Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 743
Filtrar
1.
Prostate ; 84(9): 823-831, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38606933

RESUMO

BACKGROUND: There are limited preclinical orthotopic prostate cancer models due to the technical complexity of surgical engraftment and tracking the tumor growth in the mouse prostate gland. Orthotopic xenografts recapitulate the tumor microenvironment, tumor stromal interactions, and clinical behavior to a greater extent than xenografts grown at subcutaneous or intramuscular sites. METHODS: This study describes a novel micro-surgical technique for orthotopically implanting intact tumors pieces from cell line derived (transgenic adenocarcinoma mouse prostate [TRAMP]-C2) or patient derived (neuroendocrine prostate cancer [NEPC]) tumors in the mouse prostate gland and monitoring tumor growth using magnetic resonance (MR) imaging. RESULTS: The TRAMP-C2 tumors grew rapidly to a predetermined endpoint size of 10 mm within 3 weeks, whereas the NEPC tumors grew at a slower rate over 7 weeks. The tumors were readily detected by MR and confidently identified when they were approximately 2-3 mm in size. The tumors were less well-defined on CT. The TRAMP-C2 tumors were characterized by amorphous sheets of poorly differentiated cells similar to a high-grade prostatic adenocarcinoma and frequent macroscopic peritoneal and lymph node metastases. In contrast, the NEPC's displayed a neuroendocrine morphology with polygonal cells arranged in nests and solid sheets and high count. There was a local invasion of the bladder and other adjacent tissues but no identifiable metastases. The TRAMP-C2 tumors were more hypoxic than the NEPC tumors. CONCLUSIONS: This novel preclinical orthotopic prostate cancer mouse model is suitable for either syngeneic or patient derived tumors and will be effective in developing and advancing the current selection of treatments for patients with prostate cancer.


Assuntos
Adenocarcinoma , Modelos Animais de Doenças , Neoplasias da Próstata , Animais , Masculino , Neoplasias da Próstata/patologia , Neoplasias da Próstata/terapia , Neoplasias da Próstata/diagnóstico por imagem , Camundongos , Humanos , Adenocarcinoma/patologia , Adenocarcinoma/terapia , Linhagem Celular Tumoral , Camundongos Transgênicos , Transplante de Neoplasias/métodos , Imageamento por Ressonância Magnética , Carcinoma Neuroendócrino/patologia , Carcinoma Neuroendócrino/diagnóstico por imagem , Carcinoma Neuroendócrino/terapia
2.
Oxid Med Cell Longev ; 2022: 5885342, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35140839

RESUMO

Acute erythroleukemia (AEL) is an infrequent subtype of acute myeloid leukemia (AML) with worse prognosis. Though the last decade has seen major advances in the novel features and genomic landscape in AEL, there is still a lack of specific therapeutic targets and effective treatment approaches for this disease. Here, we found a novel oncogene KEL that specifically and aberrantly expressed in patients with AEL. In this study, we demonstrated that KEL promoted cell proliferation and the downregulation of KEL reversed drug resistance in AEL cells to JQ1. Our findings suggested that KEL contributed to gain of H3K27 acetylation and promoted erythroid differentiation induced by GATA1. Additionally, GATA1 and TAL1 as cotranscription factors (TFs) modulated the expression of KEL. Maintaining cell viability and differentiation, KEL also played parts in the immune evasion of tumor cells. Our work expands the current knowledge regarding molecular mechanisms involved in cancer onset and progression, offering promising therapeutic target to broaden the treatment options.


Assuntos
Carcinogênese/genética , Progressão da Doença , Leucemia Eritroblástica Aguda/genética , Glicoproteínas de Membrana/genética , Metaloendopeptidases/genética , Oncogenes , Transdução de Sinais/genética , Animais , Estudos de Casos e Controles , Diferenciação Celular/genética , Proliferação de Células/genética , Regulação para Baixo/genética , Fator de Transcrição GATA1/metabolismo , Regulação Leucêmica da Expressão Gênica , Células HEK293 , Humanos , Células K562 , Leucemia Eritroblástica Aguda/metabolismo , Masculino , Glicoproteínas de Membrana/metabolismo , Metaloendopeptidases/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Transplante de Neoplasias/métodos , Transfecção
3.
Cell Death Dis ; 13(1): 10, 2021 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-34930901

RESUMO

As an important regulator of intracellular protein degradation, the mechanism of the deubiquitinating enzyme family in tumour metastasis has received increasing attention. Our previous study revealed that USP3 promotes tumour progression and is highly expressed in gastric cancer (GC). Herein, we report two critical targets, COL9A3 and COL6A5, downstream of USP3, via the isobaric tags for relative and absolute quantification technique. Mechanistically, we observed that USP3 interacted with and stabilised COL9A3 and COL6A5 via deubiquitination in GC. Importantly, we found that COL9A3 and COL6A5 were essential mediators of USP3-modulated oncogenic activity in vitro and in vivo. Examination of clinical samples confirmed that elevated expression of USP3, concomitant with increased COL9A3 and COL6A5 abundance, correlates with human GC progression. These data suggest that USP3 promotes GC progression and metastasis by deubiquitinating COL9A3 and COL6A5. These findings identify a mechanism of GC metastasis regarding USP3-mediated deubiquitinating enzyme activity and suggest potential therapeutic targets for GC management.


Assuntos
Carcinogênese/metabolismo , Colágeno Tipo IX/metabolismo , Colágeno Tipo VI/metabolismo , Neoplasias Pulmonares/secundário , Transdução de Sinais/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Proteases Específicas de Ubiquitina/metabolismo , Ubiquitinação/genética , Animais , Carcinogênese/genética , Linhagem Celular Tumoral , Colágeno Tipo IX/genética , Colágeno Tipo VI/genética , Modelos Animais de Doenças , Xenoenxertos , Humanos , Neoplasias Pulmonares/metabolismo , Camundongos , Camundongos Nus , Transplante de Neoplasias/métodos , Interferência de RNA , Transfecção/métodos , Proteases Específicas de Ubiquitina/genética
4.
Bull Exp Biol Med ; 171(6): 770-773, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34705181

RESUMO

The experimental model of synchronous multiple primary malignant tumors (MPMT) was created. B16/F10 melanoma (0.5 ml of suspension diluted 1:20 in saline) and sarcoma 45 (0.5 million tumor cells in 0.5 ml saline) were simultaneously subcutaneously inoculated to male BALB/c nude mice. In the model of synchronous MPMT, the tumors appeared faster by 2.4 times and had greater volumes: melanoma by 2.2 times and sarcoma by 3.2 times; melanoma metastasized into sarcoma in 71.4% cases; the survival of mice with MPMT was lower. The altered dynamics of malignant growth in the MPMT model is based on the mutual influence of tumors, which results in the exchange of "structural information".


Assuntos
Neoplasias Pulmonares/patologia , Melanoma Experimental/patologia , Transplante de Neoplasias/métodos , Neoplasias Primárias Múltiplas/patologia , Doenças da Imunodeficiência Primária/patologia , Neoplasias Cutâneas/patologia , Animais , Linhagem Celular Tumoral , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/imunologia , Masculino , Melanoma Experimental/genética , Melanoma Experimental/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Primárias Múltiplas/genética , Neoplasias Primárias Múltiplas/imunologia , Doenças da Imunodeficiência Primária/genética , Doenças da Imunodeficiência Primária/imunologia , Ratos , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/imunologia
5.
PLoS One ; 16(9): e0256730, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34495991

RESUMO

INTRODUCTION: Recently, electric cigarettes with liquid (e-liquid) were introduced as an alternative to tobacco smoking. They were promoted as possible cessation aids and were considered to be potentially less harmful than traditional tobacco-based cigarettes. However, there is little information on the toxicants present in e-liquids and their possible carcinogenic effects. METHODS: Western blot analysis was performed to identify the protein levels of cancer progression related signal transducers. Patient-derived brain tumor cells (CSC2) were injected into mouse brains and tumor growth was then observed by performing magnetic resonance imaging (MRI) and hematoxylin and eosin (H&E) staining of the whole brain. Immunohistochemistry (IHC) staining and Immunofluorescence staining were performed to study the expression of pEGFR and pERK. RESULTS: Western blotting revealed that e-liquids increased pEGFR and pERK expression in a dose dependent manner. Animal experiments revealed that the e-liquid treated group had accelerated tumor growth and poor prognosis compared to the vehicle group. Histological staining showed activation of pEGFR and pERK in the e-liquid treated group. CONCLUSION: Our study revealed that e-liquid activates pEGFR and pERK, leading to accelerated brain tumor growth and poor prognosis.


Assuntos
Neoplasias Encefálicas/metabolismo , Carcinogênese/efeitos dos fármacos , Sistemas Eletrônicos de Liberação de Nicotina , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Glioblastoma/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Nicotina/administração & dosagem , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Neoplasias Encefálicas/patologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Fumar Cigarros/metabolismo , Modelos Animais de Doenças , Receptores ErbB/metabolismo , Feminino , Glioblastoma/patologia , Xenoenxertos/efeitos dos fármacos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias/métodos , Fosforilação/efeitos dos fármacos , Prognóstico , Propilenoglicol/administração & dosagem , Soluções , Solventes/administração & dosagem , Carga Tumoral/efeitos dos fármacos
6.
J Immunol ; 207(6): 1662-1671, 2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34417261

RESUMO

The 1858C>T allele of the tyrosine phosphatase PTPN22 (causing amino acid substitution R620W in encoded protein lymphoid tyrosine phosphatase) is present in 5-10% of the North American population and is strongly associated with numerous autoimmune diseases. Although much research has been done to define how this allele potentiates autoimmunity, the influence PTPN22 and its proautoimmune allele have in tumor immunity is poorly defined. To interrogate the role this allele may have in the antitumor immune response, we used CRISPR/Cas9 to generate mice in which the ortholog of lymphoid tyrosine phosphatase, PEST domain-enriched protein (PEP), is mutated at position 619 to produce the relevant proautoimmune mutation (R619W). Results of this study show that mice homozygous for this alteration (PEP-619WW) resist tumor growth as compared with wild-type mice. Consistent with these results, tumors from PEP-619WW mice have more CD45 infiltrates containing more activated CD8 T cells and CD4 T cells. In addition, there are more conventional dendritic cell type 1 (cDC1) cells and fewer myeloid-derived suppressor cells in tumors from PEP-619WW mice. Interestingly, the tumor-infiltrating PEP-619WW cDC1 cells have decreased PD-L1 expression compared with cDC1 cells from PEP-wild-type mice. Taken together, our data show that the proautoimmune allele of Ptpn22 drives a strong antitumor response in innate and adaptive immune cells resulting in superior control of tumors.


Assuntos
Alelos , Carcinoma Pulmonar de Lewis/enzimologia , Carcinoma Pulmonar de Lewis/imunologia , Imunidade , Melanoma Experimental/enzimologia , Melanoma Experimental/imunologia , Proteína Tirosina Fosfatase não Receptora Tipo 22/genética , Neoplasias Cutâneas/enzimologia , Neoplasias Cutâneas/imunologia , Animais , Carcinoma Pulmonar de Lewis/patologia , Linhagem Celular Tumoral , Feminino , Linfócitos do Interstício Tumoral/imunologia , Masculino , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transplante de Neoplasias/métodos , Fenótipo , Neoplasias Cutâneas/patologia
7.
Int J Mol Sci ; 22(14)2021 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-34298975

RESUMO

Previously, we showed that chemotherapy paradoxically exacerbated cancer cell colonization at the secondary site in a manner dependent on Atf3, a stress-inducible gene, in the non-cancer host cells. Here, we present evidence that this phenotype is established at an early stage of colonization within days of cancer cell arrival. Using mouse breast cancer models, we showed that, in the wild-type (WT) lung, cyclophosphamide (CTX) increased the ability of the lung to retain cancer cells in the vascular bed. Although CTX did not change the WT lung to affect cancer cell extravasation or proliferation, it changed the lung macrophage to be pro-cancer, protecting cancer cells from death. This, combined with the initial increase in cell retention, resulted in higher lung colonization in CTX-treated than control-treated mice. In the Atf3 knockout (KO) lung, CTX also increased the ability of lung to retain cancer cells. However, the CTX-treated KO macrophage was highly cytotoxic to cancer cells, resulting in no increase in lung colonization-despite the initial increase in cell retention. In summary, the status of Atf3 dictates the dichotomous activity of macrophage: pro-cancer for CTX-treated WT macrophage but anti-cancer for the KO counterpart. This dichotomy provides a mechanistic explanation for CTX to exacerbate lung colonization in the WT but not Atf3 KO lung.


Assuntos
Fator 3 Ativador da Transcrição/fisiologia , Ciclofosfamida/toxicidade , Neoplasias Pulmonares/secundário , Macrófagos/fisiologia , Neoplasias Mamárias Experimentais/genética , Metástase Neoplásica/fisiopatologia , Proteínas de Neoplasias/fisiologia , Estresse Fisiológico/genética , Macrófagos Associados a Tumor/fisiologia , Animais , Peptídeos Catiônicos Antimicrobianos/biossíntese , Peptídeos Catiônicos Antimicrobianos/genética , Linhagem Celular Tumoral , Ciclofosfamida/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes Reporter , Genótipo , Humanos , Neoplasias Pulmonares/metabolismo , Ativação de Macrófagos , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Camundongos Transgênicos , Terapia Neoadjuvante/efeitos adversos , Metástase Neoplásica/genética , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Transplante de Neoplasias/métodos , Células-Tronco Neoplásicas/patologia , Migração Transendotelial e Transepitelial , Microambiente Tumoral , Macrófagos Associados a Tumor/efeitos dos fármacos , Catelicidinas
8.
JCI Insight ; 6(13)2021 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-34081628

RESUMO

Existing patient-derived xenograft (PDX) mouse models of solid tumors lack a fully tumor donor-matched, syngeneic, and functional immune system. We developed a model that overcomes these limitations by engrafting lymphopenic recipient mice with a fresh, undisrupted piece of solid tumor, whereby tumor-infiltrating lymphocytes (TILs) persisted in the recipient mice for several weeks. Successful tumor engraftment was achieved in 83% to 89% of TIL-PDX mice, and these were seen to harbor exhausted immuno-effector as well as functional immunoregulatory cells persisting for at least 6 months postengraftment. Combined treatment with interleukin-15 stimulation and immune checkpoint inhibition resulted in complete or partial tumor response in this model. Further, depletion of cytotoxic T lymphocytes and/or natural killer cells before combined immunotherapy revealed that both cell types were required for maximal tumor regression. Our TIL-PDX model provides a valuable resource for powerful mechanistic and therapeutic studies in solid tumors.


Assuntos
Xenoenxertos , Imunoterapia/métodos , Células Matadoras Naturais/imunologia , Transplante de Neoplasias , Neoplasias , Linfócitos T Citotóxicos/imunologia , Adjuvantes Imunológicos/farmacologia , Animais , Modelos Animais de Doenças , Xenoenxertos/imunologia , Xenoenxertos/patologia , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Interleucina-15/metabolismo , Camundongos , Transplante de Neoplasias/imunologia , Transplante de Neoplasias/métodos , Neoplasias/imunologia , Neoplasias/terapia , Transplante Heterólogo/métodos , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
9.
Cells ; 10(3)2021 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-33800887

RESUMO

For nearly a decade, researchers in the field of pediatric oncology have been using zebrafish as a model for understanding the contributions of genetic alternations to the pathogenesis of neuroblastoma (NB), and exploring the molecular and cellular mechanisms that underlie neuroblastoma initiation and metastasis. In this review, we will enumerate and illustrate the key advantages of using the zebrafish model in NB research, which allows researchers to: monitor tumor development in real-time; robustly manipulate gene expression (either transiently or stably); rapidly evaluate the cooperative interactions of multiple genetic alterations to disease pathogenesis; and provide a highly efficient and low-cost methodology to screen for effective pharmaceutical interventions (both alone and in combination with one another). This review will then list some of the common challenges of using the zebrafish model and provide strategies for overcoming these difficulties. We have also included visual diagram and figures to illustrate the workflow of cancer model development in zebrafish and provide a summary comparison of commonly used animal models in cancer research, as well as key findings of cooperative contributions between MYCN and diverse singling pathways in NB pathogenesis.


Assuntos
Proteínas de Neoplasias/genética , Neoplasias do Sistema Nervoso/genética , Neuroblastoma/genética , Proteínas Proto-Oncogênicas/genética , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/genética , Animais , Animais Geneticamente Modificados , Antineoplásicos/farmacologia , Modelos Animais de Doenças , Edição de Genes/métodos , Regulação Neoplásica da Expressão Gênica , Ensaios de Triagem em Larga Escala , Humanos , Mutação , Metástase Neoplásica , Proteínas de Neoplasias/metabolismo , Transplante de Neoplasias/métodos , Neoplasias do Sistema Nervoso/tratamento farmacológico , Neoplasias do Sistema Nervoso/metabolismo , Neoplasias do Sistema Nervoso/patologia , Neuroblastoma/tratamento farmacológico , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/metabolismo
10.
Cancer Sci ; 112(5): 1822-1838, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33068050

RESUMO

Biliary tract cancer (BTC) arises from biliary epithelial cells (BECs) and includes intrahepatic cholangiocarcinoma (IHCC), gallbladder cancer (GC), and extrahepatic cholangiocarcinoma (EHCC). Although frequent KRAS mutations and epigenetic changes at the INK4A/ARF locus have been identified, the molecular pathogenesis of BTC is unclear and the development of corresponding anticancer agents remains inadequate. We isolated epithelial cell adhesion molecule (EpCAM)-positive BECs from the mouse intrahepatic bile duct, gallbladder, and extrahepatic bile duct, and established organoids derived from these cells. Introduction of activated KRAS and homozygous deletion of Ink4a/Arf in the cells of each organoid type conferred the ability to form lethal metastatic adenocarcinoma with differentiated components and a pronounced desmoplastic reaction on cell transplantation into syngeneic mice, indicating that the manipulated cells correspond to BTC-initiating cells. The syngeneic mouse models recapitulate the pathological features of human IHCC, GC, and EHCC, and they should therefore prove useful for the investigation of BTC carcinogenesis and the development of new therapeutic strategies. Tumor cells isolated from primary tumors formed organoids in three-dimensional culture, and serial syngeneic transplantation of these cells revealed that their cancer stem cell properties were supported by organoid culture, but not by adherent culture. Adherent culture thus attenuated tumorigenic activity as well as the expression of both epithelial and stem cell markers, whereas the expression of epithelial-mesenchymal transition (EMT)-related transcription factor genes and mesenchymal cell markers was induced. Our data show that organoid culture is important for maintenance of epithelial cell characteristics, stemness, and tumorigenic activity of BTC-initiating cells.


Assuntos
Neoplasias do Sistema Biliar/genética , Colangiocarcinoma/genética , Células Epiteliais/fisiologia , Genes ras , Organoides , Células-Tronco/fisiologia , Fator 1 de Ribosilação do ADP/genética , Adenocarcinoma/genética , Adenocarcinoma/patologia , Animais , Neoplasias dos Ductos Biliares/genética , Neoplasias dos Ductos Biliares/patologia , Ductos Biliares Extra-Hepáticos/anatomia & histologia , Ductos Biliares Extra-Hepáticos/citologia , Ductos Biliares Intra-Hepáticos/citologia , Neoplasias do Sistema Biliar/patologia , Colangiocarcinoma/patologia , Inibidor p16 de Quinase Dependente de Ciclina/genética , Modelos Animais de Doenças , Molécula de Adesão da Célula Epitelial , Células Epiteliais/química , Transição Epitelial-Mesenquimal , Feminino , Vesícula Biliar/anatomia & histologia , Vesícula Biliar/citologia , Neoplasias da Vesícula Biliar/genética , Neoplasias da Vesícula Biliar/patologia , Deleção de Genes , Genes Supressores de Tumor , Fígado/anatomia & histologia , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias/métodos , Organoides/metabolismo , Organoides/patologia , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Análise Serial de Tecidos/métodos , Microambiente Tumoral/fisiologia
11.
J Biomed Mater Res A ; 109(6): 1027-1035, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-32862485

RESUMO

Brain cancer is a devastating disease given its extreme invasiveness and intricate location. Glioblastoma multiforme (GBM) is one of the most common forms of brain cancer, and cancer progression is often correlated with significantly altered tissue stiffness. To elucidate the effect of matrix stiffness on GBM cell fates, previous research is largely limited to 2D studies using immortalized cell lines, which has limited physiological relevance. The objective of the study is to develop gradient hydrogels with brain-mimicking stiffness range as a 3Din vitro GBM model for screening of the effects of matrix stiffness on GBM. To increase the physiological relevance, patient-derived tumor xenograft (PDTX) GBM cells were used. Our gradient platform allows formation of cell-containing hydrogels with stiffness ranging from 40 Pa to 1,300 Pa within a few minutes. By focusing on a brain-mimicking stiffness range, this gradient hydrogel platform is designed for investigating brain cancer. Increasing stiffness led to decreased GBM proliferation and less spreading, which is accompanied by downregulation of matrix-metalloproteinases (MMPs). Using temozolomide (TMZ) as a model drug, we demonstrate that increasing stiffness led to higher drug resistance by PDTX GBM cells in 3D, suggesting matrix stiffness can directly modulate how GBM cells respond to drug treatment. While the current study focuses on stiffness gradient, the setup may also be adapted for screening other cancer niche cues such as how biochemical ligand gradient modulates brain cancer progression and drug responses using reduced materials and time.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Hidrogéis , Transplante de Neoplasias/métodos , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Antineoplásicos Alquilantes/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Progressão da Doença , Xenoenxertos , Humanos , Imageamento Tridimensional , Metaloproteinases da Matriz/metabolismo , Fenômenos Mecânicos , Temozolomida/farmacologia
12.
Neoplasia ; 22(12): 809-819, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33152619

RESUMO

AIM: Evaluation of fibrin role on cancer cells implantation in injured tissues and studying the molecular mechanism of cancer cell interaction with the peritoneal damage. MATERIAL AND METHODS: Mouse colon cancer (CT26) and human mesothelial cells (HMCs) were used. CT26 cells were implanted on injured peritoneal zones. Icodextrin was used as a lubricant. For in vitro studies, fibrin clots from human plasma were used. The cell-fibrin interaction was observed by optical, electronic, and confocal microscopies. Aprotinin was used as a plasmin inhibitor. Hemostasis impact quantified by (1) the fibrin degradation product D-Dimer and PAR expression in HMCs; (2) the expression of plasminogen activator (PA) and its inhibitor (PAI-1) in cancer cells by qPCR and in supernatants through ELISA after in vitro HMC incubation with 2U of thrombin for 24 h. RESULTS: (i) Cancer cell lines were adhered and implanted into the wound area in vivo in both the incision and peeling zones of the peritoneum and on the fibrin network in vitro. (ii) Icodextrin significantly inhibited cancer nodule formation in the scar and the incision or peritoneal damaged zones after surgery. (iii) In in vitro studies, cancer cell interaction with the fibrin clot generated a lysed area, causing an increase in plasmin-dependent fibrinolysis measured by D-dimer levels in the supernatants that was inhibited by aprotinin. (iv) Aprotinin inhibited cell-fibrin interaction and invasion. (v) Thrombin upregulates PAI-1 and downregulates PA expression in HMC. CONCLUSION: Injured tissues favor cancer cell implantation through generated fibrin. Fibrin-cancer cells adhesion can be inhibited by icodextrin.


Assuntos
Cicatriz/metabolismo , Fibrina/metabolismo , Transplante de Neoplasias , Animais , Adesão Celular , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Cicatriz/etiologia , Cicatriz/patologia , Modelos Animais de Doenças , Feminino , Imunofluorescência , Humanos , Camundongos , Transplante de Neoplasias/métodos , Peritônio/metabolismo , Peritônio/patologia
13.
Cell Cycle ; 19(22): 3086-3095, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33073681

RESUMO

Pancreatic cancer (PC) is a leading cause of cancer mortality and is expected to continue increasing incidence. Abnormally expressed microRNAs have been demonstrated tightly correlated with the development and progression of PC. However, the molecular mechanisms remain largely unknown. In this study through combing both the TCGA database and our two verification PC cohorts, we found the consistent reduction of miR-3613-5p in PC tumors, which significantly correlated with reduced cumulative survival rate among PC patients. PC patients with higher lymph node metastasis rate show reduced miR-3613-5p expression. Through further mechanistic investigation, we demonstrate that miR-3613-5p down-regulated CDK6 in repressing the metastasis capacity of PC cells in vitro and in vivo. Elevated CDK6 were also found in PC samples, which also correlate with poor prognosis. Thus, our study found a novel tumor repressor miR-3613-5p in PC progression.


Assuntos
Quinase 6 Dependente de Ciclina/metabolismo , Metástase Linfática/genética , MicroRNAs/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Transdução de Sinais/genética , Animais , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Quinase 6 Dependente de Ciclina/genética , Modelos Animais de Doenças , Regulação para Baixo/genética , Seguimentos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , MicroRNAs/genética , Invasividade Neoplásica/genética , Transplante de Neoplasias/métodos , Neoplasias Pancreáticas/mortalidade , Prognóstico , Taxa de Sobrevida , Transfecção
14.
Cancer Lett ; 494: 27-39, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-32866607

RESUMO

Cells in a tumor are heterogeneous, often including a small number of tumor-initiating cells (TICs) and the majority of cancerous and non-cancerous cells. We have previously reported that constitutive activation of Notch signaling in adipocytes of mice leads to dedifferentiated liposarcoma (DDLPS), an aggressive liposarcoma (LPS) with no effective treatment. Here, we explored the role of Notch signaling in cellular heterogeneity of LPS. We performed serial transplantations to enrich for TICs, and derived cells exhibiting sustained Notch activation (mLPS1 cells) and cells with normal Notch activity (mLPS2 cells). Both mLPS1 and mLPS2 cells proliferated rapidly, and neither exhibited contact inhibition. However, only the mLPS1 cells exhibited tumorigenicity and gave rise to LPS upon engraftment into mice. The mLPS1 cells also highly expressed markers of cancer stem cells (Cd133), mesenchymal stem cells (Cd73, Cd90, Cd105, Dlk1) and the long non-coding RNA Rian. By contrast, the mLPS2 cells accumulated lipid droplets and expressed mature adipocyte markers when induced to differentiate. Most importantly, CRISPR-mediated disruption of Notch abrogated the tumorigenic properties of mLPS1 cells. These results reveal a key role of Notch signaling in maintaining TICs in LPS.


Assuntos
Lipossarcoma/patologia , Transplante de Neoplasias/métodos , Células-Tronco Neoplásicas/metabolismo , Receptores Notch/metabolismo , Animais , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Humanos , Lipossarcoma/genética , Lipossarcoma/metabolismo , Camundongos , Células-Tronco Neoplásicas/patologia , Inoculações Seriadas , Transdução de Sinais
15.
CNS Neurosci Ther ; 26(11): 1168-1177, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32734621

RESUMO

OBJECTIVE: Glioma is a devastating disease lacking effective treatment. Tumor electric field therapy is emerging as a novel non-invasive therapy. The current study evaluates the efficacy and safety of a self-designed tumor electric field therapy system (TEFTS ASCLU-300) in a rat orthotopic transplantation model of glioma. METHODS: A model of intracranial orthotopic transplantation was established in rats using glioma C6 cells. For electric field therapy, glioma-bearing rats were exposed to alternating electric fields generated by a self-developed TEFTS starting on either 1st (Group 2) or 3rd (Group 3) day after transplantation, while other conditions were maintained the same as non-treated rats (Group 1). Glioma size, body weight, and overall survival (OS) were compared between groups. Immunohistochemical staining was applied to access tumor cell death and microvessel density within the tumor. In addition, the systemic effects of TEFTS on blood cells, vital organs, and hepatorenal functions were evaluated. RESULTS: TEFTS treatment significantly elongated the OS of tumor-bearing rats compared with non-treated rats (non-treated vs treated: 24.77 ± 7.08 days vs 40.31 ± 19.11 days, P = .0031). Continuous TEFTS treatment starting on 1st or 3rd day significantly reduced glioma size at 2 and 3 weeks after tumor cell inoculation (Week 2: Group 1:289.95 ± 101.69 mm3 ; Group 2:70.45 ± 17.79 mm3 ; Group 3:73.88 ± 33.21 mm3 , P < .0001. Week 3: Group 1:544.096 ± 78.53 mm3 ; Group 2:187.58 ± 78.44 mm3 ; Group 3:167.14 ± 109.96 mm3 , P = .0005). Continuous treatment for more than 4 weeks inhibited tumor growth. The TEFTS treatment promoted tumor cell death, as demonstrated by increased number of Caspase 3+ cells within the tumor (non-treated vs treated: 38.06 ± 10.04 vs 68.57 ± 8.09 cells/field, P = .0007), but had minimal effect on microvessel density, as shown by CD31 expression (non-treated vs treated: 1.63 ± 0.09 vs 1.57 ± 0.13% of positively stained areas, P > .05). No remarkable differences were observed in hepatorenal function, blood cell counts, or other vital organs between non-treated and treated groups. CONCLUSION: The TEFTS developed by our research team was proved to be effective and safe to inhibit tumor growth and improve general outcomes in a rat model of brain glioma.


Assuntos
Neoplasias Encefálicas/terapia , Terapia por Estimulação Elétrica/métodos , Glioma/terapia , Transplante de Neoplasias/métodos , Carga Tumoral , Animais , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Glioma/patologia , Masculino , Ratos , Ratos Sprague-Dawley
16.
Prostate ; 80(14): 1263-1269, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32761950

RESUMO

BACKGROUND: Liver metastasis is not uncommon in men with metastatic castration-resistant prostate cancer (mCRPC), estimated at ~20% to 60% of advanced late-stage patients. Liver and other visceral metastases are associated with worse overall survival. Recent evidence suggests the frequency of visceral metastases may be increasing for reasons that are unclear but may be related to selective pressures induced by modern therapies, including second-generation antiandrogen receptor signaling inhibitors such as enzalutamide and abiraterone. Consequently, robust models to study the pathobiology of prostate cancer liver metastases and their response to therapy are urgently needed. METHODS: Hemi-spleen injection of human (LN95, PC3, VCaP, and MDA-PCa-2b) or syngeneic (Myc-CaP) prostate cancer cells (1 × 106 ) was performed to seed liver metastases via the splenic vessels. Plasma levels of prostate-specific antigen (PSA) were monitored longitudinally in human androgen receptor-positive (AR+) models. Immunohistochemical staining of AR and HoxB13 was performed to document the prostatic origin of hepatic lesions. RESULTS: LN95, PC3, and Myc-CaP produced distinct liver micrometastases that progressed to macrometastases by ~2 to 4 weeks postinoculation, while inoculation of MDA-PCa-2b and VCaP only produced occasional micrometastases and seeding of individual cells adjacent to blood vessels, respectively, at the time points analyzed. All lesions are characterized by positive staining for nuclear AR and/or the prostate-specific differentiation marker HoxB13 depending on the model. Circulating PSA levels are strongly correlated with overall tumor burden in mice seeded with LN95. Histologic micrometastases and low levels of circulating PSA are detected in mice seeded with MDA-PCa-2b at ~60 days postinoculation, but no circulating PSA was detected in animals inoculated with VCaP up to ~75 days despite the presence of rare AR+ cells in the liver. CONCLUSION: The studies reported herein establish intrasplenic injection as a robust model of mCRPC liver metastasis. In addition, circulating PSA was validated as a noninvasive biomarker to longitudinally monitor overall tumor burden when using PSA+ models. Therefore, this model can be used to interrogate the pathophysiology of prostate cancer liver metastases, the microenvironmental factors permissive to such growth, immunologic variables, and the response of hepatic lesions to therapy.


Assuntos
Neoplasias Hepáticas/secundário , Neoplasias de Próstata Resistentes à Castração/patologia , Baço/patologia , Animais , Linhagem Celular Tumoral , Xenoenxertos , Humanos , Calicreínas/sangue , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Transplante de Neoplasias/métodos , Antígeno Prostático Específico/sangue , Neoplasias de Próstata Resistentes à Castração/sangue , Transplante Heterólogo
17.
Int J Radiat Oncol Biol Phys ; 108(3): 745-757, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32470502

RESUMO

PURPOSE: Improved efficacy of anticancer therapy and a growing pool of survivors give rise to a question about their quality of life and return to premorbid status. Radiation is effective in brain metastasis eradication, although the optimal approach and long-term effects on brain function are largely unknown. We studied the effects of radiosurgery on brain function. METHODS AND MATERIALS: Adult C57BL/6J mice with or without brain metastases (rat 9L gliosarcoma) were treated with cone beam single-arc stereotactic radiosurgery (SRS; 40 Gy). Tumor growth was monitored using bioluminescence, whereas longitudinal magnetic resonance imaging, behavioral studies, and histologic analysis were performed to evaluate brain response to the treatment for up to 18 months. RESULTS: Stereotactic radiosurgery (SRS) resulted in 9L metastases eradication within 4 weeks with subsequent long-term survival of all treated animals, whereas all nontreated animals succumbed to the brain tumor. Behavioral impairment, as measured with a recognition memory test, was observed earlier in mice subjected to radiosurgery of tumors (6 weeks) in comparison to SRS of healthy brain tissue (10 weeks). Notably, the deficit resolved by 18 weeks only in mice not bearing a tumor, whereas tumor eradication was complicated by the persistent cognitive deficits. In addition, the results of magnetic resonance imaging were unremarkable in both groups, and histopathology revealed changes. SRS-induced tumor eradication triggered long-lasting and exacerbated neuroinflammatory response. No demyelination, neuronal loss, or hemorrhage was detected in any of the groups. CONCLUSIONS: Tumor disintegration by SRS leads to exacerbated neuroinflammation and persistent cognitive deficits; therefore, methods aiming at reducing inflammation after tumor eradication or other therapeutic methods should be sought.


Assuntos
Neoplasias Encefálicas/radioterapia , Encéfalo/efeitos da radiação , Disfunção Cognitiva/etiologia , Gliossarcoma/radioterapia , Radiocirurgia/efeitos adversos , Animais , Atenção/efeitos da radiação , Comportamento , Encéfalo/diagnóstico por imagem , Encéfalo/patologia , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/secundário , Disfunção Cognitiva/diagnóstico por imagem , Encefalite/diagnóstico por imagem , Encefalite/etiologia , Encefalite/patologia , Gliossarcoma/mortalidade , Gliossarcoma/patologia , Gliossarcoma/secundário , Gliose/etiologia , Medições Luminescentes , Ativação de Macrófagos , Imageamento por Ressonância Magnética/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias/métodos , Radiocirurgia/métodos , Dosagem Radioterapêutica , Reconhecimento Psicológico
18.
Updates Surg ; 72(4): 951-966, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32253687

RESUMO

Patient-Derived Xenografts (PDXs) are, so far, the best preclinical model to validate targets and predictors of response to therapy. While subcutaneous implantation very rarely allows metastatic dissemination, orthotopic implantation (Patient-Derived Orthotopic Xenograft-PDOX) increases metastatic capability. Using a modified tool to analyze model validity, we performed a systematic review of Embase, PubMed, and Web of Science up to December 2018 to identify all original publications describing gastric cancer (GC) PDOXs. We identified ten studies of PDOX model validation from January 1981 to December 2018 that fulfilled the inclusion and exclusion criteria. Most models (70%) were derived from human GC cell lines rather than tissue fragments. In 90% of studies, the implantation was performed in the subserosal layer. Tumour engraftment rate ranged from 0 to 100%, despite the technique. Metastases were observed in 40% of PDOX models implanted into the subserosal layer, employing either cell suspension or cell line-derived tumour fragments. According to our modified model validity tool, half of the studies were defined as unclear because one or more validation criteria were not reported. Available GC PDOX models are not adequate according to our model validity tool. There is no demonstration that the submucosal site is more effective than the subserosal layer, and that tissue fragments are better than cell suspensions for successful engraftment and metastatic spread. Further studies should strictly employ model validity tools and large samples with orthotopic implant sites mirroring as much as possible the donor tumour characteristics.


Assuntos
Modelos Animais de Doenças , Metástase Neoplásica/patologia , Transplante de Neoplasias/métodos , Neoplasias Gástricas , Animais , Linhagem Celular Tumoral , Humanos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Nus , Neoplasias Gástricas/patologia
19.
Cancer Res ; 80(11): 2286-2297, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32152150

RESUMO

Patient-derived xenografts (PDX) are tumor-in-mouse models for cancer. PDX collections, such as the NCI PDXNet, are powerful resources for preclinical therapeutic testing. However, variations in experimental and analysis procedures have limited interpretability. To determine the robustness of PDX studies, the PDXNet tested temozolomide drug response for three prevalidated PDX models (sensitive, resistant, and intermediate) across four blinded PDX Development and Trial Centers using independently selected standard operating procedures. Each PDTC was able to correctly identify the sensitive, resistant, and intermediate models, and statistical evaluations were concordant across all groups. We also developed and benchmarked optimized PDX informatics pipelines, and these yielded robust assessments across xenograft biological replicates. These studies show that PDX drug responses and sequence results are reproducible across diverse experimental protocols. In addition, we share the range of experimental procedures that maintained robustness, as well as standardized cloud-based workflows for PDX exome-sequencing and RNA-sequencing analyses and for evaluating growth. SIGNIFICANCE: The PDXNet Consortium shows that PDX drug responses and sequencing results are reproducible across diverse experimental protocols, establishing the potential for multisite preclinical studies to translate into clinical trials.


Assuntos
Transplante de Neoplasias/normas , Medicina de Precisão/métodos , Medicina de Precisão/normas , Transplante Heterólogo/normas , Ensaios Antitumorais Modelo de Xenoenxerto/normas , Animais , Humanos , Camundongos , Transplante de Neoplasias/métodos , Distribuição Aleatória , Transplante Heterólogo/métodos , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
20.
Histol Histopathol ; 35(8): 871-886, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32022242

RESUMO

Glioblastoma multiforme (GBM) is an aggressive primary brain malignancy with a very poor prognosis. Researchers employ animal models to develop potential therapies. It is important that these models have clinical relevance. This means that old models, propagated for decades in cultures, should be questioned. Parameters to be evaluated include whether animals are immune competent or not, the infiltrative growth pattern of the tumor, tumor volume resulting in symptoms and growth rate. We here describe the growth pattern of an experimental glioblastoma model in detail with GFP positive glioblastoma cells in fully immune competent animals and study tumor growth rate and tumor mass as a function of time from inoculation. We were able to correlate findings made with classical immunohistochemistry and MR findings. The tumor growth rate was fitted by a Gompertz function. The model predicted the time until onset of symptoms for 5000 inoculated cells to 18.7±0.4 days, and the tumor mass at days 10 and 14, which are commonly used as the start of treatment in therapeutic studies, were 5.97±0.62 mg and 29.1±3.0 mg, respectively. We want to raise the question regarding the clinical relevance of the outline of glioblastoma experiments, where treatment is often initiated at a very early stage. The approach presented here could potentially be modified to gain information also from other tumor models.


Assuntos
Neoplasias Encefálicas/patologia , Modelos Animais de Doenças , Glioblastoma/patologia , Transplante de Neoplasias/métodos , Animais , Linhagem Celular Tumoral , Proliferação de Células , Ratos , Ratos Endogâmicos F344
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA