1
|
Isayev O, Sokolova DV, Anisimova NY, Spirina TS, Gasimov E. Investigation of an anticancer activity of combination of interferon-alpha and gemcitabine on pancreatic cancer cells. J Cancer Res Ther 2024; 20:1494-1498. [PMID: 39412913 DOI: 10.4103/jcrt.jcrt_138_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 03/11/2023] [Indexed: 10/18/2024]
Abstract
ABSTRACT Pancreatic ductal adenocarcinoma (PDAC) is still a clinical challenge due to its deteriorated prognosis. Therefore, new combination chemotherapeutic approaches are of research interest. In this work, we attempted to characterize the effects of gemcitabine and interferon-alpha as well as the combination of both on the metabolic, pro-apoptotic, and proliferative activity of MiaPaca and Panc-1 cells. We showed that the exposure of both drugs in combination increases effectively the metabolic activity of cells of MiaPaca and Panc-1 cell lines compared to the monotherapies. Based on the data from the analysis of apoptosis, the underlying molecular effect of metabolic and proliferative inhibition is an increase in the number of cells in the early apoptosis. These data can be of interest in the context of future preclinical research.
Collapse
Affiliation(s)
- Orkhan Isayev
- Department of Histology, Embryology and Cytology, Azerbaijan Medical University, Baku, Azerbaijan
- Genetic Resources Institute, Azerbaijan National Academy of Sciences, Baku, Azerbaijan
| | - Darina V Sokolova
- N.N. Blokin National Medical Research Center of Oncology, Moscow, Russia
- People's Friendship University, Moscow, Russia
| | | | - Tatiana S Spirina
- N.N. Blokin National Medical Research Center of Oncology, Moscow, Russia
| | - Eldar Gasimov
- Department of Histology, Embryology and Cytology, Azerbaijan Medical University, Baku, Azerbaijan
| |
Collapse
|
2
|
Ding H, Wang G, Yu Z, Sun H, Wang L. Role of interferon-gamma (IFN-γ) and IFN-γ receptor 1/2 (IFNγR1/2) in regulation of immunity, infection, and cancer development: IFN-γ-dependent or independent pathway. Biomed Pharmacother 2022; 155:113683. [PMID: 36095965 DOI: 10.1016/j.biopha.2022.113683] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/27/2022] [Accepted: 09/07/2022] [Indexed: 11/02/2022] Open
Abstract
IFN-γ, a soluble cytokine being produced by T lymphocytes, macrophages, mucosal epithelial cells, or natural killer cells, is able to bind to the IFN-γ receptor (IFNγR) and in turn activate the Janus kinase (JAK)-signal transducer and transcription protein (STAT) pathway and induce expression of IFN-γ-stimulated genes. IFN-γ is critical for innate and adaptive immunity and aberrant IFN-γ expression and functions have been associated with different human diseases. However, the IFN-γ/IFNγR signaling could be a double-edged sword in cancer development because the tissue microenvironments could determine its anti- or pro-tumorigenic activities. The IFNγR protein consists of two IFNγR1 and IFNγR2 chains, subunits of which play different roles under certain conditions. This review assessed IFNγR polymorphisms, expression and functions in development and progression of various human diseases in an IFN-γ-dependent or independent manner. This review also discussed tumor microenvironment, microbial infection, and vital molecules in the IFN-γ upstream signaling that might regulate IFNγR expression, drug resistance, and druggable strategy, to provide evidence for further application of IFNγR.
Collapse
Affiliation(s)
- Huihui Ding
- School of Pharmacy, Shandong First Medical University, Jinan, Shandong, China.
| | - Gongfu Wang
- Center for Drug Evaluation, China Food and Drug Administration (CFDA), Beijing, China.
| | - Zhen Yu
- Department of Pharmacy, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| | - Huimin Sun
- School of Pharmacy, Shandong First Medical University, Jinan, Shandong, China.
