Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Immunol Res ; 3(9): 1017-29, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26014097

ABSTRACT

Renal cell carcinoma (RCC) is an immunogenic and proangiogenic cancer, and antiangiogenic therapy is the current mainstay of treatment. Patients with RCC develop innate or adaptive resistance to antiangiogenic therapy. There is a need to identify biomarkers that predict therapeutic resistance and guide combination therapy. We assessed the interaction between antiangiogenic therapy and the tumor immune microenvironment and determined their impact on clinical outcome. We found that antiangiogenic therapy-treated RCC primary tumors showed increased infiltration of CD4(+) and CD8(+) T lymphocytes, which was inversely related to patient overall survival and progression-free survival. Furthermore, specimens from patients treated with antiangiogenic therapy showed higher infiltration of CD4(+)FOXP3(+) regulatory T cells and enhanced expression of checkpoint ligand programed death-ligand 1 (PD-L1). Both immunosuppressive features were correlated with T-lymphocyte infiltration and were negatively related to patient survival. Treatment of RCC cell lines and RCC xenografts in immunodeficient mice with sunitinib also increased tumor PD-L1 expression. Results from this study indicate that antiangiogenic treatment may both positively and negatively regulate the tumor immune microenvironment. These findings generate hypotheses on resistance mechanisms to antiangiogenic therapy and will guide the development of combination therapy with PD-1/PD-L1-blocking agents.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Carcinoma, Renal Cell/secondary , Drug Resistance, Neoplasm/immunology , Kidney Neoplasms/drug therapy , Tumor Microenvironment/immunology , Angiogenesis Inhibitors/pharmacology , Animals , Antineoplastic Agents/pharmacology , B7-H1 Antigen/biosynthesis , Bevacizumab/pharmacology , Biomarkers, Tumor/biosynthesis , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/immunology , Heterografts , Humans , Immune Tolerance , Indoles/pharmacology , Kidney Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Mice, Nude , Neoplasm Transplantation , Pyrroles/pharmacology , Sunitinib , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Tumor Cells, Cultured , Tumor Microenvironment/drug effects , Up-Regulation/drug effects
2.
Oncotarget ; 6(6): 3563-77, 2015 Feb 28.
Article in English | MEDLINE | ID: mdl-25784651

ABSTRACT

MYC-induced T-ALL exhibit oncogene addiction. Addiction to MYC is a consequence of both cell-autonomous mechanisms, such as proliferative arrest, cellular senescence, and apoptosis, as well as non-cell autonomous mechanisms, such as shutdown of angiogenesis, and recruitment of immune effectors. Here, we show, using transgenic mouse models of MYC-induced T-ALL, that the loss of either p19ARF or p53 abrogates the ability of MYC inactivation to induce sustained tumor regression. Loss of p53 or p19ARF, influenced the ability of MYC inactivation to elicit the shutdown of angiogenesis; however the loss of p19ARF, but not p53, impeded cellular senescence, as measured by SA-beta-galactosidase staining, increased expression of p16INK4A, and specific histone modifications. Moreover, comparative gene expression analysis suggested that a multitude of genes involved in the innate immune response were expressed in p19ARF wild-type, but not null, tumors upon MYC inactivation. Indeed, the loss of p19ARF, but not p53, impeded the in situ recruitment of macrophages to the tumor microenvironment. Finally, p19ARF null-associated gene signature prognosticated relapse-free survival in human patients with ALL. Therefore, p19ARF appears to be important to regulating cellular senescence and innate immune response that may contribute to the therapeutic response of ALL.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/immunology , Genes, myc , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/immunology , Animals , Cellular Senescence/genetics , Cellular Senescence/immunology , Disease Models, Animal , Gene Silencing , Humans , Immunity, Innate , Mice , Mice, Knockout , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/immunology
3.
Mob DNA ; 2(1): 2, 2011 Feb 14.
Article in English | MEDLINE | ID: mdl-21320307

ABSTRACT

Long interspersed elements, type 1(LINE-1, L1) are the most abundant and only active autonomous retrotransposons in the human genome. Native L1 elements are inefficiently expressed because of a transcription elongation defect thought to be caused by high adenosine content in L1 sequences. Previously, we constructed a highly active synthetic mouse L1 element (ORFeus-Mm), partially by reducing the nucleotide composition bias. As a result, the transcript abundance of ORFeus-Mm was greatly increased, and its retrotransposition frequency was > 200-fold higher than its native counterpart. In this paper, we report a synthetic human L1 element (ORFeus-Hs) synthesized using a similar strategy. The adenosine content of the L1 open reading frames (ORFs) was reduced from 40% to 27% by changing 25% of the bases in the ORFs, without altering the amino acid sequence. By studying a series of native/synthetic chimeric elements, we observed increased levels of full-length L1 RNA and ORF1 protein and retrotransposition frequency, mostly proportional to increased fraction of synthetic sequence. Overall, the fully synthetic ORFeus-Hs has > 40-fold more RNA but is at most only ~threefold more active than its native counterpart (L1RP); however, its absolute retrotransposition activity is similar to ORFeus-Mm. Owing to the elevated expression of the L1 RNA/protein and its high retrotransposition ability, ORFeus-Hs and its chimeric derivatives will be useful tools for mechanistic L1 studies and mammalian genome manipulation.

4.
Genes Dev ; 24(12): 1281-94, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20551174

ABSTRACT

The Myc protein suppresses the transcription of several cyclin-dependent kinase inhibitors (CKIs) via binding to Miz1; whether this interaction is important for Myc's ability to induce or maintain tumorigenesis is not known. Here we show that the oncogenic potential of a point mutant of Myc (MycV394D) that is selectively deficient in binding to Miz1 is greatly attenuated. Binding of Myc to Miz1 is continuously required to repress CKI expression and inhibit accumulation of trimethylated histone H3 at Lys 9 (H3K9triMe), a hallmark of cellular senescence, in T-cell lymphomas. Lymphomas that arise express high amounts of transforming growth factor beta-2 (TGFbeta-2) and TGFbeta-3. Upon Myc suppression, TGFbeta signaling is required to induce CKI expression and cellular senescence and suppress tumor recurrence. Binding of Myc to Miz1 is required to antagonize growth suppression and induction of senescence by TGFbeta. We demonstrate that, since lymphomas express high levels of TGFbeta, they are poised to elicit an autocrine program of senescence upon Myc inactivation, demonstrating that TGFbeta is a key factor that establishes oncogene addiction of T-cell lymphomas.


Subject(s)
Autocrine Communication/physiology , Lymphoma, T-Cell/physiopathology , Nuclear Proteins/metabolism , Protein Inhibitors of Activated STAT/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Transforming Growth Factor beta/metabolism , Animals , Doxycycline/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , HeLa Cells , Humans , Mice , Mice, Transgenic , Mutation/genetics , Proto-Oncogene Proteins c-myc/genetics , Ubiquitin-Protein Ligases
5.
Nat Rev Cancer ; 10(4): 301-9, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20332779

ABSTRACT

MYC regulates the transcription of thousands of genes required to coordinate a range of cellular processes, including those essential for proliferation, growth, differentiation, apoptosis and self-renewal. Recently, MYC has also been shown to serve as a direct regulator of ribosome biogenesis. MYC coordinates protein synthesis through the transcriptional control of RNA and protein components of ribosomes, and of gene products required for the processing of ribosomal RNA, the nuclear export of ribosomal subunits and the initiation of mRNA translation. We discuss how the modulation of ribosome biogenesis by MYC may be essential to its physiological functions as well as its pathological role in tumorigenesis.


Subject(s)
Genes, myc , Proto-Oncogene Proteins c-myc/metabolism , Ribosomes/physiology , Gene Expression Regulation , Homeostasis , Humans , Models, Genetic , Neoplasms/genetics , Protein Biosynthesis , Proto-Oncogene Proteins c-myc/genetics , RNA, Messenger/genetics , Ribosomal Proteins/genetics , Ribosomes/metabolism
6.
Proc Natl Acad Sci U S A ; 104(32): 13028-33, 2007 Aug 07.
Article in English | MEDLINE | ID: mdl-17664422

ABSTRACT

Oncogene-induced senescence is an important mechanism by which normal cells are restrained from malignant transformation. Here we report that the suppression of the c-Myc (MYC) oncogene induces cellular senescence in diverse tumor types including lymphoma, osteosarcoma, and hepatocellular carcinoma. MYC inactivation was associated with prototypical markers of senescence, including acidic beta-gal staining, induction of p16INK4a, and p15INK4b expression. Moreover, MYC inactivation induced global changes in chromatin structure associated with the marked reduction of histone H4 acetylation and increased histone H3 K9 methylation. Osteosarcomas engineered to be deficient in p16INK4a or Rb exhibited impaired senescence and failed to exhibit sustained tumor regression upon MYC inactivation. Similarly, only after lymphomas were repaired for p53 expression did MYC inactivation induce robust senescence and sustained tumor regression. The pharmacologic inhibition of signaling pathways implicated in oncogene-induced senescence including ATM/ATR and MAPK did not prevent senescence associated with MYC inactivation. Our results suggest that cellular senescence programs remain latently functional, even in established tumors, and can become reactivated, serving as a critical mechanism of oncogene addiction associated with MYC inactivation.


Subject(s)
Cellular Senescence , Genes, myc/physiology , Neoplasms/pathology , Animals , Chromatin/chemistry , DNA Damage , Genes, Retinoblastoma , Genes, p53 , Humans , MAP Kinase Signaling System , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...