Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 360
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 158(3): 659-72, 2014 Jul 31.
Article in English | MEDLINE | ID: mdl-25083875

ABSTRACT

Circadian rhythms are intimately linked to cellular metabolism. Specifically, the NAD(+)-dependent deacetylase SIRT1, the founding member of the sirtuin family, contributes to clock function. Whereas SIRT1 exhibits diversity in deacetylation targets and subcellular localization, SIRT6 is the only constitutively chromatin-associated sirtuin and is prominently present at transcriptionally active genomic loci. Comparison of the hepatic circadian transcriptomes reveals that SIRT6 and SIRT1 separately control transcriptional specificity and therefore define distinctly partitioned classes of circadian genes. SIRT6 interacts with CLOCK:BMAL1 and, differently from SIRT1, governs their chromatin recruitment to circadian gene promoters. Moreover, SIRT6 controls circadian chromatin recruitment of SREBP-1, resulting in the cyclic regulation of genes implicated in fatty acid and cholesterol metabolism. This mechanism parallels a phenotypic disruption in fatty acid metabolism in SIRT6 null mice as revealed by circadian metabolome analyses. Thus, genomic partitioning by two independent sirtuins contributes to differential control of circadian metabolism.


Subject(s)
Liver/metabolism , Sirtuins/metabolism , ARNTL Transcription Factors/metabolism , Animals , CLOCK Proteins/metabolism , Chromatin , Circadian Rhythm , Gene Expression Profiling , Mice , Mice, Knockout , Sirtuin 1/genetics , Sirtuin 1/metabolism , Sirtuins/genetics , Transcription, Genetic
2.
EMBO Rep ; 25(3): 1361-1386, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38332150

ABSTRACT

Non-alcoholic fatty liver disease is a chronic liver abnormality that exhibits high variability and can lead to liver cancer in advanced stages. Hepatic ablation of SIRT6 results in fatty liver disease, yet the potential mechanism of SIRT6 deficiency, particularly in relation to downstream mediators for NAFLD, remains elusive. Here we identify Serpina12 as a key gene regulated by Sirt6 that plays a crucial function in energy homeostasis. Specifically, Sirt6 suppresses Serpina12 expression through histone deacetylation at its promoter region, after which the transcription factor, Cebpα, binds to and regulates its expression. Sirt6 deficiency results in an increased expression of Serpina12 in hepatocytes, which enhances insulin signaling and promotes lipid accumulation. Importantly, CRISPR-Cas9 mediated Serpina12 knockout in the liver ameliorated fatty liver disease caused by Sirt6 ablation. Finally, we demonstrate that Sirt6 functions as a tumor suppressor in the liver, and consequently, deletion of Sirt6 in the liver leads to not only the spontaneous development of tumors but also enhanced tumorigenesis in response to DEN treatment or under conditions of obesity.


Subject(s)
Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Sirtuins , Humans , Sirtuins/metabolism , Liver/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Hepatocytes/metabolism , Liver Neoplasms/metabolism
3.
Cell ; 141(2): 243-54, 2010 Apr 16.
Article in English | MEDLINE | ID: mdl-20362325

ABSTRACT

Defective DNA repair by homologous recombination (HR) is thought to be a major contributor to tumorigenesis in individuals carrying Brca1 mutations. Here, we show that DNA breaks in Brca1-deficient cells are aberrantly joined into complex chromosome rearrangements by a process dependent on the nonhomologous end-joining (NHEJ) factors 53BP1 and DNA ligase 4. Loss of 53BP1 alleviates hypersensitivity of Brca1 mutant cells to PARP inhibition and restores error-free repair by HR. Mechanistically, 53BP1 deletion promotes ATM-dependent processing of broken DNA ends to produce recombinogenic single-stranded DNA competent for HR. In contrast, Lig4 deficiency does not rescue the HR defect in Brca1 mutant cells but prevents the joining of chromatid breaks into chromosome rearrangements. Our results illustrate that HR and NHEJ compete to process DNA breaks that arise during DNA replication and that shifting the balance between these pathways can be exploited to selectively protect or kill cells harboring Brca1 mutations.


Subject(s)
BRCA1 Protein/genetics , DNA Repair , Intracellular Signaling Peptides and Proteins/metabolism , Animals , B-Lymphocytes/metabolism , Chromosomal Proteins, Non-Histone , DNA Breaks , DNA-Binding Proteins , Female , Genomic Instability , Humans , Mice , Tumor Suppressor p53-Binding Protein 1
4.
Stem Cells ; 41(6): 578-591, 2023 06 15.
Article in English | MEDLINE | ID: mdl-36648303

ABSTRACT

The interplay among mitogenic signaling pathways is crucial for proper embryogenesis. These pathways collaboratively act through intracellular master regulators to determine specific cell fates. Identifying the master regulators is critical to understanding embryogenesis and to developing new applications of pluripotent stem cells. In this report, we demonstrate protein kinase C (PKC) as an intrinsic master switch between embryonic and extraembryonic cell fates in the differentiation of human pluripotent stem cells (hPSCs). PKCs are essential to induce the extraembryonic lineage downstream of BMP4 and other mitogenic modulators. PKC-alpha (PKCα) suppresses BMP4-induced mesoderm differentiation, and PKC-delta (PKCδ) is required for trophoblast cell fate. PKC activation overrides mesoderm induction conditions and leads to extraembryonic fate. In contrast, PKC inhibition leads to ß-catenin (CTNNB1) activation, switching cell fate from trophoblast to mesoderm lineages. This study establishes PKC as a signaling boundary directing the segregation of extraembryonic and embryonic lineages. The manipulation of intrinsic PKC activity could greatly enhance cell differentiation under mitogenic regulation in stem cell applications.


Subject(s)
Pluripotent Stem Cells , Protein Kinase C , Humans , Protein Kinase C/metabolism , Embryonic Stem Cells/metabolism , Cell Differentiation , Pluripotent Stem Cells/metabolism , Mesoderm/metabolism , Bone Morphogenetic Protein 4/pharmacology , Bone Morphogenetic Protein 4/metabolism
5.
FASEB J ; 37(8): e23073, 2023 08.
Article in English | MEDLINE | ID: mdl-37402125

ABSTRACT

In female mammals, the oviduct and uterus are essential sites for female and male gamete transport, fertilization, implantation, and maintenance of a successful pregnancy. To delineate the reproductive function of Mothers against decapentaplegic homolog 4 (Smad4), we specifically inactivated Smad4 in ovarian granulosa cells and, oviduct and uterine mesenchymal cells using the Amhr2-cre mouse line. Deletion of exon 8 of Smad4 results in the production of an MH2-truncated SMAD4 protein. These mutant mice are infertile due to the development of oviductal diverticula and defects during the implantation process. The ovaries are fully functional as demonstrated in an ovary transfer experiment. The development of oviductal diverticula occurs shortly after puberty and is dependent on estradiol. The diverticula interfere with sperm migration and embryo transit to the uterus, reducing the number of implantation sites. Analysis of the uterus shows that, even if implantation occurs, decidualization and vascularization are defective resulting in embryo resorption as early as the seventh day of pregnancy. Thus, Smad4 plays an important function in female reproduction by controlling the structural and functional integrity of the oviduct and uterus.


Subject(s)
Estradiol , Smad4 Protein , Animals , Female , Humans , Male , Mice , Pregnancy , Embryo Implantation , Estradiol/metabolism , Mammals/metabolism , Oviducts/metabolism , Semen/metabolism , Smad4 Protein/genetics , Smad4 Protein/metabolism , Uterus/metabolism
6.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Article in English | MEDLINE | ID: mdl-33335073

ABSTRACT

The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has emerged as a major threat to global health. Although varied SARS-CoV-2-related coronaviruses have been isolated from bats and SARS-CoV-2 may infect bat, the structural basis for SARS-CoV-2 to utilize the human receptor counterpart bat angiotensin-converting enzyme 2 (bACE2) for virus infection remains less understood. Here, we report that the SARS-CoV-2 spike protein receptor binding domain (RBD) could bind to bACE2 from Rhinolophus macrotis (bACE2-Rm) with substantially lower affinity compared with that to the human ACE2 (hACE2), and its infectivity to host cells expressing bACE2-Rm was confirmed with pseudotyped SARS-CoV-2 virus and SARS-CoV-2 wild virus. The structure of the SARS-CoV-2 RBD with the bACE2-Rm complex was determined, revealing a binding mode similar to that of hACE2. The analysis of binding details between SARS-CoV-2 RBD and bACE2-Rm revealed that the interacting network involving Y41 and E42 of bACE2-Rm showed substantial differences with that to hACE2. Bats have extensive species diversity and the residues for RBD binding in bACE2 receptor varied substantially among different bat species. Notably, the Y41H mutant, which exists in many bats, attenuates the binding capacity of bACE2-Rm, indicating the central roles of Y41 in the interaction network. These findings would benefit our understanding of the potential infection of SARS-CoV-2 in varied species of bats.


Subject(s)
Angiotensin-Converting Enzyme 2 , COVID-19/genetics , COVID-19/metabolism , Chiroptera , SARS-CoV-2 , Amino Acid Substitution , Angiotensin-Converting Enzyme 2/chemistry , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/metabolism , Animals , COVID-19/epidemiology , Chiroptera/genetics , Chiroptera/metabolism , Chiroptera/virology , HEK293 Cells , Humans , Mutation, Missense , Pandemics , Protein Binding , Protein Domains , SARS-CoV-2/chemistry , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , Species Specificity
7.
Mol Cancer ; 22(1): 71, 2023 04 18.
Article in English | MEDLINE | ID: mdl-37072770

ABSTRACT

Mesenchymal gastrointestinal cancers are represented by the gastrointestinal stromal tumors (GISTs) which occur throughout the whole gastrointestinal tract, and affect human health and economy globally. Curative surgical resections and tyrosine kinase inhibitors (TKIs) are the main managements for localized GISTs and recurrent/metastatic GISTs, respectively. Despite multi-lines of TKIs treatments prolonged the survival time of recurrent/metastatic GISTs by delaying the relapse and metastasis of the tumor, drug resistance developed quickly and inevitably, and became the huge obstacle for stopping disease progression. Immunotherapy, which is typically represented by immune checkpoint inhibitors (ICIs), has achieved great success in several solid tumors by reactivating the host immune system, and been proposed as an alternative choice for GIST treatment. Substantial efforts have been devoted to the research of immunology and immunotherapy for GIST, and great achievements have been made. Generally, the intratumoral immune cell level and the immune-related gene expressions are influenced by metastasis status, anatomical locations, driver gene mutations of the tumor, and modulated by imatinib therapy. Systemic inflammatory biomarkers are regarded as prognostic indicators of GIST and closely associated with its clinicopathological features. The efficacy of immunotherapy strategies for GIST has been widely explored in pre-clinical cell and mouse models and clinical experiments in human, and some patients did benefit from ICIs. This review comprehensively summarizes the up-to-date advancements of immunology, immunotherapy and research models for GIST, and provides new insights and perspectives for future studies.


Subject(s)
Antineoplastic Agents , Gastrointestinal Neoplasms , Gastrointestinal Stromal Tumors , Sarcoma , Animals , Mice , Humans , Gastrointestinal Stromal Tumors/genetics , Gastrointestinal Stromal Tumors/therapy , Neoplasm Recurrence, Local/drug therapy , Gastrointestinal Neoplasms/therapy , Gastrointestinal Neoplasms/pathology , Sarcoma/drug therapy , Immunotherapy , Antineoplastic Agents/therapeutic use , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/therapeutic use
8.
Small ; 19(31): e2206667, 2023 08.
Article in English | MEDLINE | ID: mdl-36651015

ABSTRACT

Obesity is a major global health problem that significantly increases the risk of many other diseases. Herein, a facile method of suppressing lipogenesis and obesity using L-arginine-functionalized carbon dots (L-Arg@CDots) is reported. The prepared CDots with a negative surface charge form stronger bonds than D-arginine and lysine with L-Arg in water. The L-Arg@CDots in the aqueous solution offer a high photoluminescence quantum yield of 23.6% in the red wavelength region. The proposed L-Arg functionalization strategy not only protects the red emission of the CDots from quenching by water molecules but also enhances the intracellular uptake of L-Arg to reduce lipogenesis. Injection of L-Arg@CDots can reduce the body weight increase in ob/ob mice by suppressing their food intake and shrinking the white adipose tissue cells, thereby significantly inhibiting obesity.


Subject(s)
Carbon , Quantum Dots , Mice , Animals , Carbon/chemistry , Obesity , Arginine , Quantum Dots/chemistry
9.
Pharmacol Res ; 194: 106830, 2023 08.
Article in English | MEDLINE | ID: mdl-37343647

ABSTRACT

Drug combination therapy is a highly effective approach for enhancing the therapeutic efficacy of anti-cancer drugs and overcoming drug resistance. However, the innumerable possible drug combinations make it impractical to screen all synergistic drug pairs. Moreover, biological insights into synergistic drug pairs are still lacking. To address this challenge, we systematically analyzed drug combination datasets curated from multiple databases to identify drug pairs more likely to show synergy. We classified drug pairs based on their MoA and discovered that 110 MoA pairs were significantly enriched in synergy in at least one type of cancer. To improve the accuracy of predicting synergistic effects of drug pairs, we developed a suite of machine learning models that achieve better predictive performance. Unlike most previous methods that were rarely validated by wet-lab experiments, our models were validated using two-dimensional cell lines and three-dimensional tumor slice culture (3D-TSC) models, implying their practical utility. Our prediction and validation results indicated that the combination of the RTK inhibitors Lapatinib and Pazopanib exhibited a strong therapeutic effect in breast cancer by blocking the downstream PI3K/AKT/mTOR signaling pathway. Furthermore, we incorporated molecular features to identify potential biomarkers for synergistic drug pairs, and almost all potential biomarkers found connections between drug targets and corresponding molecular features using protein-protein interaction network. Overall, this study provides valuable insights to complement and guide rational efforts to develop drug combination treatments.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Humans , Female , Phosphatidylinositol 3-Kinases , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Signal Transduction , Breast Neoplasms/drug therapy , Drug Delivery Systems
10.
Dis Colon Rectum ; 66(5): 733-743, 2023 05 01.
Article in English | MEDLINE | ID: mdl-36898057

ABSTRACT

BACKGROUND: Recent studies have shown patient-derived tumor organoids can predict the drug response of patients with cancer. However, the prognostic value of patient-derived tumor organoid-based drug tests in predicting the progression-free survival of patients with stage IV colorectal cancer after surgery remains unknown. OBJECTIVE: This study aimed to explore the prognostic value of patient-derived tumor organoid-based drug tests in patients with stage IV colorectal cancer after surgery. DESIGN: Retrospective cohort study. SETTINGS: Surgical samples were obtained from patients with stage IV colorectal cancer at the Nanfang Hospital. PATIENTS: A total of 108 patients who underwent surgery with successful patient-derived tumor organoid culture and drug testing were recruited between June 2018 and June 2019. INTERVENTIONS: Patient-derived tumor organoid culture and chemotherapeutic drug testing. MAIN OUTCOMES MEASURES: Progression-free survival. RESULTS: According to the patient-derived tumor organoid-based drug test, 38 patients were drug sensitive and 76 patients were drug resistant. The median progression-free survival was 16.0 months in the drug-sensitive group and 9.0 months in the drug resistant group ( p < 0.001). Multivariate analyses showed that drug resistance (HR, 3.38; 95% CI, 1.84-6.21; p < 0.001), right-sided colon (HR, 3.50; 95% CI, 1.71-7.15; p < 0.001), mucinous adenocarcinoma (HR, 2.47; 95% CI, 1.34-4.55; p = 0.004), and non-R0 resection (HR, 2.70; 95% CI, 1.61-4.54; p < 0.001) were independent predictors of progression-free survival. The new patient-derived tumor organoid-based drug test model, which includes the patient-derived tumor organoid-based drug test, primary tumor location, histological type, and R0 resection, was more accurate than the traditional clinicopathological model in predicting progression-free survival ( p = 0.001). LIMITATIONS: A single-center cohort study. CONCLUSIONS: Patient-derived tumor organoids can predict progression-free survival in patients with stage IV colorectal cancer after surgery. Patient-derived tumor organoid drug resistance is associated with shorter progression-free survival, and the addition of patient-derived tumor organoid drug tests to existing clinicopathological models improves the ability to predict progression-free survival.


Subject(s)
Colorectal Neoplasms , Humans , Cohort Studies , Progression-Free Survival , Retrospective Studies , Colorectal Neoplasms/surgery , Prognosis
11.
EMBO Rep ; 21(12): e51444, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33063473

ABSTRACT

PD-1 is a highly glycosylated inhibitory receptor expressed mainly on T cells. Targeting of PD-1 with monoclonal antibodies (MAbs) to block the interaction with its ligand PD-L1 has been successful for the treatment of multiple tumors. However, polymorphisms at N-glycosylation sites of PD-1 exist in the human population that might affect antibody binding, and dysregulated glycosylation has been observed in the tumor microenvironment. Here, we demonstrate varied N-glycan composition in PD-1, and show that the binding affinity of camrelizumab, a recently approved PD-1-specific MAb, to non-glycosylated PD-1 proteins from E. coli is substantially decreased compared with glycosylated PD-1. The structure of the camrelizumab/PD-1 complex reveals that camrelizumab mainly utilizes its heavy chain to bind to PD-1, while the light chain sterically inhibits the binding of PD-L1 to PD-1. Glycosylation of asparagine 58 (N58) promotes the interaction with camrelizumab, while the efficiency of camrelizumab to inhibit the binding of PD-L1 is substantially reduced for glycosylation-deficient PD-1. These results increase our understanding of how glycosylation affects the activity of PD-1-specific MAbs during immune checkpoint therapy.


Subject(s)
Escherichia coli , Programmed Cell Death 1 Receptor , Antibodies, Monoclonal, Humanized , Escherichia coli/metabolism , Glycosylation , Humans , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism
12.
J Cell Physiol ; 236(2): 981-996, 2021 02.
Article in English | MEDLINE | ID: mdl-32617965

ABSTRACT

Cancer cells metabolize glucose through glycolysis to promote cell proliferation even with abundant oxygen. Multiple glycolysis genes are deregulated during cancer development. Despite intensive effort, the cause of their deregulation remains incompletely understood. Here in this study, we discovered that DHX33 plays a critical role in Warburg effect of cancer cells. DHX33 deficient cells have markedly reduced glycolysis activity. Through RNA-seq analysis, we found multiple critical genes involved in Warburg effect were downregulated after DHX33 deficiency. These genes include lactate dehydrogenase A (LDHA), pyruvate dehydrogenase kinase 1 (PDK1), pyruvate kinase muscle isoform 2 (PKM2), enolase 1 (ENO1), ENO2, hexokinase 1/2, among others. With LDHA, PDK1, and PKM2 as examples, we further revealed that DHX33 altered the epigenetic marks around the promoter of glycolytic genes. This is through DHX33 in complex with Gadd45a-a growth arrest and DNA damage protein. DHX33 is required for the loading of Gadd45a and DNA dioxygenase Tet1 at the promoter sites, which resulted in active DNA demethylation and enhanced histone H4 acetylation. We conclude that DHX33 changes local epigenetic marks in favor of the transcription of glycolysis genes to promote cancer cell proliferation. Our study highlights the significance of RNA helicase DHX33 in Warburg effect and cancer therapeutics.


Subject(s)
DEAD-box RNA Helicases/genetics , Glycolysis/genetics , Animals , Cell Line , Cell Line, Tumor , Cell Proliferation/genetics , DNA Damage/genetics , Down-Regulation/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic/genetics , Transcription, Genetic/genetics , Warburg Effect, Oncologic
13.
Hum Mol Genet ; 28(5): 842-857, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30445628

ABSTRACT

The mammary gland undergoes fast cell proliferation during early pregnancy, yet the mechanism to ensure genome integrity during this highly proliferative stage is largely unknown. We show that pregnancy triggers replicative stresses leading to genetic instability in mice carrying a mammary specific disruption of breast cancer associated gene-1 (BRCA1). The fast cell proliferation was correlated with enhanced expression of most genes encoding replisomes, which are positively regulated by estrogen/ERα signaling but negatively regulated by BRCA1. Our further analysis revealed two parallel signaling pathways, which are mediated by ATR-CHK1 and WEE1-MCM2 and are responsible for regulating DNA replication checkpoint. Upon DNA damage, BRCA1 deficiency markedly enhances DNA replication initiation and preferably impairs DNA replication checkpoint mediated by ATR and CHK1. Meanwhile, DNA damage also activates WEE1-MCM2 signaling, which inhibits DNA replication initiation and enables BRCA1-deficient cells to avoid further genomic instability. Finally, we demonstrated that overriding this defense by WEE1 inhibition in combination with cisplatin, which causes DNA damage, serves as a promising therapeutic approach for killing BRCA1-deficient cancer cells.


Subject(s)
BRCA1 Protein/genetics , Cell Cycle Proteins/metabolism , DNA Replication , Estrogens/metabolism , Genomic Instability , Minichromosome Maintenance Complex Component 2/metabolism , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Antineoplastic Agents, Immunological/pharmacology , Ataxia Telangiectasia Mutated Proteins/metabolism , Base Sequence , Binding Sites , Cell Cycle Checkpoints , Cell Line, Tumor , Estrogens/agonists , Female , Gene Expression Profiling , Gene Expression Regulation , Humans , Phosphorylation , Pregnancy , Promoter Regions, Genetic , Signal Transduction/drug effects
14.
Dis Colon Rectum ; 64(7): 833-850, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33709991

ABSTRACT

BACKGROUND: Patient-derived tumor organoid culture has emerged as a preclinical model that has the potential to predict individual drug response. However, the predictive accuracy of patient-derived tumor organoid culture models for responses to chemotherapy regimens in stage IV colorectal cancer remains unknown. OBJECTIVE: The purpose of this study was to evaluate the predictive accuracy of the patient-derived tumor organoid culture model for responses to chemotherapy regimens in stage IV colorectal cancer. DESIGN: A pilot study was performed to define the half-maximal inhibitory concentration of the response to chemotherapy regimens in the patient-derived tumor organoid culture model. Then, a blinded study was performed to evaluate the predictive accuracy of the patient-derived tumor organoid culture model for responses to chemotherapy regimens. SETTINGS: Cancer samples were collected from patients with stage IV colorectal cancer at Nanfang Hospital of Southern Medical University in China. PATIENTS: In the pilot study, 30 patients were enrolled, and 43 samples were collected. In the blinded study, 71 patients were enrolled, and 96 samples were collected. INTERVENTION: Patient-derived tumor organoid culture and chemotherapy regimens were tested. MAIN OUTCOME MEASURES: The predictive accuracy of the patient-derived tumor organoid model for responses to chemotherapy regimens was measured. RESULTS: The median (range) time of organoid culture and drug testing was 9 days (range, 7-14 d). In the pilot study, 30 samples (69.77% [30/43]) were successfully cultured. The half-maximal inhibitory concentration of the chemotherapy response was 10 µmol/L according to clinical chemotherapy outcomes. In the blinded study, 77 samples (80.21% [77/96]) from 57 patients were successfully cultured. The sensitivity, specificity, and accuracy of the patient-derived tumor organoid model for predicting responses to chemotherapy regimens were 63.33%, 94.12%, and 79.69%. LIMITATIONS: This was a blinded study rather than a prospective randomized controlled study. CONCLUSIONS: The patient-derived tumor organoid culture model effectively predicts responses to existing chemotherapy regimens for individual patients. Video Abstract at http://links.lww.com/DCR/B511. PRECISIN EN EL USO DE MODELOS DE CULTIVO DE ORGANOIDES TUMORALES DERIVADOS DE PACIENTES PARA PREDECIR LA RESPUESTA DEL RGIMEN DE QUIMIOTERAPIA EN CNCER COLORRECTAL ESTADIO IV ESTUDIO CIEGO: ANTECEDENTES:El cultivo de organoides tumorales derivado del paciente ha surgido como un modelo preclínico que tiene el potencial de predecir la respuesta a un fármaco individual. Sin embargo, la exactitud predictiva en los modelos de cultivo de organoides tumorales derivados de pacientes para las respuestas a los regímenes de quimioterapia en el cáncer colorrectal en estadio IV sigue siendo desconocida.OBJETIVO:Evaluar la exactitud predictiva del modelo de cultivo organoide tumoral derivado de pacientes para las respuestas a los regímenes de quimioterapia en el cáncer colorrectal en estadio IV.DISEÑO:Se realizó un estudio piloto para definir la concentración inhibitoria media máxima de la respuesta a los regímenes de quimioterapia en el modelo de cultivo organoide tumoral derivado de pacientes. Luego, se realizó un estudio ciego para evaluar la exactitud predictiva del modelo de cultivo organoide tumoral derivado de pacientes para las respuestas a los regímenes de quimioterapia.AJUSTE:Se recolectaron muestras de cáncer de pacientes con cáncer colorrectal en estadio IV en el Hospital Nanfang de la Universidad Médica del Sur en China.PACIENTES:En el estudio piloto, se inscribieron 30 pacientes y se recolectaron 43 muestras. En el estudio ciego, se inscribieron 71 pacientes y se recolectaron 96 muestras.INTERVENCIÓN:Se probaron cultivos de organoides de tumores derivados del paciente y regímenes de quimioterapia.PRINCIPALES MEDIDAS DE RESULTADO:La precisión predictiva del modelo organoide tumoral derivado del paciente para las respuestas a los regímenes de quimioterapia.RESULTADOS:La mediana (rango) de tiempo de cultivo organoide y prueba de drogas fue de 9 (7-14) días. En el estudio piloto, se cultivaron con éxito 30 (69,77% [30/43]) muestras. La concentración inhibidora media máxima de la respuesta a la quimioterapia fue de 10 µmol / L según los resultados de la quimioterapia clínica. En el estudio ciego, se cultivaron con éxito 77 muestras (80,21% [77/96]) de 57 pacientes. La sensibilidad, especificidad y precisión del modelo organoide tumoral derivado del paciente para predecir las respuestas a los regímenes de quimioterapia fueron 63,33%, 94,12% y 79,69%, respectivamente.LIMITACIONES:Este fue un estudio ciego en lugar de un estudio prospectivo, aleatorizado y controlado.CONCLUSIONES:El modelo de cultivo organoide tumoral derivado de pacientes predice eficazmente las respuestas a los regímenes de quimioterapia existentes para pacientes individuales. Consulte Video Resumen en http://links.lww.com/DCR/B511.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Organoids/drug effects , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , China/epidemiology , Culture Techniques/methods , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged , Neoplasm Staging/methods , Organoids/pathology , Pilot Projects , Predictive Value of Tests , Reproducibility of Results , Sensitivity and Specificity , Specimen Handling/methods , Treatment Outcome
15.
Int J Mol Sci ; 22(9)2021 Apr 30.
Article in English | MEDLINE | ID: mdl-33946224

ABSTRACT

Steroid receptor coactivator-3 (SRC-3), also known as amplified in breast cancer 1 (AIB1), is a member of the SRC family. SRC-3 regulates not only the transcriptional activity of nuclear receptors but also many other transcription factors. Besides the essential role of SRC-3 in physiological functions, it also acts as an oncogene to promote multiple aspects of cancer. This review updates the important progress of SRC-3 in carcinogenesis and summarizes its mode of action, which provides clues for cancer therapy.


Subject(s)
Neoplasms/metabolism , Nuclear Receptor Coactivator 3/metabolism , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/genetics , Neoplasms/pathology , Nuclear Receptor Coactivator 3/analysis , Nuclear Receptor Coactivator 3/genetics , Protein Processing, Post-Translational
16.
J Biol Chem ; 294(4): 1142-1151, 2019 01 25.
Article in English | MEDLINE | ID: mdl-30487289

ABSTRACT

CRISPR/Cas9 is a powerful technology widely used for genome editing, with the potential to be used for correcting a wide variety of deleterious disease-causing mutations. However, the technique tends to generate more indels (insertions and deletions) than precise modifications at the target sites, which might not resolve the mutation and could instead exacerbate the initial genetic disruption. We sought to develop an improved protocol for CRISPR/Cas9 that would correct mutations without unintended consequences. As a case study, we focused on achondroplasia, a common genetic form of dwarfism defined by missense mutation in the Fgfr3 gene that results in glycine to arginine substitution at position 374 in mice in fibroblast growth factor receptor 3 (Fgfr3-G374R), which corresponds to G380R in humans. First, we designed a GFP reporter system that can evaluate the cutting efficiency and specificity of single guide RNAs (sgRNAs). Using the sgRNA selected based on our GFP reporter system, we conducted targeted therapy of achondroplasia in mice. We found that we achieved higher frequency of precise correction of the Fgfr3-G374R mutation using Cas9 protein rather than Cas9 mRNA. We further demonstrated that targeting oligos of 100 and 200 nucleotides precisely corrected the mutation at equal efficiency. We showed that our strategy completely suppressed phenotypes of achondroplasia and whole genome sequencing detected no off-target effects. These data indicate that improved protocols can enable the precise CRISPR/Cas9-mediated correction of individual mutations with high fidelity.


