151
|
Ivanova K, Das PK, van den Wijngaard RM, Lenz W, Klockenbring T, Malcharzyk V, Drummer C, Gerzer R. Differential expression of functional guanylyl cyclases in melanocytes: absence of nitric-oxide-sensitive isoform in metastatic cells. J Invest Dermatol 2001; 116:409-16. [PMID: 11231315 DOI: 10.1046/j.1523-1747.2001.01255.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Nitric oxide (NO) is a reactive endogenous molecule with multiple functions and its cellular signaling activity is mainly mediated by activation of the soluble isoform of guanylyl cyclase, a heterodimeric (alpha/beta) hemeprotein. The expression of the NO-sensitive soluble isoform of guanylyl cyclase was studied in various cultured melanocytic cells by measuring the accumulation of guanosine 3',5'-cyclic monophosphate in the presence and absence of NO donors. Here we report that 3-morpholino-sydnonimine, a donor of NO redox species, and (Z)-1-[2- (2-aminoethyl)-N-(2-ammonioethyl)amino]diazen-1-ium-1,2-diolate, a direct NO donor, induced a 20-fold increase in intracellular guanosine 3',5'-cyclic monophosphate in nonmetastatic melanoma cells and normal melanocytes in culture that could be related to cellular melanin content in a concentration-dependent manner. The increased intracellular guanosine 3',5'-cyclic monophosphate was due to stimulation of the activity of soluble guanylyl cyclase as such increase was completely abolished by using a specific inhibitor of soluble guanylyl cyclase. The involvement of functional soluble guanylyl cyclase was further confirmed by the presence of alpha1 and beta1 subunits in these cells at both mRNA and protein levels. In contrast, none of the NO donors induced guanosine 3',5'-cyclic monophosphate production in metastatic melanoma cells, which could be attributed to the absence of the beta1 subunit that is essential for catalytic activity of the soluble isoform of guanylyl cyclase. Metastatic melanoma cells produced higher levels of intracellular guanosine 3',5'-cyclic monophosphate in response to natriuretic peptides than other cell types, however, due to upregulation of membrane-bound guanylyl cyclase activities, but they are less pigmented or unpigmented. The present finding suggests that NO signaling in association with melanogenesis is dependent on the soluble isoform of guanylyl cyclase, whereas absence of soluble guanylyl cyclase but the presence of membrane-bound guanylyl cyclase correlates with the metastatic behavior of melanoma cells.
Collapse
Affiliation(s)
- K Ivanova
- Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
152
|
Kong G, Kim EK, Kim WS, Lee YW, Lee JK, Paik SW, Rhee JC, Choi KW, Lee KT. Inducible nitric oxide synthase (iNOS) immunoreactivity and its relationship to cell proliferation, apoptosis, angiogenesis, clinicopathologic characteristics, and patient survival in pancreatic cancer. INTERNATIONAL JOURNAL OF PANCREATOLOGY : OFFICIAL JOURNAL OF THE INTERNATIONAL ASSOCIATION OF PANCREATOLOGY 2001; 29:133-140. [PMID: 12067216 DOI: 10.1385/ijgc:29:3:133] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND The clinicopathological and biological significance of the expression of iNOS in pancreatic cancer remains unclear. The goal of this study was to determine the possible roles and clinical significance of iNOS expression in pancreatic cancer. METHODS Seventy-two pancreatic adenocarcinoma tissue specimens were obtained by surgical resection. We investigated the immunohistochemical expression of iNOS in 72 patients with pancreatic cancer with respect to variable clinicopathological characteristics, proliferation activity (assessed by Ki-67 expression), apoptosis (assessed by TUNEL stain), and microvessel density (assessed by CD34 expression; angiogenesis). RESULTS Immunohistochemical investigations demonstrated immunolabeling of tumor cells with anti-iNOS antibody. Positivity for iNOS was observed in 48/72 (66.7%). The expression of iNOS protein did not correlate with age, bilirubin, tumor marker, location, size, AJCC stage, differentiation, distant metastasis, or patient survival. No significant association was found between iNOS expression and proliferation or microvessel density in pancreatic cancer. Apoptotic index (AI) of positive iNOS expressions were significantly higher than negative expression (p < 0.001). CONCLUSION Inducible nitric oxide synthase (iNOS) is expressed by human pancreatic cancer, and its presence is positively correlated with apoptosis of cancer cells that could provide the basis for the development of therapeutic strategies in human pancreatic cancer.
Collapse
Affiliation(s)
- G Kong
- Department of Pathology, Hanyang University School of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
153
|
Abstract
Tumor immunity involves a concerted interplay between cytokines and effector cells. Extensive efforts have focused on understanding the roles of cytokines and their interactions with effector cells for the production of effective tumor immunity. One cytokine that is well recognized to play a central role in coordinating tumor immune responses is IFN-gamma. IFN-gamma exerts its biological effects through interaction with an IFN-gamma receptor that is ubiquitously expressed on nearly all cells. In this review, we discuss the positive and negative effects of IFN-gamma signaling in the tumor cell on tumor growth.
Collapse
Affiliation(s)
- G L Beatty
- Department of Microbiology, University of Pennsylvannia, Philadelphia, USA
| | | |
Collapse
|
154
|
Goldring CE, Reveneau S, Chantome A, Pance A, Fleury C, Hume DA, Sester D, Mignotte B, Jeannin JF. Heat shock enhances transcriptional activation of the murine-inducible nitric oxide synthase gene. FASEB J 2000; 14:2393-5. [PMID: 11024011 DOI: 10.1096/fj.98-0509fje] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
There is considerable interest in determining the conditions leading to enhanced inducible nitric oxide synthase (iNOS) gene expression and nitric oxide (NO) biosynthesis. Using in vivo footprinting, we demonstrate that heat shock of murine macrophages concurrent with lipopolysaccharide (LPS) treatment stimulated changes in guanine methylation sensitivity at ?898/9, at a putative partial heat shock element (HSE) and at -893/4, a site bordering an E-box, within the iNOS gene enhancer, suggesting inducible occupation by transcription factors at these regions. LPS treatment accompanied by heat shock provoked increased iNOS gene transcription, increased levels of iNOS protein, and increased production of NO compared with LPS treatment alone. Electrophoretic mobility shift analysis revealed low constitutive levels of specific binding to an E-box and a partial HSE within the iNOS enhancer. Binding to the E-box was increased by LPS treatment or by heat shock, achieving a greater increase by a combination of both treatments. The proteins occupying this site were identified as belonging to the USF family of transcription factors. Heat shock or LPS increased binding to the HSE, and the factor responsible for this interaction was identified as heeat shock factor-1 (HSF-1). Mutations at the HSE revealed the importance of HSF-1 in the induction of iNOS by LPS. Thus, our data reveal two novel regulatory sites in the murine iNOS gene, one of which is implicated in enhancing iNOS expression via LPS stimulation, and provide the first evidence that heat shock enhances transcription of the iNOS gene. These results could have implications in the host response mechanism to fever-associated gram-negative infection.
Collapse
Affiliation(s)
- C E Goldring
- Department of Pharmacology, University of Liverpool, Liverpool L69 3BX, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
155
|
Ropponen KM, Kellokoski JK, Lipponen PK, Eskelinen MJ, Alanne L, Alhava EM, Kosma VM. Expression of inducible nitric oxide synthase in colorectal cancer and its association with prognosis. Scand J Gastroenterol 2000; 35:1204-11. [PMID: 11145294 DOI: 10.1080/003655200750056709] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND The expression of inducible nitric oxide synthase (iNOS) has been reported to be altered in a number of tumours, but its role in tumour biology is still unclear. METHODS iNOS was studied in a series of 157 colorectal carcinoma patients and its relation to tumour grade, stage, cell cycle regulators, cell proliferation as well as survival was assessed. RESULTS iNOS intensity was moderate or intense in 37% of the tumours. iNOS intensity and percentage of positive cells were higher in Dukes A and B tumours than in Dukes C and D tumours, and low iNOS expression intensity was related to high histological grade. iNOS expression correlated positively with cell cycle regulators p21 and AP-2. There was also a high iNOS expression intensity and high fraction of iNOS positive cells in tumours with a high amount of tumour infiltrating lymphocytes (TILs). The cancer related survival was significantly lower among patients with a low signal for iNOS and low iNOS percentage in tumour epithelium. In multivariate analysis iNOS was not an independent prognostic factor. CONCLUSIONS These results suggest that iNOS has a protective role in colorectal carcinogenesis, but further studies are required to establish the clinical significance of iNOS in colorectal cancer.
Collapse
Affiliation(s)
- K M Ropponen
- Dept. of Pathology and Forensic Medicine, University of Kuopio, Finland
| | | | | | | | | | | | | |
Collapse
|
156
|
Abstract
The steps required for new vessel growth are biologically complex and require coordinate regulation of contributing components, including modifications of cell--cell interactions, proliferation and migration of endothelial cells and matrix degradation. The observation that in vivo angiogenesis is accompanied by vasodilation, that many angiogenesis effectors possess vasodilating properties and that tumor vasculature is in a persistent state of vasodilation, support the existence of a molecular/biochemical link between vasodilation and angiogenesis. Several pieces of evidence converge in the indication of a role for nitric oxide (NO), the factor responsible for vasodilation, in physiological and pathological angiogenesis. Data originated in different labs indicate that NO can act both as an 'actor' of angiogenesis and as a 'director of angiogenesis', both functions being equally expressed during physiological and pathological processes. NO significantly contributes to the prosurvival/proangiogenic program of capillary endothelium by triggering and transducing cell growth and differentiation via endothelial-constitutive NO synthase (ec-NOS) activation, cyclic GMP (cGMP) elevation, mitogen activated kinase (MAPK) activation and fibroblast growth factor-2 (FGF-2) expression. Re-establishment of a balanced NO production in the central nervous system results in a reduction of cell damage during inflammatory and vascular diseases. Elevation of NOS activity in correlation with angiogenesis and tumor progression has been extensively reported in experimental and human tumors. In the brain, tumor expansion and edema formation are sensitive to NOS inhibition. On this basis, the nitric oxide pathway appears to be a promising target for consideration in pro- and anti-angiogenic therapeutic strategies. The use of NOS inhibitors seems appropriate to reduce edema, block angiogenesis and facilitate antitumor drug delivery.
