Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Life Sci ; 346: 122591, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38548013

RESUMO

As a family of cationic host defense peptides, human ß-defensins (HBDs) are ubiquitous in the oral cavity and are mainly synthesized primarily by epithelial cells, serving as the primary barrier and aiming to prevent microbial invasion, inflammation, and disease while maintaining physiological homeostasis. In recent decades, there has been great interest in their biological functions, structure-activity relationships, mechanisms of action, and therapeutic potential in oral diseases. Meanwhile, researchers are dedicated to improving the properties of HBDs for clinical application. In this review, we first describe the classification, structural characteristics, functions, and mechanisms of HBDs. Next, we cover the role of HBDs and their synthetic analogs in oral diseases, including dental caries and pulp infections, periodontitis, peri-implantitis, fungal/viral infections and oral mucosal diseases, and oral squamous cell carcinoma. Finally, we discuss the limitations and challenges of clinical translation of HBDs and their synthetic analogs, including, but not limited to, stability, bioavailability, antimicrobial activity, resistance, and toxicity. Above all, this review summarizes the biological functions, mechanisms of action, and therapeutic potential of both natural HBDs and their synthetic analogs in oral diseases, as well as the challenges associated with clinical translation, thus providing substantial insights into the laboratory development and clinical application of HBDs in oral diseases.


Assuntos
Saúde Bucal , beta-Defensinas , Humanos , beta-Defensinas/farmacologia , beta-Defensinas/química , Doenças da Boca/tratamento farmacológico , Animais , Relação Estrutura-Atividade
2.
Cancer Res ; 84(4): 517-526, 2024 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-38085180

RESUMO

The three-dimensional (3D) tumor microenvironment (TME) comprises multiple interacting cell types that critically impact tumor pathology and therapeutic response. Efficient 3D imaging assays and analysis tools could facilitate profiling and quantifying distinctive cell-cell interaction dynamics in the TMEs of a wide spectrum of human cancers. Here, we developed a 3D live-cell imaging assay using confocal microscopy of patient-derived tumor organoids and a software tool, SiQ-3D (single-cell image quantifier for 3D), that optimizes deep learning (DL)-based 3D image segmentation, single-cell phenotype classification, and tracking to automatically acquire multidimensional dynamic data for different interacting cell types in the TME. An organoid model of tumor cells interacting with natural killer cells was used to demonstrate the effectiveness of the 3D imaging assay to reveal immuno-oncology dynamics as well as the accuracy and efficiency of SiQ-3D to extract quantitative data from large 3D image datasets. SiQ-3D is Python-based, publicly available, and customizable to analyze data from both in vitro and in vivo 3D imaging. The DL-based 3D imaging analysis pipeline can be employed to study not only tumor interaction dynamics with diverse cell types in the TME but also various cell-cell interactions involved in other tissue/organ physiology and pathology. SIGNIFICANCE: A 3D single-cell imaging pipeline that quantifies cancer cell interaction dynamics with other TME cell types using primary patient-derived samples can elucidate how cell-cell interactions impact tumor behavior and treatment responses.


Assuntos
Aprendizado Profundo , Humanos , Microambiente Tumoral , Imageamento Tridimensional/métodos , Software , Comunicação Celular
3.
Mol Cancer Ther ; 22(12): 1479-1492, 2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-37710057

RESUMO

Aberrant activation of the FGF19-FGFR4 signaling pathway plays an essential role in the tumorigenesis of hepatocellular carcinoma (HCC). As such, FGFR4 inhibition has emerged as a novel therapeutic option for the treatment of HCC and has shown preliminary efficacy in recent clinical trials for patients exhibiting aberrant FGF19 expression. Resistance to kinase inhibitors is common in oncology, presenting a major challenge in the clinical treatment process. Hence, we investigated the potential mechanisms mediating and causing resistance to FGFR4 inhibition in HCC. Upon the successful establishment of a battery of cellular models developing resistance to FGFR4 inhibitors, we have identified the activation of EGFR, MAPK, and AKT signaling as the primary mechanisms mediating the acquired resistance. Combination of inhibitors against EGFR or its downstream components restored sensitivity to FGFR4 inhibitors. In parental HCC cell lines, EGF treatment also resulted in resistance to FGFR4 inhibitors. This resistance was effectively reverted by inhibitors of the EGFR signaling pathway, suggesting that EGFR activation is a potential cause of intrinsic resistance. We further confirmed the above findings in vivo in mouse xenograft tumor models. Genomic analysis of patient samples from The Cancer Genome Atlas confirmed that a segment of patients with HCC harboring FGF19 overexpression indeed exhibited increased activation of EGFR signaling. These findings conclusively indicate that both induced and innate activation of EGFR could mediate resistance to FGFR4 inhibition, suggesting that dual blockade of EGFR and FGFR4 may be a promising future therapeutic strategy for the treatment of FGF19-FGFR4 altered HCC.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Animais , Camundongos , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Fatores de Crescimento de Fibroblastos/genética , Transdução de Sinais , Receptores ErbB/metabolismo , Linhagem Celular Tumoral , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/genética
4.
Nat Metab ; 5(7): 1141-1158, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37349486

RESUMO

Redox metabolites have been observed to fluctuate through the cell cycle in cancer cells, but the functional impacts of such metabolic oscillations remain unknown. Here, we uncover a mitosis-specific nicotinamide adenine dinucleotide phosphate (NADPH) upsurge that is essential for tumour progression. Specifically, NADPH is produced by glucose 6-phosphate dehydrogenase (G6PD) upon mitotic entry, which neutralizes elevated reactive oxygen species (ROS) and prevents ROS-mediated inactivation of mitotic kinases and chromosome missegregation. Mitotic activation of G6PD depends on the phosphorylation of its co-chaperone protein BAG3 at threonine 285, which results in dissociation of inhibitory BAG3. Blocking BAG3T285 phosphorylation induces tumour suppression. A mitotic NADPH upsurge is present in aneuploid cancer cells with high levels of ROS, while nearly unobservable in near-diploid cancer cells. High BAG3T285 phosphorylation is associated with worse prognosis in a cohort of patients with microsatellite-stable colorectal cancer. Our study reveals that aneuploid cancer cells with high levels of ROS depend on a G6PD-mediated NADPH upsurge in mitosis to protect them from ROS-induced chromosome missegregation.


Assuntos
Segregação de Cromossomos , Neoplasias , Humanos , NADP/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Aneuploidia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo
5.
Methods Cell Biol ; 173: 49-64, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36653085

RESUMO

Natural Killer (NK) cells detect and eliminate virus-infected cells and cancer cells, and are crucial players of the human immune defense system. Although the relevant molecular machineries involved in NK cell activation and NK-target cell interactions are largely known, how their collective signaling modulates the dynamic behaviors of NK cells, e.g., motility and cytotoxicity, and the rate-limiting kinetics involved are still in need of comprehensive investigations. In traditional bulk killing assays, heterogeneity and kinetic details of individual NK-target cell interactions are masked, seriously limiting analysis of the underlying dynamic mechanisms. Here we present detailed protocols of a number of live-cell imaging assays using fluorescent protein reporters and/or a live-cell dye that enable the acquisition of quantitative kinetic data at the single cell level for elucidating the mechanism underlying the interaction dynamics of primary human NK cells and epithelial cancer cells. Moreover, we discuss how the imaging data can be analyzed either alone or in combination to quantify and determine the key dynamic steps/intermediates involved in specific NK cell activity, e.g., NK cell cytotoxic modes and their associated kinetics, and NK cell motility toward different cancer targets. These live-cell imaging assays can be easily adapted to analyze the rate-limiting kinetics and heterogeneity of other cell-cell interaction dynamics, e.g., in T cell function.


Assuntos
Comunicação Celular , Células Matadoras Naturais , Humanos
7.
J Biol Chem ; 298(11): 102561, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36198360