| | - Lu Wang
- School of Pharmacy, Shandong First Medical University, Jinan, Shandong, China; Department of Pharmacy, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| |
Collapse
|
3
|
Booy S, Hofland L, van Eijck C. Potentials of interferon therapy in the treatment of pancreatic cancer. J Interferon Cytokine Res 2014; 35:327-39. [PMID: 25551196 DOI: 10.1089/jir.2014.0157] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Pancreatic cancer is a highly aggressive malignancy with limited treatment options. To improve survival for patients with pancreatic cancer, research has focused on other treatment modalities like adding biological modulators such as type-I interferons (IFNs). Type I IFNs (ie, IFN-α/IFN-β) have antiproliferative, antiviral, and immunoregulatory activities. Furthermore, they are able to induce apoptosis, exert cell cycle blocking, and sensitize tumor cells for chemo- and radiotherapy. A few years ago in vitro, in vivo, and several clinical trials have been described regarding adjuvant IFN-α therapy in the treatment of pancreatic cancer. Some studies reported a remarkable increase in the 2- and 5-year survival. Unfortunately, the only randomized clinical trial did not show a significant increase in overall survival, although the increased median survival implicated that some patients in the experimental group benefited from the adjuvant IFN-α therapy. Furthermore, encouraging in vitro and in vivo data points to a possible role for adjuvant IFN therapy. However, up till now, the use of IFNs in the treatment of pancreatic cancer remains controversial. This review, therefore, aims to describe, based on the available data, whether there is a distinct role for IFN therapy in the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Stephanie Booy
- 1 Department of Surgery, Erasmus Medical Centre , Rotterdam, The Netherlands
| | | | | |
Collapse
|
4
|
Booy S, van Eijck CHJ, Dogan F, van Koetsveld PM, Hofland LJ. Influence of type-I Interferon receptor expression level on the response to type-I Interferons in human pancreatic cancer cells. J Cell Mol Med 2014; 18:492-502. [PMID: 24460759 PMCID: PMC3955155 DOI: 10.1111/jcmm.12200] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Accepted: 10/17/2013] [Indexed: 12/14/2022] Open
Abstract
Pancreatic cancer is a highly aggressive malignancy with limited treatment options. Type-I interferons (e.g. IFN-α/-β) have several anti-tumour activities. Over the past few years, clinical studies evaluating the effect of adjuvant IFN-α therapy in pancreatic cancer yielded equivocal results. Although IFN-α and-β act via the type-I IFN receptor, the role of the number of receptors present on tumour cells is still unknown. Therefore, this study associated, for the first time, in a large panel of pancreatic cancer cell lines the effects of IFN-α/-β with the expression of type-I IFN receptors. The anti-tumour effects of IFN-α or IFN-β on cell proliferation and apoptosis were evaluated in 11 human pancreatic cell lines. Type-I IFN receptor expression was determined on both the mRNA and protein level. After 7 days of incubation, IFN-α significantly reduced cell growth in eight cell lines by 5–67%. IFN-β inhibited cell growth statistically significant in all cell lines by 43–100%. After 3 days of treatment, IFN-β induced significantly more apoptosis than IFN-α. The cell lines variably expressed the type-I IFN receptor. The maximal inhibitory effect of IFN-α was positively correlated with the IFNAR-1 mRNA (P < 0.05, r = 0.63), IFNAR-2c mRNA (P < 0.05, r = 0.69) and protein expression (P < 0.05, r = 0.65). Human pancreatic cancer cell lines variably respond to IFN-α and-β. The expression level of the type-I IFN receptor is of predictive value for the direct anti-tumour effects of IFN-α treatment. More importantly, IFN-β induces anti-tumour effects already at much lower concentrations, is less dependent on interferon receptor expression and seems, therefore, more promising than IFN-α.
Collapse
Affiliation(s)
- Stephanie Booy
- Department of Internal Medicine, Division of Endocrinology, Erasmus Medical Center, Rotterdam, The Netherland; Department of Surgery, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | | | | | | |
Collapse
|
5
|
Kim DJ, Yi BR, Lee HR, Kim SU, Choi KC. Pancreatic tumor mass in a xenograft mouse model is decreased by treatment with therapeutic stem cells following introduction of therapeutic genes. Oncol Rep 2013; 30:1129-36. [PMID: 23807450 DOI: 10.3892/or.2013.2564] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 04/22/2013] [Indexed: 11/06/2022] Open
Abstract
Pancreatic cancer is the fourth most common cause of cancer-related mortality. In the present study, we employed 2 types of therapeutic stem cells expressing cytosine deaminase (CD) with or without human interferon-β (IFN‑β), HB1.F3.CD and HB1.F3.CD.IFN-β cells, respectively, to selectively treat pancreatic cancer. The CD gene converts the non-toxic prodrug, 5-flurorocytosine (5-FC), into the toxic agent, 5-fluorouracil (5-FU). In addition, human IFN-β is a potent cytokine that has antitumor effects. To generate a xenograft mouse model, PANC-1 cells (2x10(6)/mouse) cultured in DMEM containing 10% FBS were mixed with Matrigel and were subcutaneously injected into Balb/c nu/nu mice. In the migration assay, the stem cells expressing the CD or IFN-β gene effectively migrated toward the pancreatic cancer cells, suggesting the presence of chemoattractant factors secreted by the pancreatic tumors. In the co-culture and MTT assay, antitumor activity of the therapeutic stem cells was observed in the presence of 5-FC was shown that the growth of PANC-1 cells was inhibited. Furthermore, these effects were confirmed in the xenograft mouse model bearing tumors originating from PANC-1 cells. Analyses by histological and fluorescence microscopy showed that treatment with the stem cells resulted in the inhibition of pancreatic cancer growth in the presence of 5-FC. Taken together, these results indicate that stem cells expressing the CD and/or IFN-β gene can be used to effectively treat pancreatic cancer and reduce the side-effects associated with conventional therapies.
Collapse
Affiliation(s)
- Doo-Jin Kim
- Laboratory of Veterinary Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | | | | | | | | |
Collapse
|
6
|
Dembinski JL, Wilson SM, Spaeth EL, Studeny M, Zompetta C, Samudio I, Roby K, Andreeff M, Marini FC. Tumor stroma engraftment of gene-modified mesenchymal stem cells as anti-tumor therapy against ovarian cancer. Cytotherapy 2013; 15:20-32. [PMID: 23260083 PMCID: PMC3709838 DOI: 10.1016/j.jcyt.2012.10.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Accepted: 07/19/2012] [Indexed: 12/31/2022]
Abstract
BACKGROUND AIMS Many ovarian cancers originate from ovarian surface epithelium, where they develop from cysts intermixed with stroma. The stromal layer is critical to the progression and survival of the neoplasm and consequently is recruited into the tumor microenvironment. METHODS Using both syngeneic mouse tumors (ID8-R) and human xenograft (OVCAR3, SKOV3) tumor models, we first confirmed that intraperitoneally injected circulating mesenchymal stem cells (MSCs) could target, preferentially engraft and differentiate into α-smooth muscle actin-positive myofibroblasts, suggesting their role as "reactive stroma" in ovarian carcinoma development and confirming their potential as a targeted delivery vehicle for the intratumoral production of interferon-β (IFN-β). Mice with ovarian carcinomas then received weekly intraperitoneal injections of IFN-β expressing MSCs. RESULTS Intraperitoneal injections of IFN-β expressing MSCs resulted in complete eradication of tumors in 70% of treated OVCAR3 mice (P = 0.004) and an increased survival of treated SKOV3 mice compared with controls (P = 0.01). Similar tumor growth control was observed using murine IFN-β delivered by murine MSCs in ID8-R ovarian carcinoma. As a potential mechanism of tumor killing, MSCs produced IFN-β-induced caspase-dependent tumor cell apoptosis. CONCLUSIONS Our results demonstrate that ovarian carcinoma engrafts MSCs to participate in myofibrovascular networks and that IFN-β produced by MSCs intratumorally modulates tumor kinetics, resulting in prolonged survival.
Collapse
Affiliation(s)
- Jennifer L. Dembinski
- Department of Leukemia, The University of Texas M. D. Anderson Cancer Center, Houston, TX USA
| | - Shanna M. Wilson
- Department of Leukemia, The University of Texas M. D. Anderson Cancer Center, Houston, TX USA
| | - Erika L. Spaeth
- Department of Leukemia, The University of Texas M. D. Anderson Cancer Center, Houston, TX USA
| | - Matus Studeny
- Department of Leukemia, The University of Texas M. D. Anderson Cancer Center, Houston, TX USA
| | - Claudia Zompetta
- Department of Leukemia, The University of Texas M. D. Anderson Cancer Center, Houston, TX USA
- Experimental Medicine and Pathology, University of Rome La Sapienza, 00185 Rome, Italy
| | - Ismael Samudio
- Department of Leukemia, The University of Texas M. D. Anderson Cancer Center, Houston, TX USA
- Cellular and Molecular Therapy Group, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Katherine Roby
- Center for Reproductive Sciences and Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City 66160, USA
| | - Michael Andreeff
- Department of Leukemia, The University of Texas M. D. Anderson Cancer Center, Houston, TX USA
| | - Frank C. Marini
- Department of Leukemia, The University of Texas M. D. Anderson Cancer Center, Houston, TX USA
| |
Collapse
|
7
|
Zeng J, Xiong Y, Zhang LB, Ruan JD, Wu W, Sun WJ. The antitumor activity of recombinant antitumor antiviral protein and the associated molecular mechanism in pancreatic tumor cells. Cancer Biother Radiopharm 2012; 27:426-33. [PMID: 22783925 DOI: 10.1089/cbr.2012.1166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Pancreatic cancer has poor prognosis and high mortality. Currently, the therapy of pancreatic cancer remains a challenge. In this study, we compared the antitumor activity of the recombinant antitumor antiviral protein (RAAP), an improved interferon, with gemcitabine, a classic chemotherapy agent used for pancreatic cancer treatment. METHODS The proliferation of Bx-PC3 pancreatic cancer cells was evaluated by an MTT assay. Cell cycle arrest and apoptosis were evaluated by flow cytometry and annexin V-FITC/propidium iodide double staining, respectively. The expressions of matrix metalloproteinase (MMP)-2, MMP-9, caspase-3, caspase-8, and caspase-9 genes were evaluated by reverse transcription-polymerase chain reaction and the Western blot analysis. A xenograft pancreatic cancer model was established by inoculating Bx-PC3 cells into athymic nude mice. The antitumor activity of RAAP and gemcitabine was tested in the xenograft tumor model. RESULTS RAAP significantly inhibited proliferation, induced cell cycle arrest, and induced apoptosis in Bx-PC3 cells in vitro and delayed tumor growth in vivo. The antitumor activity of 20 ng/mL of RAAP was a little more effective than 10 μM of gemcitabine. The antitumor activity of RAAP was associated with its role in inducing caspase-3 and caspase-8 expression as well as downregulating MMP-2 expression. CONCLUSIONS RAAP can effectively suppress human pancreatic cancer cell growth in vitro and in vivo. The antitumor efficacy of RAAP is similar to gemcitabine.
Collapse
Affiliation(s)
- Jie Zeng
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha 410008, P.R. China
| | | | | | | | | | | |
Collapse
|
8
|
Saidi RF, Ahad A, Tilak J, Nalbantoglu I, Jacobs MJ. The expression of interferon receptor α/β in human pancreatic cancer in nude mice is essential for tumor response to interferon α treatment. J Surg Res 2010; 173:309-13. [PMID: 21067774 DOI: 10.1016/j.jss.2010.09.044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2010] [Revised: 09/17/2010] [Accepted: 09/29/2010] [Indexed: 11/19/2022]
Abstract
BACKGROUND Adjuvant interferon based chemoradiation has rendered promising results against pancreatic cancer. This study evaluated the in vivo effect of interferon α on two human pancreatic carcinoma cell lines implanted in nude. MATERIAL AND METHODS MiaPaCa-2 expressed the interferon α/β receptor and Panc-1 cells did not. Regimen I consisted of intraperitoneal single-agent gemcitabine and Regimen II consisted of IFN-α and gemcitabine biweekly for 30 d. RESULTS Regimen I and II significantly decreased median tumor volume compared with control mice (P < 0.001). However, MiaPaCa-2 showed a more dramatic response to Regimen II compared with Panc-1 implanted mice. MiaPaCa-2 and treated with Regimen II showed less metastasis and less local invasion compared with Panc-1 treated with same regimen. Regimen II was more effective on MiaPaCa-2 compared with Regimen I (P < 0.001). There were no differences between Regimens I and II in the Panc-1 group. CONCLUSIONS Treatment of human pancreatic cancer in nude mice with interferon α and gemcitabine was associated with a reduction in tumor volume. This process was more prominent in the cells that express the interferon receptors.
Collapse
Affiliation(s)
- Reza F Saidi
- Department of Surgery, University of Massachusetts Medical School, Worcester, Massachusetts S6-426, USA.
| | | | | | | | | |
Collapse
|
9
|
Jöst E, Roos WP, Kaina B, Schmidberger H. Response of pancreatic cancer cells treated with interferon-alpha or beta and co-exposed to ionising radiation. Int J Radiat Biol 2010; 86:732-41. [PMID: 20586542 DOI: 10.3109/09553002.2010.481321] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
PURPOSE Clinical trials on pancreatic cancer demonstrated that interferons (IFN) improve the therapeutic index of combined radio- and chemotherapy. This is believed to be due to radiosensitisation of cells, which, however, needs experimental verification. MATERIALS AND METHODS Here, we compared the survival response of ten pancreatic tumour cell lines following ionising radiation (IR), interferon-alpha (IFN-alpha), interferon-beta (IFN-beta) and combined treatment. The effect of combination treatment on apoptosis induction was also determined. RESULTS In most cell lines IFN treatment on its own exerted cytotoxicity, which was independent of the expression level of the IFN receptor on the cell surface. Three cell lines showed a radiosensitisation effect while two showed radioprotection. Although IFN-alpha is commonly used in the clinic, IFN-beta induced a stronger cytotoxic response than IFN-alpha in vitro. The likely mechanism of enhancement of radiosensitivity in the responsive cell lines was shown to be an increase of the radiation-induced apoptotic response by IFN pretreatment. CONCLUSIONS Given that the in vitro data do not conform to the impressive clinical results observed after combined radio- and chemotherapy with IFN-alpha, it is reasonable to conclude that the sensitising effect of IFN is not mediated through modulating the intrinsic radiosensitivity of pancreatic cancer cells.
Collapse
Affiliation(s)
- Eva Jöst
- Institute of Toxicology, University Medicine Mainz, Mainz, Germany
| | | | | | | |
Collapse
|
10
|
Abstract
Pancreatic cancer (PC) remains a life-threatening disease. Efficient therapeutic gene delivery to PC-derived cells continues to present challenges. We used self-inactivated lentiviral vectors to transduce PC-derived cells in vitro and in vivo. We showed that lentiviral vectors transduce PC-derived cell lines with high efficiency (>90%), regardless of the differentiation state of the cell. Next, we transferred human interferon beta (hIFN-beta) gene. Expression of hIFN-beta in PC cells using lentiviral vectors resulted in the inhibition of cell proliferation and the induction of cell death by apoptosis. In vivo, lentiviral administration of hIFN-beta prevented PC tumor progression for up to 15 days following gene therapy, and induced tumor regression/stabilization in 50% of the mice treated. Again, hIFN-beta expression resulted in cancer cell proliferation inhibition and apoptosis induction. We provide evidence that human immunodeficiency virus (HIV)-1-based lentiviral vectors are very efficient for gene transfer in PC-derived cells in vitro and in vivo. As a consequence, delivery of hIFN-beta stopped PC tumor progression. Thus, our approach could be applied to the 85% of PC patients with a locally advanced disease.
Collapse
|
11
|
Adjuvant Interferon-Based Chemoradiation Followed by Gemcitabine for Resected Pancreatic Adenocarcinoma. Ann Surg 2009; 249:865; author reply 865-6. [DOI: 10.1097/sla.0b013e3181a42218] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
12
|
Fitzner B, Holzhueter SA, Ibrahim S, Nizze H, Jaster R. Interferon-gamma treatment accelerates and aggravates autoimmune pancreatitis in the MRL/Mp-mouse. Pancreatology 2009; 9:233-9. [PMID: 19407476 DOI: 10.1159/000199434] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2008] [Accepted: 01/27/2009] [Indexed: 12/11/2022]
Abstract
BACKGROUND T helper 1 cell-released pro-inflammatory cytokines have been implicated in the pathogenesis of autoimmune pancreatitis (AIP), but the experimental database is small. Here, we have directly tested the effects of interferon-gamma (IFN-gamma) by applying it to AIP-prone MRL/Mp mice. METHODS MRL/Mp-mice were treated for 4 weeks with IFN-gamma. Severity of AIP was assessed by histopathology, laboratory findings and gene expression analysis. RESULTS Using a histopathological score from 0 (healthy pancreas) to 4 (severe AIP), we found that IFN-gamma treatment strongly increased severity of pancreatic lesions. IFN-gamma also caused pancreatic accumulation of CD4-, CD8-, C11b- and CD138-positive cells, and enhanced pancreatic mRNA expression of interleukin (IL)-1 beta, transforming growth factor-beta and IFN-gamma itself. In the serum of IFN-gamma-treated mice, higher lipase activities but normal glucose levels were observed. DISCUSSION IFN-gamma accelerates and aggravates AIP in MRL/Mp mice. IFN-gamma-enhanced AIP of MRL/Mp mice may serve to study pathophysiology, and to test diagnostic/therapeutic approaches.
Collapse
Affiliation(s)
- Brit Fitzner
- Department of Medicine, Division of Gastroenterology, University of Rostock, Rostock, Germany
| | | | | | | | | |
Collapse
|
13
|
Lai HS, Lin WH, Hsu WM, Chen CN, Chang KJ, Lee PH. Variations in Interferon Gamma Receptor Gene Expression during Liver Regeneration after Partial Hepatectomy in Rats. Am Surg 2009. [DOI: 10.1177/000313480907500111] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cell-mediated immunity, which includes interferon gamma (IFN-γ) expression, is activated during the process of liver regeneration; however, the genetic pathway of this activation is still unclear. The present study evaluated variations in the interferon gamma receptor (IFN-γR) gene and its mRNA expression during liver regeneration after partial hepatectomy (PH). Male Wistar rats weighing approximately 200 g were subjected to PH (70 or 40%). IFN-γR gene expression in the remnant liver was measured by cDNA microarray, and mRNA expression was verified by real-time quantitative reverse transcription-polymerase chain reaction (Q-PCR) preoperatively and at 2, 4, 6,12, 24, and 72 hours and 7 days postoperatively. The ratio of remnant liver weight to body weight increased markedly after 70 per cent PH and more gradually after 40 per cent PH. It reached near 90 per cent of the preoperative level at 72 hours after PH in both groups. The scanned spots of the genomic survey on the cDNA microarray chips were uneven and increased irregularly in number and density after PH. IFN-γR gene expression increased markedly in a single peak pattern, up to more than double the preoperative level, at 6 hours after 70 per cent PH. The curve in the 40 per cent PH group was flat and peaked at only 1.6 times the preoperative level. The variations in IFN-γR-related mRNA expression were verified by Q-PCR. Elevations in IFN-γR gene and mRNA expression were shown during the early stage of liver regeneration after PH. The genetic pathway of IFN-γ/IFN-γR expression is activated during liver regeneration.
Collapse
Affiliation(s)
- Hong-Shiee Lai
- From the Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Wen-Hsi Lin
- From the Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Wen-Ming Hsu
- From the Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chiung-Nien Chen
- From the Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - King-Jen Chang
- From the Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Po-Huang Lee
- From the Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
14
|
Nomelini RS, De Carvalho Mardegan M, Murta EFC. Utilization of Interferon in Gynecologic and Breast Cancer. Clin Med Oncol 2007. [DOI: 10.4137/cmo.s432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
The usual treatment of gynecologic cancer has been surgery, chemotherapy and radiotherapy. New therapies are being developed to improve efficacy of treatment. Interferons are inducible secretory glycoproteins that have immunomodulatory, antiviral, anti-angiogenic and anti-proliferative effects. Their potential antitumor effect has been demonstrated in many studies. Some patients obtain beneficial effects; in other patients the treatment failure can occur. IFNs can modulate the immune response and inhibition of tumor angiogenesis. When any alteration in gene expression occurs, there is modulation of the receptors of other cytokines and enzymes that control cell function. These alterations can influence the differentiation, cell proliferation rate and apoptosis. The molecular mechanisms that control apoptotic cell death can be improved through cancer management using IFN in single, combination or adjuvant treatment. Malignant cells generally present defects in programmed cell death and apoptosis. Immunomodulation and angiogenesis inhibition are indirect antitumor mechanisms mediated by apoptosis. With regard to immunomodulation, IFNs can have antitumor effects through increases in cytotoxic T cells, natural killer cells and dendritic cells. Angiogenesis inhibition can result from endothelial cell apoptosis. This factor is important in inhibiting tumor genesis and forming metastases. The aim of this review is to discuss the role of Interferon in the treatment of gynecologic malignancies/breast cancer and mechanisms of action.
Collapse
Affiliation(s)
- Rosekeila Simões Nomelini
- Discipline of Gynecology and Obstetrics, Discipline of Human Anatomy, Research Institute of Oncology (IPON), Federal University do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Marília De Carvalho Mardegan
- Obstetric and Gynecologic Pathology, Research Institute of Oncology (IPON), Federal University do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Eddie Fernando Candido Murta
- Research Institute of Oncology (IPON)/Discipline of Gynecology and Obstetrics, Federal University do Triângulo Mineiro (UFTM), Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| |
Collapse
|
15
|
Vitale G, van Eijck CHJ, van Koetsveld Ing PM, Erdmann JI, Speel EJM, van der Wansem Ing K, Mooij DM, Colao A, Lombardi G, Croze E, Lamberts SWJ, Hofland LJ. Type I interferons in the treatment of pancreatic cancer: mechanisms of action and role of related receptors. Ann Surg 2007; 246:259-68. [PMID: 17667505 PMCID: PMC1933574 DOI: 10.1097/01.sla.0000261460.07110.f2] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
OBJECTIVE We evaluated the role of type I interferons (IFNs) and IFN receptors in the regulation of cell growth in 3 human pancreatic adenocarcinoma cell lines (BxPC-3, MiaPaCa-2, and Panc-1). BACKGROUND Chemotherapy and radiotherapy have a marginal role in the management of pancreatic adenocarcinoma. The addition of IFN-alpha showed promising results in early clinical trials. METHODS Cell proliferation and apoptosis were evaluated by DNA measurement and DNA fragmentation, respectively. Type I IFN receptor (IFNAR-1 and IFNAR-2 subunits) was determined by quantitative RT-PCR and immunocytochemistry. Cell cycle distribution was evaluated by propidium iodide staining and flow-cytometric analysis. RESULTS The incubation with IFN-beta for 6 days showed a potent inhibitory effect on the proliferation of BxPC-3 (IC(50), 14 IU/mL) and MiaPaCa-2 (IC(50), 64 IU/mL). The inhibitory effect of IFN-beta was stronger than IFN-alpha in all 3 cell lines and mainly modulated by the stimulation of apoptosis, although cell cycle arrest was induced as well. The expression of the type I IFN receptors was significantly higher in BxPC-3 (the most sensitive cell line to IFN) and mainly localized on the membrane, whereas in Panc-1 (the most resistant cell line) about 60% to 70% of cells were negative for IFNAR-2c with a mainly cytoplasmic staining for IFNAR-2c. CONCLUSION The antitumor activity of IFN-beta is more potent than IFN-alpha in pancreatic cancer cell lines through the induction of apoptosis. Further studies should investigate in vivo whether the intensity and distribution of IFNAR-1 and IFNAR-2c may predict the response to therapy with IFN-alpha and IFN-beta in pancreatic cancer.
Collapse
Affiliation(s)
- Giovanni Vitale
- Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Singhal A, Jaiswal A, Arora VK, Prasad HK. Modulation of gamma interferon receptor 1 by Mycobacterium tuberculosis: a potential immune response evasive mechanism. Infect Immun 2007; 75:2500-10. [PMID: 17339358 PMCID: PMC1865798 DOI: 10.1128/iai.01743-06] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2006] [Revised: 12/01/2006] [Accepted: 02/20/2007] [Indexed: 12/27/2022] Open
Abstract
Mycobacterium tuberculosis inhibits gamma interferon (IFN-gamma)-mediated antimycobacterial action by adopting diverse mechanisms. IFN-gamma binds to its receptor, IFN-gammaR, in order to initiate proper signaling. We have observed reduced surface expression levels of IFN-gamma receptor 1 (IFN-gammaR1) in untreated pulmonary tuberculosis patients compared to those in healthy individuals (P < 0.01). Following antitubercular therapy, the expression of IFN-gammaR1 was restored in these patients. To delineate the mechanism by which M. tuberculosis modulates IFN-gammaR1, in vitro experiments were designed, wherein the down modulation of IFN-gammaR1 surface expression was observed for CD14+ cells in peripheral blood mononuclear cells (PBMCs) cocultured with live M. tuberculosis compared to that for uninfected cells (P < 0.01). No modulation of IFN-gammaR1 expression was observed for CD14+ cells in PBMCs infected with Mycobacterium smegmatis. A time-dependent decrease in IFN-gammaR1 mRNA expression was observed for PBMCs infected with M. tuberculosis. Similar down modulation of IFN-gammaR1 protein and mRNA expression in phorbol myristate acetate-differentiated THP-1 cells (pdTHP-1) by M. tuberculosis was observed (P < 0.01). Using reporter gene analysis of 5' deletion constructs of the IFN-gammaR1 gene (IFNGR1) promoter, the decrease in IFN-gammaR1 mRNA in M. tuberculosis-infected pdTHP-1 cells was shown to be due to the decreased transcription of IFNGR1. By immunoblotting and electrophoretic mobility shift assays, the down regulation of stimulating protein 1 (Sp1) expression and its recruitment on the phorbol ester-responsive element of the IFNGR1 promoter in M. tuberculosis-infected pdTHP-1 cells was observed. This down regulation of Sp1 in pdTHP-1 cells cocultured with M. tuberculosis may be responsible for the down regulation of IFN-gammaR1 expression, thereby potentially altering its receptivity to IFN-gamma.
Collapse
Affiliation(s)
- Amit Singhal
- TB Immunology Laboratory, Department of Biotechnology, All India Institute of Medical Sciences, New Delhi 110029, India
| | | | | | | |
Collapse
|
17
|
Saidi RF, Remine SG, Jacobs MJ. Interferon receptor alpha/beta is associated with improved survival after adjuvant therapy in resected pancreatic cancer. HPB (Oxford) 2007; 9:289-94. [PMID: 18345307 PMCID: PMC2215399 DOI: 10.1080/13651820701329241] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2006] [Indexed: 12/12/2022]
Abstract
AIM Interferons (IFNs) are known to have antiproliferative and immunoregulatory activities that are modulated through specific cell surface ligands, known as IFN-alpha, -beta, and -gamma receptors. The presence of these receptors and their impact on response to adjuvant therapy in patients with pancreatic cancer has not been determined. PATIENTS AND METHODS Slides were prepared from 46 patients with pancreatic adenocarcinoma. Immunohistochemistry (IHC) was subsequently used to determine the expression of IFN- alpha/beta receptor-chain 2 (IFN-alpha/betaR) and IFN-gamma receptor-chain 1 (IFN-gammaR). The correlation between IFN receptor expression, tumor characteristics, and the overall patient response to adjuvant therapy were determined analytically. RESULTS The IHC performed for pancreatic adenocarcinoma demonstrated a high IFN-alpha/betaR expression in 4% (2/46) of patients, moderate expression in 20% (9/46) of patients, and faint or no expression in 76% (35/46) of patients. IHC confirmed a high expression of IFN-gammaR in 52% (24/46) of patients, moderate expression in 35% (16/46) of patients, and faint or no expression in the remaining 13% (6/46) of patients. Thirty-two (69.7%) patients received adjuvant therapy. Clinicopathological survey did not demonstrate any significant correlation between IFN-alpha/betaR and IFN-gammaR expression with regard to tumor size, vascular invasion, perineural invasion, lymph node metastases, or stage of disease. Use of adjuvant therapy was associated with increased survival in patients with IFN-alpha/betaR-positive tumors compared with patients with IFN-alpha/betaR-negative tumors (24 months versus 14.7 months in log rank test, p=0.012). The expression of IFN-gammaR, however, had no impact on patient survival (20 months vs 17 months; p=0.656, log rank test). CONCLUSION IFN-alpha/betaR is associated with improved survival for patients with resectable pancreatic cancer who received adjuvant therapy.
Collapse
Affiliation(s)
- Reza F. Saidi
- Department of Surgery, Providence Hospital and Medical CentersSouthfield MIUSA
| | - Stephen G. Remine
- Department of Surgery, Providence Hospital and Medical CentersSouthfield MIUSA
| | - Michael J. Jacobs
- Department of Surgery, Providence Hospital and Medical CentersSouthfield MIUSA
| |
Collapse
|
18
|
Strath J, Blair GE. Adenovirus subversion of immune surveillance, apoptotic and growth regulatory pathways: a model for tumorigenesis. Acta Microbiol Immunol Hung 2006; 53:145-69. [PMID: 16956126 DOI: 10.1556/amicr.53.2006.2.3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The adenovirus system provides a novel model for evaluating the roles of multiple factors involved in tumour progression. In common with other DNA tumour viruses, adenovirus employs a variety of strategies to evade immune surveillance and perturbs cellular apoptotic and growth regulatory pathways to ensure efficient replication of progeny virions. Such subversion of cellular networks is also found in tumour cells. The mechanism behind the avoidance of immune surveillance and the extent of cellular network interference achieved by adenovirus is still being uncovered and is predicted to have ramifications for the design of cancer therapeutics.
Collapse
Affiliation(s)
- Janet Strath
- Institute of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, UK
| | | |
Collapse
|