1
|
Bekhbat M, Block AM, Dickinson SY, Tharp GK, Bosinger SE, Felger JC. Neurotransmitter and metabolic effects of interferon-alpha in association with decreased striatal dopamine in a non-human primate model of cytokine-Induced depression. Brain Behav Immun 2025; 125:308-318. [PMID: 39826580 PMCID: PMC11903159 DOI: 10.1016/j.bbi.2025.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/13/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025] Open
Abstract
Inflammatory stimuli administered to humans and laboratory animals affect mesolimbic and nigrostriatal dopaminergic pathways in association with impaired motivation and motor activity. Alterations in dopaminergic corticostriatal reward and motor circuits have also been observed in depressed patients with increased peripheral inflammatory markers. The effects of peripheral inflammation on dopaminergic pathways and associated neurobiologic mechanisms and consequences have been difficult to measure in patients. Postmortem tissue (n = 11) from an established, translationally-relevant non-human primate model of cytokine-induced depressive behavior involving chronic interferon-alpha (IFN-a) administration was utilized herein to explore the molecular mechanisms of peripheral cytokine effects on striatal dopamine. Dopamine (but not serotonin or norepinephrine) was decreased in the nucleus accumbens (NAcc) and putamen of IFN-a-treated animals (p < 0.05). IFN-a had no effect on number of striatal neurons or dopamine terminal density, suggesting no overt neurodegenerative changes. RNA sequencing examined in the caudate, putamen, substantia nigra, and prefrontal cortical subregions revealed that while IFN-a nominally up-regulated limited numbers of genes enriching inflammatory signaling pathways in all regions, robust, whole genome-significant effects of IFN-a were observed specifically in putamen. Genes upregulated in the putamen primarily enriched synaptic signaling, glutamate receptor signaling, and inflammatory/metabolic pathways downstream of IFN-a, including MAPK and PI3K/AKT cascades. Conversely, gene transcripts reduced by IFN-a enriched oxidative phosphorylation (OXPHOS), protein translation, and pathways regulated by dopamine receptors. Unsupervised clustering identified a gene co-expression module in the putamen that was associated with both IFN-a treatment and low dopamine levels, which enriched similar inflammatory, metabolic, and synaptic signaling pathways. IFN-a-induced reductions in dopamine further correlated with genes related to excitotoxic glutamate, kynurenine, and altered dopamine receptor signaling (r = 0.78-97, p < 0.05). These findings provide insight into the immunologic mechanisms and neurobiological consequences of peripheral inflammation effects on dopamine, which may inform novel treatment strategies targeting inflammatory, metabolic or neurotransmitter systems in depressed patients with high inflammation.
Collapse
Affiliation(s)
- Mandakh Bekhbat
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | - Andrew M Block
- Department of Orthopaedic Surgery, University of Connecticut, Farmington, CT 06030, USA
| | - Sarah Y Dickinson
- Neuroscience and Behavior Program, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Gregory K Tharp
- Emory Nonhuman Primate Genomics Core, Division of Microbiology and Immunology, Emory National Primate Research Center (EPC), Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Steven E Bosinger
- Emory Nonhuman Primate Genomics Core, Division of Microbiology and Immunology, Emory National Primate Research Center (EPC), Emory University School of Medicine, Atlanta, GA 30322, USA; Department of Pathology and Laboratory Medicine, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Jennifer C Felger
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA; Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
2
|
Qin A, Wu CR, Ho MC, Tsai CY, Chen PJ. Sequential Therapy with Ropeginterferon Alfa-2b and Anti-Programmed Cell Death 1 Antibody for Inhibiting the Recurrence of Hepatitis B-Related Hepatocellular Carcinoma: From Animal Modeling to Phase I Clinical Results. Int J Mol Sci 2023; 25:433. [PMID: 38203603 PMCID: PMC10778875 DOI: 10.3390/ijms25010433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 12/23/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024] Open
Abstract
Hepatocellular carcinoma (HCC) usually recurs after curative surgical resection. Currently, no approved adjuvant therapy has been shown to reduce HCC recurrence rates. In this study, the in vivo effect of sequential combination treatment with recombinant mouse interferon-alpha (rmIFN-α) and an anti-mouse-PD1 antibody on hepatitis B virus (HBV) clearance in mice was evaluated. A Phase I clinical trial was then conducted to assess the safety, tolerability, and inhibitory activity of sequential therapy with ropeginterferon alfa-2b and nivolumab in patients with HCC recurrence who underwent curative surgery for HBV-related HCC. The animal modeling study showed that HBV suppression was significantly greater with the rmIFN-α and anti-PD1 sequential combination treatment in comparison with sole treatment with rmIFN-α or anti-PD1. In the Phase I study, eleven patients completed the sequential therapy with ropeginterferon alfa-2b every two weeks for six doses at 450 µg, followed by three doses of nivolumab every two weeks up to 0.75 mg/kg. A notable decrease in or clearance of HBV surface antigen was observed in two patients. The dose-limiting toxicity of grade 3 alanine transaminase and aspartate aminotransferase increases was observed in one patient. The maximum tolerated dose was then determined. To date, no HCC recurrence has been observed. The treatment modality was well tolerated. These data support the further clinical development of sequential combination therapy as a post-surgery prophylactic measure against the recurrence of HBV-related HCC.
Collapse
Affiliation(s)
- Albert Qin
- Medical Research & Clinical Operations, PharmaEssentia Corporation, Taipei 115, Taiwan
| | - Chang-Ru Wu
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei 100, Taiwan
| | - Ming-Chih Ho
- Department of Surgery, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Chan-Yen Tsai
- Medical Research & Clinical Operations, PharmaEssentia Corporation, Taipei 115, Taiwan
| | - Pei-Jer Chen
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei 100, Taiwan
- Hepatitis Research Center, National Taiwan University Hospital, Taipei 100, Taiwan
| |
Collapse
|
3
|
Qin A. An anti-cancer surveillance by the interplay between interferon-beta and retinoblastoma protein RB1. Front Oncol 2023; 13:1173467. [PMID: 37182173 PMCID: PMC10174298 DOI: 10.3389/fonc.2023.1173467] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 04/05/2023] [Indexed: 05/16/2023] Open
Abstract
Interferon-beta (IFN-β), an extracellular cytokine that initiates signaling pathways for gene regulation, has been demonstrated to function as a tumor suppressor protein through lentiviral gene transduction. In this article, I review the relevant previous works and propose a cell cycle-based, tumor suppressor protein-mediated mechanism of anti-cancer surveillance. IFN-β induces a tumor cell cycle alteration that leads to S phase accumulation, senescence entry, and a loss of tumorigenicity in solid tumor cells. IFN-β does not show a significant cell cycle effect in their normal counterparts. Retinoblastoma protein RB1, another tumor suppressor protein, tightly controls the cell cycle and differentiation of normal cells, preventing them from being significantly impacted by the IFN-β effect. The interplay between IFN-β and RB1 acts as a mechanism of cell cycle-based, tumor suppressor protein-mediated anti-cancer surveillance that can selectively suppress solid tumor or proliferating transformed cells from the loss of control leading to cancer. This mechanism has important implications for the treatment of solid tumors.
Collapse
Affiliation(s)
- Albert Qin
- Medical Research & Clinical Operations, PharmaEssentia Corporation, Taipei, Taiwan
| |
Collapse
|
4
|
Hasselbalch HC, Holmström MO. Perspectives on interferon-alpha in the treatment of polycythemia vera and related myeloproliferative neoplasms: minimal residual disease and cure? Semin Immunopathol 2019; 41:5-19. [PMID: 30203226 PMCID: PMC6323070 DOI: 10.1007/s00281-018-0700-2] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 08/06/2018] [Indexed: 12/19/2022]
Abstract
The first clinical trials of the safety and efficacy of interferon-alpha2 (IFN-alpha2) were performed about 30 years ago. Since then, several single-arm studies have convincingly demonstrated that IFN-alpha2 is a highly potent anti-cancer agent in several cancer types but unfortunately not being explored sufficiently due to a high toxicity profile when using non-pegylated IFN-alpha2 or high dosages or due to competitive drugs, that for clinicians at first glance might look more attractive. Within the hematological malignancies, IFN-alpha2 has only recently been revived in patients with the Philadelphia-negative myeloproliferative neoplasms-essential thrombocytosis, polycythemia vera, and myelofibrosis (MPNs)-and in patients with chronic myelogenous leukemia (CML) in combination with tyrosine kinase inhibitors. In this review, we tell the IFN story in MPNs from the very beginning in the 1980s up to 2018 and describe the perspectives for IFN-alpha2 treatment of MPNs in the future. The mechanisms of actions are discussed and the impact of chronic inflammation as the driving force for clonal expansion and disease progression in MPNs is discussed in the context of combination therapies with potent anti-inflammatory agents, such as the JAK1-2 inhibitors (licensed only ruxolitinib) and statins as well. Interferon-alpha2 being the cornerstone treatment in MPNs and having the potential of inducing minimal residual disease (MRD) with normalization of the bone marrow and low-JAK2V617F allele burden, we believe that combination therapy with ruxolitinib may be even more efficacious and hopefully revert disease progression in many more patients to enter the path towards MRD. In patients with advanced and transforming disease towards leukemic transformation or having transformed to acute myeloid leukemia, "triple therapy" is proposed as a novel treatment modality to be tested in clinical trials combining IFN-alpha2, DNA-hypomethylator, and ruxolitinib. The rationale for this "triple therapy" is given, including the fact that even in AML, IFN-alpha2 as monotherapy may revert disease progression. We envisage a new and bright future with many more patients with MPNs obtaining MRD on the above therapies. From this stage-and even before-vaccination strategies may open a new horizon with cure being the goal for some patients.
Collapse
Affiliation(s)
- Hans Carl Hasselbalch
- Department of Hematology, Zealand University Hospital, Sygehusvej 10, 4000, Roskilde, Denmark.
| | - Morten Orebo Holmström
- Department of Hematology, Zealand University Hospital, Sygehusvej 10, 4000, Roskilde, Denmark
- Center for Cancer Immune Therapy, Department of Hematology, Herlev Hospital, Herlev, Denmark
| |
Collapse
|
5
|
Mehrvarz Sarshekeh A, Halperin DM, Dasari A. Update on management of midgut neuroendocrine tumors. INTERNATIONAL JOURNAL OF ENDOCRINE ONCOLOGY 2016; 3:175-189. [PMID: 27347369 PMCID: PMC4915384 DOI: 10.2217/ije-2015-0004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Midgut neuroendocrine tumors are typically indolent but can be fatal when advanced. They can also cause significant morbidity due to the characteristic carcinoid syndrome. Somatostatin analogs continue to be the mainstay of treatment given their antiproliferative properties, as well as inhibitory effects on hormones that cause carcinoid syndrome. There have been several recent advances in the systemic therapy of these tumors including consolidation of somatostatin analogs as the cornerstone of therapy, completion of pivotal trials with mTOR inhibitors, and the establishment of novel approaches including peptide receptor radionuclide therapy and oral inhibitors of peripheral tryptophan hydroxylase in tumor and symptom control, respectively. In this review article, the recent advances are summarized and an updated approach to management is proposed.
Collapse
Affiliation(s)
- Amir Mehrvarz Sarshekeh
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Daniel M Halperin
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Arvind Dasari
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| |
Collapse
|
6
|
Hasselbalch HC. A new era for IFN-α in the treatment of Philadelphia-negative chronic myeloproliferative neoplasms. Expert Rev Hematol 2014; 4:637-55. [DOI: 10.1586/ehm.11.63] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
7
|
Ozkan E, Tokmak E, Kucuk NO. Efficacy of adding high-dose In-111 octreotide therapy during Sandostatin treatment in patients with disseminated neuroendocrine tumors: clinical results of 14 patients. Ann Nucl Med 2011; 25:425-31. [PMID: 21476058 DOI: 10.1007/s12149-011-0482-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2010] [Accepted: 02/15/2011] [Indexed: 12/22/2022]
Abstract
PURPOSE This study aimed to assess the outcome of high-dose In-111 octreotide treatment and efficacy of long-acting Sandostatin LAR in patients with disseminated neuroendocrine tumors. MATERIALS AND METHODS A total of 14 patients (mean age 51.8 ± 13.2 years; 10 female, 4 male) receiving high-dose In-111 octreotide in our centre for the treatment of neuroendocrine tumors were included in the study. Monthly treatment with long-acting somatostatin analogue [Sandostatin long-acting release (Novartis Pharmaceuticals)] was continued in nine cases. RESULTS During a 3-year period, a total of 45 courses of high-dose In-111 octreotide treatment were delivered to 14 patients. In seven patients receiving an average of four treatment courses (6 carcinoid tumors, 1 thymoma, patients: 2, 4, 5, 11-14) stable disease was achieved (50%). In two patients with carcinoid tumors (patients 1 and 3) who received four treatment courses, partial response was observed (14%). Five patients (36%; 4 NET, 1 gastrinoma; patients 6-10) died due to progressive disease following on average two treatment courses. On average, deaths occurred 2 months after the last treatment dose. No complete responses were seen. Partial response was achieved in two of the nine patients receiving Sandostatin LAR, while four had stable disease. Both treatments were associated with acceptable tolerability. CONCLUSIONS High-dose In-111 octreotide can be safely administered in conjunction with somatostatin analogue in patients with disseminated NET and this treatment may help to stabilize the disease.
Collapse
Affiliation(s)
- Elgin Ozkan
- Department of Nuclear Medicine, Ankara University Faculty of Medicine, Ankara, Turkey.
| | | | | |
Collapse
|
8
|
α-2-HS-glycoprotein is a potential marker predicting hepatitis B e antigen seroconversion in patients with chronic hepatitis B during treatment with pegylated interferon alpha-2b. SCIENCE CHINA-LIFE SCIENCES 2011; 54:39-47. [PMID: 21253869 DOI: 10.1007/s11427-010-4111-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2010] [Accepted: 08/09/2010] [Indexed: 01/27/2023]
Abstract
The efficacy of interferon (IFN) is limited in about 1/3 of patients with chronic hepatitis B (CHB). We used two-dimensional electrophoresis (2-DE)-based proteomic strategies to identify potential serum markers predicting hepatitis B e antigen (HBeAg) seroconversion in these patients during IFN therapy. Two groups of patients were enrolled: training and validation. In the training group, 2-DE experiments and subsequent identification of altered levels of proteins showed that α-2-HS-glycoprotein, leucine-rich α-2-glycoprotein, and haptoglobin were significantly upregulated as compared with baseline levels in the HBeAg seroconversion group, whereas apolipoprotein C-III precursor, leucine-rich α-2-glycoprotein, and α-albumin were downregulated in the non-seroconversion group. For patients with HBeAg seroconversion in the training group, Western blot analyses showed that α-2-HS-glycoprotein levels in 75% of patients were significantly upregulated at the end of the treatment as compared with baseline levels. Subsequent experiments in the validation group showed that α-2-HS-glycoprotein levels were significantly increased at week 4 in 83.33% of patients in the HBeAg seroconversion group. Dynamic changes in the serum level of α-2-HS-glycoprotein may be a potential early marker for predicting HBeAg seroconversion during IFN treatment for CHB.
Collapse
|
9
|
Ma H, Yang RF, Wei L. Quantitative serum HBsAg and HBeAg are strong predictors of sustained HBeAg seroconversion to pegylated interferon alfa-2b in HBeAg-positive patients. J Gastroenterol Hepatol 2010; 25:1498-506. [PMID: 20796146 DOI: 10.1111/j.1440-1746.2010.06282.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND AND AIM To evaluate the usefulness of quantitative hepatitis B surface antigen (HBsAg) and hepatitis B e antigen (HBeAg) for predicting HBeAg seroconversion in chronic hepatitis B patients treated with conventional interferon (IFN) alfa-2b or PegIFN alfa-2b. METHODS Fifty-eight patients were enrolled; 29 for the training group and 29 for the validating group. Quantification of HBsAg and HBeAg was carried out at baseline, week 12, week 24, and then again at 12 and 24 weeks follow up, respectively, for two groups. Sixteen patients in the training group were followed up for 5 years. RESULTS The cutoff of 1500 IU/mL in serum HBsAg at week 12 had a positive predictive value (PPV) of 33% and a negative predictive value (NPV) of 91%, and 2890 IU/mL at week 24 had a PPV of 43% and an NPV of 95% for HBeAg seroconversion at week 48. The cutoff of 17.55 Paul Ehrlich Institute units/mL (PEI-U/mL) in serum HBeAg at week 12 had a PPV of 38% and an NPV of 95%, and 8.52 PEI-U/mL at week 24 had a PPV of 44% and a NPV of 100% for HBeAg seroconversion at week 48. Moreover the HBsAg and HBeAg levels of PegIFN alfa-2b group were lower than those of the conventional IFN alfa-2b group. During follow up, patients with HBeAg seroconversion remained HBeAg negative and none of them progressed to cirrhosis, but among the patients with non-HBeAg seroconversion, two progressed to cirrhosis. Two additional patients with negative HBeAg were observed. CONCLUSIONS On-treatment serum HBsAg and HBeAg had high predictive values to predict sustained HBeAg seroconversion by PegIFN alfa-2b. Patients who cleared HBeAg had better survival free of hepatic complications during long-term follow-up study.
Collapse
Affiliation(s)
- Hui Ma
- Peking University People's Hospital, Peking University Hepatology Institute, Beijing, China
| | | | | |
Collapse
|
10
|
Bekisz J, Baron S, Balinsky C, Morrow A, Zoon KC. Antiproliferative Properties of Type I and Type II Interferon. Pharmaceuticals (Basel) 2010; 3:994-1015. [PMID: 20664817 PMCID: PMC2907165 DOI: 10.3390/ph3040994] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2010] [Revised: 03/15/2010] [Accepted: 03/29/2010] [Indexed: 01/22/2023] Open
Abstract
The clinical possibilities of interferon (IFN) became apparent with early studies demonstrating that it was capable of inhibiting tumor cells in culture and in vivo using animal models. IFN gained the distinction of being the first recombinant cytokine to be licensed in the USA for the treatment of a malignancy in 1986, with the approval of IFN-α2a (Hoffman-La Roche) and IFN-α2b (Schering-Plough) for the treatment of Hairy Cell Leukemia. In addition to this application, other approved antitumor applications for IFN-α2a are AIDS-related Kaposi's Sarcoma and Chronic Myelogenous Leukemia (CML) and other approved antitumor applications for IFN-α2b are Malignant Melanoma, Follicular Lymphoma, and AIDS-related Kapoisi's Sarcoma. In the ensuing years, a considerable number of studies have been conducted to establish the mechanisms of the induction and action of IFN's anti-tumor activity. These include identifying the role of Interferon Regulatory Factor 9 (IRF9) as a key factor in eliciting the antiproliferative effects of IFN-α as well as identifying genes induced by IFN that are involved in recognition of tumor cells. Recent studies also show that IFN-activated human monocytes can be used to achieve >95% eradication of select tumor cells. The signaling pathways by which IFN induces apoptosis can vary. IFN treatment induces the tumor suppressor gene p53, which plays a role in apoptosis for some tumors, but it is not essential for the apoptotic response. IFN-α also activates phosphatidylinositol 3-kinase (PI3K), which is associated with cell survival. Downstream of PI3K is the mammalian target of rapamycin (mTOR) which, in conjunction with PI3K, may act in signaling induced by growth factors after IFN treatment. This paper will explore the mechanisms by which IFN acts to elicit its antiproliferative effects and more closely examine the clinical applications for the anti-tumor potential of IFN.
Collapse
Affiliation(s)
- Joseph Bekisz
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|
11
|
Redig AJ, Sassano A, Majchrzak-Kita B, Katsoulidis E, Liu H, Altman JK, Fish EN, Wickrema A, Platanias LC. Activation of protein kinase C{eta} by type I interferons. J Biol Chem 2009; 284:10301-14. [PMID: 19211565 DOI: 10.1074/jbc.m807254200] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Type I interferons (IFNs) are cytokines with diverse biological properties, including antiviral, growth inhibitory, and immunomodulatory effects. Although several signaling pathways are activated during engagement of the type I IFN receptor and participate in the induction of IFN responses, the mechanisms of generation of specific signals for distinct biological effects remain to be elucidated. We provide evidence that a novel member of the protein kinase C (PKC) family of proteins is rapidly phosphorylated and activated during engagement of the type I IFN receptor. In contrast to other members of the PKC family that are also regulated by IFN receptors, PKCeta does not regulate IFN-inducible transcription of interferon-stimulated genes or generation of antiviral responses. However, its function promotes cell cycle arrest and is essential for the generation of the suppressive effects of IFNalpha on normal and leukemic human myeloid (colony-forming unit-granulocyte macrophage) bone marrow progenitors. Altogether, our studies establish PKCeta as a unique element in IFN signaling that plays a key and essential role in the generation of the regulatory effects of type I IFNs on normal and leukemic hematopoiesis.
Collapse
Affiliation(s)
- Amanda J Redig
- Robert H. Lurie Comprehensive Cancer Center and Division of Hematology/Oncology, Northwestern University Medical School, Chicago, Illinois 60611, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Moschos S, Kirkwood JM. Present role and future potential of type I interferons in adjuvant therapy of high-risk operable melanoma. Cytokine Growth Factor Rev 2007; 18:451-8. [PMID: 17693125 DOI: 10.1016/j.cytogfr.2007.06.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Few molecular therapeutic approaches have been so rigorously investigated in relation to the pathophysiology and outcome of human diseases as type I interferons. Historically, IFNs were discovered after the phenomenon of 'interference' was first described by Isaacs and Lindenmann in 1957, and for years IFNs (IFNalpha) were considered as potential "antiviral penicillins" until the broader spectrum of effects upon normal cell physiology, the natural and adaptive immune systems, and tumor growth and proliferation were described. Interferon beta (IFNbeta) was the second human gene after insulin to be cloned, and it codes for the first cytokine used to treat human malignancies. Despite the progress in understanding and treating cancer over the last 25 years, IFN alpha (IFNalpha) remains the most commonly used biologically active cytokine in the treatment of solid tumors, and for some like melanoma, the only successful agent. In this review we discuss the role of type I interferons in the pathophysiology and treatment of melanoma, with emphasis on the 22 years of work conducted at the University of Pittsburgh. We discuss potential mechanisms that partially explain the clinical benefit, and set the groundwork for building upon, the design of more effective treatments for this disease.
Collapse
Affiliation(s)
- Stergios Moschos
- Department of Medicine, University of Pittsburgh School of Medicine, Hillman Cancer Center, Research Pavilion, Suite L1.32c, 5117 Centre Avenue, Pittsburgh, PA 15213-2584, United States
| | | |
Collapse
|
13
|
|
14
|
Wang HS, Oh DS, Ohning GV, Pisegna JR. Cyto-reduction of neuroendocrine tumours using Sandostatin LAR in combination with Infergen: results of a case series. J Pharm Pharmacol 2006; 58:1623-1628. [PMID: 17331326 PMCID: PMC6736541 DOI: 10.1211/jpp.58.12.0009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Historically, limited trials evaluating biotherapy in treating metastatic neuroendocrine tumours have yielded mixed results. In this study, the efficacy of a novel combination therapy featuring longacting Sandostatin LAR plus alpha-interferon was evaluated. In a prospective case series, 12 patients with unresectable metastatic neuroendocrine tumours refractory to treatment initiated therapy with Infergen and Sandostatin LAR. Radiological response was followed serially at 3-month intervals. A biochemical response was considered significant if marker levels decreased by > or = 50% compared with baseline. Inhibition of tumour growth lasting for greater than 3 months (mean response 22.6+/-17.7 months) was seen in eight patients. Complete tumour regression was observed in one patient, lasting for 40 months; three patients exhibited partial tumour regression (mean response 29.3+/-24.0 months), and four patients maintained a stable tumour response (mean response 13.3+/-9.2 months). Four patients showed no response to therapy (mean response 5.0+/-6.0 months). All enrolled patients are alive currently. The biochemical response seen in seven patients did not correlate with the radiological response. These results suggest that the novel combination of longacting Sandostatin LAR with an alpha-interferon may be at least as effective as either combination therapy with short-acting octreotide or monotherapy with Sandostatin LAR.
Collapse
Affiliation(s)
- Hank S Wang
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | | | | | | |
Collapse
|
15
|
Berzsenyi MD, Roberts SK, Beard MR. Genomics of Hepatitis B and C Infections: Diagnostic and Therapeutic Applications of Microarray Profiling. Antivir Ther 2006. [DOI: 10.1177/135965350601100515] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Microarray profiling offers many potential advances in diagnostic and therapeutic intervention in human disease because of its unparalleled ability to conduct high-throughput analysis of gene expression. However, limitations of this technique relate in part to issues regarding the various methodologies and experimental designs as well as difficulties in the interpretation of results. Despite this, microarray profiling has led to a better understanding of the molecular pathogenesis of hepatitis B virus (HBV) and hepatitis C virus (HCV) infection. Key events in clearance and the development of chronicity of HCV have been identified that may prove to have a role in the development of future treatments. In addition, pharmacogenomic studies of interferon-based treatment for chronic HCV and HBV have provided mechanistic insights into the therapeutic action of interferons. These advances have implications with respect to the development of improved therapeutic agents. New biomarkers for cancer screening and gene profiles with prognostic value for survival have also been developed for hepatocellular carcinoma, which frequently complicates chronic viral hepatitis. Thus, microarray profiling offers enormous potential for improvements in antiviral therapy and our understanding of blood-borne viral hepatitis.
Collapse
Affiliation(s)
- Mark D Berzsenyi
- Department of Gastroenterology, Alfred Hospital, Victoria, Australia
| | - Stuart K Roberts
- Department of Gastroenterology, Alfred Hospital, Victoria, Australia
| | - Michael R Beard
- Infectious Diseases Laboratories and Hanson Institute, Institute of Medical and Veterinary Science, Adelaide, South Australia
- School of Molecular and Biomedical Science, The University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
16
|
Li HT, Ma B, Mi JW, Jin HY, Xu LN, Wang JW. Cloning, in vitro expression and bioactivity of goose interferon-alpha. Cytokine 2006; 34:177-83. [PMID: 16757174 DOI: 10.1016/j.cyto.2006.04.013] [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: 02/11/2006] [Revised: 04/13/2006] [Accepted: 04/25/2006] [Indexed: 10/24/2022]
Abstract
The cDNA of goose interferon-alpha (goIFN-alpha) was amplified from PHA stimulated PBMCs of goose by RT-PCR. The cDNA encodes a 30-amino acid signal peptide and a 161-amino acid mature protein, respectively. Recombinant mature goIFN-alpha (rgoIFN-alpha) expressed by prokaryotic and eukaryotic system possessed antiviral activity that was neutralized by rabbit anti-rgoIFN-alpha antibody in vitro. On the other hand, rgoIFN-alpha lacks intrinsic macrophage activating factor (MAF) activity, peripheral blood leukocyte-derived macrophages (PBLMs) could not produce nitric oxide (NO) by stimulate with rgoIFN-alpha as compared to stimulate with recombinant mature goIFN-gamma (rgoIFN-gamma) that was a powerful NO stimulant in vitro.
Collapse
Affiliation(s)
- Hong-Tao Li
- College of Animal Medicine, Northeast Agricultural University, Harbin 150030, China
| | | | | | | | | | | |
Collapse
|
17
|
Kouvaraki MA, Solorzano CC, Shapiro SE, Yao JC, Perrier ND, Lee JE, Evans DB. Surgical treatment of non-functioning pancreatic islet cell tumors. J Surg Oncol 2005; 89:170-85. [PMID: 15719379 DOI: 10.1002/jso.20178] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Pancreatic endocrine tumors (PETs) are rare neoplasms originating from the amine precursor uptake and decarboxylation (APUD) stem cells. Although the majority of PETs are sporadic, they frequently occur in familial syndromes. PETs may cause a variety of functional syndromes or symptoms of local progression if they are non-functional. General neuroendocrine tumor markers are highly sensitive in the diagnostic assessment of a PET. Imaging studies for tumor localization and staging include computer tomography (CT) scan, magnetic resonance imaging (MRI), In(111)-octreotide scan, MIBG, and endoscopic ultrasonography (EUS). Treatment of PETs often requires a multi-modality approach; however, surgical resection remains the only curative therapy for localized (non-metastatic) disease. Treatment of metastatic disease includes biologic agents, cytotoxic chemotherapy, and liver-directed therapies.
Collapse
Affiliation(s)
- Maria A Kouvaraki
- Department of Surgical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
Neuroendocrine tumors (NETs) constitute a heterogeneous group of neoplasms that originate from endocrine glands such as the pituitary, the parathyroids, and the (neuroendocrine) adrenal, as well as endocrine islets within glandular tissue (thyroid or pancreatic) and cells dispersed between exocrine cells, such as endocrine cells of the digestive (gastroenteropancreatic) and respiratory tracts. Conventionally, NETs may present with a wide variety of functional or nonfunctional endocrine syndromes and may be familial and have other associated tumors. Assessment of specific or general tumor markers offers high sensitivity in establishing the diagnosis and can also have prognostic significance. Imaging modalities include endoscopic ultrasonography, computed tomography and magnetic resonance imaging, and particularly, scintigraphy with somatostatin analogs and metaiodobenzylguanidine. Successful treatment of disseminated NETs requires a multimodal approach; radical tumor surgery may be curative but is rarely possible. Well-differentiated and slow-growing gastroenteropancreatic tumors should be treated with somatostatin analogs or alpha-interferon, with chemotherapy being reserved for poorly differentiated and progressive tumors. Therapy with radionuclides may be used for tumors exhibiting uptake to a diagnostic scan, either after surgery to eradicate microscopic residual disease or later if conventional treatment or biotherapy fails. Maintenance of the quality of life should be a priority, particularly because patients with disseminated disease may experience prolonged survival.
Collapse
Affiliation(s)
- Gregory A Kaltsas
- Department of Endocrinology, St Bartholomew's Hospital, London EC1A 7BE, United Kingdom
| | | | | |
Collapse
|
19
|
Donskov F, Marcussen N, Hokland M, Fisker R, Madsen HHT, von der Maase H. In vivo assessment of the antiproliferative properties of interferon-alpha during immunotherapy: Ki-67 (MIB-1) in patients with metastatic renal cell carcinoma. Br J Cancer 2004; 90:626-31. [PMID: 14760375 PMCID: PMC2409612 DOI: 10.1038/sj.bjc.6601587] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The aim of the present study was to investigate the in vivo antiproliferative effect of interferon alpha (IFN-α) in patients with metastatic renal cell carcinoma (mRCC). Core needle biopsies of metastatic and/or the primary kidney cancer were obtained before interleukin-2 (IL-2)- and IFN-α-based immunotherapy in 34 patients and repeated after 5 weeks in 25 patients. Tumour proliferation was assessed by use of the anti-Ki-67 antibody MIB-1 and evaluated in multiple, random systematic sampled fields of vision. Ki-67 labelling index (LI) at baseline was median 13.6% (range 1.2–85.0) and median 10.6% (range 1.3–48.6%) at week 5 with a median overall decline of 15.2% (range −95 to +258%) from baseline to week 5. There was no difference between responding and nonresponding patients. Ki-67 LI at week 5 was significantly correlated to survival. Thus, median survival of patients with Ki-67 LI ⩽10.6% at week 5 was 25.1 months compared to 11.5 months for patients with Ki-67 LI >10.6% (P=0.016). Baseline or change in Ki-67 LI did not correlate to survival. These data suggest that IFN-α in vivo has only modest effect on tumour proliferation in patients with mRCC. Tumour Ki-67 (MIB-1) reactivity after 1 month of immunotherapy appears to be a significant predictor of patient survival.
Collapse
Affiliation(s)
- F Donskov
- Department of Oncology, Aarhus University Hospital, Denmark.
| | | | | | | | | | | |
Collapse
|
20
|
Barnett PV, Keel P, Reid S, Armstrong RM, Statham RJ, Voyce C, Aggarwal N, Cox SJ. Evidence that high potency foot-and-mouth disease vaccine inhibits local virus replication and prevents the ‘carrier’ state in sheep. Vaccine 2004; 22:1221-32. [PMID: 15003651 DOI: 10.1016/j.vaccine.2003.09.024] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2003] [Accepted: 09/16/2003] [Indexed: 11/30/2022]
Abstract
The ability of a single administration of a high, medium and low potency foot-and-mouth disease (FMD) vaccine to decrease or inhibit local virus replication and excretion in the oropharynx of sheep following aerosol challenge with homologous live virus 14 days later was examined. Unvaccinated sheep showed signs of clinical FMD, whereas all of the vaccinated sheep, regardless of antigen payload, were protected against clinical disease and development of viraemia. Virological and serological results confirmed that there had been no local virus replication in the oropharynx of sheep from the high potency vaccine group in contrast to moderate or substantial virus replication in the oropharynx of the low potency vaccinated or unvaccinated sheep respectively. The vaccines showed no evidence of promoting a local mucosal antibody response at the time of virus challenge, but were capable of stimulating a systemic gamma interferon response, the level of which was related to the antigen payload. This suggests that the systemic gamma interferon response could be a useful indicator of the ability of a FMD vaccine to elicit a sterile immunity and indicates that further work is warranted to investigate the role of systemic gamma interferon in this immunity. This is the first experiment to clearly show that high potency, high payload, FMD vaccines are capable of inhibiting local virus replication and consequently persistence and the carrier state in this target species.
Collapse
Affiliation(s)
- P V Barnett
- Institute for Animal Health, Pirbright Laboratory, Ash Road, Pirbright, Woking, Surrey GU24 0NF, UK.
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Abstract
BACKGROUND The optimum management of neuroendocrine liver metastases, particularly the role of liver transplantation is ill-defined. Current strategies are based on anecdotal reports and small retrospective studies, rather than prospective data. This, as well as the failure to standardize treatment, has probably contributed to the reported variations in outcome. DATA SOURCES To formulate a putative management protocol and to reevaluate the role of liver transplantation in patients with neuroendocrine liver metastases, a review of the published literature (Medline search) was conducted. CONCLUSIONS Isolated hepatic metastases should be resected when suitable. Chemoembolization of liver metastases should precede resection of bulky disease and be used to palliate those with unresectable disease. Radiofrequency ablation is suitable for smaller metastatic lesions (<3 cm diameter) in the liver. Systemic treatment with somatostatin analogues or radioactive metaiodobenzylguanidine (MIBG) is appropriate to reduce symptoms and slow disease progression, but prospective data is required to define their exact roles as adjuvant and therapeutic agents. Although current systemic chemotherapy, applied in isolation, may have a role in patients in whom other therapies have failed, its efficacy remains unproven. Restriction of liver transplantation to the treatment of patients with carcinoid metastases with biologically favorable features, limited tumor bulk and without systemic disease may make transplantation a curative rather than a palliative treatment option in selected patients.
Collapse
Affiliation(s)
- Robert Sutcliffe
- Institute of Liver Studies, King's College Hospital, Denmark Hill, London SE5 9RS, United Kingdom
| | | | | | | | | |
Collapse
|
22
|
Banno T, Adachi M, Mukkamala L, Blumenberg M. Unique Keratinocyte-Specific Effects of Interferon-γ that Protect Skin from Viruses, Identified Using Transcriptional Profiling. Antivir Ther 2003. [DOI: 10.1177/135965350300800616] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Interferon (IFN)-γ, is a multifunctional, immunomodulatory cytokine with cell type-specific antiviral activities, particularly important in skin, where it is implicated in many diseases ranging from warts to psoriasis and cancer. Since epidermis is our first line of defence against many viruses, we investigated the molecular processes regulated by IFN-γ in keratinocytes using DNA microarrays. We identified the IFN-γ-regulated keratinocyte-specific genes and antiviral processes. Exclusively in keratinocytes, IFN-γ-induced tight junction proteins, presumably to deny viruses paracellular routes of infection. Furthermore, differing from published data, we find that IFN-γ suppressed the expression of keratinocytes differentiation markers including desmosomal proteins, cornified envelope components and suprabasal cytokeratins. Inhibition of differentiation may interfere with the epidermal tropism of viruses that require differentiating cells for growth, for example, papillomaviruses. As in other cell types, IFN-γ induced HLA, cell adhesion and proteasome proteins, facilitating leukocyte attraction and antigen-presentation by keratinocytes. IFN-γ also induced chemokine/cytokines specific for mononuclear cells. IFN-γ suppressed the expression of over 100 genes responsible for cell cycle, DNA replication and RNA metabolism, thereby shutting down many nuclear processes and denying viruses a healthy cell in which to replicate. Thus, uniquely in keratinocytes, IFN-γ initiates a well-organized molecular programme boosting host antiviral defences, obstructing viral entry, suppressing cell proliferation and impeding differentiation.
Collapse
Affiliation(s)
- Tomohiro Banno
- Departments of Dermatology, Tsukuba University, Tennodai, Ibaraki, Japan
- Dermatology Departments, the Institute of Clinical Medicine, Tsukuba University, Tennodai, Ibaraki, Japan
| | - Makoto Adachi
- Departments of Dermatology, Tsukuba University, Tennodai, Ibaraki, Japan
- Faculty of Medicine, University of Tokyo, Tokyo, Japan
| | - Lakshmi Mukkamala
- Departments of Dermatology, Tsukuba University, Tennodai, Ibaraki, Japan
| | - Miroslav Blumenberg
- Departments of Dermatology, Tsukuba University, Tennodai, Ibaraki, Japan
- Biochemistry, NYU School of Medicine, New York, NY, USA
- The Cancer Institute, NYU School of Medicine, New York, NY, USA
| |
Collapse
|
23
|
Saydam G, Aydin HH, Sahin F, Selvi N, Oktem G, Terzioglu E, Buyukkececi F, Omay SB. Involvement of protein phosphatase 2A in interferon-alpha-2b-induced apoptosis in K562 human chronic myelogenous leukaemia cells. Leuk Res 2003; 27:709-17. [PMID: 12801529 DOI: 10.1016/s0145-2126(02)00347-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Interferon-alpha (IFN-alpha)-2b is known to have antiproliferative effects on hematological malignant cells, especially chronic myelogenous leukaemia (CML). However, it can induce cytogenetical remissions in a very small percentage of the patients. Also during interferon therapy, resistance can emerge in the CML clones. K562 is an in vitro model cell line transformed from a Ph positive CML patient. It can be induced to differentiate to granulocytic and/or monocytic lineages with certain molecules. IFN-alpha-2b generally exerts its effects on CML cells by Janus family kinases (Jak/Stat) pathway, mostly through tyrosine kinase system. However, there is almost no data on the relevance of serine/threonine (Ser/Thr) protein phosphatase (PP) system in the interferon induced signal transduction pathways. In this study, we investigated serine/threonine protein phosphatases in the IFN-alpha-2b induced K562 cytotoxicity. Trypan blue dye exclusion test and MTT assay were utilised for determining cytotoxicity. IC(50) of IFN-alpha-2b on K562 cells was found to be 600IU/ml. However, no differentiation was determined by analysis of cell surface antigen expressions. Serine/threonine protein phosphatase inhibitors calyculin A (Cal A) and okadaic acid (OKA) augmented the IFN-alpha-2b induced cytotoxicity. Apoptosis assay by the mono-oligonucleosome detection and acridine orange/propidium iodide dye revealed marked apoptosis underlying cytotoxicity. Phosphatase enzyme assay revealed a gradual increase in protein phosphatase 2A (PP2A) activity during interferon induced cytotoxicity. Conversely, immunoblots showed no change in the expression of PP2A catalytic and regulatory subunits. In conclusion, PP2A plays a role in IFN-alpha-2b induced apoptosis of K562 cells and should be investigated as a new window furthermore.
Collapse
Affiliation(s)
- Guray Saydam
- Department of Hematology, School of Medicine, Ege University, Bornova, TR-35100 Izmir, Turkey
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Xin H, Pramanik R, Choubey D. Retinoblastoma (Rb) protein upregulates expression of the Ifi202 gene encoding an interferon-inducible negative regulator of cell growth. Oncogene 2003; 22:4775-85. [PMID: 12894219 DOI: 10.1038/sj.onc.1206780] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Studies have indicated that ectopic expression of p202, an interferon (IFN)-inducible retinoblastoma (Rb)-binding protein, in cultured cells retards cell proliferation and modulates cell survival. Consistent with a role of p202 in cell cycle regulation, levels of p202 increase in cells arrested in the G0/G1 phase of cell cycle after withdrawal of serum growth factors. However, a role for p202 in cell growth arrest remains to be defined. Moreover, it remains unclear how levels of p202 are upregulated during the cell growth arrest. Here, we report that Rb upregulates expression of Ifi202 gene. We found that basal as well as IFN-induced levels of p202 were significantly higher in wild-type (Rb(+/+)) mouse embryonic fibroblasts (MEFs) than isogenic Rb(-/-) MEFs. Consistent with the regulation of Ifi202 gene by Rb, expression of functional Rb, but not a pocket mutant of it, stimulated the activity of a reporter whose expression was driven by the 5'-regulatory region of Ifi202 gene. Importantly, the stimulation by Rb was dependent, in part, on a JunD/AP-1 DNA-binding site present in the 5'-regulatory region of the Ifi202 gene. Moreover, basal levels of p202 were significantly higher in wild-type (JunD(+/+)) than isogenic JunD(-/-) MEFs. Additionally, we found that increased expression of p202 potentiated the Rb-mediated inhibition of cell growth and mutations in the Rb-binding motif (LxCxE) of p202 significantly reduced cell survival. Together, our observations support the idea that the transcriptional activation of Ifi202 gene by Rb/JunD may be important for the regulation of cell growth and survival.
Collapse
Affiliation(s)
- Hong Xin
- Department of Radiation Oncology, Stritch School of Medicine, Loyola University Medical Center, 2160 South First Avenue, Building No. 1, Mail code: 114B, Maywood, IL 60153, USA
| | | | | |
Collapse
|
25
|
Abstract
The interferons (IFNs), in addition to their well-known antiviral activities, have important roles in the control of cell proliferation and are effective agents for the treatment of a limited number of malignant diseases. IFNs not only regulate cell growth and division but also influence cell survival through their effects on apoptosis. This review describes the current state of knowledge about the mechanisms of action of these cytokines on the apoptotic machinery, with particular emphasis on the synergism that exists between the IFNs and other proapoptotic agents, such as members of the tumor necrosis factor (TNF) family. The review also discusses the physiologic and clinical implications of the effects of the IFNs on apoptosis for regulation of viral infection and tumor growth.
Collapse
Affiliation(s)
- Michael J Clemens
- Translational Control Group, Department of Basic Medical Sciences, St. George's Hospital Medical School, London SW17 0RE, UK.
| |
Collapse
|
26
|
Sancéau J, Poupon MF, Delattre O, Sastre-Garau X, Wietzerbin J. Strong inhibition of Ewing tumor xenograft growth by combination of human interferon-alpha or interferon-beta with ifosfamide. Oncogene 2002; 21:7700-9. [PMID: 12400012 DOI: 10.1038/sj.onc.1205881] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2002] [Revised: 07/05/2002] [Accepted: 07/18/2002] [Indexed: 11/09/2022]
Abstract
Ewing sarcoma is the second most common bone tumor in childhood. Despite aggressive chemotherapy and radiotherapy strategies, the prognosis of patients with metastatic disease remains poor. We have recently reported that Ewing tumor cell proliferation was strongly inhibited by IFN-beta and to a lesser degree by IFN-alpha. Moreover, under IFN-beta treatment, some cell lines undergo apoptosis. Since the possibility of using IFNs for Ewing tumor treatments may be of interest, we have evaluated the efficacy of Hu-IFNs in a nude mice model of Ewing tumor xenografts. The results reported here show that human type I IFNs, Hu-IFN-alpha and Hu-IFN-beta impaired tumor xenograft take and displayed an anti-growth effect toward established xenografts. Furthermore, we have also shown that combined therapy with Hu-IFNs and ifosfamide (IFO), an alkylating agent widely used in high-dose chemotherapy of Ewing tumors, results in a strong antitumor effect. Pathological analysis showed that Hu-IFN-alpha/IFO and Hu-IFN-beta/IFO were characterized by a dramatic decrease in the mitotic index and marked necrosis, as well as extensive fibrosis associated with numerous calcifications. To our knowledge, this is the first demonstration of a potential antitumor effect of human type I IFNs and IFO on Ewing tumors, providing a rational foundation for a promising therapeutic approach to Ewing sarcoma.
Collapse
Affiliation(s)
- Josiane Sancéau
- INSERM U365, Institut Curie, Section Recherche 26, rue d'Ulm, 75 248 - Paris Cedex 05, France
| | | | | | | | | |
Collapse
|
27
|
He Y, Katze MG. To interfere and to anti-interfere: the interplay between hepatitis C virus and interferon. Viral Immunol 2002; 15:95-119. [PMID: 11952150 DOI: 10.1089/088282402317340260] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
As popular strategies used by numerous viruses, interception of interferon (IFN) signaling and inhibition of IFN-induced antiviral functions allow viruses to evade the host immune response and set up successful infections. Hepatitis C virus (HCV), the leading cause of chronic liver disease worldwide and a major public health hazard, causes persistent infection in the majority of infected individuals. IFN-based therapies, currently the only ones available for HCV infection, have been unable to eliminate viral infection in the majority of patients, and many studies suggest that HCV possesses mechanisms to antagonize the IFN-induced antiviral response. Multiple viral, host, and IFN-associated factors have been implicated in the interplay between HCV and IFN. Two viral proteins, NS5A and E2, became the focus of much attention and extensive study because of their abilities to inhibit IFN-induced, double-stranded RNA-activated protein kinase (PKR), a major mediator of the IFN-induced biologic response, and to perturb the IFN signaling pathway. In this review, we discuss the significance of the interferon sensitivity determining region (ISDR) within NS5A, which has been the subject of intense debates. In addition, we discuss the potential mechanisms by which NS5A interferes with IFN signaling and the current working models. Further understanding of the molecular mechanisms underlying the interaction between HCV and IFN will likely facilitate improvement of current IFN-based therapies and development of novel treatments for the HCV pandemic. Future HCV research will benefit from both the development of efficient, convenient model systems for viral propagation, and the utilization of high throughput, genomic-scale approaches.
Collapse
Affiliation(s)
- Yupeng He
- Department of Microbiology, School of Medicine, University of Washington, 98195, USA
| | | |
Collapse
|
28
|
Abstract
The action of interferons (IFNs) on virus-infected cells and surrounding tissues elicits an antiviral state that is characterized by the expression and antiviral activity of IFN-stimulated genes. In turn, viruses encode mechanisms to counteract the host response and support efficient viral replication, thereby minimizing the therapeutic antiviral power of IFNs. In this review, we discuss the interplay between the IFN system and four medically important and challenging viruses -- influenza, hepatitis C, herpes simplex and vaccinia -- to highlight the diversity of viral strategies. Understanding the complex network of cellular antiviral processes and virus-host interactions should aid in identifying new and common targets for the therapeutic intervention of virus infection. This effort must take advantage of the recent developments in functional genomics, bioinformatics and other emerging technologies.
Collapse
Affiliation(s)
- Michael G Katze
- Department of Microbiology, University of Washington, Seattle, Washington 98195-8070, USA.
| | | | | |
Collapse
|
29
|
de Veer MJ, Holko M, Frevel M, Walker E, Der S, Paranjape JM, Silverman RH, Williams BRG. Functional classification of interferon‐stimulated genes identified using microarrays. J Leukoc Biol 2001. [DOI: 10.1189/jlb.69.6.912] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Affiliation(s)
| | - Michelle Holko
- Department of Cancer Biology, Lerner Research Institute, and Ohio
- Department of Genetics, Case Western Reserve University, Cleveland, Ohio; and
| | - Mathias Frevel
- Department of Cancer Biology, Lerner Research Institute, and Ohio
| | - Eldon Walker
- Computer Core, Cleveland Clinic Foundation, Ohio
| | - Sandy Der
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
30
|
Jin KL, Graham SH, Mao XO, He X, Nagayama T, Simon RP, Greenberg DA. Bax kappa, a novel Bax splice variant from ischemic rat brain lacking an ART domain, promotes neuronal cell death. J Neurochem 2001; 77:1508-19. [PMID: 11413234 DOI: 10.1046/j.1471-4159.2001.00361.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Bax is a pro-apoptotic Bcl-2 family protein that regulates programmed cell death through homodimerization and through heterodimerization with Bcl-2. Bax alpha is encoded by six exons and undergoes alternative splicing. Bax kappa, a splice variant of Bax with conserved BH1, BH2 and BH3 binding domains and a C-terminal transmembrane domain (TM), but with an extra 446-bp insert between exons 1 and 2 leading to loss of an N-terminal ART domain, was identified from an ischemic rat brain cDNA library. Expression of Bax kappa mRNA and protein was up-regulated in hippocampus after cerebral ischemic injury. The increased Bax kappa mRNA was distributed mainly in selectively vulnerable hippocampal CA1 neurons that are destined to die after global ischemia. Overexpression of Bax kappa protein in HN33 mouse hippocampal neuronal cells induced cell death, which was partially abrogated by co-overexpression of Bcl-2. Moreover, co-overexpression of Bax kappa and Bax alpha increased HN33 cell death. The results suggest that the Bax kappa may have a role in ischemic neuronal death.
Collapse
Affiliation(s)
- K L Jin
- Buck Institute for Age Research, Novato, California, USA.
| | | | | | | | | | | | | |
Collapse
|
31
|
Vivo C, Lévy F, Pilatte Y, Fleury-Feith J, Chrétien P, Monnet I, Kheuang L, Jaurand MC. Control of cell cycle progression in human mesothelioma cells treated with gamma interferon. Oncogene 2001; 20:1085-93. [PMID: 11314045 DOI: 10.1038/sj.onc.1204199] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2000] [Revised: 11/14/2000] [Accepted: 12/19/2000] [Indexed: 12/28/2022]
Abstract
Recombinant human interferon gamma (r-hu-IFNgamma) exerts both antitumoral activity in the early stages of human malignant mesothelioma and a cytostatic effect in human mesothelioma (HM) cell lines in vitro. The antiproliferative effect of interferons (IFNs) reported in a variety of cells has been attributed to several mechanisms. In order to progress in the understanding of HM cell growth modulation by r-hu-IFNgamma, modifications of cell cycle progression and expression of key cell cycle regulator proteins in response to r-hu-IFNgamma were examined. Nine HM cell lines were studied, including one resistant to the antiproliferative effect of r-hu-IFNgamma. Except in the resistant cell line r-hu-IFNgamma produced an arrest in the G1 and G2-M phases of the cell cycle, associated with a reduction in both cyclin A and cyclin dependent kinase inhibitors (CDKIs) expression. Moreover cyclin B1/cdc2 activity was decreased. The present study provides the first evidence of a G2-arrest in r-hu-IFNgamma-treated HM cell lines and indicates that HM cell lines, despite their tumorigenic origin still support cell cycle control. The cell cycle arrest induced by r-hu-IFNgamma seems to depend on cyclin regulation through p21(WAF1/CIP1)- and p27(Kip1)-independent mechanisms and is not directly related to the induced DNA damage.
Collapse
Affiliation(s)
- C Vivo
- INSERM E 99.09, Université Paris Val de Marne Paris XII (EA 2345), Faculté de Médecine, 8 rue du Général Sarrail, 94010, Créteil Cedex, France
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Corssmit EP, de Metz J, Sauerwein HP, Romijn JA. Biologic responses to IFN-alpha administration in humans. J Interferon Cytokine Res 2000; 20:1039-47. [PMID: 11152569 DOI: 10.1089/107999000750053690] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Although interferon-alpha (IFN-alpha) was discovered over 40 years ago, it was many years before it was registered as a therapeutic agent. Because of its unique qualities, it has been registered for both antiviral and antitumor indications. In addition to its therapeutic effects in viral diseases and cancer, IFN-alpha interferes with several important physiologic systems. It interacts with the immune system and affects several neuroendocrine and metabolic circuits. The specific mechanisms by which IFN-alpha exerts its therapeutic effects are complex, and it is very difficult to tie the biologic actions of IFN-alpha to specific clinical effects.
Collapse
Affiliation(s)
- E P Corssmit
- Department of Endocrinology, Academic Medical Center, University of Amsterdam, 1100 DE Amsterdam, The Netherlands.
| | | | | | | |
Collapse
|
33
|
Sancéau J, Hiscott J, Delattre O, Wietzerbin J. IFN-beta induces serine phosphorylation of Stat-1 in Ewing's sarcoma cells and mediates apoptosis via induction of IRF-1 and activation of caspase-7. Oncogene 2000; 19:3372-83. [PMID: 10918594 DOI: 10.1038/sj.onc.1203670] [Citation(s) in RCA: 94] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Four human cell lines derived from Ewing's sarcoma, EW-7, EW-1, COH and ORS, were investigated to establish the effects of human recombinant interferon-alpha2a and human recombinant interferon-beta on cell proliferation and apoptosis. All four cell lines were much more sensitive to the antiproliferative effects of IFN-beta than of IFN-alpha. Analysis of the early signals triggered by IFN-alpha and IFN-beta demonstrated that the two IFNs were similarly effective in inducing tyrosine phosphorylation of the Jak-1 and Tyk-2 kinases and the transcription factors Stat-1 and Stat-2. Interestingly, an additional rapid phosphorylation of Stat-1 on serine was observed after IFN-beta treatment, with concomitant activation of p38 mitogen-activated protein kinase. In these cells, Stat-1 Ser727 phosphorylation in response to IFN-beta was found to be impaired by p38 MAPkinase inhibitor (SB203580). IFN-beta induced the formation of the Interferon Stimulated Gene Factor 3 complex more efficiently than IFN-alpha, as well as sustained induction of IRF-1, which may account for its greater induction of 2'5'oligo(A)synthetase and greater inhibition of cell proliferation. IFN-beta, but not IFN-alpha, induced apoptosis in wild-type p53 EW-7 and COH cell lines, but not in the mutated p53 EW-1 or ORS cell lines. The apoptosis induced by IFN-beta in EW-7 and COH cell lines appeared to be mediated by IRF-1 and involved the activation of caspase-7. Ectopic expression of IRF-1 induced apoptosis in all four cell lines which correlated with the activation of caspase-7 and with the downregulation of the Bcl-2 oncoprotein, as observed for IFN-beta-induced apoptosis in parental EW-7 and COH cell lines.
Collapse
Affiliation(s)
- J Sancéau
- INSERM U 365, Institut Curie, Paris, France
| | | | | | | |
Collapse
|
34
|
Sun S, Sprent J. Role of type I interferons in T cell activation induced by CpG DNA. Curr Top Microbiol Immunol 2000; 247:107-17. [PMID: 10689782 DOI: 10.1007/978-3-642-59672-8_7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- S Sun
- R.W. Johnson Pharmaceutical Research Institute, San Diego, CA 92121, USA
| | | |
Collapse
|
35
|
Detjen KM, Welzel M, Farwig K, Brembeck FH, Kaiser A, Riecken EO, Wiedenmann B, Rosewicz S. Molecular mechanism of interferon alfa-mediated growth inhibition in human neuroendocrine tumor cells. Gastroenterology 2000; 118:735-48. [PMID: 10734025 DOI: 10.1016/s0016-5085(00)70143-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS Although human neuroendocrine tumors respond to interferon (IFN)-alpha treatment in vivo, the underlying mechanisms of growth inhibition are poorly understood. To characterize the antiproliferative effects at a molecular level, we explored the growth-regulatory action of IFN-alpha in the human neuroendocrine tumor cell lines BON and QGP1. METHODS IFN-alpha receptor expression and signal transduction were examined by reverse-transcription polymerase chain reaction, immunoblotting, subcellular fractionation, and transactivation assays. Growth regulation was evaluated by cell numbers, soft agar assays, and cell cycle analysis using flow cytometry. Expression and activity of cell cycle-regulatory molecules were determined by immunoblotting and histone H1-kinase assays. RESULTS Both cell lines expressed IFN-alpha receptor mRNA transcripts. Ligand binding initiated phosphorylation of Jak kinases and Stat transcription factors, resulting in Stat activation, nuclear translocation, and transcription from an ISRE-reporter construct. Prolonged IFN-alpha treatment dose-dependently inhibited both anchorage-dependent and -independent growth. Cell cycle analysis of IFN-alpha-treated, unsynchronized cultures revealed an increased S-phase population, which was further substantiated in G(1) synchronized QGP1 cells. IFN-alpha-treated cells entered S phase in parallel to control cultures, but their progress into G(2)/M phase was delayed. Both cellular cyclin B levels and CDC 2 activity were substantially reduced. The extent and time course of this reduction corresponded to the observed S-phase accumulation. CONCLUSIONS IFN-alpha directly inhibits growth of human neuroendocrine tumor cells by specifically delaying progression through S phase and into G(2)/M. These cell cycle changes are associated with inhibition of cyclin B expression, resulting in reduced CDC2 activity.
Collapse
Affiliation(s)
- K M Detjen
- Medizinische Klinik mit Schwerpunkt Hepatologie und Gastroenterologie, Universitätsklinikum Charité, Campus Virchow Klinikum, Humboldt Universität zu Berlin, Berlin, Germany
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Sprent J, Zhang X, Sun S, Tough D. T-cell proliferation in vivo and the role of cytokines. Philos Trans R Soc Lond B Biol Sci 2000; 355:317-22. [PMID: 10794049 PMCID: PMC1692740 DOI: 10.1098/rstb.2000.0568] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Unlike typical naive T cells, T cells with an activated (CD44hi) memory phenotype show a rapid rate of proliferation in vivo. The turnover of memory-phenotype CD8+ T cells can be considerably augmented by injecting mice with various compounds, including polyinosinic polycytidylic acid, lipopolysaccharide and immunostimulatory DNA (CpG DNA). Certain cytokines, notably type I (alpha, beta) interferons (IFN-I), have a similar effect. These agents appear to induce proliferation of CD44hi CD8+ cells in vivo by an indirect process involving production of effector cytokines, possibly interleukin-15, by antigen-presenting cells. Although none of the agents tested induces proliferation of naive-phenotype T cells, IFN-I has the capacity to cause upregulation of surface markers on purified naive T cells. Depending upon the experimental conditions used, IFN-I can either inhibit or enhance primary responses of naive T cells to specific antigen.
Collapse
Affiliation(s)
- J Sprent
- Department of Immunology, IMM4, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | | | | | | |
Collapse
|
37
|
Salzberg S, Vilchik S, Cohen S, Heller A, Kronfeld-Kinar Y. Expression of a PKR dominant-negative mutant in myogenic cells interferes with the myogenic process. Exp Cell Res 2000; 254:45-54. [PMID: 10623464 DOI: 10.1006/excr.1999.4721] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
In this study, we explored the possibility that PKR, a dsRNA-activated regulatory protein, is an essential component in the differentiation program of myogenic cells in vitro. For this purpose, we used a retroviral expression vector pMV7, harboring the PKR dominant-negative mutant PKRDelta6 (pMV7-p68Delta6). Murine C2C12 myogenic cells were transfected either with pMV7 or with pMV7-p68Delta6. Neomycin-resistant clones from both types were isolated and expanded and the results obtained with the representative clones C2-NEO (transfected with pMV7) and clone 17 and clone 22 (both transfected with pMV7-p68Delta6) are presented. In clone 17 and 22 cells, regardless of IFN treatment, a similar level of the transfected human p68 PKR mutant was detected. This protein was absent in C2-NEO cells. In parallel, in all types of cells, a low basal level of the endogenous murine p65 PKR protein was observed, which was further induced by IFN. However, PKR enzymatic activity was significantly induced by IFN only in C2-NEO cells, while it was hardly detected in both clones 17 and 22, even after IFN treatment. Furthermore, in contrast to C2-NEO cells, only a slight to moderate increase in enzymatic activity was observed in clone 17 and 22 differentiating cells. Next, cells were grown either in growth medium (GM) or differentiation medium (DM), and the progression of the myogenic program was studied. An inhibition in myotube formation in clone 17 versus C2-NEO cells cultivated in DM was clearly observed. Furthermore, while the growth rate and thymidine incorporation were reduced in C2-NEO cells grown in DM, both clone 17 and 22 cells were less affected under the same conditions. Similarly, a delay in the accumulation of the transcription factors MyoD and myogenin, as well as in creatine kinase activity and accumulation of troponin T, was detected in DM-cultivated clone 17 and clone 22 cells. Moreover, a delay in the induction of p21 (WAF1), in down-regulation of cyclin D1 and c-myc, and in the accumulation of the underphosphorylated form of pRb was also observed in clone 17 cells. We conclude that inhibition of endogenous PKR activity by a PKR dominant-negative mutant interferes with the myogenic program of murine C2C12 myogenic cells.
Collapse
Affiliation(s)
- S Salzberg
- Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 52900, Israel.
| | | | | | | | | |
Collapse
|
38
|
Sun S, Zhang X, Tough D, Sprent J. Multiple effects of immunostimulatory DNA on T cells and the role of type I interferons. SPRINGER SEMINARS IN IMMUNOPATHOLOGY 2000; 22:77-84. [PMID: 10944802 DOI: 10.1007/s002810000028] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
In addition to stimulating antigen-specific immune responses, infectious agents cause nonspecific activation of the innate immune system, notably up-regulation of costimulatory/adhesion molecules on APCs and cytokine production. In recent years it has become apparent that stimulation of the immune system by microorganisms is a property of a number of different cellular components, including DNA. As discussed earlier and elsewhere in this volume, the DNA of infectious agents--and indeed of all non-vertebrates tested--differs from mammalian DNA in being enriched for unmethylated CpG motifs. With appropriate flanking sequences, CpG DNA and synthetic CpG ODNs cause strong activation of APCs and other cells. In this article we have focussed on the capacity of CpG DNA/ODNs to alter T cell function. Whether these compounds act directly on T cells or function indirectly by activating other cells, especially APCs, is controversial [7, 8, 13, 14]. In contrast to other workers [8], we have yet to find definitive evidence that CpG DNA/ODNs can provide a co-stimulatory signal for purified T cells subjected to TCR ligation ([14] and unpublished data of authors). For this reason we lean to the notion that CpG DNA/ODNs modulate T cell function by inducing activation of APC rather than by acting directly on T cells. When injected in vivo in the absence of specific antigen, CpG DNA/ODNs have two striking effects on T cells, namely (1) induction of overt activation (proliferation) of memory-phenotype CD8+ cells, and (2) partial activation of all T cells, including naïve-phenotype T cells. Both actions of CpG DNA/ODNs are heavily dependent on the production of IFN-I by APC. For memory-phenotype (CD44hi) CD8+ cells, neither CpG DNA nor IFN-I can cause proliferation of purified APC-depleted T cells in vitro. Hence, under in vivo conditions, CpG DNA-induced proliferation of CD44hi CD8+ cells is probably mediated through the production of a secondary cytokine, i.e., by a cytokine that is directly stimulatory for CD44hi CD8+ cells. Based on the available evidence, it is highly likely that the effector cytokine is IL-15. With this assumption, our current model is that proliferation of CD44hi CD8+ cells induced by injection of CpG DNA/ODNs reflects production of IFN-I which, in turn, leads to synthesis of IL-15. Which particular cell types produce these two cytokines is unclear, although APCs are probably of prime importance. In addition to inducing proliferation of memory-phenotype CD8+ cells via IL-15, the IFN-I induced by CpG DNA/ODNs can also induce partial activation of naive T cells. This form of activation leads to up-regulation of CD69 and other molecules but does not cause entry into cell cycle. It is of interest that the partial activation of naive T cells induced by IFN-I is associated with decreased T proliferative responses. Thus, proliferation of purified naïve T cells elicited by combined TCR/CD28 ligation in vitro is greatly reduced by addition of IFN-I. This inhibitory effect of IFN-I does not influence cytokine production and probably reflects production of cell cycle inhibitors. Surprisingly, except at high doses, IFN-I fails to exert an anti-proliferative effect when T proliferative responses are driven by viable APCs. Indeed, in this situation, IFN-I enhances antigen-specific T proliferative responses, both in vivo and in vitro. This adjuvant effect of IFN-I is presumably a reflection of APC activation, but direct evidence on this issue is still lacking. In this article we have emphasized that contact with CpG DNA/ODNs has multiple effects on T cell function in vivo. Many of these effects seem to be related to the production of certain cytokines by APCs, notably IFN-I and IL-15. It should be stressed, however, that CpG DNA/ODNs probably lead to the production of many other cytokines. Hence, our current models of how CpG DNA/ODNs influence T cell function are undoubtedly oversimplified.
Collapse
Affiliation(s)
- S Sun
- R.W. Johnson Pharmaceutical Research Institute, La Jolla, CA 92121, USA
| | | | | | | |
Collapse
|
39
|
Smith A, Watson N, Sharma P. Frequency of trisomy 15 and loss of the Y chromosome in adult leukemia. CANCER GENETICS AND CYTOGENETICS 1999; 114:108-11. [PMID: 10549265 DOI: 10.1016/s0165-4608(99)00042-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In the interpretation of the varied and complex cytogenetic counts obtained in analysis of bone marrow (BM) samples for leukemia, loss or gain of certain chromosomes may or may not be significant for prognosis. Loss of the Y chromosome in elderly males is a benign finding. Trisomy 15 is rare and may represent another age-related abnormality, particularly in males, together with -Y. We reviewed 3,242 routine referrals sent to our laboratory for BM cytogenetics, over a period of 34 months. We detected 5 cases with uncomplicated trisomy 15, 3 in males and 2 in females. Three of these patients had the diagnosis of myelodysplastic syndrome (MDS). All 3 males showed a -Y cell line, although the 2 females did not have an X chromosome loss. All 5 patients were alive and well at times varying from 12 months to 4 years post-diagnosis. In the further analysis of our referral cohort, there were 62 males with loss of the Y chromosome as the sole abnormality, and of these 47 (76%), were referred with myeloid disease. The frequency of trisomy 15 in our laboratory was 1/475 referrals, but 1/292 in successful cultures from new patients. This is the first report providing frequency data for trisomy 15. Further data with longer term follow-up is required to establish the significance of trisomy 15 in elderly leukemic patients.
Collapse
Affiliation(s)
- A Smith
- Department of Cytogenetics, Royal Alexandra Hospital for Children, Westmead, Parramatta, Australia
| | | | | |
Collapse
|
40
|
Zella D, Barabitskaja O, Casareto L, Romerio F, Secchiero P, Reitz MS, Gallo RC, Weichold FF. Recombinant IFN-α (2b) Increases the Expression of Apoptosis Receptor CD95 and Chemokine Receptors CCR1 and CCR3 in Monocytoid Cells. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.163.6.3169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
IFN-α-2b, known as potent immune modulator, can either inhibit or enhance immune cell activity within the tightly regulated microenvironment of inflammation, depending upon the concentration of the cytokine and the activation stage of the cell. Chemokine receptors, which not only mediate chemotaxis of immune cells to the site of inflammation but also affect cellular activation by transferring corresponding signals, represent yet another level of immune regulation. Here we demonstrate that IFN-α increases the expression of CCR1 and CCR3 in primary mononuclear phagocytes, as well as in the monocytoid cell line U937. Enhanced receptor mRNA expression correlated with functional readouts such as increased intracellular calcium mobilization and cell migration in response to ligands. Expression of CCR2b, CCR4, CCR5, and CXCR4 was unchanged or decreased after IFN-α treatment. These observations indicate a differentially regulated cellular signaling relationship of IFN-α pathways and chemokine receptor expression. We also provide evidence that, under these conditions, IFN-α treatment increased the expression of CD95 (Fas, Apo1), resulting in enhanced susceptibility to apoptosis. Taken together, these data add important information for the rational application of IFN-α (2b) in immune and cancer therapies.
Collapse
Affiliation(s)
- Davide Zella
- *Institute of Human Virology, University of Maryland, Baltimore MD 21201
| | - Oxana Barabitskaja
- *Institute of Human Virology, University of Maryland, Baltimore MD 21201
| | - Luca Casareto
- †Laboratory of Basic Science, National Cancer Institute, National Institutes of Health, Bethesda MD 20814; and
| | - Fabio Romerio
- *Institute of Human Virology, University of Maryland, Baltimore MD 21201
| | - Paola Secchiero
- *Institute of Human Virology, University of Maryland, Baltimore MD 21201
- ‡Department of Morphology and Embriology, Human Anatomy Section, Ferrara, Italy
| | - Marvin S. Reitz
- *Institute of Human Virology, University of Maryland, Baltimore MD 21201
| | - Robert C. Gallo
- *Institute of Human Virology, University of Maryland, Baltimore MD 21201
| | - Frank F. Weichold
- *Institute of Human Virology, University of Maryland, Baltimore MD 21201
| |
Collapse
|
41
|
Akiyama M, Iwase S, Horiguchi-Yamada J, Saito S, Furukawa Y, Yamada O, Mizoguchi H, Ohno T, Yamada H. Interferon-alpha repressed telomerase along with G1-accumulation of Daudi cells. Cancer Lett 1999; 142:23-30. [PMID: 10424777 DOI: 10.1016/s0304-3835(99)00109-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The implications of telomerase on senescence and human carcinogenesis are widely accepted, but the changes of telomerase activity along with cell cycle modulation by anticancer treatment still remain obscure. In this paper, we issued whether the telomerase activity fluctuated along with cell cycle of cultured cancer cells using the antiproliferative effect of interferon-alpha (IFN-alpha). Daudi Burkitt lymphoma cells, treated with IFN-alpha, showed proliferation inhibition and cell cycle arrest at G1. The telomerase activity at 72 h was repressed to about 20% of control cells. Furthermore, after 72 h IFN-alpha treatment, the cells in G1 phase showed the marked decrease of telomerase activity, while cells in S and G2/M still possessed it. Among expressions of telomerase-related genes, only the catalytic subunit of telomerase (hTERT) decreased from 48 h, while the template RNA component (hTERC) and telomerase-associated protein 1 (TEP-1) were not affected. The downregulation of c-Myc preceded the change of hTERT. Moreover, the analysis of cells treated with IFN-alpha for 24 h revealed that cells in G1-to-S transition mainly expressed high hTERT, while S and G2/M cells had higher level of telomerase activity than that of G1 cells. These results indicate that (i) the expression of hTERT precedes the telomerase activity which is higher in S and G2/M phases than G1 phase, (ii) IFN-alpha repressed the telomerase activity in a cell cycle-dependent manner with the downregulation of hTERT.
Collapse
Affiliation(s)
- M Akiyama
- Department of Pediatrics, Institute of DNA Medicine, Jikei University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Hu R, Bekisz J, Hayes M, Audet S, Beeler J, Petricoin E, Zoon K. Divergence of Binding, Signaling, and Biological Responses to Recombinant Human Hybrid IFN. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.163.2.854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
Three human IFN-α hybrids, HY-1 [IFN-α21a(1-75)/α2c(76-165)], HY-2 [IFN-α21a(1-95)/α2c(96-165)], and HY-3 [IFN-α2c(1-95)/α21a(96-166)], were constructed, cloned, and expressed. The hybrids had comparable specific antiviral activities on Madin-Darby bovine kidney (MDBK)3 cells but exhibited very different antiproliferative and binding properties on human Daudi and WISH cells and primary human lymphocytes. Our data suggest that a portion of the N-terminal region of the molecule is important for interaction with components involved in binding of IFN-α2b while the C-terminal portion of IFN is critical for antiproliferative activity. A domain affecting the antiproliferative activity was found within the C-terminal region from amino acid residues 75–166. The signal transduction properties of HY-2 and HY-3 were evaluated by EMSA and RNase protection assays. Both HY-2 and HY-3 induced activation of STAT1 and 2. However, HY-2 exhibited essentially no antiproliferative effects at concentrations that activated STAT1 and 2. Additionally, at concentrations where no antiproliferative activity was seen, HY-2 induced a variety of IFN-responsive genes to the same degree as HY-3. RNase protection assays also indicate that, at concentrations where no antiproliferative activity was seen for HY-2, this construct retained the ability to induce a variety of IFN-inducible genes. These data suggest that the antiproliferative response may not be solely directed by the activation of the STAT1 and STAT2 pathway in the cells tested.
Collapse
Affiliation(s)
- Renqiu Hu
- *Division of Cytokine Biology, Office of Therapeutics Research and Review, and
| | - Joseph Bekisz
- *Division of Cytokine Biology, Office of Therapeutics Research and Review, and
| | - Mark Hayes
- *Division of Cytokine Biology, Office of Therapeutics Research and Review, and
| | - Susette Audet
- †Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Rockville, MD 20892
| | - Judy Beeler
- †Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Rockville, MD 20892
| | - Emanuel Petricoin
- *Division of Cytokine Biology, Office of Therapeutics Research and Review, and
| | - Kathryn Zoon
- *Division of Cytokine Biology, Office of Therapeutics Research and Review, and
| |
Collapse
|
43
|
Sangfelt O, Erickson S, Castro J, Heiden T, Gustafsson A, Einhorn S, Grandér D. Molecular mechanisms underlying interferon-alpha-induced G0/G1 arrest: CKI-mediated regulation of G1 Cdk-complexes and activation of pocket proteins. Oncogene 1999; 18:2798-810. [PMID: 10362250 DOI: 10.1038/sj.onc.1202609] [Citation(s) in RCA: 108] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
One prominent effect of IFNs is their cell growth-inhibitory activity. The mechanism behind this inhibition of proliferation is still not fully understood. In this study, the effect of IFN-alpha treatment on cell cycle progression has been analysed in three lymphoid cell lines, Daudi, U-266 and H9. Examination of the growth-arrested cell populations shows that Daudi cells accumulate in a G0-like state, whereas U-266 cells arrest later in G1. H9 cells are completely resistant to IFN-alpha's cell growth-inhibitory effects. The G0/G1-phase arrest is preceded by a rapid induction of the cyclin-dependent kinase inhibitors (CKIs), p21 and p15. In parallel, the activities of the G1 Cdks are significantly reduced. In addition to p21/p15 induction, IFN-alpha regulates the expression of another CKI, p27, presumably by a post-transcriptional mechanism. In the G1 Cdk-complexes, there is first an increased binding of p21 and p15 to their respective kinases. At longer exposure times, when Cdk-bound p15 and p21 decline, p27 starts to accumulate. Furthermore, we found that IFN-alpha not only suppresses the phosphorylation of pRb, but also alters the phosphorylation and expression of the other pocket proteins p130 and p107. These data suggest that induction of p21/p15 is involved in the primary IFN-alpha response inhibiting G1 Cdk activity, whereas increased p27 expression is part of a second set of events which keep these Cdks in their inactive form. Moreover, elevated levels of p27 correlated with a dissociation of cyclin E/Cdk2-p130 or p107 complexes to yield cyclin E/Cdk2-p27 complexes. In resistant H9 cells, which possess a homozygous deletion of the p15/p16 genes and lack p21 protein expression, IFN-alpha causes no detectable changes in p27 expression and, furthermore, no effects are observed on either pocket proteins in this cell line. Taken together, these data suggest that the early decline in G1 Cdk activity, subsequent changes in phosphorylation of pocket proteins, and G1/G0 arrest following IFN-alpha treatment, is not primarily due to loss of the G1 kinase components, but result from the inhibitory action of CKIs on these complexes.
Collapse
Affiliation(s)
- O Sangfelt
- Department of Oncology/Pathology, Karolinska Hospital and Institute, Stockholm, Sweden
| | | | | | | | | | | | | |
Collapse
|
44
|
Grandér D, Sangfelt O, Skoog L, Hansson J. In vivo induction of the interferon-stimulated protein 2'5'-oligoadenylate synthetase in tumor and peripheral blood cells during IFN-alpha treatment of metastatic melanoma. J Interferon Cytokine Res 1998; 18:691-5. [PMID: 9781807 DOI: 10.1089/jir.1998.18.691] [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] [Indexed: 11/13/2022] Open
Abstract
Interferon-alpha (IFN-alpha) therapy induces a response in a proportion of patients with metastatic melanoma. However, the mechanism of the antitumor action and reason(s) for resistance to IFN therapy are not known. To investigate whether lack of clinical response may be due to resistance of the melanoma cells to IFN-alpha or to an inability of IFN-alpha to reach the tumor cells during treatment, we investigated the in vivo and in vitro susceptibility of primary tumor cells obtained through fine needle aspiration biopsies and peripheral blood mononuclear cells (PBMC) to the induction of the IFN-induced enzyme 2'5'-oligoadenylate synthetase (2'5'OAS) during initiation of IFN-alpha therapy in 10 patients with metastatic melanoma. None of the patients showed an objective response to IFN-alpha treatment. The melanoma cells from 2 of the 10 patients were resistant to IFN-induced enhancement of 2'5'OAS in vitro. This correlated well with the in vivo induction of 2'5'OAS in the malignant cells, as no in vivo induction was seen in the 2 patients whose malignant cells were resistant in vitro, whereas tumor cells from 7 of 8 of the remaining patients showed enhancement also in vivo. This study shows that it is possible to monitor the cellular susceptibility of tumor cells to IFN-alpha in vivo and that melanoma cells from a small percentage of patients are resistant to the cellular effects of IFN-alpha. Furthermore, the absence of a clinical response to IFN-alpha therapy in the majority of melanoma patients can be explained neither by impaired cellular susceptibility to IFN nor by an inability of IFN-alpha to reach the tumor.
Collapse
Affiliation(s)
- D Grandér
- Department of Oncology-Pathology, Radiumhemmet, Karolinska Hospital, Stockholm, Sweden.
| | | | | | | |
Collapse
|
45
|
Selzman CH, Shames BD, Whitehill TA, Harken AH, McIntyre RC. Class II cytokine receptor ligands inhibit human vascular smooth muscle proliferation. Surgery 1998. [DOI: 10.1016/s0039-6060(98)70137-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
46
|
Koul D, Lapushin R, Xu HJ, Mills GB, Gutterman JU, Choubey D. p202 prevents apoptosis in murine AKR-2B fibroblasts. Biochem Biophys Res Commun 1998; 247:379-82. [PMID: 9642135 DOI: 10.1006/bbrc.1998.8804] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
p202 is an interferon (IFN)-inducible, primarily nuclear, phosphoprotein (52-kDa) whose overexpression in transfected cells inhibits colony formation. p202 binds to the retinoblastoma tumor suppressor protein and two other members of the pocket family proteins (p107 and p130). Moreover, overexpression of p202 in transfected cells inhibits the transcriptional activity of E2Fs (E2F-1/DP-1 and E2F-4/DP-1), p53, AP-1 c-Fos and c-Jun, NF-kappaB p50 and p65. Here we demonstrate that inhibition of endogenous p202 production in murine AKR-2B fibroblasts did not result in an increase in cell proliferation. Instead, these cells exhibited increased susceptibility to apoptosis in response to decrease in serum concentrations in the growth medium. These observations are consistent with the notion that normal levels of p202 may be needed for the regulation of cell proliferation.
Collapse
Affiliation(s)
- D Koul
- Department of Molecular Oncology, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas, 77030, USA
| | | | | | | | | | | |
Collapse
|
47
|
Kissil JL, Kimchi A. Death-associated proteins: from gene identification to the analysis of their apoptotic and tumour suppressive functions. MOLECULAR MEDICINE TODAY 1998; 4:268-74. [PMID: 9679246 DOI: 10.1016/s1357-4310(98)01263-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Aberrations of apoptosis are implicated in many diseases, including cancer, autoimmune disease, cardiovascular disease and neurodegeneration. The cell's apoptotic machinery is, therefore, an important potential target for the development of new therapies. Our laboratory has used a strategy called technical knockout (TKO) to identify novel genes involved in apoptosis. TKO is based on random inactivation of gene expression with antisense cDNA libraries, followed by selection of those cells that survive in the continuous presence of an apoptotic stimulus. Using this approach, we have isolated five novel genes, including a serine/threonine kinase, a nucleotide-binding protein and a homologue of the p220 translation initiation factor. Expression of one of these genes (DAP kinase) is lost in some cancers, and this loss appears to increase the metastatic potential of some tumours.
Collapse
|