Collapse
Affiliation(s)
- M Ziche
- Institute of Pharmacological Sciences, University of Siena, Italy.
| | | |
Collapse
|
157
|
Soler MN, Bobé P, Benihoud K, Lemaire G, Roos BA, Lausson S. Gene therapy of rat medullary thyroid cancer by naked nitric oxide synthase II DNA injection. J Gene Med 2000; 2:344-52. [PMID: 11045428 DOI: 10.1002/1521-2254(200009/10)2:5<344::aid-jgm124>3.0.co;2-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Nitric oxide (NO), produced by NO synthase II (NOS II), is the main mediator of the tumoricidal action of activated macrophages. In the present study we examined the potential of the NOS II gene as a suicide gene for medullary thyroid cancer (MTC) therapy. METHODS We orthotopically transplanted rMTC 6-23 cells into the inbred strain of Wag/Rij rats and constructed a plasmid carrying the NOS II gene under the control of the cytomegalovirus (CMV) promoter. RESULTS Successive injections of tumor cells (Day 0) and naked DNA (Day 2) caused strong inhibition of tumor growth (50%, p < 0.05). Plasmid injection into established tumors (14-day tumors) resulted in the development of large cavities due to tumor cell destruction, with a significant reduction in tumor tissue volume (35%, p < 0.05). Adjacent quiescent tissues were unaffected. Cell death occurred by apoptosis as demonstrated by specific labeling. Macrophages and CD4+ lymphocytes were recruited in the treated tumors. However, tumor-specific T lymphocytes were undetectable in the spleen of treated rats. In control experiments using Lac Z as a reporter gene, expression of beta-galactosidase was detected in only 1% of the tumor cells. CONCLUSIONS Despite a low gene transfer efficiency, NOS II plasmid produced a strong anti-tumor action resulting from its marked 'bystander' effect mainly due to NO production and diffusion. Therefore the NOS II gene appears to be a promising suicide gene therapy of human cancer.
Collapse
Affiliation(s)
- M N Soler
- Laboratoire d'Endocrinologie Cellulaire et Evolution, Université Paris-Sud, Orsay, France
| | | | | | | | | | | |
Collapse
|
158
|
Shao ZM, Radziszewski WJ, Barsky SH. Tamoxifen enhances myoepithelial cell suppression of human breast carcinoma progression in vitro by two different effector mechanisms. Cancer Lett 2000; 157:133-44. [PMID: 10936673 DOI: 10.1016/s0304-3835(00)00466-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Our previous studies have indicated that myoepithelial cells surrounding ductal and acinar epithelium of glandular organs, such as the breast, exert multiple paracrine suppressive effects on incipient and developing cancers that arise from this epithelium. Myoepithelial cells and derived cell lines (HMS 1-6) exert these effects through the secretion of a number of different effector molecules that exert anti-invasive, anti-proliferative, and anti-angiogenic activities. Since previous basic and clinical studies have examined the role of estrogen agonists and antagonists on human breast cancer cells and because issues of hormone replacement therapy (HRT) and tamoxifen chemoprevention are such timely issues in breast cancer, we wondered whether or not hormonal manipulations might affect myoepithelial cells in vitro as far as their paracrine suppressive activities on breast cancer were concerned. The present in vitro study demonstrates that treatment of myoepithelial cells with tamoxifen but not 17beta-estradiol increases both maspin secretion and invasion-blocking ability. Furthermore tamoxifen but not 17beta-estradiol increases inducible nitric oxide synthase (iNOS) expression and nitric oxide (NO) production by myoepithelial cells when they are co-cultured with conditioned media from or breast carcinoma cells directly. This increased myoepithelial NO exerts both autocrine and paracrine antiproliferative effects which can be blocked by inhibition of iNOS. 17beta-Estradiol, however, competes with all of these suppressive effects of tamoxifen suggesting that the mechanism of tamoxifen action is estrogen receptor mediated. Myoepithelial cells lack ER-alpha but express ER-beta. Tamoxifen, but not 17beta-estradiol, increases AP-1 CAT but not ERE-CAT activity. Again, 17beta-estradiol competes with the transcription-activating effects of tamoxifen. These experiments collectively suggest that the actions of tamoxifen on the increased secretion of maspin and increased production of NO by myoepithelial cells are mediated through ER-beta and the transcription-activation of an ER-dependent AP-1 response element.
Collapse
MESH Headings
- Antineoplastic Agents, Hormonal/pharmacology
- Blotting, Northern
- Blotting, Western
- Breast Neoplasms/drug therapy
- Breast Neoplasms/enzymology
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Carcinoma, Ductal, Breast/drug therapy
- Carcinoma, Ductal, Breast/metabolism
- Cell Division/drug effects
- Disease Progression
- Epithelial Cells/drug effects
- Estrogen Antagonists/pharmacology
- Female
- Gene Expression Regulation, Neoplastic
- Genes, Tumor Suppressor
- Humans
- Neoplasm Invasiveness
- Neovascularization, Pathologic/prevention & control
- Nitric Oxide/biosynthesis
- Nitric Oxide Synthase/biosynthesis
- Nitric Oxide Synthase Type II
- Precipitin Tests
- Proteins/drug effects
- Proteins/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Serine Proteinase Inhibitors/metabolism
- Serpins/drug effects
- Serpins/metabolism
- Tamoxifen/pharmacology
- Time Factors
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Z M Shao
- Department of Pathology, UCLA School of Medicine, CA 90024, Los Angeles, CA 90024, USA
| | | | | |
Collapse
|
159
|
Brennan PA, Umar T, Buckley J, Zaki GA, Langdon JD, Spedding A, Peters W. Expression of nitric oxide synthase in pleomorphic adenomas of the parotid. Br J Oral Maxillofac Surg 2000; 38:338-42. [PMID: 10922164 DOI: 10.1054/bjom.1999.0454] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The actions of nitric oxide (NO) in the pathology of solid tumours are complicated and many are poorly understood because NO has both inhibitory and tumour-promoting activities. In the current study we aimed to find out immunohistochemically whether the expression of both the inducible (iNOS) and endothelial (eNOS) forms of the enzyme nitric oxide synthase (NOS) were changed in pleomorphic adenomas of the parotid compared with normal salivary tissue. There was a significant difference in staining for iNOS between the tumour and normal salivary tissue, with tumour epithelial cells being stained in 29 cases of the 30 cases studied (P< 0.0001). The luminal cells of the salivary ducts also stained, but not the normal salivary tissue. Immunohistochemistry for the eNOS isoenzyme showed moderate staining of the tumour epithelium in only three specimens. There was also mild staining in the salivary duct cells of the normal glandular tissue and in endothelium of blood vessels in both tumour and normal glandular tissue in the same 29 cases.
Collapse
Affiliation(s)
- P A Brennan
- Oral and Maxillofacial Surgery, Queen Alexandra Hospital, Portsmouth, UK
| | | | | | | | | | | | | |
Collapse
|
160
|
Dachs GU, Tozer GM. Hypoxia modulated gene expression: angiogenesis, metastasis and therapeutic exploitation. Eur J Cancer 2000; 36:1649-60. [PMID: 10959051 DOI: 10.1016/s0959-8049(00)00159-3] [Citation(s) in RCA: 178] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Tumour hypoxia is the result of an imbalance in oxygen supply and demand. It is an adverse prognostic indicator in cancer as it modulates tumour progression and treatment. Many genes controlling tumour biology are oxygen regulated, and new ones are constantly added to the growing list of hypoxia-induced genes. Of specific importance are hypoxia-responsive transcription factors, as they can modulate the expression of numerous different genes. Similarly, growth factors which govern the formation of new blood vessels or which control blood flow are vitally important for both the maintenance of the primary tumour and metastases at distant sites. The purpose of this review is to present an update of selected issues regarding hypoxia-inducible gene expression and how this affects prognosis, angiogenesis and metastasis. It will conclude by discussing gene therapy as one possible means of exploiting tumour hypoxia for the treatment of cancer.
Collapse
Affiliation(s)
- G U Dachs
- Tumour Microcirculation Group, Gray Laboratory Cancer Research Trust, PO Box 100, Mount Vernon Hospital, HA6 2JR, Northwood, UK
| | | |
Collapse
|
161
|
Brennan PA. The actions and interactions of nitric oxide in solid tumours. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2000; 26:434-7. [PMID: 11016461 DOI: 10.1053/ejso.1999.0917] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The small molecule nitric oxide (NO) has undergone an image change since its identification as a biological messenger in 1987. It is a free radical, with a diverse range of actions in both physiological and pathological processes. Whilst over 30000 research papers have been written to date on NO, its role in tumour biology remains incompletely understood and research in this field is still in its infancy. NO would appear to have both tumour promoting and inhibiting effects which are presumed to be dependent on its local concentration within the tumour. Recently the relationships of NO to the tumour suppressor gene p53 have been experimentally elucidated, demonstrating how mutations of p53 may adversely affect the host by enhancing NO production. This review summarizes the brief history of this molecule, outlines its roles in the common solid tumours and suggests areas for future research.
Collapse
Affiliation(s)
- P A Brennan
- Maxillofacial Department, Queen Alexandra Hospital, Portsmouth, UK
| |
Collapse
|
162
|
Boscá L, Bodelón OG, Hortelano S, Casellas A, Bosch F. Anti-inflammatory action of type I interferons deduced from mice expressing interferon beta. Gene Ther 2000; 7:817-825. [PMID: 10845718 DOI: 10.1038/sj.gt.3301179] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/1999] [Accepted: 02/04/2000] [Indexed: 02/07/2023]
Abstract
Type I interferons (IFN) are widely used for the therapeutic treatment of viral infections, tumor growth and various chronic diseases such as multiple sclerosis. Antagonism between type I IFNs and IFN-gamma has been described in cells of the immune system, in particular in the activation of macrophages. To study the systemic effects of type I IFNs we used transgenic mice carrying a human IFN-beta (hIFN-beta) gene under the control of the rat insulin I promoter. These animals expressed high levels of hIFN-beta in beta-pancreatic cells, and the ability of the macrophages to respond to pro-inflammatory stimuli was analyzed. Transgenic mice exhibited an increased extravasation of cells to the peritoneal cavity after eliciting with thioglycollate broth. The expression of the inducible form of nitric oxide synthase and cyclooxygenase-2, two enzymes involved in inflammation, was impaired in transgenic animals challenged with lipopolysaccharide and IFN-gamma. Analysis of the mechanisms leading to this attenuated inflammatory response showed a decrease in the serum levels of TNF-alpha and an inhibition of the activation of the transcription factor NF-KB in various tissues. These results indicate that systemic administration of IFN-beta might influence the response to pro-inflammatory stimuli, in particular through the antagonism of IFN-gamma signaling.
Collapse
Affiliation(s)
- L Boscá
- Instituto de Bioquímica (Centro Mixto CSIC-UCM), Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| | | | | | | | | |
Collapse
|
163
|
Shichiri M, Yokokura M, Marumo F, Hirata Y. Endothelin-1 inhibits apoptosis of vascular smooth muscle cells induced by nitric oxide and serum deprivation via MAP kinase pathway. Arterioscler Thromb Vasc Biol 2000; 20:989-97. [PMID: 10764663 DOI: 10.1161/01.atv.20.4.989] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Endothelin (ET)-1, an endothelium-derived vasoconstrictor and mitogen, acts as an antiapoptotic factor against serum deprivation-induced apoptosis of endothelial cells and fibroblasts but enhances apoptosis of some cancer cells. In the present study, we examined whether nitric oxide (NO) and ET-1 modulate apoptosis of rat vascular smooth muscle cells (VSMCs) via the mitogen-activated protein (MAP) kinase pathway. Both serum deprivation and NO donors (FK409 and SNAP) caused apoptosis of VSMCs, as demonstrated by TdT-mediated dUTP-biotin nick end-labeling, appearance of fragmented DNA, and induction of caspase-3 activity. ET-1 dose-dependently antagonized apoptosis induced by serum deprivation and NO donors. A selective ET(A) receptor antagonist (BQ123) and a nonselective ET(A/B) receptor antagonist (TAK044), but not a selective ET(B) receptor antagonist (BQ788), inhibited the antiapoptotic effect of ET-1, indicating that the antiapoptotic effect of ET-1 is mediated via the ET(A) receptor. ET-1 activated MAP kinase, whose effect was inhibited by FK409. Transfection with an unphosphorylated wild-type MAP kinase kinase-1 (MAPKK-1) or its constitutively activated mutant protected VSMCs against apoptosis induced by serum deprivation and NO donors. Inhibition of MAP kinase activity with PD98059, a specific inhibitor of MAPKK-1, or by transfection of a dominant-negative MAPKK-1 mutant antagonized the antiapoptotic effect of ET-1, suggesting the involvement of MAP kinase in the antiapoptotic effect. The potent inhibitory effect of ET-1 on apoptosis of VSMCs induced by serum deprivation and NO suggests that the counterbalance between the 2 endothelium-derived factors contributes to the process of vascular remodeling by determining VSMC survival and death, respectively, via a common MAP kinase pathway.
Collapse
Affiliation(s)
- M Shichiri
- Second Department of Internal Medicine, Tokyo Medical and Dental University, Tokyo, Japan.
| | | | | | | |
Collapse
|
164
|
Chen YK, Lin LM. Immunohistochemical expression of inducible nitric oxide synthase in DMBA-induced hamster buccal pouch carcinogenesis. Oral Oncol 2000; 36:221-4. [PMID: 10745176 DOI: 10.1016/s1368-8375(99)00081-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
Nitric oxide (NO) plays a key role in the processes of inflammation and carcinogenesis. Three isoforms of NO synthase have been identified: endothelial nitric oxide synthase (NOS), neuronal NOS, and inducible NOS (iNOS). The purpose of this study was to investigate the characteristics of iNOS in 7, 12-dimethylbenz[a]anthracene (DMBA)-induced hamster buccal pouch carcinogenesis. Thirty outbred young (6-week-old) male Syrian golden hamsters were randomly divided into three groups: DMBA (0.5%) painted group (n=10); mineral oil-treated group (n=10); and non-treated group (n=10). The average number of iNOS positive foci per section in the DMBA-treated group was approximately 12.2+/-4.7. Both cytoplasmic and nuclear stainings were observed in the DMBA-treated pouch keratinocytes. No iNOS activity could be detected in the untreated or mineral oil-treated pouches. In conclusion, this study has demonstrated that iNOS is expressed in DMBA-induced hamster pouch carcinomas. This finding suggests that iNOS expression may be associated with the development of chemically induced oral carcinomas.
Collapse
Affiliation(s)
- Y K Chen
- Oral Pathology Department, School of Dentistry, Kaohsiung Medical College, 100 Shih-Chuan 1st Road, Kaohsiung, Taiwan, ROC
| | | |
Collapse
|
165
|
Ahmed B, Van Den Oord JJ. Expression of the inducible isoform of nitric oxide synthase in pigment cell lesions of the skin. Br J Dermatol 2000; 142:432-40. [PMID: 10735946 DOI: 10.1046/j.1365-2133.2000.03352.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Nitric oxide (NO) is a small molecule produced during the conversion of L-arginine to L-citrulline by NO synthase (NOS). Several isoforms of NOS exist, of which the Ca2+-independent, inducible NOS (iNOS or NOS2) is most prominently expressed by macrophages. iNOS activity and increased levels of iNOS have been found in various tumours and tumour cell lines but not in normal tissues; however, the precise role of NO in tumour progression has yet to be elucidated. We studied the expression of iNOS in paraffin sections of 41 benign naevi and 52 primary malignant melanomas (MM) of the skin, as well as in 13 metastatic MM. In addition, nitrotyrosine, indicative of NO production and formation of peroxynitrite, was studied in frozen sections of 13 naevi and 30 MM. Virtually all naevi expressed iNOS, but very few expressed nitrotyrosine, indicating either that iNOS in naevi is functionally inactive, or that naevus cells lack reactive oxygen radicals and thus do not form peroxynitrite. Normal melanocytes in adjacent uninvolved skin were unreactive for both markers. In MM, iNOS was most frequently expressed in the 'pure' and 'invasive' radial growth phase (RGP), whereas expression in the vertical growth phase (VGP) and metastatic phase occurred only in 76% of cases; moreover, in these latest phases of tumour progression, iNOS staining was weak and focal. We conclude that iNOS is expressed de novo in most benign pigment cell lesions. In MM (iNOS-generated) NO appears to play an important part in the early steps of invasion (i.e. the 'invasive' RGP), where it may stimulate neo-angiogenesis and may be a prerequisite for further tumour progression; this view is also supported by the finding of iNOS in the associated blood vessels in the papillary dermis. Finally, our data suggest that (iNOS-generated) NO plays a less significant part in the VGP and in metastatic melanoma.
Collapse
Affiliation(s)
- B Ahmed
- Department of Pathology, Laboratory of Histo- and Cytochemistry, University Hospital St Rafael, Katholieke Universiteit Leuven, B-3000 Leuven, Belgium
| | | |
Collapse
|
166
|
Sun H, Gutierrez P, Jackson MJ, Kundu N, Fulton AM. Essential role of nitric oxide and interferon-gamma for tumor immunotherapy with interleukin-10. J Immunother 2000; 23:208-14. [PMID: 10746547 DOI: 10.1097/00002371-200003000-00005] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Several laboratories have reported anti-tumor activity for high levels of interleukin-10 (IL-10) expressed as a transgene or administered as recombinant protein. The authors have reported a positive correlation for nitric oxide production and anti-tumor activity of IL-10 in a murine model of breast cancer. In the current study, they sought evidence of a mechanistic role for nitric oxide in IL-10-mediated tumor growth inhibition. They wanted to determine whether pharmacologic inhibition of nitric oxide synthase (NOS) activity reverses the therapeutic effect of IL-10. Administration of either of two NOS inhibitors, aminoguanidine (AG) or L-lysine,N6-1-iminoethyl-dihydrochloride, appears to abrogate in part the tumor growth inhibition observed when IL-10 is overexpressed as a transgene in two murine mammary tumor cell lines. Nitric oxide levels were assessed at the tumor site by measuring nitrosylated heme levels by electron spin resonance spectroscopy. Nitric oxide hemoglobin levels were lower in tumors from aminoguanidine-treated mice, indicating that effective inhibition of nitric oxide production occurred at the tumor site. Previous investigations showed that the inducible form of NOS protein (iNOS), but not constitutive NOS, was expressed at higher levels in IL-10-expressing tumors. Because iNOS is regulated at the transcriptional level, the authors compared iNOS mRNA levels in IL-10 and control tumors. Northern analysis revealed strong iNOS message expression in all six IL-10-expressing tumors examined, whereas message was faintly detected in parental or 66-neo tumors. The inducible form of NOS is responsive to induction by interferon-gamma (IFN-gamma). The role of IFN-gamma in IL-10-mediated tumor inhibition and iNOS mRNA induction was determined. When tumors were transplanted to IFN-gamma mutant mice, the tumor-inhibitory activity of IL-10 was lost. Furthermore, iNOS mRNA was no longer induced in the absence of host expression of IFN-gamma. These data indicate that nitric oxide contributes to the anti-tumor activity of IL-10 and that expression of iNOS in this context depends on IFN-gamma.
Collapse
Affiliation(s)
- H Sun
- Greenebaum Cancer Center and Department of Pathology, University of Maryland, Baltimore 21201, USA
| | | | | | | | | |
Collapse
|
167
|
Hensley K, Williamson KS, Floyd RA. Measurement of 3-nitrotyrosine and 5-nitro-gamma-tocopherol by high-performance liquid chromatography with electrochemical detection. Free Radic Biol Med 2000; 28:520-8. [PMID: 10719233 DOI: 10.1016/s0891-5849(00)00155-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Nitric oxide (NO) is a lipophilic gaseous molecule synthesized by the enzymatic oxidation of L-arginine. During periods of inflammation, phagocytic cells generate copious quantities of NO and other reactive oxygen species. The combination of NO with other reactive oxygen species promotes nitration of ambient biomolecules, including protein tyrosine residues and membrane-localized gamma-tocopherol. The oxidative chemistry of NO and derived redox congeners is reviewed. Techniques are described for the determination of 3-nitro-tyrosine and 5-nitro-gamma-tocopherol in biological samples using high-performance liquid chromatography with electrochemical detection.
Collapse
Affiliation(s)
- K Hensley
- Free Radical Biology and Aging Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA.
| | | | | |
Collapse
|
168
|
|
169
|
Bentz BG, Simmons RL, Haines GK, Radosevich JA. The yin and yang of nitric oxide: reflections on the physiology and pathophysiology of NO. Head Neck 2000; 22:71-83. [PMID: 10585608 DOI: 10.1002/(sici)1097-0347(200001)22:1<71::aid-hed11>3.0.co;2-g] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Nitric oxide (NO.) is an arginine-derived nitrogen-based radical that is rapidly becoming one of the most important molecular species to be discovered. Over the past decade, an explosion of evidence has revealed the extreme complexity of function of this seemingly simple inorganic molecule. It is now evident that NO. demonstrates a functional dualism, playing a pivotal role in numerous physiologic and pathophysiologic processes. Whether this molecule is beneficial or detrimental is dependent upon the tissue of generation, the level of production, the oxidative/reductive (redox) environment in which this radical is generated, and the presence or absence of NO. transduction elements. Nitric oxide is generated by three independent isoenzymes that resemble the p-450 enzyme superfamily in both form and function. It ultimately alters enzymatic function through covalent modification, redox interactions, and interactions with metallic functional centers. This radical is a key figure in a number of pathophysiologic processes by means of similar yet uncoordinated interactions. In consideration of the already broad spectrum of roles attributed to NO., it seems highly likely that this molecule will be implicated in an ever widening variety of functions relative to the practice of otolaryngology-head and neck surgery. This article reviews the enzymology, signal transduction mechanisms, physiology, and pathophysiology of NO. as it pertains to head and neck cancer.
Collapse
Affiliation(s)
- B G Bentz
- Department of Otolaryngology-Head & Neck Surgery, Northwestern University School of Medicine, Searle Building 12-561, 303 East Chicago Avenue, Chicago, Illinois 60611-3008, USA
| | | | | | | |
Collapse
|
170
|
Whitman S, Wang X, Shalaby R, Shtivelman E. Alternatively spliced products CC3 and TC3 have opposing effects on apoptosis. Mol Cell Biol 2000; 20:583-93. [PMID: 10611237 PMCID: PMC85138 DOI: 10.1128/mcb.20.2.583-593.2000] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/1999] [Accepted: 10/20/1999] [Indexed: 12/13/2022] Open
Abstract
The human gene CC3 is a metastasis suppressor for small cell lung carcinoma (SCLC) in vivo. The ability of CC3 to impair the apoptotic resistance of tumor cells is likely to contribute to metastasis suppression. We describe here an alternatively spliced RNA of CC3, designated TC3, that encodes an unstable protein with antiapoptotic activity. TC3 and CC3 proteins share amino-terminal sequences, but TC3 has a unique short hydrophobic carboxyl terminus. Overexpression of CC3 results in massive death of rodent fibroblasts, but TC3 protects cells from CC3-induced death and from other death stimuli such as treatment with tumor necrosis factor or overexpression of Bax protein. The death-inducing activity of CC3 resides within its amino-terminal domain, which is conserved in TC3. The carboxyl terminus of TC3 is responsible for the antiapoptotic function of TC3; mutations in this domain abolish the ability of TC3 to protect cells from apoptosis. TC3 protein is short-lived due to its rapid degradation by proteasome, and it forms complexes with a regulatory subunit of proteasome known as s5alpha. The signal for the rapid degradation of TC3 resides within its carboxyl terminus, which is capable of conferring instability on a heterologous protein. The proapoptotic activity of CC3 in SCLC cells is induced by a wide variety of signals and involves disruption of the mitochondrial membrane potential (Deltapsim). The CC3 protein has sequence similarity to bacterial short-chain dehydrogenases/reductases and might represent a phylogenetically old effector of cell death similar to the recently identified apoptosis-inducing factor. CC3 and TC3 have opposing functions in apoptosis and represent a novel dual regulator of cell death.
Collapse
Affiliation(s)
- S Whitman
- Cancer Research Institute, University of California San Francisco, San Francisco, California 94143, USA
| | | | | | | |
Collapse
|
171
|
Mitsumoto A, Kim KR, Oshima G, Kunimoto M, Okawa K, Iwamatsu A, Nakagawa Y. Glyoxalase I is a novel nitric-oxide-responsive protein. Biochem J 1999; 344 Pt 3:837-44. [PMID: 10585871 PMCID: PMC1220706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
To clarify the molecular mechanisms of nitric oxide (NO) signalling, we examined the NO-responsive proteins in cultured human endothelial cells by two-dimensional (2D) PAGE. Levels of two proteins [NO-responsive proteins (NORPs)] with different pI values responded to NO donors. One NORP (pI 5.2) appeared in response to NO, whereas another (pI 5.0) disappeared. These proteins were identified as a native form and a modified form of human glyoxalase I (Glox I; EC 4. 4.1.5) by peptide mapping, microsequencing and correlation between the activity and the isoelectric shift. Glox I lost activity in response to NO, and all NO donors tested inhibited its activity in a dose-dependent manner. Activity and normal electrophoretic mobility were restored by dithiothreitol and by the removal of sources of NO from the culture medium. Glox I was selectively inactivated by NO; compounds that induce oxidative stress (H(2)O(2), paraquat and arsenite) failed to inhibit this enzyme. Our results suggest that NO oxidatively modifies Glox I and reversibly inhibits the enzyme's activity. The inactivation of Glox I by NO was more effective than that of glyceraldehyde-3-phosphate dehydrogenase (G3PDH), another NO-sensitive enzyme. Thus Glox I seems to be a novel NO-responsive protein that is more sensitive to NO than G3PDH.
Collapse
Affiliation(s)
- A Mitsumoto
- School of Pharmaceutical Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641 Japan
| | | | | | | | | | | | | |
Collapse
|
172
|
Lesoon-Wood LA, Pierce LM, Lau AF, Cooney RV. Enhancement of methylcholanthrene-induced neoplastic transformation in murine C3H 10T1/2 fibroblasts by antisense phosphorothioate oligodeoxynucleotide sequences. Cancer Lett 1999; 147:163-73. [PMID: 10660102 DOI: 10.1016/s0304-3835(99)00292-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Antisense phosphorothioate oligodeoxynucleotides (ODNs) are increasingly used to target specific proteins for inhibition. Previous reports of antisense inhibition of the inducible nitric oxide synthase (iNOS) gene suggested its utility in defining the role of nitric oxide (NO) in carcinogenesis, as NO is mutagenic and chemical inhibitors of iNOS block neoplastic transformation in C3H 10T1/2 fibroblasts. Treatment with ODNs (0.025-25 microM) directed against 15mer sequences in the iNOS coding region decreased NO production consistent with a reduction of iNOS protein and iNOS mRNA, however, control ODNs (2.5 microM) also showed considerable nonspecific inhibition of NO synthesis. Treatment with both iNOS antisense and missense ODNs during the promotional phase of the C3H10T1/2 transformation assay significantly increased the number of neoplastic foci in 3-methylcholanthrene (MCA) treated cells which corresponded with the ability of the ODN to inhibit NO production. Enhanced neoplastic transformation and non-specific inhibition of NO synthesis resulting from exposure to antisense ODNs suggest limitations to their long-term use in humans at higher doses.
Collapse
Affiliation(s)
- L A Lesoon-Wood
- Molecular Carcinogenesis, University of Hawaii Cancer Research Center, Honolulu 96813, USA
| | | | | | | |
Collapse
|
173
|
Zhang HY, Phan SH. Inhibition of myofibroblast apoptosis by transforming growth factor beta(1). Am J Respir Cell Mol Biol 1999; 21:658-65. [PMID: 10572062 DOI: 10.1165/ajrcmb.21.6.3720] [Citation(s) in RCA: 272] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Fibroblast differentiation to the myofibroblast phenotype is associated with alpha-smooth-muscle actin (alpha-SMA) expression and regulated by cytokines. Among these, transforming growth factor (TGF)-beta(1) and interleukin (IL)-1beta can stimulate and inhibit myofibroblast differentiation, respectively. IL-1beta inhibits alpha-SMA expression by inducing apoptosis selectively in myofibroblasts via induction of nitric oxide synthase (inducible nitric oxide synthase [iNOS]). Because TGF-beta is known to inhibit iNOS expression, this study was undertaken to see if this cytokine can protect against IL-1beta-induced myofibroblast apoptosis. Rat lung fibroblasts were treated with IL-1beta and/or TGF-beta(1) and examined for expression of alpha-SMA, iNOS, and the apoptotic regulatory proteins bax and bcl-2. The results show that TGF-beta(1) caused a virtually complete suppression of IL-1beta-induced iNOS expression while preventing the decline in alpha-SMA expression or the myofibroblast subpopulation. TGF-beta(1) treatment also completely suppressed the IL-1beta-induced apoptosis in myofibroblasts. IL-1beta-induced apoptosis was associated with a significant decline in expression of the antiapoptotic protein bcl-2, which was prevented by concomitant TGF-beta(1) treatment. The level of the proapoptotic protein bax, however, was not significantly altered by either cytokine. These data suggest that TGF-beta(1) inhibits IL-1beta-induced apoptosis in myofibroblasts by at least two mechanisms, namely, the suppression of iNOS expression and the prevention of a decline in bcl-2 expression. Thus, TGF-beta(1) may be additionally important in fibrosis by virtue of this novel ability to promote myofibroblast survival by preventing the myofibroblast from undergoing apoptosis.
Collapse
Affiliation(s)
- H Y Zhang
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | |
Collapse
|
174
|
Fimiani C, Arcuri E, Santoni A, Rialas CM, Bilfinger TV, Peter D, Salzet B, Stefano GB. Mu3 opiate receptor expression in lung and lung carcinoma: ligand binding and coupling to nitric oxide release. Cancer Lett 1999; 146:45-51. [PMID: 10656608 DOI: 10.1016/s0304-3835(99)00227-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The mu3 opiate receptor subtype is expressed in human surgical specimens of both normal lung and non-small-cell lung carcinoma. Nitric oxide (NO) release is mediated through the mu3 receptor, and in lung carcinoma, morphine-stimulated NO release is significantly higher and prolonged than in normal lung. Using reverse transcriptase-polymerase chain reaction (RT-PCR) and Southern blot analysis we show that specific mu opioid receptor transcripts are present in lung carcinoma and other cells with the mu3 profile. Our findings identify a unique role for the mu3 opiate receptor in opiate-mediated NO release and suggest that endogenous opiates, through their release of NO, may play a role in cancer progression.
Collapse
Affiliation(s)
- C Fimiani
- Neuroscience Research Institute, State University of New York, College at Old Westbury, 11568, USA
| | | | | | | | | | | | | | | |
Collapse
|
175
|
Brennan PA, Downie IP, Langdon JD, Zaki GA. Emerging role of nitric oxide in cancer. Br J Oral Maxillofac Surg 1999; 37:370-3. [PMID: 10577750 DOI: 10.1054/bjom.1999.0201] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The small molecule nitric oxide has generated an exponential amount of research since its identification as a biological messenger in 1987. It is a free radical, the actions of which are diverse, and many are still poorly understood. An area of great interest is the role of nitric oxide in the growth and metastasis of solid tumours, where it seems to have a complex action including both inhibitory and tumour-promoting activity. Over 28,000 research papers have been written on nitric oxide, with only a minority concentrating on the head and neck. In this review, we give a brief history of this fascinating molecule, concentrating on its possible interest to oral and maxillofacial surgeons.
Collapse
|
176
|
Abstract
Nitric oxide has varied effects on the skin. In this review the role of nitric oxide in cutaneous wound healing, apoptosis, carcinogenesis and psoriasis is discussed.
Collapse
Affiliation(s)
- R Weller
- Department of Immunobiology, Heinrich-Heine-University, Düsseldorf, Germany
| |
Collapse
|
177
|
Tschugguel W, Schneeberger C, Unfried G, Czerwenka K, Weninger W, Mildner M, Gruber DM, Sator MO, Waldhör T, Huber JC. Expression of inducible nitric oxide synthase in human breast cancer depends on tumor grade. Breast Cancer Res Treat 1999; 56:145-51. [PMID: 10573107 DOI: 10.1023/a:1006288526311] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Expression of inducible nitric oxide synthase (iNOS) by tumor cells has been suggested to abrogate metastasis in several tumor models, whereas constitutive NOS expression correlated positively with tumor grade in human breast carcinoma. Whether or not expression of one of the various NOS isoforms could predict the prognosis of breast cancer, however, has not been established. In the present report we investigated the cellular distribution of NOS isoforms in a series of benign and malignant breast tumors and in normal breast tissue. Immunohistochemistry revealed that in samples of benign disease the number of iNOS+ epithelial cells or total epithelial cells was 69+/-16% (n = 50). In samples of grade II invasive ductal breast carcinomas the number of iNOS+ tumor cells or total tumor cells was 62+/-20% (n = 40), compared to 12+/-9% (n = 40) in samples of grade III carcinomas (P<0.0001). iNOS protein was also identifiable in most of the epithelial cells of normal breast tissue (n = 4). In contrast, eNOS protein was restricted to vascular endothelial cells in all of the specimens studied. Since the presence of tumor cell iNOS protein is inversely related to the tumor's metastatic potential, we conclude that endogenous tumor cell mediated iNOS expression might have an inhibitory effect on the metastatic process in breast cancer.
Collapse
Affiliation(s)
- W Tschugguel
- Department of Gynecology and Obstetrics, University of Vienna, School of Medicine, Austria.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
178
|
Orucevic A, Bechberger J, Green AM, Shapiro RA, Billiar TR, Lala PK. Nitric-oxide production by murine mammary adenocarcinoma cells promotes tumor-cell invasiveness. Int J Cancer 1999; 81:889-96. [PMID: 10362135 DOI: 10.1002/(sici)1097-0215(19990611)81:6<889::aid-ijc9>3.0.co;2-2] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The role of nitric oxide (NO) in tumor biology remains controversial and poorly understood. While a few reports indicate that the presence of NO in tumor cells or their micro-environment is detrimental for tumor-cell survival, and consequently their metastatic ability, a large body of data suggests that NO promotes tumor progression. The purpose of this study was to identify the source of NO in the spontaneously metastasizing C3-L5 murine mammary-adenocarcinoma model, the role of tumor-derived NO in tumor-cell invasiveness, and the mechanisms underlying the invasion-stimulating effects of tumor-derived NO. The source of NO was established by immunocytochemical localization of NO synthase (NOS) enzymes in C3-L5 cells in vitro and transplanted tumors in vivo. An in vitro transwell Matrigel invasion assay was used to test the invasiveness of C3-L5 cells in the presence or the absence of NO blocking agents or iNOS inducers (IFN-gamma and LPS). The mechanisms underlying the invasion-stimulating effects of tumor-derived NO were examined by measuring mRNA expression of matrix metalloproteinases (MMP)-2 and -9, and tissue inhibitors of metalloproteinases (TIMP) 1, 2 and 3 in C3-L5 cells in various experimental conditions. Results showed that C3-L5 cells expressed high level of eNOS protein in vitro, and in vivo, both in primary and in metastatic tumors. C3-L5 cells also expressed iNOS mRNA and protein when cultured in the presence of IFN-gamma and LPS. Constitutively produced NO promoted tumor-cell invasiveness in vitro by down-regulating TIMP 2 and TIMP 3. In addition, there was up-regulation of MMP-2, when extra NO was induced by IFN-gamma and LPS. In conclusion, NO produced by C3-L5 cells promoted tumor-cell invasiveness by altering the balance between MMP-2 and its inhibitors TIMP-2 and 3. Thus, our earlier observations of anti-tumor and anti-metastatic effects of NO inhibitors in vivo in this tumor model can be explained, at least in part, by reduced tumor-cell invasiveness.
Collapse
Affiliation(s)
- A Orucevic
- Department of Surgery, University of Pittsburgh, PA, USA
| | | | | | | | | | | |
Collapse
|
179
|
Haddad IY, Panoskaltsis-Mortari A, Ingbar DH, Yang S, Milla CE, Blazar BR. High levels of peroxynitrite are generated in the lungs of irradiated mice given cyclophosphamide and allogeneic T cells. A potential mechanism of injury after marrow transplantation. Am J Respir Cell Mol Biol 1999; 20:1125-35. [PMID: 10340931 DOI: 10.1165/ajrcmb.20.6.3460] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
In a murine bone-marrow transplant (BMT) model designed to determine risk factors for lung dysfunction in irradiated mice, we reported that cyclophosphamide (Cy)-induced injury and lethality depended on the infusion of donor spleen T cells. In the study reported here, we hypothesized that alveolar macrophage (AM)-derived reactive oxygen/nitrogen species are associated with lung dysfunction caused by allogeneic T cells, which stimulate nitric oxide (.NO) production, and by Cy, which stimulates superoxide production.NO reacts with superoxide to form peroxynitrite, a tissue-damaging oxidant. On Day 7 after allogeneic BMT, bronchoalveolar lavage fluid (BALF) obtained from mice injected with T cells contained increased levels of nitrite, which was associated with increased lactate dehydrogenase and protein levels, both of which are indices of lung injury. The injury was most severe in mice receiving both T cells and Cy. Messenger RNA (mRNA) for inducible nitric oxide synthase was detected only in murine lungs injected with T cells +/- Cy. AMs obtained on Day 7 after BMT from mice receiving T cells +/- Cy spontaneously generated between 20 and 40 microM nitrite in culture, versus < 2 microM generated by macrophages obtained from mice undergoing BMT but not receiving T cells. The level of 3-nitrotyrosine, the stable byproduct of the reaction of peroxynitrite with tyrosine residues, was increased in the BALF proteins of mice injected with both T cells and Cy. We conclude that allogeneic T cells stimulate macrophage-derived.NO, and that the addition of Cy favors peroxynitrite formation. Peroxynitrite generation clarifies the dependence of Cy-induced lung injury and lethality on the presence of allogeneic T cells.
Collapse
Affiliation(s)
- I Y Haddad
- Departments of Pediatrics and Pulmonary Medicine, University of Minnesota, Minneapolis, Minnesota 55455, USA.
| | | | | | | | | | | |
Collapse
|
180
|
Jeon YJ, Han SB, Ahn KS, Kim HM. Activation of NF-kappaB/Rel in angelan-stimulated macrophages. IMMUNOPHARMACOLOGY 1999; 43:1-9. [PMID: 10437651 DOI: 10.1016/s0162-3109(99)00032-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
In our previous studies we showed that the primary target cell of angelan, a polysaccharide purified from Angelica gigas Nakai, is a macrophage (Han et al., 1998). In the present study we examined the effect of angelan on iNOS, IL-1beta, and TNF-alpha transcription in mouse macrophage line RAW 264.7. We show that angelan produces a marked induction of iNOS, IL-1beta, and TNF-alpha transcription by RAW 264.7 cells. Since these gene transcriptions have been recently shown to be under the control of NF-kappaB/Rel family of transcription factors, we assessed the effect of angelan on NF-kappaB/Rel using a electrophoretic mobility shift assay. Treatment of RAW 264.7 cells with angelan produced strong induction of NF-kappaB/Rel binding. Treatment of RAW 264.7 cells with angelan slightly induced AP-1 binding activity, whereas Oct binding was not affected by angelan. Angelan stimulated macrophages to activate NF-kappaB/Rel, whereas neither B-cells nor T-cells were affected by the angelan. In conclusion, we demonstrate that the stimulation effect of angelan on macrophage is mediated by specific activation of NF-kappaB/Rel.
Collapse
Affiliation(s)
- Y J Jeon
- Korea Research Institute of Bioscience and Biotechnology, Yusong, Taejon, South Korea
| | | | | | | |
Collapse
|
181
|
Onier N, Hilpert S, Reveneau S, Arnould L, Saint-Giorgio V, Exbrayat JM, Jeannin JF. Expression of inducible nitric oxide synthase in tumors in relation with their regression induced by lipid A in rats. Int J Cancer 1999; 81:755-760. [PMID: 10328229 DOI: 10.1002/(sici)1097-0215(19990531)81:5<755::aid-ijc15>3.0.co;2-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2022]
Abstract
It is well documented that nitric oxide (NO) is an effector molecule of macrophage-mediated tumor cell toxicity in vitro; however, little is known about the role of NO in the antitumor immune response in vivo. We have developed a treatment protocol using lipid A. We have investigated the effects of lipid A on inducible NO synthase (NOS II) expression and evolution inside tumors during the course of treatment. Lipid A (OM-174) treatment induced tumor regression in rats bearing established colon tumors. Furthermore, NO was synthesized and secreted inside the tumors of lipid A-treated rats, as demonstrated by the increase of NOS II mRNA and NOS II content in the tumors, as well as of NOS II activity and NO production. During treatment, NOS II was localized in tumor cells only. Lipid A had no direct effect on tumor cells in vitro, while the combination of interferon gamma (IFN-gamma) plus interleukin-1 beta (IL-1beta) induced production of NO by tumor cells which was cytostatic. The content of IFN-gamma and IL-1beta in tumors was enhanced during lipid A treatment; this is in agreement with an indirect effect of lipid A in vivo via the IFN-gamma and IL-1beta pathways.
Collapse
Affiliation(s)
- N Onier
- Laboratoire d'Immunologie et d'Immunothérapie des Cancers, Ecole Pratique des Hautes Etudes, INSERM U517, Université de Bourgogne, Dijon, France
| | | | | | | | | | | | | |
Collapse
|
182
|
Jeyarajah DR, Kielar M, Penfield J, Lu CY. Docosahexaenoic acid, a component of fish oil, inhibits nitric oxide production in vitro. J Surg Res 1999; 83:147-50. [PMID: 10329109 DOI: 10.1006/jsre.1999.5586] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Docosahexaenoic acid (DHA) has been shown to be immunosuppressive in the fetus, and fish oil diets are thought to be beneficial in autoimmune disease and transplantation. This effect may be mediated through nitric oxide (NO). Here, we investigate the effect of DHA on murine macrophages. METHODS Peritoneal macrophages were subjected to stimulation with various concentrations of interferon gamma-(IFN-gamma) and tumor necrosis factor alpha (TNF-alpha). NO production was assessed by measuring nitrite (Greiss reaction). RESULTS At all doses of IFN-gamma and TNF-alpha, DHA was found to be inhibitory to NO production. CONCLUSIONS DHA inhibits macrophage-stimulated NO production in response to IFN-gamma and TNF-alpha. As NO is thought to be important in several disease processes, DHA may be a useful agent in the treatment of conditions such as autoimmune disease.
Collapse
Affiliation(s)
- D R Jeyarajah
- Department of Surgery, Department of Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75235-9156, USA
| | | | | | | |
Collapse
|
183
|
Fournier MV, Carvalho MG, Pardee AB. A strategy to identify genes associated with circulating solid tumor cell survival in peripheral blood. Mol Med 1999; 5:313-9. [PMID: 10390547 PMCID: PMC2230427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2023] Open
Abstract
Efforts in metastasis research have centered on the phenotypic and genetic differences between primary site and metastatic site tumors. However, genes that may be used as molecular markers of metastasis in circulating tumor cells remain unidentified. Genes regulating the dissemination and survival of solid tumor cells in the blood, as well as their adaptation to new environments, could be candidates for unique metastatic tumor markers. Differential display (DD) was conducted to compare the blood of tumor-free individuals with the blood of patients with lung, breast, and colon cancers. Twenty-one up-expressed genes in the tumor patient blood samples but none in the tumor-free donor blood samples were identified. Nine of these samples were isolated, amplified, and directly sequenced. A gene AB-1 homologous to a Bcl-2 family member, which might function as an apoptosis inhibitor, was identified. The overexpression of an apoptosis inhibitor in blood from patients with metastatic tumors might be correlated with the capability of solid tumor cells to survive in peripheral blood. This is the first demonstration of the usefulness of comparing control and patient blood samples by DD to find novel potential genetic markers identifying metastasis in the blood. http://link.springer-ny. com/link/service/journals/00020/bibs/5n5p313.html
Collapse
Affiliation(s)
- M V Fournier
- Cancer Biology Division, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA.
| | | | | |
Collapse
|
184
|
A Strategy to Identify Genes Associated with Circulating Solid Tumor Cell Survival in Peripheral Blood. Mol Med 1999. [DOI: 10.1007/bf03402067] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
|
185
|
Takenaga M, Igarashi R, Ochiai A, Mizushima Y. Effect of lecithinized superoxide dismutase (PC-SOD) on experimental pulmonary metastasis in mice. Free Radic Biol Med 1999; 26:1117-25. [PMID: 10381181 DOI: 10.1016/s0891-5849(98)00301-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The inhibitory effect of lecithinized superoxide dismutase (PC-SOD) on pulmonary metastasis in mice was investigated. In an experimental pulmonary metastasis model employing Meth A-T cells, significant and dose-dependent inhibition was observed after i.v. pre-administration of PC-SOD. Unmodified SOD (U-SOD) was also effective, but a 10-times higher dose was necessary to be significant. The pulmonary accumulation of Meth A-T cells labeled with 5-[125I]iodo-2'-deoxyuridine was not reduced by either PC-SOD or U-SOD, and neither of the compounds decreased pulmonary MPO activity. However, PC-SOD increased pulmonary SOD activity for longer, compared with U-SOD. In vitro addition of PC-SOD dose-dependently suppressed the growth of Meth A-T cells, while U-SOD had little effect. The combination of PC-SOD and S-nitroso-N-acetyl-D,L-penicillamine (SNAP), a nitric oxide (NO)-generating agent, had an additive effect. It was also found that PC-SOD prevented a decrease of pulmonary NOx level following tumor cell inoculation. It was concluded that PC-SOD possessed antimetastatic activity, and its potency was superior to that of U-SOD. These results suggest that PC-SOD may prevent the excessive formation of oxygen radicals and peroxynitrite (ONOO-) which cause cell damage and facilitate tumor metastasis.
Collapse
Affiliation(s)
- M Takenaga
- The Second Department of the Institute of Medical Science, St. Marianna University School of Medicine, Kawasaki, Japan
| | | | | | | |
Collapse
|
186
|
Gerecitano J, Perle MA, Vilcek J. Transcriptional basis for the differences in inducible nitric oxide synthase (iNOS) expression between nonmetastatic and metastatic murine melanoma cell lines. J Interferon Cytokine Res 1999; 19:393-405. [PMID: 10334391 DOI: 10.1089/107999099314108] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
An inverse correlation exists between expression of the inducible nitric oxide synthase (iNOS) gene and the ability of cloned K1735 murine melanoma cell lines to metastasize. We have analyzed the basis for the difference in iNOS induction by interferon-gamma (IFN-gamma) and lipopolysaccharide (LPS) in metastatic and non-metastatic K1735 cells. Nuclear run-on (NRO) assays revealed an upregulation of iNOS transcription on treatment with IFN-gamma plus LPS in nonmetastatic cells but not in a metastatic line. Transcription factors IFN regulatory factor 1 (IRF-1) and NF-kappaB were induced and functional in both metastatic and nonmetastatic K1735 lines treated with IFN-gamma plus LPS. Furthermore, a reporter construct driven by the wild-type iNOS promoter was transcriptionally activated in both nonmetastatic and metastatic cells. The iNOS-inducible phenotype was dominant in somatic cell hybrids generated by the fusion of nonmetastatic and metastatic cells, suggesting that no inhibitors of iNOS expression are present in metastatic cells. We conclude that the selective block in iNOS transcription in metastatic K1735 cells is likely due to an alteration in iNOS gene regulatory sequences. However, no such alteration was detected within the 1.7 kb iNOS promoter region in metastatic cells.
Collapse
Affiliation(s)
- J Gerecitano
- Department of Microbiology, New York University School of Medicine, New York 10016, USA
| | | | | |
Collapse
|
187
|
Tschugguel W, Pustelnik T, Lass H, Mildner M, Weninger W, Schneeberger C, Jansen B, Tschachler E, Waldhör T, Huber JC, Pehamberger H. Inducible nitric oxide synthase (iNOS) expression may predict distant metastasis in human melanoma. Br J Cancer 1999; 79:1609-12. [PMID: 10188914 PMCID: PMC2362722 DOI: 10.1038/sj.bjc.6690256] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Expression of inducible nitric oxide synthase (iNOS) and its cellular localization was investigated in subcutaneous or lymph node metastases of human melanoma. Immunohistochemistry revealed that iNOS expression was limited to melanoma cells. In samples of patients without distant metastases, the number of iNOS+ tumour cells/total tumour cells was 55% +/- 17% (n = 12) compared with 9% +/- 8% when distant metastases of lung, liver or brain occurred within an observation period of 3 years (n = 10) (P < 0.001). Western blotting confirmed the expression of iNOS protein in select cases. Notably, iNOS is expressed in regional melanoma metastases and its expression is inversely related to the tumour's metastatic potential. Thus, iNOS expression may have predictive value for the development of distant metastases of human melanoma.
Collapse
Affiliation(s)
- W Tschugguel
- Department of Gynaecology and Obstetrics, University of Vienna, School of Medicine, Austria
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
188
|
Hung K, Hayashi R, Lafond-Walker A, Lowenstein C, Pardoll D, Levitsky H. The central role of CD4(+) T cells in the antitumor immune response. J Exp Med 1998; 188:2357-68. [PMID: 9858522 PMCID: PMC2212434 DOI: 10.1084/jem.188.12.2357] [Citation(s) in RCA: 980] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The induction of optimal systemic antitumor immunity involves the priming of both CD4(+) and CD8(+) T cells specific for tumor-associated antigens. The role of CD4(+) T helper cells (Th) in this response has been largely attributed to providing regulatory signals required for the priming of major histocompatibility complex class I restricted CD8(+) cytolytic T lymphocytes, which are thought to serve as the dominant effector cell mediating tumor killing. However, analysis of the effector phase of tumor rejection induced by vaccination with irradiated tumor cells transduced to secrete granulocyte/macrophage colony-stimulating factor indicates a far broader role for CD4(+) T cells in orchestrating the host response to tumor. This form of immunization leads to the simultaneous induction of Th1 and Th2 responses, both of which are required for maximal systemic antitumor immunity. Cytokines produced by these CD4(+) T cells activate eosinophils as well as macrophages that produce both superoxide and nitric oxide. Both of these cell types then collaborate within the site of tumor challenge to cause its destruction.
Collapse
Affiliation(s)
- K Hung
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | | | | | | | | |
Collapse
|
189
|
Ambs S, Merriam WG, Ogunfusika MO, Bennett WP, Ishibe N, Hussain SP, Tzeng EE, Geller DA, Billiar TR, Harris CC. p53 and vascular endothelial growth factor regulate tumor growth of NOS2-expressing human carcinoma cells. Nat Med 1998; 4:1371-6. [PMID: 9846573 DOI: 10.1038/3957] [Citation(s) in RCA: 207] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The finding of frequent nitric oxide synthase expression in human cancers indicates that nitric oxide has a pathophysiological role in carcinogenesis. To determine the role of nitric oxide in tumor progression, we generated human carcinoma cell lines that produced nitric oxide constitutively. Cancer cells expressing inducible nitric oxide synthase that had wild-type p53 had reduced tumor growth in athymic nude mice, whereas those with mutated p53 had accelerated tumor growth associated with increased vascular endothelial growth factor expression and neovascularization. Our data indicate that tumor-associated nitric oxide production may promote cancer progression by providing a selective growth advantage to tumor cells with mutant p53, and that inhibitors of inducible nitric oxide synthase may have therapeutic activity in these tumors.
Collapse
Affiliation(s)
- S Ambs
- Laboratory of Human Carcinogenesis, National Cancer Institute, NIH, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
190
|
Wu-Hsieh BA, Chen W, Lee HJ. Nitric oxide synthase expression in macrophages of Histoplasma capsulatum-infected mice is associated with splenocyte apoptosis and unresponsiveness. Infect Immun 1998; 66:5520-6. [PMID: 9784566 PMCID: PMC108692 DOI: 10.1128/iai.66.11.5520-5526.1998] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/1998] [Accepted: 08/11/1998] [Indexed: 11/20/2022] Open
Abstract
Splenic macrophages from Histoplasma capsulatum-infected mice express inducible nitric oxide synthase (iNOS), and the iNOS expression correlates with severity of the infection. We examined whether production of NO is responsible for apoptosis and the anti-lymphoproliferative response of splenocytes from mice infected with H. capsulatum. In situ terminal deoxynucleotidyl transferase nick end labeling revealed apoptotic nuclei in cryosections of spleen from infected but not normal mice. Splenocytes of infected mice were unresponsive to stimulation by either concanavalin A or heat-killed H. capsulatum yeast cells. Splenocyte responsiveness was restored by addition to the medium of NG-monomethyl-L-arginine, a known inhibitor of NO production. The proliferative response of splenocytes from infected mice was also restored by depletion of macrophages or by replacement with macrophages from normal mice. In addition, expression of iNOS returned to its basal level when the animals had recovered from infection. These results suggest that suppressor cell activity of macrophages is associated with production of NO, which also appears to be an effector molecule for apoptosis of cultured splenocytes from infected mice.
Collapse
Affiliation(s)
- B A Wu-Hsieh
- Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei, Taiwan, Republic of China.
| | | | | |
Collapse
|
191
|
Wink DA, Mitchell JB. Chemical biology of nitric oxide: Insights into regulatory, cytotoxic, and cytoprotective mechanisms of nitric oxide. Free Radic Biol Med 1998; 25:434-56. [PMID: 9741580 DOI: 10.1016/s0891-5849(98)00092-6] [Citation(s) in RCA: 1039] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
There has been confusion as to what role(s) nitric oxide (NO) has in different physiological and pathophysiological mechanisms. Some studies imply that NO has cytotoxic properties and is the genesis of numerous diseases and degenerative states, whereas other reports suggest that NO prevents injurious conditions from developing and promotes events which return tissue to homeostasis. The primary determinant(s) of how NO affects biological systems centers on its chemistry. The chemistry of NO in biological systems is extensive and complex. To simplify this discussion, we have formulated the "chemical biology of NO" to describe the pertinent chemical reactions under specific biological conditions. The chemical biology of NO is divided into two major categories, direct and indirect. Direct effects are defined as those reactions fast enough to occur between NO and specific biological molecules. Indirect effects do not involve NO, but rather are mediated by reactive nitrogen oxide species (RNOS) formed from the reaction of NO either with oxygen or superoxide. RNOS formed from NO can mediate either nitrosative or oxidative stress. This report discusses various aspects of the chemical biology of NO relating to biological molecules such as guanylate cyclase, cytochrome P450, nitric oxide synthase, catalase, and DNA and explores the potential roles of NO in different biological events. Also, the implications of different chemical reactions of NO with cellular processes such as mitochondrial respiration, metal homeostasis, and lipid metabolism are discussed. Finally, a discussion of the chemical biology of NO in different cytotoxic mechanisms is presented.
Collapse
Affiliation(s)
- D A Wink
- Radiation Biology Branch, National Cancer Institute, Bethesda, MD 20892, USA
| | | |
Collapse
|
192
|
Reszka KJ, Matuszak Z, Chignell CF. Lactoperoxidase-catalyzed oxidation of melanin by reactive nitrogen species derived from nitrite (NO2-): an EPR study. Free Radic Biol Med 1998; 25:208-16. [PMID: 9667498 DOI: 10.1016/s0891-5849(98)00058-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The reaction of synthetic DOPA melanin (DM) with lactoperoxidase (LPO), hydrogen peroxide, and nitrite (NO2-) has been investigated using EPR. We observed that in the presence of nitrite LPO/H2O2 generated large amount of melanin radicals, as evidenced by a strong, up to 11-fold, increase in the intensity of the melanin EPR signal. In contrast, when nitrite was omitted the increase was much less, ca. 30%, which, nevertheless, indicates that DM can be metabolized directly by LPO/H2O2. When the nitrite was present, the concentration of melanin radicals was linearly dependent on [NO2-] (for [NO2-] <5 mM), and increased when [LPO] and [H2O2] increased (at constant [NO2-]). We propose that the mechanism for the generation of melanin radicals by the LPO/H2O2/nitrite system involves oxidation of NO2- by LPO/H2O2 to a reactive metabolite, most likely the nitrogen dioxide radical (.NO2), which subsequently reacts with melanin 5,6-dihydroxyindole subunits producing the respective semiquinone radicals. Because melanin and .NO2 generating systems (nitrite, peroxidase enzymes, hydrogen peroxide) may coexist in cells in vivo, our results suggest that melanin could function as a natural scavenger of this highly reactive nitrogen species. This property may be relevant to the physiological functions of the melanin pigments in vivo.
Collapse
Affiliation(s)
- K J Reszka
- Laboratory of Pharmacology & Chemistry, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, USA.
| | | | | |
Collapse
|
193
|
Zidek Z, Masek K. Erratic behavior of nitric oxide within the immune system: illustrative review of conflicting data and their immunopharmacological aspects. INTERNATIONAL JOURNAL OF IMMUNOPHARMACOLOGY 1998; 20:319-43. [PMID: 9756129 DOI: 10.1016/s0192-0561(98)00036-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The literature data assembled in this article document the variation of immunobiological effects of nitric oxide (NO). A number of factors are obviously responsible for the diversity, ranging from inactivity, alleviation, but not rarely to exacerbation of certain pathogenetic processes. A better understanding of NO interactions with the immune system can only be reached if more complex experimental designs to study the effects of reactive nitrogen species are adopted in the future. They should integrate major participating variables and take into account pharmacodynamic/kinetic aspects of NO production in triggering the ultimate effects. If manipulation of NO in the organism by means of recently developed NO inhibitors and NO donors is to become a rational tool of immunopharmacological strategies, detailed knowledge of their pharmacologies and toxicologies is urgently needed in order to differentiate between the effects of NO and other side effects. Hopefully, this approach could improve the predictability of the clinical outcomes of NO manipulation.
Collapse
Affiliation(s)
- Z Zidek
- Institute of Pharmacology, Academy of Sciences of the Czech Republic, Prague.
| | | |
Collapse
|
194
|
Caspase Activation Is Required for Nitric Oxide–Mediated, CD95(APO-1/Fas)–Dependent and Independent Apoptosis in Human Neoplastic Lymphoid Cells. Blood 1998. [DOI: 10.1182/blood.v91.11.4311.411k07_4311_4320] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Nitric oxide (NO), an important effector molecule involved in immune regulation and host defense, was shown to induce apoptosis in lymphoma cells. In the present report the NO donor glycerol trinitrate was found to induce apoptosis in Jurkat cells that are sensitive to CD95-mediated kill. In contrast, a CD95-resistant Jurkat subclone showed substantial protection from apoptosis after exposure to NO. NO induced mRNA expression of CD95 (APO-1/Fas) and TRAIL/APO-2 ligands. Moreover, NO triggered apoptosis in freshly isolated human leukemic lymphocytes which were also sensitive to anti-CD95 treatment. The ability of NO to induce apoptosis was completely blocked by a broad-spectrum ICE (interleukin-1β converting enzyme)-protease/caspase inhibitor and correlated with FLICE/caspase-8 activation. This activation was abrogated in some neoplastic lymphoid cells but not in others by the inhibitor of protein synthesis cycloheximide. Our results were confirmed using an in vitro experimental model of coculture of human lymphoid target cells with activated bovine endothelial cells generating NO as effectors. Furthermore, the inhibition of endogenous NO production with the inducible NO synthase inhibitor NG-monomethyl-L-arginine caused a complete abrogation of the apoptotic effect. Our data provide evidence that NO-induced apoptosis in human neoplastic lymphoid cells strictly requires activation of caspases, in particular FLICE, the most CD95 receptor-proximal caspase. Depending on the cell line tested this activation required or was independent of the CD95 receptor/ligand system.
Collapse
|
195
|
Caspase Activation Is Required for Nitric Oxide–Mediated, CD95(APO-1/Fas)–Dependent and Independent Apoptosis in Human Neoplastic Lymphoid Cells. Blood 1998. [DOI: 10.1182/blood.v91.11.4311] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Nitric oxide (NO), an important effector molecule involved in immune regulation and host defense, was shown to induce apoptosis in lymphoma cells. In the present report the NO donor glycerol trinitrate was found to induce apoptosis in Jurkat cells that are sensitive to CD95-mediated kill. In contrast, a CD95-resistant Jurkat subclone showed substantial protection from apoptosis after exposure to NO. NO induced mRNA expression of CD95 (APO-1/Fas) and TRAIL/APO-2 ligands. Moreover, NO triggered apoptosis in freshly isolated human leukemic lymphocytes which were also sensitive to anti-CD95 treatment. The ability of NO to induce apoptosis was completely blocked by a broad-spectrum ICE (interleukin-1β converting enzyme)-protease/caspase inhibitor and correlated with FLICE/caspase-8 activation. This activation was abrogated in some neoplastic lymphoid cells but not in others by the inhibitor of protein synthesis cycloheximide. Our results were confirmed using an in vitro experimental model of coculture of human lymphoid target cells with activated bovine endothelial cells generating NO as effectors. Furthermore, the inhibition of endogenous NO production with the inducible NO synthase inhibitor NG-monomethyl-L-arginine caused a complete abrogation of the apoptotic effect. Our data provide evidence that NO-induced apoptosis in human neoplastic lymphoid cells strictly requires activation of caspases, in particular FLICE, the most CD95 receptor-proximal caspase. Depending on the cell line tested this activation required or was independent of the CD95 receptor/ligand system.
Collapse
|
196
|
Kundu N, Dorsey R, Jackson MJ, Guiterrez P, Wilson K, Fu S, Ramanujam K, Thomas E, Fulton AM. Interleukin-10 gene transfer inhibits murine mammary tumors and elevates nitric oxide. Int J Cancer 1998; 76:713-9. [PMID: 9610731 DOI: 10.1002/(sici)1097-0215(19980529)76:5<713::aid-ijc17>3.0.co;2-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Transfection of cDNA for IL-10 into line 66.1 murine mammary tumor cells results in marked suppression of tumor growth and metastasis. Others have reported that nitric oxide has potent antitumor activity and IL-10 is known to regulate the inducible isoform of nitric oxide synthase (iNOS) expressed in macrophages. We identified nitric oxide production in mammary tumors as indicated by electron paramagnetic resonance detection of nitric oxide-hemoglobin (NO-Hb). IL-10 expression resulted in elevated levels of NO-Hb in mammary tumors. Immunohistochemical examination of mammary tumors for iNOS protein revealed few positively staining cells in parental or control neo-transfected tumors but strong iNOS staining in all IL-10 transfected tumors, consistent with the NO-Hb data. To determine if mammary epithelial tumor cells themselves, express nitric oxide synthase activity, cultured tumor cells were treated with pro-inflammatory cytokines and nitrite accumulation was assessed in the conditioned medium. All IL-10 producing cell lines accumulated uM concentrations of nitrite in response to short term (24 hr) cytokine stimulation. Cells not expressing IL-10 (parental and neo-transfectants) accumulated no nitrite under similar culture conditions. After longer stimulation (48 hr), parental and 66-neo cells accumulated lower amounts of nitrite. IL-10 gene transfer is associated with increased iNOS protein expression and enzymatic activity detected both in vitro and in vivo. Our findings suggest that the antimetastatic and antitumor activity of IL-10 is related to enhanced production of nitric oxide.
Collapse
Affiliation(s)
- N Kundu
- Greenebaum Cancer Center, University of Maryland, Baltimore, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
197
|
Thornton FJ, Schäffer MR, Witte MB, Moldawer LL, MacKay SL, Abouhamze A, Tannahill CL, Barbul A. Enhanced collagen accumulation following direct transfection of the inducible nitric oxide synthase gene in cutaneous wounds. Biochem Biophys Res Commun 1998; 246:654-9. [PMID: 9618268 DOI: 10.1006/bbrc.1998.8681] [Citation(s) in RCA: 112] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Inducible nitric oxide synthase (iNOS) is expressed during cutaneous wound repair. Mounting evidence suggests that wound nitric oxide (NO) augments collagen accumulation. We hypothesized that in vivo transfection of wound cells with the iNOS gene would increase physiological wound NO levels and thus augment collagen accumulation. Polyvinyl alcohol sponges were instilled with a mammalian expression plasmid (pMP6) containing either the chloramphenicol acetyl transferase (CAT) reporter or murine iNOS gene driven by a CMV immediate-early promoter. Plasmid DNA was injected alone or in complex with cationic liposomes, and the sponges were placed subcutaneously in male Sprague-Dawley rats which had received a longitudinal dorsal midline incision. Animals were sacrificed at different time points post-wounding and the sponges assayed for CAT activity, transfected iNOS mRNA, total nitrate and nitrite concentration (NOx) (as an index of wound NO synthesis), and hydroxyproline content (as an index of sponge collagen accumulation). The results demonstrate that wound cells were more efficiently transfected by naked DNA than by liposome mediated transfection and that maximal expression of both iNOS and CAT occurred at 48 hrs with a rapid decline after this time point. After 7 days, iNOS transfected sponges had accumulated significantly more collagen than those transfected with CAT. We conclude that cutaneous wounds can be successfully transfected by direct injection of naked DNA and that increased iNOS expression precedes an increase in collagen synthesis.
Collapse
Affiliation(s)
- F J Thornton
- Department of Surgery, Sinai Hospital of Baltimore, Maryland 21215, USA
| | | | | | | | | | | | | | | |
Collapse
|
198
|
Chen L, Xie QW, Nathan C. Alkyl hydroperoxide reductase subunit C (AhpC) protects bacterial and human cells against reactive nitrogen intermediates. Mol Cell 1998; 1:795-805. [PMID: 9660963 DOI: 10.1016/s1097-2765(00)80079-9] [Citation(s) in RCA: 169] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
In Salmonella typhimurium, ahpC encodes subunit C of alkyl hydroperoxide reductase, an enzyme that reduces organic peroxides. Here, we asked if ahpC could protect cells from reactive nitrogen intermediates (RNI). Salmonella disrupted in ahpC became hypersusceptible to RNI. ahpC from either Mycobacterium tuberculosis or S. typhimurium fully complemented the defect. Unlike protection against cumene hydroperoxide, protection afforded by ahpC against RNI was independent of the reducing flavoprotein, AhpF. Mycobacterial ahpC protected human cells from necrosis and apoptosis caused by RNI delivered exogenously or produced endogenously by transfected nitric oxide synthase. Resistance to RNI appears to be a physiologic function of ahpC. ahpC is the most widely distributed gene known that protects cells directly from RNI, and provides an enzymatic defense against an element of antitubercular immunity.
Collapse
Affiliation(s)
- L Chen
- Seaver Laboratory, Department of Medicine, Cornell University Medical College, New York, New York 10021, USA
| | | | | |
Collapse
|
199
|
Juang SH, Xie K, Xu L, Shi Q, Wang Y, Yoneda J, Fidler IJ. Suppression of tumorigenicity and metastasis of human renal carcinoma cells by infection with retroviral vectors harboring the murine inducible nitric oxide synthase gene. Hum Gene Ther 1998; 9:845-54. [PMID: 9581907 DOI: 10.1089/hum.1998.9.6-845] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The purpose of this study was to determine whether retrovirus-mediated transfer of the murine macrophage inducible nitric oxide synthase (iNOS) gene can inhibit tumorigenicity and metastasis of human renal cancer cells. Retroviral vectors encoding murine macrophage iNOS were constructed in the pLXSN retroviral vector with the iNOS gene under the control of a long terminal repeat promoter and a neomycin resistance gene under the control of an internal simian virus 40 promoter. Highly metastatic human renal carcinoma SN12PM6 cells were infected with control or iNOS retrovirus. Expression of iNOS was confirmed by Northern and Western blot analyses, and expression of the functional iNOS protein, i.e., production of nitric oxide (NO), was determined by measuring nitrite accumulation in culture supernatants. Noninfected or control cells produced large orthotopic tumors in the kidney of nude mice and a larger number of experimental lung metastases, whereas iNOS-infected cells produced small tumors in the kidneys and few to no lung metastases. The data indicate that the infection of human renal cancer cells by retroviruses harboring the murine iNOS gene can induce the production of high levels of NO, which is associated with autocytotoxicity, suppression of tumorigenicity, and abrogation of metastasis.
Collapse
Affiliation(s)
- S H Juang
- Department of Cell Biology, The University of Texas M.D. Anderson Cancer Center, Houston 77030, USA
| | | | | | | | | | | | | |
Collapse
|
200
|
Tozer GM, Everett SA. Nitric oxide in tumor biology and cancer therapy. Part 2: Therapeutic implications. Clin Oncol (R Coll Radiol) 1998; 9:357-64. [PMID: 9448964 DOI: 10.1016/s0936-6555(97)80128-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- G M Tozer
- Gray Laboratory Cancer Research Trust, Mount Vernon Hospital, Northwood, UK
| | | |
Collapse
|