolomide, or both. RESULTS: Given individually, cabozantinib and temozolomide each significantly decreased the growth and viability of cells. This inhibitory effect was more pronounced when cabozantinib (0.50 µmol/L) and temozolomide (0.25 or 0.50 mmol/L) were co-administered (P < 0.05). The combination of the drugs also significantly increased apoptosis in all cells. Moreover, this effect was consistently observed in patient-derived leiomyosarcoma cells. In vivo studies with SK-LMS-1 cell xenografts and the PDX model with leiomyosarcoma demonstrated that combined treatment with cabozantinib (5 mg/kg/d, per os administration) and temozolomide (5 mg/kg/d, per os administration) synergistically decreased tumor growth (both P < 0.05). CONCLUSIONS: The addition of cabozantinib to temozolomide offers synergistic anticancer effects in uterine sarcoma cell lines and xenograft mouse models, including PDX. These results warrant further investigation in a clinical trial.


Sujet(s)
Léiomyosarcome , Sarcomes , Tumeurs des tissus mous , Tumeurs de l'utérus , Anilides , Animaux , Apoptose , Lignée cellulaire tumorale , Femelle , Humains , Léiomyosarcome/traitement médicamenteux , Léiomyosarcome/métabolisme , Souris , Pyridines , Sarcomes/traitement médicamenteux , Sarcomes/anatomopathologie , Témozolomide/pharmacologie , Tumeurs de l'utérus/anatomopathologie , Tests d'activité antitumorale sur modèle de xénogreffe
18.
Clin Cancer Res ; 28(10): 2147-2159, 2022 05 13.
Article de Anglais | MEDLINE | ID: mdl-35302600

RÉSUMÉ

PURPOSE: Uterine leiomyosarcoma is among the most aggressive gynecological malignancies. No effective treatment strategies have been established. This study aimed to identify novel therapeutic targets for uterine leiomyosarcoma based on transcriptome analysis and assess the preclinical efficacy of novel drug candidates. EXPERIMENTAL DESIGN: Transcriptome analysis was performed using fresh-frozen samples of six uterine leiomyosarcomas and three myomas. The Ingenuity Pathway Analysis (IPA) was used to identify potential therapeutic target genes for uterine leiomyosarcoma. Afterward, our results were validated using three independent datasets, including 40 uterine leiomyosarcomas. Then, the inhibitory effects of several selective inhibitors for the candidate genes were examined using SK-UT-1, SK-LMS-1, and SKN cell lines. RESULTS: We identified 512 considerably dysregulated genes in uterine leiomyosarcoma compared with myoma. The IPA revealed that the function of several genes, including CHEK1 and PLK1, were predicted to be activated in uterine leiomyosarcoma. Through an in vitro drug screening, PLK1 or CHEK1 inhibitors (BI-2536 or prexasertib) were found to exert a superior anticancer effect against cell lines at low nanomolar concentrations and induce cell-cycle arrest. In SK-UT-1 tumor-bearing mice, BI-2536 monotherapy remarkably suppressed tumorigenicity. Moreover, the prexasertib and cisplatin combination therapy inhibited tumor proliferation and prolonged the time to tumor progression. CONCLUSIONS: We identified upregulated expressions of PLK1 and CHEK1; their kinase activity was activated in uterine leiomyosarcoma. BI-2536 and prexasertib demonstrated a significant anticancer effect. Therefore, cell-cycle-related kinases may present a promising therapeutic strategy for the treatment of uterine leiomyosarcoma.


Sujet(s)
Protéines du cycle cellulaire , Checkpoint kinase 1 , Léiomyosarcome , Protein-Serine-Threonine Kinases , Protéines proto-oncogènes , Tumeurs de l'utérus , Animaux , Points de contrôle du cycle cellulaire , Protéines du cycle cellulaire/antagonistes et inhibiteurs , Protéines du cycle cellulaire/génétique , Lignée cellulaire tumorale , Checkpoint kinase 1/antagonistes et inhibiteurs , Checkpoint kinase 1/génétique , Cisplatine/usage thérapeutique , Femelle , Humains , Léiomyosarcome/traitement médicamenteux , Léiomyosarcome/génétique , Léiomyosarcome/métabolisme , Souris , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Protein-Serine-Threonine Kinases/génétique , Protéines proto-oncogènes/antagonistes et inhibiteurs , Protéines proto-oncogènes/génétique , Tumeurs de l'utérus/traitement médicamenteux , Tumeurs de l'utérus/génétique , Tumeurs de l'utérus/métabolisme , Polo-Like Kinase 1
19.
Invest New Drugs ; 40(3): 529-536, 2022 06.
Article de Anglais | MEDLINE | ID: mdl-35201535

RÉSUMÉ

BACKGROUND: Uterine leiomyosarcoma is a rare aggressive smooth muscle cancer with poor survival rates. RNA Polymerase I (Pol I) activity is elevated in many cancers supporting tumour growth and prior studies in uterine leiomyosarcoma revealed enlarged nucleoli and upregulated Pol I activity-related genes. This study aimed to investigate the anti-tumour potential of CX-5461, a Pol I transcription inhibitor currently being evaluated in clinical trials for several cancers, against the human uterine leiomyosarcoma cell line, SK-UT-1. METHODS: SK-UT-1 was characterised using genome profiling and western blotting. The anti-tumour effects of CX-5461 were investigated using cell proliferation assays, expression analysis using qRT-PCR, and BrdU/PI based cell cycle analysis. RESULTS: Genetic analysis of SK-UT-1 revealed mutations in TP53, RB1, PTEN, APC and TSC1 & 2, all potentially associated with increased Pol I activity. Protein expression analysis showed dysregulated p53, RB1 and c-Myc. CX-5461 treatment resulted in an anti-proliferation response, G2 phase cell-cycle arrest and on-target activity demonstrated by reduced ribosomal DNA transcription. CONCLUSIONS: SK-UT-1 was confirmed as a representative model of uterine leiomyosarcoma and CX-5461 has significant potential as a novel adjuvant for this rare cancer.


Sujet(s)
Benzothiazoles , Léiomyosarcome , Naphtyridines , Tumeurs de l'utérus , Benzothiazoles/pharmacologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Antienzymes/pharmacologie , Femelle , Humains , Léiomyosarcome/traitement médicamenteux , Léiomyosarcome/génétique , Léiomyosarcome/métabolisme , Naphtyridines/pharmacologie , RNA polymerase I/antagonistes et inhibiteurs , RNA polymerase I/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Tumeurs de l'utérus/traitement médicamenteux , Tumeurs de l'utérus/génétique , Tumeurs de l'utérus/métabolisme
20.
Int J Mol Sci ; 23(4)2022 Feb 16.
Article de Anglais | MEDLINE | ID: mdl-35216305

RÉSUMÉ

The absence of standardized molecular profiling to differentiate uterine leiomyosarcomas versus leiomyomas represents a current diagnostic challenge. In this study, we aimed to search for a differential molecular signature for these myometrial tumors based on artificial intelligence. For this purpose, differential exome and transcriptome-wide research was performed on histologically confirmed leiomyomas (n = 52) and leiomyosarcomas (n = 44) to elucidate differences between and within these two entities. We identified a significantly higher tumor mutation burden in leiomyosarcomas vs. leiomyomas in terms of somatic single-nucleotide variants (171,863 vs. 81,152), indels (9491 vs. 4098), and copy number variants (8390 vs. 5376). Further, we discovered alterations in specific copy number variant regions that affect the expression of some tumor suppressor genes. A transcriptomic analysis revealed 489 differentially expressed genes between these two conditions, as well as structural rearrangements targeting ATRX and RAD51B. These results allowed us to develop a machine learning approach based on 19 differentially expressed genes that differentiate both tumor types with high sensitivity and specificity. Our findings provide a novel molecular signature for the diagnosis of leiomyoma and leiomyosarcoma, which could be helpful to complement the current morphological and immunohistochemical diagnosis and may lay the foundation for the future evaluation of malignancy risk.


Sujet(s)
Léiomyome , Léiomyosarcome , Tumeurs de l'utérus , Intelligence artificielle , Diagnostic différentiel , Femelle , Humains , Léiomyome/diagnostic , Léiomyome/génétique , Léiomyome/métabolisme , Léiomyosarcome/diagnostic , Léiomyosarcome/génétique , Léiomyosarcome/métabolisme , Transcriptome , Tumeurs de l'utérus/diagnostic , Tumeurs de l'utérus/génétique , Tumeurs de l'utérus/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE