Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
West Afr J Med ; 40(10): 1035-1040, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37906498

RESUMO

BACKGROUND: Renal cell carcinoma is the most lethal urological cancer and contributes significantly to morbidity and mortality due to cancers of the urogenital tract. In routine diagnostic surgical pathology practice of renal tumours, immunohistochemistry is a helpful ancillary technique after routine H & E. The role of renal immunohistochemistry is explored in this study. MATERIALS AND METHODS: The paraffin-embedded tissue blocks of all the confirmed cases of renal cell carcinoma seen at the University College Hospital (UCH), Ibadan, during the 10-year study period of 2007 to 2016 were retrieved, sectioned and immunohistochemistry done using monoclonal antibodies for EMA, Vimentin and CD117 following standard protocols. Frequency statistics and chi-square were applied to data to determine proportions and associations using the Statistical Package for the Social Sciences (SPSS) version 23. RESULTS: A total of 48 cases of renal cell carcinoma were seen within the study period that met the inclusion criteria for the study. The age range of the patients was between 3 to 76 years with an average age of 44.17 years. The male-to-female ratio was 1:1.3. Fuhrman Grade 2 nuclei were predominant (43.75%) while Fuhrman Grade 4 nuclei had the lowest frequency (6.25%). EMAstaining patterns for the different histological patterns of RCC showed no statistically significant difference while Vimentin and CD117 staining patterns showed a statistically significant difference. There was no statistically significant difference observed between the staining patterns of all three markers and the nuclear grades of the cases of RCC. CONCLUSION: This study demonstrated the usefulness of Vimentin and CD117 in differentiating chromophobe variant of renal cell carcinoma from other subtypes while EMA showed variable expression across the various subtypes.


CONTEXTE: Le carcinome à cellules rénales est le cancer urologique le plus mortel et contribue de manière significative à la morbidité et à la mortalité liées aux cancers du tractus urogénital. Dans la pratique courante de la pathologie chirurgicale diagnostique des tumeurs rénales, l'immunohistochimie est une technique auxiliaire utile après la coloration H & E (hématoxyline et éosine). Le rôle de l'immunohistochimie rénale est exploré dans cette étude. MATÉRIEL ET MÉTHODES: Les blocs de tissus inclus en paraffine de tous les cas confirmés de carcinome à cellules rénales observés à l'hôpital universitaire du collège (UCH) d'Ibadan, au cours de la période d'étude de 10 ans de 2007 à 2016, ont été récupérés, sectionnés et soumis à une immunohistochimie en utilisant des anticorps monoclonaux dirigés contre l'EMA, la vimentine et le CD117 suivant des protocoles standard.Des statistiques de fréquence et le test du chi-carré ont été appliqués aux données pour déterminer les proportions et les associations à l'aide du logiciel Statistical Package for the Social Sciences (SPSS) version 23. RÉSULTATS: Au cours de la période d'étude, un total de 48 cas de carcinome à cellules rénales répondant aux critères d'inclusion de l'étude ont été observés. L'âge des patients variait de 3 à 76 ans, avec un âge moyen de 44,17 ans. Le ratio hommes-femmes était de 1:1,3. Les noyaux de grade Fuhrman 2 étaient prédominants (43,75 %), tandis que les noyaux de grade Fuhrman 4 présentaient la fréquence la plus basse (6,25 %). Les schémas de coloration de l'EMA pour les différentes variantes histologiques du RCC n'ont montré aucune différence statistiquement significative, tandis que les schémas de coloration de la vimentine et du CD117 ont montré une différence statistiquement significative. Aucune différence statistiquement significative n'a été observée entre les schémas de coloration des trois marqueurs et les grades nucléaires des cas de RCC. CONCLUSION: Cette étude a démontré l'utilité de la vimentine et du CD117 pour différencier la variante chromophobe du carcinome à cellules rénales des autres sous-types, tandis que l'EMA a montré une expression variable dans les différents sous-types. Mots-clés: Carcinome à cellules rénales (CCR), antigène membranaire épithélial (EMA), vimentine, C-Kit (tyrosine kinase, CD 117), hématoxyline et éosine (H & E).


Assuntos
Carcinoma de Células Renais , Neoplasias Renais , Humanos , Masculino , Feminino , Adulto , Pré-Escolar , Criança , Adolescente , Adulto Jovem , Pessoa de Meia-Idade , Idoso , Carcinoma de Células Renais/diagnóstico , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/patologia , Estudos Retrospectivos , Vimentina/metabolismo , Centros de Atenção Terciária , Biomarcadores Tumorais/metabolismo , Nigéria , Neoplasias Renais/diagnóstico , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia
2.
Proc Natl Acad Sci U S A ; 116(45): 22746-22753, 2019 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-31636198

RESUMO

Gastrointestinal stromal tumors (GISTs) are the most common human sarcoma and are initiated by activating mutations in the KIT or PDGFRA receptor tyrosine kinases. Chromosome 22q deletions are well-recognized frequent abnormalities in GISTs, occurring in ∼50% of GISTs. These deletions are thought to contribute to the pathogenesis of this disease via currently unidentified tumor suppressor mechanisms. Using whole exome sequencing, we report recurrent genomic inactivated DEPDC5 gene mutations in GISTs (16.4%, 9 of 55 patients). The demonstration of clonal DEPDC5 inactivation mutations in longitudinal specimens and in multiple metastases from individual patients suggests that these mutations have tumorigenic roles in GIST progression. DEPDC5 inactivation promotes GIST tumor growth in vitro and in nude mice. DEPDC5 reduces cell proliferation through the mTORC1-signaling pathway and subsequently induces cell-cycle arrest. Furthermore, DEPDC5 modulates the sensitivity of GIST to KIT inhibitors, and the combination therapy with mTOR inhibitor and KIT inhibitor may work better in GIST patients with DEPDC5 inactivation. These findings of recurrent genomic alterations, together with functional data, validate the DEPDC5 as a bona fide tumor suppressor contributing to GIST progression and a biologically relevant target of the frequent chromosome 22q deletions.


Assuntos
Proteínas Ativadoras de GTPase/genética , Neoplasias Gastrointestinais/genética , Tumores do Estroma Gastrointestinal/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Mutação , Animais , Deleção Cromossômica , Cromossomos Humanos Par 22 , Progressão da Doença , Neoplasias Gastrointestinais/patologia , Tumores do Estroma Gastrointestinal/patologia , Xenoenxertos , Humanos , Sequenciamento do Exoma
3.
Proteins ; 88(11): 1434-1446, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32530065

RESUMO

Tyrosine phosphorylation, a highly regulated post-translational modification, is carried out by the enzyme tyrosine kinase (TK). TKs are important mediators in signaling cascades, facilitating diverse biological processes in response to stimuli. TKs may acquire mutations leading to malignancy and are viable targets for anti-cancer drugs. Mast/stem cell growth factor receptor KIT is a TK involved in cell differentiation, whose dysregulation leads to various types of cancer, including gastrointestinal stromal tumors, leukemia, and melanoma. KIT can be targeted by a range of inhibitors that predominantly bind to the inactive state of the enzyme. A mutation Y823D in the activation loop of KIT is known to be responsible for the loss of sensitivity to some drugs in metastatic tumors. We used all-atom molecular dynamics simulations to study the impact of Y823D on the KIT conformation and dynamics and compared it to the effect of phosphorylation of Y823. We simulated in total 6.4 µs of wild-type, mutant and phosphorylated KIT in the active- and inactive-state conformations. We found that Y823D affects the protein dynamics differently: in the active state, the mutation increases the protein stability, whereas in the inactive state it induces local destabilization, thus shifting the dynamic equilibrium towards the active state, altering the communication between distant regulatory regions. The observed dynamics of the Y823D mutant is similar to the dynamics of KIT phosphorylated at position Y823, thus we hypothesize that this mutation mimics a constitutively active kinase, which is not responsive to inhibitors that bind its inactive conformation.


Assuntos
Antineoplásicos/química , Ácido Aspártico/química , Inibidores de Proteínas Quinases/química , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas c-kit/química , Tirosina/química , Antineoplásicos/metabolismo , Ácido Aspártico/metabolismo , Sítios de Ligação , Bases de Dados de Proteínas , Resistencia a Medicamentos Antineoplásicos/genética , Humanos , Ligação de Hidrogênio , Ligantes , Simulação de Dinâmica Molecular , Mutação , Fosforilação , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Inibidores de Proteínas Quinases/metabolismo , Estabilidade Proteica , Proteínas Proto-Oncogênicas c-kit/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-kit/genética , Proteínas Proto-Oncogênicas c-kit/metabolismo , Especificidade por Substrato , Termodinâmica , Tirosina/metabolismo
4.
Int J Mol Sci ; 21(11)2020 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-32498255

RESUMO

In recent years, molecular characterization and management of patients with systemic mastocytosis (SM) have greatly benefited from the application of advanced technologies. Highly sensitive and accurate assays for KIT D816V mutation detection and quantification have allowed the switch to non-invasive peripheral blood testing for patient screening; allele burden has prognostic implications and may be used to monitor therapeutic efficacy. Progress in genetic profiling of KIT, together with the use of next-generation sequencing panels for the characterization of associated gene mutations, have allowed the stratification of patients into three subgroups differing in terms of pathogenesis and prognosis: i) patients with mast cell-restricted KIT D816V; ii) patients with multilineage KIT D816V-involvement; iii) patients with "multi-mutated disease". Thanks to these findings, new prognostic scoring systems combining clinical and molecular data have been developed. Finally, non-genetic SETD2 histone methyltransferase loss of function has recently been identified in advanced SM. Assessment of SETD2 protein levels and activity might provide prognostic information and has opened new research avenues exploring alternative targeted therapeutic strategies. This review discusses how progress in recent years has rapidly complemented previous knowledge improving the molecular characterization of SM, and how this has the potential to impact on patient diagnosis and management.


Assuntos
Alelos , Histona-Lisina N-Metiltransferase/genética , Mastocitose Sistêmica/diagnóstico , Mastocitose Sistêmica/terapia , Mutação , Proteínas Proto-Oncogênicas c-kit/genética , Linhagem da Célula , Citogenética , Variação Genética , Genômica , Sequenciamento de Nucleotídeos em Larga Escala , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Mastócitos/metabolismo , Fenótipo , Prognóstico
5.
Cell Commun Signal ; 17(1): 114, 2019 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-31484543

RESUMO

BACKGROUND: KIT tyrosine kinase is expressed in mast cells, interstitial cells of Cajal, and hematopoietic cells. Permanently active KIT mutations lead these host cells to tumorigenesis, and to such diseases as mast cell leukemia (MCL), gastrointestinal stromal tumor (GIST), and acute myeloid leukemia (AML). Recently, we reported that in MCL, KIT with mutations (D816V, human; D814Y, mouse) traffics to endolysosomes (EL), where it can then initiate oncogenic signaling. On the other hand, KIT mutants including KITD814Y in GIST accumulate on the Golgi, and from there, activate downstream. KIT mutations, such as N822K, have been found in 30% of core binding factor-AML (CBF-AML) patients. However, how the mutants are tyrosine-phosphorylated and where they activate downstream molecules remain unknown. Moreover, it is unclear whether a KIT mutant other than KITD816V in MCL is able to signal on EL. METHODS: We used leukemia cell lines, such as Kasumi-1 (KITN822K, AML), SKNO-1 (KITN822K, AML), and HMC-1.1 (KITV560G, MCL), to explore how KIT transduces signals in these cells and to examine the signal platform for the mutants using immunofluorescence microscopy and inhibition of intracellular trafficking. RESULTS: In AML cell lines, KITN822K aberrantly localizes to EL. After biosynthesis, KIT traffics to the cell surface via the Golgi and immediately migrates to EL through endocytosis in a manner dependent on its kinase activity. However, results of phosphorylation imaging show that KIT is preferentially activated on the Golgi. Indeed, blockade of KITN822K migration to the Golgi with BFA/M-COPA inhibits the activation of KIT downstream molecules, such as AKT, ERK, and STAT5, indicating that KIT signaling occurs on the Golgi. Moreover, lipid rafts in the Golgi play a role in KIT signaling. Interestingly, KITV560G in HMC-1.1 migrates and activates downstream in a similar manner to KITN822K in Kasumi-1. CONCLUSIONS: In AML, KITN822K mislocalizes to EL. Our findings, however, suggest that the mutant transduces phosphorylation signals on lipid rafts of the Golgi in leukemia cells. Unexpectedly, the KITV560G signal platform in MCL is similar to that of KITN822K in AML. These observations provide new insights into the pathogenic role of KIT mutants as well as that of other mutant molecules.


Assuntos
Complexo de Golgi/metabolismo , Leucemia Mieloide Aguda/patologia , Microdomínios da Membrana/metabolismo , Mutação , Proteínas Proto-Oncogênicas c-kit/genética , Proteínas Proto-Oncogênicas c-kit/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/genética , Endocitose/genética , Ativação Enzimática/genética , Humanos , Transporte Proteico/genética
6.
Vet Clin Pathol ; 52(2): 334-340, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36609791

RESUMO

A 10-year-old female Golden Retriever was presented for a recheck after the complete removal of low-grade complex mammary carcinoma. The in-house ProCyte Dx automated counts revealed moderate regenerative anemia and moderate eosinophilia. The ProCyte Dx WBC scattergram showed a cloud in an unusual place parallel and to the right of the monocyte dot plot location. Cells were classified as either monocytes or neutrophils with no clear separation. Complete blood count analysis performed in the laboratory on a Sysmex XT-2000iV analyzer showed moderate regenerative anemia and WBC count within RI; a differential count was not provided by the instrument. On the Sysmex XT-2000iV DIFF scattergram, neutrophil and eosinophil dot plots were present at the respective locations and appeared separated, but the instrument did not provide numerical results. In addition to the normal lymphocyte dot plot location, the second cloud of cells classified as lymphocytes was displayed to the right of the monocyte dot plot area. On the WBC/BASO scattergram, the second population of cells was present above and to the right of the leukocyte cluster. Morphologic assessment of the blood smear detected mastocytemia with 16% poorly granulated and degranulated mast cells. FNAs from the liver and spleen contained large aggregates of poorly granulated mast cells. C-kit somatic mutation screening detected the presence of point mutation S479I in exon 9 of the canine c-KIT gene. This is the first description of abnormal scattergrams from ProCyte Dx and Sysmex XT-2000iV analyzers in a dog with concurrent mastocytemia and systemic mastocytosis, and where cytologic assessments of a blood smear, liver, and spleen, and c-kit somatic mutation analysis were performed.


Assuntos
Doenças do Cão , Mastocitose Sistêmica , Feminino , Cães , Animais , Mastocitose Sistêmica/diagnóstico , Mastocitose Sistêmica/genética , Mastocitose Sistêmica/veterinária , Contagem de Leucócitos/veterinária , Contagem de Células Sanguíneas/veterinária , Leucócitos , Mutação , Doenças do Cão/diagnóstico
7.
Cell Biosci ; 9: 88, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31673329

RESUMO

BACKGROUND: Imatinib shows limited efficacy in patients with gastrointestinal stromal tumors (GISTs) carrying secondary KIT mutations. HQP1351, an orally bioavailable multikinase BCR-ABL inhibitor, is currently in clinical trials for the treatment of T315I mutant chronic myelogenous leukemia (CML), but the potential application in imatinib-resistant GISTs carrying secondary KIT mutations has not been explored. METHODS: The binding activities of HQP1351 with native or mutant KIT were first analyzed. Imatinib-sensitive GIST T1 and imatinib-resistant GIST 430 cells were employed to test the in vitro antiproliferative activity. Colony formation assay, cell migration assay and cell invasion assay were performed to evaluate the clonogenic, migration and invasion ability respectively. Flow cytometry and western blot analysis were used to detect cell apoptosis, cell cycle and signaling pathway. In vivo antitumor activity was evaluated in mouse xenograft models derived from GIST cell lines. RESULTS: HQP1351 potently inhibited both wild-type and mutant KIT kinases. In both imatinib-resistant and sensitive GIST cell lines, HQP1351 exhibited more potent or equivalent antiproliferative activity compared with ponatinib, a third generation BCR-ABL and KIT inhibitor. HQP1351 led to more profound inhibition of cell colony formation, cell migration and invasion, cell cycle arrest and cell apoptosis than ponatinib. Furthermore, HQP1351 also inhibited p-KIT, p-AKT, p-ERK1/2, and p-STAT3 to a higher extent than ponatinib. Finally, in xenograft tumor models derived from imatinib-resistant GIST cancer cell lines, HQP1351 exhibited antitumor activity superior to ponatinib. CONCLUSIONS: Collectively, our in vitro and in vivo results suggest that the therapeutic application of HQP1351 in imatinib-resistant GIST patients deserves further investigation in clinical trials.

8.
Cancer Lett ; 415: 1-10, 2018 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-29196126

RESUMO

Most gastrointestinal stromal tumours (GISTs) are caused by constitutively active mutations in Kit tyrosine kinase. The drug imatinib, a specific Kit inhibitor, improves the prognosis of metastatic GIST patients, but these patients become resistant to the drug by acquiring secondary mutations in the Kit kinase domain. We recently reported that a Kit mutant causes oncogenic signals only on the Golgi apparatus in GISTs. In this study, we show that in GIST, 2-methylcoprophilinamide (M-COPA, also known as "AMF-26"), an inhibitor of biosynthetic protein trafficking from the endoplasmic reticulum (ER) to the Golgi, suppresses Kit autophosphorylation at Y703/Y721/Y730/Y936, resulting in blockade of oncogenic signalling. Results of our M-COPA treatment assay show that Kit Y703/Y730/Y936 in the ER are dephosphorylated by protein tyrosine phosphatases (PTPs), thus the ER-retained Kit is unable to activate downstream molecules. ER-localized Kit Y721 is not phosphorylated, but not due to PTPs. Importantly, M-COPA can inhibit the activation of the Kit kinase domain mutant, resulting in suppression of imatinib-resistant GIST proliferation. Our study demonstrates that Kit autophosphorylation is spatio-temporally regulated and may offer a new strategy for treating imatinib-resistant GISTs.


Assuntos
Complexo de Golgi/metabolismo , Mutação , Naftóis/farmacologia , Proteínas Proto-Oncogênicas c-kit/genética , Piridinas/farmacologia , Linhagem Celular Tumoral , Retículo Endoplasmático/metabolismo , Tumores do Estroma Gastrointestinal/genética , Tumores do Estroma Gastrointestinal/metabolismo , Tumores do Estroma Gastrointestinal/patologia , Humanos , Microscopia Confocal , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Fosforilação/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-kit/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tirosina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA