Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Res ; 68(17): 7130-6, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18757428

ABSTRACT

Nitric oxide (NO(*)), an important signaling molecule and a component of inflammatory response, is involved in tumorigenesis. However, the quantity of NO(*) and the cellular microenvironment influences the role of NO(*) in tumor development. We used a genetic strategy to test the hypothesis that an inflammatory microenvironment with an enhanced level of NO(*) accelerates spontaneous tumor development. C. parvum-induced inflammation and increased NO(*) synthase-2 (NOS2) expression coincided with accelerated spontaneous tumor development, mostly lymphomas, in p53-/-NOS2+/+ C57BL6 mice when compared with the controls (P = 0.001). However, p53-/-NOS2-/- mice did not show any difference in tumor latency between C. parvum-treated and control groups. In C. parvum-treated p53-/-NOS2+/+ mice, tumor development was preceded by a higher expression of NOS2 and phosphorylated Akt-Ser(473) (pAkt-Ser473) in spleen, increased cell proliferation measured by Ki-67 IHC in spleen and thymus, and a lower apoptotic index and CD95-L expression in spleen and thymus. C. parvum-treated p53-/-NOS2+/+ mice showed an increase in the number of Foxp3(+) T-reg cells, dendritic cells (DC), as well as increased CD80(+), CD86(+), CD40(+), and CD83(+) on DC in the spleen. Regulatory T-cells (T-reg) and the maturation of DC may modulate tumorigenesis. An increase in the FoxP3(+)T-reg cells in C. parvum-treated p53-/-NOS2+/+ mice indicates a role of NO(*) in the regulation of T-reg cells that may contribute to a protumor shift of the immune environment favoring an accelerated tumor development. These data provide genetic and mechanistic evidence that an inflammatory microenvironment and an increased level of NO(*) can accelerate tumor development.


Subject(s)
Inflammation/pathology , Neoplasms, Experimental/pathology , Nitric Oxide/physiology , Animals , Apoptosis , Flow Cytometry , Immunohistochemistry , Interferon-gamma/metabolism , Interleukin-6/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/metabolism , Tumor Necrosis Factor-alpha/metabolism
2.
Cancer Res ; 64(19): 6849-53, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15466171

ABSTRACT

Inflammation influences the development of cancer. The nitric oxide synthase (NOS2) is induced by inflammatory cytokines, e.g., tumor necrosis factor alpha and interleukin 1beta, and produces nitric oxide (NO*), a critical mediator of the inflammatory response. Because p53 governs NO* production by transcriptionally transrepressing NOS2, we used a genetic strategy to determine whether NO* and p53 cooperatively regulate tumorigenesis. Lymphomas developed more rapidly in p53-/-NOS2-/- or p53-/-NOS2+/- mice than in p53-/-NOS2+/+ mice that were cross-bred into a >95% C57BL6 background and maintained in a pathogen-free condition. Likewise, sarcomas and lymphomas developed faster in p53+/-NOS2-/- or p53+/-NOS2+/- than in p53+/-NOS2+/+ mice. When compared with the double knockout mice, p53-/-NOS2+/+ mice showed a higher apoptotic index and a decreased proliferation index with an increased expression of death receptor ligands, CD95-L and tumor necrosis factor-related apoptosis-inducing ligand, and the cell cycle checkpoint protein, p21(waf1), in the spleen and thymus before tumor development. Furthermore, mice deficient in both p53 and NOS2 produced a high level of anti-inflammatory interleukin 10 when compared with p53-deficient mice. These studies provide genetic and mechanistic evidence that NO* can suppress tumorigenesis.


Subject(s)
Inflammation Mediators/metabolism , Lymphoma, T-Cell/metabolism , Nitric Oxide/metabolism , Sarcoma, Experimental/metabolism , Animals , Apoptosis/physiology , Apoptosis Regulatory Proteins , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/biosynthesis , Disease Models, Animal , Fas Ligand Protein , Female , Inbreeding , Interleukin-10/biosynthesis , Ki-67 Antigen/biosynthesis , Lymphoma, T-Cell/enzymology , Lymphoma, T-Cell/pathology , Male , Membrane Glycoproteins/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase/deficiency , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Sarcoma, Experimental/enzymology , Sarcoma, Experimental/pathology , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
3.
Am J Pathol ; 165(3): 1007-18, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15331424

ABSTRACT

CACNA2D2 is a putative tumor suppressor gene located in the human chromosome 3p21.3 region that shows frequent allelic imbalances in lung, breast, and other cancers. The alpha2delta-2 protein encoded by the gene is a regulatory subunit of voltage-dependent calcium channels and is expressed in brain, heart, and other tissues. Here we report that mice homozygous for targeted disruption of the Cacna2d2 gene exhibit growth retardation, reduced life span, ataxic gait with apoptosis of cerebellar granule cells followed by Purkinje cell depletion, enhanced susceptibility to seizures, and cardiac abnormalities. The Cacna2d2(tm1NCIF) null phenotype has much in common with that of Cacna1a mutants, such as cerebellar neuro-degeneration associated with ataxia, seizures, and premature death. A tendency to bradycardia and limited response of null mutants to isoflurane implicate alpha2delta-2 in sympathetic regulation of cardiac function. In summary, our findings provide genetic evidence that the alpha2delta-2 subunit serves in vivo as a component of P/Q-type calcium channels, is indispensable for the central nervous system function, and may be involved in hereditary cerebellar ataxias and epileptic disorders in humans.


Subject(s)
Calcium Channels/physiology , Cerebellar Ataxia/pathology , Disease Models, Animal , Growth Disorders/pathology , Heart Diseases/pathology , Seizures/pathology , Animals , Blood Pressure , Calcium Channels/genetics , Cerebellar Ataxia/genetics , Cerebellar Ataxia/mortality , Electrocardiography , Female , Gene Deletion , Gene Targeting , Genes, Tumor Suppressor , Growth Disorders/genetics , Growth Disorders/metabolism , Heart Diseases/genetics , Heart Diseases/metabolism , Homozygote , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Neurologic Mutants , Motor Activity , Purkinje Cells/metabolism , Purkinje Cells/pathology , Seizures/genetics , Seizures/mortality
SELECTION OF CITATIONS
SEARCH DETAIL
...