1
|
Ellis SLS, Dada S, Nohara LL, Saranchova I, Munro L, Pfeifer CG, Eyford BA, Morova T, Williams DE, Cheng P, Lack NA, Andersen RJ, Jefferies WA. Curcuphenol possesses an unusual histone deacetylase enhancing activity that counters immune escape in metastatic tumours. Front Pharmacol 2023; 14:1119620. [PMID: 37637416 PMCID: PMC10449465 DOI: 10.3389/fphar.2023.1119620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 07/03/2023] [Indexed: 08/29/2023] Open
Abstract
Curcuphenol, a common component of the culinary spices, naturally found in marine invertebrates and plants, has been identified as a novel candidate for reversing immune escape by restoring expression of the antigen presentation machinery (APM) in invasive cancers, thereby resurrecting the immune recognition of metastatic tumours. Two synthetic curcuphenol analogues, were prepared by informed design that demonstrated consistent induction of APM expression in metastatic prostate and lung carcinoma cells. Both analogues were subsequently found to possess a previously undescribed histone deacetylase (HDAC)-enhancing activity. Remarkably, the H3K27ac ChIPseq analysis of curcuphenol-treated cells reveals that the induced epigenomic marks closely resemble the changes in genome-wide pattern observed with interferon-γ, a cytokine instrumental for orchestrating innate and adaptive immunity. These observations link dietary components to modifying epigenetic programs that modulate gene expression guiding poised immunity.
Collapse
Affiliation(s)
- Samantha L. S. Ellis
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Sarah Dada
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada
- Departments of Medical Genetics, Zoology, and Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Lilian L. Nohara
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Iryna Saranchova
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada
- Departments of Medical Genetics, Zoology, and Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Lonna Munro
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada
| | - Cheryl G. Pfeifer
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada
| | - Brett A. Eyford
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Tunc Morova
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada
| | - David E. Williams
- Departments of Chemistry and Earth Ocean, and Atmospheric Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Ping Cheng
- Departments of Chemistry and Earth Ocean, and Atmospheric Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Nathan A. Lack
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada
- School of Medicine, Koç University, Istanbul, Türkiye
| | - Raymond J. Andersen
- Departments of Chemistry and Earth Ocean, and Atmospheric Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Wilfred A. Jefferies
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada
- Departments of Medical Genetics, Zoology, and Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
2
|
Anang V, Singh A, Kottarath SK, Verma C. Receptors of immune cells mediates recognition for tumors. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 194:219-267. [PMID: 36631194 DOI: 10.1016/bs.pmbts.2022.09.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Over the last few decades, the immune system has been steered toward eradication of cancer cells with the help of cancer immunotherapy. T cells, B cells, monocytes/macrophages, dendritic cells, T-reg cells, and natural killer (NK) cells are some of the numerous immune cell types that play a significant part in cancer cell detection and reduction of inflammation, and the antitumor response. Briefly stated, chimeric antigen receptors, adoptive transfer and immune checkpoint modulators are currently the subjects of research focus for successful immunotherapy-based treatments for a variety of cancers. This chapter discusses ongoing investigations on the mechanisms and recent developments by which receptors of immune cells especially that of lymphocytes and monocytes/macrophages regulate the detection of immune system leading to malignancies. We will also be looking into the treatment strategies based on these mechanisms.
Collapse
Affiliation(s)
- Vandana Anang
- International Center for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | | | - Sarat Kumar Kottarath
- Department of Experimental Therapeutics, MD Anderson Cancer Center, Huston, TX, United States.
| | - Chaitenya Verma
- Department of Pathology, Wexner Medical Center, Ohio State University, Columbus, OH, United States.
| |
Collapse
|
3
|
Song IH, Kim YA, Heo SH, Bang WS, Park HS, Choi YH, Lee H, Seo JH, Cho Y, Jung SW, Kim HJ, Ahn SH, Lee HJ, Gong G. The Association of Estrogen Receptor Activity, Interferon Signaling, and MHC Class I Expression in Breast Cancer. Cancer Res Treat 2022; 54:1111-1120. [PMID: 34942685 PMCID: PMC9582481 DOI: 10.4143/crt.2021.1017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 12/20/2021] [Indexed: 11/30/2022] Open
Abstract
PURPOSE The expression of major histocompatibility complex class I (MHC I) has previously been reported to be negatively associated with estrogen receptor (ER) expression. Furthermore, MHC I expression, level of tumor-infiltrating lymphocytes (TILs), and expression of interferon (IFN) mediator MxA are positively associated with one another in human breast cancers. This study aimed to investigate the mechanisms of association of MHC I with ER and IFN signaling. MATERIALS AND METHODS The human leukocyte antigen (HLA)-ABC protein expression was analyzed in breast cancer cell lines. The expressions of HLA-A and MxA mRNAs were analyzed in MCF-7 cells in Gene Expression Omnibus (GEO) data. ER and HLA-ABC expressions, Ki-67 labeling index and TIL levels in tumor tissue were also analyzed in ER+/ human epidermal growth factor receptor 2 (HER2)- breast cancer patients who randomly received either neoadjuvant chemotherapy or estrogen modulator treatment followed by resection. RESULTS HLA-ABC protein expression was decreased after β-estradiol treatment or hESR-GFP transfection and increased after fulvestrant or IFN-γ treatment in cell lines. In GEO data, HLA-A and MxA expression was increased after ESR1 shRNA transfection. In patients, ER Allred score was significantly lower and the HLA-ABC expression, TIL levels, and Ki-67 were significantly higher in the estrogen modulator treated group than the chemotherapy treated group. CONCLUSION MHC I expression and TIL levels might be affected by ER pathway modulation and IFN treatment. Further studies elucidating the mechanism of MHC I regulation could suggest a way to boost TIL influx in cancer in a clinical setting.
Collapse
Affiliation(s)
- In Hye Song
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | | | - Sun-Hee Heo
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Won Seon Bang
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | | | | | | | | | - Youngjin Cho
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Sung Wook Jung
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Hee Jeong Kim
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Sei Hyun Ahn
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Hee Jin Lee
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
- NeogenTC Corp., Seoul,
Korea
| | - Gyungyub Gong
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| |
Collapse
|
4
|
Aoun R, El Hadi C, Tahtouh R, El Habre R, Hilal G. Microarray analysis of breast cancer gene expression profiling in response to 2-deoxyglucose, metformin, and glucose starvation. Cancer Cell Int 2022; 22:123. [PMID: 35305635 PMCID: PMC8933915 DOI: 10.1186/s12935-022-02542-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 03/07/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Breast cancer (BC) is the most frequently diagnosed cancer in women. Altering glucose metabolism and its effects on cancer progression and treatment resistance is an emerging interest in BC research. For instance, combining chemotherapy with glucose-lowering drugs (2-deoxyglucose (2-DG), metformin (MET)) or glucose starvation (GS) has shown better outcomes than with chemotherapy alone. However, the genes and molecular mechanisms that govern the action of these glucose deprivation conditions have not been fully elucidated. Here, we investigated the differentially expressed genes in MCF-7 and MDA-MB-231 BC cell lines upon treatment with glucose-lowering drugs (2-DG, MET) and GS using microarray analysis to study the difference in biological functions between the glucose challenges and their effect on the vulnerability of BC cells. METHODS MDA-MB-231 and MCF-7 cells were treated with 20 mM MET or 4 mM 2-DG for 48 h. GS was performed by gradually decreasing the glucose concentration in the culture medium to 0 g/L, in which the cells remained with fetal bovine serum for one week. Expression profiling was carried out using Affymetrix Human Clariom S microarrays. Differentially expressed genes were obtained from the Transcriptome Analysis Console and enriched using DAVID and R packages. RESULTS Our results showed that MDA-MB-231 cells were more responsive to glucose deprivation than MCF-7 cells. Endoplasmic reticulum stress response and cell cycle inhibition were detected after all three glucose deprivations in MDA-MB-231 cells and only under the metformin and GS conditions in MCF-7 cells. Induction of apoptosis and inhibition of DNA replication were observed with all three treatments in MDA-MB-231 cells and metformin-treated MCF-7 cells. Upregulation of cellular response to reactive oxygen species and inhibition of DNA repair mechanisms resulted after metformin and GS administration in MDA-MB-231 cell lines and metformin-treated MCF-7 cells. Autophagy was induced after 2-DG treatment in MDA-MB-231 cells and after metformin in MCF-7 cells. Finally, inhibition of DNA methylation were observed only with GS in MDA-MB-231 cells. CONCLUSION The procedure used to process cancer cells and analyze their expression data distinguishes our study from others. GS had the greatest effect on breast cancer cells compared to 2-DG and MET. Combining MET and GS could restrain both cell lines, making them more vulnerable to conventional chemotherapy.
Collapse
Affiliation(s)
- Rita Aoun
- Cancer and Metabolism Laboratory, Faculty of Medicine, Saint-Joseph University, Beirut, Lebanon
| | | | - Roula Tahtouh
- Cancer and Metabolism Laboratory, Faculty of Medicine, Saint-Joseph University, Beirut, Lebanon
| | - Rita El Habre
- Cancer and Metabolism Laboratory, Faculty of Medicine, Saint-Joseph University, Beirut, Lebanon
| | - George Hilal
- Cancer and Metabolism Laboratory, Faculty of Medicine, Saint-Joseph University, Beirut, Lebanon.
| |
Collapse
|
5
|
Lv D, Chen J, Kang Y, Luo M, Chen H, Cui B, Wang L, Wang J, Zhou X, Feng Y, Huang L, Zhang P. Protein Kinase D3 Promotes the Reconstruction of OSCC Immune Escape Niche Via Regulating MHC-I and Immune Inhibit Molecules Expression. J Immunother 2021; 44:339-347. [PMID: 34545012 DOI: 10.1097/cji.0000000000000395] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 08/26/2021] [Indexed: 02/05/2023]
Abstract
Protein kinase D3 (PKD3) has been involved in various aspects of tumorigenesis and progression in many kinds of cancer types. However, whether PKD3 regulates immune escape in tumor microenvironment is rarely reported. Here, we explored the function and mechanism of PKD3 in reconstructing the immune escape niche of oral squamous cell carcinoma (OSCC). Both the Western blotting analysis in OSCC cells and the gene expression correlation analysis from The Cancer Genome Atlas shows that the expression of Fas and programmed cell death-ligand 1 (PD-L1) was positively correlated with PKD3, while major histocompatibility complex-I (MHC-I) was negatively correlated with PKD3. Knockdown of PKD3 significantly decreased the expression of Fas and PD-L1 and increased the expression of MHC-I. Furthermore, when PKD3 was overexpressed in oral precancerous cells, Fas, PD-L1, and MHC-I showed an opposite trend to that observed when PKD3 was knocked down. In addition, PKD3 knockdown decreased the secretion of transforming growth factor β, CC-chemokine ligand 21, interleukin-10 by OSCC cells. Finally, the tumor cell antigen, which was extracted from PKD3 knockdown OSCC cells, significantly induced the growth and activation of T lymphocytes. These results demonstrate that PKD3 promotes the immune escape of OSCC cells by regulating the expression of Fas, PD-L1, MHC-I, transforming growth factor β, CC-chemokine ligand 21, interleukin-10, and plays a key role in reconstructing the tumor immune escape niche.
Collapse
Affiliation(s)
- Die Lv
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
O'Connor EA, Westerdahl H. Trade-offs in expressed major histocompatibility complex diversity seen on a macroevolutionary scale among songbirds. Evolution 2021; 75:1061-1069. [PMID: 33666228 DOI: 10.1111/evo.14207] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 01/07/2021] [Accepted: 02/17/2021] [Indexed: 12/30/2022]
Abstract
To survive organisms must defend themselves against pathogens. Classical Major Histocompatibility Complex (MHC) genes play a key role in pathogen defense by encoding molecules involved in pathogen recognition. MHC gene diversity influences the variety of pathogens individuals can recognize and respond to and has consequently been a popular genetic marker for disease resistance in ecology and evolution. However, MHC diversity is predominantly estimated using genomic DNA (gDNA) with little knowledge of expressed diversity. This limits our ability to interpret the adaptive significance of variation in MHC diversity, especially in species with very many MHC genes such as songbirds. Here, we address this issue using phylogenetic comparative analyses of the number of MHC class I alleles (MHC-I diversity) in gDNA and complementary DNA (cDNA), that is, expressed alleles, across 13 songbird species. We propose three theoretical relationships that could be expected between genomic and expressed MHC-I diversity on a macroevolutionary scale and test which of these are best supported. In doing so, we show that significantly fewer MHC-I alleles than the number available are expressed, suggesting that optimal MHC-I diversity could be achieved by modulating gene expression. Understanding the relationship between genomic and expressed MHC diversity is essential for interpreting variation in MHC diversity in an evolutionary context.
Collapse
Affiliation(s)
- Emily A O'Connor
- Molecular Ecology and Evolution Lab, Department of Biology, Lund University, Lund, Sweden
| | - Helena Westerdahl
- Molecular Ecology and Evolution Lab, Department of Biology, Lund University, Lund, Sweden
| |
Collapse
|
7
|
Dennison L, Mohan AA, Yarchoan M. Tumor and Systemic Immunomodulatory Effects of MEK Inhibition. Curr Oncol Rep 2021; 23:23. [PMID: 33547983 PMCID: PMC8028056 DOI: 10.1007/s11912-020-01008-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/17/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Mitogen-activated protein kinase (MAPK) kinase (MEK) is an integral component of the RAS signaling pathway, one of the most frequently mutated pathways in cancer biology. MEK inhibitors were initially developed to directly target oncogenic signaling, but are recognized to have pleiotropic effects on both tumor cells and lymphocytes. Here, we review the preclinical and clinical evidence that MEK inhibition is immunomodulatory and discuss the potential rationale for combining MEK inhibitors with systemic immunotherapies. RECENT FINDINGS MEK inhibition may modulate the tumor microenvironment (TME) through direct effects on both tumor cells and immune cells. Despite encouraging evidence that MEK inhibition can reprogram the tumor microenvironment (TME) and augment anti-tumor immunity regardless of KRAS/BRAF status, recent clinical outcome studies combining MEK inhibition with systemic immunotherapy have yielded mixed results. The combination of MEK inhibitors plus systemic immunotherapies has been tolerable, but has thus far failed to demonstrate clear evidence of synergistic clinical activity. These results underscore the need to understand the appropriate therapeutic context for this combination. MEK inhibitors have the potential to inhibit oncogenic signaling and reprogram the tumor immune microenvironment, representing an attractive therapy to combine with systemic immunotherapies. Ongoing preclinical and clinical studies will further clarify the immunomodulatory effects of MEK inhibitors to inform the design of rational therapeutic combinations.
Collapse
Affiliation(s)
- Lauren Dennison
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, 21231, USA
| | - Aditya A Mohan
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, 21231, USA
| | - Mark Yarchoan
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, 21231, USA.
| |
Collapse
|
8
|
Retained or altered expression of major histocompatibility complex class I in patient-derived xenograft models in breast cancer. Immunol Res 2020; 67:469-477. [PMID: 31900802 DOI: 10.1007/s12026-019-09109-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The expression of major histocompatibility complex class I (MHC I) in tumor cells is regulated by interferon signaling, and it is an important factor in the efficacy of cytotoxic T cell-dependent immunotherapy. To determine the impact of immune cells in MHC I expression on tumor cells, we compared the expression of MHC I in tumor cells derived from primary breast cancers and patient-derived xenograft (PDX) models. MHC I and myxovirus resistance gene A (MxA) expression were analyzed using immunohistochemistry in 23 cases of tumor tissue and corresponding primary and secondary PDXs. The median H score of MHC I was 210 (0-300) in patient tumor tissues, 197.5 (0-300) in primary PDX tumors, and 157.5 (5-300) in secondary PDX tumors. Cases were divided into four groups based on the difference in MHC I expression between the patient tumor tissues and secondary PDXs. Eleven cases constituted the high MHC I group, four constituted the low MHC I group, six comprised the decreased MHC I group, and two comprised the increased MHC I group. MHC I and MxA expressions in each tumor were weakly correlated within patients' tumors, while strongly correlated within PDX models. Retained or altered expression of MHC I in breast cancer PDXs reveals the presence of intrinsic and extrinsic interferon signaling pathways in tumor cells. Thus, considering MHC I expression in PDX is important when using PDX models to evaluate the efficacy of cancer immunotherapy in a preclinical setting.
Collapse
|
9
|
Shen Y, Wei X, Jin S, Wu Y, Zhao W, Xu Y, Pan L, Zhou Z, Chen S. TCR-mimic antibody-drug conjugates targeting intracellular tumor-specific mutant antigen KRAS G12V mutation. Asian J Pharm Sci 2020; 15:777-785. [PMID: 33363632 PMCID: PMC7750800 DOI: 10.1016/j.ajps.2020.01.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 12/29/2019] [Accepted: 01/22/2020] [Indexed: 12/22/2022] Open
Abstract
Limited clinical application of antibody-drug conjugates (ADCs) targeting tumor associated antigens (TAAs) is usually caused by on-target off-tumor side effect. Tumor-specific mutant antigens (TSMAs) only expressed in tumor cells which are ideal targets for ADCs. In addition, intracellular somatic mutant proteins can be presented on the cell surface by human leukocyte antigen class I (HLA I)molecules forming tumor-specific peptide/HLA I complexes. KRAS G12V mutation frequently occurred in varied cancer and was verified as a promising target for cancer therapy. In this study, we generated two TCR-mimic antibody-drug conjugates (TCRm-ADCs), 2E8-MMAE and 2A5-MMAE, targeting KRAS G12V/HLA-A*0201 complex, which mediated specific antitumor activity in vitro and in vivo without obvious toxicity. Our findings are the first time validate the strategy of TCRm-ADCs targeting intracellular TSMAs, which improves the safety of antibody-based drugs and provides novel strategy for precision medicine in cancer therapy.
Collapse
Affiliation(s)
- Ying Shen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xiaoyue Wei
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,Huabo Biopharm Co., Ltd., Shanghai 201203, China
| | - Shijie Jin
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yue Wu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wenbin Zhao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yingchun Xu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Liqiang Pan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhan Zhou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Shuqing Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
10
|
Gustafsson JR, Katsioudi G, Degn M, Ejlerskov P, Issazadeh-Navikas S, Kornum BR. DNMT1 regulates expression of MHC class I in post-mitotic neurons. Mol Brain 2018; 11:36. [PMID: 29970123 PMCID: PMC6029374 DOI: 10.1186/s13041-018-0380-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 06/21/2018] [Indexed: 02/06/2023] Open
Abstract
Major Histocompability Complex I (MHC-I) molecules present cellularly derived peptides to the adaptive immune system. Generally MHC-I is not expressed on healthy post-mitotic neurons in the central nervous system, but it is known to increase upon immune activation such as viral infections and also during neurodegenerative processes. MHC-I expression is known to be regulated by the DNA methyltransferase DNMT1 in non-neuronal cells. Interestingly DNMT1 expression is high in neurons despite these being non-dividing. This suggests a role for DNMT1 in neurons beyond the classical re-methylation of DNA after cell division. We thus investigated whether DNMT1 regulates MHC-I in post-mitotic neurons. For this we used primary cultures of mouse cerebellar granule neurons (CGNs). Our results showed that knockdown of DNMT1 in CGNs caused upregulation of some, but not all subtypes of MHC-I genes. This effect was synergistically enhanced by subsequent IFNγ treatment. Overall MHC-I protein level was not affected by knockdown of DNMT1 in CGNs. Instead our results show that the relative MHC-I expression levels among the different MHC subtypes is regulated by DNMT1 activity. In conclusion, we show that while the mouse H2-D1/L alleles are suppressed in neurons by DNMT1 activity under normal circumstances, the H2-K1 allele is not. This finding is particularly important in two instances. One: in the context of CNS autoimmunity with epitope presentation by specific MHC-I subtypes where this allele specific regulation might become important; and two: in amyotropic lateral sclerosis (ALS) where H2-K but not H2-D protects motor neurons from ALS astrocyte-induced toxicity in a mouse model of ALS.
Collapse
Affiliation(s)
- Julie Ry Gustafsson
- Department of Clinical Biochemistry, Molecular Sleep Laboratory, Rigshospitalet, Glostrup, Nordre Ringvej 57, 2600, Glostrup, Denmark
| | - Georgia Katsioudi
- Department of Clinical Biochemistry, Molecular Sleep Laboratory, Rigshospitalet, Glostrup, Nordre Ringvej 57, 2600, Glostrup, Denmark
| | - Matilda Degn
- Department of Clinical Biochemistry, Molecular Sleep Laboratory, Rigshospitalet, Glostrup, Nordre Ringvej 57, 2600, Glostrup, Denmark
| | - Patrick Ejlerskov
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Ole Maaløes Vej 5, 2200, Copenhagen N, Denmark
| | - Shohreh Issazadeh-Navikas
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Ole Maaløes Vej 5, 2200, Copenhagen N, Denmark
| | - Birgitte Rahbek Kornum
- Department of Clinical Biochemistry, Molecular Sleep Laboratory, Rigshospitalet, Glostrup, Nordre Ringvej 57, 2600, Glostrup, Denmark. .,Department of Clinical Neurophysiology, Danish Center for Sleep Medicine, Rigshospitalet, Glostrup, Denmark. .,Molecular Sleep Laboratory, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
11
|
Watt SK, Hasselbalch HC, Skov V, Kjær L, Thomassen M, Kruse TA, Burton M, Gögenur I. Whole Blood Gene Expression Profiling in patients undergoing colon cancer surgery identifies differential expression of genes involved in immune surveillance, inflammation and carcinogenesis. Surg Oncol 2018; 27:208-215. [PMID: 29937173 DOI: 10.1016/j.suronc.2018.03.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 03/13/2018] [Accepted: 03/26/2018] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Cancer surgery may represent a potential risk of enhanced growth and metastatic ability of residual cancer cells due to post-operative immune dysfunction. This study identifies changes in transcription of genes involved in immune surveillance, immune suppression and carcinogenesis in the post-operative period of laparoscopic colon-cancer surgery within an ERAS regime. METHODS Patients undergoing elective, curatively intended laparoscopic surgery for colon cancer stage I-III UICC were included in the study. Patients followed standard of care in an ERAS setting. Whole blood gene expression profiling (WBGP) was performed on the day prior to surgery and 1, 2, 3 and 10-14 days after surgery. Samples were collected in Paxgene tubes and Labeled cDNA was fragmented and hybridized to Affymetrix GeneChip™ 2.0. Results were corrected for multiple hypothesis testing using the false discovery rate. Pathway analysis was performed through the Molecular Signature Database. Paired fold changes of gene expression were calculated for post-operative compared to pre-operative samples. A mixed effect model was used to test differential gene expression by repeated-measures ANOVA. RESULTS WBGP of 33,804 genes at five timepoints in six patients showed 302 significantly differentially expressed genes between samples from the day prior to surgery and the day after surgery. Pathway gene enrichment analysis showed a downregulation of immunologically relevant pathways. There was a significant downregulation of genes involved in T-cell receptor signaling, antigen presentation and NK-cell activity after surgery. Furthermore, there was an upregulation of cytokines related to metastatic ability, growth and angiogenesis. CONCLUSION Whole blood gene expression profiling revealed dysregulation of genes involved in immune surveillance, inflammation and carcinogenesis, after laparoscopic colon cancer surgery.
Collapse
Affiliation(s)
- Sara Kehlet Watt
- Zealand University Hospital, Department of Surgery, Sygehusvej 10, 4000 Roskilde, Denmark.
| | - Hans Carl Hasselbalch
- Zealand University Hospital, Department of Hematology, Sygehusvej 10, 4000 Roskilde, Denmark
| | - Vibe Skov
- Zealand University Hospital, Department of Hematology, Sygehusvej 10, 4000 Roskilde, Denmark
| | - Lasse Kjær
- Zealand University Hospital, Department of Hematology, Sygehusvej 10, 4000 Roskilde, Denmark
| | - Mads Thomassen
- Odense University Hospital, Department of Clinical Genetics, Denmark
| | - Torben A Kruse
- Odense University Hospital, Department of Clinical Genetics, Denmark
| | - Mark Burton
- Odense University Hospital, Department of Clinical Genetics, Denmark
| | - Ismail Gögenur
- Zealand University Hospital, Department of Surgery, Sygehusvej 10, 4000 Roskilde, Denmark; Institute for Clinical Medicine, Copenhagen University and Danish Colorectal Cancer Group, Copenhagen, Denmark
| |
Collapse
|
12
|
Differential expression of major histocompatibility complex class I in subtypes of breast cancer is associated with estrogen receptor and interferon signaling. Oncotarget 2017; 7:30119-32. [PMID: 27121061 PMCID: PMC5058668 DOI: 10.18632/oncotarget.8798] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 04/03/2016] [Indexed: 01/09/2023] Open
Abstract
Tumor-infiltrating lymphocytes (TILs) in triple-negative breast cancer (TNBC) have a strong prognostic and predictive significance. However, the mechanism of TIL influx in TNBC is unclear. Expression of major histocompatibility complex class I (MHC I) on the tumor cell is essential for the effective killing of tumor by cytotoxic TILs. In our current study, human leukocyte antigen (HLA) expression was inversely correlated with estrogen receptor (ER) expression in normal and cancerous breast tissue and positively correlated with TILs in breast cancer. The ER score was inversely correlated with TILs in breast cancer. HLA-A and CD8B gene expression was negatively correlated with ESR1 and positively correlated with interferon-associated gene expression in The Cancer Genome Atlas (TCGA) data. Negative correlation between ESR1 and HLA and positive correlation between interferon-associated and HLA gene expression were also confirmed in Cancer Cell Line Encyclopedia (CCLE) data. Taken together, our data suggest that a lower expression of HLA in luminal-type tumors might be associated with low level of TILs in those tumors. Further investigation of the mechanism of higher HLA expression and TIL influx in TNBC may help to boost the host immune response.
Collapse
|
13
|
Hegde PS, Wallin JJ, Mancao C. Predictive markers of anti-VEGF and emerging role of angiogenesis inhibitors as immunotherapeutics. Semin Cancer Biol 2017; 52:117-124. [PMID: 29229461 DOI: 10.1016/j.semcancer.2017.12.002] [Citation(s) in RCA: 306] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 12/02/2017] [Accepted: 12/07/2017] [Indexed: 12/11/2022]
Abstract
The critical role of angiogenesis in promoting tumor growth and metastasis has been well established scientifically, and consequently blocking this pathway as a therapeutic strategy has demonstrated great clinical success for the treatment of cancer. The holy grail however, has been the identification of patients who derive significant survival benefit from this class of agents. Here we attempt to delineate the diverse mechanisms related to anti-VEGF including its role as an anti-vascular, anti-angiogenic or an anti-permeability factor and review the most promising predictive biomarkers interrogated in large clinical trials, that identify patients who may derive significant survival advantage with VEGF inhibition. Lastly, we describe the function of VEGF as an immunomodulator and illustrate the evidence for anti-VEGF in reprogramming the tumor milieu from an immunosuppressive to an immune permissive microenvironment in human cancers, thus elucidating the role of anti-VEGF as an optimal combination partner for immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Priti S Hegde
- Genentech, 1 DNA Way, South San Francisco, CA 94080, USA.
| | | | | |
Collapse
|
14
|
Kim A, Lee SJ, Kim YK, Park WY, Park DY, Kim JY, Lee CH, Gong G, Huh GY, Choi KU. Programmed death-ligand 1 (PD-L1) expression in tumour cell and tumour infiltrating lymphocytes of HER2-positive breast cancer and its prognostic value. Sci Rep 2017; 7:11671. [PMID: 28916815 PMCID: PMC5601941 DOI: 10.1038/s41598-017-11905-7] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 08/31/2017] [Indexed: 12/31/2022] Open
Abstract
Immunotherapy targeting PD-1/PD-L1 axis showed benefits in cancer. Prognostic significance of tumour infiltrating lymphocytes (TILs) has been determined. We evaluated PD-L1 protein expression in tumour cells and TILs, PD-L1 mRNA level and various histopathologic factors including TILs using 167 formalin-fixed paraffin embedded tissues and 39 fresh tissue of HER2-positive breast cancer. TILs level and PD-L1 expression in tumour cells and TILs were significantly correlated one another. PD-L1 positivity in tumour cells was associated with high histologic grade and high TILs level (p < 0.001, both). High PD-L1 immunoscore in TILs and high total immunoscore (in tumour cells and TILs) of PD-L1 were correlated with high histologic grade (p = 0.001 and p < 0.001, respectively), absence of lymphovascular invasion (p = 0.012 and p = 0.007, respectively), negative hormone receptor expression (p = 0.044 and p = 0.001, respectively) and high TILs level (p < 0.001, both). High PD-L1 mRNA expression was associated with high TILs level (p < 0.001, both). PD-L1 positivity in tumour cells was associated with better disease-free survival in HR−/HER2+ breast cancer (p = 0.039). PD-L1 expression in tumour cells and TILs are significantly associated with TILs level in HER2-positive breast cancer. PD-L1 expression in tumour cells might be positive prognostic factor in HR−/HER2+ breast cancers.
Collapse
Affiliation(s)
- Ahrong Kim
- Department of Pathology, BioMedical Research Institute, Pusan National University Hospital, Pusan, Korea
| | - So Jeong Lee
- Department of Pathology, BioMedical Research Institute, Pusan National University Hospital, Pusan, Korea
| | - Young Keum Kim
- Department of Pathology, BioMedical Research Institute, Pusan National University Hospital, Pusan, Korea
| | - Won Young Park
- Department of Pathology, BioMedical Research Institute, Pusan National University Hospital, Pusan, Korea
| | - Do Youn Park
- Department of Pathology, BioMedical Research Institute, Pusan National University Hospital, Pusan, Korea
| | - Jee Yeon Kim
- Department of Pathology, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - Chang Hun Lee
- Department of Pathology, BioMedical Research Institute, Pusan National University Hospital, Pusan, Korea
| | - Gyungyub Gong
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Gi Yeong Huh
- Department of Pathology, BioMedical Research Institute, Pusan National University Hospital, Pusan, Korea
| | - Kyung Un Choi
- Department of Pathology, BioMedical Research Institute, Pusan National University Hospital, Pusan, Korea.
| |
Collapse
|
15
|
Reuben A, Chung JW, Lapointe R, Santos MM. The hemochromatosis protein HFE 20 years later: An emerging role in antigen presentation and in the immune system. IMMUNITY INFLAMMATION AND DISEASE 2017; 5:218-232. [PMID: 28474781 PMCID: PMC5569368 DOI: 10.1002/iid3.158] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 01/30/2017] [Accepted: 02/10/2017] [Indexed: 12/13/2022]
Abstract
Introduction Since its discovery, the hemochromatosis protein HFE has been primarily defined by its role in iron metabolism and homeostasis, and its involvement in the genetic disease termed hereditary hemochromatosis (HH). While HH patients are typically afflicted by dysregulated iron levels, many are also affected by several immune defects and increased incidence of autoimmune diseases that have thereby implicated HFE in the immune response. Growing evidence has supported an immunological role for HFE with recent studies describing HFE specifically as it relates to MHC I antigen presentation. Methods/Results Here, we present a comprehensive overview of the relationship between iron metabolism, HFE, and the immune system to better understand the origin and cause of immune defects in HH patients. We further describe the role of HFE in MHC I antigen presentation and its potential to impair autoimmune responses in homeostatic conditions, a mechanism which may be exploited by tumors to evade immune surveillance. Conclusion Overall, this increased understanding of the role of HFE in the immune response sets the stage for better treatment and management of HH and other iron‐related diseases, as well as of the immune defects related to this condition.
Collapse
Affiliation(s)
- Alexandre Reuben
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada.,Département de Médicine, Université de Montréal, Montréal, Québec, Canada.,Institut du Cancer de Montréal, Montréal, Québec, Canada
| | - Jacqueline W Chung
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Réjean Lapointe
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada.,Département de Médicine, Université de Montréal, Montréal, Québec, Canada.,Institut du Cancer de Montréal, Montréal, Québec, Canada
| | - Manuela M Santos
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada.,Département de Médicine, Université de Montréal, Montréal, Québec, Canada.,Institut du Cancer de Montréal, Montréal, Québec, Canada
| |
Collapse
|
16
|
Ping Y, Liu C, Zhang Y. T-cell receptor-engineered T cells for cancer treatment: current status and future directions. Protein Cell 2017; 9:254-266. [PMID: 28108950 PMCID: PMC5829268 DOI: 10.1007/s13238-016-0367-1] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 12/16/2016] [Indexed: 12/20/2022] Open
Abstract
T-cell receptor (TCR)-engineered T cells are a novel option for adoptive cell therapy used for the treatment of several advanced forms of cancer. Work using TCR-engineered T cells began more than two decades ago, with numerous preclinical studies showing that such cells could mediate tumor lysis and eradication. The success of these trials provided the foundation for clinical trials, including recent clinical successes using TCR-engineered T cells to target New York esophageal squamous cell carcinoma (NY-ESO-1). These successes demonstrate the potential of this approach to treat cancer. In this review, we provide a perspective on the current and future applications of TCR-engineered T cells for the treatment of cancer. Our summary focuses on TCR activation and both pre-clinical and clinical applications of TCR-engineered T cells. We also discuss how to enhance the function of TCR-engineered T cells and prolong their longevity in the tumor microenvironment.
Collapse
Affiliation(s)
- Yu Ping
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Chaojun Liu
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China. .,Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China. .,Engineering Key Laboratory for Cell Therapy of Henan Province, Zhengzhou, 450052, China.
| |
Collapse
|
17
|
Gehrke M, Junge V, Jaśkowski JM, Polak MP, Urbaniak K, Żuraw A. Bovine Neonatal Pancytopenia - correlation between clinical symptoms, hematological parameters and lifespan. Tierarztl Prax Ausg G Grosstiere Nutztiere 2016; 44:355-359. [PMID: 27805246 DOI: 10.15653/tpg-160098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 06/17/2016] [Indexed: 11/22/2022]
Abstract
OBJECTIVE To compare the hematological parameters and clinical symptoms between Bovine Neonatal Pancytopenia (BNP) diseased calves dying before and after 14 days of life. MATERIAL AND METHODS Clinical observations included 47 calves from dams which underwent a 3-year vaccination program with the inactivated PregSure® BVD vaccine. In 25 of these 47 BNP affected calves blood examinations were performed and in 22 dead calves diagnosis was mainly based on post-mortem findings. RESULTS Cutaneous bleeding was the predominant clinical manifestation in 32 from 47 calves (68.1%). Seven from 47 calves (14.9%) developed cutaneous bleeding as the only symptom and 17 from 47 calves (36.2%) demonstrated these alterations in combination with hemorrhagic lesions of the oral mucosa. In 66.0% (31/47) of calves petechiae of the oral mucosa were seen and petechiation without any other BNP related symptoms occurred in eight from 47 calves (17.0%). The hematological analysis revealed thrombocytopenia in all 25 cases (n = 23: PLT < 60 x 109/l, n = 2: PLT 139-164 x 109/l). Nineteen from 25 calves (76.0%) developed thrombocytopenia and leukocytopenia (WBC < 3.5 x 109/l). In nine of them a decrease of erythrocyte count (RBC < 4.5 x 109/l), hemoglobin concentration (Hb < 8 g/dl) and packed cell volume (PCV < 24%) was measured. Three BNP affected calves without clinical symptoms were identified by hematological examination. The average life time of BNP affected calves was 14.7 ± 6.2 days. Clinical findings, especially multifocal cutaneous hemorrhages were more frequently recognized in calves living longer than 14 days. CONCLUSION AND CLINICAL RELEVANCE At the time of falling ill with BNP, older calves displayed more numerous symptoms, especially bleeding in the skin. Thrombocytopenia and erythropenia occur as well as a decreased hemoglobin concentration and a low PCV. The time between outbreak of symptoms and death of calves which fell ill later, did not differ from the survival time of BNP calves, which displayed symptoms at a younger age. A decrease of thrombocytes was the cardinal laboratory finding.
Collapse
Affiliation(s)
- Marek Gehrke
- Marek Gehrke, Institute of Veterinary Sciences, Faculty of Veterinary Medicine and Animal Sciences, Poznan University of Life Sciences, Wołyńska 35, 60-637 Poznan, Poland,
| | | | | | | | | | | |
Collapse
|
18
|
Brea EJ, Oh CY, Manchado E, Budhu S, Gejman RS, Mo G, Mondello P, Han JE, Jarvis CA, Ulmert D, Xiang Q, Chang AY, Garippa RJ, Merghoub T, Wolchok JD, Rosen N, Lowe SW, Scheinberg DA. Kinase Regulation of Human MHC Class I Molecule Expression on Cancer Cells. Cancer Immunol Res 2016; 4:936-947. [PMID: 27680026 DOI: 10.1158/2326-6066.cir-16-0177] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 09/01/2016] [Indexed: 12/13/2022]
Abstract
The major histocompatibility complex I (MHC-1) presents antigenic peptides to tumor-specific CD8+ T cells. The regulation of MHC-I by kinases is largely unstudied, even though many patients with cancer are receiving therapeutic kinase inhibitors. Regulators of cell-surface HLA amounts were discovered using a pooled human kinome shRNA interference-based approach. Hits scoring highly were subsequently validated by additional RNAi and pharmacologic inhibitors. MAP2K1 (MEK), EGFR, and RET were validated as negative regulators of MHC-I expression and antigen presentation machinery in multiple cancer types, acting through an ERK output-dependent mechanism; the pathways responsible for increased MHC-I upon kinase inhibition were mapped. Activated MAPK signaling in mouse tumors in vivo suppressed components of MHC-I and the antigen presentation machinery. Pharmacologic inhibition of MAPK signaling also led to improved peptide/MHC target recognition and killing by T cells and TCR-mimic antibodies. Druggable kinases may thus serve as immediately applicable targets for modulating immunotherapy for many diseases. Cancer Immunol Res; 4(11); 936-47. ©2016 AACR.
Collapse
Affiliation(s)
- Elliott J Brea
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center New York, New York.,Weill Cornell Medicine, New York, New York
| | - Claire Y Oh
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center New York, New York.,Weill Cornell Medicine, New York, New York
| | - Eusebio Manchado
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center New York, New York
| | - Sadna Budhu
- Immunology Program, Memorial Sloan Kettering Cancer Center New York, New York
| | - Ron S Gejman
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center New York, New York.,Weill Cornell Medicine, New York, New York
| | - George Mo
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center New York, New York
| | - Patrizia Mondello
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center New York, New York
| | - James E Han
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center New York, New York.,Weill Cornell Medicine, New York, New York
| | - Casey A Jarvis
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center New York, New York
| | - David Ulmert
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center New York, New York
| | - Qing Xiang
- RNAi Core Facility, Memorial Sloan Kettering Cancer Center New York, New York
| | - Aaron Y Chang
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center New York, New York.,Weill Cornell Medicine, New York, New York
| | - Ralph J Garippa
- RNAi Core Facility, Memorial Sloan Kettering Cancer Center New York, New York
| | - Taha Merghoub
- Immunology Program, Memorial Sloan Kettering Cancer Center New York, New York
| | - Jedd D Wolchok
- Weill Cornell Medicine, New York, New York.,Immunology Program, Memorial Sloan Kettering Cancer Center New York, New York
| | - Neal Rosen
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center New York, New York.,Weill Cornell Medicine, New York, New York
| | - Scott W Lowe
- Weill Cornell Medicine, New York, New York.,Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center New York, New York.,Howard Hughes Medical Institute, New York, New York
| | - David A Scheinberg
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center New York, New York. .,Weill Cornell Medicine, New York, New York
| |
Collapse
|
19
|
Reches A, Nachmani D, Berhani O, Duev-Cohen A, Shreibman D, Ophir Y, Seliger B, Mandelboim O. HNRNPR Regulates the Expression of Classical and Nonclassical MHC Class I Proteins. THE JOURNAL OF IMMUNOLOGY 2016; 196:4967-76. [PMID: 27194785 DOI: 10.4049/jimmunol.1501550] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 04/20/2016] [Indexed: 11/19/2022]
Abstract
MHC class I molecules, in addition to their role in specific activation of the CTL of adaptive immune system, function also as the main ligands for NK cell inhibitory receptors, which prevent NK cells from killing normal, healthy cells. MHC class I proteins are divided into classical and nonclassical proteins. The former group consists of hundreds of HLA-A, B, and C alleles, which are universally expressed, whereas several alleles of the latter group, such as HLA-G, manifest a restricted expression pattern. Despite the important role played by these molecules in innate and adaptive immune responses, their complex expression regulation is not fully known. In our study, we investigated the regulation processes controlling the expression of MHC class I molecules, with a particular focus on their 3' untranslated regions. We identified heterogeneous nuclear ribonucleoprotein R (HNRNPR) as an important positive regulator of classical and nonclassical MHC class I molecules. HNRNPR is a RNA-binding protein belonging to the heterogeneous nuclear ribonucleoprotein family, which has a known role in processing of precursor mRNA. We demonstrated that HNRNPR binds MHC class I mRNAs in their 3' untranslated regions and enhances their stability and consequently their expression. Furthermore, regulation by HNRNPR modulates the cytotoxic activity of NK cells. In conclusion, we show that HNRNPR acts as a general positive regulator of MHC class I expression.
Collapse
Affiliation(s)
- Adi Reches
- Lautenberg Center for General and Tumor Immunology, Institute of Medical Research Israel-Canada, Faculty of Medicine, Hebrew University Hadassah Medical School, 9112001 Jerusalem, Israel; and
| | - Daphna Nachmani
- Lautenberg Center for General and Tumor Immunology, Institute of Medical Research Israel-Canada, Faculty of Medicine, Hebrew University Hadassah Medical School, 9112001 Jerusalem, Israel; and
| | - Orit Berhani
- Lautenberg Center for General and Tumor Immunology, Institute of Medical Research Israel-Canada, Faculty of Medicine, Hebrew University Hadassah Medical School, 9112001 Jerusalem, Israel; and
| | - Alexandra Duev-Cohen
- Lautenberg Center for General and Tumor Immunology, Institute of Medical Research Israel-Canada, Faculty of Medicine, Hebrew University Hadassah Medical School, 9112001 Jerusalem, Israel; and
| | - Dorin Shreibman
- Lautenberg Center for General and Tumor Immunology, Institute of Medical Research Israel-Canada, Faculty of Medicine, Hebrew University Hadassah Medical School, 9112001 Jerusalem, Israel; and
| | - Yael Ophir
- Lautenberg Center for General and Tumor Immunology, Institute of Medical Research Israel-Canada, Faculty of Medicine, Hebrew University Hadassah Medical School, 9112001 Jerusalem, Israel; and
| | - Barbara Seliger
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany
| | - Ofer Mandelboim
- Lautenberg Center for General and Tumor Immunology, Institute of Medical Research Israel-Canada, Faculty of Medicine, Hebrew University Hadassah Medical School, 9112001 Jerusalem, Israel; and
| |
Collapse
|
20
|
Erdoğan Ö, Xie L, Wang L, Wu B, Kong Q, Wan Y, Chen X. Proteomic dissection of LPS-inducible, PHF8-dependent secretome reveals novel roles of PHF8 in TLR4-induced acute inflammation and T cell proliferation. Sci Rep 2016; 6:24833. [PMID: 27112199 PMCID: PMC4845005 DOI: 10.1038/srep24833] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 04/01/2016] [Indexed: 11/09/2022] Open
Abstract
Endotoxin (LPS)-induced changes in histone lysine methylation contribute to the gene-specific transcription for control of inflammation. Still unidentified are the chromatin regulators that drive the transition from a transcriptional-repressive to a transcriptional-active chromatin state of pro-inflammatory genes. Here, using combined approaches to analyze LPS-induced changes in both gene-specific transcription and protein secretion to the extracellular compartment, we characterize novel functions of the lysine demethylase PHF8 as a pro-inflammatory, gene-specific chromatin regulator. First, in the LPS-induced, acute-inflamed macrophages, PHF8 knockdown led to both a reduction of pro-inflammatory factors and an increase in a transcriptional-repressive code (H3K9me2) written by the methyltransferase G9a. Through unbiased quantitative secretome screening we discovered that LPS induces the secretion of a cluster of PHF8-dependent, 'tolerizable' proteins that are related to diverse extracellular pathways/processes including those for the activation of adaptive immunity. Specifically, we determined that PHF8 promotes T-cell activation and proliferation, thus providing the first link between the epigenetic regulation of inflammation and adaptive immunity. Further, we found that, in the acute-inflamed macrophages, the acute-active PHF8 opposes the H3K9me1/2-writing activity of G9a to activate specific protein secretions that are suppressed by G9a in the endotoxin-tolerant cells, revealing the inflammatory-phenotypic chromatin drivers that regulate the gene-specific chromatin plasticity.
Collapse
Affiliation(s)
- Özgün Erdoğan
- Department of Biochemistry and Biophysics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, US
| | - Ling Xie
- Department of Biochemistry and Biophysics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, US
| | - Li Wang
- Department of Biochemistry and Biophysics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, US
- Department of Chemistry, Fudan University, Shanghai, China
| | - Bing Wu
- Departement of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, US
| | - Qing Kong
- Department of Biochemistry and Biophysics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, US
| | - Yisong Wan
- Departement of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, US
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, US
| | - Xian Chen
- Department of Biochemistry and Biophysics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, US
- Department of Chemistry, Fudan University, Shanghai, China
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, US
| |
Collapse
|
21
|
Cortical beta amyloid protein triggers an immune response, but no synaptic changes in the APPswe/PS1dE9 Alzheimer's disease mouse model. Neurobiol Aging 2012; 34:1328-42. [PMID: 23245294 DOI: 10.1016/j.neurobiolaging.2012.11.008] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Revised: 11/14/2012] [Accepted: 11/16/2012] [Indexed: 01/06/2023]
Abstract
Using microarray technology we studied the genome-wide gene expression profiles in the frontal cortex of APPswe/PS1dE9 mice and age and sex-matched littermates at the age of 2, 3, 6, 9, 12, and 15-18 months to investigate transcriptional changes that are associated with beta amyloid protein (Aβ) plaque formation and buildup. We observed the occurrence of an immune response with glial activation, but no changes in genes involved in synaptic transmission or plasticity. Comparison of the mouse gene expression data set with a human data set representing the course of Alzheimer's disease revealed a strikingly limited overlap between gene expression in the APPswe/PS1dE9 and human Alzheimer's disease prefrontal cortex. Only plexin domain containing 2, complement component 4b, and solute carrier family 14 (urea transporter) member 1 were significantly upregulated in the mouse and human brain which might suggest a function in Aβ pathology for these 3 genes. In both data sets we detected clusters of upregulated genes involved in immune-related processes. We conclude that the APPswe/PS1dE9 mouse can be a good model to study the immune response associated with cortical Aβ plaques.
Collapse
|
22
|
Assad A, Amann B, Friedrich A, Deeg CA. Immunophenotyping and characterization of BNP colostra revealed pathogenic alloantibodies of IgG1 subclass with specifity to platelets, granulocytes and monocytes of all maturation stages. Vet Immunol Immunopathol 2012; 147:25-34. [PMID: 22554492 DOI: 10.1016/j.vetimm.2012.04.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Accepted: 04/10/2012] [Indexed: 12/18/2022]
Abstract
Bovine neonatal pancytopenia (BNP) is mainly characterized by multiple haemorrhages, thrombocytopenia and leukocytopenia as a result of bone marrow depletion. BNP can be induced in healthy calves through application of colostrum from BNP donors, proofing that BNP is mediated to maternal alloantibodies. Alloantibody binding to bovine blood cells is present in sera and colostra of BNP donors and is probably initialized by vaccination with a certain BVD vaccine. To understand etiology and pathomechanisms of BNP, we closely characterized disease inducing antibodies regarding immunoglobulin subclass and binding specificities to peripheral blood derived leukocytes and platelets. By exact phenotyping the targeted blood cell subsets, including platelets for the first time, we investigated that BNP alloantibodies are exclusively of IgG1 subclass. Interestingly, IgG1 of BNP colostra bound to 70% leukocytes and 100% platelets irrespective of different bovine breeds and cellular maturity of all specimens tested. Furthermore, staining pattern on platelets as well as leukocyte subsets by BNP-IgG1 alloantibody exposed 100% reactivity to platelets, granulocytes and monocytes. Interestingly, the main part of T-helper cells was not bound by colostral alloantibodies. Our results point to a crucial role of IgG1 antibodies in BNP and to a target antigen that is expressed by all cells of myeloid lineage, but only partially by the lymphoid lineage.
Collapse
Affiliation(s)
- Aryan Assad
- Clinic for Ruminants, Centre of Clinical Veterinary Medicine, Ludwig-Maximilians University, Sonnenstraße 16, D-85764 Oberschleißheim, Germany.
| | | | | | | |
Collapse
|
23
|
Medulloblasoma: challenges for effective immunotherapy. J Neurooncol 2011; 108:1-10. [PMID: 22173741 DOI: 10.1007/s11060-011-0776-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2011] [Accepted: 11/29/2011] [Indexed: 01/21/2023]
Abstract
For medulloblastoma patients, the current therapeutic paradigm of surgery followed by radiation and chemotherapy can lead to long-term remission. However, the sequelae of treatment can be very debilitating, particularly in young children. Immunotherapy is an attractive treatment approach to optimize the targeting of tumor cells while sparing the vulnerable surrounding brain that is still developing in children. Understanding the relationship between medulloblastoma and the immune system is critical to develop effective immunologic-based treatment strategies for these patients. This review focuses on current knowledge of tumor immunology and the factors that contribute to the lack of immune system recognition of these tumors. The specificity of tumor antigens present in medulloblastoma is also discussed along with a summary of early clinical immunotherapy results.
Collapse
|
24
|
Deutskens F, Lamp B, Riedel CM, Wentz E, Lochnit G, Doll K, Thiel HJ, Rümenapf T. Vaccine-induced antibodies linked to bovine neonatal pancytopenia (BNP) recognize cattle major histocompatibility complex class I (MHC I). Vet Res 2011; 42:97. [PMID: 21878124 PMCID: PMC3180656 DOI: 10.1186/1297-9716-42-97] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Accepted: 08/30/2011] [Indexed: 01/26/2023] Open
Abstract
A mysterious disease affecting calves, named bovine neonatal pancytopenia (BNP), emerged in 2007 in several European countries. Epidemiological studies revealed a connection between BNP and vaccination with an inactivated vaccine against bovine virus diarrhea (BVD). Alloantibodies reacting with blood leukocytes of calves were detected in serum and colostrum of dams, which have given birth to calves affected by BNP. To understand the linkage between vaccination and the development of alloantibodies, we determined the antigens reacting with these alloantibodies. Immunoprecipitation of surface proteins from bovine leukocytes and kidney cells using sera from dams with a confirmed case of BNP in their gestation history reacted with two dominant protein species of 44 and 12 kDa. These proteins were not detected by sera from dams, free of BVDV and not vaccinated against BVD, and from sera of animals vaccinated with a different inactivated BVD vaccine. The 44 kDa protein was identified by mass spectrometry analysis as MHC I, the other as β-2-microglobulin. The presence of major histocompatibility complex class I (MHC I) in the vaccine was confirmed by Western blot using a MHC I specific monoclonal antibody. A model of BNP pathogenesis is proposed.
Collapse
Affiliation(s)
- Fabian Deutskens
- Institute of Virology, Faculty of Veterinary Medicine, Justus-Liebig-University Giessen, Germany
| | - Benjamin Lamp
- Institute of Virology, Faculty of Veterinary Medicine, Justus-Liebig-University Giessen, Germany
| | - Christiane M Riedel
- Institute of Virology, Faculty of Veterinary Medicine, Justus-Liebig-University Giessen, Germany
| | - Eveline Wentz
- Institute of Virology, Faculty of Veterinary Medicine, Justus-Liebig-University Giessen, Germany
| | - Günter Lochnit
- Institute of Biochemistry, Justus-Liebig-University Giessen, Germany
| | - Klaus Doll
- Clinic for Ruminants, Faculty of Veterinary Medicine, Justus-Liebig-University Giessen, Germany
| | - Heinz-Jürgen Thiel
- Institute of Virology, Faculty of Veterinary Medicine, Justus-Liebig-University Giessen, Germany
| | - Till Rümenapf
- Institute of Virology, Faculty of Veterinary Medicine, Justus-Liebig-University Giessen, Germany
| |
Collapse
|
25
|
Lask A, Goichberg P, Cohen A, Goren-Arbel R, Milstein O, Aviner S, Feine I, Ophir E, Reich-Zeliger S, Hagin D, Klein T, Nagler A, Berrebi A, Reisner Y. TCR-independent killing of B cell malignancies by anti-third-party CTLs: the critical role of MHC-CD8 engagement. THE JOURNAL OF IMMUNOLOGY 2011; 187:2006-14. [PMID: 21753148 DOI: 10.4049/jimmunol.1100095] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We previously demonstrated that anti-third-party CTLs (stimulated under IL-2 deprivation against cells with an MHC class I [MHC-I] background different from that of the host and the donor) are depleted of graft-versus-host reactivity and can eradicate B cell chronic lymphocytic leukemia cells in vitro or in an HU/SCID mouse model. We demonstrated in the current study that human allogeneic or autologous anti-third-party CTLs can also efficiently eradicate primary non-Hodgkin B cell lymphoma by inducing slow apoptosis of the pathological cells. Using MHC-I mutant cell line as target cells, which are unrecognizable by the CTL TCR, we demonstrated directly that this killing is TCR independent. Strikingly, this unique TCR-independent killing is induced through lymphoma MHC-I engagement. We further showed that this killing mechanism begins with durable conjugate formation between the CTLs and the tumor cells, through rapid binding of tumor ICAM-1 to the CTL LFA-1 molecule. This conjugation is followed by a slower second step of MHC-I-dependent apoptosis, requiring the binding of the MHC-I α2/3 C region on tumor cells to the CTL CD8 molecule for killing to ensue. By comparing CTL-mediated killing of Daudi lymphoma cells (lacking surface MHC-I expression) to Daudi cells with reconstituted surface MHC-I, we demonstrated directly for the first time to our knowledge, in vitro and in vivo, a novel role for MHC-I in the induction of lymphoma cell apoptosis by CTLs. Additionally, by using different knockout and transgenic strains, we further showed that mouse anti-third-party CTLs also kill lymphoma cells using similar unique TCR-independence mechanism as human CTLs, while sparing normal naive B cells.
Collapse
Affiliation(s)
- Assaf Lask
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Sigalov AB. The SCHOOL of nature: IV. Learning from viruses. SELF/NONSELF 2010; 1:282-298. [PMID: 21487503 PMCID: PMC3062383 DOI: 10.4161/self.1.4.13279] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2010] [Revised: 08/04/2010] [Accepted: 08/05/2010] [Indexed: 02/05/2023]
Abstract
During the co-evolution of viruses and their hosts, the latter have equipped themselves with an elaborate immune system to defend themselves from the invading viruses. In order to establish a successful infection, replicate and persist in the host, viruses have evolved numerous strategies to counter and evade host antiviral immune responses as well as exploit them for productive viral replication. These strategies include those that modulate signaling mediated by cell surface receptors. Despite tremendous advancement in recent years, the exact molecular mechanisms underlying these critical points in viral pathogenesis remain unknown. In this work, based on a novel platform of receptor signaling, the Signaling Chain HOmoOLigomerization (SCHOOL) platform, I suggest specific mechanisms used by different viruses such as human immunodeficiency virus (HIV), cytomegalovirus (CMV), severe acute respiratory syndrome coronavirus, human herpesvirus 6 and others, to modulate receptor signaling. I also use the example of HIV and CMV to illustrate how two unrelated enveloped viruses use a similar SCHOOL mechanism to modulate the host immune response mediated by two functionally different receptors: T cell antigen receptor and natural killer cell receptor, NKp30. This suggests that it is very likely that similar general mechanisms can be or are used by other viral and possibly non-viral pathogens. Learning from viruses how to target cell surface receptors not only helps us understand viral strategies to escape from the host immune surveillance, but also provides novel avenues in rational drug design and the development of new therapies for immune disorders.
Collapse
|
27
|
Bukur J, Herrmann F, Handke D, Recktenwald C, Seliger B. Identification of E2F1 as an important transcription factor for the regulation of tapasin expression. J Biol Chem 2010; 285:30419-26. [PMID: 20663889 PMCID: PMC2945534 DOI: 10.1074/jbc.m109.094284] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Revised: 06/23/2010] [Indexed: 11/06/2022] Open
Abstract
HER-2/neu overexpression in tumor cells caused abnormalities of MHC class I surface expression due to impaired expression of components of the antigen-processing machinery (APM) including the low molecular weight proteins, the transporter associated with antigen processing (TAP), and the chaperone tapasin, whereas the expression of MHC class I heavy chain as well as β(2)-microglobulin was only marginally affected. This oncogene-mediated deficient APM component expression could be reverted by interferon-γ treatment, suggesting a deregulation rather than structural alterations as underlying molecular mechanisms. To determine the level of regulation, the transcriptional activity of APM components was analyzed in HER-2/neu(-) and HER-2/neu(+) cells. All major APM components were transcriptionally down-regulated in HER-2/neu(+) when compared with HER-2/neu(-) cells, which was accompanied by a reduced binding of RNA polymerase II to the APM promoters. Site-directed mutagenesis of the p300- and E2F-binding sites in the APM promoters did not reconstitute the oncogene-mediated decreased transcription rate with the exception of tapasin, which was restored in HER-2/neu(+) cells to levels of wild type tapasin promoter activity in HER-2/neu(-) fibroblasts. The E2F-directed control of tapasin expression was further confirmed by chromatin immunoprecipitation analyses showing that E2F1 and p300 bind to the tapasin and APM promoters in both cell lines. Moreover, siRNA-mediated silencing of E2F1 was associated with an increased tapasin expression, whereas transient overexpression of E2F1 launch a reduced tapasin transcription, suggesting that E2F1 is an essential transcription factor for tapasin.
Collapse
Affiliation(s)
- Juergen Bukur
- From the Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany
| | - Felix Herrmann
- From the Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany
| | - Diana Handke
- From the Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany
| | - Christian Recktenwald
- From the Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany
| | - Barbara Seliger
- From the Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany
| |
Collapse
|
28
|
Characterization of signaling function and expression of HLA class I molecules in medulloblastoma. J Neurooncol 2010; 103:197-206. [PMID: 20811766 DOI: 10.1007/s11060-010-0378-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2010] [Accepted: 08/19/2010] [Indexed: 12/15/2022]
Abstract
Although known for the important function in the immune system, MHC class I molecules are increasingly ascribed an alternative role in modifying signal transduction. In medulloblastoma, HLA class I molecules are associated with poor prognosis, and can induce ERK1/2 activation upon engagement with ligands that bind to incompletely assembled complexes (so called open conformers). We here demonstrate that ERK1/2 activation in medulloblastoma can occur in the absence of endogenously synthesized β2m, formally excluding involvement of closed HLA class conformation. In addition, several experimental observations suggest that heterogeneity of HLA class I expression may be a reflection of the status of original cells before transformation, rather than a consequence of immune-based selection of HLA-loss mutants. These results contribute to our understanding of an immune system-independent role of HLA class I in the pathology of medulloblastoma, and cancer in general.
Collapse
|
29
|
Li H, Zhan T, Li C, Liu M, Wang QK. Repression of MHC class I transcription by HPV16E7 through interaction with a putative RXRbeta motif and NF-kappaB cytoplasmic sequestration. Biochem Biophys Res Commun 2009; 388:383-8. [PMID: 19665994 DOI: 10.1016/j.bbrc.2009.08.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2009] [Accepted: 08/04/2009] [Indexed: 11/17/2022]
Abstract
Down-regulation of transcription of the MHC class I genes in HPV16 tumorigenic cells is partly due to HPV16E7 associated with the MHC class I promoter and repressed chromatin activation. In this study, we further demonstrated that HPV16E7 is physically associated with a putative RXRbeta binding motif (GGTCA) of the proximal promoter of the MHC class I genes by using reporter transcriptional assays and chromatin immunoprecipitation assays. Our data also provide evidence that HPV16E7 inhibits TNF-alpha-induced up-regulation of MHC class I transcription by impaired nuclear translocation of NF-kappaB. More importantly, CaSki tumor cells treated with TSA and transfected with the constitutively active mutant form of IKK-alpha (which can activate NF-kappaB directly) showed a maximal level of up-regulation of MHC-I expression. Taken together, our results suggest that HPV16E7 may employ two independent mechanisms to ensure that either the constitutive or inducible transcription of MHC class I genes is down-regulated.
Collapse
Affiliation(s)
- Hui Li
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, PR China
| | | | | | | | | |
Collapse
|
30
|
Smith C, Santi M, Rajan B, Rushing EJ, Choi MR, Rood BR, Cornelison R, MacDonald TJ, Vukmanovic S. A novel role of HLA class I in the pathology of medulloblastoma. J Transl Med 2009; 7:59. [PMID: 19594892 PMCID: PMC2714836 DOI: 10.1186/1479-5876-7-59] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2009] [Accepted: 07/12/2009] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND MHC class I expression by cancer cells enables specific antigen recognition by the immune system and protection of the host. However, in some cancer types MHC class I expression is associated with an unfavorable outcome. We explored the basis of MHC class I association with unfavorable prognostic marker expression in the case of medulloblastoma. METHODS We investigated expression of four essential components of MHC class I (heavy chain, beta2m, TAP1 and TAP2) in 10 medulloblastoma mRNA samples, a tissue microarray containing 139 medulloblastoma tissues and 3 medulloblastoma cell lines. Further, in medulloblastoma cell lines we evaluated the effects of HLA class I engagement on activation of ERK1/2 and migration in vitro. RESULTS The majority of specimens displayed undetectable or low levels of the heavy chains. Medulloblastomas expressing high levels of HLA class I displayed significantly higher levels of anaplasia and c-myc expression, markers of poor prognosis. Binding of beta2m or a specific antibody to open forms of HLA class I promoted phosphorylation of ERK1/2 in medulloblastoma cell line with high levels, but not in the cell line with low levels of HLA heavy chain. This treatment also promoted ERK1/2 activation dependent migration of medulloblastoma cells. CONCLUSION MHC class I expression in medulloblastoma is associated with anaplasia and c-myc expression, markers of poor prognosis. Peptide- and/or beta2m-free forms of MHC class I may contribute to a more malignant phenotype of medulloblastoma by modulating activation of signaling molecules such as ERK1/2 that stimulates cell mobility.
Collapse
Affiliation(s)
- Courtney Smith
- Center for Cancer and Immunology Research, Children's Research Institute, Children's National Medical Center, Washington, DC, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Kim WM, Sigalov AB. Viral pathogenesis, modulation of immune receptor signaling and treatment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2008; 640:325-49. [PMID: 19065800 PMCID: PMC7122915 DOI: 10.1007/978-0-387-09789-3_22] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
During the co-evolution of viruses and their hosts, the latter have equipped themselves with an elaborate immune system to defend themselves from the invading viruses. In order to establish a successful infection, replicate and persist in the host, viruses have evolved numerous strategies to counter and evade host antiviral immune responses as well as exploit them for productive viral replication. These strategies include those that target immune receptor transmembrane signaling. Uncovering the exact molecular mechanisms underlying these critical points in viral pathogenesis will not only help us understand strategies used by viruses to escape from the host immune surveillance but also reveal new therapeutic targets for antiviral as well as immunomodulatory therapy. In this chapter, based on our current understanding of transmembrane signal transduction mediated by multichain immune recognition receptors (MIRRs) and the results of sequence analysis, we discuss the MIRR-targetingviral strategies of immune evasion and suggest their possible mechanisms that, in turn, reveal new points of antiviral intervention. We also show how two unrelated enveloped viruses, human immunodeficiency virus and human cytomegalovirus, use a similar mechanism to modulate the host immune response mediated by two functionally different MIRRs-T-cell antigen receptor and natural killer cell receptor, NKp30. This suggests that it is very likely that similar general mechanisms can be or are used by other viral and possibly nonviral pathogens.
Collapse
Affiliation(s)
- Walter M Kim
- Department of Pathology, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655, USA
| | | |
Collapse
|
32
|
Abstract
In the present report we describe the laborious identification of the A*02010102L allele found in three healthy individuals of a French family who have shown a reduced A2 antigen expression using serological tests since the 1980s. PCR-SSP typing showed a classical A*0201 allele. Sequencing of exons 2, 3 and 4 confirmed this assignment. Sequencing of the whole gene (promoter, introns and exons 1-8) revealed one single point mutation (T to C) at position -101 in the enhancer B element region compared to the A*02010101 allele. This single mutation appears to be related to the reduced expression of the A2 antigen. This allele segregates with the haplotype Cw*12, B44, DR7, DQ2, which is different to the one described earlier.
Collapse
Affiliation(s)
- P Perrier
- Laboratoire d'histocompatibilité, Vandoeuvre-les-Nancy, France.
| | | | | | | |
Collapse
|
33
|
Marchetti B, Ashrafi GH, Dornan ES, Araibi EH, Ellis SA, Campo MS. The E5 protein of BPV-4 interacts with the heavy chain of MHC class I and irreversibly retains the MHC complex in the Golgi apparatus. Oncogene 2006; 25:2254-63. [PMID: 16288210 DOI: 10.1038/sj.onc.1209245] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BPV-4 E5 inhibits transcription of the bovine MHC class I heavy chain (HC) gene, increases degradation of HC and downregulates surface expression of MHC class I by retaining the complex in the Golgi apparatus (GA). Here we report that transcription inhibition can be alleviated by interferon treatment and the degradation of HC can be reversed by treatment with inhibitors of proteasomes and lysosomes. However, the inhibition of transport of MHC class I to the cell surface is irreversible. We show that E5 is capable of physically interacting with HC. Together with the inhibition of the vacuolar ATPase (due to the interaction between E5 and 16k subunit c), the interaction between E5 and HC is likely to be responsible for retention of MHC class I in the GA. C-terminus deletion mutants of E5 are incapable of either downregulating surface MHC class I or interacting with HC, establishing that the C-terminus domain of E5 is important in the inhibition of MHC class I.
Collapse
Affiliation(s)
- B Marchetti
- Division of Pathological Sciences, Institute of Comparative Medicine, University of Glasgow, Glasgow, UK
| | | | | | | | | | | |
Collapse
|
34
|
Ashrafi GH, Haghshenas MR, Marchetti B, O'Brien PM, Campo MS. E5 protein of human papillomavirus type 16 selectively downregulates surface HLA class I. Int J Cancer 2005; 113:276-83. [PMID: 15386416 DOI: 10.1002/ijc.20558] [Citation(s) in RCA: 152] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Papillomaviruses have evolved mechanisms that result in escape from host immune surveillance. The E5 protein is expressed early in papillomavirus infection in the deep layers of the infected epithelium. It is localized to the Golgi apparatus (GA) and endoplasmic reticulum. The E5 protein of bovine papillomavirus (BPV) impairs the synthesis and stability of major histocompatibility (MHC) class I complexes and prevents their transport to the cell surface due to retention in the GA. Here we show that human papillomavirus type 16 (HPV-16) E5 also causes the retention of MHC (HLA) class I complexes in the GA and impedes their transport to the cell surface, which is rescued by treatment with interferon. Unlike BPV E5, HPV-16 E5 does not affect the synthesis of HLA class I heavy chains or the expression of the transporter associated with antigen processing TAP. These results show that downregulation of surface MHC class I molecules is common to both BPV and HPV E5 proteins. Moreover, we determined that HPV-16 E5 downregulates surface expression of HLA-A and HLA-B, which present viral peptides to MHC class I-restricted cytotoxic T lymphocytes (CTLs), but not the natural killer (NK) cell inhibitory ligands HLA-C and HLA-E. Selective downregulation of cell surface HLA class I molecules may allow the virus to establish infection by avoiding immune clearance of virus-infected cells by both CTLs and NK cells.
Collapse
Affiliation(s)
- G Hossein Ashrafi
- Institute of Comparative Medicine, Department of Pathological Sciences, Glasgow University, Glasgow G61 1QH, Scotland, UK
| | | | | | | | | |
Collapse
|
35
|
Merritt RE, Yamada RE, Crystal RG, Korst RJ. Augmenting major histocompatibility complex class I expression by murine tumors in vivo enhances antitumor immunity induced by an active immunotherapy strategy. J Thorac Cardiovasc Surg 2004; 127:355-64. [PMID: 14762342 DOI: 10.1016/j.jtcvs.2003.09.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
OBJECTIVE Tumors down-regulate major histocompatibility complex class I expression, escaping recognition by the cellular immune response. We hypothesized that augmentation of tumor cell class I expression by interferon-gamma would enhance the cellular antitumor immune response and cure rate of an active immunotherapy strategy. METHODS B16.F10 tumor cells were exposed to interferon-gamma in culture, and class I expression was quantified using flow cytometry. Syngeneic mice bearing established tumors were injected with interferon-gamma (5000 U, intraperitoneal), and class I expression was assessed using immunohistochemistry. Tumor-specific cytotoxic T lymphocytes were induced in mice by an intratumoral injection of AdCD40L (5 x 10(10) particles), an adenovirus gene transfer vector-based immunotherapy strategy previously demonstrated to augment cellular antitumor immunity. A conjugate-formation assay and the enzyme-linked immunospot assay were used to evaluate the binding and activation of cytotoxic T lymphocytes, respectively. Interferon-gamma was administered to tumor-bearing mice concomitantly with intratumoral AdCD40L. End points measured included the frequencies of cytotoxic T lymphocytes using the enzyme-linked immunospot assay, tumor size, and mouse survival. The role of class I expression was further evaluated by monoclonal antibody blockade in both in vitro and in vivo experiments. RESULTS B16.F10 cells exposed to interferon-gamma expressed significantly more class I, both in vitro and in vivo, and were able to bind to and activate cytotoxic T lymphocytes more efficiently than untreated cells. Cytotoxic T-lymphocyte frequencies, tumor regression, and the cure rate induced by AdCD40L were augmented by the addition of a single dose of interferon-gamma in tumor-bearing mice. These in vitro and in vivo effects of interferon-gamma were attenuated by class I monoclonal antibody blockade. CONCLUSIONS Up-regulation of class I expression using interferon-gamma enhances the cellular antitumor immune response and cure rate of AdCD40L, an active immunotherapy strategy. This approach may be useful for human tumors that lack class I expression.
Collapse
MESH Headings
- Animals
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/immunology
- Carcinoma, Lewis Lung/immunology
- Carcinoma, Lewis Lung/metabolism
- Carcinoma, Lewis Lung/therapy
- Cytotoxicity, Immunologic/drug effects
- Cytotoxicity, Immunologic/immunology
- Dose-Response Relationship, Immunologic
- Drug Therapy, Combination
- Enzyme-Linked Immunosorbent Assay
- Female
- Genes, MHC Class I/drug effects
- Genes, MHC Class I/immunology
- H-2 Antigens/drug effects
- H-2 Antigens/immunology
- H-2 Antigens/metabolism
- Immunity, Cellular/drug effects
- Immunity, Cellular/immunology
- Immunotherapy, Active
- Interferon-gamma/administration & dosage
- Lung Neoplasms/immunology
- Lung Neoplasms/metabolism
- Lung Neoplasms/therapy
- Lymphocyte Activation/drug effects
- Lymphocyte Activation/immunology
- Mice
- Mice, Inbred C57BL
- Models, Animal
- Recombinant Proteins
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
- Treatment Outcome
- Tumor Cells, Cultured
- Up-Regulation/immunology
Collapse
Affiliation(s)
- Robert E Merritt
- Department of Cardiothoracic Surgery, Weill Medical College of Cornell University, New York, NY 10021, USA
| | | | | | | |
Collapse
|
36
|
Herrmann F, Trowsdale J, Huber C, Seliger B. Cloning and functional analyses of the mouse tapasin promoter. Immunogenetics 2003; 55:379-88. [PMID: 12942211 DOI: 10.1007/s00251-003-0597-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2003] [Revised: 07/23/2003] [Indexed: 12/17/2022]
Abstract
The expression of tapasin is critical for an optimized MHC class I assembly and stable MHC class I surface expression. Thus, impaired MHC class I antigen expression of tumors can be attributable to tapasin downregulation. In order to understand the molecular mechanisms of deficient tapasin expression, the mouse tapasin promoter region and its 5'-flanking sequences were characterized. The mouse tapasin promoter lacks the TATA box and its transcription is initiated at multiple sites within a 51-nucleotide stretch. Sequence analyses revealed transcription factor binding motifs for NF-kappaB, GATA, E2F, p300, AP1, SP1 and IRF-1/2. Detailed analysis of deletion mutants and elimination of transcription factor binding motifs demonstrated an important role of NF-kappaB at position -468 for its basal activity, whereas E2F at position -229 represses constitutive promoter activity. Furthermore, the IRF-1/2 binding site is required for gamma interferon inducibility of the tapasin promoter in vitro, but also negatively interferes with its constitutive activity. Thus, characterization of the tapasin promoter represents the molecular basis for the understanding of the heterogeneous expression levels of tapasin under physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Felix Herrmann
- 3rd Department of Internal Medicine, Johannes Gutenberg University, 55131 Mainz, Germany
| | | | | | | |
Collapse
|
37
|
Kimchi-Sarfaty C, Arora M, Sandalon Z, Oppenheim A, Gottesman MM. High cloning capacity of in vitro packaged SV40 vectors with no SV40 virus sequences. Hum Gene Ther 2003; 14:167-77. [PMID: 12614568 DOI: 10.1089/104303403321070865] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
In vitro packaging of plasmid DNA using recombinant SV40 capsid proteins is a potentially useful procedure that overcomes some restrictions of the other SV40 systems such as the requirement for SV40 sequences and the limitation in size of DNA that can be packaged. The in vitro packaging system uses the four SV40 proteins (VP1, VP2, VP3, and agno) or VP1 only. The ability to confer drug resistance by three ABC transporter genes (MDR 1, MRP 1, or MXR) was determined using the surrogate fluorescent substrates rhodamine-123 or calcein AM and their specific inhibitors, or by using specific antibodies to the transporters to detect cell surface expression by fluorescence-activated cell sorter analysis (FACS). A green fluorescent protein plasmid (EGFP-C1) was also used to monitor gene transfer. The packaged plasmids ranged in size from 4.2 to 17.6 kb, and only slightly affected particle size as determined by electron microscopy. When 9.5 kb and larger plasmids were packaged using all SV40 proteins, MDR1 expression was decreased compared to VP1 alone. The size of the 15.2 kb DNA after packaging was the same as the original DNA. Packaging with SV40 capsid proteins in vitro does not require any SV40 sequences. Using either the MDR1 or the GFP gene we could demonstrate enhanced expression when cells were pretreated with phorbol 12-myristate 13-acetate (PMA) at low concentrations. Interferon-gamma did not alter expression. We conclude that in vitro packaging is more flexible then previously realized, permitting packaging of at least 17 kb plasmid DNA without the requirement for any viral sequences. This system combines efficient gene delivery of the SV40 viral vector with the presumed safety of nonviral vectors.
Collapse
Affiliation(s)
- Chava Kimchi-Sarfaty
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-4254, USA
| | | | | | | | | |
Collapse
|
38
|
Marchetti B, Ashrafi GH, Tsirimonaki E, O'Brien PM, Campo MS. The bovine papillomavirus oncoprotein E5 retains MHC class I molecules in the Golgi apparatus and prevents their transport to the cell surface. Oncogene 2002; 21:7808-16. [PMID: 12420217 DOI: 10.1038/sj.onc.1205885] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2002] [Revised: 07/16/2002] [Accepted: 07/18/2002] [Indexed: 11/08/2022]
Abstract
During papillomavirus infection, the E5 protein localizes in the cell Golgi apparatus and other endomembrane compartments. Cells transformed by E5 do not express major histocompatibility class I complex (MHC I) on the cell surface, while cells transformed by the other transforming proteins E6 and E7 do. In addition, the total amount of both MHC I protein and mRNA is reduced in E5-transformed cells. Here we show that expression of bovine papillomavirus E5 causes the retention of MHC I in the Golgi apparatus, thus preventing its transport to the cell surface. We ascribe this effect to a failure of acidification of the Golgi apparatus, as similar effects are observed in control cells treated with the ionophore monensin. Treatment of E5-transformed cells with either beta- or gamma-interferon increases the synthesis of MHC I, showing that inhibition of MHC I expression by E5 is not irreversible. However, even after interferon treatment, MHC I, although increased in quantity, is not transported to the cell surface. E5 therefore affects MHC I at several levels, but prevention of MHC I transport to the cell surface appears to be the dominant effect. Lack of surface MHC I would have profound consequences for presentation of viral peptides to the immune system.
Collapse
Affiliation(s)
- Barbara Marchetti
- Institute of Comparative Medicine, Glasgow University Veterinary School, Garscube Estate, Glasgow G61 1QH, UK
| | | | | | | | | |
Collapse
|
39
|
Foster JA, Quan N, Stern EL, Kristensson K, Herkenham M. Induced neuronal expression of class I major histocompatibility complex mRNA in acute and chronic inflammation models. J Neuroimmunol 2002; 131:83-91. [PMID: 12458039 DOI: 10.1016/s0165-5728(02)00258-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Studies have demonstrated neuronal expression of class I major histocompatibility complex (MHC) mRNA and protein in normal and developing brain and in response to injury or viral infection. We report neuronal expression of class I MHC mRNA in hypothalamic paraventricular nucleus (PVN) neurons in rats following systemic infection with Trypanosoma brucei brucei parasites (chronic) and in response to intravenous 1 mg/kg lipopolysaccharide administration (acute peripheral) and in striatal neurons following intrastriatal 5 microg lipopolysaccharide injection (acute central). These results demonstrate that neurons can be a source of immune signaling molecules and establish class I MHC as part of the neuronal component of immune responses.
Collapse
Affiliation(s)
- Jane A Foster
- Section on Functional Neuroanatomy, National Institute of Mental Health, 36 Convent Drive, Building 36, Room 2D15, Bethesda, MD 20892-4070, USA
| | | | | | | | | |
Collapse
|