Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 185(1): 169-183.e19, 2022 01 06.
Article in English | MEDLINE | ID: mdl-34963055

ABSTRACT

Non-small cell lung cancers (NSCLCs) harboring KEAP1 mutations are often resistant to immunotherapy. Here, we show that KEAP1 targets EMSY for ubiquitin-mediated degradation to regulate homologous recombination repair (HRR) and anti-tumor immunity. Loss of KEAP1 in NSCLC induces stabilization of EMSY, producing a BRCAness phenotype, i.e., HRR defects and sensitivity to PARP inhibitors. Defective HRR contributes to a high tumor mutational burden that, in turn, is expected to prompt an innate immune response. Notably, EMSY accumulation suppresses the type I interferon response and impairs innate immune signaling, fostering cancer immune evasion. Activation of the type I interferon response in the tumor microenvironment using a STING agonist results in the engagement of innate and adaptive immune signaling and impairs the growth of KEAP1-mutant tumors. Our results suggest that targeting PARP and STING pathways, individually or in combination, represents a therapeutic strategy in NSCLC patients harboring alterations in KEAP1.


Subject(s)
Carcinoma, Non-Small-Cell Lung/immunology , Interferon Type I/metabolism , Lung Neoplasms/immunology , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Recombinational DNA Repair/genetics , Repressor Proteins/metabolism , Tumor Escape/genetics , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Female , HEK293 Cells , Humans , Immunity, Innate/genetics , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mutation , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Repressor Proteins/genetics , Signal Transduction/genetics , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Xenograft Model Antitumor Assays
2.
Nature ; 592(7856): 789-793, 2021 04.
Article in English | MEDLINE | ID: mdl-33854235

ABSTRACT

D-type cyclins are central regulators of the cell division cycle and are among the most frequently deregulated therapeutic targets in human cancer1, but the mechanisms that regulate their turnover are still being debated2,3. Here, by combining biochemical and genetics studies in somatic cells, we identify CRL4AMBRA1 (also known as CRL4DCAF3) as the ubiquitin ligase that targets all three D-type cyclins for degradation. During development, loss of Ambra1 induces the accumulation of D-type cyclins and retinoblastoma (RB) hyperphosphorylation and hyperproliferation, and results in defects of the nervous system that are reduced by treating pregnant mice with the FDA-approved CDK4 and CDK6 (CDK4/6) inhibitor abemaciclib. Moreover, AMBRA1 acts as a tumour suppressor in mouse models and low AMBRA1 mRNA levels are predictive of poor survival in cancer patients. Cancer hotspot mutations in D-type cyclins abrogate their binding to AMBRA1 and induce their stabilization. Finally, a whole-genome, CRISPR-Cas9 screen identified AMBRA1 as a regulator of the response to CDK4/6 inhibition. Loss of AMBRA1 reduces sensitivity to CDK4/6 inhibitors by promoting the formation of complexes of D-type cyclins with CDK2. Collectively, our results reveal the molecular mechanism that controls the stability of D-type cyclins during cell-cycle progression, in development and in human cancer, and implicate AMBRA1 as a critical regulator of the RB pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Division , Cyclin D1/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , CRISPR-Cas Systems , Cyclin D2/metabolism , Cyclin D3/metabolism , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Female , Gene Knockout Techniques , Genes, Tumor Suppressor , HCT116 Cells , HEK293 Cells , Humans , Male , Mice , Neoplasms/genetics , Ubiquitin/metabolism
3.
Blood ; 142(5): 460-476, 2023 08 03.
Article in English | MEDLINE | ID: mdl-37267505

ABSTRACT

The chromosome 9p21 locus comprises several tumor suppressor genes including MTAP, CDKN2A, and CDKN2B, and its homo- or heterozygous deletion is associated with reduced survival in multiple cancer types. We report that mice with germ line monoallelic deletion or induced biallelic deletion of the 9p21-syntenic locus (9p21s) developed a fatal myelodysplastic syndrome/myeloproliferative neoplasm (MDS/MPN)-like disease associated with aberrant trabecular bone formation and/or fibrosis in the bone marrow (BM). Reciprocal BM transfers and conditional targeting of 9p21s suggested that the disease originates in the BM stroma. Single-cell analysis of 9p21s-deficient BM stroma revealed the expansion of chondrocyte and osteogenic precursors, reflected in increased osteogenic differentiation in vitro. It also showed reduced expression of factors maintaining hematopoietic stem/progenitor cells, including Cxcl12. Accordingly, 9p21s-deficient mice showed reduced levels of circulating Cxcl12 and concomitant upregulation of the profibrotic chemokine Cxcl13 and the osteogenesis- and fibrosis-related multifunctional glycoprotein osteopontin/Spp1. Our study highlights the potential of mutations in the BM microenvironment to drive MDS/MPN-like disease.


Subject(s)
Bone Marrow , Osteogenesis , Mice , Animals , Bone Marrow/pathology , Hematopoietic Stem Cells/metabolism , Genes, Tumor Suppressor , Cell Differentiation
4.
Biochim Biophys Acta Bioenerg ; 1858(8): 686-699, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28161329

ABSTRACT

Mitochondria, known for more than a century as the energy powerhouse of a cell, represent key intracellular signaling hub that are emerging as important determinants of several aspects of cancer development and progression, including metabolic reprogramming, acquisition of metastatic capability, and response to chemotherapeutic drugs. The majority of cancer cells harbors somatic mutations in the mitochondrial genome (mtDNA) and/or alterations in the mtDNA content, leading to mitochondrial dysfunction. Decreased mtDNA content is also detected in tumor-initiating cells, a subpopulation of cancer cells that are believed to play an integral role in cancer recurrence following chemotherapy. Although mutations in mitochondrial genes are common in cancer cells, they do not shut down completely the mitochondrial energy metabolism and functionality. Instead, they promote rewiring of the bioenergetics and biosynthetic profile of a cancer cell through a mitochondria-to-nucleus signaling activated by "dysfunctional" mitochondria that results in changes in transcription and/or activity of cancer-related genes and signaling pathways. Different cancer cell types may undergo different bioenergetic changes, some to more glycolytic and some to more oxidative. These different metabolic signatures may coexist within the same tumor mass (intra-tumor heterogeneity). In this review we describe the current understanding of mitochondrial dysfunction in the context of cancer chemoresistance with special attention to the role of mtDNA alterations. We put emphasis on potential therapeutic strategies targeting different metabolic events specific to cancer cells, including glycolysis, glutaminolysis, oxidative phosphorylation, and the retrograde signaling, to prevent chemoresistance. We also highlight novel genome-editing strategies aimed at "correcting" mtDNA defects in cancer cells. We conclude on the importance of considering intratumor metabolic heterogeneity to develop effective metabolism-based cancer therapy that can overcome chemoresistance. This article is part of a Special Issue entitled Mitochondria in Cancer, edited by Giuseppe Gasparre, Rodrigue Rossignol and Pierre Sonveaux.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/physiology , Energy Metabolism/drug effects , Mitochondria/drug effects , Neoplasms/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Transformation, Neoplastic , DNA, Mitochondrial/genetics , Disease Progression , Drug Design , Drug Resistance, Neoplasm/drug effects , Gene Transfer, Horizontal , Humans , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/physiology , Models, Biological , Neoplasm Metastasis , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Oxidative Phosphorylation , Sequence Deletion , Signal Transduction/drug effects , Tumor Microenvironment/drug effects
5.
J Magn Reson Imaging ; 39(6): 1401-10, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24123697

ABSTRACT

PURPOSE: To characterize the uptake and elimination of ferumoxytol, an ultrasmall superparamagnetic iron oxide (USPIO) agent, in bone marrow of healthy human subjects. MATERIALS AND METHODS: Four men and two postmenopausal women, aged 22 to 57 years, were prospectively included. Simultaneous fat, water, and T2* mapping of the proximal femora was performed at 1.5 Tesla using a three-dimensional multiple gradient echo sequence. After baseline imaging, ferumoxytol (Feraheme/Rienso) was injected intravenously at a dose of 5 mg Fe/kg body weight. Imaging was repeated at 3 days, 1 month, 3 months, and 5 months after administration. RESULTS: Imaging at 3 days revealed large increases in R2* ( =1/T2*) in hematopoietic marrow and lower average responses in fatty marrow, consistent with macrophage-specific uptake. However, certain regions of the diaphysis exhibited substantial R2* enhancement despite having very high fat content. This suggests the persistence of residual marrow stroma following adipose conversion, and may reflect the ability of diaphyseal marrow to adapt dynamically to fluctuating demand for hematopoiesis. Follow-up imaging demonstrated almost complete R2* recovery within 3 months. CONCLUSION: The observed R2* enhancement characteristics support applications for ferumoxytol in distinguishing normal or hypercellular marrow from neoplasms, infection and inflammation. Further studies are warranted in specific patient populations.


Subject(s)
Bone Marrow/metabolism , Contrast Media/pharmacokinetics , Dextrans/pharmacokinetics , Magnetic Resonance Imaging/methods , Adult , Cohort Studies , Female , Follow-Up Studies , Humans , Image Processing, Computer-Assisted/methods , Imaging, Three-Dimensional/methods , Magnetite Nanoparticles , Male , Middle Aged , Prospective Studies , Reference Values , Young Adult
6.
Mult Scler Relat Disord ; 79: 105047, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37832255

ABSTRACT

OBJECTIVES: To compare proportions of B-cell lineage CD19+ and CD20+ cells in CSF of African-American (AA) and White (W) patients with MS. BACKGROUND: AA MS patients are more likely to have oligoclonal bands in CSF, higher IgG index in CSF, and higher circulating plasmablasts in blood than W MS patients. It is unknown whether the proportion of B-cells in CSF differs between AA and W patients in MS. METHODS: Demographics, disease-related information, treatment history were retrospectively collected on patients with MS who self-identified as AA or W and underwent flow cytometry of CSF during diagnostic work-up. Proportion of B-lymphocytes, T-lymphocytes, NK cells, monocytes, and plasma cells were analyzed with flow cytometry. RESULTS: 20 AA and 56 W MS patients fulfilled our inclusion criteria. The groups had similar demographics, CSF cell counts, protein and glucose CSF concentrations, and oligoclonal band number. IgG index was higher in AA compared to W (1.08 vs. 0.85, p = 0.031). AA had higher proportions of CD19+ (5.46 % AA vs. 2.26 % W, p = 0.006) and CD20+ (4.64 % AA vs. 1.91 % W, p = 0.004) cells but did not significantly differ in proportion of CD4+, CD8+, CD38+ bright B-cells, NK cells and monocytes. CONCLUSIONS: B-cells are overrepresented in the CSF of African American patients with MS relative to Whites.


Subject(s)
B-Lymphocytes , Black or African American , Multiple Sclerosis , Humans , Cell Lineage , Immunoglobulin G , Oligoclonal Bands/cerebrospinal fluid , Retrospective Studies , White
7.
Am J Pathol ; 178(5): 2367-76, 2011 May.
Article in English | MEDLINE | ID: mdl-21514447

ABSTRACT

BRCA2 (breast cancer 2, early onset) is a tumor suppressor gene that confers increased susceptibility for prostate cancer (PCa). Previous in vitro experiments demonstrated that Skp2, an E3 ubiquitin ligase aberrantly overexpressed in PCa, is involved in the proteolytic degradation of BRCA2 in PCa cells, suggesting that the BRCA2-Skp2 interaction may play a role in prostate tumorigenesis. Herein, we investigated BRCA2 and Skp2 expression during PCa development using a prostate TMA. Although luminal and basal benign prostate epithelium exhibited moderate to strong nuclear BRCA2 immunostaining, the intensity and number of positive nuclei decreased significantly in high-grade prostatic intraepithelial neoplasia and PCa. Decreased frequency and intensity of nuclear BRCA2 labeling were inversely correlated with Skp2 expression in high-grade prostatic intraepithelial neoplasia and PCa. To functionally assess the effects of BRCA2 and Skp2 expression on prostate malignant transformation, we overexpressed Skp2 in normal immortalized prostate cells. Skp2 overexpression reduced BRCA2 protein and promoted cell growth and migration. A similar phenotype was observed after reduction of BRCA2 protein levels using specific BRCA2 small-interfering RNA. Forced BRCA2 expression in Skp2-overexpressing stable transfectants inhibited the migratory and growth properties by >60%. These results show that loss of BRCA2 expression during prostate tumor development is strongly correlated with both migratory behavior and cancer growth and include Skp2 as a BRCA2 proteolytic partner in vivo.


Subject(s)
BRCA2 Protein/biosynthesis , Cell Transformation, Neoplastic/genetics , Prostatic Neoplasms/metabolism , S-Phase Kinase-Associated Proteins/biosynthesis , BRCA2 Protein/genetics , Blotting, Western , Cell Movement , Cell Proliferation , Humans , Immunohistochemistry , Male , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , S-Phase Kinase-Associated Proteins/genetics , Tissue Array Analysis , Transfection , Up-Regulation
8.
Mult Scler Relat Disord ; 63: 103830, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35490448

ABSTRACT

This retrospective, single-center study aimed to characterize and compare the kinetics of B-cell reemergence following anti-CD20 infusion (anti-CD20i) in African American (AA) and white patients with MS or NMOSD. In a logistic regression model that included race, time since anti-CD20i, body mass index, and diagnosis, only AA race (p=0.01) and time since anti-CD20i (p=0.0003) were significant predictors of B-cell repletion. However, B-cell subset composition was similar between AA and white patients with detectable CD19+ B-cell counts. These findings highlight the importance of including a diverse study population in future studies of anti-CD20 therapies.


Subject(s)
Multiple Sclerosis , Nervous System Diseases , Neuromyelitis Optica , Antigens, CD20 , B-Lymphocytes , Cell Count , Humans , Immunologic Factors , Multiple Sclerosis/therapy , Neuromyelitis Optica/therapy , Retrospective Studies
9.
Cell Rep ; 37(3): 109870, 2021 10 19.
Article in English | MEDLINE | ID: mdl-34686346

ABSTRACT

FBXO31 is the substrate receptor of one of many CUL1-RING ubiquitin ligase (CRL1) complexes. Here, we show that low FBXO31 mRNA levels are associated with high pre-operative prostate-specific antigen (PSA) levels and Gleason grade in human prostate cancer. Mechanistically, the ubiquitin ligase CRL1FBXO31 promotes the ubiquitylation-mediated degradation of DUSP6, a dual specificity phosphatase that dephosphorylates and inactivates the extracellular-signal-regulated kinase-1 and -2 (ERK1/2). Depletion of FBXO31 stabilizes DUSP6, suppresses ERK signaling, and activates the PI3K-AKT signaling cascade. Moreover, deletion of FBXO31 promotes tumor development in a mouse orthotopic model of prostate cancer. Treatment with BCI, a small molecule inhibitor of DUSP6, suppresses AKT activation and prevents tumor formation, suggesting that the FBXO31 tumor suppressor activity is dependent on DUSP6. Taken together, our studies highlight the relevance of the FBXO31-DUSP6 axis in the regulation of ERK- and PI3K-AKT-mediated signaling pathways, as well as its therapeutic potential in prostate cancer.


Subject(s)
Dual Specificity Phosphatase 6/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , F-Box Proteins/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Prostatic Neoplasms/enzymology , Proto-Oncogene Proteins c-akt/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cullin Proteins/genetics , Cullin Proteins/metabolism , Cyclohexylamines/pharmacology , Dual Specificity Phosphatase 6/antagonists & inhibitors , Dual Specificity Phosphatase 6/genetics , Enzyme Activation , Enzyme Inhibitors/pharmacology , Enzyme Stability , F-Box Proteins/genetics , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Indenes/pharmacology , Male , Mice, Inbred NOD , Mice, SCID , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Proteolysis , Signal Transduction , Tumor Suppressor Proteins/genetics , Xenograft Model Antitumor Assays
10.
Nat Cell Biol ; 22(9): 1130-1142, 2020 09.
Article in English | MEDLINE | ID: mdl-32839549

ABSTRACT

Epigenetic plasticity is a pivotal factor that drives metastasis. Here, we show that the promoter of the gene that encodes the ubiquitin ligase subunit FBXL7 is hypermethylated in advanced prostate and pancreatic cancers, correlating with decreased FBXL7 mRNA and protein levels. Low FBXL7 mRNA levels are predictive of poor survival in patients with pancreatic and prostatic cancers. FBXL7 mediates the ubiquitylation and proteasomal degradation of active c-SRC after its phosphorylation at Ser 104. The DNA-demethylating agent decitabine recovers FBXL7 expression and limits epithelial-to-mesenchymal transition and cell invasion in a c-SRC-dependent manner. In vivo, FBXL7-depleted cancer cells form tumours with a high metastatic burden. Silencing of c-SRC or treatment with the c-SRC inhibitor dasatinib together with FBXL7 depletion prevents metastases. Furthermore, decitabine reduces metastases derived from prostate and pancreatic cancer cells in a FBXL7-dependent manner. Collectively, this research implicates FBXL7 as a metastasis-suppressor gene and suggests therapeutic strategies to counteract metastatic dissemination of pancreatic and prostatic cancer cells.


Subject(s)
Epigenesis, Genetic/genetics , Epithelial-Mesenchymal Transition/genetics , F-Box Proteins/genetics , Gene Silencing/physiology , Neoplasm Metastasis/genetics , Protein Subunits/genetics , src-Family Kinases/genetics , Animals , Cell Line , Gene Expression Regulation, Neoplastic/genetics , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, SCID , PC-3 Cells , Signal Transduction/genetics , Ubiquitin/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination/genetics
12.
Cancer Res ; 79(6): 1098-1112, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30504123

ABSTRACT

IFNγ, a potent cytokine known to modulate tumor immunity and tumoricidal effects, is highly elevated in patients with prostate cancer after radiation. In this study, we demonstrate that IFNγ can induce epithelial-to-mesenchymal transition (EMT) in prostate cancer cells via the JAK-STAT signaling pathway, leading to the transcription of IFN-stimulated genes (ISG) such as IFN-induced tetratricopeptide repeat 5 (IFIT5). We unveil a new function of IFIT5 complex in degrading precursor miRNAs (pre-miRNA) that includes pre-miR-363 from the miR-106a-363 cluster as well as pre-miR-101 and pre-miR-128, who share a similar 5'-end structure with pre-miR-363. These suppressive miRNAs exerted a similar function by targeting EMT transcription factors in prostate cancer cells. Depletion of IFIT5 decreased IFNγ-induced cell invasiveness in vitro and lung metastasis in vivo. IFIT5 was highly elevated in high-grade prostate cancer and its expression inversely correlated with these suppressive miRNAs. Altogether, this study unveils a prometastatic role of the IFNγ pathway via a new mechanism of action, which raises concerns about its clinical application.Significance: A unique IFIT5-XRN1 complex involved in the turnover of specific tumor suppressive microRNAs is the underlying mechanism of IFNγ-induced epithelial-to-mesenchymal transition in prostate cancer.See related commentary by Liu and Gao, p. 1032.


Subject(s)
Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Interferon-gamma/pharmacology , Lung Neoplasms/secondary , MicroRNAs/genetics , Neoplasm Proteins/metabolism , Prostatic Neoplasms/pathology , Animals , Antiviral Agents/pharmacology , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Proliferation , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Mice , Mice, SCID , Neoplasm Proteins/genetics , Prognosis , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
Cancer Sci ; 99(3): 553-63, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18167127

ABSTRACT

BRCA2 is a multifunctional tumor suppressor protein which plays critical roles in DNA repair, transcription, and cell proliferation, and the loss of which has been linked to the biology of several types of cancers. Here, on prostate adenocarcinoma specimens from 80 patients, we demonstrate that BRCA2 protein is lost in carcinoma cells compared to normal and hyperplastic prostate epithelium. Using highly metastatic prostate cancer PC-3 cells, we show that while BRCA2 depletion by small-interfering RNA promoted migration onto the extracellular matrix proteins fibronectin, laminin, and collagens, as well as invasion through the reconstituted basement membrane matrix Matrigel by more than 140%, recombinant BRCA2 overexpression decreased both phenomena by 57-80% and changed cell morphology from angular and spindle to round and compact. The BRCA2 inhibitory effect on cancer cell migration and invasion resulted from down-regulation of matrix metalloproteinase (MMP)-9 protein levels due to increased MMP-9 proteolysis, and was signaled through inhibition of PI3-kinase/AKT and activation of MAPK/ERK pathway. In BRCA2-overexpressing PC-3 cells, transient transfection with a constitutively active PI3-kinase mutant or treatment with the MAPK/ERK inhibitor PD98059 rescued MMP-9 levels and restored the migratory and invasive capabilities. Consistently, PI3-kinase inhibition with a dominant-negative mutant or MAPK/ERK activation with a gain-of-function mutant reduced MMP-9 levels and prevented migration and invasion in wild-type PC-3 cells. These results provide novel evidence showing that a functional BRCA2 protein may limit the metastatic potential of neoplastic cells by down-regulating MMP-9 production through inhibition of PI3-kinase/AKT and activation of MAPK/ERK, effectively hindering cancer cell migration and invasion.


Subject(s)
BRCA2 Protein/metabolism , Matrix Metalloproteinase 9/metabolism , Prostatic Neoplasms/enzymology , Up-Regulation , BRCA2 Protein/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , Humans , Male , Matrix Metalloproteinase 9/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Neoplasm Invasiveness , Phosphatidylinositol 3-Kinases/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
14.
Am J Clin Pathol ; 129(2): 300-8, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18208811

ABSTRACT

Diagnosis of myelodysplastic syndromes (MDS) could be difficult. We explored the usefulness of the enumeration of maturing B-lineage precursors (hematogones) by multiparameter flow cytometric analysis in the diagnosis of MDS in bone marrow (BM) specimens. We evaluated 111 MDS, 120 non-MDS (most with cytopenias; control group 1), and 41 noncytopenic lymphoma staging BM (control group 2) specimens. The percentage of total hematogones was significantly lower in MDS (median, 0%; mean, 0.10%) compared with non-MDS (control group 1, median, 0.38%, and mean, 0.91%; control group 2, median, 0.38%, and mean, 0.60%; P < .0001), as was the percentage of the most immature (stage I) hematogones. Thus, hematogone enumeration may serve as a biomarker to aid in the diagnosis of MDS. Interestingly, the percentage of hematogones was not significantly different between MDS subgroups or patients with MDS with and without chromosomal abnormalities, implying that a defect in maturing B-cell precursors may be an early event in the pathogenesis of MDS.


Subject(s)
B-Lymphocytes/pathology , Bone Marrow Cells/pathology , Flow Cytometry/methods , Hematopoietic Stem Cells/pathology , Myelodysplastic Syndromes/pathology , Adult , Aged , Aged, 80 and over , Case-Control Studies , Female , Humans , Immunophenotyping , Male , Middle Aged
15.
Cell Oncol ; 30(4): 307-22, 2008.
Article in English | MEDLINE | ID: mdl-18607066

ABSTRACT

Mitochondrial dysfunction resulting from mitochondrial DNA (mtDNA) mutations and/or depletion has been correlated with cancer progression and drug resistance. To investigate the role of mtDNA in prostate cancer progression, we used LNCaP and PC-3 prostate carcinoma cells as experimental model. Compared to minimally invasive androgen-dependent LNCaP cells, highly invasive androgen-independent PC-3 cells, as well as androgen-independent DU145 and C4-2 cells, exhibited significantly reduced mtDNA content. In PC-3 cells, reduction of mtDNA was accompanied by decreased mitochondrial membrane potential (DeltaPsi(m)), increased migration onto the basement membrane protein laminin-1, reduced chemosensitivity to paclitaxel (IC(50)=110 nM vs. 22 nM) and decreased expression of poly(ADP-ribose) polymerase (PARP)-1. To investigate the relationship between mtDNA depletion and these phenotypic characteristics, we established mtDNA-depleted LNCaP cells [Rho(-)] by long-term exposure to ethidium bromide or treated wild-type LNCaP cells with a mitochondrial ionophore, carbonyl cyanide m-chlorophenylhydrazone. Both manipulations resulted in DeltaPsi(m) loss, acquisition of invasive cytology, increased motility onto laminin-1, reduced sensitivity to paclitaxel (IC(50)= approximately 100 nM) and approximately 75% reduction in PARP-1 protein levels, resembling PC-3 cells. Overall, these results provide novel evidence demonstrating that mtDNA depletion in early prostate carcinoma may contribute to the acquisition of a more invasive phenotype that is less sensitive to paclitaxel-induced apoptosis.


Subject(s)
Androgens/metabolism , DNA, Mitochondrial/genetics , Neoplasms, Hormone-Dependent/genetics , Poly(ADP-ribose) Polymerases/genetics , Prostatic Neoplasms/genetics , Apoptosis/drug effects , Carcinogens/pharmacology , Cell Line, Tumor , Cell Migration Assays , Cell Movement/genetics , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , DNA, Mitochondrial/antagonists & inhibitors , DNA, Mitochondrial/biosynthesis , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Ethidium/pharmacology , Humans , Male , Membrane Potential, Mitochondrial , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasms, Hormone-Dependent/pathology , Paclitaxel/pharmacology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/biosynthesis , Prostatic Neoplasms/pathology
16.
Int J Oncol ; 30(1): 217-24, 2007 01.
Article in English | MEDLINE | ID: mdl-17143532

ABSTRACT

BRCA2 is central to an utterly diverse biological behavior elicited after integrin-mediated normal and prostate cancer cell adhesion to basement membrane (BM) and extracellular matrix (ECM) proteins. Unlike normal cells, adhesive stimuli in cancer cells activate PI 3-kinase/AKT signaling resulting in BRCA2 degradation and unchecked cancer cell proliferation and metastasis. However, the precise mechanisms involved in normal BRCA2 homeostasis are unknown. We investigated ERK and AKT phosphorylation in normal (PNT1A) and cancer (PC-3) prostate cells after adhesion to ECM and the effects upon BRCA2 and cell proliferation. PNT1A cell adhesion to ECM triggered MAPK/ERK signaling resulting in upregulation of BRCA2 mRNA and protein, with negligible effects upon cell proliferation. Disruption of MAPK/ERK with PD98059 prevented any BRCA2 upregulation inhibiting DNA synthesis below basal levels. PC-3 cells exhibited a defective MAPK/ERK pathway that was unresponsive to adhesion to the ECM, which instead triggered PI 3-kinase/AKT signaling leading to BRCA2 protein depletion and cell proliferation. Reconstitution of MAPK/ERK by recombinant expression of a constitutively active form of MAPK kinase 1 (MEK1) effectively reversed the neoplastic phenotype by increasing BRCA2 expression and preventing any aberrant cell proliferation at rest and upon interaction with ECM proteins. Our results suggest that aberrant loss of MAPK/ERK activity in prostate cancer may play a pivotal role in the malignant phenotype, and provide evidence that interventions aimed at bypassing the signaling block are able to effectively reverse neoplastic unchecked cell proliferation.


Subject(s)
BRCA2 Protein/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Prostatic Neoplasms/pathology , Cell Adhesion , Cell Division , Cell Line, Tumor , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/genetics , Humans , Immunoblotting , Male , Mitogen-Activated Protein Kinases/genetics , RNA, Small Interfering/genetics , Thymidine/metabolism , Transfection
17.
Autops Case Rep ; 7(4): 30-36, 2017.
Article in English | MEDLINE | ID: mdl-29259930

ABSTRACT

Intravascular large B-cell lymphoma (IVLBCL) is a very rare extra nodal lymphoma that tends to proliferate within small blood vessels, particularly capillaries and postcapillary venules while sparing the organ parenchyma. The cause of its affinity for the vascular bed remains unknown. Because of its rarity and unremarkable clinical presentation, a timely diagnosis of IVLBCL is very challenging. Here, we describe a case of IVLBCL presenting as pancreatic mass that was ultimately diagnosed at autopsy. A 71-year-old Caucasian female presented with a 3-month history of fatigue, abdominal pain, and weight loss. She was referred to the emergency room with a new diagnosis of portal vein thrombosis and lactic acidosis. During her hospital course she was found to have a 1.9 × 1.8 cm lesion in the pancreatic tail on imaging; The cytologic specimen on the mass showed a high-grade lymphoma. A bone marrow biopsy showed no involvement. The patient's condition rapidly deteriorated and she, later, died due to multi-organ failure. An autopsy revealed diffuse intravascular invasion in multiple organs by the lymphoma cells. Based on our literature review-and to the best of our knowledge-there are virtually no reports describing the presentation of this lymphoma with a discernible tissue mass and associated multi-organ failure. The immunophenotypic studies performed revealed de novo CD5+ intravascular large B-cell lymphoma, which is known to be aggressive with very poor prognosis. Although it is a very rare lymphoma, it should be considered as a potential cause of multi-organ failure when no other cause has been identified. A prompt tissue diagnosis, appropriate high-dose chemotherapy and stem cell transplantation remain the only viable alternative to achieve some kind of remission.

18.
Front Oncol ; 7: 295, 2017.
Article in English | MEDLINE | ID: mdl-29250487

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) allows epithelial cancer cells to assume mesenchymal features, endowing them with enhanced motility and invasiveness, thus enabling cancer dissemination and metastatic spread. The induction of EMT is orchestrated by EMT-inducing transcription factors that switch on the expression of "mesenchymal" genes and switch off the expression of "epithelial" genes. Mitochondrial dysfunction is a hallmark of cancer and has been associated with progression to a metastatic and drug-resistant phenotype. The mechanistic link between metastasis and mitochondrial dysfunction is gradually emerging. The discovery that mitochondrial dysfunction owing to deregulated mitophagy, depletion of the mitochondrial genome (mitochondrial DNA) or mutations in Krebs' cycle enzymes, such as succinate dehydrogenase, fumarate hydratase, and isocitrate dehydrogenase, activate the EMT gene signature has provided evidence that mitochondrial dysfunction and EMT are interconnected. In this review, we provide an overview of the current knowledge on the role of different types of mitochondrial dysfunction in inducing EMT in cancer cells. We place emphasis on recent advances in the identification of signaling components in the mito-nuclear communication network initiated by dysfunctional mitochondria that promote cellular remodeling and EMT activation in cancer cells.

19.
Leuk Res ; 30(5): 547-51, 2006 May.
Article in English | MEDLINE | ID: mdl-16246419

ABSTRACT

Various subsets of extranodal marginal zone lymphomas of mucosa-associated lymphoid tissues (MALT lymphomas) have been associated with infectious organisms. Most notable of these is the association of gastric MALT lymphomas with Helicobacter pylori infection. In a recent publication Ferreri et al. [Ferreri AJ, Guidoboni M, Ponzoni M, De Conciliis C, Dell'Oro S, Fleischhauer K, et al. Evidence for an association between Chlamydia psittaci and ocular adnexal lymphomas. J Natl Cancer Inst 2004;96:586-94] reported the presence of C. psittaci DNA in 80% of 40 ocular adnexal lymphomas. Similar to the gastric MALT lymphoma data, a subset of these patients responded well to antibiotic treatment. We analyzed a set of ocular adnexal lymphomas and benign (non-neoplastic) lesions for evidence of C. psittaci DNA in patients from New York State. No evidence of C. psittaci DNA was seen in seven MALT-type ocular adnexal lymphomas, four non-MALT ocular lymphomas, one Langerhans histiocytosis, and five reactive lymphoproliferations. We eliminated several possible reasons that would cause our study to fail to find C. psittaci DNA, including the presence of PCR inhibitors, inadequate template DNA, and sequence diversity in the target region in C. psittaci. The positive data were based primarily on patients from Italy, while our study involved only patients living in the Northeastern United States. This would suggest possible geographic differences in the etiology of ocular adnexal lymphomas.


Subject(s)
Chlamydophila psittaci/genetics , Eye Neoplasms/microbiology , Lymphoma, B-Cell, Marginal Zone/microbiology , Psittacosis/microbiology , Adolescent , Adult , Aged , Aged, 80 and over , Comorbidity , DNA, Bacterial/analysis , DNA, Bacterial/isolation & purification , Eye Neoplasms/epidemiology , Eye Neoplasms/pathology , Female , Humans , Lymphoma, B-Cell, Marginal Zone/epidemiology , Lymphoma, B-Cell, Marginal Zone/pathology , Male , Middle Aged , Polymerase Chain Reaction/methods , Psittacosis/diagnosis , Psittacosis/epidemiology , United States/epidemiology
20.
Int J Oncol ; 48(6): 2722, 2016 06.
Article in English | MEDLINE | ID: mdl-27082245

ABSTRACT

After the publication of the article the authors noted the following errors in the assembling of the figures. In Fig. 3A the tubulin panel for PC-3 cells is incorrect. The correct panel is reported below. In Figs. 3B and 5B the panels are incorrect. The correct panels are shown below. These changes do not affect the interpretation of the data or conclusions of this work [the original article was published in the International Journal of Oncology 30: 217-224, 2007; DOI: 10.3892/ijo.30.1.217].

SELECTION OF CITATIONS
SEARCH DETAIL