151
|
Liu X, Ye L, Bai Y, Mojidi H, Simister NE, Zhu X. Activation of the JAK/STAT-1 signaling pathway by IFN-gamma can down-regulate functional expression of the MHC class I-related neonatal Fc receptor for IgG. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 181:449-63. [PMID: 18566411 PMCID: PMC2667120 DOI: 10.4049/jimmunol.181.1.449] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Expression of many MHC genes is enhanced at the transcriptional or posttranscriptional level following exposure to the cytokine IFN-gamma. However, in this study we found that IFN-gamma down-regulated the constitutive expression of the neonatal Fc receptor (FcRn), an MHC class I-related molecule that functions to transport maternal IgG and protect IgG and albumin from degradation. Epithelial cell, macrophage-like THP-1 cell, and freshly isolated human PBMC exposure to IFN-gamma resulted in a significant decrease of FcRn expression as assessed by real-time RT-PCR and Western blotting. The down-regulation of FcRn was not caused by apoptosis or the instability of FcRn mRNA. Chromatin immunoprecipitation and gel mobility shift assays showed that STAT-1 bound to an IFN-gamma activation site in the human FcRn promoter region. Luciferase expression from an FcRn promoter-luciferase reporter gene construct was not altered in JAK1- and STAT-1-deficient cells following exposure to IFN-gamma, whereas expression of JAK1 or STAT-1 protein restored the IFN-gamma inhibitory effect on luciferase activity. The repressive effect of IFN-gamma on the FcRn promoter was selectively reversed or blocked by mutations of the core nucleotides in the IFN-gamma activation site sequence and by overexpression of the STAT-1 inhibitor PIAS1 or the dominant negative phospho-STAT-1 mutations at Tyr-701 and/or Ser-727 residues. Furthermore, STAT-1 might down-regulate FcRn transcription through sequestering the transcriptional coactivator CREB binding protein/p300. Functionally, IFN-gamma stimulation dampened bidirectional transport of IgG across a polarized Calu-3 lung epithelial monolayer. Taken together, our results indicate that the JAK/STAT-1 signaling pathway was necessary and sufficient to mediate the down-regulation of FcRn gene expression by IFN-gamma.
Collapse
Affiliation(s)
- Xindong Liu
- Laboratory of Immunology, Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, MD 20742
| | - Lilin Ye
- Laboratory of Immunology, Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, MD 20742
| | - Yu Bai
- Laboratory of Immunology, Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, MD 20742
| | - Habi Mojidi
- Laboratory of Immunology, Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, MD 20742
- Maryland Pathogen Research Institute, Graduate Program in Molecular and Cell Biology, University of Maryland, College Park, MD 20742
| | - Neil E. Simister
- Rosenstiel Center for Basic Biomedical Sciences and Biology Department, Brandeis University, Waltham, MA 02254
| | - Xiaoping Zhu
- Laboratory of Immunology, Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, MD 20742
- Maryland Pathogen Research Institute, Graduate Program in Molecular and Cell Biology, University of Maryland, College Park, MD 20742
| |
Collapse
|
152
|
Amplification of the signal transducer and activator of transcription I signaling pathway and its association with apoptosis in monocytes from HIV-infected patients. AIDS 2008; 22:1137-44. [PMID: 18525259 DOI: 10.1097/qad.0b013e3283013d42] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Monocytes/macrophages play a major role in inflammation and pathogen clearance. However, chronic immune activation observed during HIV infection may also cause cellular dysfunction and tissue pathology. Indeed, several defects have been reported in these cells during HIV infections. As cytokine responsiveness via the signal transducer and activator of transcription (STAT1) signaling pathway is critical for these functions, we hypothesized that its activation in monocytes from HIV-positive patients may be disrupted. OBJECTIVES To evaluate cytokine-dependent STAT signaling in monocytes from HIV-positive patients and study the biological impact and molecular mechanisms responsible for the alterations in the interferon (IFN)-gamma-induced STAT1 pathway observed. METHODS Monocytes from chronically infected HIV-positive patients on and off antiretroviral therapy were assayed respectively for STAT activation, apoptosis, and other downstream effects by flow cytometry, real-time PCR and enzyme-linked immunosorbent assay. RESULTS Unlike IFN-alpha, interleukin-10, granulocyte macrophage colony-stimulating factor, and interleukin-4, only IFN-gamma-induced STAT1 activation was upregulated in monocytes from off-therapy patients compared with those on antiretroviral therapy and HIV-negative controls, correlating with increased total STAT1 expression. Among the IFN-gamma responsive genes (IRF-1, CXCL9, CXCL10) studied, differential effects were observed, likely reflecting the more complex regulatory control over their expression. Interestingly, spontaneous monocyte apoptosis was elevated in HIV-positive patients off-therapy compared with HIV-negative controls and correlated with STAT1 expression. IFN-gamma-induced apoptosis was also increased and persisted despite seemingly effective antiretroviral therapy. CONCLUSION Amplification of STAT1 signaling and apoptosis may reflect the chronic nature of immune activation in HIV-positive patients and contribute to the functional impairment observed in monocytes through the course of the disease.
Collapse
|
153
|
Kang TJ, Basu S, Zhang L, Thomas KE, Vogel SN, Baillie L, Cross AS. Bacillus anthracis spores and lethal toxin induce IL-1beta via functionally distinct signaling pathways. Eur J Immunol 2008; 38:1574-84. [PMID: 18493980 PMCID: PMC3681412 DOI: 10.1002/eji.200838141] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Previous reports suggested that lethal toxin (LT)-induced caspase-1 activity and/or IL-1beta accounted for Bacillus anthracis (BA) infection lethality. In contrast, we now report that caspase-1-mediated IL-1beta expression in response to BA spores is required for anti-BA host defenses. Caspase-1(-/-) and IL-1beta(-/-) mice are more susceptible than wild-type (WT) mice to lethal BA infection, are less able to kill BA both in vivo and in vitro, and addition of rIL-1beta to macrophages from these mice restored killing in vitro. Non-germinating BA spores induced caspase-1 activity, IL-1beta and nitric oxide, by which BA are killed in WT but not in caspase-1(-/-) mice, suggesting that the spore itself stimulated inflammatory responses. While spores induced IL-1beta in LT-susceptible and -resistant macrophages, LT induced IL-1beta only in LT-susceptible macrophages. Cooperation between MyD88-dependent and -independent signaling pathways was required for spore-induced, but not LT-induced, IL-1beta. While both spores and LT induced caspase-1 activity and IL-1beta, LT did not induce IL-1beta mRNA, and spores did not induce cell death. Thus different components of the same bacterium each induce IL-1beta by distinct signaling pathways. Whereas the spore-induced IL-1beta limits BA infection, LT-induced IL-1beta enables BA to escape host defenses.
Collapse
Affiliation(s)
- Tae Jin Kang
- Center for Vaccine Development, Department of Medicine, University of Maryland School of Medicine, Baltimore, USA
| | - Subhendu Basu
- Center for Vaccine Development, Department of Medicine, University of Maryland School of Medicine, Baltimore, USA
| | - Lei Zhang
- Center for Vaccine Development, Department of Medicine, University of Maryland School of Medicine, Baltimore, USA
| | - Karen E. Thomas
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, USA
| | - Stefanie N. Vogel
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, USA
| | - Les Baillie
- Medical Biotechnology Center, University of Maryland Biotechnology Institute, Baltimore, USA
| | - Alan S. Cross
- Center for Vaccine Development, Department of Medicine, University of Maryland School of Medicine, Baltimore, USA
| |
Collapse
|
154
|
Carson WE, Liang MI. Current immunotherapeutic strategies in breast cancer. Surg Oncol Clin N Am 2008; 16:841-60, ix. [PMID: 18022548 DOI: 10.1016/j.soc.2007.07.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Despite significant advances in the administration of combination cytotoxic chemotherapy, the overall 5-year survival rate is about 75% for a woman who has node-positive breast cancer, and metastatic disease is considered incurable. Recent advances in our understanding of the immune system have led to the hope that manipulation of this organ system could be used as a cancer treatment. Strategies that have been used in the immune therapy of breast cancer include the administration of exogenous cytokines, vaccines, and humanized monoclonal antibodies (mAb). Each of these approaches is discussed in turn in this article.
Collapse
Affiliation(s)
- William E Carson
- Division of Surgical Oncology, The Ohio State University School of Medicine, 410 West 10th Avenue, Columbus, OH 43210-1228, USA.
| | | |
Collapse
|
155
|
Takaoka A, Tamura T, Taniguchi T. Interferon regulatory factor family of transcription factors and regulation of oncogenesis. Cancer Sci 2008; 99:467-78. [PMID: 18190617 PMCID: PMC11159419 DOI: 10.1111/j.1349-7006.2007.00720.x] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2007] [Revised: 11/21/2007] [Accepted: 11/25/2007] [Indexed: 01/03/2023] Open
Abstract
A family of transcription factors, the interferon regulatory factors (IRF), was identified originally in the context of the regulation of the type I interferon (IFN)-alpha/beta system. The IRF family has now expanded to nine members, and gene-disruption studies have revealed the critical involvement of these members in multiple facets of host defense systems, such as innate and adaptive immune responses and tumor suppression. In the present review article, we aim at summarizing our current knowledge of the roles of IRF in host defense, with special emphasis on their involvement in the regulation of oncogenesis.
Collapse
Affiliation(s)
- Akinori Takaoka
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
| | | | | |
Collapse
|
156
|
Guruvayoorappan C, Kuttan G. Biophytum sensitivum (L.) DC Inhibits Tumor Cell Invasion and Metastasis Through a Mechanism Involving Regulation of MMPs, Prolyl Hydroxylase, Lysyl Oxidase, nm23, ERK-1, ERK-2, STAT-1, and Proinflammatory Cytokine Gene Expression in Metastatic Lung Tissue. Integr Cancer Ther 2008; 7:42-50. [DOI: 10.1177/1534735407313744] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Biophytum sensitivum is a traditional oriental herbal medicine that is known for its immunostimulatory and antitumor effects. Tumor metastasis is the most important cause of cancer death. Although B sensitivum was shown to inhibit metastasis, the mechanism underlying this action is not well understood. In the present report, the authors had studied the effect of B sensitivum on the invasion and motility of B16F-10 melanoma cells and investigate the regulatory effect on the expression of matrix metalloproteases (MMPs), prolyl hydoxylase, lysyl oxidase, nm23, extracellular signal-regulated kinase (ERK)—1, ERK-2, signal transducer and activator of transcription (STAT)—1, and proinflammatory cytokines in metastatic tumor-bearing lungs. B sensitivum inhibited the invasion and motility of B16F-10 cells in a dose-dependent manner. B sensitivum inhibited the expression of MMP-2 and MMP-9, whereas it activated STAT-1 expression in metastatic tumor-bearing lungs. Similarly, inhibition of prolyl hydroxylase, lysyl oxidase, ERK-1, ERK-2, and vascular endothelial growth factor (VEGF) expression but activation of nm23 by B sensitivum was observed in metastatic tumor-bearing lungs. B sensitivum treatment also downregulated the expression of tumor necrosis factor—α, interleukin (IL)—1β, IL-6, and granulocyte monocyte—colony stimulating factor in metastatic tumor-bearing lungs. In B16F-10 cells, B sensitivum also inhibited the production of proinflammatory cytokines. Overall, the results indicate that B sensitivum exhibits antimetastatic effects through the inhibition of invasion and motility. The results also suggest that MMPs, prolyl hydroxylase, lysyl oxidase, nm23, ERKs, VEGF, STAT, and proinflammatory cytokines are critical regulators of the B sensitivum—mediated antimetastatic effect.
Collapse
Affiliation(s)
| | - Girija Kuttan
- Department of Immunology, Amala Cancer Research Centre, Kerala State, India,
| |
Collapse
|
157
|
Randall RE, Goodbourn S. Interferons and viruses: an interplay between induction, signalling, antiviral responses and virus countermeasures. J Gen Virol 2008; 89:1-47. [PMID: 18089727 DOI: 10.1099/vir.0.83391-0] [Citation(s) in RCA: 1233] [Impact Index Per Article: 72.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The interferon (IFN) system is an extremely powerful antiviral response that is capable of controlling most, if not all, virus infections in the absence of adaptive immunity. However, viruses can still replicate and cause disease in vivo, because they have some strategy for at least partially circumventing the IFN response. We reviewed this topic in 2000 [Goodbourn, S., Didcock, L. & Randall, R. E. (2000). J Gen Virol 81, 2341-2364] but, since then, a great deal has been discovered about the molecular mechanisms of the IFN response and how different viruses circumvent it. This information is of fundamental interest, but may also have practical application in the design and manufacture of attenuated virus vaccines and the development of novel antiviral drugs. In the first part of this review, we describe how viruses activate the IFN system, how IFNs induce transcription of their target genes and the mechanism of action of IFN-induced proteins with antiviral action. In the second part, we describe how viruses circumvent the IFN response. Here, we reflect upon possible consequences for both the virus and host of the different strategies that viruses have evolved and discuss whether certain viruses have exploited the IFN response to modulate their life cycle (e.g. to establish and maintain persistent/latent infections), whether perturbation of the IFN response by persistent infections can lead to chronic disease, and the importance of the IFN system as a species barrier to virus infections. Lastly, we briefly describe applied aspects that arise from an increase in our knowledge in this area, including vaccine design and manufacture, the development of novel antiviral drugs and the use of IFN-sensitive oncolytic viruses in the treatment of cancer.
Collapse
Affiliation(s)
- Richard E Randall
- School of Biology, University of St Andrews, The North Haugh, St Andrews KY16 9ST, UK
| | - Stephen Goodbourn
- Division of Basic Medical Sciences, St George's, University of London, London SW17 0RE, UK
| |
Collapse
|
158
|
Yang D, Stewart TJ, Smith KK, Georgi D, Abrams SI, Liu K. Downregulation of IFN-gammaR in association with loss of Fas function is linked to tumor progression. Int J Cancer 2008; 122:350-62. [PMID: 17918178 DOI: 10.1002/ijc.23090] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The host immune system functions as an intrinsic surveillance network in the recognition and destruction of tumor cells, and it has been demonstrated that lymphocytes and IFN-gamma are the primary tumor suppressors of the immune system. However, the immune system can concurrently select for tumor variants with reduced immunogenicity and aggressive phenotypes. We report here that tumor escape variants that have survived CTL adoptive immunotherapy exhibited decreased expression levels of both Fas and IFN-gammaR in vitro. Furthermore, examination of spontaneously arising mouse primary mammary carcinoma and lung metastases revealed that both Fas and IFN-gammaR protein levels were dramatically lower in lung metastases than in primary tumors in vivo. Functional disruption of either the Fas- or the IFN-gamma signaling pathway enhanced the colonization efficiency of preexisting metastatic tumor cells, whereas disruption of both Fas and IFN-gammaR pathways resulted in synergistic augmentation of the colonization efficiency of the preexisting metastatic tumor cells, as determined by experimental lung metastases assay. Gene expression profiling revealed that altered expression of genes involved in immediate IFN-gammaR signaling, the interferon primary response, apoptosis and tumor colonization is associated with loss of IFN-gammaR function and enhanced metastatic potential. Interestingly, disruption of IFN-gammaR function did not alter tumor cell susceptibility to CTL-mediated cytotoxicity, but is linked to enhanced infiltration of endogenous T cells in the tumor microenvironment in vivo. These findings suggest that coordinate downregulation of Fas and IFN-gammaR, 2 key components of cancer immunosurveillance system on tumor cells, leads to a more aggressive metastatic phenotype.
Collapse
Affiliation(s)
- Dafeng Yang
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912, USA
| | | | | | | | | | | |
Collapse
|
159
|
Huffman LC, Koch SE, Butler KL. Coronary effluent from a preconditioned heart activates the JAK-STAT pathway and induces cardioprotection in a donor heart. Am J Physiol Heart Circ Physiol 2008; 294:H257-62. [DOI: 10.1152/ajpheart.00769.2007] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Preconditioning (PC) protects against ischemia-reperfusion (I/R) injury via the activation of the JAK-STAT pathway. We hypothesized that the mediators responsible for PC can be transferred to naive myocardium through the coronary effluent. Langendorff-perfused hearts from male Sprague-Dawley rats were randomized to paired donor/acceptor protocols with or without PC in the presence or absence of the JAK-2 inhibitor AG-490 ( n = 6 for each group). Warmed, oxygenated coronary effluent collected during the reperfusion phases of PC (3 cycles of 5 min ischemia and 5 min reperfusion) was administered to acceptor hearts. The hearts were then subjected to 30 min ischemia and 40 min reperfusion. The left ventricles were analyzed for phosphorylated (p)STAT-1, pSTAT-3, Bax, Bcl, Bcl-XL/Bcl-2-associated protein (BAD), and caspase-3 expression by Western blot. A separate group of hearts ( n = 6) was analyzed for STAT activation immediately after the transfer of the PC effluent (no I-R). Baseline cardiodynamics were not different among the groups. End-reperfusion maximal change in pressure over time (+dP/d tmax) was significantly ( P < 0.05) improved in acceptor PC (3,637 ± 199 mmHg/s) and donor PC (4,304 ± 347 mmHg/s) hearts over non-PC donor (2,020 ± 363 mmHg/s) and acceptor (2,624 ± 345 mmHg/s) hearts. Similar differences were seen for minimal change in pressure over time (−dP/d tmin). STAT-3 activation was significantly increased in donor and acceptor PC hearts compared with non-PC hearts. Conversely, pSTAT-1 and Bax expression was decreased in donor and acceptor PC hearts compared with non-PC hearts. No differences in Bcl, BAD, or caspase-3 expression were observed. Treatment with AG-490 attenuated the recovery of ±dP/d t in acceptor PC hearts and significantly reduced pSTAT-3 expression. The PC coronary effluent activates JAK-STAT signaling, limits apoptosis, and protects myocardial performance from I/R injury.
Collapse
|
160
|
Abstract
Signal Transducers and Activators of Transcription (STATs) comprise a family of transcription factors that mediate a wide variety of biological functions in the central and peripheral nervous systems. Injury to neural tissue induces STAT activation, and STATs are increasingly recognized for their role in neuronal survival. In this review, we discuss the role of STAT3 during neural development and following ischemic and traumatic injury in brain, spinal cord and peripheral nerves. We focus on STAT3 because of the expanding body of literature that investigates protective and regenerative effects of growth factors, hormones and cytokines that use STAT3 to mediate their effect, in part through transcriptional upregulation of neuroprotective and neurotrophic genes. Defining the endogenous molecular mechanisms that lead to neuroprotection by STAT3 after injury might identify novel therapeutic targets against acute neural tissue damage as well as chronic neurodegenerative disorders.
Collapse
Affiliation(s)
- Suzan Dziennis
- Department of Anesthesiology & Peri-Operative Medicine, University, Portland, OR, USA
| | - Nabil J. Alkayed
- Department of Anesthesiology & Peri-Operative Medicine, University, Portland, OR, USA
- Physiology & Pharmacology, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
161
|
Barker BR, Parvani JG, Meyer D, Hey AS, Skak K, Letvin NL. IL-21 induces apoptosis of antigen-specific CD8+ T lymphocytes. THE JOURNAL OF IMMUNOLOGY 2007; 179:3596-603. [PMID: 17785794 DOI: 10.4049/jimmunol.179.6.3596] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
IL-21, a member of the common gamma-chain family of cytokines, has pleiotropic effects on T, B, and NK cells. We found that IL-21 and the prototype common gamma-chain cytokine IL-2 can stimulate proliferation and cytokine secretion by Ag-specific rhesus monkey CD8+ T cells. However, unique among the members of this family of cytokines, we found that IL-21 drives these cells to apoptosis by down-regulation of Bcl-2. These findings suggest that IL-21 may play an important role in the contraction of CD8+ T cell responses.
Collapse
Affiliation(s)
- Brianne R Barker
- Division of Viral Pathogenesis, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | | | | | | | | | | |
Collapse
|
162
|
Yu HM, Zhi JL, Cui Y, Tang EH, Sun SN, Feng JQ, Chen PX. Role of the JAK-STAT pathway in protection of hydrogen peroxide preconditioning against apoptosis induced by oxidative stress in PC12 cells. Apoptosis 2007; 11:931-41. [PMID: 16547593 DOI: 10.1007/s10495-006-6578-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The aim of this study was to investigate the role of JAK-STAT pathway in the cytoprotection afforded by preconditioning with H(2)O(2). It was shown that (1) Preconditioning with 100 micromol/L H(2)O(2) can markedly protect PC12 cells against apoptosis and cytotoxicity induced by 300 micromol/L H(2)O(2); (2) The expression and tyrosine phosphorylation of JAK2, not JAK1 were rapidly increased at 5 min after H(2)O(2) preconditioning; (3) The expression of STAT1 and STAT3 were significantly increased at 15 min after H(2)O(2) preconditioning, and the pTyr-STAT1 and pTyr-STAT3 were markedly increased at 60 min after H(2)O(2) preconditioning; (4) Pretreatment with the JAK inhibitor AG-490 (10 micromol/L) 20 min before H(2)O(2) preconditioning blocked not only the activation of JAK2, STAT1 and STAT3, but also the cytoprotection of H(2)O(2) preconditioning against apoptosis and cytotoxicity induced by oxidative stress. These findings suggested that preconditioning with H(2)O(2) activated the JAK-STAT pathway that played an important role in the cytoprotection induced by H(2)O(2) preconditioning.
Collapse
Affiliation(s)
- H M Yu
- Department of Physiology, Zhongshan Medical College, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
163
|
Abstract
Tumour necrosis factor-alpha (TNF-alpha) is a cytokine that is involved in many functions, including the inflammatory response, immunity and apoptosis. Some of the responses of TNF-alpha are mediated by caspase-1, which is involved in the production of the pro-inflammatory cytokines interleukin-1beta, interleukin-18 and interleukin-33. The molecular mechanisms involved in TNF-alpha-induced caspase-1 gene expression remain poorly defined, despite the fact that signaling by TNF-alpha has been well studied. The present study was undertaken to investigate the mechanisms involved in the induction of caspase-1 gene expression by TNF-alpha. Treatment of A549 cells with TNF-alpha resulted in an increase in caspase-1 mRNA and protein expression, which was preceded by an increase in interferon regulatory factor-1 and p73 protein levels. Caspase-1 promoter reporter was activated by the treatment of cells with TNF-alpha. Mutation of the interferon regulatory factor-1 binding site resulted in the almost complete loss of basal as well as of TNF-alpha-induced caspase-1 promoter activity. Mutation of the p53/p73 responsive site resulted in reduced TNF-alpha-induced promoter activity. Blocking of p73 function by a dominant negative mutant or by a p73-directed small hairpin RNA reduced basal as well as TNF-alpha-induced caspase-1 promoter activity. TNF-alpha-induced caspase-1 mRNA and protein levels were reduced when p73 mRNA was down-regulated by small hairpin RNA. Caspase-5 gene expression was induced by TNF-alpha, which was inhibited by the small hairpin RNA-mediated down-regulation of p73. Our results show that TNF-alpha induces p73 gene expression, which, together with interferon regulatory factor-1, plays an important role in mediating caspase-1 promoter activation by TNF-alpha.
Collapse
Affiliation(s)
- Nishant Jain
- Centre for Cellular and Molecular Biology, Hyderabad, India
| | | | | |
Collapse
|
164
|
Roda JM, Joshi T, Butchar JP, McAlees JW, Lehman A, Tridandapani S, Carson WE. The activation of natural killer cell effector functions by cetuximab-coated, epidermal growth factor receptor positive tumor cells is enhanced by cytokines. Clin Cancer Res 2007; 13:6419-28. [PMID: 17962339 DOI: 10.1158/1078-0432.ccr-07-0865] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Natural killer (NK) cells express an activating Fc receptor (FcgammaRIIIa) that mediates antibody-dependent cellular cytotoxicity (ADCC) and production of immune modulatory cytokines in response to antibody-coated targets. Cetuximab is a therapeutic monoclonal antibody directed against the HER1 antigen. We hypothesized that the NK cell response to cetuximab-coated tumor cells could be enhanced by the administration of NK cell-stimulatory cytokines. EXPERIMENTAL DESIGN Human NK cells stimulated with cetuximab-coated tumor cells and interleukin-2 (IL-2), IL-12, or IL-21 were assessed for ADCC and secretion of IFN-gamma and T cell-recruiting chemokines. IL-21 and cetuximab were given to nude mice bearing HER1-positive xenografts. RESULTS Stimulation of human NK cells with cetuximab-coated tumor cells and IL-2, IL-12, or IL-21 resulted in 3-fold to 10-fold higher IFN-gamma production than was observed with either agent alone. NK cell-derived IFN-gamma significantly enhanced monocyte ADCC against cetuximab-coated tumor cells. Costimulated NK cells also secreted elevated levels of chemokines (IL-8, macrophage inflammatory protein-1alpha, and RANTES) that could direct the migration of naive and activated T cells. IL-2, IL-12, and IL-21 enhanced NK cell ADCC against tumor cells treated with cetuximab. The combination of cetuximab, trastuzumab (an anti-HER2 monoclonal antibody), and IL-21 mediated greater NK cell cytokine secretion and ADCC than any agent alone. Furthermore, administration of IL-21 enhanced the effects of cetuximab in a murine tumor model. CONCLUSIONS These results show that cetuximab-mediated NK cell activity can be significantly enhanced in the presence of NK cell-stimulatory cytokines. These factors, therefore, may be effective adjuvants to administer, in combination with cetuximab, to patients with HER1-positive malignancies.
Collapse
Affiliation(s)
- Julie M Roda
- Integrated Biomedical Sciences Graduate Program, Arthur G. James Comprehensive Cancer Center and Solove Research Institute, Ohio State University, Columbus, Ohio 43210, USA
| | | | | | | | | | | | | |
Collapse
|
165
|
Wang H, Zhou M, Brand J, Huang L. Inflammation activates the interferon signaling pathways in taste bud cells. J Neurosci 2007; 27:10703-13. [PMID: 17913904 PMCID: PMC2096741 DOI: 10.1523/jneurosci.3102-07.2007] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2007] [Revised: 08/20/2007] [Accepted: 08/20/2007] [Indexed: 11/21/2022] Open
Abstract
Patients with viral and bacterial infections or other inflammatory illnesses often experience taste dysfunctions. The agents responsible for these taste disorders are thought to be related to infection-induced inflammation, but the mechanisms are not known. As a first step in characterizing the possible role of inflammation in taste disorders, we report here evidence for the presence of interferon (IFN)-mediated signaling pathways in taste bud cells. IFN receptors, particularly the IFN-gamma receptor IFNGR1, are coexpressed with the taste cell-type markers neuronal cell adhesion molecule and alpha-gustducin, suggesting that both the taste receptor cells and synapse-forming cells in the taste bud can be stimulated by IFN. Incubation of taste bud-containing lingual epithelia with recombinant IFN-alpha and IFN-gamma triggered the IFN-mediated signaling cascades, resulting in the phosphorylation of the downstream STAT1 (signal transducer and activator of transcription protein 1) transcription factor. Intraperitoneal injection of lipopolysaccharide or polyinosinic:polycytidylic acid into mice, mimicking bacterial and viral infections, respectively, altered gene expression patterns in taste bud cells. Furthermore, the systemic administration of either IFN-alpha or IFN-gamma significantly increased the number of taste bud cells undergoing programmed cell death. These findings suggest that bacterial and viral infection-induced IFNs can act directly on taste bud cells, affecting their cellular function in taste transduction, and that IFN-induced apoptosis in taste buds may cause abnormal cell turnover and skew the representation of different taste bud cell types, leading to the development of taste disorders. To our knowledge, this is the first study providing direct evidence that inflammation can affect taste buds through cytokine signaling pathways.
Collapse
Affiliation(s)
- Hong Wang
- Monell Chemical Senses Center, Philadelphia, Pennsylvania 19104-3308, and
| | - Minliang Zhou
- Monell Chemical Senses Center, Philadelphia, Pennsylvania 19104-3308, and
| | - Joseph Brand
- Monell Chemical Senses Center, Philadelphia, Pennsylvania 19104-3308, and
- Department of Biochemistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Liquan Huang
- Monell Chemical Senses Center, Philadelphia, Pennsylvania 19104-3308, and
| |
Collapse
|
166
|
Mohib K, Guan Q, Diao H, Du C, Jevnikar AM. Proapoptotic activity of indoleamine 2,3-dioxygenase expressed in renal tubular epithelial cells. Am J Physiol Renal Physiol 2007; 293:F801-12. [PMID: 17609291 DOI: 10.1152/ajprenal.00044.2007] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Exposure of renal tubular epithelial cells (TEC) to IFN-γ/TNF-α leads to Fas/FasL-mediated self-injury, which contributes to allograft rejection. Indoleamine 2,3-dioxygenase (IDO) converts tryptophan to N-formyl-kynurenine and contributes to immune privilege in tissues by increasing Fas-mediated T cell apoptosis. However, renal expression of IDO and its role in promoting Fas-mediated TEC death have not been examined. IDO expression was analyzed by RT-PCR and Western blot. Apoptosis was measured by fluorescence-activated cell sorting analysis and terminal deoxytransferase-mediated dUTP nick end labeling. We demonstrated that functional IDO is expressed in TEC and is increased by IFN-γ/TNF-α exposure. Increased IDO activity promoted TEC apoptosis, whereas inhibition of IDO by its specific inhibitor 1-methyl-d-tryptophan attenuated IFN-γ/TNF-α-mediated TEC apoptosis and augmented TEC survival. Transgenic expression of IDO resulted in increased TEC apoptosis in the absence of proinflammatory cytokine exposure, supporting a central role for IDO in TEC injury. Inhibition of IDO-mediated TEC death by a caspase-8-specific inhibitor (Z-IETD-FMK), as well as the absence of an IDO effect in Fas-deficient and FasL-deficient TEC, supports a Fas/FasL-dependent, caspase-8-mediated mechanism for IDO-enhanced TEC death. These data suggest that renal IDO expression may be deleterious during renal inflammation, because it enhances TEC self-injury through Fas/FasL interactions. Thus attenuation of IDO may represent a novel strategy to promote kidney function following ischemia and renal allograft rejection.
Collapse
Affiliation(s)
- Kanishka Mohib
- Department of Medicine and Microbiology, The University of Western Ontario, London, Ontario, Canada
| | | | | | | | | |
Collapse
|
167
|
Ghazani AA, Arneson N, Warren K, Pintilie M, Bayani J, Squire JA, Done SJ. Genomic alterations in sporadic synchronous primary breast cancer using array and metaphase comparative genomic hybridization. Neoplasia 2007; 9:511-20. [PMID: 17603634 PMCID: PMC1899252 DOI: 10.1593/neo.07301] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2007] [Revised: 05/03/2007] [Accepted: 05/03/2007] [Indexed: 01/19/2023] Open
Abstract
Synchronous primary breast cancer describes the occurrence of multiple tumors affecting one or both breasts at initial diagnosis. This provides a unique opportunity to identify tissue-specific genomic markers that characterize each tumor while controlling for the individual genetic background of a patient. The aim of this study was to examine the genomic alterations and degree of similarity between synchronous cancers. Using metaphase comparative genomic hybridization and array comparative genomic hybridization (aCGH), the genomic alterations of 23 synchronous breast cancers from 10 patients were examined at both chromosomal and gene levels. Synchronous breast cancers, when compared to their matched counterparts, were found to have a common core set of genetic alterations, with additional unique changes present in each. They also frequently exhibited features distinct from the more usual solitary primary breast cancers. The most frequent genomic alterations included chromosomal gains of 1q, 3p, 4q, and 8q, and losses of 11q, 12q, 16q, and 17p. aCGH identified copy number amplification in regions that are present in all 23 tumor samples, including 1p31.3-1p32.3 harboring JAK1. Our findings suggest that synchronous primary breast cancers represent a unique type of breast cancer and, at least in some instances, one tumor may give rise to the other.
Collapse
MESH Headings
- Breast Neoplasms/genetics
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/genetics
- Carcinoma, Ductal, Breast/pathology
- Chromosome Aberrations
- Chromosomes, Human
- DNA, Neoplasm
- Genome, Human
- Humans
- In Situ Hybridization, Fluorescence
- Lasers
- Metaphase
- Microdissection
- Mitosis
- Neoplasm Invasiveness/genetics
- Neoplasm Invasiveness/pathology
- Nucleic Acid Hybridization
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Tissue Array Analysis
Collapse
Affiliation(s)
- Arezou A Ghazani
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
168
|
Harada D, Yamanaka Y, Ueda K, Nishimura R, Morishima T, Seino Y, Tanaka H. Sustained phosphorylation of mutated FGFR3 is a crucial feature of genetic dwarfism and induces apoptosis in the ATDC5 chondrogenic cell line via PLCgamma-activated STAT1. Bone 2007; 41:273-81. [PMID: 17561467 DOI: 10.1016/j.bone.2006.11.030] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2006] [Revised: 11/03/2006] [Accepted: 11/14/2006] [Indexed: 12/23/2022]
Abstract
The most frequent type of rhizomelic dwarfism, achondroplasia (ACH), is caused by mutations in the fibroblast growth factor receptor 3 (FGFR3) gene. Mutations in FGFR3 result in skeletal dysplasias of variable severity, including mild phenotypic effects in hypochondroplasia (HCH), severe phenotypic effects in thanatophoric dysplasia types I (TDI) and II (TDII), and severe but survivable phenotypic effects in severe achondroplasia with developmental delay and acanthosis nigricans (SADDAN). To explore the molecular mechanisms that result in the different phenotypes, we investigated the kinetics of mutated versions of FGFR3. First, we assayed the phosphorylation states of the mutated FGFR3s and found that the level of phosphorylation in TDI-FGFR3 was lower than in ACH-FGFR3, although the other mutants were phosphorylated according to phenotypic severity. Second, we analyzed the duration of the phosphorylation. TDI-FGFR3 was not highly phosphorylated under ligand-free conditions, but the peak phosphorylation levels of TDI-FGFR3 and ACH-FGFR3 were maintained for 30 min after stimulation with FGF-1. Moreover, ligand-dependent phosphorylation of TDI-FGFR3, but not ACH-FGFR3, lasted for more than 8 h after FGF-1 administration. The other mutant proteins showed sustained phosphorylation independent of ligand presence. Third, we investigated the intracellular localization of the mutant proteins. Immunofluorescence analysis showed accumulations of TDII-FGFR3, SADDAN-FGFR3, and a portion of TDI-FGFR3 in the endoplasmic reticulum (ER). Based on these data, we concluded that sustained phosphorylation of FGFR3 causes chondrodysplasia, and the phenotypic severity depends on the proportion of ER-localized mutant FGFR3. In FGFR3 signaling, the transcription factor, signal transducer and activator of transcription 1 (STAT1) inhibit proliferation and induce apoptosis of chondrocytes. Here we reveal that phospholipase C gamma (PLCgamma) mediates FGFR3-induced STAT1 activation. Both PLCgamma and STAT1 were activated by FGFR3 signaling, but a dominant-negative form of PLCgamma (DN-PLCgamma) remarkably reduced STAT1 phosphorylation. Apoptosis assays revealed that the constitutively active forms of FGFR3 (TDII-FGFR3) and STAT1 (STAT1-C) induce apoptosis of chondrogenic ATDC5 cells via caspase activity. DN-PLCgamma reduced the apoptosis of ATDC5 cells expressing TDII-FGFR3, but over-expression of both DN-PLCgamma and STAT1-C induced apoptosis. Therefore, we conclude that a PLCgamma-STAT1 pathway mediates apoptotic signaling by FGFR3.
Collapse
Affiliation(s)
- Daisuke Harada
- Department of Pediatrics, Okayama University, Graduate School of Medicine and Dentistry, 2-5-1 Shikata-Cho, Okayama 700-8558, Japan
| | | | | | | | | | | | | |
Collapse
|
169
|
Grudinkin PS, Zenin VV, Kropotov AV, Dorosh VN, Nikolsky NN. EGF-induced apoptosis in A431 cells is dependent on STAT1, but not on STAT3. Eur J Cell Biol 2007; 86:591-603. [PMID: 17646016 DOI: 10.1016/j.ejcb.2007.05.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2006] [Revised: 05/22/2007] [Accepted: 05/24/2007] [Indexed: 11/21/2022] Open
Abstract
EGF in high concentrations has a growth-inhibitory effect on human epidermoid carcinoma cells A431. The transcription factor STAT1 is the most probable candidate for mediating this effect. In the present study, we demonstrated a strong reduction of the expression level of STAT1 in EGF-resistant sub-clones of A431 cells. EGF resistance was reversed by introducing wild-type STAT1, but not its Y701F mutant. Moreover, blocking the activity of Src family kinases reduced tyrosine phosphorylation of STAT1 and STAT3 and protected A431 cells from the EGF-induced growth inhibition. To further elucidate roles of STATs in A431 cell growth and survival, clones of A431 cells expressing short hairpin RNA (shRNA) against STAT1 or STAT3 were generated. Neither STAT1 nor STAT3 knockdown exerted any effect on growth rate or apoptotic death of A431 cells in the absence of EGF. However, upon EGF treatment A431 cells with knocked down STAT1 continued to grow and demonstrated a significantly lower level of apoptosis as compared to A431 cells. The knockdown of STAT3 did not alter cell growth or apoptosis. Taken together, our experiments prove the essential role of tyrosine phosphorylated STAT1, but not of STAT3, in EGF-induced apoptosis in A431 cells.
Collapse
Affiliation(s)
- Pavel S Grudinkin
- Department of Intracellular Signalling and Transport, Institute of Cytology RAS, 194064 St. Petersburg, Russia.
| | | | | | | | | |
Collapse
|
170
|
Akamatsu N, Yamada Y, Hasegawa H, Makabe K, Asano R, Kumagai I, Murata K, Imaizumi Y, Tsukasaki K, Tsuruda K, Sugahara K, Atogami S, Yanagihara K, Kamihira S. High IL-21 receptor expression and apoptosis induction by IL-21 in follicular lymphoma. Cancer Lett 2007; 256:196-206. [PMID: 17624663 DOI: 10.1016/j.canlet.2007.06.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2007] [Revised: 05/25/2007] [Accepted: 06/05/2007] [Indexed: 11/28/2022]
Abstract
We surveyed IL-21 receptor (IL-21R) in leukemia and lymphoma and found that follicular lymphoma cells showed exceptionally high IL-21R expression. Notably, IL-21 showed divergent effects depending on the cell origin: growth stimulation in Burkitt lymphoma cell lines and adult T cell leukemia/lymphoma cell lines but induction of apoptosis in B lymphoma cell lines with t(14;18)(q32;q21), a marker karyotype of follicular lymphoma. IL-21 activated caspase-8 and -3 and reduced mitochondrial membrane potential. More importantly, IL-21 decreased Bcl-2 expression but increased Bax expression. These results support a new therapeutic approach using the IL-21/IL-21R system in follicular lymphoma.
Collapse
Affiliation(s)
- Norihiko Akamatsu
- Department of Laboratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
171
|
Dinis-Oliveira RJ, Sousa C, Remião F, Duarte JA, Ferreira R, Sánchez Navarro A, Bastos ML, Carvalho F. Sodium salicylate prevents paraquat-induced apoptosis in the rat lung. Free Radic Biol Med 2007; 43:48-61. [PMID: 17561093 DOI: 10.1016/j.freeradbiomed.2007.03.014] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2006] [Revised: 03/11/2007] [Accepted: 03/13/2007] [Indexed: 11/24/2022]
Abstract
The nonselective contact herbicide, paraquat (PQ), is a strong pneumotoxicant, especially due to its accumulation in the lung through a polyamine uptake system and to its capacity to induce redox cycling, leading to oxidative stress-related damage. In the present study, we aimed to investigate the occurrence of apoptotic events in the lungs of male Wistar rats, 24, 48, and 96 h after PQ exposure (25 mg/kg ip) as well as the putative healing effects provided by sodium salicylate [(NaSAL), 200 mg/kg ip] when administered 2 h after PQ. PQ exposure resulted in marked lung apoptosis, in a time-dependent manner, characterized by the "ladder-like" pattern of DNA observed through electrophoresis and by the presence of terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end-labeling (TUNEL)-positive cells (TPC) as revealed by immunohistochemistry. The two main caspase cascades (the extrinsic receptor-mediated and the intrinsic mitochondria-mediated) and the expressions of p53 and activator protein-1 (AP-1) were also evaluated, to obtain an insight into apoptotic cellular signaling. PQ-exposed rats suffered a time-dependent increase of caspase-3 and caspase-8 and a decrease of caspase-1 activities in lungs compared to the control group. A marked mitochondrial dysfunction evidenced by cytochrome c (Cyt c) release was also observed as a consequence of PQ exposure. In addition, fluorescence electrophoretic mobility shift assay (fEMSA) revealed a transcriptional induction of the p53 and AP-1 transcription factors in a time-dependent manner as a consequence of PQ exposure. NaSAL treatment resulted in the remission of the observed apoptotic signaling and consequently of lung apoptosis. Taken together, the present results showed that PQ activates several events involved in the apoptotic pathways, which might contribute to its lung toxicodynamics. NaSAL, a recently implemented antidote for PQ intoxications, proved to protect lungs from PQ-induced apoptosis.
Collapse
Affiliation(s)
- R J Dinis-Oliveira
- REQUIMTE, Departamento de Toxicologia, Faculdade de Farmácia, Universidade do Porto, Rua Aníbal Cunha, 164, 4099-030 Porto, Portugal.
| | | | | | | | | | | | | | | |
Collapse
|
172
|
Stout BA, Melendez K, Seagrave J, Holtzman MJ, Wilson B, Xiang J, Tesfaigzi Y. STAT1 activation causes translocation of Bax to the endoplasmic reticulum during the resolution of airway mucous cell hyperplasia by IFN-gamma. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2007; 178:8107-16. [PMID: 17548649 DOI: 10.4049/jimmunol.178.12.8107] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Disruption of the normal resolution process of inflammation-induced mucous cell hyperplasia may lead to sustained mucous hypersecretion in chronic diseases. During prolonged exposure of mice to allergen, IFN-gamma reduces mucous cell hyperplasia, but the signaling responsible for the cell death is largely unknown. A brief phosphorylation of STAT1 by IFN-gamma was required for cell death in airway epithelial cells (AEC), and during prolonged exposure to allergen, mucous cell hyperplasia remained elevated in STAT1(-/-) but was resolved in STAT1(+/+) mice. Although IFN-gamma treatment of primary human AECs and other airway cell lines left Bax protein levels unchanged, it caused translocation of Bax from the cytosol to the endoplasmic reticulum (ER) but not to the mitochondria. Localization of Bax to the ER was observed in IFN-gamma-treated primary AECs isolated from STAT1(+/+) mice but not in cells from STAT1(-/-) mice. In addition, ER Bax was detected in mucous cells of STAT1(+/+) but not STAT1(-/-) airways of mice exposed to allergen for prolonged periods. IFN-gamma did not release cytochrome c from mitochondria but reduced ER calcium stores and dilated the ER, confirming that the IFN-gamma-induced cell death is mediated through changes localized in the ER. Collectively, these observations suggest that STAT1-dependent translocation of Bax to the ER is crucial for IFN-gamma-induced cell death of AECs and the resolution of allergen-induced mucous cell hyperplasia.
Collapse
Affiliation(s)
- Barbara A Stout
- Lovelace Respiratory Research Institute, 2425 Ridgecrest Drive SE, Albuquerque, NM 87185, USA
| | | | | | | | | | | | | |
Collapse
|
173
|
Lui VWY, Boehm AL, Koppikar P, Leeman RJ, Johnson D, Ogagan M, Childs E, Freilino M, Grandis JR. Antiproliferative mechanisms of a transcription factor decoy targeting signal transducer and activator of transcription (STAT) 3: the role of STAT1. Mol Pharmacol 2007; 71:1435-43. [PMID: 17325127 DOI: 10.1124/mol.106.032284] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
We previously developed a transcription factor decoy targeting signal transducer and activator of transcription 3 (STAT3) and reported antitumor activity in both in vitro and in vivo models of squamous cell carcinoma of the head and neck (SCCHN). Based on the known existence of STAT1-STAT3 heterodimers, the high sequence homology between STAT1 and STAT3, as well as expression of both STAT1 and STAT3 in SCCHN, we examined whether the STAT3 decoy interferes with STAT1 signaling. SCCHN cell lines with different STAT1 expression levels (but similar STAT3 levels) were used. Both cell lines were sensitive to the growth-inhibitory effects of the STAT3 decoy compared with a mutant control decoy. Intact STAT1 signaling was demonstrated by interferon-gamma (IFN-gamma)-mediated induction of STAT1 phosphorylation (Tyr701) and interferon-regulatory factor-1 (IRF-1) expression. Treatment with the STAT3 decoy (but not a mutant control decoy) resulted in inhibition of IRF-1 protein expression in both cell lines, indicating specific inhibition of STAT1 signaling by the STAT3 decoy. Because STAT1 is a potential tumor suppressor, we also investigated whether STAT1 signaling mitigated the therapeutic efficacy of the STAT3 decoy. In both PCI-15B and UM-22B cells, STAT1 siRNA treatment resulted in decreased STAT1 expression, without altering the antitumor activity of the STAT3 decoy. Likewise, the antitumor effects of the STAT3 decoy were not altered by STAT1 activation upon IFN-gamma treatment. These results suggest that the therapeutic mechanisms of STAT3 blockade using a transcription factor decoy are independent of STAT1 activation.
Collapse
Affiliation(s)
- Vivian Wai Yan Lui
- Department of Clinical Oncology, Chinese University of Hong Kong, Shatin, Hong Kong
| | | | | | | | | | | | | | | | | |
Collapse
|
174
|
Abstract
It is firmly established that interferon-gamma (IFN-gamma) plays a mandatory role in acquired protective immunity to pathogenic mycobacteria and other intracellular pathogens. Therefore, it seems conceivable that application of recombinant IFN-gamma could be exploited for the treatment of tuberculosis. However, the results of experimental studies and clinical trials, conducted mostly in patients with multidrug resistant (MDR) disease, have thus far been only moderately encouraging. Further studies are now needed to determine if a greater clinical benefit from IFN-gamma could be obtained for the prophylactic treatment of latent tuberculosis infection and for shortening of the protracted standard chemotherapy regimen. Thus, aerosolized IFN-gamma treatment could be particularly beneficial to AIDS patients at high risk of developing mycobacterial infections, that is, those with significantly declined CD4(+) T cell counts. This review describes the current state of research on IFN-gamma interventions in tuberculosis and related infections and highlights some of the future opportunities.
Collapse
Affiliation(s)
- Rajko Reljic
- Department of Cellular and Molecular Medicine, St George's Hospital Medical School, University of London, 43 Cranmer Terrace, London, England.
| |
Collapse
|
175
|
Vadrot N, Legrand A, Nello E, Bringuier AF, Guillot R, Feldmann G. Inducible nitric oxide synthase (iNOS) activity could be responsible for resistance or sensitivity to IFN-gamma-induced apoptosis in several human hepatoma cell lines. J Interferon Cytokine Res 2007; 26:901-13. [PMID: 17238833 DOI: 10.1089/jir.2006.26.901] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Response to interferon-gamma (IFN-gamma)-induced apoptosis of human hepatoma cell lines (HHCLs) is variable. We analyzed this different behavior in Hep3B, Chang-liver, HepG2, and HuH7 cells. We studied (1) IFN-gamma-induced apoptosis, (2) protein expression of Stat1, (3) binding of nuclear proteins to IFN-gamma activated sequence (GAS), (4) mRNA and expression of proteins acting in apoptosis, and (5) HuH7 sensitivity after inducible nitric oxide synthase (iNOS) siRNA transfection. IFN-gamma induced apoptosis in Hep3B and Chang-liver cells only. In all HHCLs, Stat1 protein increased. Binding of proteins and transactivation activity of GAS increased much more in HuH7. In all HHCLs, caspase activity and apoptotic proteins were not implicated in resistance or sensitivity. iNOS mRNA and protein expression increased in HuH7, disappeared in Hep3B, and remained unchanged in Chang-liver and HepG2. We compared the role of iNOS in Hep3B and HuH7. The iNOS inhibitor, L-NAME, sensitized HuH7 to IFN-gamma, Hep3B/HuH7 coculture partially inhibited Hep3B apoptosis, and HuH7 transfection with iNOS siRNA induced a 50% inhibition of iNOS protein and cell apoptosis. GAS activity and overexpression of iNOS in HuH7, but not in the other HHCLs, suggest that this enzyme could play an important role in the resistance of HuH7 to IFN-gamma-induced apoptosis, perhaps by the antiapoptotic action of NO.
Collapse
Affiliation(s)
- Nathalie Vadrot
- INSERM U773, Centre de Recherche Biologique CRB3, Equipe 5, U.F.R. de Médecine Denis Diderot, site Xavier Bichat (Université Paris 7-Denis Diderot), France
| | | | | | | | | | | |
Collapse
|
176
|
Burova E, Vassilenko K, Dorosh V, Gonchar I, Nikolsky N. Interferon gamma-dependent transactivation of epidermal growth factor receptor. FEBS Lett 2007; 581:1475-80. [PMID: 17362940 DOI: 10.1016/j.febslet.2007.03.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2007] [Accepted: 03/01/2007] [Indexed: 11/16/2022]
Abstract
The present report provides evidence that, in A431 cells, interferon gamma (IFNgamma) induces the rapid (within 5 min), and reversible, tyrosine phosphorylation of the epidermal growth factor receptor (EGFR). IFNgamma-induced EGFR transactivation requires EGFR kinase activity, as well as activity of the Src-family tyrosine kinases and JAK2. Here, we show that IFNgamma-induced STAT1 activation in A431 and HeLa cells partially depends on the kinase activity of both EGFR and Src. Furthermore, in these cells, EGFR kinase activity is essential for IFNgamma-induced ERK1,2 activation. This study is the first to demonstrate that EGFR is implicated in IFNgamma-dependent signaling pathways.
Collapse
Affiliation(s)
- Elena Burova
- Department of Intracellular Signaling and Transport, Institute of Cytology of Russian Academy of Sciences, St.-Petersburg 194064, Russia.
| | | | | | | | | |
Collapse
|
177
|
Cuesta N, Nhu QM, Zudaire E, Polumuri S, Cuttitta F, Vogel SN. IFN Regulatory Factor-2 Regulates Macrophage Apoptosis through a STAT1/3- and Caspase-1-Dependent Mechanism. THE JOURNAL OF IMMUNOLOGY 2007; 178:3602-11. [PMID: 17339457 DOI: 10.4049/jimmunol.178.6.3602] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
IFN regulatory factor (IRF)-2(-/-) mice are significantly more resistant to LPS challenge than wild-type littermates, and this was correlated with increased numbers of apoptotic Kupffer cells. To assess the generality of this observation, and to understand the role of IRF-2 in apoptosis, responses of peritoneal macrophages from IRF-2(+/+) and IRF-2(-/-) mice to apoptotic stimuli, including the fungal metabolite, gliotoxin, were compared. IRF-2(-/-) macrophages exhibited a consistently higher incidence of apoptosis that failed to correlate with caspase-3/7 activity. Using microarray gene expression profiling of liver RNA samples derived from IRF-2(+/+) and IRF-2(-/-) mice treated with saline or LPS, we identified >40 genes that were significantly down-regulated in IRF-2(-/-) mice, including Stat3, which has been reported to regulate apoptosis. Compared with IRF-2(+/+) macrophages, STAT3alpha mRNA was up-regulated constitutively or after gliotoxin treatment of IRF-2(-/-) macrophages, whereas STAT3beta mRNA was down-regulated. Phospho-Y705-STAT3, phospho-S727-STAT1, and phospho-p38 protein levels were also significantly higher in IRF-2(-/-) than control macrophages. Activation of the STAT signaling pathway has been shown to elicit expression of CASP1 and apoptosis. IRF-2(-/-) macrophages exhibited increased basal and gliotoxin-induced caspase-1 mRNA expression and enhanced caspase-1 activity. Pharmacologic inhibition of STAT3 and caspase-1 abolished gliotoxin-induced apoptosis in IRF-2(-/-) macrophages. A novel IFN-stimulated response element, identified within the murine promoter of Casp1, was determined to be functional by EMSA and supershift analysis. Collectively, these data support the hypothesis that IRF-2 acts as a transcriptional repressor of Casp1, and that the absence of IRF-2 renders macrophages more sensitive to apoptotic stimuli in a caspase-1-dependent process.
Collapse
Affiliation(s)
- Natalia Cuesta
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 600 West Baltimore Street, Baltimore, MD 21201, USA
| | | | | | | | | | | |
Collapse
|
178
|
Abstract
Signal transducers and activators of transcription (STATs) are latent cytoplasmic transcription factors that mediate various biological responses, including cell proliferation, survival, apoptosis, and differentiation. Among the members of the STAT family, accumulating evidence now indicates an important role for STAT1 in various forms of cell death. Depending upon stimuli or cell types, STAT1 can modulate a broad spectrum of cell death, comprising both apoptotic and non-apoptotic pathways. STAT1-dependent regulation of cell death is largely dependent on a transcriptional mechanism such as the activation of death-promoting genes. However, non-transcriptional mechanisms such as STAT1 interaction with TRADD, p53, or HDAC have been implicated in the regulation of cell death by STAT1. Furthermore, STAT1 itself is also subject to complex forms of regulation such as post-translational protein modification, which can critically affect STAT1 signaling and STAT1-dependent cell death. Given the reports showing that dysregulation of STAT1 signaling is associated with various pathological conditions, including the development of cancer, a better understanding of the mechanism underlying STAT1 regulation of cell death may lead to successful strategies for targeting STAT1 in such pathological settings.
Collapse
Affiliation(s)
- Hun Sik Kim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | | |
Collapse
|
179
|
Abstract
BACKGROUND/PURPOSE In previous studies, incubation of doxorubicin-resistant neuroblastoma SK-N-SH (Dox-R) cells with epidermal growth factor (EGF) decreased extracellular signal-regulated kinase activation. Because extracellular signal-regulated kinase activation is associated with cell proliferation, we hypothesized that EGF could induce apoptosis and decrease the rate of cell growth in these cells. METHODS The growth of wild-type (WT) SK-N-SH and Dox-R cells after incubation with EGF concentrations ranging from 1 to 100 ng/mL was determined by a colorimetric assay. Apoptosis was assessed by Hoechst staining and DNA laddering in WT, Dox-R, and cisplatin-resistant cells treated with EGF (100 ng/mL). Cleaved caspase-3 and EGF receptor (human epidermal growth factor receptor [HER1-HER4]) expression were verified by Western blot and reverse transcriptase-polymerase chain reaction. RESULTS Epidermal growth factor decreased WT cell growth at concentrations between 50 and 100 ng/mL; Dox-R cell growth was attenuated at all EGF concentrations. Apoptosis was observed in WT and Dox-R cells incubated with EGF. Maximal cleaved caspase-3 expression occurred in WT cells treated with EGF 100 ng/mL and in Dox-R treated with EGF 5 to 10 ng/mL. Epidermal growth factor did not induce apoptosis in cisplatin-resistant cells. HER2 and HER3 transcription was maximal in WT and Dox-R cells, respectively. CONCLUSIONS Wild-type and Dox-R cells exhibited decreased cell growth after EGF treatment because of apoptosis. This involved caspase-3 activation and could work through HER2 and HER3 receptors.
Collapse
Affiliation(s)
- Bill Chiu
- Department of Surgery, Children's Memorial Hospital, Northwestern University Feinberg School of Medicine, Chicago, IL 60614, USA
| | | | | |
Collapse
|
180
|
Moiseeva EP, Heukers R, Manson MM. EGFR and Src are involved in indole-3-carbinol-induced death and cell cycle arrest of human breast cancer cells. Carcinogenesis 2007; 28:435-45. [PMID: 16956907 DOI: 10.1093/carcin/bgl171] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Indole-3-carbinol (I3C), a dietary chemopreventive compound, induced marked reduction in epidermal growth factor receptor (EGFR) prior to cell death in cells representing three breast cancer subtypes. Signalling pathways, linking these events were investigated in detail. I3C modulated tyrosine phosphorylation from 30 min in four cell lines. In MDA-MB-468 and HBL100 cells, it induced Src activation after 5 h. In MDA-MB-468 cells, I3C induced signalling between 4.5 and 7 h, which involved sequential activation of Src, EGFR, STAT-1 and STAT-3, followed by EGFR degradation. It also induced physical association between activated Src and EGFR. In MCF7 and MDA-MB-231 cells, I3C modulated expression of cell cycle-related proteins, p21Cip1, p27Kip1, cyclin E, cyclin D1 and CDK6, with upregulation of p21Cip1 and cyclin E being dependent on Src. Inhibition of EGFR by specific inhibitors PD153035 or ZD1839 increased susceptibility to I3C-induced apoptosis of MCF7, MDA-MB-468 and MDA-MB-231 cells. Inhibition of Src sensitized MDA-MB-468 and MDA-MB-231 cells to I3C, whereas overexpression of c-Src increased resistance to I3C in MDA-MB-468 and HBL100 cells. Modulation of Src in MDA-MB-468 cells influenced the basal level of EGFR expression and cell viability; the latter being positively correlated with EGFR activation levels. Therefore, EGFR and Src activities are essential for I3C-induced cell cycle arrest and death; however, I3C-induced pathways depend on specific features of breast cancer cells. The cancer types, which rely on 'EGFR addiction' or Src deregulation, are likely to be susceptible to I3C.
Collapse
Affiliation(s)
- Elena P Moiseeva
- Cancer Biomarkers and Prevention Group, Department of Biochemistry, University of Leicester, Leicester LE1 7RH, UK.
| | | | | |
Collapse
|
181
|
Hückel M, Schurigt U, Wagner AH, Stöckigt R, Petrow PK, Thoss K, Gajda M, Henzgen S, Hecker M, Bräuer R. Attenuation of murine antigen-induced arthritis by treatment with a decoy oligodeoxynucleotide inhibiting signal transducer and activator of transcription-1 (STAT-1). Arthritis Res Ther 2007; 8:R17. [PMID: 16507120 PMCID: PMC1526583 DOI: 10.1186/ar1869] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2005] [Revised: 10/27/2005] [Accepted: 11/21/2005] [Indexed: 01/05/2023] Open
Abstract
The transcription factor STAT-1 (signal transducer and activator of transcription-1) plays a pivotal role in the expression of inflammatory gene products involved in the pathogenesis of arthritis such as various cytokines and the CD40/CD40 ligand (CD40/CD40L) receptor-ligand dyad. The therapeutic efficacy of a synthetic decoy oligodeoxynucleotide (ODN) binding and neutralizing STAT-1 was tested in murine antigen-induced arthritis (AIA) as a model for human rheumatoid arthritis (RA). The STAT-1 decoy ODN was injected intra-articularly in methylated bovine serum albumin (mBSA)-immunized mice 4 h before arthritis induction. Arthritis was evaluated by joint swelling measurement and histological evaluation and compared to treatment with mutant control ODN. Serum levels of pro-inflammatory cytokines, mBSA-specific antibodies and auto-antibodies against matrix constituents were assessed by enzyme-linked immunosorbent assay (ELISA). The transcription factor neutralizing efficacy of the STAT-1 decoy ODN was verified in vitro in cultured synoviocytes and macrophages. Single administration of STAT-1 decoy ODN dose-dependently suppressed joint swelling and histological signs of acute and chronic arthritis. Delayed-type hypersensitivity (DTH) reaction, serum levels of interleukin-6 (IL-6) and anti-proteoglycan IgG titres were significantly reduced in STAT-1 decoy ODN-treated mice, whereas mBSA, collagen type I and type II specific immunoglobulins were not significantly affected. Intra-articular administration of an anti-CD40L (anti-CD154) antibody was similarly effective. Electrophoretic mobility shift analysis (EMSA) of nuclear extracts from synoviocytes incubated with the STAT-1 decoy ODN in vitro revealed an inhibitory effect on STAT-1. Furthermore, the STAT-1 decoy ODN inhibited the expression of CD40 mRNA in stimulated macrophages. The beneficial effects of the STAT-1 decoy ODN in experimental arthritis presumably mediated in part by affecting CD40 signalling in macrophages may provide the basis for a novel treatment of human RA.
Collapse
Affiliation(s)
- Marion Hückel
- Institute of Pathology, Friedrich Schiller University, Jena, Germany
| | - Uta Schurigt
- Institute of Pathology, Friedrich Schiller University, Jena, Germany
| | - Andreas H Wagner
- Institute of Physiology and Pathophysiology, Ruprecht Karls University, Heidelberg, Germany
| | - Renate Stöckigt
- Institute of Pathology, Friedrich Schiller University, Jena, Germany
| | - Peter K Petrow
- Institute of Pathology, Friedrich Schiller University, Jena, Germany
| | - Klaus Thoss
- Institute of Pathology, Friedrich Schiller University, Jena, Germany
| | - Mieczyslaw Gajda
- Institute of Pathology, Friedrich Schiller University, Jena, Germany
| | - Steffen Henzgen
- Institute of Pathology, Dietrich Bonhoeffer Clinical Centre, Neubrandenburg, Germany
| | - Markus Hecker
- Institute of Physiology and Pathophysiology, Ruprecht Karls University, Heidelberg, Germany
| | - Rolf Bräuer
- Institute of Pathology, Friedrich Schiller University, Jena, Germany
| |
Collapse
|
182
|
Kaganoi J, Watanabe G, Okabe M, Nagatani S, Kawabe A, Shimada Y, Imamura M, Sakai Y. STAT1 activation-induced apoptosis of esophageal squamous cell carcinoma cells in vivo. Ann Surg Oncol 2006; 14:1405-15. [PMID: 17195908 DOI: 10.1245/s10434-006-9274-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2006] [Revised: 10/04/2006] [Accepted: 10/18/2006] [Indexed: 11/18/2022]
Abstract
BACKGROUND The induction of apoptosis might be a promising treatment for cancers refractory to conventional therapies, such as esophageal cancer. In this study, we examined whether epidermal growth factor-induced growth inhibition results from apoptosis of esophageal squamous cell carcinoma (SCC) cells as a result of STAT1 activation and evaluated whether interferon gamma (IFN-gamma) can induce apoptosis of cancer cells in vivo. METHODS To assess the function of STAT1, we established stable transfectants expressing dominant-negative STAT1. Apoptosis was assessed by several experimental techniques, including flow cytometry. Differentiation was evaluated by Western blot test with involucrin used as a marker. In vivo, cancer cells were injected into male BALB/c nu/nu mice. Two weeks later, the mice started to receive injections of IFN-gamma or saline into a tail vein four times per week. Concentrations of IFN-gamma in the tumors were analyzed by enzyme-linked immunosorbent assay. Apoptosis was evaluated by TUNEL (terminal deoxynucleotidyl transferase dUTP nick-end labeling) staining. RESULTS Epidermal growth factor inhibited the growth of esophageal SCC cells by causing apoptosis through several pathways involving STAT1 activation. IFN-gamma induced the apoptosis of cancer cells, but it also promoted the differentiation (not apoptosis) of primary cultured cells derived from normal esophageal epithelium. IFN-gamma also inhibited the growth of xenograft tumors of esophageal SCC cells in vivo. CONCLUSIONS Our results suggest that IFN-gamma is one candidate for cytokine-based therapy of cancer. IFN-gamma-induced STAT1 activation might be involved in the apoptosis of esophageal SCC cells and in the terminal differentiation of normal squamous cells. Further studies of STAT1 signaling pathways may provide the basis for new targeted therapeutic strategies for esophageal SCC.
Collapse
Affiliation(s)
- Junichi Kaganoi
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | | | | | | | | | | | | | | |
Collapse
|
183
|
Barry SP, Townsend PA, Latchman DS, Stephanou A. Role of the JAK-STAT pathway in myocardial injury. Trends Mol Med 2006; 13:82-9. [PMID: 17194625 DOI: 10.1016/j.molmed.2006.12.002] [Citation(s) in RCA: 134] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2006] [Revised: 11/24/2006] [Accepted: 12/14/2006] [Indexed: 01/03/2023]
Abstract
Cardiovascular pathologies are an enormous burden in human health and despite the vast amount of research; the molecular mechanisms and pathways that control the underlying pathologies are still not fully appreciated. The Janus kinase (JAK)-signal transducers and activators of transcription (STAT) pathway has recently been shown to be an integral part of the response of the myocardium to various cardiac insults, including myocardial infarction, oxidative damage, myocarditis, hypertrophy and remodeling, in addition to having a prominent role in cardioprotective therapies such as ischaemic preconditioning. Here, recent advances in the understanding of how the JAK-STAT pathway orchestrates the response to cellular damage in the myocardium are discussed, along with the potential benefits and challenges in manipulating this pathway in cardiovascular therapy.
Collapse
Affiliation(s)
- Seán P Barry
- Medical Molecular Biology Unit, The Institute of Child Health, University College London, 30 Guilford Street, London, UK
| | | | | | | |
Collapse
|
184
|
Abstract
A clear picture of the dynamic relationship between the host immune system and cancer is emerging as the cells and molecules that participate in naturally occurring antitumour immune responses are being identified. The interferons (IFNs) - that is, the type I IFNs (IFNalpha and IFNbeta) and type II IFN (IFNgamma) - have emerged as central coordinators of tumour-immune-system interactions. Indeed, the decade-old finding that IFNgamma has a pivotal role in promoting antitumour responses became the focus for a renewed interest in the largely abandoned concept of cancer immunosurveillance. More recently, type I IFNs have been found to have distinct functions in this process. In this Review, we discuss the roles of the IFNs, not only in cancer immunosurveillance but also in the broader process of cancer immunoediting.
Collapse
Affiliation(s)
- Gavin P Dunn
- Washington University School of Medicine, Department of Pathology and Immunology, Box 8118, 660 South Euclid Avenue, St. Louis, Missouri 63110, USA
| | | | | |
Collapse
|
185
|
Giunta B, Obregon D, Hou H, Zeng J, Sun N, Nikolic V, Ehrhart J, Shytle D, Fernandez F, Tan J. EGCG mitigates neurotoxicity mediated by HIV-1 proteins gp120 and Tat in the presence of IFN-gamma: role of JAK/STAT1 signaling and implications for HIV-associated dementia. Brain Res 2006; 1123:216-225. [PMID: 17078933 PMCID: PMC4487677 DOI: 10.1016/j.brainres.2006.09.057] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2006] [Accepted: 09/12/2006] [Indexed: 11/19/2022]
Abstract
Human immunodeficiency virus (HIV)-1 infection of the central nervous system occurs in the vast majority of HIV-infected patients. HIV-associated dementia (HAD) represents the most severe form of HIV-related neuropsychiatric impairment and is associated with neuropathology involving HIV proteins and activation of proinflammatory cytokine circuits. Interferon-gamma (IFN-gamma) activates the JAK/STAT1 pathway, a key regulator of inflammatory and apoptotic signaling, and is elevated in HIV-1-infected brains progressing to HAD. Recent reports suggest green tea-derived (-)-epigallocatechin-3-gallate (EGCG) can attenuate neuronal damage mediated by this pathway in conditions such as brain ischemia. In order to investigate the therapeutic potential of EGCG to mitigate the neuronal damage characteristic of HAD, IFN-gamma was evaluated for its ability to enhance well-known neurotoxic properties of HIV-1 proteins gp120 and Tat in primary neurons and mice. Indeed, IFN-gamma enhanced the neurotoxicity of gp120 and Tat via increased JAK/STAT signaling. Additionally, primary neurons pretreated with a JAK1 inhibitor, or those derived from STAT1-deficient mice, were largely resistant to the IFN-gamma-enhanced neurotoxicity of gp120 and Tat. Moreover, EGCG treatment of primary neurons from normal mice reduced IFN-gamma-enhanced neurotoxicity of gp120 and Tat by inhibiting JAK/STAT1 pathway activation. EGCG was also found to mitigate the neurotoxic properties of HIV-1 proteins in the presence of IFN-gamma in vivo. Taken together, these data suggest EGCG attenuates the neurotoxicity of IFN-gamma augmented neuronal damage from HIV-1 proteins gp120 and Tat both in vitro and in vivo. Thus EGCG may represent a novel natural copound for the prevention and treatment of HAD.
Collapse
Affiliation(s)
- Brian Giunta
- Neuroimmunology Laboratory, Silver Child Development Center, Institute for Research in Psychiatry, Department of Psychiatry and Behavioral Medicine, University of South Florida, 3515 East Fletcher Avenue Tampa, Florida, 33613, USA
| | - Demian Obregon
- Neuroimmunology Laboratory, Silver Child Development Center, Institute for Research in Psychiatry, Department of Psychiatry and Behavioral Medicine, University of South Florida, 3515 East Fletcher Avenue Tampa, Florida, 33613, USA
| | - Hauyan Hou
- Neuroimmunology Laboratory, Silver Child Development Center, Institute for Research in Psychiatry, Department of Psychiatry and Behavioral Medicine, University of South Florida, 3515 East Fletcher Avenue Tampa, Florida, 33613, USA
| | | | - Nan Sun
- Neuroimmunology Laboratory, Silver Child Development Center, Institute for Research in Psychiatry, Department of Psychiatry and Behavioral Medicine, University of South Florida, 3515 East Fletcher Avenue Tampa, Florida, 33613, USA
| | - Veljko Nikolic
- Neuroimmunology Laboratory, Silver Child Development Center, Institute for Research in Psychiatry, Department of Psychiatry and Behavioral Medicine, University of South Florida, 3515 East Fletcher Avenue Tampa, Florida, 33613, USA
| | - Jared Ehrhart
- Neuroimmunology Laboratory, Silver Child Development Center, Institute for Research in Psychiatry, Department of Psychiatry and Behavioral Medicine, University of South Florida, 3515 East Fletcher Avenue Tampa, Florida, 33613, USA
| | - Douglas Shytle
- Neuroimmunology Laboratory, Silver Child Development Center, Institute for Research in Psychiatry, Department of Psychiatry and Behavioral Medicine, University of South Florida, 3515 East Fletcher Avenue Tampa, Florida, 33613, USA
- Center for Excellence in Aging and Brain Repair, Department of Neurosurgery; University of South Florida, 3515 East Fletcher Avenue Tampa, Florida, 33613, USA
| | - Francisco Fernandez
- Neuroimmunology Laboratory, Silver Child Development Center, Institute for Research in Psychiatry, Department of Psychiatry and Behavioral Medicine, University of South Florida, 3515 East Fletcher Avenue Tampa, Florida, 33613, USA
| | - Jun Tan
- Neuroimmunology Laboratory, Silver Child Development Center, Institute for Research in Psychiatry, Department of Psychiatry and Behavioral Medicine, University of South Florida, 3515 East Fletcher Avenue Tampa, Florida, 33613, USA
- Center for Excellence in Aging and Brain Repair, Department of Neurosurgery; University of South Florida, 3515 East Fletcher Avenue Tampa, Florida, 33613, USA
- Corresponding author: Jun Tan, M.D., Ph.D., Neuroimmunology Laboratory, Institute for Reseach in Psychiatry, Department of Psychiatry, College of Medicine University of South Florida. 3515 E Fletcher Ave. Tampa, FL 33613, USA. Phone: 813-974-9326; Fax: 813-974-1130;
| |
Collapse
|
186
|
Joshi VD, Kalvakolanu DV, Chen W, Zhang L, Kang TJ, Thomas KE, Vogel SN, Cross AS. A role for Stat1 in the regulation of lipopolysaccharide-induced interleukin-1beta expression. J Interferon Cytokine Res 2006; 26:739-47. [PMID: 17032168 DOI: 10.1089/jir.2006.26.739] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Because the induction of interleukin-1beta (IL-1beta) is critical to antibacterial host defenses and its excessive generation is a prominent component of sepsis, regulation of this proinflammatory cytokine is a critical factor in the immune response to lipopolysaccharide (LPS). We previously showed that LPS-induced IL-1beta expression was regulated by a Stat1-dependent, nitric oxide (NO)-mediated mechanism. Subsequent in vivo studies showed that whereas Stat1 had a role in the downregulation of IL-1beta expression, it had a more significant effect on its initial induction. Although both interferon-beta (IFN-beta) and IFN-gamma activate Stat1, the early appearance of IFN-beta in the circulation after LPS administration suggested its pivotal role in Stat1-mediated IL-1beta expression in vivo. Further in vitro analysis of peritoneal macrophages from IFN-beta (/), Stat1(/), and caspase-1(/) mice and their wild-type controls following LPS stimulation demonstrated that IL-1beta mRNA was expressed in these mice but not in macrophages from MyD88(/) mice. Despite the presence of IL-1beta mRNA, IL-1beta protein was markedly reduced in the absence of Stat1 activation in macrophages derived from IFN-beta (/) and Stat1(/) mice or in the absence of caspase-1 activity, which itself was dependent on Stat1 activation. These studies support the hypothesis that the expression of IL-1beta requires both the MyD88-dependent induction of IL-1beta mRNA and pro-IL-1beta as well as the MyD88-independent, Stat1-mediated processing of that gene product into active cytokine.
Collapse
Affiliation(s)
- Vishwas D Joshi
- Department of Medicine, Division of Geographic Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | | | | | | | | | | | |
Collapse
|
187
|
Frigg R, Wenzel A, Samardzija M, Oesch B, Wariwoda H, Navarini AA, Seeliger MW, Tanimoto N, Remé C, Grimm C. The prion protein is neuroprotective against retinal degeneration in vivo. Exp Eye Res 2006; 83:1350-8. [PMID: 16952355 DOI: 10.1016/j.exer.2006.07.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2006] [Revised: 07/06/2006] [Accepted: 07/14/2006] [Indexed: 01/14/2023]
Abstract
A common feature of neurodegenerative disorders is acute or progressive loss of neurons due to apoptosis. The pathological isoform of the prion protein is associated with retinal apoptosis and the cellular isoform (PrPc) has been shown to mediate protection from apoptosis in cell culture and in neonatal retinal explants. Using a model of light-induced photoreceptor apoptosis, we show in vivo that the levels of PrPc expression in the retina inversely correlate with the susceptibility of photoreceptors to light damage. Dissection of apoptotic signalling cascades suggests that PrPc acts neuroprotectively downstream of AP-1 induction. Our results reveal PrP as a neuroprotective/anti-apoptotic factor in vivo and suggest that PrPc may function as a guardian of neuronal integrity.
Collapse
Affiliation(s)
- Rico Frigg
- Lab for Retinal Cell Biology, Department of Ophthalmology, Center for Integrative Human Physiology (CIHP) and Neuroscience Center (ZNZ), University of Zurich, Frauenklinikstrasse 24, Zürich, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
188
|
Yang Y, Pan X, Lei W, Wang J, Shi J, Li F, Song J. Regulation of Transforming Growth Factor-β1–Induced Apoptosis and Epithelial-to-Mesenchymal Transition by Protein Kinase A and Signal Transducers and Activators of Transcription 3. Cancer Res 2006; 66:8617-24. [PMID: 16951175 DOI: 10.1158/0008-5472.can-06-1308] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Apoptosis and epithelial-to-mesenchymal transdifferentiation or transition (EMT) are crucial for normal development and body homeostasis. The alterations of these events are closely related to some pathologic processes, such as tumor formation and metastasis, fibrotic diseases of liver and kidney, and abnormal development of embryos. The mechanism that underlies the simultaneously occurring apoptosis and EMT induced by transforming growth factor-beta (TGF-beta) has not been well studied. In this report, we investigated the potential mechanism that underlies TGF-beta1-induced apoptosis and EMT. TGF-beta1-induced apoptosis and EMT were associated with the activation of protein kinase A (PKA) and signal transducers and activators of transcription 3 (STAT3). Inhibition of PKA by specific PKA inhibitor H89 or by PKA inhibitor peptide blocked STAT3 activation and suppressed TGF-beta1-induced apoptosis and EMT. Furthermore, overexpression of a phosphorylation-deficient form of STAT3, but not wild-type STAT3, produced an inhibitory effect on TGF-beta1-induced apoptosis and EMT. The results indicate that PKA is an upstream regulator for TGF-beta1-induced STAT3 activation and plays an important role in TGF-beta1-mediated apoptosis and EMT. These studies provided a new insight into the signaling mechanism underlying the apoptosis and EMT, which could be of importance in understanding some related physiologic and pathologic processes.
Collapse
Affiliation(s)
- Yanan Yang
- Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
189
|
Deng H, Wu R, Chen Y, Liu L. STAT1 and survivin expression in full lymph node examined gastric cancer by using tissue microarray technique. ACTA ACUST UNITED AC 2006. [DOI: 10.1007/s10330-006-0468-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
190
|
Salaun B, Coste I, Rissoan MC, Lebecque SJ, Renno T. TLR3 Can Directly Trigger Apoptosis in Human Cancer Cells. THE JOURNAL OF IMMUNOLOGY 2006; 176:4894-901. [PMID: 16585585 DOI: 10.4049/jimmunol.176.8.4894] [Citation(s) in RCA: 360] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
TLRs function as molecular sensors to detect pathogen-derived products and trigger protective responses ranging from secretion of cytokines that increase the resistance of infected cells and chemokines that recruit immune cells to cell death that limits microbe spreading. Viral dsRNA participate in virus-infected cell apoptosis, but the signaling pathway involved remains unclear. In this study we show that synthetic dsRNA induces apoptosis of human breast cancer cells in a TLR3-dependent manner, which involves the molecular adaptor Toll/IL-1R domain-containing adapter inducing IFN-beta and type I IFN autocrine signaling, but occurs independently of the dsRNA-activated kinase. Moreover, detailed molecular analysis of dsRNA-induced cell death established the proapoptotic role of IL-1R-associated kinase-4 and NF-kappaB downstream of TLR3 as well as the activation of the extrinsic caspases. The direct proapoptotic activity of endogenous human TLR3 expressed by cancerous cells reveals a novel aspect of the multiple-faced TLR biology, which may open new clinical prospects for using TLR3 agonists as cytotoxic agents in selected cancers.
Collapse
Affiliation(s)
- Bruno Salaun
- Laboratory for Immunological Research Schering-Plough Research Institute, Dardilly, France
| | | | | | | | | |
Collapse
|
191
|
Stehr W, Bernal NP, Bernabe KQ, Erwin CR, Warner BW. Absent STAT-1 expression perturbs adaptation and apoptosis after massive intestinal resection. J Pediatr Surg 2006; 41:713-8; discussion 713-8. [PMID: 16567182 DOI: 10.1016/j.jpedsurg.2005.12.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND We have previously established the significance of epidermal growth factor receptor (EGFR) activity and the cyclin-dependent kinase inhibitor p21waf1/cip1 (p21) for the adaptive response of the intestine to massive small bowel resection (SBR). In this study, we tested the role of the signal transducer and activator of transcription 1 (STAT-1) as this transcription factor is activated by the EGFR and known to induce p21 expression. METHODS Control (n = 40; C57/Bl6) and STAT-1-null mice (n = 40) underwent 50% proximal SBR or sham operation. After 3 days, the remnant ileum was harvested and the villus and crypt morphology was measured along with changes in rates of enterocyte proliferation and apoptosis. RESULTS The magnitude of resection-induced adaptation was greater in STAT-1-null animals as verified by taller villi and deeper crypts. The expected increase in enterocyte apoptosis did not occur after SBR in the background of STAT-1 deficiency. Western blotting revealed elevated expression of p21 protein in both STAT-1-null and controls after SBR. CONCLUSION Increased p21 expression after SBR in the absence of STAT-1 suggests an alternate mechanism for resection-induced regulation of p21. Enhanced adaptation in STAT-1-null animals suggests that this transcription factor serves an inhibitor to the process of adaptation, perhaps via regulation of enterocyte apoptosis.
Collapse
Affiliation(s)
- Wolfgang Stehr
- Division of Pediatric General and Thoracic Surgery, Department of Surgery, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229-3039, USA
| | | | | | | | | |
Collapse
|
192
|
Tang L, Roberts PC, Kraniak JM, Li Q, Tainsky MA. Stat1 expression is not sufficient to regulate the interferon signaling pathway in cellular immortalization. J Interferon Cytokine Res 2006; 26:14-26. [PMID: 16426144 DOI: 10.1089/jir.2006.26.14] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
DNA hypermethylation in gene promoters is an epigenetic mechanism regulating gene expression in cellular immortalization, an important step in carcinogenesis. Previously, we studied the genes dysregulated during immortalization using spontaneously immortalized fibroblasts from patients with Li-Fraumeni syndrome (LFS), who carry a germline mutation in the tumor suppressor gene p53. We found that multiple interferon (IFN) signaling pathway genes were regulated by epigenetic silencing. In this study we focused on a key regulator of that pathway, the signal transducer and transcription activator 1 (Stat1) gene. Although Stat1 is downregulated after cellular immortalization and upregulated in immortal MDAH041 cells after 5-aza-2'-deoxycytidine (5-aza-dC) treatment, we detected no methylation of the Stat1 promoter region in these cells before or after immortalization. To analyze the function of Stat1 in immortalization, we expressed Stat1 in immortal MDAH041 cells by stable infection, expecting to induce IFN-regulated genes or cellular senescence or both. However, the overexpression of Stat1 alone was not sufficient to repress the proliferation rate of immortal MDAH041 cells or induce senescence in immortal MDAH041 cells. We concluded that factor(s) additional to Stat1 (whether IFN dependent or not) are required for the immortalization of LFS fibroblasts.
Collapse
Affiliation(s)
- Lin Tang
- Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | | | | | | | | |
Collapse
|
193
|
Yan X, Orentas RJ, Johnson BD. Tumor-derived macrophage migration inhibitory factor (MIF) inhibits T lymphocyte activation. Cytokine 2006; 33:188-98. [PMID: 16522371 PMCID: PMC2018658 DOI: 10.1016/j.cyto.2006.01.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2005] [Revised: 12/05/2005] [Accepted: 01/13/2006] [Indexed: 11/20/2022]
Abstract
Macrophage migration inhibitory factor (MIF) is a multi-functional cytokine that is considered a pro-inflammatory cytokine. However, our studies show that MIF, when produced in super-physiological levels by a murine neuroblastoma cell line (Neuro-2a) exceeding those normally seen during an immune response, inhibits cytokine-, CD3-, and allo-induced T-cell activation. MIF is also able to inhibit T cells that have already received an activation signal. The T-cell inhibitory effects of culture supernatants from neuroblastoma cells were reversed when the cells were transfected with dicer-generated si-RNA to MIF. When T cells were activated in vitro by co-culture with interleukin (IL)-2 and IL-15 and analyzed for cytokine production in the presence or absence of MIF-containing culture supernatant, inhibition of T-cell proliferation and induced cell death were observed even as the treated T cells produced high levels of interferon-gamma (IFN-gamma). The inhibitory effects of MIF were partially reversed when lymphocytes from IFN-gamma knockout mice were tested. We propose that the high levels of MIF produced by neuroblastoma cause activation induced T-cell death through an IFN-gamma pathway and may eliminate activated T cells from the tumor microenvironment and thus contribute to escape from immune surveillance.
Collapse
Affiliation(s)
- Xiaocai Yan
- Department of Pediatrics, Section of Hematology-Oncology, MFRC 6011, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | | | | |
Collapse
|
194
|
Xi S, Dyer KF, Kimak M, Zhang Q, Gooding WE, Chaillet JR, Chai RL, Ferrell RE, Zamboni B, Hunt J, Grandis JR. Decreased STAT1 expression by promoter methylation in squamous cell carcinogenesis. J Natl Cancer Inst 2006; 98:181-9. [PMID: 16449678 DOI: 10.1093/jnci/djj020] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Dysregulation of signal transducers and activators of transcription (STATs) is associated with many cancers, but no role of STAT1 in human tumor progression has been demonstrated. METHODS We compared STAT1 protein expression in squamous cell carcinoma of the head and neck (SCCHN) tumors (n = 28) and normal oropharyngeal mucosa samples (n = 10) from patients without cancer as assessed by immunoblotting. Stable clones were established from SCCHN 1483 cells that were transfected with a STAT1 expression construct; cell growth and cisplatin-induced apoptosis of the clones and vector control-transfected 1483 cells were compared using trypan blue exclusion and Annexin V staining and expression of the cyclin-dependent kinase inhibitor p21 was assayed by immunoblotting. The growth of STAT1-overexpressing SCCHN 1483 xenograft tumors was compared with that of xenograft tumors derived from cells transfected with vector control DNA. DNA from SCCHN tumors (n = 16) and paired peripheral blood lymphocytes were analyzed for STAT1 mutations and promoter methylation using methylation-specific polymerase chain reaction and bisulfite sequencing. SCCHN cell lines (PCI-15b, 1483, and UM-22B) were treated with the demethylating agent azacytidine alone or in combination with the cytotoxic drug cisplatin, and expression of STAT1 and p21 were monitored by immunoblotting. All statistical tests were two-sided. RESULTS STAT1 levels were statistically significantly lower in the SCCHN tumors than normal mucosa (median = 0.8 relative units versus 2.4, difference = 1.6, 95% confidence interval [CI] = 1.3 to 2.0, P < .001). Overexpression of STAT1 abrogated the growth of SCCHN cells and xenograft tumors and increased p21 expression. STAT1 expression levels of the tumors with STAT1 promoter methylation (n = 12) were lower than those of tumors (n = 4) without promoter methylation of STAT1 (P = .008). Azacytidine treatment increased expression of STAT1 and p21 in SCCHN cell lines and increased apoptosis in cisplatin-treated 1483 cells compared with cisplatin treatment alone (mean = 61.3% versus 25.8%, difference = 35.5%, 95% CI = 24.5% to 43.4%; P = .028). CONCLUSION STAT1 can function as a tumor suppressor in SCCHN cells. Silencing of the STAT1 gene via promoter methylation may contribute to SCCHN tumor cell growth.
Collapse
Affiliation(s)
- Sichuan Xi
- Department of Otolaryngology, University of Pittsburgh School of Medicine, Cancer Institute, Pittsburgh, PA, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
195
|
Lee H, Yu H. Methylation of Stat1 promoter can contribute to squamous cell carcinogenesis. J Natl Cancer Inst 2006; 98:154-5. [PMID: 16449669 DOI: 10.1093/jnci/djj038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
196
|
Sabbagh L, Bourbonnière M, Denis F, Sékaly RP. Cloning and Functional Characterization of the Murine Caspase-3 Gene Promoter. DNA Cell Biol 2006; 25:104-15. [PMID: 16460234 DOI: 10.1089/dna.2006.25.104] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Several studies have shown that the levels of caspase-3 are upregulated under different conditions of apoptosis. Previously, we have shown that activation of T cells through the TCR leads to the upregulation of caspase-3 levels. These findings highlight the importance of regulating the expression of caspase-3 in order to prevent premature cell death. To better understand the regulation of the caspase-3 gene, a portion of the 5'- untranslated region was cloned, sequenced, and characterized. The segment of the 5'-flanking region of the caspase-3 gene was also cloned upstream of a luciferase reporter gene, demonstrating that this fragment contains promoter activity. Higher luciferase expression was found with several of the promoter deletion constructs in Jurkat T cells but not the mouse Neuro-2A neuroblastoma cell line, suggesting the presence of a T-cell-specific regulated region. The importance of these sequences is further supported by the genomic organization of the human and mouse caspase-3 promoter regions. These findings demonstrated that the -2245/+14 region of the caspase-3 promoter shows constitutive levels of expression, and that several regions of the promoter play a role in basal regulation. Finally, some of the conserved transcription factor binding sites identified between the human and mouse promoters appear to play an important role in lymphoid cells.
Collapse
Affiliation(s)
- Laurent Sabbagh
- Laboratoire d'Immunologie, Centre de Recherche du CHUM, Campus St.-Luc, Pavillon Edouard-Asselin, 264 Boulevard René Lévesque Est, #1370D, Montreal, Quebec H3C 3J7, Canada
| | | | | | | |
Collapse
|
197
|
Yu S, Sharp GC, Braley-Mullen H. Thyrocytes responding to IFN-gamma are essential for development of lymphocytic spontaneous autoimmune thyroiditis and inhibition of thyrocyte hyperplasia. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2006; 176:1259-65. [PMID: 16394017 DOI: 10.4049/jimmunol.176.2.1259] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
IFN-gamma promotes the development of lymphocytic spontaneous autoimmune thyroiditis (L-SAT) in NOD.H-2h4 mice and inhibits the development of thyrocyte hyperplasia and proliferation (TEC H/P). The precise mechanisms by which IFN-gamma promotes L-SAT and inhibits TEC H/P are unknown. To determine whether responsiveness of lymphocytes or thyrocytes to IFN-gamma is important for the development of these lesions, IFN-gammaR-/- mice, which develop TEC H/P similar to IFN-gamma-/- mice, were used as recipients for adoptive cell transfer. Wild-type (WT) splenocytes or bone marrow induced L-SAT and inhibited TEC H/P in IFN-gamma-/-, but not IFN-gammaR-/- recipients. IFN-gammaR-/- recipients of WT cells developed severe TEC H/P, but did not develop L-SAT, suggesting that thyrocytes responding to IFN-gamma are important for inhibition of TEC H/P. Unexpectedly, IFN-gammaR-/- splenocytes or bone marrow did not induce L-SAT in IFN-gamma-/- or WT mice even though IFN-gammaR-/- lymphocyte donors produced as much IFN-gamma as lymphocytes from WT donors, and thyrocytes could respond to IFN-gamma. Real-time PCR indicated that recipients of IFN-gammaR-/- bone marrow expressed less mRNA for IFN-gamma-inducible chemokines compared with recipients of WT bone marrow. This might limit the migration of IFN-gammaR-/- lymphocytes to thyroids. Few IFN-gammaR-/- lymphocytes infiltrated thyroids even in the presence of WT lymphocytes, suggesting that lymphocytes unable to respond to IFN-gamma are not induced to migrate to thyroids. These results suggest that thyrocytes must be able to respond to IFN-gamma for the development of L-SAT and inhibition of TEC H/P, and lymphocytes must be able to respond to IFN-gamma to induce L-SAT.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Base Sequence
- Chemokines/genetics
- Hyperplasia
- Interferon-gamma/deficiency
- Interferon-gamma/genetics
- Interferon-gamma/physiology
- Lymphocytes/immunology
- Lymphocytes/pathology
- Male
- Mice
- Mice, Inbred NOD
- Mice, Knockout
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Interferon/deficiency
- Receptors, Interferon/genetics
- Receptors, Interferon/physiology
- Thyroid Gland/immunology
- Thyroid Gland/pathology
- Thyroiditis, Autoimmune/etiology
- Thyroiditis, Autoimmune/immunology
- Thyroiditis, Autoimmune/pathology
- Interferon gamma Receptor
Collapse
Affiliation(s)
- Shiguang Yu
- Department of Internal Medicine, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | | | | |
Collapse
|
198
|
Dudley AC, Thomas D, Best J, Jenkins A. A VEGF/JAK2/STAT5 axis may partially mediate endothelial cell tolerance to hypoxia. Biochem J 2006; 390:427-36. [PMID: 15918795 PMCID: PMC1198922 DOI: 10.1042/bj20050351] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Perturbation of oxygen flow occurs in disease states such as diabetic retinopathy and cancer. To maintain oxygen homoeostasis, the mammalian microvascular endothelium undergoes a dramatic reorganization to assist in bringing oxygen and nutrients to oxygen-starved tissues. This process is termed angiogenesis and is common in certain cancers with hypoxic foci and in areas of focal ischaemia in the diabetic retina. In the present study, we report on the activation of the JAK2/STAT5 pathway (where JAK stands for Janus kinase and STAT stands for signal transduction and activator of transcription) by low oxygen in microvascular endothelial cells. This activation appears to occur downstream of VEGF (vascular endothelial growth factor), a well-known proangiogenic factor, and is related to repression of proapoptotic FAS(CD95)/FASL(CD95L). These results indicate that the JAK/STAT pathway may play a pivotal role during tumour-associated or retinal angiogenesis in which endothelial cell survival during tissue hypoxia is critical for maintaining either the growth of neoplasms or the inappropriate retinal neovascularization common in diabetic retinopathy.
Collapse
Affiliation(s)
- Andrew C Dudley
- Department of Medicine, St. Vincent's Hospital, University of Melbourne, Corner of Princes and Regent Streets, Fitzroy, VIC, 3065, Australia.
| | | | | | | |
Collapse
|
199
|
Smyth MJ, Dunn GP, Schreiber RD. Cancer Immunosurveillance and Immunoediting: The Roles of Immunity in Suppressing Tumor Development and Shaping Tumor Immunogenicity. Adv Immunol 2006; 90:1-50. [PMID: 16730260 DOI: 10.1016/s0065-2776(06)90001-7] [Citation(s) in RCA: 576] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cellular transformation and tumor development result from an accumulation of mutational and epigenetic changes that alter normal cell growth and survival pathways. For the last 100 years, there has been a vigorous debate as to whether the unmanipulated immune system can detect and eliminate such altered host derived cells despite the fact that cancer cells frequently express either abnormal proteins or abnormal levels of normal cellular proteins that function as tumor antigens. In this review, we discuss the current state of this argument and point out some of the recent key experiments demonstrating that immunity not only protects the host from cancer development (i.e., provides a cancer immunosurveillance function) but also can promote tumor growth, sometimes by generating more aggressive tumors. The terminology "cancer immunoediting" has been used to describe this dual host protective and tumor promoting action of immunity, and herein we summarize the ever-increasing experimental and clinical data that support the validity of this concept.
Collapse
Affiliation(s)
- Mark J Smyth
- Cancer Immunology Program, Peter MacCallum Cancer Centre, East Melbourne, 3002 Victoria, Australia
| | | | | |
Collapse
|
200
|
Krause CD, He W, Kotenko S, Pestka S. Modulation of the activation of Stat1 by the interferon-gamma receptor complex. Cell Res 2006; 16:113-23. [PMID: 16467883 DOI: 10.1038/sj.cr.7310015] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The activation of Stat1 by the interferon-gamma (IFN-gamma) receptor complex is responsible for the transcription of a significant portion of IFN-gamma induced genes. Many of these genes are responsible for the induction of an apoptotic state in response to IFN-gamma. In the absence of Stat1 activation, IFN-gamma instead induces a proliferative response. Modifying Stat1 activation by IFN-gamma may have pharmacological benefits. We report that the rate of activation of Stat1 can be altered in HeLa cells by overexpressing either the IFN-gammaR1 chain or the IFN-gammaR2 chain. These alterations occur in hematopoietic cell lines: Raji cells and monocytic cell lines, which have average and above-average IFN-gammaR2 surface expression, activate Stat1 similarly to HeLa cells and HeLa cells overexpressing IFNgammaR2, respectively. The rapid Stat1 activation seen in HeLa cells can be inhibited by overexpressing a chimeric IFN-gammaR2 chain that does not bind Jak2 or (when high concentrations of IFN-gamma are used) by overexpressing IFN-gammaR1. These data are consistent with a model in which the recruitment of additional Jak2 activity to a signaling complex accelerates the rate of Stat1 activation. We conclude that the rate of activation of Stat1 in cells by IFN-gamma can be modified by regulating either receptor chain and speculate that pharmacological agents which modify receptor chain expression may alter IFN-gamma receptor signal transduction.
Collapse
Affiliation(s)
- Christopher D Krause
- Department of Molecular Genetics, Microbiology and Immunology, Robert Wood Johnson Medical School, The University of Medicine and Dentistry of New Jersey, 675 Hoes Lane West, Piscataway, 08855, USA
| | | | | | | |
Collapse
|