Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Urol Oncol ; 42(7): 220.e1-220.e8, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38570271

ABSTRACT

BACKGROUND: Patients with lymph node positive (pN+) disease found at the time of radical prostatectomy with pelvic lymphadenectomy for clinically localized prostate cancer (CaP) are at high risk of disease persistence and progression. Contemporary management trends of pN+ CaP are not well described. MATERIALS AND METHODS: Patients in the Michigan Urologic Surgery Improvement Collaborative (MUSIC) with clinically localized prostate cancer who underwent radical prostatectomy between 2012 and 2023 with cN0/pN+ disease were identified. The primary outcome was to evaluate patient and practice-level factors associated with time to secondary post-RP treatment. Secondary outcomes included practice-level variation in management of pN+ CaP and rates of secondary treatment modality. To assess factors associated with secondary treatment, a Cox proportional hazards model of a 60-day landmark analysis was performed. RESULTS: We identified 666 patients with pN+ disease. Overall, 66% underwent secondary treatment within 12 months post-RP. About 19% of patients with detectable post-RP PSA did not receive treatment. Of patients receiving secondary treatment after 60-days post-RP, 34% received androgen deprivation therapy (ADT) alone, 27% received radiation (RT) alone, 36% received combination, and 4% received other systemic therapies. In the multivariable model, pathologic grade group (GG)3 (HR 1.5; 95%CI: 1.05-2.14), GG4-5 (HR 1.65; 95%CI: 1.16-2.34), positive margins (HR 1.46; 95%CI: 1.13-1.88), and detectable postoperative PSA ≥0.1 ng/ml (HR 3.46; 95%CI: 2.61-4.59) were significantly associated with secondary post-RP treatment. There was wide variation in adjusted practice-level 12-month secondary treatment utilization (28%-79%). CONCLUSIONS: The majority pN+ patients receive treatment within 12 months post-RP which was associated with high-risk pathological features and post-RP PSA. Variation in management of pN+ disease highlights the uncertainty regarding the optimal management. Understanding which patients will benefit from secondary treatment, and which type, will be critical to minimize variation in care.


Subject(s)
Prostatectomy , Prostatic Neoplasms , Quality Improvement , Humans , Male , Prostatic Neoplasms/surgery , Prostatic Neoplasms/pathology , Prostatectomy/methods , Middle Aged , Aged , Lymph Node Excision , Lymphatic Metastasis , Retrospective Studies , Lymph Nodes/pathology , Michigan
2.
Cancers (Basel) ; 15(23)2023 Nov 22.
Article in English | MEDLINE | ID: mdl-38067234

ABSTRACT

There is a need to optimize the treatment of clear cell renal cell carcinoma (ccRCC) patients at high recurrence risk after nephrectomy. We sought to elucidate the tumor immune microenvironment (TIME) of localized ccRCC and understand the prognostic and predictive characteristics of certain features. The discovery cohort was clinically localized patients in the TCGA-Kidney Renal Clear Cell Carcinoma (KIRC) project (n = 382). We identified an M0 macrophage-enriched cluster (n = 25) in the TCGA-KIRC cohort. This cluster's median progression-free survival (PFS) and overall survival (OS) were 40.4 and 45.3 months, respectively, but this was not reached in the others (p = 0.0003 and <0.0001, respectively). Gene set enrichment (GSEA) analysis revealed an enrichment of epithelial to mesenchymal transition and cell cycle progression genes within this cluster, and these patients also had a lower predicted response to immune checkpoint blockade (ICB) (4% vs. 20-34%). An M0-enriched cluster (n = 9) with shorter PFS (p = 0.0006) was also identified in the Clinical Proteomics Tumor Analysis Consortium (CPTAC) cohort (n = 94). Through this characterization of the TIME in ccRCC, a cluster of patients defined by enrichment in M0 macrophages was identified that demonstrated poor prognosis and lower predicted ICB response. Pending further validation, this signature can identify localized ccRCC patients at high risk of recurrence after nephrectomy and who may require therapeutic approaches beyond ICB monotherapy.

3.
J Urol ; 209(1): 268-269, 2023 01.
Article in English | MEDLINE | ID: mdl-36200355
4.
Urology ; 167: 109-114, 2022 09.
Article in English | MEDLINE | ID: mdl-35772487

ABSTRACT

OBJECTIVE: To understand how the lack of a physical examination during new patient video visits can impact urological surgery planning during the COVID-19 pandemic. METHODS: We retrospectively reviewed 590 consecutive urology patients who underwent new patient video visits from March through May 2020 at a single academic center. Our primary outcome was procedural plan concordance, the proportion of video visit surgical plans that remained the same after the patient was seen in-person, either in clinic or on day of surgery. Median days between video and in-person visits were compared between concordant and discordant cases using the Mann-Whitney U test; P < .05 was significant. RESULTS: Overall, 195 (33%) were evaluated by new patient video visits and had a procedure scheduled, of which, 186 (95%) had concordant plans after in-person evaluation. Further, 99% of plans for in-office procedures and 91% for operating room procedures were unchanged. Four patients (2.1%) had surgical plans altered after changes in clinical course, two (1%) due to additional imaging, and three (1.5%) based on genitourinary examination findings. Days between video visit and in-person evaluation did not differ significantly in concordant cases (median 37.5 [IQR, 16 - 80.5]) as compared to discordant cases (median 58.0 [IQR, 20 - 224]; P = .12). CONCLUSIONS: Most surgical plans developed during new patient video visits remain unchanged after in-person examination. However, changes in clinical course or updated imaging can alter operative plans. Likewise, certain urologic conditions (eg, penile cancer) rely on the genitourinary examination to dictate surgical approach.


Subject(s)
COVID-19 , Telemedicine , COVID-19/epidemiology , COVID-19/prevention & control , Humans , Operating Rooms , Pandemics/prevention & control , Physical Examination , Retrospective Studies
6.
Int J Mol Sci ; 20(17)2019 Aug 27.
Article in English | MEDLINE | ID: mdl-31461847

ABSTRACT

Neutrophils have been well-characterized for their role in the host anti-microbial response. However, it is now appreciated that neutrophils have a critical role in tumorigenesis and tumor progression in the majority of solid tumors. Recent studies have indicated a critical role for hypoxia in regulating neutrophil function in tumors. Furthermore, neutrophil-specific expression of hypoxia-inducible transcription factors may represent a novel therapeutic target for human cancer. In this review, we highlight the function of neutrophils in cancer and the role of the neutrophil hypoxic response in regulating the neoplastic progression of cancer.


Subject(s)
Neoplasms/immunology , Neutrophils/immunology , Oxygen/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Hypoxia , Humans , Tumor Microenvironment/immunology
7.
Nat Commun ; 10(1): 660, 2019 02 08.
Article in English | MEDLINE | ID: mdl-30737385

ABSTRACT

Microfold cells (M-cells) are specialized cells of the intestine that sample luminal microbiota and dietary antigens to educate the immune cells of the intestinal lymphoid follicles. The function of M-cells in systemic inflammatory responses are still unclear. Here we show that epithelial non-canonical NFkB signaling mediated by NFkB-inducing kinase (NIK) is highly active in intestinal lymphoid follicles, and is required for M-cell maintenance. Intestinal NIK signaling modulates M-cell differentiation and elicits both local and systemic IL-17A and IgA production. Importantly, intestinal NIK signaling is active in mouse models of colitis and patients with inflammatory bowel diseases; meanwhile, constitutive NIK signaling increases the susceptibility to inflammatory injury by inducing ectopic M-cell differentiation and a chronic increase of IL-17A. Our work thus defines an important function of non-canonical NFkB and M-cells in immune homeostasis, inflammation and polymicrobial sepsis.


Subject(s)
NF-kappa B/metabolism , Animals , B-Lymphocytes/metabolism , Blotting, Western , Colitis/immunology , Colitis/metabolism , Colon/metabolism , Colon/pathology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunoglobulin A/metabolism , Interleukin-17/metabolism , Intestines/immunology , Mice , Protein Serine-Threonine Kinases , RNA, Ribosomal, 16S/genetics , Sepsis/genetics , Sepsis/metabolism , Signal Transduction/physiology
8.
Gastroenterology ; 156(5): 1467-1482, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30550822

ABSTRACT

BACKGROUND & AIMS: Neutrophils are among the most prevalent immune cells in the microenvironment of colon tumors; they are believed to promote growth of colon tumors, and their numbers correlate with outcomes of patients with colon cancer. Trials of inhibitors of neutrophil trafficking are underway in patients with cancer, but it is not clear how neutrophils contribute to colon tumorigenesis. METHODS: Colitis-associated colon cancer was induced in mice with conditional deletion of neutrophils (LysMCre;Mcl1fl/fl) and wild-type littermates (LysMCre;Mcl1wt/wt, control mice) by administration of azoxythmethane and/or dextran sulfate sodium. Sporadic colon tumorigenesis was assessed in neutrophil-deficient and neutrophil-replete mice with conditional deletion of colon epithelial Apc (Cdx2-CreERT2;Apcfl/fl). Primary colon tumor tissues from these mice were assessed by histology, RNA sequencing, quantitative polymerase chain reaction, and fluorescence in situ hybridization analyses. Fecal and tumor-associated microbiota were assessed by 16s ribosomal RNA sequencing. RESULTS: In mice with inflammation-induced and sporadic colon tumors, depletion of neutrophils increased the growth, proliferation, and invasiveness of the tumors. RNA sequencing analysis identified genes that regulate antimicrobial and inflammatory processes that were dysregulated in neutrophil-deficient colon tumors compared with colon tumors from control mice. Neutrophil depletion correlated with increased numbers of bacteria in tumors and proliferation of tumor cells, tumor-cell DNA damage, and an inflammatory response mediated by interleukin 17 (IL17). The 16s ribosomal RNA sequencing identified significant differences in the composition of the microbiota between colon tumors from neutrophil-deficient vs control mice. Administration of antibiotics or a neutralizing antibody against IL17 to neutrophil-deficient mice resulted in development of less-invasive tumors compared with mice given vehicle. We found bacteria in tumors to induce production of IL17, which promotes influx of intratumor B cells that promote tumor growth and progression. CONCLUSIONS: In comparisons of mice with vs without neutrophils, we found neutrophils to slow colon tumor growth and progression by restricting numbers of bacteria and tumor-associated inflammatory responses.


Subject(s)
Adenocarcinoma/immunology , Bacteria/growth & development , Cell Movement , Cell Proliferation , Colonic Neoplasms/immunology , Neutrophils/immunology , Adenocarcinoma/genetics , Adenocarcinoma/microbiology , Adenocarcinoma/pathology , Animals , Anti-Bacterial Agents/pharmacology , Antibodies, Neutralizing/pharmacology , Azoxymethane , Bacteria/drug effects , Bacteria/immunology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/pathology , Colonic Neoplasms/genetics , Colonic Neoplasms/microbiology , Colonic Neoplasms/pathology , Dextran Sulfate , Disease Models, Animal , Disease Progression , Female , Host-Pathogen Interactions , Interleukin-17/antagonists & inhibitors , Interleukin-17/immunology , Male , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Invasiveness , Neutrophils/drug effects , Tumor Burden , Tumor Microenvironment
9.
Mol Cell Biol ; 38(22)2018 11 15.
Article in English | MEDLINE | ID: mdl-30181395

ABSTRACT

Myc-associated zinc finger (MAZ) is a transcription factor highly upregulated in chronic inflammatory disease and several human cancers. In the present study, we found that MAZ protein is highly expressed in human ulcerative colitis and colon cancer. However, the precise role for MAZ in the progression of colitis and colon cancer is not well defined. To determine the function of MAZ, a novel mouse model of intestinal epithelial cell-specific MAZ overexpression was generated. Expression of MAZ in intestinal epithelial cells was sufficient to enhance inflammatory injury in two complementary models of colitis. Moreover, MAZ expression increased tumorigenesis in an in vivo model of inflammation-induced colon cancer and was important for growth of human colon cancer cell lines in vitro and in vivo Mechanistically, MAZ is critical in the regulation of oncogenic STAT3 signaling. MAZ-expressing mice have enhanced STAT3 activation in the acute response to colitis. Moreover, MAZ was essential for cytokine- and bacterium-induced STAT3 signaling in colon cancer cells. Furthermore, we show that STAT3 is essential for MAZ-induced colon tumorigenesis using a chemical inhibitor. These data indicate an important functional role for MAZ in the inflammatory progression of colon cancer through regulation of STAT3 signaling and suggest that MAZ is a potential therapeutic target to dampen STAT3 signaling in colon cancer.


Subject(s)
Colitis/metabolism , Colonic Neoplasms/metabolism , DNA-Binding Proteins/metabolism , Inflammation/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism , Zinc Fingers/physiology , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Cell Proliferation/physiology , Colitis/pathology , Colonic Neoplasms/pathology , Cytokines/metabolism , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/pathology , HCT116 Cells , HT29 Cells , Humans , Inflammation/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Up-Regulation/physiology
10.
Cell Metab ; 28(1): 69-86.e6, 2018 Jul 03.
Article in English | MEDLINE | ID: mdl-29972798

ABSTRACT

Although breast cancer stem cells (BCSCs) display plasticity transitioning between quiescent mesenchymal-like (M) and proliferative epithelial-like (E) states, how this plasticity is regulated by metabolic or oxidative stress remains poorly understood. Here, we show that M- and E-BCSCs rely on distinct metabolic pathways and display markedly different sensitivities to inhibitors of glycolysis and redox metabolism. Metabolic or oxidative stress generated by 2DG, H2O2, or hypoxia promotes the transition of ROSlo M-BCSCs to a ROShi E-state. This transition is reversed by N-acetylcysteine and mediated by activation of the AMPK-HIF1α axis. Moreover, E-BCSCs exhibit robust NRF2-mediated antioxidant responses, rendering them vulnerable to ROS-induced differentiation and cytotoxicity following suppression of NRF2 or downstream thioredoxin (TXN) and glutathione (GSH) antioxidant pathways. Co-inhibition of glycolysis and TXN and GSH pathways suppresses tumor growth, tumor-initiating potential, and metastasis by eliminating both M- and E-BCSCs. Exploiting metabolic vulnerabilities of distinct BCSC states provides a novel therapeutic approach targeting this critical tumor cell population.


Subject(s)
Acetylcysteine/metabolism , Breast Neoplasms/pathology , Cell Transformation, Neoplastic , Neoplastic Stem Cells/metabolism , Protein Kinases/metabolism , Reactive Oxygen Species/metabolism , AMP-Activated Protein Kinase Kinases , Animals , Antioxidants/metabolism , Cell Line, Tumor , Female , Glucose/metabolism , Glutathione/metabolism , Glycolysis , Humans , Mice, Inbred NOD , NF-E2-Related Factor 2/metabolism , Neoplastic Stem Cells/cytology , Oxidation-Reduction , Oxidative Stress , Signal Transduction , Stress, Physiological , Thioredoxins/metabolism , Xenograft Model Antitumor Assays
11.
Mol Cell Biol ; 37(5)2017 03 01.
Article in English | MEDLINE | ID: mdl-27956697

ABSTRACT

Inflammation is a significant risk factor for colon cancer. Recent work has demonstrated essential roles for several infiltrating immune populations in the metaplastic progression following inflammation. Hypoxia and stabilization of hypoxia-inducible factors (HIFs) are hallmark features of inflammation and solid tumors. Previously, we demonstrated an important role for tumor epithelial HIF-2α in colon tumors; however, the function of epithelial HIF-2α as a critical link in the progression of inflammation to cancer has not been elucidated. In colitis-associated colon cancer models, epithelial HIF-2α was essential in tumor growth. Concurrently, epithelial disruption of HIF-2α significantly decreased neutrophils in the colon tumor microenvironment. Intestinal epithelial HIF-2α-overexpressing mice demonstrated that neutrophil recruitment was a direct response to increased epithelial HIF-2α signaling. High-throughput RNA sequencing (RNA-seq) analysis of HIF-2α-overexpressing mice in conjunction with data mining from the Cancer Genome Atlas showed that the neutrophil chemokine CXCL1 gene was highly upregulated in colon tumor epithelium in a HIF-2α-dependent manner. Using selective peptide inhibitors of the CXCL1-CXCR2 signaling axis identified HIF-2α-dependent neutrophil recruitment as an essential mechanism to increase colon carcinogenesis. These studies demonstrate that HIF-2α is a novel regulator of neutrophil recruitment to colon tumors and that it is essential in shaping the protumorigenic inflammatory microenvironment in colon cancer.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Disease Progression , Epithelial Cells/metabolism , Neutrophil Infiltration , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Chemokine CXCL1/genetics , Chemokine CXCL1/metabolism , Chemotaxis , Colitis/complications , Colitis/pathology , Colonic Neoplasms/complications , Colonic Neoplasms/genetics , DNA-Binding Proteins/metabolism , Epithelial Cells/pathology , Gene Expression Regulation, Neoplastic , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation/metabolism , Inflammation/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Neutrophils/metabolism , Neutrophils/pathology , Promoter Regions, Genetic/genetics , Transcription Factors/metabolism
12.
Cell Metab ; 24(3): 447-461, 2016 09 13.
Article in English | MEDLINE | ID: mdl-27546461

ABSTRACT

Dietary iron intake and systemic iron balance are implicated in colorectal cancer (CRC) development, but the means by which iron contributes to CRC are unclear. Gene expression and functional studies demonstrated that the cellular iron importer, divalent metal transporter 1 (DMT1), is highly expressed in CRC through hypoxia-inducible factor 2α-dependent transcription. Colon-specific Dmt1 disruption resulted in a tumor-selective inhibitory effect of proliferation in mouse colon tumor models. Proteomic and genomic analyses identified an iron-regulated signaling axis mediated by cyclin-dependent kinase 1 (CDK1), JAK1, and STAT3 in CRC progression. A pharmacological inhibitor of DMT1 antagonized the ability of iron to promote tumor growth in a CRC mouse model and a patient-derived CRC enteroid orthotopic model. Our studies implicate a growth-promoting signaling network instigated by elevated intracellular iron levels in tumorigenesis, offering molecular insights into how a key dietary component may contribute to CRC.


Subject(s)
Carcinogenesis/pathology , Cation Transport Proteins/metabolism , Cell Cycle , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Iron/metabolism , Janus Kinases/metabolism , STAT3 Transcription Factor/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , CDC2 Protein Kinase/metabolism , Carcinogenesis/metabolism , Cation Transport Proteins/genetics , Cell Cycle/drug effects , Cell Proliferation/drug effects , Colitis/complications , Colitis/pathology , Colon/pathology , Colorectal Neoplasms/genetics , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Humans , Inflammation/complications , Inflammation/pathology , Iron/pharmacology , Signal Transduction/drug effects
13.
J Clin Invest ; 126(10): 3689-3698, 2016 10 03.
Article in English | MEDLINE | ID: mdl-27525434

ABSTRACT

The tumor immune response is in a dynamic balance between antitumor mechanisms, which serve to decrease cancer growth, and the protumor inflammatory response, which increases immune tolerance, cell survival, and proliferation. Hypoxia and expression of HIF-1α and HIF-2α are characteristic features of all solid tumors. HIF signaling serves as a major adaptive mechanism in tumor growth in a hypoxic microenvironment. HIFs represent a critical signaling node in the switch to protumorigenic inflammatory responses through recruitment of protumor immune cells and altered immune cell effector functions to suppress antitumor immune responses and promote tumor growth through direct growth-promoting cytokine production, angiogenesis, and ROS production. Modulating HIF function will be an important mechanism to dampen the tumor-promoting inflammatory response and inhibit cancer growth.


Subject(s)
Gene Expression Regulation, Neoplastic , Hypoxia-Inducible Factor 1/physiology , Neoplasms/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/physiology , Cell Hypoxia , Humans , Inflammation/metabolism , Neoplasms/immunology , Neoplasms/pathology , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/metabolism , Signal Transduction , Tumor Microenvironment/immunology
14.
Sci Signal ; 8(397): ra98, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26443705

ABSTRACT

Yes-associated protein 1 (YAP1) is a transcriptional coactivator in the Hippo signaling pathway. Increased YAP1 activity promotes the growth of tumors, including that of colorectal cancer (CRC). Verteporfin, a drug that enhances phototherapy to treat neovascular macular degeneration, is an inhibitor of YAP1. We found that verteporfin inhibited tumor growth independently of its effects on YAP1 or the related protein TAZ in genetically or chemically induced mouse models of CRC, in patient-derived xenografts, and in enteroid models of CRC. Instead, verteporfin exhibited in vivo selectivity for killing tumor cells in part by impairing the global clearance of high-molecular weight oligomerized proteins, particularly p62 (a sequestrome involved in autophagy) and STAT3 (signal transducer and activator of transcription 3; a transcription factor). Verteporfin inhibited cytokine-induced STAT3 activity and cell proliferation and reduced the viability of cultured CRC cells. Although verteporfin accumulated to a greater extent in normal cells than in tumor cells in vivo, experiments with cultured cells indicated that the normal cells efficiently cleared verteporfin-induced protein oligomers through autophagic and proteasomal pathways. Culturing CRC cells under hypoxic or nutrient-deprived conditions (modeling a typical CRC microenvironment) impaired the clearance of protein oligomers and resulted in cell death, whereas culturing cells under normoxic or glucose-replete conditions protected cell viability and proliferation in the presence of verteporfin. Furthermore, verteporfin suppressed the proliferation of other cancer cell lines even in the absence of YAP1, suggesting that verteporfin may be effective against multiple types of solid cancers.


Subject(s)
Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adenocarcinoma/drug therapy , Adenoma/drug therapy , Antineoplastic Agents/pharmacology , Colonic Neoplasms/drug therapy , Neoplasm Proteins/drug effects , Phosphoproteins/antagonists & inhibitors , Porphyrins/pharmacology , Acyltransferases , Adaptor Proteins, Signal Transducing/physiology , Adenocarcinoma/pathology , Adenoma/pathology , Adenomatous Polyposis Coli/drug therapy , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/pathology , Animals , Apoptosis/drug effects , Autophagy/drug effects , Cell Division/drug effects , Cell Line, Tumor , Colonic Neoplasms/chemically induced , Colonic Neoplasms/pathology , Genes, APC , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Weight , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/physiology , Phosphoproteins/physiology , Phosphorylation , Proteasome Endopeptidase Complex/drug effects , Protein Multimerization/drug effects , Protein Processing, Post-Translational , STAT3 Transcription Factor/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Transcription, Genetic/drug effects , Verteporfin , Xenograft Model Antitumor Assays , YAP-Signaling Proteins
15.
Blood ; 126(20): 2291-301, 2015 Nov 12.
Article in English | MEDLINE | ID: mdl-26400962

ABSTRACT

The serine-threonine kinase ataxia-telangiectasia mutated (ATM) plays a central role in maintaining genomic integrity. In mice, ATM deficiency is exclusively associated with T-cell lymphoma development, whereas B-cell tumors predominate in human ataxia-telangiectasia patients. We demonstrate in this study that when T cells are removed as targets for lymphomagenesis and as mediators of immune surveillance, ATM-deficient mice exclusively develop early-onset immunoglobulin M(+) B-cell lymphomas that do not transplant to immunocompetent mice and that histologically and genetically resemble the activated B cell-like (ABC) subset of human diffuse large B-cell lymphoma (DLBCL). These B-cell lymphomas show considerable chromosomal instability and a recurrent genomic amplification of a 4.48-Mb region on chromosome 18 that contains Malt1 and is orthologous to a region similarly amplified in human ABC DLBCL. Of importance, amplification of Malt1 in these lymphomas correlates with their dependence on nuclear factor (NF)-κB, MALT1, and B-cell receptor (BCR) signaling for survival, paralleling human ABC DLBCL. Further, like some human ABC DLBCLs, these mouse B-cell lymphomas also exhibit constitutive BCR-dependent NF-κB activation. This study reveals that ATM protects against development of B-cell lymphomas that model human ABC DLBCL and identifies a potential role for T cells in preventing the emergence of these tumors.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/deficiency , Immunologic Surveillance , Lymphoma, Large B-Cell, Diffuse/immunology , Tumor Suppressor Proteins/deficiency , Animals , Ataxia Telangiectasia Mutated Proteins/immunology , Caspases/genetics , Caspases/immunology , Cell Line, Tumor , Chromosomal Instability/immunology , Genetic Loci/immunology , Humans , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Mice , Mice, Knockout , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , NF-kappa B/genetics , NF-kappa B/immunology , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Tumor Suppressor Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...