RESUMO

Cancer cells have distinctive demands for intermediates from glucose metabolism for biosynthesis and energy in different cell cycle phases. However, how cell cycle regulators and glycolytic enzymes coordinate to orchestrate the essential metabolic processes are still poorly characterized. Here, we report a novel interaction between the mitotic kinase, Aurora A, and the glycolytic enzyme, pyruvate kinase M2 (PKM2), in the interphase of the cell cycle. We found Aurora A-mediated phosphorylation of PKM2 at threonine 45. This phosphorylation significantly attenuated PKM2 enzymatic activity by reducing its tetramerization and also promoted glycolytic flux and the branching anabolic pathways. Replacing the endogenous PKM2 with a nonphosphorylated PKM2 T45A mutant inhibited glycolysis, glycolytic branching pathways, and tumor growth in both in vitro and in vivo models. Together, our study revealed a new protumor function of Aurora A through modulating a rate-limiting glycolytic enzyme, PKM2, mainly during the S phase of the cell cycle. Our findings also showed that although both Aurora A and Aurora B kinase phosphorylate PKM2 at the same residue, the spatial and temporal regulations of the specific kinase and PKM2 interaction are context dependent, indicating intricate interconnectivity between cell cycle and glycolytic regulators.


Assuntos
Leucemia Mieloide Aguda , Piruvato Quinase , Humanos , Piruvato Quinase/metabolismo , Fosforilação , Ácido Pirúvico/metabolismo , Linhagem Celular Tumoral , Glicólise , Divisão Celular
8.
Clin Implant Dent Relat Res ; 24(4): 468-474, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35576245

RESUMO

OBJECTIVE: This study was aimed to evaluate bone healing after jaw cyst enucleation with or without bone substitutes by cone beam computed tomography, and to analyze potential influence factors for bone formation as well. MATERIALS AND METHODS: Sixty seven jaw cyst patients were randomly assigned to two groups. Thirty three patients in control group accepted cystectomy without any filling material. The rest 34 bone cavities which filled with xenograft (DBBM, Bio-Oss®) and covered by absorbable membrane (Bio-Gide®) were included in the guided bone regeneration (GBR) group. All patients were examined with cone bean computerized tomography before operation, 3 and 6 months after surgery. Linear regression analysis was applied to evaluate the influence factors of bone healing. RESULTS: There was no significant difference in bone formation rate at 3 months after enucleation, with shrinkage rate (SR) of cystic lesion in control group and GBR group of 26.43 ± 14.98% and 20.78 ± 10.80%, respectively (p > 0.05). Larger shrinkage area in GBR group was detected on postoperative radiographs after 6 months with SR of 60.11 ± 19.23%, when compared to those in patients without filling (6 months SR: 48.63 ± 19.39%, p = 0.018, <0.05). Linear regression analysis showed that cyst size was negatively correlated with bone formation. CONCLUSION: GBR with bovine xenograft and absorbable membrane showed considerable bone regeneration property in the healing of jaw cystic defects after enucleation of radicular cysts. Cyst size showed a suppressive influence on bone formation.


Assuntos
Substitutos Ósseos , Cistos Maxilomandibulares , Animais , Regeneração Óssea , Substitutos Ósseos/uso terapêutico , Transplante Ósseo/métodos , Bovinos , Humanos , Cistos Maxilomandibulares/cirurgia , Estudos Prospectivos
9.
Adv Sci (Weinh) ; 9(16): e2103230, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35403834

RESUMO

Tumor heterogeneity plays a key role in cancer relapse and metastasis, however, the distinct cellular behaviors and kinetics of interactions among different cancer cell subclones and the tumor microenvironment are poorly understood. By profiling an isogenic model that resembles spontaneous human ovarian cancer metastasis with an highly metastatic (HM) and non-metastatic (NM) tumor cell pair, one finds an upregulation of Wnt/ß-catenin signaling uniquely in HM. Using humanized immunocompetent mice, one shows for the first time that activated ß-catenin acts nonautonomously to modulate the immune microenvironment by enhancing infiltrating tumor-associated macrophages (TAM) at the metastatic site. Single-cell time-lapse microscopy further reveals that upon contact with macrophages, a significant subset of HM, but not NM, becomes polyploid, a phenotype pivotal for tumor aggressiveness and therapy resistance. Moreover, HM, but not NM, polarizes macrophages to a TAM phenotype. Mechanistically, ß-catenin upregulates cancer cell surface metadherin, which communicates through CEACAM1 expressed on macrophages to produce CCL3. Tumor xenografts in humanized mice and clinical patient samples both corroborate the relevance of enhanced metastasis, TAM activation, and polyploidy in vivo. The results thus suggest that targeting the ß-catenin-metadherin/CEACAM1-CCL3 positive feedback cascade holds great therapeutic potential to disrupt polyploidization of the cancer subclones that drive metastasis.


Assuntos
Via de Sinalização Wnt , beta Catenina , Animais , Antígenos CD , Antígeno Carcinoembrionário/metabolismo , Moléculas de Adesão Celular , Linhagem Celular Tumoral , Quimiocina CCL3/metabolismo , Humanos , Macrófagos/metabolismo , Camundongos , Recidiva Local de Neoplasia/metabolismo , Fatores de Transcrição/metabolismo , Microambiente Tumoral , Via de Sinalização Wnt/fisiologia , beta Catenina/genética , beta Catenina/metabolismo
10.
BMC Biol ; 20(1): 85, 2022 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-35410287

RESUMO

BACKGROUND: To understand functional changes of complex biological networks, mathematical modeling of network topologies provides a quantitative measure of the way biological systems adapt to external stimuli. However, systemic network topology-based analysis often generates conflicting evidence depending on specific experimental conditions, leading to a limited mechanistic understanding of signaling networks and their differential dynamic outputs, an example of which is the regulation of p53 pathway responses to different stress stimuli and in variable mammalian cell types. Here, we employ a network motif approach to dissect key regulatory units of the p53 pathway and elucidate how network activities at the motif level generate context-specific dynamic responses. RESULTS: By combining single-cell imaging and mathematical modeling of dose-dependent p53 dynamics induced by three chemotherapeutics of distinct mechanism-of-actions, including Etoposide, Nutlin-3a and 5-fluorouracil, and in five cancer cell types, we uncovered novel and highly variable p53 dynamic responses, in particular p53 transitional dynamics induced at intermediate drug concentrations, and identified the functional roles of distinct positive and negative feedback motifs of the p53 pathway in modulating the central p53-Mdm2 negative feedback to generate stimulus- and cell type-specific signaling responses. The mechanistic understanding of p53 network dynamics also revealed previously unknown mediators of anticancer drug actions and phenotypic variations in cancer cells that impact drug sensitivity. CONCLUSIONS: Our results demonstrate that transitional dynamics of signaling proteins such as p53, activated at intermediate stimulus levels, vary the most between the dynamic outputs of different generic network motifs and can be employed as novel quantitative readouts to uncover and elucidate the key building blocks of large signaling networks. Our findings also provide new insight on drug mediators and phenotypic heterogeneity that underlie differential drug responses.


Assuntos
Análise de Célula Única , Proteína Supressora de Tumor p53 , Animais , Mamíferos , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo
11.
BMC Biol ; 19(1): 140, 2021 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-34325694

RESUMO

BACKGROUND: Natural killer (NK) cells play an important role in cancer immunosurveillance and therapy. However, the target selectivity of NK cell activity is still poorly understood. RESULTS: Here, we used live-cell reporters to unravel differential epithelial cancer target killing by primary human NK cells. We found highly variable fractions of killing by distinct NK cell cytotoxic modes that were not determined by NK ligand expression. Rather, epithelial plasma membrane dynamics driven by ROCK-mediated blebs and/or Rac1-mediated lamellipodia promoted necrotic mode in preference to the apoptotic mode of killing. Inhibition of granzyme B and key necroptosis regulators RIP1, RIP3, and MLKL significantly attenuated the necrotic killing, revealing a novel NK cell cytotoxic pathway by granzyme-induced necroptosis that conferred target selectivity. CONCLUSIONS: Our results not only elucidate a new NK cell effector mechanism but also suggest that tissue microenvironment and oncogenic signaling pathways that promote membrane dynamics, e.g., Rac1 and Rho/ROCK, could be exploited to enhance proinflammatory NK cell killing.


Assuntos
Células Matadoras Naturais , Necroptose , Granzimas/metabolismo , Humanos , Células Matadoras Naturais/metabolismo , Necrose , Transdução de Sinais , Proteínas rac1 de Ligação ao GTP
12.
Front Surg ; 8: 678696, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34250007

RESUMO

In the past half-century, considerable attention has been paid to oral and maxillofacial skeletal cyst, however, aneurysmal bone cyst (ABC), unlike other common bone diseases, still contours numerous unanswered questions in terms of classification, etiology and pathological mechanism. The purpose of this article was to evaluate the proportion of primary ABC and secondary ABC, and to assess the recurrence of ABC and related factors. A methodical search of Embase, MEDLINE, Cochrane Library, Web of Science was conducted for well-documented jaw aneurysmal bone cyst (JABC) cases. One hundred thirty-one articles were identified after database searching and 31 of them were included in our study for further research with 44 JABC cases. All the articles were analyzed by two separate authors. About 25% of the reported jaw aneurysmal bone cyst was secondary. Both the pathological classification and surgical treatment had a significant influence on recurrence rate (P = 0.0082, P = 0.0022), while patients' age or radiographic features rarely affected prognosis. Jaw aneurysmal bone cysts can present variable clinical and histological presentations. Recurrence may be attributed to omittance of underlying potential blood supply or conservative surgical protocol.

13.
Cell Cycle ; 20(12): 1195-1208, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34048314

RESUMO

ATP metabolism during mitosis needs to be coordinated with numerous energy-demanding activities, especially in cancer cells whose metabolic pathways are reprogramed to sustain rapid proliferation in a nutrient-deficient environment. Although strategies targeting the energy metabolic pathways have shown therapeutic efficacy in preclinical cancer models, how normal cells and cancer cells differentially respond to energy shortage is unclear. In this study, using time-lapse microscopy, we found that cancer cells displayed unique mitotic phenotypes in a dose-dependent manner upon decreasing ATP (i.e. energy) supply. When reduction in ATP concentration was moderate, chromosome movements in mitosis were barely affected, while the metaphase-anaphase transition was significantly prolonged due to reduced tension between the sister-kinetochores, which delayed the satisfaction of the spindle assembly checkpoint. Further reduction in ATP concentration led to a decreased level of Aurora-B at the centromere, resulting in increased chromosome mis-segregation after metaphase delay. In contrast to cancer cells, ATP restriction in non-transformed cells induced cell cycle arrest in interphase, rather than causing mitotic defects. In addition, data mining of cancer patient database showed a correlation between signatures of energy production and chromosomal instability possibly resulted from mitotic defects. Together, these results reveal that energy restriction induces differential responses in normal and cancer cells, with chromosome mis-segregation only observed in cancer cells. This points to targeting energy metabolism as a potentially cancer-selective therapeutic strategy.


Assuntos
Trifosfato de Adenosina/metabolismo , Trifosfato de Adenosina/farmacologia , Segregação de Cromossomos/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Metáfase/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Neoplasias do Colo do Útero/metabolismo , Anáfase/efeitos dos fármacos , Aurora Quinase B/metabolismo , Feminino , Células HeLa , Humanos , Interfase/efeitos dos fármacos , Cinetocoros/metabolismo , Microscopia/métodos , NAD/farmacologia , Fuso Acromático/metabolismo , Imagem com Lapso de Tempo/métodos , Neoplasias do Colo do Útero/patologia
14.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 41(2): 220-227, 2019 Apr 28.
Artigo em Chinês | MEDLINE | ID: mdl-31060678

RESUMO

Objective To investigate the expressions,roles,and clinical significance of microRNA-365(miR-365)and E74-like factor 4(ELF4)in cervical cancer. Methods The expressions of miR-365 in normal cervical tissues(n=34),cervical intraepithelial neoplasia 1(CIN 1)(n=31),cervical intraepithelial neoplasia2-3(CIN 2-3)(n=37),squamous cell carcinoma of the cervix(SCC)(n=33),and three cervical cancer cell lines(C33A cells,Hela cells,and SiHa cells)were detected by real-time quantitative polymerase chain reaction(qPCR).Bioinformatic prediction and luciferase reporter gene assay were performed to verify whether ELF4 was a direct target of miR-365.Western blot and immunohistochemistry were used to detect ELF4 expression in cervical cancer cells and in different pathological cervix tissues.CCK8 assay was used to detect the effect of overexpression or inhibition of miR-365 on the proliferation of cervical cancer cells at different time points.The relationships among the miR-365 expression,ELF4 expression,and clinicopathological parameters of cervical cancer were analyzed by correlation analysis. Results qPCR results showed that compared with the normal cervical cell HcerEpic,the expressions of miR-365 in CIN1,CIN2-3,and cervical cancer tissues gradually decreased with the increased pathologic grade,and its expressions also decreased in different cervical cancer cell lines.The luciferase reporter gene assay confirmed that ELF4 was the direct target of miR-365.Western blot showed that the expression of ELF4 increased in all three cervical cancer cell lines compared with normal cervical epidermal cell(P=0.013,P=0.002,P=0.004).Immunohistochemistry showed that ELF4 expression was up-regulated in CIN and cervical cancer tissues.CCK8 assay showed that overexpression of miR-365 inhibited cell proliferation,while inhibition of miR-365 promoted the proliferation of three cervical cancer cells(P<0.05).Further analysis confirmed that there was a negative correlation between the expression levels of miR-365 and ELF4 in CIN2-3 and SCC(r=-0.351,P=0.045;r=-0.349,P=0.035).Clinical analysis showed that the expressions of both miR-365 and ELF4 were correlated with tumor size,pathological grade,and clinical stage in SCC(all P < 0.05).Conclusion The decreased expression of miR-365 in human cervical cancer cells relieves its inhibitory effect on ELF4,which promotes the proliferation of cervical cancer cells and the formation of tumor.


Assuntos
Proteínas de Ligação a DNA/genética , MicroRNAs/genética , Fatores de Transcrição/genética , Neoplasias do Colo do Útero/genética , Proliferação de Células , Feminino , Células HeLa , Humanos
16.
Sci Adv ; 4(12): eaat5077, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30585287

RESUMO

Studies of drug resistance mostly characterize genetic mutation, and we know much less about phenotypic mechanisms of drug resistance, especially at a quantitative level. p53 is an important mediator of cellular response to chemotherapy, but even p53 wild-type cells vary in drug sensitivity for unclear reasons. Here, we elucidated a new resistance mechanism to a DNA-damaging chemotherapeutic through bimodal modulation of p53 activation dynamics. By combining single-cell imaging with computational modeling, we characterized a four-component regulatory module, which generates bimodal p53 dynamics through coupled feed-forward and feedback, and found that the inhibitory strength between ATM and Mdm2 determined the differential modular output between drug-sensitive and drug-resistant cancer cell lines. We further showed that the combinatorial inhibition of Mdm2 and Wip1 was an effective strategy to alter p53 dynamics in resistant cancer cells and sensitize their apoptotic response. Our results point to p53 pulsing as a potentially druggable mechanism that mediates chemoresistance.


Assuntos
Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Algoritmos , Apoptose/genética , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Linhagem Celular Tumoral , Dano ao DNA , Etoposídeo/farmacologia , Humanos , Modelos Biológicos , Especificidade de Órgãos/genética , Fosforilação , Proteólise , Proteínas Proto-Oncogênicas c-mdm2/metabolismo
17.
Cell Stem Cell ; 23(6): 882-897.e11, 2018 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-30344100

RESUMO

Gastric cancer displays marked molecular heterogeneity with aggressive behavior and treatment resistance. Therefore, good in vitro models that encompass unique subtypes are urgently needed for precision medicine development. Here, we have established a primary gastric cancer organoid (GCO) biobank that comprises normal, dysplastic, cancer, and lymph node metastases (n = 63) from 34 patients, including detailed whole-exome and transcriptome analysis. The cohort encompasses most known molecular subtypes (including EBV, MSI, intestinal/CIN, and diffuse/GS, with CLDN18-ARHGAP6 or CTNND1-ARHGAP26 fusions or RHOA mutations), capturing regional heterogeneity and subclonal architecture, while their morphology, transcriptome, and genomic profiles remain closely similar to in vivo tumors, even after long-term culture. Large-scale drug screening revealed sensitivity to unexpected drugs that were recently approved or in clinical trials, including Napabucasin, Abemaciclib, and the ATR inhibitor VE-822. Overall, this new GCO biobank, with linked genomic data, provides a useful resource for studying both cancer cell biology and precision cancer therapy.


Assuntos
Antineoplásicos/farmacologia , Bancos de Espécimes Biológicos , Ensaios de Seleção de Medicamentos Antitumorais , Organoides/efeitos dos fármacos , Organoides/patologia , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/patologia , Aminopiridinas/farmacologia , Benzimidazóis/farmacologia , Benzofuranos/farmacologia , Proliferação de Células/efeitos dos fármacos , Feminino , Humanos , Isoxazóis/farmacologia , Masculino , Naftoquinonas/farmacologia , Medicina de Precisão , Pirazinas/farmacologia , Neoplasias Gástricas/classificação , Neoplasias Gástricas/genética
18.
Nat Commun ; 8(1): 1940, 2017 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-29208897

RESUMO

Cancer is a complex disease involving multiple genomic alterations that disrupt the dynamic response of signaling networks. The heterogeneous nature of cancer, which results in highly variable drug response, is a major obstacle to developing effective cancer therapy. Previous studies of cancer therapeutic response mostly focus on static analysis of genome-wide alterations, thus they are unable to unravel the dynamic, network-specific origin of variation. Here we present a network dynamics-based approach to integrate cancer genomics with dynamics of biological network for drug response prediction and design of drug combination. We select the p53 network as an example and analyze its cancer-specific state transition dynamics under distinct anticancer drug treatments by attractor landscape analysis. Our results not only enable stratification of cancer into distinct drug response groups, but also reveal network-specific drug targets that maximize p53 network-mediated cell death, providing a basis to design combinatorial therapeutic strategies for distinct cancer genomic subtypes.


Assuntos
Antineoplásicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Genômica , Neoplasias/tratamento farmacológico , Proteína Supressora de Tumor p53/genética , Morte Celular , Linhagem Celular Tumoral , Redes Reguladoras de Genes , Humanos , Neoplasias/genética , Transdução de Sinais
19.
Oncotarget ; 8(31): 51253-51263, 2017 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-28881645

RESUMO

Though the signaling events involved in radiation induced bystander effects (RIBE) have been investigated both in vitro and in vivo, the spatial function of these communications, especially the related signaling pathways, is not fully elucidated. In the current study, significant increases of DNA damage were clearly observed in C. elegans germline upon irradiation to both intra-system of posterior pharynx and inter-system of vulva, in which more severe damage, even to F1 generation worms, was shown for vulva irradiation. Spatial function assay indicated the DDR key components of mrt-2/hus-1/cep-1/ced-4 were indispensable in germ cells for both sites irradiation, while those components in somatic cells were either not (cep-1/ced-4) or partially (mrt-2/hus-1) required to promote apoptosis. Moreover, production of reactive oxygen species (ROS) indicated by the superoxide dismutase expression and the unfolded protein response of the mitochondria was found systemically involved in the initiation of these processes for both two site irradiation. These results will give a better understanding of the RIBE mechanisms in vivo, and invaluable to assess the clinical relevance to radiotherapy.

20.
Endocr Relat Cancer ; 24(9): T83-T96, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28249963

RESUMO

Anti-mitotic cancer drugs include classic microtubule-targeting drugs, such as taxanes and vinca alkaloids, and the newer spindle-targeting drugs, such as inhibitors of the motor protein; Kinesin-5 (aka KSP, Eg5, KIF11); and Aurora-A, Aurora-B and Polo-like kinases. Microtubule-targeting drugs are among the first line of chemotherapies for a wide spectrum of cancers, but patient responses vary greatly. We still lack understanding of how these drugs achieve a favorable therapeutic index, and why individual patient responses vary. Spindle-targeting drugs have so far shown disappointing results in the clinic, but it is possible that certain patients could benefit if we understand their mechanism of action better. Pre-clinical data from both cell culture and mouse tumor models showed that the cell death response is the most variable point of the drug action. Hence, in this review we focus on current mechanistic understanding of the cell death response to anti-mitotics. We first draw on extensive results from cell culture studies, and then cross-examine them with the more limited data from animal tumor models and the clinic. We end by discussing how cell type variation in cell death response might be harnessed to improve anti-mitotic chemotherapy by better patient stratification, new drug combinations and identification of novel targets for drug development.


Assuntos
Antimitóticos/farmacologia , Antimitóticos/uso terapêutico , Morte Celular/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Animais , Dano ao DNA , Modelos Animais de Doenças , Humanos , Camundongos , Microtúbulos , Neoplasias/genética , Fuso Acromático , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA