Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Cells ; 13(9)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38727270

ABSTRACT

Self-renewal and differentiation are two characteristics of hematopoietic stem cells (HSCs). Under steady physiological conditions, most primitive HSCs remain quiescent in the bone marrow (BM). They respond to different stimuli to refresh the blood system. The transition from quiescence to activation is accompanied by major changes in metabolism, a fundamental cellular process in living organisms that produces or consumes energy. Cellular metabolism is now considered to be a key regulator of HSC maintenance. Interestingly, HSCs possess a distinct metabolic profile with a preference for glycolysis rather than oxidative phosphorylation (OXPHOS) for energy production. Byproducts from the cellular metabolism can also damage DNA. To counteract such insults, mammalian cells have evolved a complex and efficient DNA damage repair (DDR) system to eliminate various DNA lesions and guard genomic stability. Given the enormous regenerative potential coupled with the lifetime persistence of HSCs, tight control of HSC genome stability is essential. The intersection of DDR and the HSC metabolism has recently emerged as an area of intense research interest, unraveling the profound connections between genomic stability and cellular energetics. In this brief review, we delve into the interplay between DDR deficiency and the metabolic reprogramming of HSCs, shedding light on the dynamic relationship that governs the fate and functionality of these remarkable stem cells. Understanding the crosstalk between DDR and the cellular metabolism will open a new avenue of research designed to target these interacting pathways for improving HSC function and treating hematologic disorders.


Subject(s)
DNA Damage , DNA Repair , Hematopoietic Stem Cells , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/cytology , Humans , Animals , Genomic Instability , Energy Metabolism , Oxidative Phosphorylation
2.
Sci Rep ; 13(1): 18962, 2023 11 03.
Article in English | MEDLINE | ID: mdl-37923775

ABSTRACT

Host immunity involves various immune cells working in concert to achieve balanced immune response. Host immunity interacts with tumorigenic process impacting disease outcome. Clusters of different immune cells may reveal unique host immunity in relation to breast cancer progression. CIBERSORT algorithm was used to estimate relative abundances of 22 immune cell types in 3 datasets, METABRIC, TCGA, and our study. The cell type data in METABRIC were analyzed for cluster using unsupervised hierarchical clustering (UHC). The UHC results were employed to train machine learning models. Kaplan-Meier and Cox regression survival analyses were performed to assess cell clusters in association with relapse-free and overall survival. Differentially expressed genes by clusters were interrogated with IPA for molecular signatures. UHC analysis identified two distinct immune cell clusters, clusters A (83.2%) and B (16.8%). Memory B cells, plasma cells, CD8 positive T cells, resting memory CD4 T cells, activated NK cells, monocytes, M1 macrophages, and resting mast cells were more abundant in clusters A than B, whereas regulatory T cells and M0 and M2 macrophages were more in clusters B than A. Patients in cluster A had favorable survival. Similar survival associations were also observed in other independent studies. IPA analysis showed that pathogen-induced cytokine storm signaling pathway, phagosome formation, and T cell receptor signaling were related to the cell type clusters. Our finding suggests that different immune cell clusters may indicate distinct immune responses to tumor growth, suggesting their potential for disease management.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/genetics , Neoplasm Recurrence, Local , Survival Analysis , Cluster Analysis , Machine Learning
3.
Int J Mol Sci ; 22(23)2021 Nov 30.
Article in English | MEDLINE | ID: mdl-34884777

ABSTRACT

Among human genetic diseases, Fanconi Anemia (FA) tops all with its largest number of health complications in nearly all human organ systems, suggesting the significant roles played by FA genes in the maintenance of human health. With the accumulated research on FA, the encoded protein products by FA genes have been building up to the biggest cell defense signaling network, composed of not only 22+ FA proteins but also ATM, ATR, and many other non-FA proteins. The FA D2 group protein (FANCD2) and its paralog form the focal point of FA signaling to converge the effects of its upstream players in response to a variety of cellular insults and simultaneously with downstream players to protect humans from contracting diseases, including aging and cancer. In this review, we update and discuss how the FA signaling crucially eases cellular stresses through understanding its focal point.


Subject(s)
DNA Damage/genetics , DNA Repair/genetics , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group Proteins/genetics , Fanconi Anemia/genetics , DNA Replication/genetics , Fanconi Anemia/pathology , Fanconi Anemia Complementation Group D2 Protein/metabolism , Fanconi Anemia Complementation Group Proteins/metabolism , Female , Genomic Instability/genetics , Humans , Male , Signal Transduction/genetics
4.
Front Cell Dev Biol ; 9: 754507, 2021.
Article in English | MEDLINE | ID: mdl-34722540

ABSTRACT

Extrinsic factors such as expression of PD-L1 (programmed dealth-ligand 1) in the tumor microenvironment (TME) have been shown to correlate with responses to checkpoint blockade therapy. More recently two intrinsic factors related to tumor genetics, microsatellite instability (MSI), and tumor mutation burden (TMB), have been linked to high response rates to checkpoint blockade drugs. These response rates led to the first tissue-agnostic approval of any cancer therapy by the FDA for the treatment of metastatic, MSI-H tumors with anti-PD-1 immunotherapy. But there are still very few studies focusing on the association of miRNAs with immune therapy through checkpoint inhibitors. Our team sought to explore the biology of such tumors further and suggest potential companion therapeutics to current checkpoint inhibitors. Analysis by Pearson Correlation revealed 41 total miRNAs correlated with mutation burden, 62 miRNAs correlated with MSI, and 17 miRNAs correlated with PD-L1 expression. Three miRNAs were correlated with all three of these tumor features as well as M1 macrophage polarization. No miRNAs in any group were associated with overall survival. TGF-ß was predicted to be influenced by these three miRNAs (p = 0.008). Exploring miRNA targets as companions to treatment by immune checkpoint blockade revealed three potential miRNA targets predicted to impact TGF-ß. M1 macrophage polarization state was also associated with tumors predicted to respond to therapy by immune checkpoint blockade.

5.
Aging (Albany NY) ; 12(20): 20268-20284, 2020 10 24.
Article in English | MEDLINE | ID: mdl-33099537

ABSTRACT

Emerging Fanconi Anemia (FA) signaling in the field of cancer research annotates the extreme importance of its center player, Fanconi Anemia complementation group D2 (FANCD2) in protecting human cells from going awry. However, a previously-unrecognized form of FANCD2, namely FANCD2-V2, is understudied. We report TRK-Fused Gene (TFG) is critical for roles played by FANCD2-V2 in early responses to DNA damage, but not for FANCD2-V1, the long-known form of FANCD2. FANCD2-V2 forms nuclear foci upon DNA damage, and both its focus appearance and disappearance are earlier than FANCD2-V1. The amino acid/aa 5-100 of TFG and the aa1437-1442 of FANCD2-V2 were identified to contribute to their interaction, which maintains the steady-state level of FANCD2-V2 protein. TFGΔaa5-100 or FANCD2-V2Δaa1437-1442-carrying cells could not show timely focus formation of FANCD2-V2 upon DNA damage and gained carcinogenicity over time. This study provides a previously-unknown key to unlock in-depth insights into maintaining genome stability, fostering translational studies on preventing, diagnosing and/or treating related diseases.


Subject(s)
Cell Transformation, Neoplastic/metabolism , DNA Damage , Fanconi Anemia Complementation Group D2 Protein/metabolism , Genomic Instability , Proteins/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Fanconi Anemia Complementation Group D2 Protein/genetics , HEK293 Cells , Humans , Protein Binding , Protein Interaction Domains and Motifs , Protein Stability , Proteins/genetics , Time Factors , Ultraviolet Rays
6.
Breast Cancer Res Treat ; 175(2): 353-368, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30830488

ABSTRACT

PURPOSE: Low expression of long intergenic non-coding RNA LINC00472 in breast cancer is associated with aggressive tumors and unfavorable disease outcomes in multiple clinical datasets, but the reasons for these associations were unknown. METHODS: To study the mechanisms underlying the lncRNA's connection to breast cancer, we investigated the molecular targets and regulation of LINC00472 in breast cancer cells, and analyzed relevant molecular features in relation to patient survival. Gene expression profiles of breast cancer cells overexpressing LINC00472 were analyzed for its regulatory pathways and downstream targets. Effects of LINC00472 overexpression on cell behaviors were evaluated in vitro and in vivo. Meta-analysis was performed using online datasets and our own study. RESULTS: Analysis of LINC00472 transcriptome revealed ERα upregulation of LINC00472 expression, and an ERα-binding site in the LINC00472 promoter was identified. Evaluation of LINC00472 overexpression also indicated a possible link between LINC00472 and NF-κB. Cell experiments confirmed that LINC00472 suppressed the phosphorylation of p65 and IκBα through binding to IKKß, inhibiting its phosphorylation. High LINC00472 expression inhibited tumor growth both in vitro and in vivo and suppressed aggressive tumor cell behaviors in vitro. Suppressing LINC00472 expression in ER-positive tumor cells increased cell aggressive behaviors. Tamoxifen treatment of ER-positive cells inhibited ERα and LINC00472 expression and increased p65 and IκBα phosphorylation. Meta-analysis showed that LINC00472 expression were higher in ER-positive than ER-negative tumors and that high expression was associated with better disease outcomes in ER-positive patients. CONCLUSIONS: The study demonstrates that ERα upregulates LINC00472 which suppresses the phosphorylation of NF-κB, and suggests that endocrine treatment may lower LINC00472 and increase NF-κB activities, leading to tumor progression and disease recurrence.


Subject(s)
Breast Neoplasms/genetics , Estrogen Receptor alpha/genetics , Neoplasm Recurrence, Local/genetics , RNA, Long Noncoding/genetics , Adult , Aged , Aged, 80 and over , Animals , Breast Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic/genetics , Heterografts , Humans , MCF-7 Cells , Mice , Middle Aged , NF-kappa B/genetics , Neoplasm Recurrence, Local/pathology , Phosphorylation/drug effects , Tamoxifen/pharmacology , eIF-2 Kinase/genetics
7.
Gastroenterology ; 156(8): 2297-2312, 2019 06.
Article in English | MEDLINE | ID: mdl-30836096

ABSTRACT

BACKGROUND & AIMS: Interleukin 6 (IL6) and tumor necrosis factor contribute to the development of colitis-associated cancer (CAC). We investigated these signaling pathways and the involvement of G protein subunit alpha i1 (GNAI1), GNAI2, and GNAI3 in the development of CAC in mice and humans. METHODS: B6;129 wild-type (control) or mice with disruption of Gnai1, Gnai2, and/or Gnai3 or conditional disruption of Gnai2 in CD11c+ or epithelial cells were given dextran sulfate sodium (DSS) to induce colitis followed by azoxymethane (AOM) to induce carcinogenesis; some mice were given an antibody against IL6. Feces were collected from mice, and the compositions of microbiomes were analyzed by polymerase chain reactions. Dendritic cells (DCs) and myeloid-derived suppressor cells (MDSCs) isolated from spleen and colon tissues were analyzed by flow cytometry. We performed immunoprecipitation and immunoblot analyses of colon tumor tissues, MDSCs, and mouse embryonic fibroblasts to study the expression levels of GNAI1, GNAI2, and GNAI3 and the interactions of GNAI1 and GNAI3 with proteins in the IL6 signaling pathway. We analyzed the expression of Gnai2 messenger RNA by CD11c+ cells in the colonic lamina propria by PrimeFlow, expression of IL6 in DCs by flow cytometry, and secretion of cytokines in sera and colon tissues by enzyme-linked immunosorbent assay. We obtained colon tumor and matched nontumor tissues from 83 patients with colorectal cancer having surgery in China and 35 patients with CAC in the United States. Mouse and human colon tissues were analyzed by histology, immunoblot, immunohistochemistry, and/or RNA-sequencing analyses. RESULTS: GNAI1 and GNAI3 (GNAI1;3) double-knockout (DKO) mice developed more severe colitis after administration of DSS and significantly more colonic tumors than control mice after administration of AOM plus DSS. Development of increased tumors in DKO mice was not associated with changes in fecal microbiomes but was associated with activation of nuclear factor (NF) κB and signal transducer and activator of transcription (STAT) 3; increased levels of GNAI2, nitric oxide synthase 2, and IL6; increased numbers of CD4+ DCs and MDSCs; and decreased numbers of CD8+ DCs. IL6 was mainly produced by CD4+/CD11b+, but not CD8+, DCs in DKO mice. Injection of DKO mice with a blocking antibody against IL6 reduced the expansion of MDSCs and the number of tumors that developed after CAC induction. Incubation of MDSCs or mouse embryonic fibroblasts with IL6 induced activation of either NF-κB by a JAK2-TRAF6-TAK1-CHUK/IKKB signaling pathway or STAT3 by JAK2. This activation resulted in expression of GNAI2, IL6 signal transducer (IL6ST, also called GP130) and nitric oxide synthase 2, and expansion of MDSCs; the expression levels of these proteins and expansion of MDSCs were further increased by the absence of GNAI1;3 in cells and mice. Conditional disruption of Gnai2 in CD11c+ cells of DKO mice prevented activation of NF-κB and STAT3 and changes in numbers of DCs and MDSCs. Colon tumor tissues from patients with CAC had reduced levels of GNAI1 and GNAI3 and increased levels of GNAI2 compared with normal tissues. Further analysis of a public human colorectal tumor DNA microarray database (GSE39582) showed that low Gani1 and Gnai3 messenger RNA expression and high Gnai2 messenger RNA expression were significantly associated with decreased relapse-free survival. CONCLUSIONS: GNAI1;3 suppresses DSS-plus-AOM-induced colon tumor development in mice, whereas expression of GNAI2 in CD11c+ cells and IL6 in CD4+/CD11b+ DCs appears to promote these effects. Strategies to induce GNAI1;3, or block GNAI2 and IL6, might be developed for the prevention or therapy of CAC in patients.


Subject(s)
Cell Transformation, Neoplastic/genetics , Colitis/pathology , Colonic Neoplasms/pathology , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Animals , Biopsy, Needle , Carcinogenesis , Colitis/genetics , Colonic Neoplasms/genetics , Disease Models, Animal , Down-Regulation/genetics , Female , Gene Expression Regulation, Neoplastic , Immunohistochemistry , Interleukin-16/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Random Allocation , Reference Values , Sensitivity and Specificity , Signal Transduction/genetics
8.
Cancer Biol Ther ; 20(1): 1-5, 2019.
Article in English | MEDLINE | ID: mdl-30188759

ABSTRACT

In the past 25 years, incidence rates of breast cancer have risen about 30% in westernized countries. Mutations in BRCA1 and BRCA2 are the most prominent cause of breast cancer. However, these cancer susceptibility genes (BRCAs) only account for a few percent of women suffering breast tumor. With our understanding that BRCAs are Fanconi Anemia (FA) genes, investigations into the FA signaling network should provide a previously unrecognized key to unlock in-depth insights into both etiology and treatment of breast cancer. Here, we discuss utilization of the FA signaling as a unique genetic model system to expand our knowledge about the molecular biology of breast cancer and potential applications of the gained knowledge to enable preventive and therapeutic approaches for breast cancer patient care.


Subject(s)
Breast Neoplasms/genetics , Fanconi Anemia Complementation Group Proteins/genetics , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Signal Transduction/genetics , Breast Neoplasms/pathology , Carcinogenesis/genetics , Chromosomal Instability , Fanconi Anemia Complementation Group Proteins/metabolism , Female , Humans , Models, Genetic , Mutation
10.
Aging (Albany NY) ; 10(6): 1506-1522, 2018 Jun 21.
Article in English | MEDLINE | ID: mdl-29930218

ABSTRACT

Given importance of 22-Fanconi Anemia (FA) proteins together to act in a signaling pathway in preventing deleterious clinical symptoms, e.g. severe bone marrow failure, congenital defects, an early onset of aging and cancer, studies on each FA protein become increasingly attractive. However, an unbiased and systematic investigation of cellular effects resulting from each FA protein is missing. Here, we report roles of FA complementation C group protein (FANCC) in the protection from metabolic disorders. This study was prompted by the diabetes-prone feature displayed in FANCC knockout mice, which is not typically shown in patients with FA. We found that in cells expressing FANCC at different levels, there are representative alterations in metabolites associated with aging (glycine, citrulline, ornithine, L-asparagine, L-tyrosine, L-arginine, L-glutamine, L-leucine, L-isoleucine, L-valine, L-proline and L-alanine), Diabetes Mellitus (DM) (carbon monoxide, collagens, fatty acids, D-glucose, fumaric acid, 2-oxoglutaric acid, C3), inflammation (inosine, L-arginine, L-isoleucine, L-leucine, L-lysine, L-phenylalanine, hypoxanthine, L-methionine), and cancer ( L-methionine, sphingomyelin, acetyl-L-carnitine, L-aspartic acid, L-glutamic acid, niacinamide, phospho-rylethanolamine). We also found that FANCC can act in an FA-pathway-independent manner in tumor suppression. Collectively, featured-metabolic alterations are readouts of functional mechanisms underlying reduced tumorigenicity driven by FANCC, demonstrating close links among cancer, aging, inflammation and DM.


Subject(s)
Fanconi Anemia Complementation Group C Protein/metabolism , Metabolomics/methods , Animals , Biomarkers , Breast Neoplasms , Cell Line, Tumor , Cloning, Molecular , Female , Gene Expression Regulation , Humans , Principal Component Analysis
11.
Trends Genet ; 34(3): 171-183, 2018 03.
Article in English | MEDLINE | ID: mdl-29254745

ABSTRACT

In 1927 Guido Fanconi described a hereditary condition presenting panmyelopathy accompanied by short stature and hyperpigmentation, now better known as Fanconi anemia (FA). With this discovery the genetic and molecular basis underlying FA has emerged as a field of great interest. FA signaling is crucial in the DNA damage response (DDR) to mediate the repair of damaged DNA. This has attracted a diverse range of investigators, especially those interested in aging and cancer. However, recent evidence suggests FA signaling also regulates functions outside the DDR, with implications for many other frontiers of research. We discuss here the characteristics of FA functions and expand upon current perspectives regarding the genetics of FA, indicating that FA plays a role in a myriad of molecular and cellular processes.


Subject(s)
DNA Damage , DNA Repair , Fanconi Anemia/genetics , Signal Transduction/genetics , Fanconi Anemia/metabolism , Fanconi Anemia/pathology , Fanconi Anemia Complementation Group Proteins/genetics , Fanconi Anemia Complementation Group Proteins/metabolism , Humans , Models, Genetic , Mutation
12.
Trends Cancer ; 3(12): 840-856, 2017 12.
Article in English | MEDLINE | ID: mdl-29198440

ABSTRACT

The extremely high cancer incidence associated with patients suffering from a rare human genetic disease, Fanconi anemia (FA), demonstrates the importance of FA genes. Over the course of human tumor development, FA genes perform critical tumor-suppression roles. In doing so, FA provides researchers with a unique genetic model system to study cancer etiology. Here, we review how aberrant function of the 22 FA genes and their signaling network contributes to malignancy. From this perspective, we will also discuss how the knowledge discovered from FA research serves basic and translational cancer research.


Subject(s)
Fanconi Anemia Complementation Group Proteins/genetics , Fanconi Anemia/genetics , Neoplasms/genetics , DNA Damage/genetics , Fanconi Anemia/complications , Fanconi Anemia/pathology , Humans , Neoplasms/complications , Neoplasms/pathology , Protein Processing, Post-Translational/genetics , Signal Transduction/genetics
13.
Int J Mol Sci ; 18(8)2017 Aug 19.
Article in English | MEDLINE | ID: mdl-28825622

ABSTRACT

Investigators have dedicated considerable effort to understanding the molecular basis underlying Fanconi Anemia (FA), a rare human genetic disease featuring an extremely high incidence of cancer and many congenital defects. Among those studies, FA group D2 protein (FANCD2) has emerged as the focal point of FA signaling and plays crucial roles in multiple aspects of cellular life, especially in the cellular responses to DNA damage. Here, we discuss the recent and relevant studies to provide an updated review on the roles of FANCD2 in the DNA damage response.


Subject(s)
DNA Damage/genetics , DNA Repair/genetics , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia/genetics , Fanconi Anemia/pathology , Humans , Signal Transduction
14.
Sci Rep ; 7(1): 4921, 2017 07 07.
Article in English | MEDLINE | ID: mdl-28687786

ABSTRACT

Growing evidence supports a general hypothesis that aging and cancer are diseases related to energy metabolism. However, the involvement of Fanconi Anemia (FA) signaling, a unique genetic model system for studying human aging or cancer, in energy metabolism remains elusive. Here, we report that FA complementation group D2 protein (FANCD2) functionally impacts mitochondrial ATP production through its interaction with ATP5α, whereas this relationship was not observed in the mutant FANCD2 (K561R)-carrying cells. Moreover, while ATP5α is present within the mitochondria in wild-type cells, it is instead located mostly outside in cells that carry the non-monoubiquitinated FANCD2. In addition, mitochondrial ATP production is significantly reduced in these cells, compared to those cells carrying wtFANCD2. We identified one region (AA42-72) of ATP5α, contributing to the interaction between ATP5α and FANCD2, which was confirmed by protein docking analysis. Further, we demonstrated that mtATP5α (∆AA42-72) showed an aberrant localization, and resulted in a decreased ATP production, similar to what was observed in non-monoubiquitinated FANCD2-carrying cells. Collectively, our study demonstrates a novel role of FANCD2 in governing cellular ATP production, and advances our understanding of how defective FA signaling contributes to aging and cancer at the energy metabolism level.


Subject(s)
Adenosine Triphosphate/chemistry , Energy Metabolism/genetics , Fanconi Anemia Complementation Group D2 Protein/chemistry , Mitochondria/genetics , Mitochondrial Proton-Translocating ATPases/chemistry , Adenosine Triphosphate/biosynthesis , Amino Acid Sequence , Binding Sites , Cell Line, Tumor , Crystallography, X-Ray , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group D2 Protein/metabolism , Gene Expression , HEK293 Cells , Humans , Mitochondria/metabolism , Mitochondrial Proton-Translocating ATPases/genetics , Mitochondrial Proton-Translocating ATPases/metabolism , Molecular Docking Simulation , Mutation , Osteoblasts/cytology , Osteoblasts/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Thermodynamics
15.
Oncotarget ; 8(14): 22490-22500, 2017 Apr 04.
Article in English | MEDLINE | ID: mdl-28157704

ABSTRACT

Fanconi Anemia (FA) complementation group D2 protein (FANCD2) is the center of the FA tumor suppressor pathway, which has become an important field of investigation in human aging and cancer. Here we report an overlooked central player in the FA pathway, FANCD2 variant 2 (FANCD2-V2), which appears to perform more potent tumor suppressor-function compared to the known variant of FANCD2, namely, FANCD2-V1. Detailed analysis of the FANCD2 gene structure indicated a proximal and distal polyadenylation site (PAS), associated with V2 and V1 transcripts accordingly. RNA polymerase II Chromatin immunoprecipitation (ChIP) targeting the two PAS-regions determined lesser binding of RNA pol II to DNA fragments in the distal PAS region in non-malignant cells compared to malignant cells. Conversely, the opposite occurred in the proximal PAS region. Moreover, RNA immunoprecipitation (RIP) identified that U2 snRNP, a major component of RNA splicing complex that interacts with the 3'end of an intron, showed greater binding to the last intron of the FANCD2-V1 transcript in malignant cells compared to the non-malignant cells. Importantly, our data showed that in human tissue samples, the ratio of V2 /V1 expression in lung, bladder, or ovarian cancer correlates inversely with the tumor stages/grades. Therefore, these findings provide a previously unrecognized central player FANCD2-V2 and thus novel insights into human tumorigenesis, and indicate that V2/V1 can act as an effective biomarker in assisting the recognition of tumor malignance.


Subject(s)
Biomarkers, Tumor/genetics , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia/genetics , Lung Neoplasms/genetics , Ovarian Neoplasms/genetics , Tumor Suppressor Proteins/genetics , Urinary Bladder Neoplasms/genetics , Alternative Splicing , Carcinogenesis/genetics , Fanconi Anemia/diagnosis , Fanconi Anemia/pathology , Female , Gene Expression Regulation , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/pathology , Neoplasm Staging , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/pathology , Polyadenylation/genetics , Transcriptome , Urinary Bladder Neoplasms/diagnosis , Urinary Bladder Neoplasms/pathology
16.
Org Lett ; 18(10): 2335-8, 2016 05 20.
Article in English | MEDLINE | ID: mdl-27135759

ABSTRACT

Three unusual polyketide-sesquiterpene metabolites peyronellins A-C (1-3), along with the new epoxyphomalin analog 11-dehydroxy epoxyphomalin A (4), have been isolated from the endophytic fungus Peyronellaea coffeae-arabicae FT238, which was isolated from the native Hawaiian plant Pritchardia lowreyana. The structures of compounds 1-4 were characterized based on NMR and MS spectroscopic analysis. The absolute configuration (AC) of the compounds was determined by electronic circular dichroism (ECD). Compound 4 showed antiproliferative activity with an IC50 of 0.5 µM against OVCAR3, and it also strongly inhibited Stat3 at 5 µM.


Subject(s)
Antineoplastic Agents/isolation & purification , Arecaceae/microbiology , Ascomycota/chemistry , Cell Proliferation/drug effects , Endophytes/chemistry , Terpenes/isolation & purification , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Humans , Inhibitory Concentration 50 , Molecular Structure , Terpenes/pharmacology
17.
Oncotarget ; 7(22): 32351-61, 2016 May 31.
Article in English | MEDLINE | ID: mdl-27083049

ABSTRACT

Mutations in the human RecQ helicase, BLM, causes Bloom Syndrome, which is a rare autosomal recessive disorder and characterized by genomic instability and an increased risk of cancer. Fanconi Anemia (FA), resulting from mutations in any of the 19 known FA genes and those yet to be known, is also characterized by chromosomal instability and a high incidence of cancer. BLM helicase and FA proteins, therefore, may work in a common tumor-suppressor signaling pathway. To date, it remains largely unclear as to how BLM and FA proteins work concurrently in the maintenance of genome stability. Here we report that BLM is involved in the early activation of FA group D2 protein (FANCD2). We found that FANCD2 activation is substantially delayed and attenuated in crosslinking agent-treated cells harboring deficient Blm compared to similarly treated control cells with sufficient BLM. We also identified that the domain VI of BLM plays an essential role in promoting FANCD2 activation in cells treated with DNA crosslinking agents, especially ultraviolet B. The similar biological effects performed by ΔVI-BLM and inactivated FANCD2 further confirm the relationship between BLM and FANCD2. Mutations within the domain VI of BLM detected in human cancer samples demonstrate the functional importance of this domain, suggesting human tumorigenicity resulting from mtBLM may be at least partly attributed to mitigated FANCD2 activation. Collectively, our data show a previously unknown regulatory liaison in advancing our understanding of how the cancer susceptibility gene products act in concert to maintain genome stability.


Subject(s)
Bloom Syndrome/enzymology , Fanconi Anemia Complementation Group L Protein/metabolism , Fanconi Anemia/enzymology , Neoplasms/enzymology , RecQ Helicases/metabolism , Signal Transduction , Bloom Syndrome/genetics , Bloom Syndrome/pathology , Bone Neoplasms/enzymology , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Survival , Cross-Linking Reagents/pharmacology , Fanconi Anemia/genetics , Fanconi Anemia/pathology , Fanconi Anemia Complementation Group L Protein/chemistry , Fanconi Anemia Complementation Group L Protein/genetics , Female , Humans , Mutation , Neoplasms/genetics , Neoplasms/pathology , Osteosarcoma/enzymology , Osteosarcoma/genetics , Osteosarcoma/pathology , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Protein Binding , Protein Interaction Domains and Motifs , RNA Interference , RecQ Helicases/chemistry , RecQ Helicases/genetics , Signal Transduction/drug effects , Signal Transduction/radiation effects , Transfection , Ultraviolet Rays
18.
J Proteome Res ; 15(4): 1333-41, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26956768

ABSTRACT

Metabolic profiling has great potential to help the diagnosis and prognosis of cancer patients. Fanconi Anemia (FA) tumor-suppressor signaling has been instrumental in understanding human tumorigenesis. However, this instrumental understanding has never been demonstrated at the metabolic level. Here, we show that impaired FA signaling can lead cells to exhibit metabolic signatures of tumorigenesis. This is consistent with our original studies of the roles of FA signaling in suppressing non-FA tumorigenesis at functional and genetic levels. Using ultraperformance liquid chromatography-mass spectroscopy and gas chromatography-mass spectrometry, we characterized metabolic alterations in bladder cancer cells carrying an intact or impaired FA pathway. The latter was obtained by ectopically expressing FAVL (FAVL-high), which we previously found to be capable of inactivating FA signaling. A total of 18 metabolites, end products of cell proliferation or apoptosis, were significantly different between FAVL-high and -low cells. Methionine, phenylalanine, and threonine, resulting from a tumorigenic process, were substantially increased in FAVL-high cells. With this study, we achieved genomic, functional, and metabolomic characterization of the roles of FA signaling in the development of human cancer. Furthermore, this study provides novel insights into how to translate FA basic research into strategies for producing effective biomarkers in human cancer diagnosis and prognosis.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Epithelial Cells/metabolism , Fanconi Anemia Complementation Group L Protein/genetics , Metabolome , Signal Transduction , Alternative Splicing , Apoptosis , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Chromatography, Liquid , Epithelial Cells/pathology , Fanconi Anemia Complementation Group L Protein/metabolism , Gene Expression , Humans , Mass Spectrometry , Methionine/metabolism , Phenylalanine/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Threonine/metabolism , Transgenes
19.
Article in English | MEDLINE | ID: mdl-26640811

ABSTRACT

Fanconi anemia (FA) is a rare human genetic disease, resulting from dysfunction in any of 17 known complementation proteins: FANC-A, B, C, D1, D2, E, F, G, I, J, L, M, N, O, P, Q & S, and other unknowns. Besides the severe bone marrow failure, an extremely high incidence of cancer as well as many other clinic symptoms associated with FA patients, FA cells are known of insufficiency in homologous recombination, DNA mismatch repair, nucleotide excision repair, translesion DNA synthesis, and other molecular defects, leading to genome instability. Those similar molecular and cellular/tissue features show that all FA proteins function in one common signaling pathway, namely, the FA pathway. The monoubiquitination of FANCD2 is the central step of the FA pathway activation upon DNA damage or during DNA replication. The molecular functions of FANCD2 emerge as a very attractive filed of investigation in cancer research. Herein, we review the recent progresses in FANCD2 functions at these rapidly progressed aspects.

20.
Breast Cancer Res Treat ; 154(3): 473-82, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26564482

ABSTRACT

Long non-coding RNAs (lncRNAs) are a class of newly recognized DNA transcripts that have diverse biological activities. Dysregulation of lncRNAs may be involved in many pathogenic processes including cancer. Recently, we found an intergenic lncRNA, LINC00472, whose expression was correlated with breast cancer progression and patient survival. Our findings were consistent across multiple clinical datasets and supported by results from in vitro experiments. To evaluate further the role of LINC00472 in breast cancer, we used various online databases to investigate possible mechanisms that might affect LINC00472 expression in breast cancer. We also analyzed associations of LINC00472 with estrogen receptor, tumor grade, and molecular subtypes in additional online datasets generated by microarray platforms different from the one we investigated previously. We found that LINC00472 expression in breast cancer was regulated more possibly by promoter methylation than by the alteration of gene copy number. Analysis of additional datasets confirmed our previous findings of high expression of LINC00472 associated with ER-positive and low-grade tumors and favorable molecular subtypes. Finally, in nine datasets, we examined the association of LINC00472 expression with disease-free survival in patients with grade 2 tumors. Meta-analysis of the datasets showed that LINC00472 expression in breast tumors predicted the recurrence of breast cancer in patients with grade 2 tumors. In summary, our analyses confirm that LINC00472 is functionally a tumor suppressor, and that assessing its expression in breast tumors may have clinical implications in breast cancer management.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/mortality , DNA Methylation , RNA, Long Noncoding/genetics , Breast Neoplasms/pathology , Comparative Genomic Hybridization , Databases, Genetic , Disease-Free Survival , Female , Gene Dosage , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Humans , Promoter Regions, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...