Subject(s)
Achondroplasia/therapy , CRISPR-Cas Systems , Gene Targeting , Mutation , Receptor, Fibroblast Growth Factor, Type 3/physiology , Achondroplasia/genetics , Animals , Female , Gene Editing , Male , Mice , Mice, Knockout , Receptor, Fibroblast Growth Factor, Type 3/genetics
17.
Ann Rheum Dis ; 79(1): 112-122, 2020 01.
Article in English | MEDLINE | ID: mdl-31662319

ABSTRACT

OBJECTIVES: This study aims to investigate the role and mechanism of FGFR3 in macrophages and their biological effects on the pathology of arthritis. METHODS: Mice with conditional knockout of FGFR3 in myeloid cells (R3cKO) were generated. Gait behaviours of the mice were monitored at different ages. Spontaneous synovial joint destruction was evaluated by digital radiographic imaging and µCT analysis; changes of articular cartilage and synovitis were determined by histological analysis. The recruitment of macrophages in the synovium was examined by immunostaining and monocyte trafficking assay. RNA-seq analysis, Western blotting and chemotaxis experiment were performed on control and FGFR3-deficient macrophages. The peripheral blood from non-osteoarthritis (OA) donors and patients with OA were analysed. Mice were treated with neutralising antibody against CXCR7 to investigate the role of CXCR7 in arthritis. RESULTS: R3cKO mice but not control mice developed spontaneous cartilage destruction in multiple synovial joints at the age of 13 months. Moreover, the synovitis and macrophage accumulation were observed in the joints of 9-month-old R3cKO mice when the articular cartilage was not grossly destructed. FGFR3 deficiency in myeloid cells also aggravated joint destruction in DMM mouse model. Mechanically, FGFR3 deficiency promoted macrophage chemotaxis partly through activation of NF-κB/CXCR7 pathway. Inhibition of CXCR7 could significantly reverse FGFR3-deficiency-enhanced macrophage chemotaxis and the arthritic phenotype in R3cKO mice. CONCLUSIONS: Our study identifies the role of FGFR3 in synovial macrophage recruitment and synovitis, which provides a new insight into the pathological mechanisms of inflammation-related arthritis.


Subject(s)
Cartilage, Articular/pathology , Chemokine CXCL12/metabolism , Macrophages/metabolism , Osteoarthritis/genetics , Receptor, Fibroblast Growth Factor, Type 3/genetics , Receptors, CXCR/genetics , Synovitis/genetics , Animals , Chemotaxis/genetics , Gait , Gene Expression Regulation , Humans , Joints/metabolism , Joints/pathology , Mice , Mice, Knockout , Monocytes/metabolism , Myeloid Cells , NF-kappa B/metabolism , Osteoarthritis/metabolism , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Receptors, CXCR/metabolism , Synovial Membrane/metabolism , Synovial Membrane/pathology , Synovitis/pathology
18.
Nature ; 510(7506): 556-9, 2014 Jun 26.
Article in English | MEDLINE | ID: mdl-24776801

ABSTRACT

Replication fork stalling can promote genomic instability, predisposing to cancer and other diseases. Stalled replication forks may be processed by sister chromatid recombination (SCR), generating error-free or error-prone homologous recombination (HR) outcomes. In mammalian cells, a long-standing hypothesis proposes that the major hereditary breast/ovarian cancer predisposition gene products, BRCA1 and BRCA2, control HR/SCR at stalled replication forks. Although BRCA1 and BRCA2 affect replication fork processing, direct evidence that BRCA gene products regulate homologous recombination at stalled chromosomal replication forks is lacking, due to a dearth of tools for studying this process. Here we report that the Escherichia coli Tus/Ter complex can be engineered to induce site-specific replication fork stalling and chromosomal HR/SCR in mouse cells. Tus/Ter-induced homologous recombination entails processing of bidirectionally arrested forks. We find that the Brca1 carboxy (C)-terminal tandem BRCT repeat and regions of Brca1 encoded by exon 11-two Brca1 elements implicated in tumour suppression-control Tus/Ter-induced homologous recombination. Inactivation of either Brca1 or Brca2 increases the absolute frequency of 'long-tract' gene conversions at Tus/Ter-stalled forks, an outcome not observed in response to a site-specific endonuclease-mediated chromosomal double-strand break. Therefore, homologous recombination at stalled forks is regulated differently from homologous recombination at double-strand breaks arising independently of a replication fork. We propose that aberrant long-tract homologous recombination at stalled replication forks contributes to genomic instability and breast/ovarian cancer predisposition in BRCA mutant cells.


Subject(s)
BRCA1 Protein/metabolism , DNA Replication , Escherichia coli Proteins/metabolism , Homologous Recombination , Animals , BRCA1 Protein/chemistry , BRCA1 Protein/genetics , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , DNA Breaks, Double-Stranded , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Exons/genetics , Gene Conversion/genetics , Genomic Instability/genetics , Hereditary Breast and Ovarian Cancer Syndrome/genetics , Mice
19.
J Biol Chem ; 293(22): 8315-8329, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29666189

ABSTRACT

The mammary gland is very intricately and well organized into distinct tissues, including epithelia, endothelia, adipocytes, and stromal and immune cells. Many mammary gland diseases, such as breast cancer, arise from abnormalities in the mammary epithelium, which is mainly composed of two distinct lineages, the basal and luminal cells. Because of the limitation of traditional transcriptome analysis of bulk mammary cells, the hierarchy and heterogeneity of mammary cells within these two lineages remain unclear. To this end, using single-cell RNA-Seq coupled with FACS analysis and principal component analysis, we determined gene expression profiles of mammary epithelial cells of virgin and pregnant mice. These analyses revealed a much higher heterogeneity among the mammary cells than has been previously reported and enabled cell classification into distinct subgroups according to signature gene markers present in each group. We also identified and verified a rare CDH5+ cell subpopulation within a basal cell lineage as quiescent mammary stem cells (MaSCs). Moreover, using pseudo-temporal analysis, we reconstructed the developmental trajectory of mammary epithelia and uncovered distinct changes in gene expression and in biological functions of mammary cells along the developmental process. In conclusion, our work greatly refines the resolution of the cellular hierarchy in developing mammary tissues. The discovery of CDH5+ cells as MaSCs in these tissues may have implications for our understanding of the initiation, development, and pathogenesis of mammary tumors.


Subject(s)
Cell Lineage , Epithelial Cells/cytology , Epithelial Cells/metabolism , High-Throughput Nucleotide Sequencing/methods , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Single-Cell Analysis/methods , Animals , Biomarkers/metabolism , Cell Differentiation , Cells, Cultured , Female , Gene Expression Profiling , Mice , Stem Cells/cytology , Stem Cells/metabolism , Transcriptome
20.
Gastroenterology ; 154(1): 195-210, 2018 01.
Article in English | MEDLINE | ID: mdl-28918914

ABSTRACT

BACKGROUND & AIMS: Development of hepatocellular carcinoma (HCC) is associated with alterations in the transforming growth factor-beta (TGF-ß) signaling pathway, which regulates liver inflammation and can have tumor suppressor or promoter activities. Little is known about the roles of specific members of this pathway at specific of HCC development. We took an integrated approach to identify and validate the effects of changes in this pathway in HCC and identify therapeutic targets. METHODS: We performed transcriptome analyses for a total of 488 HCCs that include data from The Cancer Genome Atlas. We also screened 301 HCCs reported in the Catalogue of Somatic Mutations in Cancer and 202 from Cancer Genome Atlas for mutations in genome sequences. We expressed mutant forms of spectrin beta, non-erythrocytic 1 (SPTBN1) in HepG2, SNU398, and SNU475 cells and measured phosphorylation, nuclear translocation, and transcriptional activity of SMAD family member 3 (SMAD3). RESULTS: We found somatic mutations in at least 1 gene whose product is a member of TGF-ß signaling pathway in 38% of HCC samples. SPTBN1 was mutated in the largest proportion of samples (12 of 202, 6%). Unsupervised clustering of transcriptome data identified a group of HCCs with activation of the TGF-ß signaling pathway (increased transcription of genes in the pathway) and a group of HCCs with inactivation of TGF-ß signaling (reduced expression of genes in this pathway). Patients with tumors with inactivation of TGF-ß signaling had shorter survival times than patients with tumors with activation of TGF-ß signaling (P = .0129). Patterns of TGF-ß signaling correlated with activation of the DNA damage response and sirtuin signaling pathways. HepG2, SNU398, and SNU475 cells that expressed the D1089Y mutant or with knockdown of SPTBN1 had increased sensitivity to DNA crosslinking agents and reduced survival compared with cells that expressed normal SPTBN1 (controls). CONCLUSIONS: In genome and transcriptome analyses of HCC samples, we found mutations in genes in the TGF-ß signaling pathway in almost 40% of samples. These correlated with changes in expression of genes in the pathways; up-regulation of genes in this pathway would contribute to inflammation and fibrosis, whereas down-regulation would indicate loss of TGF-ß tumor suppressor activity. Our findings indicate that therapeutic agents for HCCs can be effective, based on genetic features of the TGF-ß pathway; agents that block TGF-ß should be used only in patients with specific types of HCCs.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Mutation/genetics , Signal Transduction/physiology , Transforming Growth Factor beta/physiology , Aged , Carcinoma, Hepatocellular/mortality , Case-Control Studies , Cluster Analysis , Female , Humans , Liver Neoplasms/mortality , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL