1
|
Chen S, Li P, Shi K, Tang S, Zhang W, Peng C, Li T, Xie H, Liu C, Zhou J. Tanshinone IIA promotes ferroptosis in cutaneous melanoma via STAT1-mediated upregulation of PTGS2 expression. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156702. [PMID: 40222167 DOI: 10.1016/j.phymed.2025.156702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 03/10/2025] [Accepted: 03/26/2025] [Indexed: 04/15/2025]
Abstract
BACKGROUND Melanoma is highly aggressive, metastatic with a poor prognosis. Despite significant advances in targeted therapies and immunotherapies, their efficiency limited by drug resistance. Tanshinone IIA (Tan IIA), a bioactive compound derived from Traditional Chinese plant, exhibits significant anticancer potential, which still needs more research in its complex regulatory mechanisms. PURPOSE This study aimed to elucidate the putative targets and regulatory mechanisms of Tan IIA in anti-melanoma, with a focus on its role in inducing ferroptosis. STUDY DESIGN We designed the experiment to explore the effects of Tan IIA on melanoma through both in vitro and in vivo experiments and to investigate the underlying mechanisms through transcriptomics combining network pharmacology analysis. METHOD Ferroptosis monitored by Malondialdehyde (MDA), Fe2+, reactive oxygen species (ROS) and glutathione (GSH) in vivo and in vitro. RNA sequence was performed to explore the key regulatory pathways involved in Tan IIA-induced ferroptosis. Chromatin immunoprecipitation (ChIP) and Luciferase assays were used to validate transcription factor responsible for prostaglandin-endoperoxide synthase 2 (PTGS2) regulation. Additionally, RT-qPCR, western blot, IF, IHC were aimed to evaluate the expression of target gene. RESULT Tan IIA markedly suppresses melanoma growth in a xenograft model. The same effect performed on inhibition melanoma cells and promotion to ferroptosis with accumulation of ROS, MDA, and Fe²⁺levels and GSH consumption. RNA sequencing and public database analysis revealed that Tan IIA regulates PTGS2, the critical marker of ferroptosis, and PTGS2-knockdown attenuates Tan IIA -induced ferroptosis in melanoma cells. Furthermore, we identified that Tan IIA stimulate signal transducer and activator of transcription 1 (STAT1), a transcription factor, promoting PTGS2 expression and localized in the cell cytoplasm. Moreover, downregulation of the transcription factor STAT1 lead to PTGS2 downregulation and also inhibit ferroptosis in melanoma. CONCLUSION This study, the first to link Tan IIA-induced ferroptosis to the STAT1/PTGS2 axis in melanoma, identifies STAT1 and PTGS2 as novel therapeutic targets for melanoma, which demonstrates the potential of natural compounds Tan IIA in overcoming drug resistance and integrates traditional medicine with advanced molecular techniques for mechanistic exploration.
Collapse
Affiliation(s)
- Shuyue Chen
- Department of Plastic and Reconstructive Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Peiting Li
- Department of Plastic and Reconstructive Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Ke Shi
- Department of Plastic and Reconstructive Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Shijie Tang
- Department of Burns and Plastic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Wancong Zhang
- Department of Burns and Plastic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Cong Peng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Tianyu Li
- Department of Burns and Plastic Surgery, Nanshi Hospital of Nanyang, Nanyang, Henan Province 473000, China
| | - Huiqing Xie
- Department of Rehabilitation, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Can Liu
- Department of Plastic and Reconstructive Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China.
| | - Jianda Zhou
- Department of Plastic and Reconstructive Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China.
| |
Collapse
|
2
|
Nygrén P, Bouhlal J, Jokinen E, Forstén S, Laajala E, Dias D, Adnan-Awad S, Ianevski A, Klievink J, Lähteenmäki H, Kuusanmäki H, Myllymäki M, Kasanen T, Saeed K, Lee DA, iCAN Study Group, Hjorth-Hansen H, Aittokallio T, Dufva O, Mustjoki S. High-throughput drug screening identifies SMAC mimetics as enhancers of NK-cell cytotoxicity in chronic myeloid leukemia. Blood 2025; 145:1670-1686. [PMID: 39792962 PMCID: PMC12000656 DOI: 10.1182/blood.2024025286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 10/31/2024] [Accepted: 12/05/2024] [Indexed: 01/12/2025] Open
Abstract
ABSTRACT Natural killer (NK) cells have proven to be safe and effective immunotherapies, associated with favorable treatment responses in chronic myeloid leukemia (CML). Augmenting NK-cell function with oncological drugs could improve NK-cell-based immunotherapies. Here, we used a high-throughput drug screen consisting of >500 small-molecule compounds, to systematically evaluate the effects of oncological drugs on primary NK cells against CML cells. We identified second mitochondrially derived activator of caspases (SMAC) mimetics as potent enhancers of NK-cell cytotoxicity in both cell lines and primary patient samples. In contrast, several drug classes, including glucocorticoids and tyrosine kinase inhibitors such as dasatinib, inhibited NK-cell cytotoxicity. Single-cell RNA sequencing revealed drug-induced transcriptomic changes in both NK and target CML cells. SMAC mimetics upregulated NF-κB target genes in NK cells, potentially contributing to their enhanced cytotoxicity. Inhibitory drugs dexamethasone, dasatinib, and sotrastaurin prevented NK-cell transition to an activated state and suppressed the expression of interferon gamma (IFN-γ) by NK cells, thus preventing IFN-γ-mediated target cell transcriptomic response. In conclusion, we discovered that SMAC mimetics sensitize cancer cells to NK-cell-mediated killing, with potential clinical applications especially in patients with advanced phase CML.
Collapse
MESH Headings
- Humans
- Killer Cells, Natural/immunology
- Killer Cells, Natural/drug effects
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- High-Throughput Screening Assays
- Cytotoxicity, Immunologic/drug effects
- Antineoplastic Agents/pharmacology
- Mitochondrial Proteins
- Apoptosis Regulatory Proteins
- Cell Line, Tumor
- Intracellular Signaling Peptides and Proteins
Collapse
Affiliation(s)
- Petra Nygrén
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Jonas Bouhlal
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Emmi Jokinen
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Sofia Forstén
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Essi Laajala
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Diogo Dias
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland
| | - Shady Adnan-Awad
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Aleksandr Ianevski
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland
| | - Jay Klievink
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Hanna Lähteenmäki
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Heikki Kuusanmäki
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland
| | - Mikko Myllymäki
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Tiina Kasanen
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Khalid Saeed
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Dean A. Lee
- Division of Hematology, Oncology, and Blood and Marrow Transplant, Nationwide Children's Hospital, Columbus, OH
| | - iCAN Study Group
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland
- Division of Hematology, Oncology, and Blood and Marrow Transplant, Nationwide Children's Hospital, Columbus, OH
- Department of Hematology, St. Olavs Hospital, Trondheim, Norway
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Oslo Centre for Biostatistics and Epidemiology, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
| | | | - Tero Aittokallio
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Oslo Centre for Biostatistics and Epidemiology, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Olli Dufva
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Satu Mustjoki
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
- Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
3
|
Young AP, Denovan-Wright EM. Microglia-mediated neuron death requires TNF and is exacerbated by mutant Huntingtin. Pharmacol Res 2024; 209:107443. [PMID: 39362509 DOI: 10.1016/j.phrs.2024.107443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/28/2024] [Accepted: 09/28/2024] [Indexed: 10/05/2024]
Abstract
Microglia, the resident immune cells of the brain, regulate the balance of inflammation in the central nervous system under healthy and pathogenic conditions. Huntington's disease (HD) is a chronic neurodegenerative disease characterized by activated microglia and elevated concentrations of pro-inflammatory cytokines within the brain. Chronic hyperactivation of microglia is associated with brain pathology and eventual neuron death. However, it is unclear which specific cytokines are required for neuron death and whether HD neurons may be hypersensitive to neuroinflammation. We assessed the profile of microglia-secreted proteins in response to LPS and IFNγ, and a conditioned media paradigm was used to examine the effects of these secreted proteins on cultured neuronal cells. STHdhQ7/Q7 and STHdhQ111/Q111 neuronal cells were used to model wild-type and HD neurons, respectively. We determined that STHdhQ111/Q111 cells were hypersensitive to pro-inflammatory factors secreted by microglia, and that TNF was required to induce neuronal death. Microglia-mediated neuronal death could be effectively halted through the use of JAK-STAT or TNF inhibitors which supported the requirement for TNF as well as IFNγ in the process of secondary neurotoxicity. Further data derived from human HD patients as well as HD mice were suggestive of enhanced receptor density for TNF (TNFR1) and IFNγ (IFNGR) which could sensitize the HD brain to these cytokines. This highlights several potential mechanisms by which microglia may induce neuronal death and suggests that these mechanisms may be upregulated in the brain of HD patients.
Collapse
Affiliation(s)
- Alexander P Young
- Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada.
| | | |
Collapse
|
4
|
Rao D, Lacroix R, Rooker A, Gomes T, Stunnenberg JA, Valenti M, Dimitriadis P, Lin CP, de Bruijn B, Krijgsman O, Ligtenberg MA, Peeper DS, Blank CU. MeVa2.1.dOVA and MeVa2.2.dOVA: two novel BRAFV600E-driven mouse melanoma cell lines to study tumor immune resistance. Melanoma Res 2023; 33:12-26. [PMID: 36545919 DOI: 10.1097/cmr.0000000000000863] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
While immunotherapy has become standard-of-care for cutaneous melanoma patients, primary and acquired resistance prevent long-term benefits for about half of the late-stage patients. Pre-clinical models are essential to increase our understanding of the resistance mechanisms of melanomas, aiming to improve the efficacy of immunotherapy. Here, we present two novel syngeneic transplantable murine melanoma cell lines derived from the same primary tumor induced on BrafV600E Pten-/- mice: MeVa2.1 and MeVa2.2. Derivatives of these cell lines expressing the foreign antigen ovalbumin (dOVA) showed contrasting immune-mediated tumor control. MeVa2.2.dOVA melanomas were initially controlled in immune-competent hosts until variants grew out that had lost their antigens. By contrast, MeVa2.1.dOVA tumors were not controlled despite presenting the strong OVA antigen, as well as infiltration of tumor-reactive CD8+ T cells. MeVa2.1.dOVA displayed reduced sensitivity to T cell-mediated killing and growth inhibition in vitro by both IFN-γ and TNF-α. MeVa2.1.dOVA tumors were transiently controlled in vivo by either targeted therapy, adoptive T cell transfer, regulatory T cell depletion, or immune checkpoint blockade. MeVa2.1.dOVA could thus become a valuable melanoma model to evaluate novel immunotherapy combinations aiming to overcome immune resistance mechanisms.
Collapse
Affiliation(s)
- Disha Rao
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam
| | - Ruben Lacroix
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam
| | - Alex Rooker
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam
| | - Tainá Gomes
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam
| | - Johanna A Stunnenberg
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam
| | - Mesele Valenti
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam
| | - Petros Dimitriadis
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam
| | - Chun-Pu Lin
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam
| | - Beaunelle de Bruijn
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam
| | - Oscar Krijgsman
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam
| | - Maarten A Ligtenberg
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam
| | - Daniel S Peeper
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam
- Oncode Institute, Utrecht
| | - Christian U Blank
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam
- Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
5
|
Pretreatment of umbilical cord derived MSCs with IFN-γ and TNF-α enhances the tumor-suppressive effect on acute myeloid leukemia. Biochem Pharmacol 2022; 199:115007. [DOI: 10.1016/j.bcp.2022.115007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 03/09/2022] [Accepted: 03/13/2022] [Indexed: 01/27/2023]
|
6
|
Sun X, Bieber JM, Hammerlindl H, Chalkley RJ, Li KH, Burlingame AL, Jacobson MP, Wu LF, Altschuler SJ. Modulating environmental signals to reveal mechanisms and vulnerabilities of cancer persisters. SCIENCE ADVANCES 2022; 8:eabi7711. [PMID: 35089788 PMCID: PMC8797778 DOI: 10.1126/sciadv.abi7711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 12/08/2021] [Indexed: 06/14/2023]
Abstract
Cancer persister cells are able to survive otherwise lethal doses of drugs through nongenetic mechanisms, which can lead to cancer regrowth and drug resistance. The broad spectrum of molecular differences observed between persisters and their treatment-naïve counterparts makes it challenging to identify causal mechanisms underlying persistence. Here, we modulate environmental signals to identify cellular mechanisms that promote the emergence of persisters and to pinpoint actionable vulnerabilities that eliminate them. We found that interferon-γ (IFNγ) can induce a pro-persistence signal that can be specifically eliminated by inhibition of type I protein arginine methyltransferase (PRMT) (PRMTi). Mechanistic investigation revealed that signal transducer and activator of transcription 1 (STAT1) is a key component connecting IFNγ's pro-persistence and PRMTi's antipersistence effects, suggesting a previously unknown application of PRMTi to target persisters in settings with high STAT1 expression. Modulating environmental signals can accelerate the identification of mechanisms that promote and eliminate cancer persistence.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Lani F. Wu
- Corresponding author. (S.J.A.); (L.F.W.)
| | | |
Collapse
|
7
|
Freeman AJ, Vervoort SJ, Michie J, Ramsbottom KM, Silke J, Kearney CJ, Oliaro J. HOIP limits anti-tumor immunity by protecting against combined TNF and IFN-gamma-induced apoptosis. EMBO Rep 2021; 22:e53391. [PMID: 34467615 DOI: 10.15252/embr.202153391] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/11/2021] [Accepted: 08/18/2021] [Indexed: 12/15/2022] Open
Abstract
The success of cancer immunotherapy is limited to a subset of patients, highlighting the need to identify the processes by which tumors evade immunity. Using CRISPR/Cas9 screening, we reveal that melanoma cells lacking HOIP, the catalytic subunit of LUBAC, are highly susceptible to both NK and CD8+ T-cell-mediated killing. We demonstrate that HOIP-deficient tumor cells exhibit increased sensitivity to the combined effect of the inflammatory cytokines, TNF and IFN-γ, released by NK and CD8+ T cells upon target recognition. Both genetic deletion and pharmacological inhibition of HOIP augment tumor cell sensitivity to combined TNF and IFN-γ. Together, we unveil a protective regulatory axis, involving HOIP, which limits a transcription-dependent form of cell death that engages both intrinsic and extrinsic apoptotic machinery upon exposure to TNF and IFN-γ. Our findings highlight HOIP inhibition as a potential strategy to harness and enhance the killing capacity of TNF and IFN-γ during immunotherapy.
Collapse
Affiliation(s)
- Andrew J Freeman
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Vic., Australia
| | - Stephin J Vervoort
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Vic., Australia.,Translational Haematology Program, Peter MacCallum Cancer Centre, Melbourne, Vic., Australia
| | - Jessica Michie
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Vic., Australia
| | - Kelly M Ramsbottom
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
| | - John Silke
- Inflammation Department, Walter and Eliza Hall Institute of Medical Research, Parkville, Vic., Australia.,Department of Medical Biology, The University of Melbourne, Melbourne, Vic., Australia
| | - Conor J Kearney
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Vic., Australia.,Translational Haematology Program, Peter MacCallum Cancer Centre, Melbourne, Vic., Australia
| | - Jane Oliaro
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Vic., Australia.,Department of Immunology and Pathology, Monash University, Melbourne, Vic., Australia
| |
Collapse
|
8
|
Wang Y, Liu R, Liao J, Jiang L, Jeong GH, Zhou L, Polite M, Duong D, Seyfried NT, Wang H, Kiyokawa H, Yin J. Orthogonal ubiquitin transfer reveals human papillomavirus E6 downregulates nuclear transport to disarm interferon-γ dependent apoptosis of cervical cancer cells. FASEB J 2021; 35:e21986. [PMID: 34662469 DOI: 10.1096/fj.202101232rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 02/05/2023]
Abstract
The E6 protein of the human papillomavirus (HPV) underpins important protein interaction networks between the virus and host to promote viral infection. Through its interaction with E6AP, a host E3 ubiquitin (UB) ligase, E6 stirs the protein ubiquitination pathways toward the oncogenic transformation of the infected cells. For a systematic measurement of E6 reprogramming of the substrate pool of E6AP, we performed a proteomic screen based on "orthogonal UB transfer (OUT)" that allowed us to identify the ubiquitination targets of E6AP dependent on the E6 protein of HPV-16, a high-risk viral subtype for the development of cervical cancer. The OUT screen identified more than 200 potential substrates of the E6-E6AP pair based on the transfer of UB from E6AP to the substrate proteins. Among them, we verified that E6 would induce E6AP-catalyzed ubiquitination of importin proteins KPNA1-3, protein phosphatase PGAM5, and arginine methyltransferases CARM1 to trigger their degradation by the proteasome. We further found that E6 could significantly reduce the cellular level of KPNA1 that resulted in the suppression of nuclear transport of phosphorylated STAT1 and the inhibition of interferon-γ-induced apoptosis in cervical cancer cells. Overall, our work demonstrates OUT as a powerful proteomic platform to probe the interaction of E6 and host cells through protein ubiquitination and reveals a new role of E6 in down-regulating nuclear transport proteins to attenuate tumor-suppressive signaling.
Collapse
Affiliation(s)
- Yiyang Wang
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, China
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, USA
| | - Ruochuan Liu
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, USA
| | - Jia Liao
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, China
| | - Lucen Jiang
- Department of Pathology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Geon H Jeong
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, USA
| | - Li Zhou
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, USA
| | - Monica Polite
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, USA
| | - Duc Duong
- Integrated Proteomics Core, Emory University, Atlanta, Georgia, USA
| | - Nicholas T Seyfried
- Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Huadong Wang
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, China
| | - Hiroaki Kiyokawa
- Department of Pharmacology, Northwestern University, Chicago, Illinois, USA
| | - Jun Yin
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, USA
| |
Collapse
|
9
|
Lee HS, Lee IH, Kang K, Park SI, Jung M, Yang SG, Kwon TW, Lee DY. A Comprehensive Understanding of the Anticancer Mechanisms of FDY2004 Against Cervical Cancer Based on Network Pharmacology. Nat Prod Commun 2021. [DOI: 10.1177/1934578x211004304] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Herbal drugs are continuously being developed and used as effective therapeutics for various cancers, such as cervical cancer (CC); however, their mechanisms of action at a systemic level have not been explored fully. To study such mechanisms, we conducted a network pharmacological investigation of the anti-CC mechanisms of FDY2004, an herbal drug consisting of Moutan Radicis Cortex, Persicae Semen , and Rhei Radix et Rhizoma. We found that FDY2004 inhibited the viability of human CC cells. By performing pharmacokinetic evaluation and network analysis of the phytochemical components of FDY2004, we identified 29 bioactive components and their 116 CC-associated pharmacological targets. Gene ontology enrichment analysis showed that the modulation of cellular functions, such as apoptosis, growth, proliferation, and survival, might be mediated through the FDY2004 targets. The therapeutic targets were also key components of CC-associated oncogenic and tumor-suppressive pathways, including PI3K-Akt, human papillomavirus infection, IL-17, MAPK, TNF, focal adhesion, and viral carcinogenesis pathways. In conclusion, our data present a comprehensive insight for the mechanisms of the anti-CC properties of FDY2004.
Collapse
Affiliation(s)
- Ho-Sung Lee
- The Fore, Songpa-gu, Seoul, Republic of Korea
- Forest Hospital, Songpa-gu, Seoul, Republic of Korea
| | - In-Hee Lee
- The Fore, Songpa-gu, Seoul, Republic of Korea
| | - Kyungrae Kang
- Forest Hospital, Songpa-gu, Seoul, Republic of Korea
| | - Sang-In Park
- Forestheal Hospital, Songpa-gu, Seoul, Republic of Korea
| | - Minho Jung
- Forest Hospital, Songpa-gu, Seoul, Republic of Korea
| | - Seung Gu Yang
- Kyunghee Naro Hospital, Bundang-gu, Seongnam, Republic of Korea
| | - Tae-Wook Kwon
- Forest Hospital, Songpa-gu, Seoul, Republic of Korea
| | - Dae-Yeon Lee
- The Fore, Songpa-gu, Seoul, Republic of Korea
- Forest Hospital, Songpa-gu, Seoul, Republic of Korea
| |
Collapse
|
10
|
Kauffman MR, Nazemidashtarjandi S, Ghazanfari D, Allen AE, Reynolds NM, Faik A, Burdick MM, McCall KD, Goetz DJ. Evidence that knock down of GSK-3β in Chronic Myelogenous Leukemia cells augments IFN-γ-induced apoptosis. Leuk Res 2020; 99:106464. [PMID: 33130330 PMCID: PMC7740760 DOI: 10.1016/j.leukres.2020.106464] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 10/15/2020] [Accepted: 10/17/2020] [Indexed: 12/25/2022]
Abstract
The role of interferon-gamma (IFN-γ) in Chronic Myelogenous/Myeloid Leukemia (CML) and in the treatment of CML remains unclear; specifically, the effect of IFN-γ on apoptosis. There is reported interplay between IFN-γ and glycogen synthase kinase-3 (GSK-3), a kinase which has been implicated in both cell death and, conversely, cell survival. Thus, we utilized the CML-derived HAP1 cell line and a mutant HAP1 GSK-3β knocked-down cell line (GSK-3β 31bp) to investigate whether GSK-3 modulates IFN-γ's action on CML cells. Significantly less GSK-3β 31bp cells, relative to HAP1 cells, were present after 48 h treatment with IFN-γ. IFN-γ treatment significantly decreased GSK-3β 31bp substrate adhesiveness (relative to HAP1 cells); an observation often correlated with cell death. Fluorescence microscopy revealed that IFN-γ induces a modest level of apoptosis in the HAP1 cells and that IFN-γ induced apoptosis is significantly enhanced in GSK-3β 31bp cells. Utilizing a complementary GSK-3β knocked-down cell line (8bp) we found, via flow cytometric analysis, that IFN-γ induced apoptosis is significantly enhanced in GSK-3β 8bp cells relative to HAP1 cells. Combined, our findings suggest that IFN-γ induces apoptosis of CML cells and that loss of GSK-3β significantly augments IFN-γ-induced apoptosis.
Collapse
MESH Headings
- Antineoplastic Agents/pharmacology
- Apoptosis/drug effects
- CRISPR-Cas Systems
- Cell Adhesion/drug effects
- Cell Line, Tumor/drug effects
- Codon, Nonsense
- Drug Interactions
- Flow Cytometry
- Frameshift Mutation
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Gene Knockdown Techniques
- Glycogen Synthase Kinase 3 beta/antagonists & inhibitors
- Glycogen Synthase Kinase 3 beta/genetics
- Humans
- Interferon-gamma/pharmacology
- Interferon-gamma/therapeutic use
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Male
- Neoplasm Proteins/antagonists & inhibitors
- Protein Kinase Inhibitors/pharmacology
- Spectrometry, Fluorescence
- RNA, Guide, CRISPR-Cas Systems
Collapse
Affiliation(s)
- Melissa R Kauffman
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH 45701, USA; Biomedical Engineering Program, Ohio University, Athens, OH 45701, USA
| | | | - Davoud Ghazanfari
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH 45701, USA
| | - Abigail E Allen
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH 45701, USA; Biomedical Engineering Program, Ohio University, Athens, OH 45701, USA
| | - Nathan M Reynolds
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH 45701, USA; Translational Biomedical Sciences Program, Ohio University, Athens, OH 45701, USA
| | - Ahmed Faik
- Environmental and Plant Biology Department, Ohio University, Athens, OH 45701, USA; Molecular and Cellular Biology Program, Ohio University, Athens, OH 45701, USA
| | - Monica M Burdick
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH 45701, USA; Biomedical Engineering Program, Ohio University, Athens, OH 45701, USA; Molecular and Cellular Biology Program, Ohio University, Athens, OH 45701, USA; Translational Biomedical Sciences Program, Ohio University, Athens, OH 45701, USA
| | - Kelly D McCall
- Biomedical Engineering Program, Ohio University, Athens, OH 45701, USA; Molecular and Cellular Biology Program, Ohio University, Athens, OH 45701, USA; Translational Biomedical Sciences Program, Ohio University, Athens, OH 45701, USA; Department of Specialty Medicine, Ohio University, Athens, OH 45701, USA; The Diabetes Institute, Ohio University, Athens, OH 45701, USA
| | - Douglas J Goetz
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH 45701, USA; Biomedical Engineering Program, Ohio University, Athens, OH 45701, USA; Molecular and Cellular Biology Program, Ohio University, Athens, OH 45701, USA; Translational Biomedical Sciences Program, Ohio University, Athens, OH 45701, USA.
| |
Collapse
|
11
|
Martí Navia A, Dal Ben D, Lambertucci C, Spinaci A, Volpini R, Marques-Morgado I, Coelho JE, Lopes LV, Marucci G, Buccioni M. Adenosine Receptors as Neuroinflammation Modulators: Role of A 1 Agonists and A 2A Antagonists. Cells 2020; 9:cells9071739. [PMID: 32708189 PMCID: PMC7409197 DOI: 10.3390/cells9071739] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 07/17/2020] [Indexed: 01/07/2023] Open
Abstract
The pathological condition of neuroinflammation is caused by the activation of the neuroimmune cells astrocytes and microglia. The autacoid adenosine seems to be an important neuromodulator in this condition. Its main receptors involved in the neuroinflammation modulation are A1AR and A2AAR. Evidence suggests that A1AR activation produces a neuroprotective effect and A2AARs block prevents neuroinflammation. The aim of this work is to elucidate the effects of these receptors in neuroinflammation using the partial agonist 2'-dCCPA (2-chloro-N6-cyclopentyl-2'-deoxyadenosine) (C1 KiA1AR = 550 nM, KiA2AAR = 24,800 nM, and KiA3AR = 5560 nM, α = 0.70, EC50A1AR = 832 nM) and the newly synthesized in house compound 8-chloro-9-ethyl-2-phenethoxyadenine (C2 KiA2AAR = 0.75 nM; KiA1AR = 17 nM and KiA3AR = 227 nM, IC50A2AAR = 251 nM unpublished results). The experiments were performed in in vitro and in in vivo models of neuroinflammation. Results showed that C1 was able to prevent the inflammatory effect induced by cytokine cocktail (TNF-α, IL-1β, and IFN-γ) while C2 possess both anti-inflammatory and antioxidant properties, counteracting both neuroinflammation in mixed glial cells and in an animal model of neuroinflammation. In conclusion, C2 is a potential candidate for neuroinflammation therapy.
Collapse
Affiliation(s)
- Aleix Martí Navia
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, 62032 Camerino (MC), Italy; (A.M.N.); (D.D.B.); (C.L.); (A.S.); (R.V.); (M.B.)
| | - Diego Dal Ben
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, 62032 Camerino (MC), Italy; (A.M.N.); (D.D.B.); (C.L.); (A.S.); (R.V.); (M.B.)
| | - Catia Lambertucci
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, 62032 Camerino (MC), Italy; (A.M.N.); (D.D.B.); (C.L.); (A.S.); (R.V.); (M.B.)
| | - Andrea Spinaci
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, 62032 Camerino (MC), Italy; (A.M.N.); (D.D.B.); (C.L.); (A.S.); (R.V.); (M.B.)
| | - Rosaria Volpini
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, 62032 Camerino (MC), Italy; (A.M.N.); (D.D.B.); (C.L.); (A.S.); (R.V.); (M.B.)
| | - Inês Marques-Morgado
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, 1649-028 Lisbon, Portugal; (I.M.-M.); (J.E.C.); (L.V.L.)
| | - Joana E. Coelho
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, 1649-028 Lisbon, Portugal; (I.M.-M.); (J.E.C.); (L.V.L.)
| | - Luísa V. Lopes
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, 1649-028 Lisbon, Portugal; (I.M.-M.); (J.E.C.); (L.V.L.)
| | - Gabriella Marucci
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, 62032 Camerino (MC), Italy; (A.M.N.); (D.D.B.); (C.L.); (A.S.); (R.V.); (M.B.)
- Correspondence: ; Tel.: +39-073-7402-223
| | - Michela Buccioni
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, 62032 Camerino (MC), Italy; (A.M.N.); (D.D.B.); (C.L.); (A.S.); (R.V.); (M.B.)
| |
Collapse
|
12
|
Wang LY, Li RL, Guo M, Huang LX, Chen YM. Long Noncoding RNA HULC Promoter Polymorphism rs1041279 Is Associated with an Increased Risk of Cervical Squamous Cell Carcinoma. Reprod Sci 2020; 27:93-99. [PMID: 32046387 DOI: 10.1007/s43032-019-00013-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 02/26/2019] [Indexed: 12/12/2022]
Abstract
Hepatocellular carcinoma upregulated long noncoding RNA (HULC), identified as an oncogene in cervical cancer, is involved in not only the clinical stage, lymph node metastasis, and depth of cervical invasion but also outcome. In this study, we aimed to investigate the association between 3 polymorphisms (i.e., rs1041279, rs3005167, and rs7770772) in the promoter of HULC and the risk of cervical squamous cell carcinoma (CSCC). The polymorphisms were genotyped using the multiplex ligase detection reaction assay. The promoter activity was measured using the dual-luciferase reporter assay kit. The rs1041279 GG genotype and G allele revealed a significantly higher risk of CSCC compared with the rs1041279 CC genotype and C allele (GG vs. CC, adjusted OR = 1.79, 95% CI, 1.17-2.73, P = 0.007; G vs. C, adjusted OR = 1.36, 95% CI, 1.09-1.69, P = 0.006). Haplotype analysis revealed that the rs3005167C-rs7770772G-rs1041279C or rs3005167C-rs7770772G-rs1041279G haplotype had a significantly higher risk of CSCC compared to the rs3005167G-rs7770772G-rs1041279C haplotype (CGC vs. GGC, OR = 2.38, 95% CI, 1.53-3.75, P < 0.001; CGG vs. GGC, OR = 3.76, 95% CI, 2.12-6.68, P < 0.001). Dual-luciferase reporter assay showed that the rs1041279 G promoter resulted in higher transcriptional activity compared with the rs1041279 C (P < 0.01). Additionally, the rs1041279 GG genotype carriers had an increased level of HULC expression (P = 0.03). These findings suggest that the HULC rs1041279 may be a useful marker for the etiology of CSCC.
Collapse
Affiliation(s)
- Long-Yi Wang
- Department of Obstetrics and Gynecology, Wenzhou People's Hospital, Wenzhou, 325000, People's Republic of China
| | - Ren-Liang Li
- Department of Obstetrics and Gynecology, Wenzhou People's Hospital, Wenzhou, 325000, People's Republic of China.
| | - Min Guo
- Department of Obstetrics and Gynecology, Wenzhou People's Hospital, Wenzhou, 325000, People's Republic of China
| | - Ling-Xiao Huang
- Department of Obstetrics and Gynecology, Wenzhou People's Hospital, Wenzhou, 325000, People's Republic of China
| | - Yu-Mei Chen
- Department of Obstetrics and Gynecology, Wenzhou People's Hospital, Wenzhou, 325000, People's Republic of China
| |
Collapse
|
13
|
Cardenas H, Jiang G, Thomes Pepin J, Parker JB, Condello S, Nephew KP, Nakshatri H, Chakravarti D, Liu Y, Matei D. Interferon-γ signaling is associated with BRCA1 loss-of-function mutations in high grade serous ovarian cancer. NPJ Precis Oncol 2019; 3:32. [PMID: 31840082 PMCID: PMC6897992 DOI: 10.1038/s41698-019-0103-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 10/29/2019] [Indexed: 12/17/2022] Open
Abstract
Loss-of-function mutations of the breast cancer type 1 susceptibility protein (BRCA1) are associated with breast (BC) and ovarian cancer (OC). To identify gene signatures regulated by epigenetic mechanisms in OC cells carrying BRCA1 mutations, we assessed cellular responses to epigenome modifiers and performed genome-wide RNA- and chromatin immunoprecipitation-sequencing in isogenic OC cells UWB1.289 (carrying a BRCA1 mutation, BRCA1-null) and UWB1.289 transduced with wild-type BRCA1 (BRCA1+). Increased sensitivity to histone deacetylase inhibitors (HDACi) was observed in BRCA1-null vs. BRCA1+ cells. Gene expression profiles of BRCA1-null vs. BRCA1+ cells and treated with HDACi were integrated with chromatin mapping of histone H3 lysine 9 or 27 acetylation. Gene networks activated in BRCA1-null vs. BRCA1 + OC cells related to cellular movement, cellular development, cellular growth and proliferation, and activated upstream regulators included TGFβ1, TNF, and IFN-γ. The IFN-γ pathway was altered by HDACi in BRCA1+ vs. BRCA1-null cells, and in BRCA1-mutated/or low vs. BRCA1-normal OC tumors profiled in the TCGA. Key IFN-γ-induced genes upregulated at baseline in BRCA1-null vs. BRCA1+OC and BC cells included CXCL10, CXCL11, and IFI16. Increased localization of STAT1 in the promoters of these genes occurred in BRCA1-null OC cells, resulting in diminished responses to IFN-γ or to STAT1 knockdown. The IFN-γ signature was associated with improved survival among OC patients profiled in the TCGA. In all, our results support that changes affecting IFN-γ responses are associated with inactivating BRCA1 mutations in OC. This signature may contribute to altered responses to anti-tumor immunity in BRCA1-mutated cells or tumors.
Collapse
Affiliation(s)
- Horacio Cardenas
- Department of Obstetrics and Gynecology, Northwestern University, Chicago, IL USA
| | - Guanglong Jiang
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, IN USA
- Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN USA
| | - Jessica Thomes Pepin
- Department of Obstetrics and Gynecology, Indiana University, Indianapolis, IN USA
| | - J. Brandon Parker
- Department of Obstetrics and Gynecology, Northwestern University, Chicago, IL USA
| | - Salvatore Condello
- Department of Obstetrics and Gynecology, Northwestern University, Chicago, IL USA
| | - Kenneth P. Nephew
- Department of Obstetrics and Gynecology, Indiana University, Indianapolis, IN USA
- Melvin and Bren Simon Cancer Center, Indianapolis, IN USA
- Medical Sciences, Indiana University School of Medicine, Bloomington, IN USA
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN USA
| | - Harikrishna Nakshatri
- Melvin and Bren Simon Cancer Center, Indianapolis, IN USA
- Departments of Surgery, Biochemistry and Molecular Biology, Indiana University, Indianapolis, IN USA
| | - Debabrata Chakravarti
- Department of Obstetrics and Gynecology, Northwestern University, Chicago, IL USA
- Robert H Lurie Comprehensive Cancer Center, Chicago, IL USA
| | - Yunlong Liu
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, IN USA
- Melvin and Bren Simon Cancer Center, Indianapolis, IN USA
| | - Daniela Matei
- Department of Obstetrics and Gynecology, Northwestern University, Chicago, IL USA
- Robert H Lurie Comprehensive Cancer Center, Chicago, IL USA
- Jesse Brown VA Medical Center, Chicago, IL USA
| |
Collapse
|
14
|
Xie M, Zheng H, Madan-Lala R, Dai W, Gimbrone NT, Chen Z, Kinose F, Blackstone SA, Smalley KSM, Cress WD, Haura EB, Rix U, Beg AA. MEK Inhibition Modulates Cytokine Response to Mediate Therapeutic Efficacy in Lung Cancer. Cancer Res 2019; 79:5812-5825. [PMID: 31362929 DOI: 10.1158/0008-5472.can-19-0698] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 06/19/2019] [Accepted: 07/23/2019] [Indexed: 11/16/2022]
Abstract
Activating mutations in BRAF, a key mediator of RAS signaling, are present in approximately 50% of melanoma patients. Pharmacologic inhibition of BRAF or the downstream MAP kinase MEK is highly effective in treating BRAF-mutant melanoma. In contrast, RAS pathway inhibitors have been less effective in treating epithelial malignancies, such as lung cancer. Here, we show that treatment of melanoma patients with BRAF and MEK inhibitors (MEKi) activated tumor NF-κB activity. MEKi potentiated the response to TNFα, a potent activator of NF-κB. In both melanoma and lung cancer cells, MEKi increased cell-surface expression of TNFα receptor 1 (TNFR1), which enhanced NF-κB activation and augmented expression of genes regulated by TNFα and IFNγ. Screening of 289 targeted agents for the ability to increase TNFα and IFNγ target gene expression demonstrated that this was a general activity of inhibitors of MEK and ERK kinases. Treatment with MEKi led to acquisition of a novel vulnerability to TNFα and IFNγ-induced apoptosis in lung cancer cells that were refractory to MEKi killing and augmented cell-cycle arrest. Abolishing the expression of TNFR1 on lung cancer cells impaired the antitumor efficacy of MEKi, whereas the administration of TNFα and IFNγ in MEKi-treated mice enhanced the antitumor response. Furthermore, immunotherapeutics known to induce expression of these cytokines synergized with MEKi in eradicating tumors. These findings define a novel cytokine response modulatory function of MEKi that can be therapeutically exploited. SIGNIFICANCE: Lung cancer cells are rendered sensitive to MEK inhibitors by TNFα and IFNγ, providing a strong mechanistic rationale for combining immunotherapeutics, such as checkpoint blockers, with MEK inhibitor therapy for lung cancer.See related commentary by Havel, p. 5699.
Collapse
Affiliation(s)
- Mengyu Xie
- Department of Immunology, Moffitt Cancer Center, Tampa, Florida.,Cancer Biology PhD Program, University of South Florida, Tampa, Florida
| | - Hong Zheng
- Department of Immunology, Moffitt Cancer Center, Tampa, Florida
| | | | - Wenjie Dai
- Department of Immunology, Moffitt Cancer Center, Tampa, Florida
| | - Nicholas T Gimbrone
- Cancer Biology PhD Program, University of South Florida, Tampa, Florida.,Department of Molecular Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Zhihua Chen
- Department of Bioinformatics, Moffitt Cancer Center, Tampa, Florida
| | - Fumi Kinose
- Department of Thoracic Oncology, Moffitt Cancer Center, Tampa, Florida
| | | | | | - W Douglas Cress
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, Florida.,Department of Thoracic Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Eric B Haura
- Department of Thoracic Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Uwe Rix
- Department of Thoracic Oncology, Moffitt Cancer Center, Tampa, Florida.,Department of Drug Discovery, Moffitt Cancer Center, Tampa, Florida
| | - Amer A Beg
- Department of Immunology, Moffitt Cancer Center, Tampa, Florida. .,Department of Thoracic Oncology, Moffitt Cancer Center, Tampa, Florida
| |
Collapse
|
15
|
Benbenishty A, Gadrich M, Cottarelli A, Lubart A, Kain D, Amer M, Shaashua L, Glasner A, Erez N, Agalliu D, Mayo L, Ben-Eliyahu S, Blinder P. Prophylactic TLR9 stimulation reduces brain metastasis through microglia activation. PLoS Biol 2019; 17:e2006859. [PMID: 30921319 PMCID: PMC6469801 DOI: 10.1371/journal.pbio.2006859] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Revised: 04/17/2019] [Accepted: 02/28/2019] [Indexed: 02/07/2023] Open
Abstract
Brain metastases are prevalent in various types of cancer and are often terminal, given the low efficacy of available therapies. Therefore, preventing them is of utmost clinical relevance, and prophylactic treatments are perhaps the most efficient strategy. Here, we show that systemic prophylactic administration of a toll-like receptor (TLR) 9 agonist, CpG-C, is effective against brain metastases. Acute and chronic systemic administration of CpG-C reduced tumor cell seeding and growth in the brain in three tumor models in mice, including metastasis of human and mouse lung cancer, and spontaneous melanoma-derived brain metastasis. Studying mechanisms underlying the therapeutic effects of CpG-C, we found that in the brain, unlike in the periphery, natural killer (NK) cells and monocytes are not involved in controlling metastasis. Next, we demonstrated that the systemically administered CpG-C is taken up by endothelial cells, astrocytes, and microglia, without affecting blood-brain barrier (BBB) integrity and tumor brain extravasation. In vitro assays pointed to microglia, but not astrocytes, as mediators of CpG- C effects through increased tumor killing and phagocytosis, mediated by direct microglia-tumor contact. In vivo, CpG-C-activated microglia displayed elevated mRNA expression levels of apoptosis-inducing and phagocytosis-related genes. Intravital imaging showed that CpG-C-activated microglia cells contact, kill, and phagocytize tumor cells in the early stages of tumor brain invasion more than nonactivated microglia. Blocking in vivo activation of microglia with minocycline, and depletion of microglia with a colony-stimulating factor 1 inhibitor, indicated that microglia mediate the antitumor effects of CpG-C. Overall, the results suggest prophylactic CpG-C treatment as a new intervention against brain metastasis, through an essential activation of microglia.
Collapse
Affiliation(s)
- Amit Benbenishty
- School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
- Neurobiology Department, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Meital Gadrich
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- School for Molecular Cell Biology & Biotechnology, Tel Aviv University, Tel Aviv, Israel
| | - Azzurra Cottarelli
- Department of Neurology, Columbia University Medical Center, New York, New York, United States of America
| | - Alisa Lubart
- Neurobiology Department, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - David Kain
- Neurobiology Department, Tel Aviv University, Tel Aviv, Israel
| | - Malak Amer
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Lee Shaashua
- School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ariella Glasner
- The Lautenberg Centre for General and Tumor Immunology, The Hebrew University Hadassah Medical School, Jerusalem, Israel
| | - Neta Erez
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dritan Agalliu
- Department of Neurology, Columbia University Medical Center, New York, New York, United States of America
| | - Lior Mayo
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- School for Molecular Cell Biology & Biotechnology, Tel Aviv University, Tel Aviv, Israel
| | - Shamgar Ben-Eliyahu
- School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Pablo Blinder
- Neurobiology Department, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
16
|
Lo UG, Pong RC, Yang D, Gandee L, Hernandez E, Dang A, Lin CJ, Santoyo J, Ma S, Sonavane R, Huang J, Tseng SF, Moro L, Arbini AA, Kapur P, Raj GV, He D, Lai CH, Lin H, Hsieh JT. IFNγ-Induced IFIT5 Promotes Epithelial-to-Mesenchymal Transition in Prostate Cancer via miRNA Processing. Cancer Res 2018; 79:1098-1112. [PMID: 30504123 DOI: 10.1158/0008-5472.can-18-2207] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 10/23/2018] [Accepted: 11/27/2018] [Indexed: 11/16/2022]
Abstract
IFNγ, a potent cytokine known to modulate tumor immunity and tumoricidal effects, is highly elevated in patients with prostate cancer after radiation. In this study, we demonstrate that IFNγ can induce epithelial-to-mesenchymal transition (EMT) in prostate cancer cells via the JAK-STAT signaling pathway, leading to the transcription of IFN-stimulated genes (ISG) such as IFN-induced tetratricopeptide repeat 5 (IFIT5). We unveil a new function of IFIT5 complex in degrading precursor miRNAs (pre-miRNA) that includes pre-miR-363 from the miR-106a-363 cluster as well as pre-miR-101 and pre-miR-128, who share a similar 5'-end structure with pre-miR-363. These suppressive miRNAs exerted a similar function by targeting EMT transcription factors in prostate cancer cells. Depletion of IFIT5 decreased IFNγ-induced cell invasiveness in vitro and lung metastasis in vivo. IFIT5 was highly elevated in high-grade prostate cancer and its expression inversely correlated with these suppressive miRNAs. Altogether, this study unveils a prometastatic role of the IFNγ pathway via a new mechanism of action, which raises concerns about its clinical application.Significance: A unique IFIT5-XRN1 complex involved in the turnover of specific tumor suppressive microRNAs is the underlying mechanism of IFNγ-induced epithelial-to-mesenchymal transition in prostate cancer.See related commentary by Liu and Gao, p. 1032.
Collapse
Affiliation(s)
- U-Ging Lo
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Rey-Chen Pong
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Diane Yang
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Leah Gandee
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Elizabeth Hernandez
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Andrew Dang
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Chung-Jung Lin
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - John Santoyo
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Shihong Ma
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Rajni Sonavane
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jun Huang
- Department of Urology, The First Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an China
| | - Shu-Fen Tseng
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas
| | - Loredana Moro
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council, Bari, Italy
| | - Arnaldo A Arbini
- Department of Pathology, NYU Langone Medical Center, New York, New York
| | - Payal Kapur
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ganesh V Raj
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Dalin He
- Department of Urology, The First Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an China
| | - Chih-Ho Lai
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ho Lin
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Jer-Tsong Hsieh
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas.
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan, Republic of China
| |
Collapse
|
17
|
SnapshotDx Quiz: October 2018. J Invest Dermatol 2018. [DOI: 10.1016/j.jid.2018.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
18
|
Lively S, Schlichter LC. Microglia Responses to Pro-inflammatory Stimuli (LPS, IFNγ+TNFα) and Reprogramming by Resolving Cytokines (IL-4, IL-10). Front Cell Neurosci 2018; 12:215. [PMID: 30087595 PMCID: PMC6066613 DOI: 10.3389/fncel.2018.00215] [Citation(s) in RCA: 245] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 06/28/2018] [Indexed: 12/11/2022] Open
Abstract
Microglia respond to CNS injuries and diseases with complex reactions, often called "activation." A pro-inflammatory phenotype (also called classical or M1 activation) lies at one extreme of the reactivity spectrum. There were several motivations for this study. First, bacterial endotoxin (lipopolysaccharide, LPS) is the most commonly used pro-inflammatory stimulus for microglia, both in vitro and in vivo; however, pro-inflammatory cytokines (e.g., IFNγ, TNFα) rather than LPS will be encountered with sterile CNS damage and disease. We lack direct comparisons of responses between LPS and such cytokines. Second, while transcriptional profiling is providing substantial data on microglial responses to LPS, these studies mainly use mouse cells and models, and there is increasing evidence that responses of rat microglia can differ. Third, the cytokine milieu is dynamic after acute CNS damage, and an important question in microglial biology is: How malleable are their responses? There are very few studies of effects of resolving cytokines, particularly for rat microglia, and much of the work has focused on pro-inflammatory outcomes. Here, we first exposed primary rat microglia to LPS or to IFNγ+TNFα (I+T) and compared hallmark functional (nitric oxide production, migration) and molecular responses (almost 100 genes), including surface receptors that can be considered part of the sensome. Protein changes for exemplary molecules were also quantified: ARG1, CD206/MRC1, COX-2, iNOS, and PYK2. Despite some similarities, there were notable differences in responses to LPS and I+T. For instance, LPS often evoked higher pro-inflammatory gene expression and also increased several anti-inflammatory genes. Second, we compared the ability of two anti-inflammatory, resolving cytokines (IL-4, IL-10), to counteract responses to LPS and I+T. IL-4 was more effective after I+T than after LPS, and IL-10 was surprisingly ineffective after either stimulus. These results should prove useful in modeling microglial reactivity in vitro; and comparing transcriptional responses to sterile CNS inflammation in vivo.
Collapse
Affiliation(s)
- Starlee Lively
- Division of Genetics & Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Lyanne C Schlichter
- Division of Genetics & Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
19
|
Stimulation of chemokines in human endometrial stromal cells by tumor necrosis factor-α and interferon-γ is similar under apoptotic and non-apoptotic conditions. Arch Gynecol Obstet 2017; 297:505-512. [DOI: 10.1007/s00404-017-4586-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Accepted: 11/06/2017] [Indexed: 11/26/2022]
|
20
|
Feldhaus B, Dietzel ID, Heumann R, Berger R. Effects of Interferon-γ and Tumor Necrosis Factor-α on Survival and Differentiation of Oligodendrocyte Progenitors. ACTA ACUST UNITED AC 2016; 11:89-96. [PMID: 14980310 DOI: 10.1016/j.jsgi.2003.08.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
OBJECTIVE There is strong evidence from recent clinical studies that ascending intrauterine infection is associated with an increased incidence of periventricular leukomalacia in very premature fetuses. Periventricular leukomalacia is characterized by disrupted myelination from a loss of oligodendrocyte progenitors. We investigated the effects of proinflammatory cytokines on the survival and differentiation of this cell type. METHODS Cultures of more than 90% A2B5-positive progenitors were prepared from neonatal rats and kept for 3 days in medium supplemented with factors that stimulate cell proliferation. After 1 day in proliferation medium, cells were treated with interferon-gamma (100 U/mL) and tumor necrosis factor-alpha (100 ng/mL) for 48 hours triggering an increase in apoptotic A2B5 progenitor cells from 3.2 +/- 2.3% to 11.0 +/- 2.6%. After cytokine treatment cultures were transferred to medium containing factors to promote differentiation of progenitors into the myelinating phenotype. RESULTS In cytokine pretreated cultures, only 2.6 +/- 1.1% of total cells survived after a total of 9 days in vitro, whereas in untreated cultures most cells differentiated as shown by expression of myelin basic protein, myelin-associated glycoprotein, 2,3-cyclic nucleotide 3-phosphodiesterase, and myelin oligodendrocyte-specific protein. Using ten-fold reduced concentrations of combined interferon-gamma (10 U/mL) and tumor necrosis factor-alpha (10 ng/mL) pretreatment resulted in a survival to 11.2 +/- 4.9% of total cells with 36.3 +/- 11.6% A2B5-positive cells at day 9. This indicates a major enrichment of undifferentiated cells compared with untreated controls which harbored only 1.0 +/- 0.3% A2B5-positive cells. CONCLUSION Inflammatory cytokines not only induced apoptotic cell death but also prevented the differentiation of immature A2B5 oligodendrocyte progenitors into the myelinating phenotype.
Collapse
Affiliation(s)
- Beatrix Feldhaus
- Departments ofDepartment of Obstetrics and Gynecology, University of Bochum, Bochum, Germany
| | | | | | | |
Collapse
|
21
|
Antiseptic effect of sea cucumber ( Holothuria atra) against multi-organ failure induced by sepsis: Molecular and histopathological study. Exp Ther Med 2016; 12:222-230. [PMID: 27347042 PMCID: PMC4906945 DOI: 10.3892/etm.2016.3321] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 03/10/2016] [Indexed: 12/11/2022] Open
Abstract
Sepsis is a systemic inflammatory response to infection and severe sepsis patients can develop acute lung and liver injury. The aim of the present study was to evaluate the efficacy of Holothuria atra methanolic body wall extract (HaE), as an antioxidant and anti-inflammatory agent against induced sepsis in a cecal ligation and puncture (CLP) rat model. In total, 30 males albino rats were divided into three groups (n=10 each) as follows: Sham (Sh), which was used as negative control; sepsis (Se), which was used as a positive control and was subjected to CLP surgery; and Ho, which was subjected to CLP and fed with 200 mg/kg (body weight) of HaE, once daily for 7 days. Subsequently, the expression of various genes was detected by polymerase chain reaction, while liver and lung tissues were examined by immunohistochemistry. The expression of Caspase-3 was significantly reduced in liver and lung tissues in the Ho group, while the expression levels of Gsta2, Cat and Sod1 genes were slightly reduced in the Ho group, when compared with the Se group. In addition, expression levels of tumor necrosis factor, interferon-γ, liver interleukin (IL)1b and lung IL1a were reduced in the Ho group compared with the Se group. Furthermore, histopathological changes were observed in liver tissues of the Se group, including congestion of hepatoportal blood vessel and focal hepatic necrosis, while lung tissues showed marked edema, hemorrhage and alveolar septal thickening. The Ho group showed apparent normal hepatic parenchyma and slight interstitial pneumonia. Immunohistochemical staining of caspase-3 in liver and lung tissues showed no expression in the Sh group, strong expression in the Se group and moderate expression in the Ho group. In conclusion, HaE demonstrated beneficial effect against induced sepsis, which may be attributed to its antioxidant and antiapoptotic activities.
Collapse
|
22
|
Shen J, Li ZJ, Li LF, Lu L, Xiao ZG, Wu WKK, Zhang L, Li MX, Hu W, Chan KM, Cho CH. Vascular-targeted TNFα and IFNγ inhibits orthotopic colorectal tumor growth. J Transl Med 2016; 14:187. [PMID: 27342639 PMCID: PMC4919862 DOI: 10.1186/s12967-016-0944-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 06/15/2016] [Indexed: 02/06/2023] Open
Abstract
Background Tumor necrosis factor alpha (TNFα) and interferon gamma (IFNγ) were originally identified to show potent anti-tumor activity and immunomodulatory capability. Unfortunately, several clinical studies of relevant cancer therapy did not observe significant response in maximum tolerated dose whether given alone or in combination. We have identified a tumor vasculature homing peptide (TCP-1 peptide) which targets only the vasculature of colorectal tumors but not normal blood vessels in animals and humans. In the current study, the antitumor effect of TCP-1/TNFα and TCP-1/IFNγ alone or in combination was studied in orthotopic colorectal tumor model. Methods TCP-1/TNFα and TCP-1/IFNγ recombinant proteins were prepared and i.v. injected to study the in vivo anticancer effect in orthotopic colorectal tumor model. Tumor apoptosis was determined by TUNEL staining and cleaved caspase-3 immunofluorescent staining. Tumor infiltrating lymphocytes were analyzed by immunofluorescent staining and flow cytometry. Western-blot was performed to examine the expression of proteins. Cell apoptosis was measured by Annexin V/PI flow cytometry. Results Targeted delivery of TNFα or IFNγ by TCP-1 peptide exhibited better antitumor activity than unconjugated format by inducing more tumor apoptosis and also enhancing antitumor immunity shown by increased infiltration of T lymphocytes inside the tumor. More importantly, combination therapy of TCP-1/TNFα and TCP-1/IFNγ synergistically suppressed tumor growth and alleviated systematic toxicity associated with untargeted therapy. This combination therapy induced massive apoptosis/secondary necrosis in the tumor. Conclusions Taken together, our data demonstrate TCP-1 is an efficient drug carrier for targeted therapy of colorectal cancer (CRC). TCP-1/TNFα combined with TCP-1/IFNγ is a promising combination therapy for CRC. Electronic supplementary material The online version of this article (doi:10.1186/s12967-016-0944-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jing Shen
- Laboratory for Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, People's Republic of China
| | - Zhi Jie Li
- Laboratory for Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, People's Republic of China. .,Harry Perkins Institute of Medical Research, University of Western Australia, Crawley, WA, 6009, Australia.
| | - Long Fei Li
- Laboratory for Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, People's Republic of China
| | - Lan Lu
- Laboratory for Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, People's Republic of China.,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong, People's Republic of China
| | - Zhan Gang Xiao
- Laboratory for Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, People's Republic of China
| | - William Ka Kei Wu
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Lin Zhang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong, People's Republic of China
| | - Ming Xing Li
- Laboratory for Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, People's Republic of China
| | - Wei Hu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong, People's Republic of China
| | - Kam Ming Chan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong, People's Republic of China
| | - Chi Hin Cho
- Laboratory for Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, People's Republic of China. .,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong, People's Republic of China.
| |
Collapse
|
23
|
Armstrong MJ, Stang MT, Liu Y, Yan J, Pizzoferrato E, Yim JH. IRF-1 inhibits NF-κB activity, suppresses TRAF2 and cIAP1 and induces breast cancer cell specific growth inhibition. Cancer Biol Ther 2016; 16:1029-41. [PMID: 26011589 DOI: 10.1080/15384047.2015.1046646] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Interferon Regulatory Factor (IRF)-1, originally identified as a transcription factor of the human interferon (IFN)-β gene, mediates tumor suppression and may inhibit oncogenesis. We have shown that IRF-1 in human breast cancer cells results in the down-regulation of survivin, tumor cell death, and the inhibition of tumor growth in vivo in xenogeneic mouse models. In this current report, we initiate studies comparing the effect of IRF-1 in human nonmalignant breast cell and breast cancer cell lines. While IRF-1 in breast cancer cells results in growth inhibition and cell death, profound growth inhibition and cell death are not observed in nonmalignant human breast cells. We show that TNF-α or IFN-γ induces IRF-1 in breast cancer cells and results in enhanced cell death. Abrogation of IRF-1 diminishes TNF-α and IFN-γ-induced apoptosis. We test the hypothesis that IRF-1 augments TNF-α-induced apoptosis in breast cancer cells. Potential signaling networks elicited by IRF-1 are investigated by evaluating the NF-κB pathway. TNF-α and/or IFN-γ results in decreased presence of NF-κB p65 in the nucleus of breast cancer cells. While TNF-α and/or IFN-γ can induce IRF-1 in nonmalignant breast cells, a marked change in NF-κB p65 is not observed. Moreover, the ectopic expression of IRF-1 in breast cancer cells results in caspase-3, -7, -8 cleavage, inhibits NF-κB activity, and suppresses the expression of molecules involved in the NF-κB pathway. These data show that IRF-1 in human breast cancer cells elicits multiple signaling networks including intrinsic and extrinsic cell death and down-regulates molecules involved in the NF-κB pathway.
Collapse
Key Words
- Ad, adenovirus
- Cdk, cyclin-dependent kinase
- DISC, death-inducing signaling complex
- DMEM, Dulbecco's Modified Eagle's Medium
- DR, death receptor
- EGFP, enhanced green fluorescent protein
- ER, estrogen receptor
- FADD, fas-associated death domain
- FBS, Fetal Bovine Serum
- FITC, fluorescein isothiocyanate
- FLICE, fas-associated death domain protein interleukin-1 β-converting enzyme
- IAP
- IFN-β, interferon-β
- IFN-γ, interferon-gamma
- IKK, IκB, kinase complex
- IRF-1
- IRF-1, interferon regulatory factor-1
- IκB, Inhibitory kappaB
- MOI, multiplicity of infection
- MTT, methylthiazoltetrazolium
- NEMO, NF-κB essential modulator
- NF-κB
- NF-κB, nuclear factor of kappa Beta
- RIP1, receptor interacting protein 1
- SCID, severe combined immunodeficiency
- STAT, signal transducer and activator of transcription
- Smac/DIABLO, Second mitochondria-derived activator of caspase/Direct IAP-binding protein with low pI
- TNF-α, tumor necrosis factor-α
- TNFR, tumor necrosis factor receptor
- TRADD, TNF receptor associated protein with a death domain
- TRAF2, tumor necrosis factor receptor-associated factor 2
- TRAIL, tumor necrosis factor-related apoptosis-inducing ligand
- XIAP, X-linked inhibitor of apoptosis protein
- apoptosis
- breast cancer
- cFLIP, cellular FLICE inhibitory protein
- cIAP1, c-inhibitor of apoptosis
- p53
- siRNA, small interfering RNA
- tumor suppressor
- β-gal, β-galactosidase
Collapse
Affiliation(s)
- Michaele J Armstrong
- a Department of Surgery; University of Pittsburgh School of Medicine ; Pittsburgh , PA , USA
| | | | | | | | | | | |
Collapse
|
24
|
Nunes C, Teixeira N, Serra D, Freitas V, Almeida L, Laranjinha J. Red wine polyphenol extract efficiently protects intestinal epithelial cells from inflammation via opposite modulation of JAK/STAT and Nrf2 pathways. Toxicol Res (Camb) 2016; 5:53-65. [PMID: 30090326 PMCID: PMC6061778 DOI: 10.1039/c5tx00214a] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 10/01/2015] [Indexed: 12/17/2022] Open
Abstract
The development of therapeutic approaches combining efficacy and safety represents an important goal in intestinal inflammation research. Recently, evidence has supported dietary polyphenols as useful tools in the treatment and prevention of chronic inflammatory diseases, but the mechanisms of action are still poorly understood. We here reveal molecular mechanisms underlying the anti-inflammatory action of a non-alcoholic polyphenol red wine extract (RWE), operating at complementary levels via the Janus kinase/signal transducer and activator of transcription (JAK/STAT) and Nuclear factor-erythroid 2-related factor-2 (Nrf2) pathways. RWE significantly reduced the nuclear levels of phosphorylated STAT1 and also the cellular levels of phosphorylated JAK1 induced by cytokines, suppressing the JAK/STAT inflammatory signalling cascade. In turn, RWE increased the Nrf2 nuclear level, activating the Nrf2 pathway, leading not only to an up-regulation of the heme oxygenase-1 (HO-1) expression but also to an increase of the glutamate-cysteine ligase subunit catalytic (GCLc) gene expression, enhancing the GSH synthesis, thereby counteracting GSH depletion that occurs under inflammatory conditions. Overall, data indicate that the anti-inflammatory action of RWE is exerted at complementary levels, via suppression of the JAK/STAT inflammatory pathway and positive modulation of the activity of Nrf2. These results point to the potential use of the RWE as an efficient, readily available and inexpensive therapeutic strategy in the context of gastrointestinal inflammation.
Collapse
Affiliation(s)
- Carla Nunes
- Center for Neurosciences and Cell Biology and Faculty of Pharmacy , University of Coimbra , Health Sciences Campus , Azinhaga de Santa Comba , 3000-548 Coimbra , Portugal .
| | - Natércia Teixeira
- Department of Chemistry , Faculty of Sciences , University of Porto , Portugal
| | - Diana Serra
- Center for Neurosciences and Cell Biology and Faculty of Pharmacy , University of Coimbra , Health Sciences Campus , Azinhaga de Santa Comba , 3000-548 Coimbra , Portugal .
| | - Víctor Freitas
- Department of Chemistry , Faculty of Sciences , University of Porto , Portugal
| | - Leonor Almeida
- Center for Neurosciences and Cell Biology and Faculty of Pharmacy , University of Coimbra , Health Sciences Campus , Azinhaga de Santa Comba , 3000-548 Coimbra , Portugal .
| | - João Laranjinha
- Center for Neurosciences and Cell Biology and Faculty of Pharmacy , University of Coimbra , Health Sciences Campus , Azinhaga de Santa Comba , 3000-548 Coimbra , Portugal .
| |
Collapse
|
25
|
Meissl K, Macho-Maschler S, Müller M, Strobl B. The good and the bad faces of STAT1 in solid tumours. Cytokine 2015; 89:12-20. [PMID: 26631912 DOI: 10.1016/j.cyto.2015.11.011] [Citation(s) in RCA: 185] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 11/09/2015] [Indexed: 12/13/2022]
Abstract
Signal transducer and activator of transcription (STAT) 1 is part of the Janus kinase (JAK)/STAT signalling cascade and is best known for its essential role in mediating responses to all types of interferons (IFN). STAT1 regulates a variety of cellular processes, such as antimicrobial activities, cell proliferation and cell death. It exerts important immune modulatory activities both in the innate and the adaptive arm of the immune system. Based on studies in mice and data from human patients, STAT1 is generally considered a tumour suppressor but there is growing evidence that it can also act as a tumour promoter. This review aims at contrasting the two faces of STAT1 in tumourigenesis and providing an overview on the current knowledge of the underlying mechanisms or pathways.
Collapse
Affiliation(s)
- Katrin Meissl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria
| | - Sabine Macho-Maschler
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria
| | - Mathias Müller
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria
| | - Birgit Strobl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria.
| |
Collapse
|
26
|
Spratte J, Schönborn M, Treder N, Bornkessel F, Zygmunt M, Fluhr H. Heparin modulates chemokines in human endometrial stromal cells by interaction with tumor necrosis factor α and thrombin. Fertil Steril 2015; 103:1363-9. [PMID: 25813285 DOI: 10.1016/j.fertnstert.2015.02.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Revised: 02/02/2015] [Accepted: 02/17/2015] [Indexed: 01/11/2023]
Abstract
OBJECTIVE To study the impact of heparins on chemokines in decidualized human endometrial stromal cells (ESCs) in vitro. DESIGN In vitro experiment. SETTING Research laboratory. PATIENT(S) Premenopausal women undergoing hysterectomy for benign reasons. INTERVENTION(S) ESCs were isolated from hysterectomy specimens, decidualized in vitro and incubated with unfractionated heparin and low-molecular-weight heparins (LMWHs) as well as tumor necrosis factor (TNF) α or thrombin with or without heparins. MAIN OUTCOME MEASURE(S) Chemokines CXCL1, CXCL5, CXCL8, CCL2, and CCL5 were measured with the use of ELISA, and CXCL5, CXCL8, CCL2, and CCL5 were detected with the use of real-time reverse-transcription polymerase chain reaction. Cell viability was determined with the use of a fluorometric assay. RESULT(S) TNF-α and thrombin stimulated distinct patterns of chemokines in ESCs. Unfractionated heparin and LMWHs attenuated the TNF-α-mediated induction of CXCL8 and enhanced CXCL5, CCL2, and CCL5. The stimulating effect of thrombin on CXCL8 could be inhibited by heparin, whereas heparin had no impact on thrombin-induced CXCL1 and CCL2. Nuclear factor of transcription κB signaling mediated the effects of TNF-α. The effects of thrombin were mediated via extracellular signal-regulated protein kinases 1/2. CONCLUSION(S) Heparins have modulating effects on TNF-α- and thrombin-induced endometrial chemokines, which might have implications in the regulation of endometrial receptivity and early implantation.
Collapse
Affiliation(s)
- Julia Spratte
- Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany.
| | - Magdalena Schönborn
- Department of Obstetrics and Gynecology, University of Greifswald, Greifswald, Germany
| | - Nora Treder
- Department of Obstetrics and Gynecology, University of Greifswald, Greifswald, Germany
| | - Frauke Bornkessel
- Department of Obstetrics and Gynecology, University of Greifswald, Greifswald, Germany
| | - Marek Zygmunt
- Department of Obstetrics and Gynecology, University of Greifswald, Greifswald, Germany
| | - Herbert Fluhr
- Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
27
|
Zhang Y, Molavi O, Su M, Lai R. The clinical and biological significance of STAT1 in esophageal squamous cell carcinoma. BMC Cancer 2014; 14:791. [PMID: 25355139 PMCID: PMC4233059 DOI: 10.1186/1471-2407-14-791] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 10/13/2014] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Loss of STAT1 (Signal Transducer and Activator of Transcription-1) has been implicated in the pathobiology of a number of cancer types. Nonetheless, the biological and clinical significance of STAT1 in esophageal squamous cell carcinomas (ESCC) has not been comprehensively studied. METHODS Using immunohistochemistry, we detected the STAT1 expression in a cohort of ESCC patients; In-vitro experiments, we used enforced gene transfection of STAT1C into two STAT1- weak/negative ESCC cell lines and siRNA knockdown of STAT1 in two STAT1-strong ESCC cell lines to detect STAT1 function in ESCC. RESULTS We found that the expression of STAT1 was heterogeneous in ESCC, with 64 (49.0%) strongly positive cases, 59 (45.0%) weakly positive cases and 8 (6.1%) negative cases. STAT1 expression inversely correlated with the depth of tumor invasion and tumor size (p=0.047 and p=0.029, respectively, Chi square). Furthermore, patients with STAT1-strong/weak tumors had a significantly longer survival compared to those with STAT1-negative tumors (33.6 months versus 13.1 months, p=0.019). In patients carrying tumors of aggressive cytology (n=50), those with STAT1-strong tumors survived significantly longer than those with STAT1-weak/negative tumors (34.6 months versus 20.5 months, p=0.011). Our in-vitro experiments revealed that STAT1 is proapoptotic and inhibitory to cell-cycle progression and colony formation. Lastly, we found evidence that STAT1 signaling in ESCC cells down-regulated the expression and/or activity of NF-κB and STAT3, both of which are known to have oncogenic potential. CONCLUSION To conclude, our findings suggest that STAT1 is a tumor suppressor in ESCC. Loss of STAT1, which is frequent in ESCC, contributes to the pathogenesis of these tumors.
Collapse
Affiliation(s)
- Ying Zhang
- />Department of Pathology, Shantou University Medical College, 22 Xinling Road, Shantou, 515031 Guangdong Province China
- />Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta Canada
| | - Ommoleila Molavi
- />Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta Canada
| | - Min Su
- />Department of Pathology, Shantou University Medical College, 22 Xinling Road, Shantou, 515031 Guangdong Province China
| | - Raymond Lai
- />Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta Canada
- />Department of Oncology, University of Alberta, Edmonton, Alberta Canada
- />DynaLIFEDX Medical Laboratories, Edmonton, Alberta Canada
- />Department of Laboratory Medicine and Pathology, University of Alberta, 5142 Katz Group Centre for Pharmacy and Health Research, Edmonton, Alberta T6G 2E1 Canada
| |
Collapse
|
28
|
Zhang J, Zhong Q. Histone deacetylase inhibitors and cell death. Cell Mol Life Sci 2014; 71:3885-901. [PMID: 24898083 PMCID: PMC4414051 DOI: 10.1007/s00018-014-1656-6] [Citation(s) in RCA: 160] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 04/23/2014] [Accepted: 05/20/2014] [Indexed: 12/14/2022]
Abstract
Histone deacetylases (HDACs) are a vast family of enzymes involved in chromatin remodeling and have crucial roles in numerous biological processes, largely through their repressive influence on transcription. In addition to modifying histones, HDACs also target many other non-histone protein substrates to regulate gene expression. Recently, HDACs have gained growing attention as HDAC-inhibiting compounds are being developed as promising cancer therapeutics. Histone deacetylase inhibitors (HDACi) have been shown to induce differentiation, cell cycle arrest, apoptosis, autophagy and necrosis in a variety of transformed cell lines. In this review, we mainly discuss how HDACi may elicit a therapeutic response to human cancers through different cell death pathways, in particular, apoptosis and autophagy.
Collapse
Affiliation(s)
- Jing Zhang
- Center for Autophagy Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Qing Zhong
- Center for Autophagy Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| |
Collapse
|
29
|
Choudhury S, Schnell M, Bühler T, Reinke Y, Lüdemann J, Nießner F, Brinkmeier H, Herda LR, Staudt A, Kroemer HK, Völker U, Felix SB, Landsberger M. Antibodies against potassium channel interacting protein 2 induce necrosis in isolated rat cardiomyocytes. J Cell Biochem 2014; 115:678-89. [PMID: 24453044 DOI: 10.1002/jcb.24707] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 10/24/2013] [Indexed: 12/26/2022]
Abstract
Auto-antibodies against cardiac proteins have been described in patients with dilated cardiomyopathy. Antibodies against the C-terminal part of KChIP2 (anti-KChIP2 [C-12]) enhance cell death of rat cardiomyocytes. The underlying mechanisms are not fully understood. Therefore, we wanted to explore the mechanisms responsible for anti-KChIP2-mediated cell death. Rat cardiomyocytes were treated with anti-KChIP2 (C-12). KChIP2 RNA and protein expressions, nuclear NF-κB, mitochondrial membrane potential Δψm, caspase-3 and -9 activities, necrotic and apoptotic cells, total Ca(2+) and K(+) concentrations, and the effects on L-type Ca(2+) channels were quantified. Anti-KChIP2 (C-12) induced nuclear translocation of NF-κB. Anti-KChIP2 (C-12)-treatment for 2 h significantly reduced KChIP2 mRNA and protein expression. Anti-KChIP2 (C-12) induced nuclear translocation of NF-κB after 1 h. After 6 h, Δψm and caspase-3 and -9 activities were not significantly changed. After 24 h, anti-KChIP2 (C-12)-treated cells were 75 ± 3% necrotic, 2 ± 1% apoptotic, and 13 ± 2% viable. Eighty-six ± 1% of experimental buffer-treated cells were viable. Anti-KChIP2 (C-12) induced significant increases in total Ca(2+) (plus 11 ± 2%) and K(+) (plus 18 ± 2%) concentrations after 5 min. Anti-KChIP2 (C-12) resulted in an increased Ca(2+) influx through L-type Ca(2+) channels. In conclusion, our results suggest that anti-KChIP2 (C-12) enhances cell death of rat cardiomyocytes probably due to necrosis.
Collapse
Affiliation(s)
- Sangita Choudhury
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Jansson D, Rustenhoven J, Feng S, Hurley D, Oldfield RL, Bergin PS, Mee EW, Faull RLM, Dragunow M. A role for human brain pericytes in neuroinflammation. J Neuroinflammation 2014; 11:104. [PMID: 24920309 PMCID: PMC4105169 DOI: 10.1186/1742-2094-11-104] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 05/19/2014] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Brain inflammation plays a key role in neurological disease. Although much research has been conducted investigating inflammatory events in animal models, potential differences in human brain versus rodent models makes it imperative that we also study these phenomena in human cells and tissue. METHODS Primary human brain cell cultures were generated from biopsy tissue of patients undergoing surgery for drug-resistant epilepsy. Cells were treated with pro-inflammatory compounds IFNγ, TNFα, IL-1β, and LPS, and chemokines IP-10 and MCP-1 were measured by immunocytochemistry, western blot, and qRT-PCR. Microarray analysis was also performed on late passage cultures treated with vehicle or IFNγ and IL-1β. RESULTS Early passage human brain cell cultures were a mixture of microglia, astrocytes, fibroblasts and pericytes. Later passage cultures contained proliferating fibroblasts and pericytes only. Under basal culture conditions all cell types showed cytoplasmic NFκB indicating that they were in a non-activated state. Expression of IP-10 and MCP-1 were significantly increased in response to pro-inflammatory stimuli. The two chemokines were expressed in mixed cultures as well as cultures of fibroblasts and pericytes only. The expression of IP-10 and MCP-1 were regulated at the mRNA and protein level, and both were secreted into cell culture media. NFκB nuclear translocation was also detected in response to pro-inflammatory cues (except IFNγ) in all cell types. Microarray analysis of brain pericytes also revealed widespread changes in gene expression in response to the combination of IFNγ and IL-1β treatment including interleukins, chemokines, cellular adhesion molecules and much more. CONCLUSIONS Adult human brain cells are sensitive to cytokine challenge. As expected 'classical' brain immune cells, such as microglia and astrocytes, responded to cytokine challenge but of even more interest, brain pericytes also responded to such challenge with a rich repertoire of gene expression. Immune activation of brain pericytes may play an important role in communicating inflammatory signals to and within the brain interior and may also be involved in blood brain barrier (BBB) disruption . Targeting brain pericytes, as well as microglia and astrocytes, may provide novel opportunities for reducing brain inflammation and maintaining BBB function and brain homeostasis in human brain disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Mike Dragunow
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, 85 Park Road, Auckland 1023, New Zealand.
| |
Collapse
|
31
|
Messina NL, Banks KM, Vidacs E, Martin BP, Long F, Christiansen AJ, Smyth MJ, Clarke CJP, Johnstone RW. Modulation of antitumour immune responses by intratumoural
Stat1
expression. Immunol Cell Biol 2013; 91:556-67. [DOI: 10.1038/icb.2013.41] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 07/12/2013] [Accepted: 07/12/2013] [Indexed: 01/09/2023]
Affiliation(s)
- Nicole L Messina
- Cancer Therapeutics Program, Peter MacCallum Cancer CentreEast MelbourneVictoriaAustralia
- Deptartment of Pathology, University of MelbourneParkvilleVictoriaAustralia
| | - Kellie M Banks
- Cancer Therapeutics Program, Peter MacCallum Cancer CentreEast MelbourneVictoriaAustralia
| | - Eva Vidacs
- Cancer Therapeutics Program, Peter MacCallum Cancer CentreEast MelbourneVictoriaAustralia
| | - Ben P Martin
- Cancer Therapeutics Program, Peter MacCallum Cancer CentreEast MelbourneVictoriaAustralia
| | - Fennella Long
- Cancer Therapeutics Program, Peter MacCallum Cancer CentreEast MelbourneVictoriaAustralia
| | - Ailsa J Christiansen
- Institute of Pharmaceutical Science, Swiss Federal Institute of Technology (ETHZ)ZurichSwitzerland
| | - Mark J Smyth
- Immunology in Cancer and Infection Laboratory, Queensland Institute of Medical ResearchHerstonQueenslandAustralia
- School of Medicine, University of QueenslandHerstonQueenslandAustralia
| | - Christopher J P Clarke
- Cancer Therapeutics Program, Peter MacCallum Cancer CentreEast MelbourneVictoriaAustralia
- Deptartment of Pathology, University of MelbourneParkvilleVictoriaAustralia
| | - Ricky W Johnstone
- Cancer Therapeutics Program, Peter MacCallum Cancer CentreEast MelbourneVictoriaAustralia
- Sir Peter MacCallum Department of Oncology, University of MelbourneParkvilleVictoriaAustralia
| |
Collapse
|
32
|
Okazawa S, Furusawa Y, Kariya A, Hassan MA, Arai M, Hayashi R, Tabuchi Y, Kondo T, Tobe K. Inactivation of DNA-dependent protein kinase promotes heat-induced apoptosis independently of heat-shock protein induction in human cancer cell lines. PLoS One 2013; 8:e58325. [PMID: 23505488 PMCID: PMC3594312 DOI: 10.1371/journal.pone.0058325] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 02/01/2013] [Indexed: 12/14/2022] Open
Abstract
The inhibition of DNA damage response pathway seems to be an attractive strategy for cancer therapy. It was previously reported that in rodent cells exposed to heat stress, cell growth was promoted by the activity of DNA-dependent protein kinase (DNA-PK), an enzyme involved in DNA non-homologous end joining (NHEJ) required for double-strand break repair. The absence of a functioning DNA-PK was associated with down regulation of heat shock protein 70 (HSP70). The objective of this study is thus to investigate the role of DNA-PK inhibition in heat-induced apoptosis in human cell lines. The inhibitors of phosphorylation of the DNA-PK catalytic subunit (DNA-PKcs) at Ser2056, such as NU7026 and NU7441, were utilized. Furthermore, knock down of DNA-PKcs was carried out using small interfering RNA (siDNA-PKcs). For heat exposure, cells were placed in water bath at 44°C for 60 min. Apoptosis was evaluated after 24 h incubation flow cytometrically. Proteins were extracted after 24 h and analyzed for HSP70 and HSP40 expression by Western blotting. Total RNA was extracted 6 h after treatment and analyzed using a GeneChip® microarray system to identify and select the up-regulated genes (≥1.5 fold). The results showed an enhancement in heat-induced apoptosis in absence of functioning DNA-PKcs. Interestingly, the expression levels of HSP70 and HSP40 were elevated in the absence of DNA-PKcs under heat stress. The results of genetic network analysis showed that HSPs and JUN genes were up-regulated independently of DNA-PKcs in exposed parent and knock out cells. In the presence of functioning DNA-PKcs, there was an observed up-regulation of anti-apoptotic genes, such as NR1D1, whereas in the absence of DNA-PKcs the pro-apoptotic genes, such as EGR2, were preferentially up-regulated. From these findings, we concluded that in human cells, the inactivation of DNA-PKcs can promote heat-induced apoptosis independently of heat-shock proteins.
Collapse
Affiliation(s)
- Seisuke Okazawa
- First Department of Internal Medicine, Graduate School of Medicine Pharmaceutical Sciences, University of Toyama, Toyama, Japan
- Department of Radiological Sciences, Graduate School of Medicine Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Yukihiro Furusawa
- Department of Radiological Sciences, Graduate School of Medicine Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Ayako Kariya
- Department of Radiological Sciences, Graduate School of Medicine Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Mariame Ali Hassan
- Department of Radiological Sciences, Graduate School of Medicine Pharmaceutical Sciences, University of Toyama, Toyama, Japan
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Mie Arai
- Department of Radiological Sciences, Graduate School of Medicine Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Ryuji Hayashi
- First Department of Internal Medicine, Graduate School of Medicine Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Yoshiaki Tabuchi
- Division of Molecular Genetics Research, Life Science Research Center, Graduate School of Medicine Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Takashi Kondo
- Department of Radiological Sciences, Graduate School of Medicine Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Kazuyuki Tobe
- First Department of Internal Medicine, Graduate School of Medicine Pharmaceutical Sciences, University of Toyama, Toyama, Japan
- * E-mail:
| |
Collapse
|
33
|
TWEAK signals through JAK-STAT to induce tumor cell apoptosis. Cytokine 2012; 61:210-7. [PMID: 23107828 DOI: 10.1016/j.cyto.2012.09.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Revised: 08/22/2012] [Accepted: 09/25/2012] [Indexed: 01/24/2023]
Abstract
The TWEAK receptor Fn14 (TNFRSF12), a member of the TNF Receptor superfamily, can mediate many processes, including apoptosis. Fn14 agonists have therefore been the subject of interest as potential cancer therapeutics. In cell culture experiments, interferon gamma (IFNγ) is typically required for induction of apoptotic activity by either TWEAK or Fn14 agonistic antibodies in most cell lines. We have investigated the mechanism of IFNγ signaling and the role of JAK-STAT signaling in TWEAK/Fn14-mediated tumor cell killing. We found that IFNγ-mediated enhancement of tumor cell killing is JAK-STAT dependent, as JAK inhibitors block IFNγ-dependent TWEAK induced apoptosis. Exposure of tumor cells to IFNγ results in an increase in Fn14 expression on the cell surface, which may be a mechanism by which IFNγ induces sensitivity to TWEAK. In a reciprocal fashion, we observed that IFNγ receptor levels increase in response to TWEAK treatment in WiDr cells. Significantly, we found that TWEAK alone can induce STAT1 phosphorylation in WiDr tumor cells. Moreover, TWEAK induction of tumor cell apoptosis in WiDr cells in the absence of IFNγ is mediated by the JAK-STAT pathway. Correspondingly, we show that treatment of tumor bearing mice with mBIIB036, an Fn14 agonistic antibody, results in STAT1 phosphorylation in the tumors. Notably, the level of STAT1 phosphorylation appears to correlate with the degree of tumor growth inhibition by BIIB036 in vivo. Additionally, in WiDr cells, TWEAK induces a soluble factor, which we have identified as IFNβ, capable of independently inducing STAT1 phosphorylation when transferred to naïve cells. Finally, either IFNα or IFNβ can partially substitute for IFNγ in sensitizing tumor cells to Fn14 agonists. In summary, we show that TWEAK/Fn14 can signal through the JAK-STAT pathway to induce IFNβ, and that the ability of TWEAK to induce tumor cell apoptosis is mediated by JAK-STAT signaling. We also demonstrate that IFNγ enhancement of TWEAK/FN14-mediated tumor cell death is JAK-dependent and may occur by IFNγ-dependent upregulation of Fn14 on tumor cells. These findings may have implications for the appropriately targeted clinical development of Fn14 agonists as anti-cancer therapy.
Collapse
|
34
|
Montinaro A, Forte G, Sorrentino R, Luciano A, Palma G, Arra C, Adcock IM, Pinto A, Morello S. Adoptive immunotherapy with Cl-IB-MECA-treated CD8+ T cells reduces melanoma growth in mice. PLoS One 2012; 7:e45401. [PMID: 23028986 PMCID: PMC3454429 DOI: 10.1371/journal.pone.0045401] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 08/22/2012] [Indexed: 12/17/2022] Open
Abstract
Cl-IB-MECA is a selective A3 adenosine receptor agonist, which plays a crucial role in limiting tumor progression. In mice, Cl-IB-MECA administration enhances the anti-tumor T cell-mediated response. However, little is known about the activity of Cl-IB-MECA on CD8+ T cells. The aim of this study was to investigate the effect of ex vivo Cl-IB-MECA treatment of CD8+ T cells, adoptively transferred in melanoma-bearing mice. Adoptive transfer of Cl-IB-MECA-treated CD8+ T cells or a single administration of Cl-IB-MECA (20 ng/mouse) inhibited tumor growth compared with the control group and significantly improved mouse survival. This was associated with the release of Th1-type cytokines and a greater influx of mature Langerin+ dendritic cells (LCs) into the tumor microenvironment. CD8+ T cells treated with Cl-IB-MECA released TNF-α which plays a critical role in the therapeutic efficacy of these cells when injected to mice. Indeed, neutralization of TNF-α by a specific monoclonal Ab significantly blocked the anti-tumor activity of Cl-IB-MECA-treated T cells. This was due to the reduction in levels of cytotoxic cytokines and the presence of fewer LCs. In conclusion, these studies reveal that ex vivo treatment with Cl-IB-MECA improves CD8+ T cell adoptive immunotherapy for melanoma in a TNF-α-dependent manner.
Collapse
Affiliation(s)
- Antonella Montinaro
- Department of Pharmaceutical and Biomedical Sciences, University of Salerno, Salerno, Italy
| | - Giovanni Forte
- Department of Pharmaceutical and Biomedical Sciences, University of Salerno, Salerno, Italy
| | - Rosalinda Sorrentino
- Department of Pharmaceutical and Biomedical Sciences, University of Salerno, Salerno, Italy
| | - Antonio Luciano
- National Cancer Institute “G. Pascale” Naples, Naples, Italy
| | - Giuseppe Palma
- National Cancer Institute “G. Pascale” Naples, Naples, Italy
| | - Claudio Arra
- National Cancer Institute “G. Pascale” Naples, Naples, Italy
| | - Ian M. Adcock
- NHLI, Imperial College of London, London, United Kingdom
| | - Aldo Pinto
- Department of Pharmaceutical and Biomedical Sciences, University of Salerno, Salerno, Italy
| | - Silvana Morello
- Department of Pharmaceutical and Biomedical Sciences, University of Salerno, Salerno, Italy
| |
Collapse
|
35
|
Kim S, Kim KA, Suk K, Kim YH, Oh SH, Lee MK, Kim KW, Lee MS. Apoptosis of human islet cells by cytokines. Immune Netw 2012; 12:113-7. [PMID: 22916047 PMCID: PMC3422709 DOI: 10.4110/in.2012.12.3.113] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Revised: 05/21/2012] [Accepted: 05/23/2012] [Indexed: 12/01/2022] Open
Abstract
FasL, perforin, TNFα, IL-1 and NO have been considered as effector molecule(s) leading to β-cell death in autoimmune diabetes. However, the real culprit(s) of β-cell destruction have long been elusive despite intense investigation. Previously we have suggested IFNγ/TNFα synergism as the final effector molecules in autoimmune diabetes of NOD mice. A combination of IFNγ and TNFα but neither cytokine alone, induced classical caspase-dependent apoptosis in murine insulinoma and pancreatic islet cells. IFNγ treatment conferred susceptibility to TNFα-induced apoptosis on otherwise resistant murine insulinoma cells by STAT1 activation followed by IRF-1 induction. Here we report that IFNγ/TNFα synergism induces apoptosis of human pancreatic islet cells. We also observed STAT1 activation followed by IRF-1 induction by IFNγ treatment in human islet cells. Taken together, we suggest that IFNγ/TNFα synergism could be involved in human islet cell death in type 1 diabetes, similar to murine type 1 diabetes.
Collapse
Affiliation(s)
- Sunshin Kim
- New Experimental Therapeutics Branch, Division of Convergence Technology, National Cancer Center, Goyang 410-769, Korea
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Spencer JPE, Vafeiadou K, Williams RJ, Vauzour D. Neuroinflammation: modulation by flavonoids and mechanisms of action. Mol Aspects Med 2012; 33:83-97. [PMID: 22107709 DOI: 10.1016/j.mam.2011.10.016] [Citation(s) in RCA: 229] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2011] [Accepted: 10/14/2011] [Indexed: 01/01/2023]
Abstract
Neuroinflammatory processes are known to contribute to the cascade of events culminating in the neuronal damage that underpins neurodegenerative disorders such as Parkinson's and Alzheimer's disease. Recently, there has been much interest in the potential neuroprotective effects of flavonoids, a group of plant secondary metabolites known to have diverse biological activity in vivo. With respect to the brain, flavonoids, such as those found in cocoa, tea, berries and citrus, have been shown to be highly effective in preventing age-related cognitive decline and neurodegeneration in both animals and humans. Evidence suggests that flavonoids may express such ability through a multitude of physiological functions, including an ability to modulate the brains immune system. This review will highlight the evidence for their potential to inhibit neuroinflammation through an attenuation of microglial activation and associated cytokine release, iNOS expression, nitric oxide production and NADPH oxidase activity. We will also detail the current evidence indicting that their regulation of these immune events appear to be mediated by their actions on intracellular signaling pathways, including the nuclear factor-κB (NF-κB) cascade and mitogen-activated protein kinase (MAPK) pathway. As such, flavonoids represent important precursor molecules in the quest to develop of a new generation of drugs capable of counteracting neuroinflammation and neurodegenerative disease.
Collapse
Affiliation(s)
- Jeremy P E Spencer
- Molecular Nutrition Group, Centre for Integrative Neuroscience and Neurodynamics, School of Chemistry, Food and Pharmacy, University of Reading, Reading RG6 6AP, UK.
| | | | | | | |
Collapse
|
37
|
Adams LS, Kanaya N, Phung S, Liu Z, Chen S. Whole blueberry powder modulates the growth and metastasis of MDA-MB-231 triple negative breast tumors in nude mice. J Nutr 2011; 141:1805-12. [PMID: 21880954 PMCID: PMC3174855 DOI: 10.3945/jn.111.140178] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Revised: 04/14/2011] [Accepted: 07/27/2011] [Indexed: 12/20/2022] Open
Abstract
Previous studies in our laboratory demonstrated that blueberry (BB) extract exhibited antitumor activity against MDA-MB-231 triple negative breast cancer (TNBC) cells and decreased metastatic potential in vitro. The current study tested 2 doses of whole BB powder, 5 and 10% (wt:wt) in the diet, against MDA-MB-231 tumor growth in female nude mice. In this study, tumor volume was 75% lower in mice fed the 5% BB diet and 60% lower in mice fed the 10% BB diet than in control mice (P ≤ 0.05). Tumor cell proliferation (Ki-67) was lower in the 5 and 10% BB-fed mice and cell death (Caspase 3) was greater in the 10% BB-fed mice compared to control mice (P ≤ 0.05). Gene analysis of tumor tissues from the 5% BB-fed mice revealed significantly altered expression of genes important to inflammation, cancer, and metastasis, specifically, Wnt signaling, thrombospondin-2, IL-13, and IFNγ. To confirm effects on Wnt signaling, analysis of tumor tissues from 5% BB-fed mice revealed lower β-catenin expression and glycogen synthase kinase-3β phosphorylation with greater expression of the β-catenin inhibitory protein adenomatous polyposis coli compared to controls. A second study tested the ability of the 5% BB diet to inhibit MDA-MB-231-luc-D3H2LN metastasis in vivo. In this study, 5% BB-fed mice developed 70% fewer liver metastases (P = 0.04) and 25% fewer lymph node metastases (P = 0.09) compared to control mice. This study demonstrates the oral antitumor and metastasis activity of whole BB powder against TNBC in mice.
Collapse
Affiliation(s)
- Lynn S. Adams
- Division of Tumor Cell Biology, Beckman Research Institute of the City of Hope, Duarte, CA
| | - Noriko Kanaya
- Division of Tumor Cell Biology, Beckman Research Institute of the City of Hope, Duarte, CA
| | - Sheryl Phung
- Division of Tumor Cell Biology, Beckman Research Institute of the City of Hope, Duarte, CA
| | - Zheng Liu
- Division of Tumor Cell Biology, Beckman Research Institute of the City of Hope, Duarte, CA
| | - Shiuan Chen
- Division of Tumor Cell Biology, Beckman Research Institute of the City of Hope, Duarte, CA
| |
Collapse
|
38
|
Ascierto ML, Giorgi VD, Liu Q, Bedognetti D, Spivey TL, Murtas D, Uccellini L, Ayotte BD, Stroncek DF, Chouchane L, Manjili MH, Wang E, Marincola FM. An immunologic portrait of cancer. J Transl Med 2011; 9:146. [PMID: 21875439 PMCID: PMC3175185 DOI: 10.1186/1479-5876-9-146] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Accepted: 08/29/2011] [Indexed: 12/31/2022] Open
Abstract
The advent of high-throughput technology challenges the traditional histopathological classification of cancer, and proposes new taxonomies derived from global transcriptional patterns. Although most of these molecular re-classifications did not endure the test of time, they provided bulk of new information that can reframe our understanding of human cancer biology. Here, we focus on an immunologic interpretation of cancer that segregates oncogenic processes independent from their tissue derivation into at least two categories of which one bears the footprints of immune activation. Several observations describe a cancer phenotype where the expression of interferon stimulated genes and immune effector mechanisms reflect patterns commonly observed during the inflammatory response against pathogens, which leads to elimination of infected cells. As these signatures are observed in growing cancers, they are not sufficient to entirely clear the organism of neoplastic cells but they sustain, as in chronic infections, a self-perpetuating inflammatory process. Yet, several studies determined an association between this inflammatory status and a favorable natural history of the disease or a better responsiveness to cancer immune therapy. Moreover, these signatures overlap with those observed during immune-mediated cancer rejection and, more broadly, immune-mediated tissue-specific destruction in other immune pathologies. Thus, a discussion concerning this cancer phenotype is warranted as it remains unknown why it occurs in immune competent hosts. It also remains uncertain whether a genetically determined response of the host to its own cancer, the genetic makeup of the neoplastic process or a combination of both drives the inflammatory process. Here we reflect on commonalities and discrepancies among studies and on the genetic or somatic conditions that may cause this schism in cancer behavior.
Collapse
Affiliation(s)
- Maria Libera Ascierto
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, Maryland, 20892, USA
- Department of Internal Medicine, University of Genoa, Italy
- Center of Excellence for Biomedical Research (CEBR), Genoa, Italy
| | - Valeria De Giorgi
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - Qiuzhen Liu
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - Davide Bedognetti
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, Maryland, 20892, USA
- Center of Excellence for Biomedical Research (CEBR), Genoa, Italy
- Department of Oncology, Biology and Genetics and National Cancer Research Institute of Genoa, Italy
| | - Tara L Spivey
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - Daniela Murtas
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - Lorenzo Uccellini
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - Ben D Ayotte
- Department of Biology, Northern Michigan University, Marquette, MI 49855,USA
| | - David F Stroncek
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - Lotfi Chouchane
- Weill Cornell Medical College in Qatar, Education City, Doha Qatar Box 24144
| | - Masoud H Manjili
- Department of Microbiology & Immunology, Virginia Commonwealth University Massey Cancer Center, Richmond, VA 23298, USA
| | - Ena Wang
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - Francesco M Marincola
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, Maryland, 20892, USA
| |
Collapse
|
39
|
Schwartz JL, Shajahan AN, Clarke R. The Role of Interferon Regulatory Factor-1 (IRF1) in Overcoming Antiestrogen Resistance in the Treatment of Breast Cancer. Int J Breast Cancer 2011; 2011:912102. [PMID: 22295238 PMCID: PMC3262563 DOI: 10.4061/2011/912102] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2011] [Revised: 04/29/2011] [Accepted: 05/09/2011] [Indexed: 12/20/2022] Open
Abstract
Resistance to endocrine therapy is common among breast cancer patients with estrogen receptor alpha-positive (ER+) tumors and limits the success of this therapeutic strategy. While the mechanisms that regulate endocrine responsiveness and cell fate are not fully understood, interferon regulatory factor-1 (IRF1) is strongly implicated as a key regulatory node in the underlying signaling network. IRF1 is a tumor suppressor that mediates cell fate by facilitating apoptosis and can do so with or without functional p53. Expression of IRF1 is downregulated in endocrine-resistant breast cancer cells, protecting these cells from IRF1-induced inhibition of proliferation and/or induction of cell death. Nonetheless, when IRF1 expression is induced following IFNγ treatment, antiestrogen sensitivity is restored by a process that includes the inhibition of prosurvival BCL2 family members and caspase activation. These data suggest that a combination of endocrine therapy and compounds that effectively induce IRF1 expression may be useful for the treatment of many ER+ breast cancers. By understanding IRF1 signaling in the context of endocrine responsiveness, we may be able to develop novel therapeutic strategies and better predict how patients will respond to endocrine therapy.
Collapse
Affiliation(s)
- J L Schwartz
- Georgetown University Medical Center, W401 Research Building, 3970 Reservoir Road, NW, Washington, DC 20057, USA
| | | | | |
Collapse
|
40
|
Wang E, Bussom S, Chen J, Quinn C, Bedognetti D, Lam W, Guan F, Jiang Z, Mark Y, Zhao Y, Stroncek DF, White J, Marincola FM, Cheng YC. Interaction of a traditional Chinese Medicine (PHY906) and CPT-11 on the inflammatory process in the tumor microenvironment. BMC Med Genomics 2011; 4:38. [PMID: 21569348 PMCID: PMC3117677 DOI: 10.1186/1755-8794-4-38] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Accepted: 05/11/2011] [Indexed: 12/18/2022] Open
Abstract
Background - Traditional Chinese Medicine (TCM) has been used for thousands of years to treat or prevent diseases, including cancer. Good manufacturing practices (GMP) and sophisticated product analysis (PhytomicsQC) to ensure consistency are now available allowing the assessment of its utility. Polychemical Medicines, like TCM, include chemicals with distinct tissue-dependent pharmacodynamic properties that result in tissue-specific bioactivity. Determining the mode of action of these mixtures was previously unsatisfactory; however, information rich RNA microarray technologies now allow for thorough mechanistic studies of the effects complex mixtures. PHY906 is a long used four herb TCM formula employed as an adjuvant to relieve the side effects associated with chemotherapy. Animal studies documented a decrease in global toxicity and an increase in therapeutic effectiveness of chemotherapy when combined with PHY906. Methods - Using a systems biology approach, we studied tumor tissue to identify reasons for the enhancement of the antitumor effect of CPT-11 (CPT-11) by PHY906 in a well-characterized pre-clinical model; the administration of PHY906 and CPT-11 to female BDF-1 mice bearing subcutaneous Colon 38 tumors. Results - We observed that 1) individually PHY906 and CPT-11 induce distinct alterations in tumor, liver and spleen; 2) PHY906 alone predominantly induces repression of transcription and immune-suppression in tumors; 3) these effects are reverted in the presence of CPT-11, with prevalent induction of pro-apoptotic and pro-inflammatory pathways that may favor tumor rejection. Conclusions - PHY906 together with CPT-11 triggers unique changes not activated by each one alone suggesting that the combination creates a unique tissue-specific response.
Collapse
Affiliation(s)
- Ena Wang
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Guan YQ, Li Z, Chen J, Tao H, Wang W, Zheng Z, Li L, Liu JM. Pathway of programmed cell death in HeLa cells induced by polymeric anti-cancer drugs. Biomaterials 2011; 32:3637-46. [DOI: 10.1016/j.biomaterials.2011.01.060] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Accepted: 01/20/2011] [Indexed: 02/02/2023]
|
42
|
Morello S, Sorrentino R, Montinaro A, Luciano A, Maiolino P, Ngkelo A, Arra C, Adcock IM, Pinto A. NK1.1 cells and CD8 T cells mediate the antitumor activity of Cl-IB-MECA in a mouse melanoma model. Neoplasia 2011; 13:365-73. [PMID: 21472141 PMCID: PMC3071085 DOI: 10.1593/neo.101628] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Revised: 01/10/2011] [Accepted: 01/17/2011] [Indexed: 11/18/2022]
Abstract
Cl-IB-MECA, synthetic A(3) adenosine receptor agonist, is a potential anticancer agent. In this study, we have examined the effect of Cl-IB-MECA in a mouse melanoma model. Cl-IB-MECA significantly inhibited tumor growth in immune-competent mice. Notably, the number of tumor-infiltrating NK1.1(+) cells and CD8(+) T cells was significantly increased in Cl-IB-MECA-treated mice. This effect was correlated with high levels of tumor necrosis factor α (TNF-α) and interferon γ in melanoma tissue. Depletion of either CD8(+) T cells or NK1.1(+) cells completely abrogated the antitumor effect of Cl-IB-MECA. Accordingly, Cl-IB-MECA did not affect tumor growth in nude mice. In addition, we also found that the number of mature and active conventional dendritic cells at the tumor site was increased after Cl-IB-MECA administration. Moreover, Cl-IB-MECA significantly increased TNF-α and IL-12p40 release from splenic CD11c(+) cells. In conclusion, our study provides novel insights into the mechanism by which Cl-IB-MECA leads to an effective antitumor response that involves the activation of natural killer cells and CD8(+) T cells and further highlights its therapeutic potential.
Collapse
MESH Headings
- Adenosine/analogs & derivatives
- Adenosine/pharmacology
- Adenosine/therapeutic use
- Animals
- Antimetabolites, Antineoplastic/pharmacology
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/physiology
- Cell Line, Tumor
- Chemotaxis, Leukocyte/drug effects
- Female
- Immunity, Cellular/drug effects
- Immunity, Cellular/physiology
- Killer Cells, Natural/metabolism
- Killer Cells, Natural/physiology
- Lymphocyte Activation/drug effects
- Melanoma, Experimental/drug therapy
- Melanoma, Experimental/immunology
- Melanoma, Experimental/pathology
- Mice
- Mice, Inbred C57BL
- Mice, Nude
- Skin Neoplasms/drug therapy
- Skin Neoplasms/immunology
- Skin Neoplasms/pathology
- Tumor Microenvironment/drug effects
Collapse
Affiliation(s)
- Silvana Morello
- Department of Pharmaceutical Sciences, Biomedical Section, University of Salerno, Fisciano (SA), Italy
| | - Rosalinda Sorrentino
- Department of Pharmaceutical Sciences, Biomedical Section, University of Salerno, Fisciano (SA), Italy
| | - Antonella Montinaro
- Department of Pharmaceutical Sciences, Biomedical Section, University of Salerno, Fisciano (SA), Italy
| | | | | | - Anta Ngkelo
- National Heart and Lung Institute, Imperial College, London, UK
| | - Claudio Arra
- National Cancer Institute “G. Pascale,” Naples, Italy
| | - Ian M Adcock
- National Heart and Lung Institute, Imperial College, London, UK
| | - Aldo Pinto
- Department of Pharmaceutical Sciences, Biomedical Section, University of Salerno, Fisciano (SA), Italy
| |
Collapse
|
43
|
Kravchenko-Balasha N, Klein S, Safrai M, Levitzki A. Contribution of gross chromosomal changes to HPV16-induced transformation. MOLECULAR BIOSYSTEMS 2011; 7:1501-11. [DOI: 10.1039/c0mb00284d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
44
|
Guan YQ, Li Z, Liu JM. Death signal transduction induced by co-immobilized TNF-α plus IFN-γ and the development of polymeric anti-cancer drugs. Biomaterials 2010; 31:9074-85. [DOI: 10.1016/j.biomaterials.2010.08.044] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2010] [Accepted: 08/19/2010] [Indexed: 01/22/2023]
|
45
|
Abstract
Susceptibility to most autoimmune diseases is dependent on polygenic inheritance, environmental factors, and poorly defined stochastic events. One of the significant challenges facing autoimmune disease research is in identifying the specific events that trigger loss of tolerance and autoimmunity. Although many intrinsic factors, including age, sex, and genetics, contribute to autoimmunity, extrinsic factors such as drugs, chemicals, microbes, or other environmental factors can also act as important initiators. This review explores how certain extrinsic factors, namely, drugs and chemicals, can promote the development of autoimmunity, focusing on a few better characterized agents that, in most instances, have been shown to produce autoimmune manifestations in human populations. Mechanisms of autoimmune disease induction are discussed in terms of research obtained using specific animal models. Although a number of different pathways have been delineated for drug/chemical-induced autoimmunity, some similarities do exist, and a working model is proposed.
Collapse
Affiliation(s)
- K Michael Pollard
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California, 92037, USA
| | | | | |
Collapse
|
46
|
Cheon H, Cho JM, Kim S, Baek SH, Lee MK, Kim KW, Yu SW, Solinas G, Kim SS, Lee MS. Role of JNK activation in pancreatic beta-cell death by streptozotocin. Mol Cell Endocrinol 2010; 321:131-7. [PMID: 20176078 DOI: 10.1016/j.mce.2010.02.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2009] [Revised: 01/14/2010] [Accepted: 02/12/2010] [Indexed: 01/18/2023]
Abstract
c-Jun N-terminal kinase (JNK) is activated by cellular stress and plays critical roles in diverse types of cell death. However, role of JNK in beta-cell injury is obscure. We investigated the role for JNK in streptozotocin (STZ)-induced beta-cell death. STZ induced JNK activation in insulinoma or islet cells. JNK inhibitors attenuated insulinoma or islet cell death by STZ. STZ-induced JNK activation was decreased by PARP inhibitors, suggesting that JNK activation is downstream of PARP-1. Phosphatase inhibitors induced activation of JNK and abrogated the suppression of STZ-induced JNK activation by PARP inhibitors, suggesting that the inhibition of phosphatases is involved in the activation of JNK by STZ. STZ induced production of reactive oxygen species (ROS) as potential inhibitors of phosphatases, which was suppressed by PARP inhibitors. PARP-1 siRNA attenuated insulinoma cell death and JNK activation after STZ treatment, which was reversed by MKP (MAP kinase phosphatase)-1 siRNA. These results suggest that JNK is activated by STZ downstream of PARP-1 through inactivation of phosphatases such as MKP, which plays important roles in STZ-induced beta-cell death.
Collapse
Affiliation(s)
- Hwanju Cheon
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Najjar I, Fagard R. STAT1 and pathogens, not a friendly relationship. Biochimie 2010; 92:425-44. [PMID: 20159032 PMCID: PMC7117016 DOI: 10.1016/j.biochi.2010.02.009] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2009] [Accepted: 02/09/2010] [Indexed: 12/21/2022]
Abstract
STAT1 belongs to the STAT family of transcription factors, which comprises seven factors: STAT1, STAT2, STAT3, STAT4, STAT5A, STAT5B and STAT6. STAT1 is a 91 kDa protein originally identified as the mediator of the cellular response to interferon (IFN) α, and thereafter found to be a major component of the cellular response to IFNγ. STAT1 is, in fact, involved in the response to several cytokines and to growth factors. It is activated by cytokine receptors via kinases of the JAK family. STAT1 becomes phosphorylated and forms a dimer which enters the nucleus and triggers the transcription of its targets. Although not lethal at birth, selective gene deletion of STAT1 in mice leads to rapid death from severe infections, demonstrating its major role in the response to pathogens. Similarly, in humans who do not express STAT1, there is a lack of resistance to pathogens leading to premature death. This indicates a key, non-redundant function of STAT1 in the defence against pathogens. Thus, to successfully infect organisms, bacterial, viral or parasitic pathogens must overcome the activity of STAT1, and almost all the steps of this pathway can be blocked or inhibited by proteins produced in infected cells. Interestingly, some pathogens, like the oncogenic Epstein–Barr virus, have evolved a strategy which uses STAT1 activation.
Collapse
Affiliation(s)
- Imen Najjar
- INSERM Unité 978, SMBH, 74 rue Marcel Cachin, Bobigny-cedex 93017, France.
| | | |
Collapse
|
48
|
IRF-1 transcriptionally upregulates PUMA, which mediates the mitochondrial apoptotic pathway in IRF-1-induced apoptosis in cancer cells. Cell Death Differ 2009; 17:699-709. [PMID: 19851330 PMCID: PMC2838929 DOI: 10.1038/cdd.2009.156] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Interferon Regulatory Factor-1 (IRF-1) is a transcription factor which acts as a tumor suppressor and causes apoptosis in cancer cells. We evaluated IRF-1 induced apoptosis in gastric cancer cell lines. We established stable clones in AGS cells that have a tetracycline inducible IRF-1 expression system. We used these clones and recombinant adenovirus expressing IRF-1 to explore the mechanism of IRF-1 induced apoptosis in gastric cancer. Expression of IRF-1 causes apoptosis in gastric cancer cell lines as demonstrated by phosphatidylserine exposure and cleavage of caspase-8, caspase-3, and Bid with mitochondrial release of cytochrome c. However, inhibition of caspase-8 and Bid did not inhibit apoptosis and did not decrease cleaved caspase-9 or mitochondrial release of cytochrome c. We then demonstrate that IRF-1 up-regulates PUMA (p53 up-regulated modulator of apoptosis), that is known to activate apoptosis by the intrinsic pathway; this can be p53 independent. IRF-1 binds to distinct sites in the promoter of PUMA and activates PUMA transcription. Moreover, molecular markers of mitochondrial apoptosis are eliminated in PUMA knockout and knockdown cells and phospatidylserine exposure is decreased dramatically. Finally, we demonstrate that IFN-γ induces IRF-1 mediated up-regulation of PUMA in cancer cells. We conclude that IRF-1 can induce apoptosis by the intrinsic pathway independent of the extrinsic pathway by up-regulation of PUMA.
Collapse
|
49
|
Yi JY, Jung YJ, Choi SS, Chung E. TNF-alpha downregulates E-cadherin and sensitizes response to γ-irradiation in Caco-2 cells. Cancer Res Treat 2009; 41:164-70. [PMID: 19809566 DOI: 10.4143/crt.2009.41.3.164] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2009] [Accepted: 06/01/2009] [Indexed: 12/17/2022] Open
Abstract
PURPOSE The purpose of the present study was to assess the biological effects of TNF-alpha in Caco-2 well-differentiated colon adenocarcinoma cells and to determine radiation sensitivity in order to develop TNF-alpha into a cancer therapeutic agent. MATERIALS AND METHODS A cell viability test was conducted via a colorimetric and colony forming assay after 1 day and 3 days of incubation with TNF-alpha. Western blotting analysis and immunofluorescence staining were conducted to explore TNF-alpha-induced morphological and molecular changes in the adhesion molecules, E-cadherin and claudin-4. The effects of γ-irradiation at a dose of 2 Gy on cell survival were evaluated by a clonogenic assay. The molecular changes in apoptosis-regulatory proteins were assessed by Western blotting. RESULTS Caco-2 cells were highly resistant to TNF alpha-induced cell death and 2 Gy of γ-irradiation. However, we observed the downregulation of the adherens junctional protein, E-cadherin and translocation of tight junctional protein, claudin-4 from the membrane to the cytosol induced by TNF-alpha treatment which would indicate cell-cell junction disruptions. These alterations of junctional proteins influenced the regulation of cell death in response to 2 Gy of γ-irradiation. The combined treatment of TNF-alpha with 2 Gy of γ-irradiation reduced the survival of Caco-2 cells by down-regulating bcl-xl and activating JNK pathways. CONCLUSION These results suggest that TNF-alpha might be potentially applied as a therapeutic agent in order to enhance sensitivity to 2 Gy of γ-irradiation administered in radiotherapy for the treatment of human colon cancer.
Collapse
Affiliation(s)
- Jae Youn Yi
- Lab of Modulation of Radiobiological Response, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | | | | | | |
Collapse
|
50
|
Cavalli LR, Riggins RB, Wang A, Clarke R, Haddad BR. Frequent loss of heterozygosity at the interferon regulatory factor-1 gene locus in breast cancer. Breast Cancer Res Treat 2009; 121:227-31. [PMID: 19697121 DOI: 10.1007/s10549-009-0509-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2009] [Accepted: 08/08/2009] [Indexed: 12/23/2022]
Abstract
The interferon regulatory factor-1 (IRF1) gene, localized on chromosome 5q31.1, is mutated or rearranged in several cancers including some hematopoietic and gastric cancers. However, whether loss of IRF1 occurs in sporadic breast cancer is unknown. Loss of 5q12-31 is reported in 11% of sporadic breast cancers, and high-resolution array-CGH studies have shown loss at 5q31.1 in 50% of breast cancers with a mutated BRCA1 gene. Functionally, overexpression of IRF1 reduces, and a dominant negative IRF1 construct increases, tumorigenesis of human breast cancer xenografts. Taken together, these observations indicate that the IRF1 gene may play a potentially important role as a breast cancer tumor suppressor gene. In this study, we investigated allelic loss of the IRF1 gene in breast tumor specimens from 52 women with invasive breast cancer using an IRF1 intragenic dinucleotide polymorphic marker. Thirty-seven cases were informative. LOH at the IRF1 locus was detected in 32% of these informative cases (12/37). There was a significant association between IRF1 loss and both older age (P = 0.0167) and earlier stage (Stages 1 and 2) (P = 0.0165). To assess the association of IRF1 mRNA expression with clinical outcomes in breast cancer, we studied data from two published gene expression microarray datasets. In breast cancer patients, low IRF1 mRNA expression is strongly correlated with both risk of recurrence (OR = 3.00; P = 0.003; n = 273 cases) and risk of death (OR = 4.18; P = 0.004; n = 191 cases). Our findings strongly imply a tumor suppressor role for the IRF1 gene in breast cancer.
Collapse
Affiliation(s)
- Luciane R Cavalli
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University School of Medicine, Washington, DC 20057, USA
| | | | | | | | | |
Collapse
|