Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Am J Physiol Cell Physiol ; 327(1): C34-C47, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38646787

ABSTRACT

The dystrophin gene (Dmd) is recognized for its significance in Duchenne muscular dystrophy (DMD), a lethal and progressive skeletal muscle disease. Some patients with DMD and model mice with muscular dystrophy (mdx) spontaneously develop various types of tumors, among which rhabdomyosarcoma (RMS) is the most prominent. By contrast, spindle cell sarcoma (SCS) has rarely been reported in patients or mdx mice. In this study, we aimed to use metabolomics to better understand the rarity of SCS development in mdx mice. Gas chromatography-mass spectrometry was used to compare the metabolic profiles of spontaneously developed SCS and RMS tumors from mdx mice, and metabolite supplementation assays and silencing experiments were used to assess the effects of metabolic differences in SCS tumor-derived cells. The levels of 75 metabolites exhibited differences between RMS and SCS, 25 of which were significantly altered. Further characterization revealed downregulation of nonessential amino acids, including alanine, in SCS tumors. Alanine supplementation enhanced the growth, epithelial mesenchymal transition, and invasion of SCS cells. Reduction of intracellular alanine via knockdown of the alanine transporter Slc1a5 reduced the growth of SCS cells. Lower metabolite secretion and reduced proliferation of SCS tumors may explain the lower detection rate of SCS in mdx mice. Targeting of alanine depletion pathways may have potential as a novel treatment strategy.NEW & NOTEWORTHY To the best of our knowledge, SCS has rarely been identified in patients with DMD or mdx mice. We observed that RMS and SCS tumors that spontaneously developed from mdx mice with the same Dmd genetic background exhibited differences in metabolic secretion. We proposed that, in addition to dystrophin deficiency, the levels of secreted metabolites may play a role in the determination of tumor-type development in a Dmd-deficient background.


Subject(s)
Mice, Inbred mdx , Rhabdomyosarcoma , Sarcoma , Animals , Rhabdomyosarcoma/metabolism , Rhabdomyosarcoma/pathology , Rhabdomyosarcoma/genetics , Mice , Sarcoma/metabolism , Sarcoma/pathology , Sarcoma/genetics , Metabolomics/methods , Cell Line, Tumor , Mice, Inbred C57BL , Disease Models, Animal , Cell Proliferation , Male , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Duchenne/genetics , Epithelial-Mesenchymal Transition , Amino Acid Transport System ASC/metabolism , Amino Acid Transport System ASC/genetics
2.
Histochem Cell Biol ; 161(5): 435-444, 2024 May.
Article in English | MEDLINE | ID: mdl-38396247

ABSTRACT

Rhabdomyosarcoma (RMS) is the most common soft tissue malignancy in children and adolescents. Respecting the age of the patients and the tumor aggressiveness, investigation of the molecular mechanisms of RMS tumorigenesis is directed toward the identification of novel therapeutic targets. To contribute to a better understanding of the molecular pathology of RMS, we investigated ankyrin repeat domain 1 (ANKRD1), designated as a potential marker for differential diagnostics. In this study, we used three RMS cell lines (SJRH30, RD, and HS-729) to assess its expression profile, intracellular localization, and turnover. They express wild-type ANKRD1, as judged by the sequencing of the open reading frame. Each cell line expressed a different amount of ANKRD1 protein, although the transcript level was similar. According to western blot analysis, ANKRD1 protein was expressed at detectable levels in the SJRH30 and RD cells (SJRH30 > RD), but not in the HS-729, even after immunoprecipitation. Immunocytochemistry revealed nuclear and cytoplasmic localization of ANKRD1 in all examined cell lines. Moreover, the punctate pattern of ANKRD1 staining in the nuclei of RD and HS-729 cells overlapped with coilin, indicating its association with Cajal bodies. We have shown that RMS cells are not able to overexpress ANKRD1 protein, which can be attributed to its proteasomal degradation. The unsuccessful attempt to overexpress ANKRD1 in RMS cells indicates the possibility that its overexpression may have detrimental effects for RMS cells and opens a window for further research into its role in RMS pathogenesis and for potential therapeutic targeting.


Subject(s)
Nuclear Proteins , Proteasome Endopeptidase Complex , Repressor Proteins , Rhabdomyosarcoma , Humans , Rhabdomyosarcoma/metabolism , Rhabdomyosarcoma/pathology , Rhabdomyosarcoma/genetics , Proteasome Endopeptidase Complex/metabolism , Repressor Proteins/metabolism , Nuclear Proteins/metabolism , Muscle Proteins/metabolism , Muscle Proteins/analysis , Cell Line, Tumor
3.
BMC Cancer ; 24(1): 79, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38225540

ABSTRACT

BACKGROUND: GEFT is a key regulator of tumorigenesis in rhabdomyosarcoma (RMS), and overexpression of GEFT is significantly correlated with distant metastasis, lymph node metastasis, and a poor prognosis, yet the underlying molecular mechanism is still poorly understood. This study aimed to investigate and validate the molecular mechanism of GEFT-activated lncRNAs in regulating mTOR expression to promote the progression of RMS. METHODS: GEFT-regulated lncRNAs were identified through microarray analysis. The effects of GEFT-regulated lncRNAs on the proliferation, apoptosis, invasion, and migration of RMS cells were confirmed through cell functional experiments. The target miRNAs of GEFT-activated lncRNAs in the regulation of mTOR expression were predicted by bioinformatics analysis combined with quantitative real-time polymerase chain reaction (qRT-PCR) analysis. The expression of lnc-PSMA8-1, miR-144-3p, and mTOR was measured by qRT-PCR in RMS tissue samples and cell lines. The regulatory mechanisms of the lnc-PSMA8-1-miR-144-3p-mTOR signaling axis were verified by RNA-binding protein immunoprecipitation (RIP), a luciferase reporter assay, qRT-PCR analysis, Western blot analysis, and cell functional experiments. RESULTS: The microarray-based analysis identified 31 differentially expressed lncRNAs (fold change > 2.0, P < 0.05). Silencing the 4 upregulated lncRNAs (lnc-CEACAM19-1, lnc-VWCE-2, lnc-GPX7-1, and lnc-PSMA8-1) and overexpressing the downregulated lnc-FAM59A-1 inhibited the proliferation, invasion, and migration and induced the apoptosis of RMS cells. Among the factors analyzed, the expression of lnc-PSMA8-1, miR-144-3p, and mTOR in RMS tissue samples and cells was consistent with the correlations among their expression indicated by the lncRNA-miRNA-mRNA regulatory network based on the ceRNA hypothesis. lnc-PSMA8-1 promoted RMS progression by competitively binding to miR-144-3p to regulate mTOR expression. CONCLUSION: Our research demonstrated that lnc-PSMA8-1 was activated by GEFT and that the former positively regulated mTOR expression by sponging miR-144-3p to promote the progression of RMS. Therefore, targeting this network may constitute a potential therapeutic approach for the management of RMS.


Subject(s)
MicroRNAs , RNA, Long Noncoding , Rhabdomyosarcoma , TOR Serine-Threonine Kinases , Humans , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , MicroRNAs/metabolism , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Up-Regulation
4.
Transgenic Res ; 33(4): 229-241, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38851650

ABSTRACT

Rhabdomyosarcoma (RMS) is a solid tumor whose metastatic progression can be accelerated through interleukin-4 receptor alpha (Il4ra) mediated interaction with normal muscle stem cells (satellite cells). To understand the function of Il4ra in this tumor initiation phase of RMS, we conditionally deleted Il4ra in genetically-engineered RMS mouse models. Nullizygosity of Il4ra altered the latency, site and/or stage distribution of RMS tumors compared to IL4RA intact models. Primary tumor cell cultures taken from the genetically-engineered models then used in orthotopic allografts further defined the interaction of satellite cells and RMS tumor cells in the context of tumor initiation: in alveolar rhabdomyosarcoma (ARMS), satellite cell co-injection was necessary for Il4ra null tumor cells engraftment, whereas in embryonal rhabdomyosarcoma (ERMS), satellite cell co-injection decreased latency of engraftment of Il4ra wildtype tumor cells but not Il4ra null tumor cells. When refocusing on Il4ra wildtype tumors by single cell sequencing and cytokine studies, we have uncovered a putative signaling interplay of Il4 from T-lymphocytes being received by Il4ra + rhabdomyosarcoma tumor cells, which in turn express Ccl2, the ligand for Ccr2 and Ccr5. Taken together, these results suggest that mutations imposed during tumor initiation have different effects than genetic or therapeutic intervention imposed once tumors are already formed. We also propose that CCL2 and its cognate receptors CCR2 and/or CCR5 are potential therapeutic targets in Il4ra mediated RMS progression.


Subject(s)
Interleukin-4 Receptor alpha Subunit , Animals , Mice , Interleukin-4 Receptor alpha Subunit/genetics , Interleukin-4 Receptor alpha Subunit/metabolism , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/pathology , Rhabdomyosarcoma/metabolism , Humans , Satellite Cells, Skeletal Muscle/metabolism , Rhabdomyosarcoma, Alveolar/genetics , Rhabdomyosarcoma, Alveolar/pathology , Rhabdomyosarcoma, Alveolar/metabolism , Disease Models, Animal , Rhabdomyosarcoma, Embryonal/genetics , Rhabdomyosarcoma, Embryonal/pathology , Rhabdomyosarcoma, Embryonal/metabolism , Signal Transduction , Receptors, Cell Surface
5.
PLoS One ; 19(1): e0295629, 2024.
Article in English | MEDLINE | ID: mdl-38277404

ABSTRACT

Targeted therapies for inhibiting the growth of cancer cells or inducing apoptosis are urgently needed for effective rhabdomyosarcoma (RMS) treatment. However, identifying cancer-targeting compounds with few side effects, among the many potential compounds, is expensive and time-consuming. A computational approach to reduce the number of potential candidate drugs can facilitate the discovery of attractive lead compounds. To address this and obtain reliable predictions of novel cell-line-specific drugs, we apply prediction models that have the potential to improve drug discovery approaches for RMS treatment. The results of two prediction models were ensemble and validated via in vitro experiments. The computational models were trained using data extracted from the Genomics of Drug Sensitivity in Cancer database and tested on two RMS cell lines to select potential RMS drug candidates. Among 235 candidate drugs, 22 were selected following the result of the computational approach, and three candidate drugs were identified (NSC207895, vorinostat, and belinostat) that showed selective effectiveness in RMS cell lines in vitro via the induction of apoptosis. Our in vitro experiments have demonstrated that our proposed methods can effectively identify and repurpose drugs for treating RMS.


Subject(s)
Rhabdomyosarcoma , Humans , Cell Line, Tumor , Rhabdomyosarcoma/drug therapy , Rhabdomyosarcoma/metabolism , Apoptosis , Genomics , Treatment Outcome
6.
Nat Commun ; 15(1): 6307, 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39060228

ABSTRACT

Rhabdomyosarcoma (RMS) is a pediatric tumor that resembles undifferentiated muscle cells; yet the extent to which cell state heterogeneity is shared with human development has not been described. Using single-cell/nucleus RNA sequencing from patient tumors, patient-derived xenografts, primary in vitro cultures, and cell lines, we identify four dominant muscle-lineage cell states: progenitor, proliferative, differentiated, and ground cells. We stratify these RMS cells/nuclei along the continuum of human muscle development and show that they share expression patterns with fetal/embryonal myogenic precursors rather than postnatal satellite cells. Fusion-negative RMS (FN-RMS) have a discrete stem cell hierarchy that recapitulates fetal muscle development and contain therapy-resistant FN-RMS progenitors that share transcriptomic similarity with bipotent skeletal mesenchymal cells. Fusion-positive RMS have tumor-acquired cells states, including a neuronal cell state, that are not found in myogenic development. This work identifies previously underappreciated cell state heterogeneity including unique treatment-resistant and tumor-acquired cell states that differ across RMS subtypes.


Subject(s)
Gene Expression Profiling , Rhabdomyosarcoma , Single-Cell Analysis , Transcriptome , Humans , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/pathology , Rhabdomyosarcoma/metabolism , Single-Cell Analysis/methods , Animals , Gene Expression Profiling/methods , Cell Line, Tumor , Mice , Child , Drug Resistance, Neoplasm/genetics , Cell Differentiation , Muscle Development/genetics , Gene Expression Regulation, Neoplastic
7.
Sci Rep ; 14(1): 18936, 2024 08 15.
Article in English | MEDLINE | ID: mdl-39147820

ABSTRACT

Rhabdomyosarcoma (RMS) is the most common childhood soft tissue sarcoma. For the alveolar subtype (ARMS), the presence of the PAX3::FOXO1 fusion gene and/or metastases are strong predictors of poor outcome. Metastatic PAX3::FOXO1+ ARMS often responds to chemotherapies initially, only to subsequently relapse and become resistant with most patients failing to survive beyond 8 years post-diagnosis. No curative intent phase II or phase III clinical trial has been available for patients in the past 10 years (ARST0921). Thus, metastatic ARMS represents a significantly unmet clinical need. Chemotherapy resistance in ARMS has previously been attributed to PAX3::FOXO1-mediated cell cycle checkpoint adaptation, which is mediated by an HDAC3-SMARCA4-miR-27a-PAX3::FOXO1 circuit that can be disrupted by HDAC3 inhibition. In this study, we investigated the therapeutic efficacy of combining the epigenetic regulator entinostat, a Class I Histone Deacetylase (HDAC1-3) inhibitor, with RMS-specific chemotherapies in patient derived xenograft (PDX) models of RMS. We identified single agent, additive or synergistic relationships between relapse-specific chemotherapies and clinically relevant drug exposures of entinostat in three PAX3::FOXO1+ ARMS mouse models. This preclinical data provides further rationale for clinical investigation of entinostat, already known to be well tolerated in a pediatric phase I clinical trial (ADVL1513).


Subject(s)
Benzamides , Pyridines , Rhabdomyosarcoma , Xenograft Model Antitumor Assays , Humans , Pyridines/pharmacology , Pyridines/therapeutic use , Animals , Benzamides/therapeutic use , Benzamides/pharmacology , Mice , Rhabdomyosarcoma/drug therapy , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/pathology , Rhabdomyosarcoma/metabolism , Cell Line, Tumor , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology
8.
J Exp Clin Cancer Res ; 43(1): 135, 2024 May 04.
Article in English | MEDLINE | ID: mdl-38702792

ABSTRACT

BACKGROUND: Rhabdomyosarcoma (RMS) is a rare malignancy and the most common soft tissue sarcoma in children. Vasculogenic mimicry (VM) is a novel tumor microcirculation model different from traditional tumor angiogenesis, which does not rely on endothelial cells to provide sufficient blood supply for tumor growth. In recent years, VM has been confirmed to be closely associated with tumor progression. However, the ability of RMS to form VM has not yet been reported. METHODS: Immunohistochemistry, RT-qPCR and western blot were used to test the expression level of SNAI2 and its clinical significance. The biological function in regulating vasculogenic mimicry and malignant progression of SNAI2 was examined both in vitro and in vivo. Mass spectrometry, co-immunohistochemistry, immunofluorescence staining, and ubiquitin assays were performed to explore the regulatory mechanism of SNAI2. RESULTS: Our study indicated that SNAI2 was abnormally expressed in patients with RMS and RMS cell lines and promoted the proliferation and metastasis of RMS. Through cell tubule formation experiments, nude mice Matrigel plug experiments, and immunohistochemistry (IHC), we confirmed that RMS can form VM and that SNAI2 promotes the formation of VM. Due to SNAI2 is a transcription factor that is not easily drugged, we used Co-IP combined with mass spectrometry to screen for the SNAI2-binding protein USP7 and TRIM21. USP7 depletion inhibited RMS VM formation, proliferation and metastasis by promoting SNAI2 degradation. We further demonstrated that TRIM21 is expressed at low levels in human RMS tissues and inhibits VM in RMS cells. TRIM21 promotes SNAI2 protein degradation through ubiquitination in the RMS. The deubiquitinase USP7 and E3 ligase TRIM21 function in an antagonistic rather than competitive mode and play a key role in controlling the stability of SNAI2 to determine the VM formation and progression of RMS. CONCLUSION: Our findings reveal a previously unknown mechanism by which USP7 and TRIM21 balance the level of SNAI2 ubiquitination, determining RMS vasculogenic mimicry, proliferation, and migration. This new mechanism may provide new targeted therapies to inhibit the development of RMS by restoring TRIM21 expression or inhibiting USP7 expression in RMS patients with high SNAI2 protein levels.


Subject(s)
Neovascularization, Pathologic , Rhabdomyosarcoma , Ribonucleoproteins , Snail Family Transcription Factors , Ubiquitin-Specific Peptidase 7 , Humans , Snail Family Transcription Factors/metabolism , Snail Family Transcription Factors/genetics , Animals , Mice , Ubiquitin-Specific Peptidase 7/metabolism , Ubiquitin-Specific Peptidase 7/genetics , Rhabdomyosarcoma/metabolism , Rhabdomyosarcoma/pathology , Rhabdomyosarcoma/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Female , Disease Progression , Cell Proliferation , Male , Homeostasis , Cell Line, Tumor , Mice, Nude , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination
9.
Chem Biol Interact ; 399: 111149, 2024 Aug 25.
Article in English | MEDLINE | ID: mdl-39032852

ABSTRACT

Rhabdomyosarcoma (RMS) represents one of the most lethal soft-tissue sarcomas in children. The toxic trace element arsenic has been reported to function as a radiosensitizer in sarcomas. To investigate the role of arsenic sulfide (As4S4) in enhancing radiation sensitization in RMS, this study was conducted to elucidate its underlying mechanism in radiotherapy. The combination of As4S4 and radiotherapy showed significant inhibition in RMS cells, as demonstrated by the cell counting kit-8 (CCK-8) assay and flow cytometry. Subsequently, we demonstrated for the first time that As4S4, as well as the knockdown of NFATc3 led to double-strand break (DSB) through increased expression of RAG1. In vivo experiment confirmed that co-treatment efficiently inhibited RMS growth. Furthermore, survival analysis of a clinical cohort consisting of 59 patients revealed a correlation between NFATc3 and RAG1 expression and overall survival (OS). Cox regression analysis also confirmed the independent prognostic significance of NFATc3 and RAG1.Taken together, As4S4 enhances radiosensitivity in RMS via activating NFATc3-RAG1 mediated DSB. NFATc3 and RAG1 are potential therapeutic targets. As4S4 will hopefully serve as a prospective radio-sensitizing agent for RMS.


Subject(s)
Arsenicals , DNA Breaks, Double-Stranded , NFATC Transcription Factors , Radiation Tolerance , Rhabdomyosarcoma , Sulfides , Humans , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Double-Stranded/radiation effects , Sulfides/pharmacology , Sulfides/therapeutic use , Rhabdomyosarcoma/metabolism , Rhabdomyosarcoma/drug therapy , Rhabdomyosarcoma/radiotherapy , Rhabdomyosarcoma/pathology , Rhabdomyosarcoma/genetics , Cell Line, Tumor , Male , Female , Arsenicals/pharmacology , Arsenicals/therapeutic use , Animals , Radiation Tolerance/drug effects , NFATC Transcription Factors/metabolism , Mice , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Mice, Nude , Child , Radiation-Sensitizing Agents/pharmacology , Radiation-Sensitizing Agents/therapeutic use , Mice, Inbred BALB C
10.
Biomed Pharmacother ; 174: 116562, 2024 May.
Article in English | MEDLINE | ID: mdl-38626518

ABSTRACT

Rhabdomyosarcoma (RMS), a mesenchymal tumor occurring in the soft tissue of children, is associated with a defect in differentiation. This study unveils a novel anti-tumor mechanism of dimethylaminomicheliolide (DMAMCL), which is a water-soluble derivative of Micheliolide. First, we demonstrate that DMAMCL inhibits RMS cell growth without obvious cell death, leading to morphological alterations, enhanced expression of muscle differentiation markers, and a shift from a malignant to a more benign metabolic phenotype. Second, we detected decreased expression of DLL1 in RMS cells after DMAMCL treatment, known as a pivotal ligand in the Notch signaling pathway. Downregulation of DLL1 inhibits RMS cell growth and induces morphological changes similar to the effects of DMAMCL. Furthermore, DMAMCL treatment or loss of DLL1 expression also inhibits RMS xenograft tumor growth and augmented the expression of differentiation markers. Surprisingly, in C2C12 cells DMAMCL treatment or DLL1 downregulation also induces cell growth inhibition and an elevation in muscle differentiation marker expression. These data indicated that DMAMCL induced RMS differentiation and DLL1 is an important factor for RMS differentiation, opening a new window for the clinical use of DMAMCL as an agent for differentiation-inducing therapy for RMS treatment.


Subject(s)
Calcium-Binding Proteins , Cell Differentiation , Cell Proliferation , Down-Regulation , Rhabdomyosarcoma , Cell Differentiation/drug effects , Rhabdomyosarcoma/pathology , Rhabdomyosarcoma/drug therapy , Rhabdomyosarcoma/metabolism , Animals , Down-Regulation/drug effects , Humans , Cell Line, Tumor , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/genetics , Cell Proliferation/drug effects , Mice , Xenograft Model Antitumor Assays , Membrane Proteins/metabolism , Membrane Proteins/genetics , Mice, Nude , Intercellular Signaling Peptides and Proteins/metabolism , Signal Transduction/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Antineoplastic Agents/pharmacology
11.
Biomed Pharmacother ; 177: 117126, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38996706

ABSTRACT

BACKGROUND: Rhabdomyosarcoma (RMS) is one of the most common soft tissue sarcomas in children and adolescents, in which PAX3-FOXO1 fusion gene positive patients have very poor prognosis. PAX3-FOXO1 has been identified as an independent prognostic predictor in RMS, with no currently available targeted therapeutic intervention. The novel tyrosine kinase inhibitor anlotinib exhibits a wide range of anticancer effects in multiple types of cancers; however, there have been no relevant studies regarding its application in RMS. MATERIALS AND METHODS: We investigated the effects of PAX3-FOXO1 on the therapeutic efficacy of anlotinib using the CCK-8 assay, flow cytometry, invasion assay, wound healing assay, western blotting, quantitative polymerase chain reaction(qPCR), and xenograft experiments. RNA-seq and co-immunoprecipitation assays were conducted to determine the specific mechanism by which anlotinib regulates PAX3-FOXO1 expression. RESULTS: Anlotinib effectively inhibited RMS cell proliferation and promoted apoptosis and G2/M phase arrest while impeding tumor growth in vivo. Downregulation of PAX3-FOXO1 enhances the antitumor effects of anlotinib. Anlotinib upregulates protein kinase NEK2 and increases the degradation of PAX3-FOXO1 via the ubiquitin-proteasome pathway, leading to a reduction in PAX3-FOXO1 protein levels. CONCLUSION: Anlotinib effectively inhibited the malignant progression of RMS and promoted degradation of the fusion protein PAX3-FOXO1. Anlotinib could be a targeted therapeutic approach to treat PAX3-FOXO1 fusion-positive RMS.


Subject(s)
Apoptosis , Cell Proliferation , Indoles , NIMA-Related Kinases , Oncogene Proteins, Fusion , Quinolines , Rhabdomyosarcoma , Up-Regulation , Humans , Indoles/pharmacology , Indoles/therapeutic use , Animals , Cell Line, Tumor , Rhabdomyosarcoma/drug therapy , Rhabdomyosarcoma/pathology , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/metabolism , Up-Regulation/drug effects , Quinolines/pharmacology , NIMA-Related Kinases/metabolism , NIMA-Related Kinases/genetics , Apoptosis/drug effects , Oncogene Proteins, Fusion/metabolism , Oncogene Proteins, Fusion/genetics , Cell Proliferation/drug effects , Mice, Nude , Xenograft Model Antitumor Assays , Mice , Gene Expression Regulation, Neoplastic/drug effects , Mice, Inbred BALB C , Antineoplastic Agents/pharmacology , G2 Phase Cell Cycle Checkpoints/drug effects , Protein Kinase Inhibitors/pharmacology , Paired Box Transcription Factors
12.
Rev. cuba. obstet. ginecol ; 42(4): 524-529, sep.-dic. 2016. ilus
Article in Spanish | LILACS | ID: biblio-845037

ABSTRACT

El rabdomiosarcoma es un tumor complejo y de gran malignidad que se origina en las células del mesénquima embrionario con capacidad para diferenciarse en células musculares esqueléticas. Este es el tumor maligno de tejido blando más frecuente. Representa aproximadamente 3,5 por ciento de los casos de cáncer en niños de 0 a 14 años de edad. La presentación clínica del rabdomiosarcoma embrionario variedad botrioides es, en general, una masa que protruye por la uretra o el introito vaginal, o por la presencia de flujo fétido o sangrado vaginal en niñas menores de 2 años. Se presenta el caso de un rabdomiosarcoma botrioides de la vagina diagnosticado es una paciente de 16 años y virgen. El apoyo diagnóstico con inmunohistoquímica es de vital importancia y la evaluación médica multidisciplinaria precoz y oportuna permitirá siempre establecer un diagnóstico y tratamiento adecuados que mejoren el pronóstico de quienes padecen esta enfermedad(AU)


Rhabdomyosarcoma is a complex tumor of great malignity that originates in the embryonary mesenchymal cells with the capacity of differentiating into skeletal muscle cells. This is the most frequent malignant tumor in the soft tissue. It roughly represents 3.5 percent of cancer in children aged 0 to 14 years. Generally, the clinical presentation of botryoid-type embryonary rhabdomyosarcoma is a mass that protrudes from the urethra or the vaginal introit or the presence of fetid fluid or vaginal bleeding in girls under 2 years-old. This is the case of a 16 years-old virgin female patient diagnosed with botryoid rhabdomyosarcoma of the vagina. The diagnostic support with immunohistochemistry is of vital importance in addition to the early and timely multidisciplinary medical assessment for setting adequate diagnosis and treatment that improve the prognosis of persons suffering this disease(AU)


Subject(s)
Humans , Female , Adolescent , Physical Examination/methods , Rhabdomyosarcoma/metabolism , Vagina/metabolism , Vaginal Neoplasms/surgery , Rhabdomyosarcoma/diagnosis
SELECTION OF CITATIONS
SEARCH DETAIL