Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 101
Filter
Add more filters

Publication year range
1.
Cell ; 152(1-2): 340-51, 2013 Jan 17.
Article in English | MEDLINE | ID: mdl-23332765

ABSTRACT

Monitoring cancer and aging in vivo remains experimentally challenging. Here, we describe a luciferase knockin mouse (p16(LUC)), which faithfully reports expression of p16(INK4a), a tumor suppressor and aging biomarker. Lifelong assessment of luminescence in p16(+/LUC) mice revealed an exponential increase with aging, which was highly variable in a cohort of contemporaneously housed, syngeneic mice. Expression of p16(INK4a) with aging did not predict cancer development, suggesting that the accumulation of senescent cells is not a principal determinant of cancer-related death. In 14 of 14 tested tumor models, expression of p16(LUC) was focally activated by early neoplastic events, enabling visualization of tumors with sensitivity exceeding other imaging modalities. Activation of p16(INK4a) was noted in the emerging neoplasm and surrounding stromal cells. This work suggests that p16(INK4a) activation is a characteristic of all emerging cancers, making the p16(LUC) allele a sensitive, unbiased reporter of neoplastic transformation.


Subject(s)
Aging/genetics , Biomarkers , Cell Transformation, Neoplastic , Cyclin-Dependent Kinase Inhibitor p16/genetics , Luciferases/genetics , Neoplasms/genetics , Animals , Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Female , Gene Knock-In Techniques , Mice , Neoplasms/physiopathology , Wounds and Injuries/genetics
2.
Nature ; 569(7756): E4, 2019 May.
Article in English | MEDLINE | ID: mdl-31043737

ABSTRACT

Further analysis has revealed that the signal reported in Extended Data Fig. 1c of this Letter is attributed to phosphorylethanolamine, not carbamoyl phosphate. A newly developed derivatization method revealed that the level of carbamoyl phosphate in these NSCLC extracts is below the detection threshold of approximately 10 nanomoles. These findings do not alter the overall conclusions of the Letter; see associated Amendment for full details. The Letter has not been corrected online.

3.
Proc Natl Acad Sci U S A ; 119(40): e2201738119, 2022 10 04.
Article in English | MEDLINE | ID: mdl-36161943

ABSTRACT

Mismatch repair (MMR) is a replication-coupled DNA repair mechanism and plays multiple roles at the replication fork. The well-established MMR functions include correcting misincorporated nucleotides that have escaped the proofreading activity of DNA polymerases, recognizing nonmismatched DNA adducts, and triggering a DNA damage response. In an attempt to determine whether MMR regulates replication progression in cells expressing an ultramutable DNA polymerase ɛ (Polɛ), carrying a proline-to-arginine substitution at amino acid 286 (Polɛ-P286R), we identified an unusual MMR function in response to hydroxyurea (HU)-induced replication stress. Polɛ-P286R cells treated with hydroxyurea exhibit increased MRE11-catalyzed nascent strand degradation. This degradation by MRE11 depends on the mismatch recognition protein MutSα and its binding to stalled replication forks. Increased MutSα binding at replication forks is also associated with decreased loading of replication fork protection factors FANCD2 and BRCA1, suggesting blockage of these fork protection factors from loading to replication forks by MutSα. We find that the MutSα-dependent MRE11-catalyzed fork degradation induces DNA breaks and various chromosome abnormalities. Therefore, unlike the well-known MMR functions of ensuring replication fidelity, the newly identified MMR activity of promoting genome instability may also play a role in cancer avoidance by eliminating rogue cells.


Subject(s)
DNA-Binding Proteins , Hydroxyurea , Amino Acids/genetics , Arginine/genetics , DNA Adducts , DNA Mismatch Repair , DNA Replication , DNA-Binding Proteins/metabolism , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , Hydroxyurea/pharmacology , MRE11 Homologue Protein/genetics , MRE11 Homologue Protein/metabolism , Nucleotides/metabolism , Proline/genetics
4.
Mod Pathol ; 37(8): 100532, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38848896

ABSTRACT

Endometrial serous carcinoma (ESC) is an uncommon, aggressive type of endometrial cancer. While immune checkpoint blockade has emerged as a promising treatment option for endometrial carcinomas, research on the expression of immune checkpoints that could serve as prospective immunotherapy targets in ESC is limited. We examined the prevalence and prognostic value of lymphocyte-activation gene 3 (LAG-3), T-cell immunoglobulin and ITIM domain (TIGIT), V-domain immunoglobulin (Ig) suppressor of T-cell activation (VISTA), and indoleamine 2,3-dioxygenase 1 in 94 cases of ESC and correlated their expression with CD8+ and FOXP3+ tumor-infiltrating lymphocytes (TILs). We observed a positive correlation among LAG-3, TIGIT, and VISTA expressed on immune cells, and among these markers and CD8+ and FOXP3+ TIL densities. In Kaplan-Meier survival analysis, tumors with high levels of LAG-3 and TIGIT expression had better progression-free survival (PFS) and overall survival (OS) than those with lower levels of expression (LAG-3: PFS, P = .03, OS, P = .04; TIGIT: PFS, P = .01, OS, P = .009). In multivariate analysis, only high TIGIT expression was of independent prognostic value for better OS. VISTA expression in immune or tumor cells, and indoleamine 2,3-dioxygenase 1 expression in tumor cells, did not show a significant association with survival. Our data indicate that LAG-3, TIGIT, and VISTA immune checkpoints have roles in the microenvironment of ESC, and their expression patterns highlight the complex interactions among the different components of this system. High levels of these markers, together with high CD8+ TIL, suggest the potential immunogenicity of a subset of these tumors. Further studies are needed to elucidate the roles of various immune components in the ESC microenvironment and their association with intrinsic tumor properties.

5.
Nature ; 546(7656): 168-172, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28538732

ABSTRACT

Metabolic reprogramming by oncogenic signals promotes cancer initiation and progression. The oncogene KRAS and tumour suppressor STK11, which encodes the kinase LKB1, regulate metabolism and are frequently mutated in non-small-cell lung cancer (NSCLC). Concurrent occurrence of oncogenic KRAS and loss of LKB1 (KL) in cells specifies aggressive oncological behaviour. Here we show that human KL cells and tumours share metabolomic signatures of perturbed nitrogen handling. KL cells express the urea cycle enzyme carbamoyl phosphate synthetase-1 (CPS1), which produces carbamoyl phosphate in the mitochondria from ammonia and bicarbonate, initiating nitrogen disposal. Transcription of CPS1 is suppressed by LKB1 through AMPK, and CPS1 expression correlates inversely with LKB1 in human NSCLC. Silencing CPS1 in KL cells induces cell death and reduces tumour growth. Notably, cell death results from pyrimidine depletion rather than ammonia toxicity, as CPS1 enables an unconventional pathway of nitrogen flow from ammonia into pyrimidines. CPS1 loss reduces the pyrimidine to purine ratio, compromises S-phase progression and induces DNA-polymerase stalling and DNA damage. Exogenous pyrimidines reverse DNA damage and rescue growth. The data indicate that the KL oncological genotype imposes a metabolic vulnerability related to a dependence on a cross-compartmental pathway of pyrimidine metabolism in an aggressive subset of NSCLC.


Subject(s)
Carbamoyl-Phosphate Synthase (Ammonia)/metabolism , DNA/biosynthesis , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Pyrimidines/metabolism , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases/metabolism , Ammonia/metabolism , Animals , Bicarbonates/metabolism , Carbamoyl-Phosphate Synthase (Ammonia)/deficiency , Carbamoyl-Phosphate Synthase (Ammonia)/genetics , Carbamyl Phosphate/metabolism , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Death , Cell Proliferation , DNA Damage/drug effects , DNA Replication , DNA-Directed DNA Polymerase/metabolism , Female , Gene Silencing , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Male , Metabolomics , Mice , Mitochondria/metabolism , Nitrogen/metabolism , Protein Serine-Threonine Kinases/metabolism , Purines/metabolism , Pyrimidines/pharmacology , S Phase , Transcription, Genetic , Xenograft Model Antitumor Assays
6.
Genes Dev ; 29(16): 1707-20, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26302789

ABSTRACT

Inactivation of phosphatase and tensin homology deleted on chromosome 10 (PTEN) is linked to increased PI3K-AKT signaling, enhanced organismal growth, and cancer development. Here we generated and analyzed Pten knock-in mice harboring a C2 domain missense mutation at phenylalanine 341 (Pten(FV)), found in human cancer. Despite having reduced levels of PTEN protein, homozygous Pten(FV/FV) embryos have intact AKT signaling, develop normally, and are carried to term. Heterozygous Pten(FV/+) mice develop carcinoma in the thymus, stomach, adrenal medulla, and mammary gland but not in other organs typically sensitive to Pten deficiency, including the thyroid, prostate, and uterus. Progression to carcinoma in sensitive organs ensues in the absence of overt AKT activation. Carcinoma in the uterus, a cancer-resistant organ, requires a second clonal event associated with the spontaneous activation of AKT and downstream signaling. In summary, this PTEN noncatalytic missense mutation exposes a core tumor suppressor function distinct from inhibition of canonical AKT signaling that predisposes to organ-selective cancer development in vivo.


Subject(s)
Carcinoma/genetics , Mutation, Missense/genetics , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Signal Transduction , Animals , Carcinoma/enzymology , Carcinoma/physiopathology , Cell Nucleus/metabolism , Cells, Cultured , Embryo, Mammalian , Enzyme Activation , Female , Gene Knock-In Techniques , Mice , Oncogene Protein v-akt/genetics , Oncogene Protein v-akt/metabolism , Protein Stability
7.
Mod Pathol ; 35(12): 1955-1965, 2022 12.
Article in English | MEDLINE | ID: mdl-35804040

ABSTRACT

Endometrial serous carcinoma (ESC) is an aggressive type of endometrial carcinoma with a poor prognosis. Immune checkpoint blockade has evolved as a novel treatment option for endometrial cancers; however, data on expression of immune checkpoints that may be potential targets for immunotherapy in ESC are limited. We analyzed the prevalence and prognostic significance of PD-L1, TIM-3 and B7-H3 immune checkpoints in 99 ESC and evaluated their correlation with CD8 + tumor infiltrating lymphocytes. Applying the tumor proportion score (TPS) with a cutoff of 1%, PD-L1, TIM-3 and B7-H3 expression was present in 17%, 10% and 93% of cases, respectively. Applying the combined positive score (CPS) with a cutoff of 1, PD-L1, TIM-3 and B7-H3 expression was present in 63%, 67% and 94% of cases, respectively. Expression of these markers was largely independent of one another. PD-L1 correlated with higher CD8 + T-cell density when evaluated by either TPS (p = 0.02) or CPS (p < 0.0001). TIM-3 correlated with CD8 + T-cell density when evaluated by CPS (p < 0.0001). PD-L1 positivity was associated with improved overall survival (p = 0.038) when applying CPS. No association between PD-L1 expression and survival was found using TPS, and there was no association between TIM-3 or B7-H3 positivity and survival by either TPS or CPS. Using TPS, PD-L1 correlated with a higher tumor stage but not with survival, whereas the converse was true when PD-L1 was evaluated by CPS, suggesting that PD-L1 expression in immune cells correlates with prognosis and is independent of tumor stage. In conclusion, PD-L1, TIM-3 and B7-H3 may be potential therapeutic targets in selected patients with ESC. Further investigation of their roles as predictive biomarkers is needed.


Subject(s)
Cystadenocarcinoma, Serous , Endometrial Neoplasms , Female , Humans , B7-H1 Antigen/metabolism , Prognosis , Hepatitis A Virus Cellular Receptor 2/metabolism , Prevalence , Biomarkers, Tumor/metabolism , Lymphocytes, Tumor-Infiltrating , Endometrial Neoplasms/pathology , Cystadenocarcinoma, Serous/pathology
8.
Mod Pathol ; 35(11): 1702-1712, 2022 11.
Article in English | MEDLINE | ID: mdl-35798968

ABSTRACT

Endometrial polyps (EMPs) are common exophytic masses associated with abnormal uterine bleeding and infertility. Unlike normal endometrium, which is cyclically shed, EMPs persist over ovulatory cycles and after the menopause. Despite their usual classification as benign entities, EMPs are paradoxically associated with endometrial carcinomas of diverse histologic subtypes, which frequently arise within EMPs. The etiology and potential origins of EMPs as clonally-derived neoplasms are uncertain, but previous investigations suggested that EMPs are neoplasms of stromal origin driven by recurring chromosomal rearrangements. To better define benign EMPs at the molecular genetic level, we analyzed individual EMPs from 31 women who underwent hysterectomy for benign indications. The 31 EMPs were subjected to comprehensive genomic profiling by exome sequencing of a large panel of tumor-related genes including oncogenes, tumor suppressors, and chromosomal translocation partners. There were no recurring chromosomal rearrangements, and copy-number analyses did not reveal evidence of significant chromosome-level events. Surprisingly, there was a high incidence of single nucleotide variants corresponding to classic oncogenic drivers (i.e., definitive cancer drivers). The spectrum of known oncogenic driver events matched that of endometrial cancers more closely than any other common cancer. Further analyses including laser-capture microdissection showed that these mutations were present in the epithelial compartment at low allelic frequencies. These results establish a link between EMPs and the acquisition of endometrial cancer driver mutations. Based on these findings, we propose a model where the association between EMPs and endometrial cancer is explained by the age-related accumulation of endometrial cancer drivers in a protected environment that-unlike normal endometrium-is not subject to cyclical shedding.


Subject(s)
Endometrial Neoplasms , Polyps , Uterine Neoplasms , Female , Humans , Neoplasm Recurrence, Local/pathology , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Polyps/genetics , Polyps/pathology , Uterine Neoplasms/pathology , Mutation , Carcinogenesis/pathology , Nucleotides , Endometrium/pathology
9.
Int J Gynecol Pathol ; 41(1): 45-50, 2022 Jan 01.
Article in English | MEDLINE | ID: mdl-33900230

ABSTRACT

Complete hydatidiform mole (CHM) is a premalignant proliferative disease of the placenta characterized by misexpression of imprinted gene products, most notably p57. The majority of CHM exhibit immunohistochemical absence of p57 protein in villous mesenchyme (VM) and cytotrophoblast (CT) and are thus p57 VM/CT concordant. However, some gestations show loss of p57 in only VM or CT, either in all chorionic villi or a subset thereof (VM/CT discordant). Here, we present a rare case of a p57 VM/CT-discordant CHM with diffuse retention of p57 expression in VM but complete absence in CT. Histologically, the case exhibited typical features of CHM including trophoblast hyperplasia and severe nuclear atypia, but was unusual in the presence of gestational membranes identified ultrasonographically and histologically. Ploidy determination by FISH and genotyping by short tandem repeat analyses showed that this was a diploid gestation with variable allelic ratios and with an androgenetic lineage, similar to previously reported p57 VM/CT-discordant cases.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p57/metabolism , Hydatidiform Mole/diagnostic imaging , Uterine Neoplasms/diagnostic imaging , Adult , Chorionic Villi/diagnostic imaging , Chorionic Villi/pathology , Cyclin-Dependent Kinase Inhibitor p57/genetics , Female , Genotyping Techniques , Humans , Hydatidiform Mole/pathology , Immunohistochemistry , Mesoderm/diagnostic imaging , Mesoderm/pathology , Placenta/diagnostic imaging , Placenta/pathology , Pregnancy , Trophoblasts/pathology , Uterine Neoplasms/pathology
10.
J Pathol ; 254(1): 20-30, 2021 05.
Article in English | MEDLINE | ID: mdl-33506979

ABSTRACT

The endometrium is unique as an accessible anatomic location that can be repeatedly biopsied and where diagnostic biopsies do not extirpate neoplastic lesions. We exploited these features to retrospectively characterize serial genomic alterations along the precancer/cancer continuum in individual women. Cases were selected based on (1) endometrial cancer diagnosis/hysterectomy and (2) preceding serial endometrial biopsies including for some patients an early biopsy before a precancer histologic diagnosis. A comprehensive panel was designed for endometrial cancer genes. Formalin-fixed, paraffin-embedded specimens for each cancer, preceding biopsies, and matched germline samples were subjected to barcoded high-throughput sequencing to identify mutations and track their origin and allelic frequency progression. In total, 92 samples from 21 patients were analyzed, providing an opportunity for new insights into early endometrial cancer progression. Definitive invasive endometrial cancers exhibited expected mutational spectra, and canonical driver mutations were detectable in preceding biopsies. Notably, ≥1 cancer mutations were detected prior to the histopathologic diagnosis of an endometrial precancer in the majority of patients. In 18/21 cases, ≥1 mutations were confirmed by abnormal protein levels or subcellular localization by immunohistochemistry, confirming genomic data and providing unique views of histologic correlates. In 19 control endometria, mutation counts were lower, with a lack of canonical endometrial cancer hotspot mutations. Our study documents the existence of endometrial lesions that are histologically indistinct but are bona fide endometrial cancer precursors. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Endometrium/pathology , Precancerous Conditions/genetics , Precancerous Conditions/pathology , Adult , Aged , DNA Mutational Analysis/methods , Female , High-Throughput Nucleotide Sequencing/methods , Humans , Middle Aged , Retrospective Studies , Sequence Analysis, DNA/methods
11.
Semin Diagn Pathol ; 39(3): 137-147, 2022 May.
Article in English | MEDLINE | ID: mdl-34920905

ABSTRACT

Early detection of endometrial cancer, especially its precancers, remains a critical and evolving issue in patient management and the quest to decrease mortality due to endometrial cancer. Due to many factors such as specimen fragmentation, the confounding influence of endogenous or exogenous hormones, and variable or overlapping histologic features, identification of bona fide endometrial precancers and their reliable discrimination from benign mimics remains one of the most challenging areas in diagnostic pathology. At the same time, the diagnosis of endometrial precancer, or the presence of suspicious but subdiagnostic features in an endometrial biopsy, can lead to long clinical follow-up with multiple patient visits and serial endometrial sampling, emphasizing the need for accurate diagnosis. Our understanding of endometrial precancers and their diagnosis has improved due to systematic investigations into morphologic criteria, the molecular genetics of endometrial cancer and their precursors, the validation of novel biomarkers and their use in panels, and more recent methods such digital image analysis. Although precancers for both endometrioid and non-endometrioid carcinomas will be reviewed, emphasis will be placed on the former. We review these advances and their relevance to the histopathologic diagnosis of endometrial precancers, and the recently updated 2020 World Health Organization (WHO) Classification of Female Genital Tumors.


Subject(s)
Endometrial Hyperplasia , Endometrial Neoplasms , Precancerous Conditions , Biomarkers , Biopsy , Endometrial Hyperplasia/diagnosis , Endometrial Hyperplasia/pathology , Endometrial Neoplasms/diagnosis , Endometrial Neoplasms/pathology , Female , Humans , PTEN Phosphohydrolase , Precancerous Conditions/diagnosis , Precancerous Conditions/pathology
12.
Proc Natl Acad Sci U S A ; 116(51): 25880-25890, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31772025

ABSTRACT

Uterine carcinosarcoma is an aggressive variant of endometrial carcinoma characterized by unusual histologic features including discrete malignant epithelial and mesenchymal components (carcinoma and sarcoma). Recent studies have confirmed a monoclonal origin, and comprehensive genomic characterizations have identified mutations such as Tp53 and Pten However, the biological origins and specific combination of driver events underpinning uterine carcinosarcoma have remained mysterious. Here, we explored the role of the tumor suppressor Fbxw7 in endometrial cancer through defined genetic model systems. Inactivation of Fbxw7 and Pten resulted in the formation of precancerous lesions (endometrioid intraepithelial neoplasia) and well-differentiated endometrioid adenocarcinomas. Surprisingly, all adenocarcinomas eventually developed into definitive uterine carcinosarcomas with carcinomatous and sarcomatous elements including heterologous differentiation, yielding a faithful genetically engineered model of this cancer type. Genomic analysis showed that most tumors spontaneously acquired Trp53 mutations, pointing to a triad of pathways (p53, PI3K, and Fbxw7) as the critical combination underpinning uterine carcinosarcoma, and to Fbxw7 as a key driver of this enigmatic endometrial cancer type. Lineage tracing provided formal genetic proof that the uterine carcinosarcoma cell of origin is an endometrial epithelial cell that subsequently undergoes a prominent epithelial-mesenchymal transition underlying the attainment of a highly invasive phenotype specifically driven by Fbxw7.


Subject(s)
Endometrial Neoplasms/metabolism , Epithelial-Mesenchymal Transition/physiology , F-Box-WD Repeat-Containing Protein 7/genetics , F-Box-WD Repeat-Containing Protein 7/metabolism , Uterine Neoplasms/metabolism , Animals , Cell Movement , Cell Proliferation , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Endometrium/pathology , Epithelial Cells , Female , Humans , Mice , Mutation , PTEN Phosphohydrolase/metabolism , Phenotype , Tumor Suppressor Protein p53/metabolism , Uterine Neoplasms/genetics , Uterine Neoplasms/pathology
13.
Nat Immunol ; 10(2): 176-84, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19136962

ABSTRACT

Foxo transcription factors have a conserved role in the adaptation of cells and organisms to nutrient and growth factor availability. Here we show that Foxo1 has a crucial, nonredundant role in T cells. In naive T cells, Foxo1 controlled the expression of the adhesion molecule L-selectin, the chemokine receptor CCR7 and the transcription factor Klf2, and its deletion was sufficient to alter lymphocyte trafficking. Furthermore, Foxo1 deficiency resulted in a severe defect in interleukin 7 receptor alpha-chain (IL-7Ralpha) expression associated with its ability to bind an Il7r enhancer. Finally, growth factor withdrawal induced a Foxo1-dependent increase in Sell, Klf2 and Il7r expression. These data suggest that Foxo1 regulates the homeostasis and life span of naive T cells by sensing growth factor availability and regulating homing and survival signals.


Subject(s)
Chemotaxis, Leukocyte/immunology , Forkhead Transcription Factors/metabolism , L-Selectin/biosynthesis , Receptors, CCR7/biosynthesis , Receptors, Interleukin-7/biosynthesis , T-Lymphocytes/metabolism , Animals , Blotting, Western , Cell Differentiation/immunology , Cell Survival , Flow Cytometry , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Gene Expression , Gene Expression Regulation/immunology , Homeostasis/immunology , Immunoprecipitation , L-Selectin/immunology , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , RNA, Messenger/analysis , Receptors, CCR7/immunology , Receptors, Interleukin-7/immunology , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/cytology , T-Lymphocytes/immunology
14.
Nat Immunol ; 10(5): 504-13, 2009 May.
Article in English | MEDLINE | ID: mdl-19363483

ABSTRACT

Foxo transcription factors regulate cell cycle progression, cell survival and DNA-repair pathways. Here we demonstrate that deficiency in Foxo3 resulted in greater expansion of T cell populations after viral infection. This exaggerated expansion was not T cell intrinsic. Instead, it was caused by the enhanced capacity of Foxo3-deficient dendritic cells to sustain T cell viability by producing more interleukin 6. Stimulation of dendritic cells mediated by the coinhibitory molecule CTLA-4 induced nuclear localization of Foxo3, which in turn inhibited the production of interleukin 6 and tumor necrosis factor. Thus, Foxo3 acts to constrain the production of key inflammatory cytokines by dendritic cells and to control T cell survival.


Subject(s)
Dendritic Cells/immunology , Forkhead Transcription Factors/immunology , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Animals , Antigen Presentation/immunology , Antigens, CD/immunology , Antigens, CD/metabolism , Arenaviridae Infections/immunology , Blotting, Western , CTLA-4 Antigen , Dendritic Cells/metabolism , Flow Cytometry , Forkhead Box Protein O3 , Forkhead Transcription Factors/metabolism , Interleukin-6/immunology , Interleukin-6/metabolism , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Congenic , Mice, Transgenic , Protein Transport/immunology , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/metabolism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
15.
Hum Mol Genet ; 27(7): 1136-1149, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29360988

ABSTRACT

The continuity of life depends on mechanisms in the germline that ensure the integrity of the genome. The DNA damage response/checkpoint kinases ATM and ATR are essential signaling factors in the germline. However, it remains unknown how a downstream transducer, Checkpoint Kinase 1 (CHEK1 or CHK1), mediates signaling in the male germline. Here, we show that CHEK1 has distinct functions in both the mitotic and meiotic phases of the male germline in mice. In the mitotic phase, CHEK1 is required for the resumption of prospermatogonia proliferation after birth and the maintenance of spermatogonia. In the meiotic phase, we uncovered two functions for CHEK1: one is the stage-specific attenuation of DNA damage signaling on autosomes, and the other is coordination of meiotic stage progression. On autosomes, the loss of CHEK1 delays the removal of DNA damage signaling that manifests as phosphorylation of histone variant H2AX at serine 139 (γH2AX). Importantly, CHEK1 does not have a direct function in meiotic sex chromosome inactivation (MSCI), an essential event in male meiosis, in which ATR is a key regulator. Thus, the functions of ATR and CHEK1 are uncoupled in MSCI, in contrast to their roles in DNA damage signaling in somatic cells. Our study reveals stage-specific functions for CHEK1 that ensure the integrity of the male germline.


Subject(s)
Checkpoint Kinase 1/metabolism , DNA Damage , Meiosis , Signal Transduction , Spermatogonia/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Checkpoint Kinase 1/genetics , Male , Mice , Mice, Knockout , Spermatogonia/cytology
16.
Nat Immunol ; 9(12): 1388-98, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18978794

ABSTRACT

The transcription factors Foxo1, Foxo3 and Foxo4 modulate cell fate 'decisions' in diverse systems. Here we show that Foxo1-dependent gene expression was critical at many stages of B cell differentiation. Early deletion of Foxo1 caused a substantial block at the pro-B cell stage due to a failure to express interleukin 7 receptor-alpha. Foxo1 inactivation in late pro-B cells resulted in an arrest at the pre-B cell stage due to lower expression of the recombination-activating genes Rag1 and Rag2. Deletion of Foxo1 in peripheral B cells led to fewer lymph node B cells due to lower expression of L-selectin and failed class-switch recombination due to impaired upregulation of the gene encoding activation-induced cytidine deaminase. Thus, Foxo1 regulates a transcriptional program that is essential for early B cell development and peripheral B cell function.


Subject(s)
B-Lymphocytes/cytology , Cell Differentiation/immunology , Forkhead Transcription Factors/immunology , Animals , B-Lymphocytes/immunology , Blotting, Southern , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression/immunology , Gene Rearrangement, B-Lymphocyte/genetics , Homeodomain Proteins/immunology , Homeodomain Proteins/metabolism , Immunohistochemistry , Mice , Mice, Mutant Strains , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/immunology , Stem Cells/metabolism , Transcription, Genetic/immunology
17.
J Immunol ; 201(3): 940-949, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29950509

ABSTRACT

Central tolerance checkpoints are critical for the elimination of autoreactive B cells and the prevention of autoimmunity. When autoreactive B cells encounter their Ag at the immature B cell stage, BCR cross-linking induces receptor editing, followed by apoptosis if edited cells remain autoreactive. Although the transcription factor Foxo1 is known to promote receptor editing, the role of the related factor Foxo3 in central B cell tolerance is poorly understood. We find that BCR-stimulated immature B cells from Foxo3-deficient mice demonstrate reduced apoptosis compared with wild type cells. Despite this, Foxo3-/- mice do not develop increased autoantibodies. This suggests that the increased survival of Foxo3-/- immature B cells allows additional rounds of receptor editing, resulting in more cells "redeeming" themselves by becoming nonautoreactive. Indeed, increased Igλ usage and increased recombining sequence recombination among Igλ-expressing cells were observed in Foxo3-/- mice, indicative of increased receptor editing. We also observed that deletion of high-affinity autoreactive cells was intact in the absence of Foxo3 in the anti-hen egg lysozyme (HEL)/membrane-bound HEL model. However, Foxo3 levels in B cells from systemic lupus erythematosus (SLE) patients were inversely correlated with disease activity and reduced in patients with elevated anti-dsDNA Abs. Although this is likely due in part to increased B cell activation in these SLE patients, it is also possible that low-affinity B cells that remain autoreactive after editing may survive inappropriately in the absence of Foxo3 and become activated to secrete autoantibodies in the context of other SLE-associated defects.


Subject(s)
Apoptosis/immunology , B-Lymphocytes/immunology , Forkhead Box Protein O3/immunology , Receptors, Antigen, B-Cell/immunology , Animals , Autoantibodies/immunology , Autoantigens/immunology , Autoimmunity/immunology , Cell Differentiation/immunology , Female , Immune Tolerance/immunology , Lupus Erythematosus, Systemic/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Precursor Cells, B-Lymphoid/immunology
18.
Int J Gynecol Pathol ; 38(6): 503-513, 2019 Nov.
Article in English | MEDLINE | ID: mdl-30256235

ABSTRACT

The diagnosis of endometrioid intraepithelial neoplasia (EIN) is challenging owing to limited sampling, hormonal status, and other confounding histologic variables. Markers such as PTEN or PAX2 can delineate EIN in some cases, but are not wholly reliable. Clearly, new markers of EIN are needed. We explored several potential markers of EIN based rationally on molecular pathways most frequently misregulated in endometrial cancer: the 3-phosphoinositide kinase (PI3K)/AKT, ß-catenin, and mismatch repair pathways. We studied PTEN, PAX2, ß-catenin, and MLH1, in conjunction with 2 new markers-FOXO1 and phosphorylated AKT (pAKT)-not previously investigated in EIN. Benign (n=14) and EIN (n=35) endometria were analyzed by immunohistochemistry. Staining patterns were interpreted, tabulated, and scored by "clonal distinctiveness" in neoplastic lesions; that is, pattern alterations relative to normal glands. In normal endometria, FOXO1 was cytoplasmic in proliferative phase, but nuclear in secretory phase, showing that PI3K/FOXO1 participates in endometrial cycling and that FOXO1 is a readout of PI3K status. pAKT expression was low across normal endometria. FOXO1 or pAKT expression was altered in the majority of EINs (27/35, 77%), with FOXO1 and pAKT being co-altered only in some (20/35, 57%). ß-catenin or MLH1 also exhibited clonal distinctiveness in EINs, showing that these are also useful markers in some cases. This is the first study to demonstrate the potential of pAKT and FOXO1 as biomarkers in the histopathologic evaluation of EIN. However, variability in expression poses challenges in interpretation.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma in Situ/metabolism , Endometrial Hyperplasia/metabolism , Endometrial Neoplasms/metabolism , Forkhead Box Protein O1/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Carcinoma in Situ/pathology , Endometrial Hyperplasia/pathology , Endometrial Neoplasms/pathology , Endometrium/metabolism , Endometrium/pathology , Female , Humans , Immunohistochemistry , Mice , Mice, Knockout , PAX2 Transcription Factor/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphorylation , Signal Transduction , beta Catenin/metabolism
19.
Int J Gynecol Pathol ; 38(6): 533-542, 2019 Nov.
Article in English | MEDLINE | ID: mdl-30383610

ABSTRACT

Screening for Lynch syndrome (LS) is routinely performed in patients with endometrial carcinoma. Currently, no screening recommendations exist for LS in precancerous lesions. The study goal was to determine the incidence of abnormal protein expression in endometrioid intraepithelial neoplasia/atypical hyperplasia (EIN/AH). We analyzed mismatch repair (MMR) protein expression by immunohistochemistry in EIN/AH concurrent with MMR-deficient endometrial carcinomas, and in endometrial biopsy/curettage specimens with EIN/AH from an unselected group of patients. Of 63 patients with MMR-deficient endometrial carcinoma, 34 demonstrated loss of MLH1/PMS2 expression; 1 showed loss of PMS2 alone; 12 showed loss of MSH2/MSH6, and 15 had loss of MSH6 alone. Genetic testing identified deleterious mutations in 14 cases (LS). 15 tumors demonstrated MLH1 promoter hypermethylation. Abnormal MMR expression in EIN/AH and adjacent carcinoma was concordant in 100% of LS cases and 71% of MLH1 promoter hypermethylation cases. Of 118 patients from the unselected group with EIN/AH, 4 (3%) cases demonstrated absent expression of one or more MMR proteins. Of these, 2 patients were later confirmed to have deleterious mutations in subsequent specimens with endometrial carcinoma. The prevalence of abnormal MMR expression in EIN/AH adjacent to carcinoma and in the unselected group of patients with EIN/AH is similar to the reported prevalence of LS in endometrial carcinoma. Identifying patients at high risk for LS through abnormal MMR expression in EIN/AH provides the benefit of early surveillance, treatment and timely diagnosis for the patient and affected family members.


Subject(s)
Carcinoma in Situ/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , DNA Mismatch Repair/genetics , Endometrial Hyperplasia/genetics , Endometrial Neoplasms/genetics , MutL Protein Homolog 1/genetics , Precancerous Conditions/genetics , Adult , Carcinoma in Situ/pathology , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , DNA Methylation , DNA-Binding Proteins/genetics , Endometrial Hyperplasia/pathology , Endometrial Neoplasms/pathology , Female , Humans , Immunohistochemistry , Middle Aged , Mismatch Repair Endonuclease PMS2/genetics , Precancerous Conditions/pathology , Promoter Regions, Genetic/genetics
20.
PLoS Genet ; 12(8): e1006215, 2016 08.
Article in English | MEDLINE | ID: mdl-27500836

ABSTRACT

In mammals, females are born with finite numbers of oocytes stockpiled as primordial follicles. Oocytes are "reawakened" via an ovarian-intrinsic process that initiates their growth. The forkhead transcription factor Foxo3 controls reawakening downstream of PI3K-AKT signaling. However, the identity of the presumptive upstream cell surface receptor controlling the PI3K-AKT-Foxo3 axis has been questioned. Here we show that the receptor tyrosine kinase Kit controls reawakening. Oocyte-specific expression of a novel constitutively-active KitD818V allele resulted in female sterility and ovarian failure due to global oocyte reawakening. To confirm this result, we engineered a novel loss-of-function allele, KitL. Kit inactivation within oocytes also led to premature ovarian failure, albeit via a contrasting phenotype. Despite normal initial complements of primordial follicles, oocytes remained dormant with arrested oocyte maturation. Foxo3 protein localization in the nucleus versus cytoplasm explained both mutant phenotypes. These genetic studies provide formal genetic proof that Kit controls oocyte reawakening, focusing future investigations into the causes of primary ovarian insufficiency and ovarian aging.


Subject(s)
Forkhead Box Protein O3/genetics , Phosphatidylinositol 3-Kinases/genetics , Primary Ovarian Insufficiency/genetics , Proto-Oncogene Proteins c-kit/genetics , Animals , Cell Nucleus/genetics , Cytoplasm/genetics , Female , Forkhead Box Protein O3/biosynthesis , Gene Expression Regulation, Developmental , Mice , Mice, Knockout , Oocytes/growth & development , Oocytes/metabolism , Ovarian Follicle/growth & development , Ovarian Follicle/metabolism , Ovary/growth & development , Ovary/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Primary Ovarian Insufficiency/pathology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL