1
|
Wang D, Dou L, Sui L, Xue Y, Xu S. Natural killer cells in cancer immunotherapy. MedComm (Beijing) 2024; 5:e626. [PMID: 38882209 PMCID: PMC11179524 DOI: 10.1002/mco2.626] [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: 12/21/2023] [Revised: 05/30/2024] [Accepted: 05/30/2024] [Indexed: 06/18/2024] Open
Abstract
Natural killer (NK) cells, as innate lymphocytes, possess cytotoxic capabilities and engage target cells through a repertoire of activating and inhibitory receptors. Particularly, natural killer group 2, member D (NKG2D) receptor on NK cells recognizes stress-induced ligands-the MHC class I chain-related molecules A and B (MICA/B) presented on tumor cells and is key to trigger the cytolytic response of NK cells. However, tumors have developed sophisticated strategies to evade NK cell surveillance, which lead to failure of tumor immunotherapy. In this paper, we summarized these immune escaping strategies, including the downregulation of ligands for activating receptors, upregulation of ligands for inhibitory receptors, secretion of immunosuppressive compounds, and the development of apoptosis resistance. Then, we focus on recent advancements in NK cell immune therapies, which include engaging activating NK cell receptors, upregulating NKG2D ligand MICA/B expression, blocking inhibitory NK cell receptors, adoptive NK cell therapy, chimeric antigen receptor (CAR)-engineered NK cells (CAR-NK), and NKG2D CAR-T cells, especially several vaccines targeting MICA/B. This review will inspire the research in NK cell biology in tumor and provide significant hope for improving cancer treatment outcomes by harnessing the potent cytotoxic activity of NK cells.
Collapse
Affiliation(s)
- DanRu Wang
- National Key Lab of Immunity and Inflammation and Institute of Immunology Naval Medical University Shanghai China
| | - LingYun Dou
- National Key Lab of Immunity and Inflammation and Institute of Immunology Naval Medical University Shanghai China
| | - LiHao Sui
- National Key Lab of Immunity and Inflammation and Institute of Immunology Naval Medical University Shanghai China
| | - Yiquan Xue
- National Key Lab of Immunity and Inflammation and Institute of Immunology Naval Medical University Shanghai China
| | - Sheng Xu
- National Key Lab of Immunity and Inflammation and Institute of Immunology Naval Medical University Shanghai China
- Shanghai Institute of Stem Cell Research and Clinical Translation Dongfang Hospital Shanghai China
| |
Collapse
|
2
|
Liu J, Jiang Y, Chen L, Qian Z, Zhang Y. Associations between HIFs and tumor immune checkpoints: mechanism and therapy. Discov Oncol 2024; 15:2. [PMID: 38165484 PMCID: PMC10761656 DOI: 10.1007/s12672-023-00836-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 11/21/2023] [Indexed: 01/03/2024] Open
Abstract
Hypoxia, which activates a variety of signaling pathways to enhance tumor cell growth and metabolism, is among the primary features of tumor cells. Hypoxia-inducible factors (HIFs) have a substantial impact on a variety of facets of tumor biology, such as epithelial-mesenchymal transition, metabolic reprogramming, angiogenesis, and improved radiation resistance. HIFs induce hypoxia-adaptive responses in tumor cells. Many academics have presented preclinical and clinical research targeting HIFs in tumor therapy, highlighting the potential applicability of targeted HIFs. In recent years, the discovery of numerous pharmacological drugs targeting the regulatory mechanisms of HIFs has garnered substantial attention. Additionally, HIF inhibitors have attained positive results when used in conjunction with traditional oncology radiation and/or chemotherapy, as well as with the very promising addition of tumor immunotherapy. Immune checkpoint inhibitors (CPIs), which are employed in a range of cancer treatments over the past decades, are essential in tumor immunotherapy. Nevertheless, the use of immunotherapy has been severely hampered by tumor resistance and treatment-related toxicity. According to research, HIF inhibitors paired with CPIs may be game changers for multiple malignancies, decreasing malignant cell plasticity and cancer therapy resistance, among other things, and opening up substantial new pathways for immunotherapy drug development. The structure, activation mechanisms, and pharmacological sites of action of the HIF family are briefly reviewed in this work. This review further explores the interactions between HIF inhibitors and other tumor immunotherapy components and covers the potential clinical use of HIF inhibitors in combination with CPIs.
Collapse
Affiliation(s)
- Jiayu Liu
- Department of Oncology, Wuxi Maternal and Child Health Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, 214002, Jiangsu, China
| | - Ying Jiang
- Department of Oncology, Wuxi Maternal and Child Health Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, 214002, Jiangsu, China
| | - Lingyan Chen
- Wuxi Maternal and Child Health Hospital, Nanjing Medical University, Nanjing, 214000, Jiangsu, China
| | - Zhiwen Qian
- Wuxi Maternal and Child Health Hospital, Nanjing Medical University, Nanjing, 214000, Jiangsu, China
| | - Yan Zhang
- Department of Oncology, Wuxi Maternal and Child Health Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, 214002, Jiangsu, China.
- Wuxi Maternal and Child Health Hospital, Nanjing Medical University, Nanjing, 214000, Jiangsu, China.
| |
Collapse
|
3
|
Kałuzińska-Kołat Ż, Kołat D, Kośla K, Płuciennik E, Bednarek AK. Molecular landscapes of glioblastoma cell lines revealed a group of patients that do not benefit from WWOX tumor suppressor expression. Front Neurosci 2023; 17:1260409. [PMID: 37781246 PMCID: PMC10540236 DOI: 10.3389/fnins.2023.1260409] [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: 07/17/2023] [Accepted: 08/28/2023] [Indexed: 10/03/2023] Open
Abstract
Introduction Glioblastoma (GBM) is notorious for its clinical and molecular heterogeneity, contributing to therapeutic failure and a grim prognosis. WWOX is one of the tumor suppressor genes important in nervous tissue or related pathologies, which was scarcely investigated in GBM for reliable associations with prognosis or disease progression despite known alterations. Recently, we observed a phenotypic heterogeneity between GBM cell lines (U87MG, T98G, U251MG, DBTRG-05MG), among which the anti-GBM activity of WWOX was generally corresponding, but colony growth and formation were inconsistent in DBTRG-05MG. This prompted us to investigate the molecular landscapes of these cell lines, intending to translate them into the clinical context. Methods U87MG/T98G/U251MG/DBTRG-05MG were subjected to high-throughput sequencing, and obtained data were explored via weighted gene co-expression network analysis, differential expression analysis, functional annotation, and network building. Following the identification of the most relevant DBTRG-distinguishing driver genes, data from GBM patients were employed for, e.g., differential expression analysis, survival analysis, and principal component analysis. Results Although most driver genes were unique for each cell line, some were inversely regulated in DBTRG-05MG. Alongside driver genes, the differentially-expressed genes were used to build a WWOX-related network depicting protein-protein interactions in U87MG/T98G/U251MG/DBTRG-05MG. This network revealed processes distinctly regulated in DBTRG-05MG, e.g., microglia proliferation or neurofibrillary tangle assembly. POLE4 and HSF2BP were selected as DBTRG-discriminating driver genes based on the gene significance, module membership, and fold-change. Alongside WWOX, POLE4 and HSF2BP expression was used to stratify patients into cell lines-resembling groups that differed in, e.g., prognosis and treatment response. Some differences from a WWOX-related network were certified in patients, revealing genes that clarify clinical outcomes. Presumably, WWOX overexpression in DBTRG-05MG resulted in expression profile change resembling that of patients with inferior prognosis and drug response. Among these patients, WWOX may be inaccessible for its partners and does not manifest its anti-cancer activity, which was proposed in the literature but not regarding glioblastoma or concerning POLE4 and HSF2BP. Conclusion Cell lines data enabled the identification of patients among which, despite high expression of WWOX tumor suppressor, no advantageous outcomes were noted due to the cancer-promoting profile ensured by other genes.
Collapse
Affiliation(s)
| | - Damian Kołat
- Department of Molecular Carcinogenesis, Medical University of Lodz, Lodz, Poland
| | - Katarzyna Kośla
- Department of Molecular Carcinogenesis, Medical University of Lodz, Lodz, Poland
| | | | - Andrzej K. Bednarek
- Department of Molecular Carcinogenesis, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
4
|
Bandopadhyay S, Patranabis S. Mechanisms of HIF-driven immunosuppression in tumour microenvironment. J Egypt Natl Canc Inst 2023; 35:27. [PMID: 37646847 DOI: 10.1186/s43046-023-00186-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/12/2023] [Indexed: 09/01/2023] Open
Abstract
Hypoxia arises due to insufficient oxygen delivery to rapidly proliferating tumour cells that outpace the available blood supply. It is a characteristic feature of most solid tumour microenvironments and plays a critical role in regulating anti-tumour immunity, enhancing tumoral heterogeneity, and promoting therapeutic resistance and poor clinical outcomes. Hypoxia-inducible factors (HIFs) are the major hypoxia-responsive transcription factors that are activated under low oxygenation conditions and have been identified to drive multifunctional roles in tumour immune evasion. The HIF signalling network serves as an attractive target for targeted therapeutic approaches. This review aims to provide a comprehensive overview of the most crucial mechanisms by which HIF controls the expression of immunosuppressive molecules and immune checkpoints, disrupts cancer immunogenicity, and induces immunotherapeutic resistance.
Collapse
Affiliation(s)
| | - Somi Patranabis
- Amity Institute of Biotechnology, Amity University, Kolkata, West Bengal, India.
| |
Collapse
|
5
|
Missiaen R, Lesner NP, Simon MC. HIF: a master regulator of nutrient availability and metabolic cross-talk in the tumor microenvironment. EMBO J 2023; 42:e112067. [PMID: 36808622 PMCID: PMC10015374 DOI: 10.15252/embj.2022112067] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 12/13/2022] [Accepted: 12/16/2022] [Indexed: 02/22/2023] Open
Abstract
A role for hypoxia-inducible factors (HIFs) in hypoxia-dependent regulation of tumor cell metabolism has been thoroughly investigated and covered in reviews. However, there is limited information available regarding HIF-dependent regulation of nutrient fates in tumor and stromal cells. Tumor and stromal cells may generate nutrients necessary for function (metabolic symbiosis) or deplete nutrients resulting in possible competition between tumor cells and immune cells, a result of altered nutrient fates. HIF and nutrients in the tumor microenvironment (TME) affect stromal and immune cell metabolism in addition to intrinsic tumor cell metabolism. HIF-dependent metabolic regulation will inevitably result in the accumulation or depletion of essential metabolites in the TME. In response, various cell types in the TME will respond to these hypoxia-dependent alterations by activating HIF-dependent transcription to alter nutrient import, export, and utilization. In recent years, the concept of metabolic competition has been proposed for critical substrates, including glucose, lactate, glutamine, arginine, and tryptophan. In this review, we discuss how HIF-mediated mechanisms control nutrient sensing and availability in the TME, the competition for nutrients, and the metabolic cross-talk between tumor and stromal cells.
Collapse
Affiliation(s)
- Rindert Missiaen
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Nicholas P Lesner
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - M Celeste Simon
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
6
|
Alves CC, Arns T, Oliveira ML, Moreau P, Antunes DA, Castelli EC, Mendes-Junior CT, Giuliatti S, Donadi EA. Computational and atomistic studies applied to the understanding of the structural and behavioral features of the immune checkpoint HLA-G molecule and gene. Hum Immunol 2023:S0198-8859(23)00004-6. [PMID: 36710086 DOI: 10.1016/j.humimm.2023.01.004] [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: 10/11/2022] [Revised: 01/12/2023] [Accepted: 01/16/2023] [Indexed: 01/29/2023]
Abstract
We took advantage of the increasingly evolving approaches for in silico studies concerning protein structures, protein molecular dynamics (MD), protein-protein and protein-DNA docking to evaluate: (i) the structure and MD characteristics of the HLA-G well-recognized isoforms, (ii) the impact of missense mutations at HLA-G receptor genes (LILRB1/2), and (iii) the differential binding of the hypoxia-inducible factor 1 (HIF1) to hypoxia-responsive elements (HRE) at the HLA-G gene. Besides reviewing these topics, they were revisited including the following novel results: (i) the HLA-G6 isoforms were unstable docked or not with β2-microglobulin or peptide, (ii) missense mutations at LILRB1/2 genes, exchanging amino acids at the intracellular domain, particularly those located within and around the ITIM motifs, may impact the HLA-G binding strength, and (iii) HREs motifs at the HLA-G promoter or exon 2 regions exhibiting a guanine at their third position present a higher affinity for HIF1 when compared to an adenine at the same position. These data shed some light into the functional aspects of HLA-G, particularly how polymorphisms may influence the role of the molecule. Computational and atomistic studies have provided alternative tools for experimental physical methodologies, which are time-consuming, expensive, demanding large quantities of purified proteins, and exhibit low output.
Collapse
Affiliation(s)
- Cinthia C Alves
- Department of Medicine, Division of Clinical Immunology, Ribeirão Preto Medical School, University of São Paulo, SP, Brazil
| | - Thaís Arns
- Luxembourg Centre for Systems Biomedicine, Luxembourg
| | - Maria L Oliveira
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, SP, Brazil
| | - Philippe Moreau
- CEA, DRF-Institut François Jacob, Service de Recherches en Hémato-Immunologie, Hôpital Saint-Louis, Paris, France; U976 HIPI Unit, IRSL, Université Paris-Cité, Paris, France
| | - Dinler A Antunes
- Department of Biology and Biochemistry, University of Houston, Houston, USA
| | - Erick C Castelli
- Department of Pathology, School of Medicine, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Celso T Mendes-Junior
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil
| | - Silvana Giuliatti
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, SP, Brazil
| | - Eduardo A Donadi
- Department of Medicine, Division of Clinical Immunology, Ribeirão Preto Medical School, University of São Paulo, SP, Brazil.
| |
Collapse
|
7
|
Zheng Z, Bian C, Wang H, Su J, Meng L, Xin Y, Jiang X. Prediction of immunotherapy efficacy and immunomodulatory role of hypoxia in colorectal cancer. Ther Adv Med Oncol 2022; 14:17588359221138383. [PMID: 36425871 PMCID: PMC9679351 DOI: 10.1177/17588359221138383] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 10/26/2022] [Indexed: 11/26/2023] Open
Abstract
Immunotherapy has been used in the clinical treatment of colorectal cancer (CRC); however, most patients fail to achieve satisfactory survival benefits. Biomarkers with high specificity and sensitivity are being increasingly developed to predict the efficacy of CRC immunotherapy. In addition to DNA alteration markers, such as microsatellite instability/mismatch repair and tumor mutational burden, immune cell infiltration and immune checkpoints (ICs), epigenetic changes and no-coding RNA, and gut microbiomes all show potential predictive ability. Recently, the hypoxic tumor microenvironment (TME) has been identified as a key factor mediating CRC immune evasion and resistance to treatment. Hypoxia-inducible factor-1α is the central transcription factor in the hypoxia response that drives the expression of a vast number of survival genes by binding to the hypoxia response element in cancer and immune cells in the TME. Hypoxia regulates angiogenesis, immune cell infiltration and activation, expression of ICs, and secretion of various immune molecules in the TME and is closely associated with the immunotherapeutic efficacy of CRC. Currently, various agents targeting hypoxia have been found to improve the TME and enhance the efficacy of immunotherapy. We reviewed current markers commonly used in CRC to predict therapeutic efficacy and the mechanisms underlying hypoxia-induced angiogenesis and tumor immune evasion. Exploring the mechanisms by which hypoxia affects the TME will assist the discovery of new immunotherapeutic predictive biomarkers and development of more effective combinations of agents targeting hypoxia and immunotherapy.
Collapse
Affiliation(s)
- Zhuangzhuang Zheng
- Department of Radiation Oncology, the First Hospital of Jilin University, Changchun China
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, Changchun, China
- NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun, China
| | - Chenbin Bian
- Department of Radiation Oncology, the First Hospital of Jilin University, Changchun China
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, Changchun, China
- NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun, China
| | - Huanhuan Wang
- Department of Radiation Oncology, the First Hospital of Jilin University, Changchun China
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, Changchun, China
- NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun, China
| | - Jing Su
- Department of Radiation Oncology, the First Hospital of Jilin University, Changchun China
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, Changchun, China
- NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun, China
| | - Lingbin Meng
- Department of Hematology and Medical Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Ying Xin
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, 126 Xinmin Street, Changchun 130021, China
| | - Xin Jiang
- Department of Radiation Oncology, the First Hospital of Jilin University, 71 Xinmin Street, Changchun 130021, China
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, Changchun, China
- NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun, China
| |
Collapse
|
8
|
Zhang Y, Dai X, Li Z. Molecular subtypes of cuproptosis regulators and their correlation with clinical prognosis and immune response in glioma. Am J Transl Res 2022; 14:8085-8102. [PMID: 36505293 PMCID: PMC9730092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 10/26/2022] [Indexed: 12/15/2022]
Abstract
Cuproptosis is a newly described form of cell death. However, nothing is known about the roles of cuproptosis regulators in glioma. First, we explored the characteristics of cuproptosis molecular subtypes and relevant tumor microenvironment (TME) immune cell infiltration patterns in glioma. Using unsupervised clustering analysis, we identified two cuproptosis subtypes and three gene clusters that exhibited different clinical characteristics and TME cell infiltration patterns. Then, we developed and validated a cuproptosis-related prognostic model for predicting the overall survival of glioma patients. We established a risk score tool based on a nomogram to assess the clinical applicability of the cuproptosis model. A high cuproptosis risk score with high immune cell infiltration level, tumor mutation burden, gene alterations, and immunity activation had an unfavorable overall survival. Next, we identified possible competing endogenous ribonucleic acid regulatory networks based on significantly differentially expressed genes between high-risk and low-risk groups and screened several candidate small molecular compounds that may improve chemotherapy. Data from IMvigor and GSE78200 showed that the cuproptosis score affected the prognosis of patients who received immunotherapy. Our study indicated that cuproptosis regulators are involved in TME immune infiltration and impact the clinical prognosis in glioma. It is necessary for clinical practice to develop different therapeutic strategies according to the different phenotypes associated with immune response. The present findings provide new insight for improving immunotherapy strategies and individualized treatment in glioma.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Pathology, The Second Affiliated Hospital of Zhengzhou UniversityZhengzhou, Henan Province, China
| | - Xuehui Dai
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou UniversityZhengzhou, Henan Province, China
| | - Zhanzhan Li
- Department of Oncology, Xiangya Hospital, Central South UniversityChangsha, Hunan Province, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South UniversityChangsha, Hunan Province, China
| |
Collapse
|
9
|
Almeida RS, Gomes TT, Araújo FS, de Oliveira SAV, Santos JF, Donadi EA, Lucena-Silva N. Differentially Expressed Bone Marrow microRNAs Are Associated With Soluble HLA-G Bone Marrow Levels in Childhood Leukemia. Front Genet 2022; 13:871972. [PMID: 35774498 PMCID: PMC9237524 DOI: 10.3389/fgene.2022.871972] [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: 02/08/2022] [Accepted: 04/15/2022] [Indexed: 11/15/2022] Open
Abstract
HLA-G is a nonclassical histocompatibility class I molecule that plays a role in immune vigilance in cancer and infectious diseases. We previously reported that highly soluble HLA-G (sHLA-G) levels in the bone marrow were associated with a high blood cell count in T-acute lymphoblastic leukemia, a marker associated with a poor prognosis. To understand the posttranscriptional HLA-G gene regulation in leukemia, we evaluated the bone marrow microRNA profile associated with the HLA-G bone marrow mRNA expression and sHLA-G bone marrow levels in children exhibiting acute leukemia (B-ALL, T-ALL, and AML) using massively parallel sequencing. Ten differentially expressed miRNAs were associated with high sHLA-G bone marrow levels, and four of them (hsa-miR-4516, hsa-miR-486-5p, hsa-miR-4488, and hsa-miR-5096) targeted HLA-G, acting at distinct HLA-G gene segments. For qPCR validation, these miRNA expression levels (ΔCt) were correlated with HLA-G5 and RREB1 mRNA expressions and sHLA-G bone marrow levels according to the leukemia subtype. The hsa-miR-4488 and hsa-miR-5096 expression levels were lower in B-ALL than in AML, while that of hsa-miR-486-5p was lower in T-ALL than in AML. In T-ALL, hsa-miR-5096 correlated positively with HLA-G5 and negatively with sHLA-G. In addition, hsa-miR-4516 correlated negatively with sHLA-G levels. In AML, hsa-miR-4516 and hsa-miR-4488 correlated positively with HLA-G5 mRNA, but the HLA-G5 negatively correlated with sHLA-G. Our findings highlight the need to validate the findings of massively parallel sequencing since the experiment generally uses few individuals, and the same type of leukemia can be molecularly quite variable. We showed that miRNA's milieu in leukemia's bone marrow environment varies according to the type of leukemia and that the regulation of sHLA-G expression exerted by the same miRNA may act by a distinct mechanism in different types of leukemia.
Collapse
Affiliation(s)
- Renata Santos Almeida
- Laboratory of Immunogenetics, Department of Immunology, Aggeu Magalhães Institute, Oswaldo Cruz Foundation (Fiocruz), Recife, Brazil
| | - Thailany Thays Gomes
- Laboratory of Immunogenetics, Department of Immunology, Aggeu Magalhães Institute, Oswaldo Cruz Foundation (Fiocruz), Recife, Brazil
| | - Felipe Souza Araújo
- Laboratory of Immunogenetics, Department of Immunology, Aggeu Magalhães Institute, Oswaldo Cruz Foundation (Fiocruz), Recife, Brazil
| | - Sávio Augusto Vieira de Oliveira
- Laboratory of Immunogenetics, Department of Immunology, Aggeu Magalhães Institute, Oswaldo Cruz Foundation (Fiocruz), Recife, Brazil
| | - Jair Figueredo Santos
- Laboratory of Immunogenetics, Department of Immunology, Aggeu Magalhães Institute, Oswaldo Cruz Foundation (Fiocruz), Recife, Brazil
| | - Eduardo Antônio Donadi
- Clinical Immunology Division, Department of Medicine, School of Medicine of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, Brazil
| | - Norma Lucena-Silva
- Laboratory of Immunogenetics, Department of Immunology, Aggeu Magalhães Institute, Oswaldo Cruz Foundation (Fiocruz), Recife, Brazil
- Laboratory of Molecular Biology, Pediatric Oncology Service, IMIP Hospital, Recife, Brazil
| |
Collapse
|
10
|
Zoehler B, Fracaro L, Boldrini-Leite LM, da Silva JS, Travers PJ, Brofman PRS, Bicalho MDG, Senegaglia AC. HLA-G and CD152 Expression Levels Encourage the Use of Umbilical Cord Tissue-Derived Mesenchymal Stromal Cells as an Alternative for Immunosuppressive Therapy. Cells 2022; 11:cells11081339. [PMID: 35456019 PMCID: PMC9032010 DOI: 10.3390/cells11081339] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/30/2022] [Accepted: 04/11/2022] [Indexed: 12/04/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) have been used in immunosuppressive therapy due to their therapeutic effects, with the HLA-G molecule seeming to play a fundamental role. This work evaluated alternative MSC sources to bone marrow (BM), namely, umbilical cord tissue (UC), adipose tissue (AD) and dental pulp tissue (DP), and the influence of interferon-γ (IFN-γ) and hypoxia on the cultivation of these cells for use in immunosuppression therapies. Expression of costimulatory markers CD40, CD80 and CD86 and immunosuppressive molecules CD152 and HLA-G was analyzed. Lymphocyte inhibition assays were also performed. Sequencing of the HLA-G gene from exons 1 to 5 was performed using next-generation sequencing to determine the presence of alleles. UC-derived MSCs (UCMSCs) expressed higher CD152 and HLA-G1 under standard cultivation. UCMSCs and DP-derived MSCs (DPSCs) secreted similar levels of HLA-G5. All MSC sources inhibited the proliferation of peripheral blood mononuclear cells (PBMCs); growth under regular versus hypoxic conditions resulted in similar levels of inhibition. When IFN-γ was added, PBMC growth was inhibited to a lesser extent by UCMSCs. The HLA-G*01:04:01:01 allele appears to generate a more efficient MSC response in inhibiting lymphocyte proliferation. However, the strength of this conclusion was limited by the small sample size. UCMSCs are an excellent alternative to BM in immunosuppressive therapy: they express high concentrations of inhibitory molecules and can be cultivated without stimuli, which minimizes cost.
Collapse
Affiliation(s)
- Bernardo Zoehler
- Immunogenetics and Histocompatibility Laboratory, Department of Genetics, Universidade Federal do Paraná (UFPR), Curitiba 81530-001, PR, Brazil; (J.S.d.S.); (M.d.G.B.)
- Correspondence: (B.Z.); (A.C.S.)
| | - Letícia Fracaro
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba 80910-215, PR, Brazil; (L.F.); (L.M.B.-L.); (P.R.S.B.)
- National Institute of Science and Technology for Regenerative Medicine, INCT-REGENERA, Rio de Janeiro 21941-902, RJ, Brazil
| | - Lidiane Maria Boldrini-Leite
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba 80910-215, PR, Brazil; (L.F.); (L.M.B.-L.); (P.R.S.B.)
- National Institute of Science and Technology for Regenerative Medicine, INCT-REGENERA, Rio de Janeiro 21941-902, RJ, Brazil
| | - José Samuel da Silva
- Immunogenetics and Histocompatibility Laboratory, Department of Genetics, Universidade Federal do Paraná (UFPR), Curitiba 81530-001, PR, Brazil; (J.S.d.S.); (M.d.G.B.)
| | - Paul J. Travers
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK;
| | - Paulo Roberto Slud Brofman
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba 80910-215, PR, Brazil; (L.F.); (L.M.B.-L.); (P.R.S.B.)
- National Institute of Science and Technology for Regenerative Medicine, INCT-REGENERA, Rio de Janeiro 21941-902, RJ, Brazil
| | - Maria da Graça Bicalho
- Immunogenetics and Histocompatibility Laboratory, Department of Genetics, Universidade Federal do Paraná (UFPR), Curitiba 81530-001, PR, Brazil; (J.S.d.S.); (M.d.G.B.)
| | - Alexandra Cristina Senegaglia
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba 80910-215, PR, Brazil; (L.F.); (L.M.B.-L.); (P.R.S.B.)
- National Institute of Science and Technology for Regenerative Medicine, INCT-REGENERA, Rio de Janeiro 21941-902, RJ, Brazil
- Correspondence: (B.Z.); (A.C.S.)
| |
Collapse
|
11
|
Syukri A, Budu, Hatta M, Amir M, Rohman MS, Mappangara I, Kaelan C, Wahyuni S, Bukhari A, Junita AR, Primaguna MR, Dwiyanti R, Febrianti A. Doxorubicin induced immune abnormalities and inflammatory responses via HMGB1, HIF1-α and VEGF pathway in progressive of cardiovascular damage. Ann Med Surg (Lond) 2022; 76:103501. [PMID: 35340325 PMCID: PMC8943401 DOI: 10.1016/j.amsu.2022.103501] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 03/08/2022] [Accepted: 03/08/2022] [Indexed: 02/06/2023] Open
Abstract
Background Doxorubicin (DOX) is a commonly used treatment for cancer and the mechanism of DOX-induced cardiomyocyte damage in cardiovascular disease is not fully understood. High-mobility group box 1 (HMGB1), strong induce proinflammatory cytokines via damage associated molecular pattern (DAMP) which its interaction with the receptor of advanced glycation end products (RAGE), that affect cytokine release, and angiogenesis via the role of HMBG1, HIF-1α and VEGF as an important regulator in these cardiac failure processes. Hypoxia-inducible factor-1α (HIF-1α) is plays an important role in the cellular response to systemic oxygen levels of cells and VEGF is an angiogenic factor and can stimulate cellular responses on the surface of endothelial cells will be described Objective The aim of this article is to comprehensively review the role of HMGB1, HIF-1α, and VEGF in DOX-induced Cardiovascular Disease and its molecular mechanisms. Methods The data in this study were collect by search the keyword combinations of medical subject headings (MeSH) of “HMGB1”, “HIF-1 α”, “VEGF”, “DOX” and “Cardiovascular disease” and relevant reference lists were manually searched in PubMed, EMBASE and Scopus database. All relevant articles in data base above were included and narratively discussed in this review article. Results Several articles were revealed that molecular mechanisms of the DOX in cardiomyocyte damage and related to HMGB1, HIF-1α and VEGF and may potential treatment and prevention to cardiovascular disease in DOX intervention. Conclusion HMGB1, HIF-1α and VEGF has a pivotal regulator in DOX-induce cardiomyocyte damage and predominantly acts through different pathways. The role of HMGB1 in DOX-induced myocardial damage suggests that HMGB1 is a mediator of DOX-induced damage. In addition, DOX can inhibit HIF-1α activity where DOX can decrease HIF-1α expression and HIF-1α is also responsible for upregulation of several angiogenic factors, including VEGF. VEGF plays an important role in angiogenesis and anti-angiogenesis both in vitro and in vivo and reduces the side effects of DOX markedly. In addition, the administration of anti-angiogenesis will show an inhibitory effect on angiogenesis mediated by the VEGF signaling pathway and triggered by DOX in cells. The effect of Doxorubicin (DOX) induced cardiovascular damage via several pathways. Cardiovascular damage can involve HMGB1, HIF-1α, and VEGF. HMGB1, HIF-1α, and VEGF as a pivotal regulator in DOX-induce cardiomyocyte damage. HMGB1, HIF-1α, and VEGF in cardiovascular diseases will be predominantly acting through different pathways.
Collapse
|
12
|
HLA-G and Other Immune Checkpoint Molecules as Targets for Novel Combined Immunotherapies. Int J Mol Sci 2022; 23:ijms23062925. [PMID: 35328349 PMCID: PMC8948858 DOI: 10.3390/ijms23062925] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/01/2022] [Accepted: 03/04/2022] [Indexed: 12/14/2022] Open
Abstract
HLA-G is an HLA-class Ib molecule that is involved in the establishment of tolerance at the maternal/fetal interface during pregnancy. The expression of HLA-G is highly restricted in adults, but the de novo expression of this molecule may be observed in different hematological and solid tumors and is related to cancer progression. Indeed, tumor cells expressing high levels of HLA-G are able to suppress anti-tumor responses, thus escaping from the control of the immune system. HLA-G has been proposed as an immune checkpoint (IC) molecule due to its crucial role in tumor progression, immune escape, and metastatic spread. We here review data available in the literature in which the interaction between HLA-G and other IC molecules is reported, in particular PD-1, CTLA-4, and TIM-3, but also IDO and TIGIT. Clinical trials using monoclonal antibodies against HLA-G and other IC are currently ongoing with cancer patients where antibodies and inhibitors of PD-1 and CTLA-4 showed encouraging results. With this background, we may envisage that combined therapies using antibodies targeting HLA-G and another IC may be successful for clinical purposes. Indeed, such immunotherapeutic protocols may achieve a better rescue of effective anti-tumor immune response, thus improving the clinical outcome of patients.
Collapse
|
13
|
Dietz S, Schwarz J, Velic A, González-Menéndez I, Quintanilla-Martinez L, Casadei N, Marmé A, Poets CF, Gille C, Köstlin-Gille N. Human Leucocyte Antigen G and Murine Qa-2 Are Critical for Myeloid Derived Suppressor Cell Expansion and Activation and for Successful Pregnancy Outcome. Front Immunol 2022; 12:787468. [PMID: 35111157 PMCID: PMC8801456 DOI: 10.3389/fimmu.2021.787468] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/27/2021] [Indexed: 12/03/2022] Open
Abstract
During pregnancy, maternal immune system has to balance tightly between protection against pathogens and tolerance towards a semi-allogeneic organism. Dysfunction of this immune adaptation can lead to severe complications such as pregnancy loss, preeclampsia or fetal growth restriction. In the present study we analyzed the impact of the murine MHC class Ib molecule Qa-2 on pregnancy outcome in vivo. We demonstrate that lack of Qa-2 led to intrauterine growth restriction and increased abortion rates especially in late pregnancy accompanied by a disturbed trophoblast invasion and altered spiral artery remodeling as well as protein aggregation in trophoblast cells indicating a preeclampsia-like phenotype. Furthermore, lack of Qa-2 caused imbalanced immunological adaptation to pregnancy with altered immune cell and especially T-cell homeostasis, reduced Treg numbers and decreased accumulation and functional activation of myeloid-derived suppressor cells. Lastly, we show that application of sHLA-G reduced abortion rates in Qa-2 deficient mice by inducing MDSC. Our results highlight the importance of an interaction between HLA-G and MDSC for pregnancy success and the therapeutic potential of HLA-G for treatment of immunological pregnancy complications.
Collapse
Affiliation(s)
- Stefanie Dietz
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany
| | - Julian Schwarz
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany
| | - Ana Velic
- Interfaculty Institute for Cell Biology, Proteome Center Tuebingen (PCT), University of Tuebingen, Tübingen, Germany
| | | | | | - Nicolas Casadei
- Next Generation Sequencing (NGS) Competence Center Tuebingen (NCCT), Tuebingen, Germany; Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany
| | - Alexander Marmé
- Gynecology and Obstetrics Practice, Am Lustnauer Tor, Tuebingen, Germany
| | - Christian F Poets
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany
| | - Christian Gille
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany.,Department of Neonatology, Heidelberg University Children's Hospital, Heidelberg, Germany
| | - Natascha Köstlin-Gille
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany.,Department of Neonatology, Heidelberg University Children's Hospital, Heidelberg, Germany
| |
Collapse
|
14
|
Ray SK, Mukherjee S. Directing hypoxic tumor microenvironment and HIF to illuminate cancer immunotherapy's existing prospects and challenges in drug targets. Curr Drug Targets 2022; 23:471-485. [PMID: 35021970 DOI: 10.2174/1389450123666220111114649] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 11/22/2022]
Abstract
Cancer is now also reflected as a disease of the tumor microenvironment, primarily supposed to be a decontrolled genetic and cellular expression disease. Over the past two decades, significant and rapid progress has been made in recognizing the dynamics of the tumor's microenvironment and its contribution to influencing the response to various anti-cancer therapies and drugs. Modulations in the tumor microenvironment and immune checkpoint blockade are interesting in cancer immunotherapy and drug targets. Simultaneously, the immunotherapeutic strategy can be done by modulating the immune regulatory pathway; however, the tumor microenvironment plays an essential role in suppressing the antitumor's immunity by its substantial heterogeneity. Hypoxia inducible factor (HIF) is a significant contributor to solid tumor heterogeneity and a key stressor in the tumor microenvironment to drive adaptations to prevent immune surveillance. Checkpoint inhibitors here halt the ability of cancer cells to stop the immune system from activating, and in turn, amplify your body's immune system to help destroy cancer cells. Common checkpoints that these inhibitors affect are the PD-1/PD-L1 and CTLA-4 pathways and important drugs involved are Ipilimumab and Nivolumab, mainly along with other drugs in this group. Targeting the hypoxic tumor microenvironment may provide a novel immunotherapy strategy, break down traditional cancer therapy resistance, and build the framework for personalized precision medicine and cancer drug targets. We hope that this knowledge can provide insight into the therapeutic potential of targeting Hypoxia and help to develop novel combination approaches of cancer drugs to increase the effectiveness of existing cancer therapies, including immunotherapy.
Collapse
Affiliation(s)
| | - Sukhes Mukherjee
- Department of Biochemistry. All India Institute of Medical Sciences. Bhopal, Madhya pradesh-462020. India
| |
Collapse
|
15
|
Xu Y, He L, Fu Q, Hu J. Metabolic Reprogramming in the Tumor Microenvironment With Immunocytes and Immune Checkpoints. Front Oncol 2021; 11:759015. [PMID: 34858835 PMCID: PMC8632143 DOI: 10.3389/fonc.2021.759015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 10/27/2021] [Indexed: 12/19/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs), Ipilimumab, Nivolumab, Pembrolizumab and Atezolizumab, have been applied in anti-tumor therapy and demonstrated exciting performance compared to conventional treatments. However, the unsatisfactory response rates, high recurrence and adaptive resistance limit their benefits. Metabolic reprogramming appears to be one of the crucial barriers to immunotherapy. The deprivation of required nutrients and altered metabolites not only promote tumor progression but also confer dysfunction on immune cells in the tumor microenvironment (TME). Glycolysis plays a central role in metabolic reprogramming and immunoregulation in the TME, and many therapies targeting glycolysis have been developed, and their combinations with ICIs are in preclinical and clinical trials. Additional attention has been paid to the role of amino acids, lipids, nucleotides and mitochondrial biogenesis in metabolic reprogramming and clinical anti-tumor therapy. This review attempts to describe reprogramming metabolisms within tumor cells and immune cells, from the aspects of glycolysis, amino acid metabolism, lipid metabolism, nucleotide metabolism and mitochondrial biogenesis and their impact on immunity in the TME, as well as the significance of targeting metabolism in anti-tumor therapy, especially in combination with ICIs. In particular, we highlight the expression mechanism of programmed cell death (ligand) 1 [PD-(L)1] in tumor cells and immune cells under reprogramming metabolism, and discuss in detail the potential of targeting key metabolic pathways to break resistance and improve the efficacy of ICIs based on results from current preclinical and clinical trials. Besides, we draw out biomarkers of potential predictive value in ICIs treatment from a metabolic perspective.
Collapse
Affiliation(s)
- Yaolin Xu
- Department of Oncology, The People's Hospital of China Medical University/The People's Hospital of LiaoNing Province, Shenyang, China
| | - Lijie He
- Department of Oncology, The People's Hospital of China Medical University/The People's Hospital of LiaoNing Province, Shenyang, China
| | - Qiang Fu
- Department of Cardiology, The People's Hospital of China Medical University/The People's Hospital of LiaoNing Province, Shenyang, China
| | - Junzhe Hu
- The Second Clinic Medical College, China Medical University, Shenyang, China
| |
Collapse
|
16
|
Alves CC, Donadi EA, Giuliatti S. Structural Characterization of the Interaction of Hypoxia Inducible Factor-1 with Its Hypoxia Responsive Element at the -964G > A Variation Site of the HLA-G Promoter Region. Int J Mol Sci 2021; 22:ijms222313046. [PMID: 34884849 PMCID: PMC8657931 DOI: 10.3390/ijms222313046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/08/2021] [Accepted: 11/12/2021] [Indexed: 11/16/2022] Open
Abstract
Human Antigen Leukocyte-G (HLA-G) gene encodes an immune checkpoint molecule that has restricted tissue expression in physiological conditions; however, the gene may be induced in hypoxic conditions by the interaction with the hypoxia inducible factor-1 (HIF1). Hypoxia regulatory elements (HRE) located at the HLA-G promoter region and at exon 2 are the major HIF1 target sites. Since the G allele of the −964G > A transversion induces higher HLA-G expression when compared to the A allele in hypoxic conditions, here we analyzed HIF1-HRE complex interaction at the pair-atom level considering both −964G > A polymorphism alleles. Mouse HIF2 dimer crystal (Protein Data Bank ID: 4ZPK) was used as template to perform homology modelling of human HIF1 quaternary structure using MODELLER v9.14. Two 3D DNA structures were built from 5′GCRTG’3 HRE sequence containing the −964G/A alleles using x3DNA. Protein-DNA docking was performed using the HADDOCK v2.4 server, and non-covalent bonds were computed by DNAproDB server. Molecular dynamic simulation was carried out per 200 ns, using Gromacs v.2019. HIF1 binding in the HRE containing −964G allele results in more hydrogen bonds and van der Waals contact formation than HRE with −964A allele. Protein-DNA complex trajectory analysis revealed that HIF1-HRE-964G complex is more stable. In conclusion, HIF1 binds in a more stable and specific manner at the HRE with G allele.
Collapse
Affiliation(s)
- Cinthia C. Alves
- Department of Genetic, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, Brazil;
| | - Eduardo A. Donadi
- Department of Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, Brazil;
| | - Silvana Giuliatti
- Department of Genetic, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, Brazil;
- Correspondence:
| |
Collapse
|
17
|
Castelli EC, de Almeida BS, Muniz YCN, Silva NSB, Passos MRS, Souza AS, Page AE, Dyble M, Smith D, Aguileta G, Bertranpetit J, Migliano AB, Duarte YAO, Scliar MO, Wang J, Passos-Bueno MR, Naslavsky MS, Zatz M, Mendes-Junior CT, Donadi EA. HLA-G genetic diversity and evolutive aspects in worldwide populations. Sci Rep 2021; 11:23070. [PMID: 34845256 PMCID: PMC8629979 DOI: 10.1038/s41598-021-02106-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 11/01/2021] [Indexed: 12/15/2022] Open
Abstract
HLA-G is a promiscuous immune checkpoint molecule. The HLA-G gene presents substantial nucleotide variability in its regulatory regions. However, it encodes a limited number of proteins compared to classical HLA class I genes. We characterized the HLA-G genetic variability in 4640 individuals from 88 different population samples across the globe by using a state-of-the-art method to characterize polymorphisms and haplotypes from high-coverage next-generation sequencing data. We also provide insights regarding the HLA-G genetic diversity and a resource for future studies evaluating HLA-G polymorphisms in different populations and association studies. Despite the great haplotype variability, we demonstrated that: (1) most of the HLA-G polymorphisms are in introns and regulatory sequences, and these are the sites with evidence of balancing selection, (2) linkage disequilibrium is high throughout the gene, extending up to HLA-A, (3) there are few proteins frequently observed in worldwide populations, with lack of variation in residues associated with major HLA-G biological properties (dimer formation, interaction with leukocyte receptors). These observations corroborate the role of HLA-G as an immune checkpoint molecule rather than as an antigen-presenting molecule. Understanding HLA-G variability across populations is relevant for disease association and functional studies.
Collapse
Affiliation(s)
- Erick C Castelli
- Molecular Genetics and Bioinformatics Laboratory, Experimental Research Unit, School of Medicine, São Paulo State University (UNESP), Botucatu, State of São Paulo, Brazil.
- Department of Pathology, School of Medicine, São Paulo State University (UNESP), Botucatu, State of São Paulo, CEP: 18618970, Brazil.
| | - Bibiana S de Almeida
- Division of Clinical Immunology, Department of Medicine, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, CEP: 14049-900, Brazil
- Laboratório Multiusuário de Estudos em Biologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina (UFSC), Florianópolis, Brazil
| | - Yara C N Muniz
- Departamento de Biologia Celular, Embriologia e Genética, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina (UFSC), Florianópolis, Brazil
| | - Nayane S B Silva
- Molecular Genetics and Bioinformatics Laboratory, Experimental Research Unit, School of Medicine, São Paulo State University (UNESP), Botucatu, State of São Paulo, Brazil
| | - Marília R S Passos
- Molecular Genetics and Bioinformatics Laboratory, Experimental Research Unit, School of Medicine, São Paulo State University (UNESP), Botucatu, State of São Paulo, Brazil
| | - Andreia S Souza
- Molecular Genetics and Bioinformatics Laboratory, Experimental Research Unit, School of Medicine, São Paulo State University (UNESP), Botucatu, State of São Paulo, Brazil
| | - Abigail E Page
- Department of Population Health, London School of Hygiene and Tropical Medicine, London, UK
| | - Mark Dyble
- Departament of Anthropology, University College London (UCL), London, UK
| | - Daniel Smith
- Bristol Medical School (PHS), University of Bristol, Bristol, UK
| | - Gabriela Aguileta
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Jaume Bertranpetit
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Andrea B Migliano
- Departament of Anthropology, Unversity of Zurich, Zurich, Switzerland
| | - Yeda A O Duarte
- Escola de Enfermagem e Faculdade de Saúde Pública, Universidade de São Paulo (USP), São Paulo, State of São Paulo, Brazil
| | - Marília O Scliar
- Human Genome and Stem Cell Research Center, Biosciences Institute, University of São Paulo (USP), São Paulo, State of São Paulo, Brazil
| | - Jaqueline Wang
- Human Genome and Stem Cell Research Center, Biosciences Institute, University of São Paulo (USP), São Paulo, State of São Paulo, Brazil
| | - Maria Rita Passos-Bueno
- Human Genome and Stem Cell Research Center, Biosciences Institute, University of São Paulo (USP), São Paulo, State of São Paulo, Brazil
- Department of Genetics and Evolutionary Biology, Biosciences Institute, University of São Paulo (USP), São Paulo, State of São Paulo, Brazil
| | - Michel S Naslavsky
- Human Genome and Stem Cell Research Center, Biosciences Institute, University of São Paulo (USP), São Paulo, State of São Paulo, Brazil
- Department of Genetics and Evolutionary Biology, Biosciences Institute, University of São Paulo (USP), São Paulo, State of São Paulo, Brazil
- Hospital Israelita Albert Einstein, São Paulo, State of São Paulo, Brazil
| | - Mayana Zatz
- Human Genome and Stem Cell Research Center, Biosciences Institute, University of São Paulo (USP), São Paulo, State of São Paulo, Brazil
- Department of Genetics and Evolutionary Biology, Biosciences Institute, University of São Paulo (USP), São Paulo, State of São Paulo, Brazil
| | - Celso Teixeira Mendes-Junior
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, 14040-901, Ribeirão Preto, SP, Brazil
| | - Eduardo A Donadi
- Division of Clinical Immunology, Department of Medicine, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, CEP: 14049-900, Brazil.
| |
Collapse
|
18
|
Bouleftour W, Rowinski E, Louati S, Sotton S, Wozny AS, Moreno-Acosta P, Mery B, Rodriguez-Lafrasse C, Magne N. A Review of the Role of Hypoxia in Radioresistance in Cancer Therapy. Med Sci Monit 2021; 27:e934116. [PMID: 34728593 PMCID: PMC8573967 DOI: 10.12659/msm.934116] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Hypoxia involves neoplastic cells. Unlike normal tissue, solid tumors are composed of aberrant vasculature, leading to a hypoxic microenvironment. Hypoxia is also known to be involved in both metastasis initiation and therapy resistance. Radiotherapy is the appropriate treatment in about half of all cancers, but loco-regional control failure and a disease recurrence often occur due to clinical radioresistance. Hypoxia induces radioresistance through a number of molecular pathways, and numerous strategies have been developed to overcome this. Nevertheless, these strategies have resulted in disappointing results, including adverse effects and limited efficacy. Additional clinical studies are needed to achieve a better understanding of the complex hypoxia pathways. This review presents an update on the mechanisms of hypoxia in radioresistance in solid tumors and the potential therapeutic solutions.
Collapse
Affiliation(s)
- Wafa Bouleftour
- Radiotherapy Department, Lucien Neuwirth Cancerology Institute, Saint Priest en Jarez, France
| | - Elise Rowinski
- Radiotherapy Department, Lucien Neuwirth Cancerology Institute, Saint Priest en Jarez, France
| | - Safa Louati
- Université Lyon 1, Faculté de Médecine-Lyon-Sud, Oullins, France.,Laboratoire de Radiobiologie Cellulaire et Moléculaire, CNRS UMR 5822, Institut de Physique Nucléaire de Lyon, IPNL, Villeurbanne, France.,Hospices Civils de Lyon, Lyon, France
| | - Sandrine Sotton
- Radiotherapy Department, Lucien Neuwirth Cancerology Institute, Saint Priest en Jarez, France
| | - Anne-Sophie Wozny
- Université Lyon 1, Faculté de Médecine-Lyon-Sud, Oullins, France.,Laboratoire de Radiobiologie Cellulaire et Moléculaire, CNRS UMR 5822, Institut de Physique Nucléaire de Lyon, IPNL, Villeurbanne, France.,Hospices Civils de Lyon, Lyon, France
| | - Pablo Moreno-Acosta
- Research Group in Cancer Biology, National Cancer Institute, Bogotá, Colombia
| | - Benoite Mery
- Radiotherapy Department, Lucien Neuwirth Cancerology Institute, Saint Priest en Jarez, France
| | - Claire Rodriguez-Lafrasse
- Université Lyon 1, Faculté de Médecine-Lyon-Sud, Oullins, France.,Laboratoire de Radiobiologie Cellulaire et Moléculaire, CNRS UMR 5822, Institut de Physique Nucléaire de Lyon, IPNL, Villeurbanne, France.,Hospices Civils de Lyon, Lyon, France
| | - Nicolas Magne
- Radiotherapy Department, Lucien Neuwirth Cancerology Institute, Saint Priest en Jarez, France.,Université Lyon 1, Faculté de Médecine-Lyon-Sud, Oullins, France.,Laboratoire de Radiobiologie Cellulaire et Moléculaire, CNRS UMR 5822, Institut de Physique Nucléaire de Lyon, IPNL, Villeurbanne, France.,Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
19
|
Desaulniers D, Vasseur P, Jacobs A, Aguila MC, Ertych N, Jacobs MN. Integration of Epigenetic Mechanisms into Non-Genotoxic Carcinogenicity Hazard Assessment: Focus on DNA Methylation and Histone Modifications. Int J Mol Sci 2021; 22:10969. [PMID: 34681626 PMCID: PMC8535778 DOI: 10.3390/ijms222010969] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/30/2021] [Accepted: 10/04/2021] [Indexed: 12/15/2022] Open
Abstract
Epigenetics involves a series of mechanisms that entail histone and DNA covalent modifications and non-coding RNAs, and that collectively contribute to programing cell functions and differentiation. Epigenetic anomalies and DNA mutations are co-drivers of cellular dysfunctions, including carcinogenesis. Alterations of the epigenetic system occur in cancers whether the initial carcinogenic events are from genotoxic (GTxC) or non-genotoxic (NGTxC) carcinogens. NGTxC are not inherently DNA reactive, they do not have a unifying mode of action and as yet there are no regulatory test guidelines addressing mechanisms of NGTxC. To fil this gap, the Test Guideline Programme of the Organisation for Economic Cooperation and Development is developing a framework for an integrated approach for the testing and assessment (IATA) of NGTxC and is considering assays that address key events of cancer hallmarks. Here, with the intent of better understanding the applicability of epigenetic assays in chemical carcinogenicity assessment, we focus on DNA methylation and histone modifications and review: (1) epigenetic mechanisms contributing to carcinogenesis, (2) epigenetic mechanisms altered following exposure to arsenic, nickel, or phenobarbital in order to identify common carcinogen-specific mechanisms, (3) characteristics of a series of epigenetic assay types, and (4) epigenetic assay validation needs in the context of chemical hazard assessment. As a key component of numerous NGTxC mechanisms of action, epigenetic assays included in IATA assay combinations can contribute to improved chemical carcinogen identification for the better protection of public health.
Collapse
Affiliation(s)
- Daniel Desaulniers
- Environmental Health Sciences and Research Bureau, Hazard Identification Division, Health Canada, AL:2203B, Ottawa, ON K1A 0K9, Canada
| | - Paule Vasseur
- CNRS, LIEC, Université de Lorraine, 57070 Metz, France;
| | - Abigail Jacobs
- Independent at the Time of Publication, Previously US Food and Drug Administration, Rockville, MD 20852, USA;
| | - M. Cecilia Aguila
- Toxicology Team, Division of Human Food Safety, Center for Veterinary Medicine, US Food and Drug Administration, Department of Health and Human Services, Rockville, MD 20852, USA;
| | - Norman Ertych
- German Centre for the Protection of Laboratory Animals (Bf3R), German Federal Institute for Risk Assessment, Diedersdorfer Weg 1, 12277 Berlin, Germany;
| | - Miriam N. Jacobs
- Centre for Radiation, Chemical and Environmental Hazards, Public Health England, Chilton OX11 0RQ, UK;
| |
Collapse
|
20
|
Blocking HIF to Enhance NK Cells: Hints for New Anti-Tumor Therapeutic Strategies? Vaccines (Basel) 2021; 9:vaccines9101144. [PMID: 34696251 PMCID: PMC8539190 DOI: 10.3390/vaccines9101144] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 09/30/2021] [Accepted: 10/01/2021] [Indexed: 12/18/2022] Open
Abstract
Natural Killer (NK) cells are becoming an ever more promising tool to design new anti-tumor strategies. However, two major issues are still a challenge to obtain versatile and effective NK-based therapies: the way to maximize the persistency of powerful NK effectors in the patient, and the way to overcome the multiple escape mechanisms that keep away or suppress NK cells at the tumor site. In this regard, targeting the hypoxia-inducible factors (HIFs), which is important for both tumor progression and immune suppression, may be an opportunity. Especially, in the context of the ongoing studies focused on more effective NK-based therapeutic products.
Collapse
|
21
|
Bhushan A, Kumari R, Srivastava T. Scouting for common genes in the heterogenous hypoxic tumor microenvironment and their validation in glioblastoma. 3 Biotech 2021; 11:451. [PMID: 34631352 DOI: 10.1007/s13205-021-02987-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 09/04/2021] [Indexed: 12/17/2022] Open
Abstract
Investigating the therapeutic and prognostic potential of genes in the heterogeneous hypoxic niche of glioblastoma. We have analyzed RNA expression of U87MG cells cultured in hypoxia compared to normoxia. Common differentially expressed genes (DEGs) from GSE45301 and GSE18494 and their functional enrichment was performed using MetaScape and PANTHER. Hub genes and their ontology were identified using MCode cytoHubba and ClueGO and validated with GlioVis, Oncomine, HPA and PrognoScan. Using the GEO2R analysis of GSE45301 and GSE18494 datasets, we have found a total of 246 common DEGs (180 upregulated and 66 downregulated) and identified 2 significant modules involved in ribosome biogenesis and TNF signaling. Meta-analysis of key genes of each module in cytoHubba identified 17 hub genes (ATF3, BYSL, DUSP1, EGFR, JUN, ETS1, LYAR, NIP7, NOLC1, NOP2, NOP56, PNO1, RRS1, TNFAIP3, TNFRSF1B, UTP15, VEGFA). Of the 17 hub genes, ATF3, BYSL, EGFR, JUN, NIP7, NOLC1, PNO1, RRS1, TNFAIP3 and VEGFA were identified as hypoxia signatures associated with poor prognosis in Glioma. Ribosome biogenesis emerged as a vital contender of possible therapeutic potential with BYSL, NIP7, NOLC1, PNO1 and RRS1 showing prognostic value. SUPPLEMENTARY INFORMATION The online version contains supplementary material available at 10.1007/s13205-021-02987-2.
Collapse
Affiliation(s)
- Ashish Bhushan
- Department of Genetics, University of Delhi South Campus, Benito Juarez Road, New Delhi, 110021 India
| | - Ranbala Kumari
- National Institute of Pathology (ICMR), Safdarjung Hospital Campus, New Delhi, India
| | - Tapasya Srivastava
- Department of Genetics, University of Delhi South Campus, Benito Juarez Road, New Delhi, 110021 India
| |
Collapse
|
22
|
Jacovas VC, Michita RT, Bisso-Machado R, Reales G, Tarazona-Santos EM, Sandoval JR, Salazar-Granara A, Chies JAB, Bortolini MC. HLA-G 3'UTR haplotype frequencies in highland and lowland South Native American populations. Hum Immunol 2021; 83:27-38. [PMID: 34563386 DOI: 10.1016/j.humimm.2021.09.002] [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: 03/18/2021] [Revised: 08/09/2021] [Accepted: 09/01/2021] [Indexed: 11/30/2022]
Abstract
Human Leukocyte Antigen (HLA)-G participates in several biological processes, including reproduction, vascular remodeling, immune tolerance, and hypoxia response. HLA-G is a potential candidate gene for high altitude adaptation since its expression is modulated in both micro and macro environment under hypoxia and constant cellular stress. Besides the promoter region, the HLA-G 3'untranslated region (UTR) influences HLA-G expression patterns through several post-transcriptional mechanisms. Currently, the 3'UTR genetic diversity in terms of altitude adaptation of Native American populations is still unexplored, particularly at high altitude ecoregions. Here, we evaluated 288 Native Americans from 9 communities located in the Andes [highland (HL); ≥2,500 m (range = 2,838-4,433 m)] and 8 populations located in lowland (LL) regions [<2,500 m (range = 80-431 m); Amazonian tropical forest, Brazilian central plateau, and Chaco] of South America. In total, nine polymorphic sites and ten haplotypes were observed. The most frequent haplotypes (UTR-1, UTR-2, and UTR-3) accounted for ∼ 77% of haplotypes found in LL, while in the HL, the same haplotypes reach ∼ 93%. Also, a remarkable high frequency of putative ancestral UTR-5 haplotype was observed in LL (21.5%), while in HL UTR-2 reaches up to 47%. Further, UTR-2 frequency positively correlates with altitude-related variables, while a negative correlation for UTR-5 was observed. From an evolutionary perspective, we observed a tendency towards balancing selection in HL and LL populations thus suggesting that haplotypes of ancient and more derived alleles may have been co-opted for relatively recent adaptations such as those experienced by modern humans in the highland and lowland of South America. We also discuss how long-term balancing selection can be a reservoir of genetic variants that can be positively selected. Finally, our study provides some pieces of evidence that HLA-G 3'UTR haplotypes may have contributed to high altitude adaptation in the Andes.
Collapse
Affiliation(s)
- Vanessa Cristina Jacovas
- Departamento de Genética, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Rafael Tomoya Michita
- Departamento de Genética, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil; Human Molecular Genetics Laboratory, Universidade Luterana do Brasil (ULBRA), Canoas, Rio Grande do Sul, Brazil
| | - Rafael Bisso-Machado
- Departamento de Genética, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Guillermo Reales
- Departamento de Genética, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Eduardo M Tarazona-Santos
- Departamento de Biologia Geral, Universidade Federal de Minas Gerais, Instituto de Ciências Biológicas, Belo Horizonte, Minas Gerais, Brazil
| | - José Raul Sandoval
- Departamento de Biologia Geral, Universidade Federal de Minas Gerais, Instituto de Ciências Biológicas, Belo Horizonte, Minas Gerais, Brazil
| | | | - José Artur Bogo Chies
- Departamento de Genética, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil.
| | - Maria Cátira Bortolini
- Departamento de Genética, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil.
| |
Collapse
|
23
|
Hazini A, Fisher K, Seymour L. Deregulation of HLA-I in cancer and its central importance for immunotherapy. J Immunother Cancer 2021; 9:e002899. [PMID: 34353849 PMCID: PMC8344275 DOI: 10.1136/jitc-2021-002899] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2021] [Indexed: 12/28/2022] Open
Abstract
It is now well accepted that many tumors undergo a process of clonal selection which means that tumor antigens arising at various stages of tumor progression are likely to be represented in just a subset of tumor cells. This process is thought to be driven by constant immunosurveillance which applies selective pressure by eliminating tumor cells expressing antigens that are recognized by T cells. It is becoming increasingly clear that the same selective pressure may also select for tumor cells that evade immune detection by acquiring deficiencies in their human leucocyte antigen (HLA) presentation pathways, allowing important tumor antigens to persist within cells undetected by the immune system. Deficiencies in antigen presentation pathway can arise by a variety of mechanisms, including genetic and epigenetic changes, and functional antigen presentation is a hard phenomenon to assess using our standard analytical techniques. Nevertheless, it is likely to have profound clinical significance and could well define whether an individual patient will respond to a particular type of therapy or not. In this review we consider the mechanisms by which HLA function may be lost in clinical disease, we assess the implications for current immunotherapy approaches using checkpoint inhibitors and examine the prognostic impact of HLA loss demonstrated in clinical trials so far. Finally, we propose strategies that might be explored for possible patient stratification.
Collapse
Affiliation(s)
- Ahmet Hazini
- Department of Oncology, University of Oxford, Oxford, Oxfordshire, UK
| | - Kerry Fisher
- Department of Oncology, University of Oxford, Oxford, Oxfordshire, UK
| | - Len Seymour
- Department of Oncology, University of Oxford, Oxford, Oxfordshire, UK
| |
Collapse
|
24
|
Cruz-Bermúdez A, Laza-Briviesca R, Casarrubios M, Sierra-Rodero B, Provencio M. The Role of Metabolism in Tumor Immune Evasion: Novel Approaches to Improve Immunotherapy. Biomedicines 2021; 9:361. [PMID: 33807260 PMCID: PMC8067102 DOI: 10.3390/biomedicines9040361] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/25/2021] [Accepted: 03/28/2021] [Indexed: 12/16/2022] Open
Abstract
The tumor microenvironment exhibits altered metabolic properties as a consequence of the needs of tumor cells, the natural selection of the most adapted clones, and the selfish relationship with other cell types. Beyond its role in supporting uncontrolled tumor growth, through energy and building materials obtention, metabolism is a key element controlling tumor immune evasion. Immunotherapy has revolutionized the treatment of cancer, being the first line of treatment for multiple types of malignancies. However, many patients either do not benefit from immunotherapy or eventually relapse. In this review we overview the immunoediting process with a focus on the metabolism-related elements that are responsible for increased immune evasion, either through reduced immunogenicity or increased resistance of tumor cells to the apoptotic action of immune cells. Finally, we describe the main molecules to modulate these immune evasion processes through the control of the metabolic microenvironment as well as their clinical developmental status.
Collapse
Affiliation(s)
- Alberto Cruz-Bermúdez
- Medical Oncology Department, Health Research Institute Puerta de Hierro–Segovia de Arana (IDIPHISA) & Puerta de Hierro Hospital, Manuel de Falla Street #1, 28222 Madrid, Spain; (R.L.-B.); (M.C.); (B.S.-R.)
| | - Raquel Laza-Briviesca
- Medical Oncology Department, Health Research Institute Puerta de Hierro–Segovia de Arana (IDIPHISA) & Puerta de Hierro Hospital, Manuel de Falla Street #1, 28222 Madrid, Spain; (R.L.-B.); (M.C.); (B.S.-R.)
- PhD Programme in Molecular Biosciences, Faculty of Medicine Doctoral School, Universidad Autónoma de Madrid, 28222 Madrid, Spain
| | - Marta Casarrubios
- Medical Oncology Department, Health Research Institute Puerta de Hierro–Segovia de Arana (IDIPHISA) & Puerta de Hierro Hospital, Manuel de Falla Street #1, 28222 Madrid, Spain; (R.L.-B.); (M.C.); (B.S.-R.)
- PhD Programme in Molecular Biosciences, Faculty of Medicine Doctoral School, Universidad Autónoma de Madrid, 28222 Madrid, Spain
| | - Belén Sierra-Rodero
- Medical Oncology Department, Health Research Institute Puerta de Hierro–Segovia de Arana (IDIPHISA) & Puerta de Hierro Hospital, Manuel de Falla Street #1, 28222 Madrid, Spain; (R.L.-B.); (M.C.); (B.S.-R.)
- PhD Programme in Molecular Biosciences, Faculty of Medicine Doctoral School, Universidad Autónoma de Madrid, 28222 Madrid, Spain
| | - Mariano Provencio
- Medical Oncology Department, Health Research Institute Puerta de Hierro–Segovia de Arana (IDIPHISA) & Puerta de Hierro Hospital, Manuel de Falla Street #1, 28222 Madrid, Spain; (R.L.-B.); (M.C.); (B.S.-R.)
| |
Collapse
|
25
|
Yuen VWH, Wong CCL. Hypoxia-inducible factors and innate immunity in liver cancer. J Clin Invest 2021; 130:5052-5062. [PMID: 32750043 DOI: 10.1172/jci137553] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The liver has strong innate immunity to counteract pathogens from the gastrointestinal tract. During the development of liver cancer, which is typically driven by chronic inflammation, the composition and biological roles of the innate immune cells are extensively altered. Hypoxia is a common finding in all stages of liver cancer development. Hypoxia drives the stabilization of hypoxia-inducible factors (HIFs), which act as central regulators to dampen the innate immunity of liver cancer. HIF signaling in innate immune cells and liver cancer cells together favors the recruitment and maintenance of pro-tumorigenic immune cells and the inhibition of anti-tumorigenic immune cells, promoting immune evasion. HIFs represent attractive therapeutic targets to inhibit the formation of an immunosuppressive microenvironment and growth of liver cancer.
Collapse
Affiliation(s)
| | - Carmen Chak-Lui Wong
- Department of Pathology and.,State Key Laboratory of Liver Research, University of Hong Kong, Hong Kong, China
| |
Collapse
|
26
|
Kumar R, Singh AK, Starokadomskyy P, Luo W, Theiss AL, Burstein E, Venuprasad K. Cutting Edge: Hypoxia-Induced Ubc9 Promoter Hypermethylation Regulates IL-17 Expression in Ulcerative Colitis. THE JOURNAL OF IMMUNOLOGY 2021; 206:936-940. [PMID: 33504619 DOI: 10.4049/jimmunol.2000015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 12/28/2020] [Indexed: 12/24/2022]
Abstract
Dysregulated IL-17 expression is central to the pathogenesis of several inflammatory disorders, including ulcerative colitis. We have shown earlier that SUMOylation of ROR-γt, the transcription factor for IL-17, regulates colonic inflammation. In this study, we show that the expression of Ubc9, the E2 enzyme that targets ROR-γt for SUMOylation, is significantly reduced in the colonic mucosa of ulcerative colitis patients. Mechanistically, we demonstrate that hypoxia-inducible factor 1α (HIF-1α) binds to a CpG island within the Ubc9 gene promoter, resulting in its hypermethylation and reduced Ubc9 expression. CRISPR-Cas9-mediated inhibition of HIF-1α normalized Ubc9 and attenuated IL-17 expression in Th17 cells and reduced diseases severity in Rag1 -/- mice upon adoptive transfer. Collectively, our study reveals a novel epigenetic mechanism of regulation of ROR-γt that could be exploited in inflammatory diseases.
Collapse
Affiliation(s)
- Ritesh Kumar
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390.,Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Amir Kumar Singh
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390.,Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Petro Starokadomskyy
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Weibo Luo
- Department of Pathology and Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Arianne L Theiss
- Division of Gastroenterology and Hepatology, School of Medicine, University of Colorado, Aurora, CO 80045; and
| | - Ezra Burstein
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390.,Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - K Venuprasad
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390; .,Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| |
Collapse
|
27
|
Wang B, Zhao Q, Zhang Y, Liu Z, Zheng Z, Liu S, Meng L, Xin Y, Jiang X. Targeting hypoxia in the tumor microenvironment: a potential strategy to improve cancer immunotherapy. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:24. [PMID: 33422072 PMCID: PMC7796640 DOI: 10.1186/s13046-020-01820-7] [Citation(s) in RCA: 134] [Impact Index Per Article: 44.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 12/20/2020] [Indexed: 12/14/2022]
Abstract
With the success of immune checkpoint inhibitors (ICIs), significant progress has been made in the field of cancer immunotherapy. Despite the long-lasting outcomes in responders, the majority of patients with cancer still do not benefit from this revolutionary therapy. Increasing evidence suggests that one of the major barriers limiting the efficacy of immunotherapy seems to coalesce with the hypoxic tumor microenvironment (TME), which is an intrinsic property of all solid tumors. In addition to its impact on shaping tumor invasion and metastasis, the hypoxic TME plays an essential role in inducing immune suppression and resistance though fostering diverse changes in stromal cell biology. Therefore, targeting hypoxia may provide a means to enhance the efficacy of immunotherapy. In this review, the potential impact of hypoxia within the TME, in terms of key immune cell populations, and the contribution to immune suppression are discussed. In addition, we outline how hypoxia can be manipulated to tailor the immune response and provide a promising combinational therapeutic strategy to improve immunotherapy.
Collapse
Affiliation(s)
- Bin Wang
- Department of Radiation Oncology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China.,Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun, 130021, China.,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, China
| | - Qin Zhao
- Department of Radiation Oncology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China.,Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun, 130021, China.,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, China
| | - Yuyu Zhang
- Department of Radiation Oncology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China.,Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun, 130021, China.,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, China
| | - Zijing Liu
- Department of Radiation Oncology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China.,Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun, 130021, China.,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, China
| | - Zhuangzhuang Zheng
- Department of Radiation Oncology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China.,Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun, 130021, China.,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, China
| | - Shiyu Liu
- Department of Radiation Oncology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China.,Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun, 130021, China.,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, China
| | - Lingbin Meng
- Department of Hematology and Medical Oncology, Moffitt Cancer Center, Tampa, FL, 33612, USA
| | - Ying Xin
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, 126 Xinmin Street, Changchun, 130021, China.
| | - Xin Jiang
- Department of Radiation Oncology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China. .,Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun, 130021, China. .,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, China.
| |
Collapse
|
28
|
Abstract
Over the last few years, cancer immunotherapy experienced tremendous developments and it is nowadays considered a promising strategy against many types of cancer. However, the exclusion of lymphocytes from the tumor nest is a common phenomenon that limits the efficiency of immunotherapy in solid tumors. Despite several mechanisms proposed during the years to explain the immune excluded phenotype, at present, there is no integrated understanding about the role played by different models of immune exclusion in human cancers. Hypoxia is a hallmark of most solid tumors and, being a multifaceted and complex condition, shapes in a unique way the tumor microenvironment, affecting gene transcription and chromatin remodeling. In this review, we speculate about an upstream role for hypoxia as a common biological determinant of immune exclusion in solid tumors. We also discuss the current state of ex vivo and in vivo imaging of hypoxic determinants in relation to T cell distribution that could mechanisms of immune exclusion and discover functional-morphological tumor features that could support clinical monitoring.
Collapse
|
29
|
Characteristics of the Tumor Microenvironment That Influence Immune Cell Functions: Hypoxia, Oxidative Stress, Metabolic Alterations. Cancers (Basel) 2020; 12:cancers12123802. [PMID: 33348579 PMCID: PMC7765870 DOI: 10.3390/cancers12123802] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/06/2020] [Accepted: 12/10/2020] [Indexed: 12/20/2022] Open
Abstract
Immunotherapy (IMT) is now a core component of cancer treatment, however, many patients do not respond to these novel therapies. Investigating the resistance mechanisms behind this differential response is now a critical area of research. Immune-based therapies, particularly immune checkpoint inhibitors (ICI), rely on a robust infiltration of T-cells into the tumor microenvironment (TME) for an effective response. While early efforts relied on quantifying tumor infiltrating lymphocytes (TIL) in the TME, characterizing the functional quality and degree of TIL exhaustion correlates more strongly with ICI response. Even with sufficient TME infiltration, immune cells face a harsh metabolic environment that can significantly impair effector function. These tumor-mediated metabolic perturbations include hypoxia, oxidative stress, and metabolites of cellular energetics. Primarily through HIF-1-dependent processes, hypoxia invokes an immunosuppressive phenotype via altered molecular markers, immune cell trafficking, and angiogenesis. Additionally, oxidative stress can promote lipid peroxidation, ER stress, and Treg dysfunction, all associated with immune dysregulation. Finally, the metabolic byproducts of lipids, amino acids, glucose, and cellular energetics are associated with immunosuppression and ICI resistance. This review will explore these biochemical pathways linked to immune cell dysfunction in the TME and highlight potential adjunctive therapies to be used alongside current IMT.
Collapse
|
30
|
Jacobsen DP, Lekva T, Moe K, Fjeldstad HES, Johnsen GM, Sugulle M, Staff AC. Pregnancy and postpartum levels of circulating maternal sHLA-G in preeclampsia. J Reprod Immunol 2020; 143:103249. [PMID: 33254097 DOI: 10.1016/j.jri.2020.103249] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 11/09/2020] [Accepted: 11/12/2020] [Indexed: 12/14/2022]
Abstract
Preeclampsia is a leading cause of maternal and offspring mortality and morbidity, and predicts increased future cardiovascular disease risk. Placental dysfunction and immune system dysregulation are likely key pathophysiological factors. Soluble human leukocyte antigen G (sHLA-G) may dampen the specific immune response towards placental trophoblasts. Previous studies have shown low sHLA-G levels in preeclampsia, but postpartum, levels are unknown. Furthermore, the relationship between sHLA-G and sFlt-1 and PlGF, placental function markers, is unknown. We hypothesized that low maternal sHLA-G during pregnancy would be associated with placental dysfunction, including preeclampsia, gestational hypertension, and dysregulated sFlt-1 and PlGF, and that sHLA-G would remain decreased following preeclampsia. We included 316 pregnant women: 58 with early-onset preeclampsia (<34 weeks' gestation), 81 with late-onset preeclampsia (≥34 weeks' gestation), 25 with gestational hypertension, and 152 normotensive controls. Postpartum (1 or 3 years), we included 321 women: 29 with early-onset preeclampsia, 98 with late-onset preeclampsia, 57 with gestational hypertension, and 137 who were normotensive during their index pregnancies. In pregnancy, plasma sHLA-G was significantly lower both in the early- and late-onset preeclampsia groups compared to controls. In women with preeclampsia or gestational hypertension, sHLA-G was inversely correlated with serum sFlt-1. Postpartum, plasma sHLA-G levels were significantly higher in women who had had early-onset preeclampsia compared to controls. Our results support that sHLA-G may be important for placental function. Unexpectedly, sHLA-G was elevated up to 3 years after early-onset preeclampsia, suggesting an excessively activated immune system following this severe preeclampsia form, potentially contributing to future cardiovascular disease risk.
Collapse
Affiliation(s)
- Daniel P Jacobsen
- Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway.
| | - Tove Lekva
- Research Institute of Internal Medicine, Oslo University Hospital, Norway
| | - Kjartan Moe
- Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway; Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Norway; Department of Obstetrics and Gynaecology, Bærum Hospital, Vestre Viken HF, Norway
| | - Heidi E S Fjeldstad
- Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway; Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Norway
| | - Guro Mørk Johnsen
- Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway
| | - Meryam Sugulle
- Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway; Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Norway
| | - Anne Cathrine Staff
- Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway; Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Norway
| |
Collapse
|
31
|
Rashidi S, Farhadi L, Ghasemi F, Sheikhesmaeili F, Mohammadi A. The potential role of HLA-G in the pathogenesis of HBV infection: Immunosuppressive or immunoprotective? INFECTION GENETICS AND EVOLUTION 2020; 85:104580. [PMID: 33022425 DOI: 10.1016/j.meegid.2020.104580] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 09/20/2020] [Accepted: 10/01/2020] [Indexed: 02/07/2023]
Abstract
The non-classical human leukocyte antigens (HLA)-G could be generally considered as a potent tolerogenic molecule, which modulates immune responses. HLA-G due to the immunosuppressive properties may play an important role in the pathogenesis of infections related to the liver. HLA-G may display two distinct activities in the pathological conditions so that it could be protective in the autoimmune and inflammatory diseases or could be suppressive of the immune system in the infections or cancers. HLA-G might be used as a novel therapeutic target for liver diseases in the future. Indeed, new therapeutic agents targeting HLA-G expression or antibodies which block HLA-G activity are being developed and tested. However, further consideration of the HLA-G function in liver disease is required. This review aims to summarize the role of HLA-G in the liver of patients with HBV infection.
Collapse
Affiliation(s)
- Saadyeh Rashidi
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Leila Farhadi
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Faezeh Ghasemi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Farshad Sheikhesmaeili
- Liver and Digestive Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Asadollah Mohammadi
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| |
Collapse
|
32
|
de Carvalho GTC, da Silva-Martins WC, de Magalhães KCSF, Nunes CB, Soares AN, Tafuri LSDA, Simões RT. Recurrence/Regrowth in Grade I Meningioma: How to Predict? Front Oncol 2020; 10:1144. [PMID: 32903787 PMCID: PMC7438949 DOI: 10.3389/fonc.2020.01144] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 06/08/2020] [Indexed: 11/22/2022] Open
Abstract
The HLA-G and HLA-E molecules, Ki67, progesterone (PR), estrogen (ER) and androgen receptors (AR), p53, COX-2, and HER2 were studied to assess whether the biological behavior of grade I meningiomas is related to their expression. Tissue samples from 96 patients with grade I intracranial meningiomas were analyzed by immunohistochemistry on tissue microarray blocks (TMA) using antibodies specific for HLA-G, HLA-E, Ki67, PR, ER, AR, p53, COX-2, and HER2. Meningiomas were classified as small (≤2 cm, 1.0%), medium (>2 and ≤4 cm, 32.3%), and large (>4 cm, 66.7%). Tumor size was not related to recurrence/regrowth (p = 0.486), but was significantly correlated with peritumoral edema (p = 0.031) and intratumoral calcifications (p = 0.018). Recurrent meningiomas were observed in 14.6% of cases. Immunostaining for each marker was: HLA-G 100%; HLA-E 95.6%; PR 62%; ER 2.1%; AR 6.5%; p53 92.6%; COX-2 100%; HER2 0%; Ki67, mean 2.61 ± 2.29%, median 2.1%. Primary and recurrent meningiomas showed no significant relation with HLA-E and hormone receptors (p > 0.05), except for Ki67, where a higher median was observed in recurrent tumors than in primary (p = 0.014). The larger the tumor, the more severe the peritumoral edema, and the greater the presence of calcifications. Ki67 appears to be a good biomarker of recurrence/regrowth in grade I meningiomas.
Collapse
Affiliation(s)
- Gervásio Teles Cardoso de Carvalho
- Laboratory of Molecular Biology and Biomarkers, Santa Casa de Belo Horizonte Ensino e Pesquisa - EP/SCBH, Belo Horizonte, Brazil.,Department of Neurosurgery, Hospital Santa Casa de Belo Horizonte, Belo Horizonte, Brazil.,Faculdade de Ciências Médicas de Minas Gerais (FCMMG), Belo Horizonte, Brazil
| | | | | | - Cristiana Buzelin Nunes
- Departamento de Anatomia Patológica e Medicina Legal, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Centro Universitário de Belo Horizonte - UniBH, Belo Horizonte, Brazil
| | - Aleida Nazareth Soares
- Laboratory of Molecular Biology and Biomarkers, Santa Casa de Belo Horizonte Ensino e Pesquisa - EP/SCBH, Belo Horizonte, Brazil
| | - Luciene Simões de Assis Tafuri
- Laboratory of Molecular Biology and Biomarkers, Santa Casa de Belo Horizonte Ensino e Pesquisa - EP/SCBH, Belo Horizonte, Brazil.,Departamento de Fisiologia e Patologia, Centro de Ciências da Saúde, Universidade Federal da Paraíba, DFP/CCS/UFPB, João Pessoa, Brazil
| | - Renata Toscano Simões
- Laboratory of Molecular Biology and Biomarkers, Santa Casa de Belo Horizonte Ensino e Pesquisa - EP/SCBH, Belo Horizonte, Brazil
| |
Collapse
|
33
|
Liu Z, Zhang R, Sun Z, Yao J, Yao P, Chen X, Wang X, Gao M, Wan J, Du Y, Zhao S. Identification of hub genes and small-molecule compounds in medulloblastoma by integrated bioinformatic analyses. PeerJ 2020; 8:e8670. [PMID: 32328342 PMCID: PMC7164431 DOI: 10.7717/peerj.8670] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 01/30/2020] [Indexed: 01/03/2023] Open
Abstract
Background Medulloblastoma (MB) is the most common intracranial malignant tumor in children. The genes and pathways involved in the pathogenesis of MB are relatively unknown. We aimed to identify potential biomarkers and small-molecule drugs for MB. Methods Gene expression profile data sets were obtained from the Gene Expression Omnibus (GEO) database and the differentially expressed genes (DEGs) were identified using the Limma package in R. Functional annotation, and cell signaling pathway analysis of DEGs was carried out using DAVID and Kobas. A protein-protein interaction network was generated using STRING. Potential small-molecule drugs were identified using CMap. Result We identified 104 DEGs (29 upregulated; 75 downregulated). Gene ontology analysis showed enrichment in the mitotic cell cycle, cell cycle, spindle, and DNA binding. Cell signaling pathway analysis identified cell cycle, HIF-1 signaling pathway, and phospholipase D signaling pathway as key pathways. SYN1, CNTN2, FAIM2, MT3, and SH3GL2 were the prominent hub genes and their expression level were verified by RT-qPCR. Vorinostat, resveratrol, trichostatin A, pyrvinium, and prochlorperazine were identified as potential drugs for MB. The five hub genes may be targets for diagnosis and treatment of MB, and the small-molecule compounds are promising drugs for effective treatment of MB. Conclusion In this study we obtained five hub genes of MB, SYN1, CNTN2, FAIM2, MT3, and SH3GL2 were confirmed as hub genes. Meanwhile, Vorinostat, resveratrol, trichostatin A, pyrvinium, and prochlorperazine were identified as potential drugs for MB.
Collapse
Affiliation(s)
- Zhendong Liu
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Ruotian Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Zhenying Sun
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Jiawei Yao
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Penglei Yao
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Xin Chen
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Xinzhuang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Ming Gao
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Jinzhao Wan
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Yiming Du
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Shiguang Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| |
Collapse
|
34
|
Ziliotto M, Rodrigues RM, Chies JAB. Controlled hypobaric hypoxia increases immunological tolerance by modifying HLA-G expression, a potential therapy to inflammatory diseases. Med Hypotheses 2020; 140:109664. [PMID: 32155542 DOI: 10.1016/j.mehy.2020.109664] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/26/2020] [Accepted: 03/03/2020] [Indexed: 12/22/2022]
Abstract
One of the most striking characteristics of human beings is the incredible capacity to adapt to different environments. This capacity allowed humans to spread all over our planet, occupying habitats as diverse as deserts, tropical forests or tundra regions. Interactions with the environment, climate, food and water availability shaped our evolution and define our survival. Essential to human life, oxygen availability also controls human dispersion and adaptation. For example, low oxygen availability can lead to physiological adaptations in populations living in highlands. Moreover, the consequences of differential oxygen availability (or even exposure to hypoxia) are evident in process as fine-tuned controlled as gene regulation. Physiological responses to fluctuations in oxygen availability are crucial already from the early days of life, since the human fetal environment is characterized by hypoxia. Hypoxia-Inducible Factors (HIFs) act as major regulators of pathways involved in glycolysis, erythropoiesis, angiogenesis, cell proliferation and stem cells function. Here we explore the physiological consequences of hypoxia in the human organism. In this sense, and considering the existence of HIF sequences in promoter regions of genes important to immune regulation, we hypothesize that exposure to induced hypoxia through the use of hypobaric chambers can be used as a complementary therapy to control chronic inflammation in several diseases characterized by systemic inflammatory conditions. Among these inflammatory conditions we highlight autoimmune diseases and chronic inflammation in HIV infected individuals under antiretroviral treatment. Several experiments, including arthritis animal models, the evaluation of athletes that already use hypobaric chambers to induce erythropoiesis, and the potential consequences of hypoxia as an immunotolerogenic inducer in the HIV infection context are approached and discussed here.
Collapse
Affiliation(s)
- Marina Ziliotto
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Raul Marques Rodrigues
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - José Artur Bogo Chies
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Rio Grande do Sul, Brazil.
| |
Collapse
|
35
|
Ajith A, Portik-Dobos V, Horuzsko DD, Kapoor R, Mulloy LL, Horuzsko A. HLA-G and humanized mouse models as a novel therapeutic approach in transplantation. Hum Immunol 2020; 81:178-185. [PMID: 32093884 DOI: 10.1016/j.humimm.2020.02.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 02/15/2020] [Accepted: 02/17/2020] [Indexed: 01/12/2023]
Abstract
HLA-G is a nonclassical MHC-Class I molecule whose expression, along the feto-maternal barrier contributes towards tolerance of the semiallogeneic fetus during pregnancy. In light of its inhibitory properties, recent research has established HLA-G involvement in mechanisms responsible for directing allogeneic immune responses towards tolerance during allogeneic situations such as organ transplantation. Here, we critically review the data supporting the tolerogenic role of HLA-G in organ transplantation, the various factors influencing its expression, and the introduction of novel humanized mouse models that are one of the best approaches to assess the utility of HLA-G as a therapeutic tool in organ transplantation.
Collapse
Affiliation(s)
- Ashwin Ajith
- Georgia Cancer Canter, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Vera Portik-Dobos
- Georgia Cancer Canter, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Daniel D Horuzsko
- Philadelphia College of Osteopathic Medicine South Georgia, Moultrie, GA, USA
| | - Rajan Kapoor
- Division of Nephrology, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Laura L Mulloy
- Division of Nephrology, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Anatolij Horuzsko
- Georgia Cancer Canter, Medical College of Georgia, Augusta University, Augusta, GA, USA.
| |
Collapse
|
36
|
Mohapatra SR, Sadik A, Tykocinski LO, Dietze J, Poschet G, Heiland I, Opitz CA. Hypoxia Inducible Factor 1α Inhibits the Expression of Immunosuppressive Tryptophan-2,3-Dioxygenase in Glioblastoma. Front Immunol 2019; 10:2762. [PMID: 31866995 PMCID: PMC6905408 DOI: 10.3389/fimmu.2019.02762] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 11/12/2019] [Indexed: 12/12/2022] Open
Abstract
Abnormal circulation in solid tumors results in hypoxia, which modulates both tumor intrinsic malignant properties as well as anti-tumor immune responses. Given the importance of hypoxia in glioblastoma (GBM) biology and particularly in shaping anti-tumor immunity, we analyzed which immunomodulatory genes are differentially regulated in response to hypoxia in GBM cells. Gene expression analyses identified the immunosuppressive enzyme tryptophan-2,3-dioxygenase (TDO2) as the second most downregulated gene in GBM cells cultured under hypoxic conditions. TDO2 catalyses the oxidation of tryptophan to N-formyl kynurenine, which is the first and rate-limiting step of Trp degradation along the kynurenine pathway (KP). In multiple GBM cell lines hypoxia reduced TDO2 expression both at mRNA and protein levels. The downregulation of TDO2 through hypoxia was reversible as re-oxygenation rescued TDO2 expression. Computational modeling of tryptophan metabolism predicted reduced flux through the KP and lower intracellular concentrations of kynurenine and its downstream metabolite 3-hydroxyanthranilic acid under hypoxia. Metabolic measurements confirmed the predicted changes, thus demonstrating the ability of the mathematical model to infer intracellular tryptophan metabolite concentrations. Moreover, we identified hypoxia inducible factor 1α (HIF1α) to regulate TDO2 expression under hypoxic conditions, as the HIF1α-stabilizing agents dimethyloxalylglycine (DMOG) and cobalt chloride reduced TDO2 expression. Knockdown of HIF1α restored the expression of TDO2 upon cobalt chloride treatment, confirming that HIF1α controls TDO2 expression. To investigate the immunoregulatory effects of this novel mechanism of TDO2 regulation, we co-cultured isolated T cells with TDO2-expressing GBM cells under normoxic and hypoxic conditions. Under normoxia TDO2-expressing GBM cells suppressed T cell proliferation, while hypoxia restored the proliferation of the T cells, likely due to the reduction in kynurenine levels produced by the GBM cells. Taken together, our data suggest that the regulation of TDO2 expression by HIF1α may be involved in modulating anti-tumor immunity in GBM.
Collapse
Affiliation(s)
- Soumya R Mohapatra
- DKTK Brain Cancer Metabolism Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ahmed Sadik
- DKTK Brain Cancer Metabolism Group, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Bioscience, Heidelberg University, Heidelberg, Germany
| | - Lars-Oliver Tykocinski
- Division of Rheumatology, Department of Medicine V, University Hospital of Heidelberg, Heidelberg, Germany
| | - Jørn Dietze
- Department of Arctic and Marine Biology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Gernot Poschet
- Centre for Organismal Studies (COS), University of Heidelberg, Heidelberg, Germany
| | - Ines Heiland
- Department of Arctic and Marine Biology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Christiane A Opitz
- DKTK Brain Cancer Metabolism Group, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Neurology Clinic and National Center for Tumor Diseases, University Hospital of Heidelberg, Heidelberg, Germany
| |
Collapse
|
37
|
Melo‐Lima BL, Poras I, Passos GA, Carosella ED, Donadi EA, Moreau P. The Autoimmune Regulator (Aire) transactivates HLA-G gene expression in thymic epithelial cells. Immunology 2019; 158:121-135. [PMID: 31322727 PMCID: PMC6742766 DOI: 10.1111/imm.13099] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 07/11/2019] [Indexed: 12/12/2022] Open
Abstract
The Autoimmune Regulator (Aire) protein coordinates the negative selection of developing thymocytes by inducing the expression of hundreds of tissue-specific antigens within the thymic medulla, which is also a primary site of the expression of the immune checkpoint HLA-G molecule. Considering the immunomodulatory properties of Aire and HLA-G, and considering that the role of the constitutive thymus expression of HLA-G has not been elucidated, we studied the effect of AIRE cDNA transfection on HLA-G expression in 4D6 thymic cells and in the HLA-G-positive JEG-3 choriocarcinoma cells. Aire promoted the transactivation of HLA-G gene by increasing the overall transcription, inducing the transcription of at least G1 and G2/G4 isoforms, and incrementing the occurrence and distribution of intracellular HLA-G protein solely in 4D6 thymic cells. Luciferase-based assays and chromatin immunoprecipitation experiments performed in 4D6 cells revealed that Aire targeted at least two regions within the 5'-untranslated regulatory region (5'-URR) extending 1·4 kb from the first ATG initiation codon. The interaction occurs independently of three putative Aire-binding sites. These results indicate that the Aire-induced upregulation of HLA-G in thymic cells is likely to act through the interaction of Aire with specific HLA-G 5'-URR DNA-binding factors. Such a multimeric transcriptional complex might operate in the thymus during the process of promiscuous gene expression.
Collapse
Affiliation(s)
- Breno Luiz Melo‐Lima
- Direction de la Recherche FondamentaleInstitut de Biologie François JacobService de Recherches en Hémato‐ImmunologieHôpital Saint‐LouisCommissariat à l'Energie Atomique et aux Energies AlternativesParisFrance
- Institut de Recherche Saint‐LouisUniversité de ParisUMR976 HIPIHôpital Saint‐LouisUniversité Paris‐DiderotParisFrance
- Division of Clinical ImmunologyDepartment of MedicineRibeirao Preto Medical SchoolUniversity of São PauloRibeirão PretoSão PauloBrazil
| | - Isabelle Poras
- Direction de la Recherche FondamentaleInstitut de Biologie François JacobService de Recherches en Hémato‐ImmunologieHôpital Saint‐LouisCommissariat à l'Energie Atomique et aux Energies AlternativesParisFrance
- Institut de Recherche Saint‐LouisUniversité de ParisUMR976 HIPIHôpital Saint‐LouisUniversité Paris‐DiderotParisFrance
| | - Geraldo Aleixo Passos
- Molecular Immunogenetics GroupDepartment of GeneticsRibeirão Preto Medical SchoolUniversity of São PauloRibeirão PretoSão PauloBrazil
| | - Edgardo D. Carosella
- Direction de la Recherche FondamentaleInstitut de Biologie François JacobService de Recherches en Hémato‐ImmunologieHôpital Saint‐LouisCommissariat à l'Energie Atomique et aux Energies AlternativesParisFrance
- Institut de Recherche Saint‐LouisUniversité de ParisUMR976 HIPIHôpital Saint‐LouisUniversité Paris‐DiderotParisFrance
| | - Eduardo Antonio Donadi
- Division of Clinical ImmunologyDepartment of MedicineRibeirao Preto Medical SchoolUniversity of São PauloRibeirão PretoSão PauloBrazil
| | - Philippe Moreau
- Direction de la Recherche FondamentaleInstitut de Biologie François JacobService de Recherches en Hémato‐ImmunologieHôpital Saint‐LouisCommissariat à l'Energie Atomique et aux Energies AlternativesParisFrance
- Institut de Recherche Saint‐LouisUniversité de ParisUMR976 HIPIHôpital Saint‐LouisUniversité Paris‐DiderotParisFrance
| |
Collapse
|
38
|
Cammarata FP, Torrisi F, Forte GI, Minafra L, Bravatà V, Pisciotta P, Savoca G, Calvaruso M, Petringa G, Cirrone GAP, Fallacara AL, Maccari L, Botta M, Schenone S, Parenti R, Cuttone G, Russo G. Proton Therapy and Src Family Kinase Inhibitor Combined Treatments on U87 Human Glioblastoma Multiforme Cell Line. Int J Mol Sci 2019; 20:E4745. [PMID: 31554327 PMCID: PMC6801826 DOI: 10.3390/ijms20194745] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 09/17/2019] [Accepted: 09/24/2019] [Indexed: 12/22/2022] Open
Abstract
Glioblastoma Multiforme (GBM) is the most common of malignant gliomas in adults with an exiguous life expectancy. Standard treatments are not curative and the resistance to both chemotherapy and conventional radiotherapy (RT) plans is the main cause of GBM care failures. Proton therapy (PT) shows a ballistic precision and a higher dose conformity than conventional RT. In this study we investigated the radiosensitive effects of a new targeted compound, SRC inhibitor, named Si306, in combination with PT on the U87 glioblastoma cell line. Clonogenic survival assay, dose modifying factor calculation and linear-quadratic model were performed to evaluate radiosensitizing effects mediated by combination of the Si306 with PT. Gene expression profiling by microarray was also conducted after PT treatments alone or combined, to identify gene signatures as biomarkers of response to treatments. Our results indicate that the Si306 compound exhibits a radiosensitizing action on the U87 cells causing a synergic cytotoxic effect with PT. In addition, microarray data confirm the SRC role as the main Si306 target and highlights new genes modulated by the combined action of Si306 and PT. We suggest, the Si306 as a new candidate to treat GBM in combination with PT, overcoming resistance to conventional treatments.
Collapse
Affiliation(s)
- Francesco P Cammarata
- Institute of Molecular Bioimaging and Physiology, National Research Council, IBFM-CNR, 90015 Cefalù, Italy.
- National Institute for Nuclear Physics, Laboratori Nazionali del Sud, INFN-LNS, 95123 Catania, Italy.
| | - Filippo Torrisi
- National Institute for Nuclear Physics, Laboratori Nazionali del Sud, INFN-LNS, 95123 Catania, Italy.
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, 95123 Catania, Italy.
| | - Giusi I Forte
- Institute of Molecular Bioimaging and Physiology, National Research Council, IBFM-CNR, 90015 Cefalù, Italy.
- National Institute for Nuclear Physics, Laboratori Nazionali del Sud, INFN-LNS, 95123 Catania, Italy.
| | - Luigi Minafra
- Institute of Molecular Bioimaging and Physiology, National Research Council, IBFM-CNR, 90015 Cefalù, Italy.
- National Institute for Nuclear Physics, Laboratori Nazionali del Sud, INFN-LNS, 95123 Catania, Italy.
| | - Valentina Bravatà
- Institute of Molecular Bioimaging and Physiology, National Research Council, IBFM-CNR, 90015 Cefalù, Italy.
- National Institute for Nuclear Physics, Laboratori Nazionali del Sud, INFN-LNS, 95123 Catania, Italy.
| | - Pietro Pisciotta
- National Institute for Nuclear Physics, Laboratori Nazionali del Sud, INFN-LNS, 95123 Catania, Italy.
- Departments of Physics and Astronomy, University of Catania, 95123 Catania, Italy.
| | - Gaetano Savoca
- Institute of Molecular Bioimaging and Physiology, National Research Council, IBFM-CNR, 90015 Cefalù, Italy.
| | - Marco Calvaruso
- Institute of Molecular Bioimaging and Physiology, National Research Council, IBFM-CNR, 90015 Cefalù, Italy.
- National Institute for Nuclear Physics, Laboratori Nazionali del Sud, INFN-LNS, 95123 Catania, Italy.
| | - Giada Petringa
- National Institute for Nuclear Physics, Laboratori Nazionali del Sud, INFN-LNS, 95123 Catania, Italy.
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, 95123 Catania, Italy.
| | - Giuseppe A P Cirrone
- National Institute for Nuclear Physics, Laboratori Nazionali del Sud, INFN-LNS, 95123 Catania, Italy.
| | - Anna L Fallacara
- Lead Discovery Siena s.r.l. (LDS), 53100 Siena, Italy.
- Department of Biotechnology, Chemistry and Pharmacy, Università degli Studi di Siena, 53100 Siena, Italy.
| | - Laura Maccari
- Lead Discovery Siena s.r.l. (LDS), 53100 Siena, Italy.
| | - Maurizio Botta
- Lead Discovery Siena s.r.l. (LDS), 53100 Siena, Italy.
- Department of Biotechnology, Chemistry and Pharmacy, Università degli Studi di Siena, 53100 Siena, Italy.
| | - Silvia Schenone
- Department of Pharmacy, Università degli Studi di Genova, 16126 Genova, Italy.
| | - Rosalba Parenti
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, 95123 Catania, Italy.
| | - Giacomo Cuttone
- National Institute for Nuclear Physics, Laboratori Nazionali del Sud, INFN-LNS, 95123 Catania, Italy.
| | - Giorgio Russo
- Institute of Molecular Bioimaging and Physiology, National Research Council, IBFM-CNR, 90015 Cefalù, Italy.
- National Institute for Nuclear Physics, Laboratori Nazionali del Sud, INFN-LNS, 95123 Catania, Italy.
| |
Collapse
|
39
|
Improving Cancer Immunotherapy by Targeting the Hypoxic Tumor Microenvironment: New Opportunities and Challenges. Cells 2019; 8:cells8091083. [PMID: 31540045 PMCID: PMC6770817 DOI: 10.3390/cells8091083] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 08/30/2019] [Accepted: 09/12/2019] [Indexed: 02/06/2023] Open
Abstract
Initially believed to be a disease of deregulated cellular and genetic expression, cancer is now also considered a disease of the tumor microenvironment. Over the past two decades, significant and rapid progress has been made to understand the complexity of the tumor microenvironment and its contribution to shaping the response to various anti-cancer therapies, including immunotherapy. Nevertheless, it has become clear that the tumor microenvironment is one of the main hallmarks of cancer. Therefore, a major challenge is to identify key druggable factors and pathways in the tumor microenvironment that can be manipulated to improve the efficacy of current cancer therapies. Among the different tumor microenvironmental factors, this review will focus on hypoxia as a key process that evolved in the tumor microenvironment. We will briefly describe our current understanding of the molecular mechanisms by which hypoxia negatively affects tumor immunity and shapes the anti-tumor immune response. We believe that such understanding will provide insight into the therapeutic value of targeting hypoxia and assist in the design of innovative combination approaches to improve the efficacy of current cancer therapies, including immunotherapy.
Collapse
|
40
|
Veiga-Castelli L, de Oliveira ML, Pereira A, Debortoli G, Marcorin L, Fracasso N, Silva G, Souza A, Massaro J, Simões AL, Sabbagh A, Cardili R, Donadi E, Castelli E, Mendes-Junior C. HLA-G Polymorphisms Are Associated with Non-segmental Vitiligo among Brazilians. Biomolecules 2019; 9:biom9090463. [PMID: 31505868 PMCID: PMC6769860 DOI: 10.3390/biom9090463] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 07/15/2019] [Accepted: 07/18/2019] [Indexed: 12/31/2022] Open
Abstract
(1) Background: Vitiligo is characterized by white patches on the skin caused by loss of melanocyte activity or the absence of these cells. The available treatments minimize the symptoms by retarding the process of skin depigmentation or re-pigmenting the affected regions. New studies are required for a better comprehension of the mechanisms that trigger the disease and for the development of more efficient treatments. Studies have suggested an autoimmune feature for vitiligo, based on the occurrence of other autoimmune diseases in vitiligo patients and their relatives, and on the involvement of genes related to the immune response. (2) Methods: We evaluated, by massive parallel sequencing, polymorphisms of the HLA-G gene in vitiligo patients and control samples, to verify if variants of this gene could influence the susceptibility to vitiligo. (3) Results: We detected an association with non-segmental vitiligo regarding the haplotype Distal-010101a/G*01:01:01:01/UTR-1, adjusting for population stratification by using ancestry-informative markers (AIMs). (4) Conclusions: It remains unclear whether the HLA-G variants associated with vitiligo were detected because of the high linkage disequilibrium (LD) with HLA-A*02, or if the HLA-A variants previously reported as associated with vitiligo were detected because of the high LD with HLA-G*01:01:01:01/UTR-1, or if both genes jointly contribute to vitiligo susceptibility.
Collapse
Affiliation(s)
- Luciana Veiga-Castelli
- Departamento de Genética, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto-SP 14049-900, Brazil; (M.L.d.O.); (A.P.); (G.D.); (L.M.); (N.F.); (A.L.S.)
- Correspondence: ; Tel.: +55-16-3315-0417; Fax: +55-16-3315-4838
| | - Maria Luiza de Oliveira
- Departamento de Genética, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto-SP 14049-900, Brazil; (M.L.d.O.); (A.P.); (G.D.); (L.M.); (N.F.); (A.L.S.)
| | - Alison Pereira
- Departamento de Genética, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto-SP 14049-900, Brazil; (M.L.d.O.); (A.P.); (G.D.); (L.M.); (N.F.); (A.L.S.)
| | - Guilherme Debortoli
- Departamento de Genética, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto-SP 14049-900, Brazil; (M.L.d.O.); (A.P.); (G.D.); (L.M.); (N.F.); (A.L.S.)
| | - Letícia Marcorin
- Departamento de Genética, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto-SP 14049-900, Brazil; (M.L.d.O.); (A.P.); (G.D.); (L.M.); (N.F.); (A.L.S.)
| | - Nádia Fracasso
- Departamento de Genética, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto-SP 14049-900, Brazil; (M.L.d.O.); (A.P.); (G.D.); (L.M.); (N.F.); (A.L.S.)
| | - Guilherme Silva
- Departamento de Química, Laboratório de Pesquisas Forenses e Genômicas, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto-SP 14040-901, Brazil; (G.S.); (C.M.-J.)
| | - Andreia Souza
- Molecular Genetics and Bioinformatics Laboratory, Experimental Research Unit (UNIPEX), School of Medicine, São Paulo State University (UNESP), Botucatu, State of São Paulo 18618-687, Brazil; (A.S.); (E.C.)
| | - Juliana Massaro
- Departamento de Clínica Médica, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto-SP 14049-900, Brazil; (J.M.); (R.C.); (E.D.)
| | - Aguinaldo Luiz Simões
- Departamento de Genética, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto-SP 14049-900, Brazil; (M.L.d.O.); (A.P.); (G.D.); (L.M.); (N.F.); (A.L.S.)
| | - Audrey Sabbagh
- UMR 216 MERIT IRD, Faculté de Pharmacie de Paris, Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France;
| | - Renata Cardili
- Departamento de Clínica Médica, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto-SP 14049-900, Brazil; (J.M.); (R.C.); (E.D.)
| | - Eduardo Donadi
- Departamento de Clínica Médica, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto-SP 14049-900, Brazil; (J.M.); (R.C.); (E.D.)
| | - Erick Castelli
- Molecular Genetics and Bioinformatics Laboratory, Experimental Research Unit (UNIPEX), School of Medicine, São Paulo State University (UNESP), Botucatu, State of São Paulo 18618-687, Brazil; (A.S.); (E.C.)
| | - Celso Mendes-Junior
- Departamento de Química, Laboratório de Pesquisas Forenses e Genômicas, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto-SP 14040-901, Brazil; (G.S.); (C.M.-J.)
| |
Collapse
|
41
|
Riera-Domingo C, Audigé A, Granja S, Cheng WC, Ho PC, Baltazar F, Stockmann C, Mazzone M. Immunity, Hypoxia, and Metabolism-the Ménage à Trois of Cancer: Implications for Immunotherapy. Physiol Rev 2019; 100:1-102. [PMID: 31414610 DOI: 10.1152/physrev.00018.2019] [Citation(s) in RCA: 184] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
It is generally accepted that metabolism is able to shape the immune response. Only recently we are gaining awareness that the metabolic crosstalk between different tumor compartments strongly contributes to the harsh tumor microenvironment (TME) and ultimately impairs immune cell fitness and effector functions. The major aims of this review are to provide an overview on the immune system in cancer; to position oxygen shortage and metabolic competition as the ground of a restrictive TME and as important players in the anti-tumor immune response; to define how immunotherapies affect hypoxia/oxygen delivery and the metabolic landscape of the tumor; and vice versa, how oxygen and metabolites within the TME impinge on the success of immunotherapies. By analyzing preclinical and clinical endeavors, we will discuss how a metabolic characterization of the TME can identify novel targets and signatures that could be exploited in combination with standard immunotherapies and can help to predict the benefit of new and traditional immunotherapeutic drugs.
Collapse
Affiliation(s)
- Carla Riera-Domingo
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium; Institute of Anatomy, University of Zurich, Zurich, Switzerland; Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal; Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland; and Ludwig Cancer Research Institute, Epalinges, Switzerland
| | - Annette Audigé
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium; Institute of Anatomy, University of Zurich, Zurich, Switzerland; Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal; Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland; and Ludwig Cancer Research Institute, Epalinges, Switzerland
| | - Sara Granja
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium; Institute of Anatomy, University of Zurich, Zurich, Switzerland; Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal; Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland; and Ludwig Cancer Research Institute, Epalinges, Switzerland
| | - Wan-Chen Cheng
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium; Institute of Anatomy, University of Zurich, Zurich, Switzerland; Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal; Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland; and Ludwig Cancer Research Institute, Epalinges, Switzerland
| | - Ping-Chih Ho
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium; Institute of Anatomy, University of Zurich, Zurich, Switzerland; Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal; Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland; and Ludwig Cancer Research Institute, Epalinges, Switzerland
| | - Fátima Baltazar
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium; Institute of Anatomy, University of Zurich, Zurich, Switzerland; Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal; Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland; and Ludwig Cancer Research Institute, Epalinges, Switzerland
| | - Christian Stockmann
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium; Institute of Anatomy, University of Zurich, Zurich, Switzerland; Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal; Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland; and Ludwig Cancer Research Institute, Epalinges, Switzerland
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium; Institute of Anatomy, University of Zurich, Zurich, Switzerland; Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal; Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland; and Ludwig Cancer Research Institute, Epalinges, Switzerland
| |
Collapse
|
42
|
Lequeux A, Noman MZ, Xiao M, Sauvage D, Van Moer K, Viry E, Bocci I, Hasmim M, Bosseler M, Berchem G, Janji B. Impact of hypoxic tumor microenvironment and tumor cell plasticity on the expression of immune checkpoints. Cancer Lett 2019; 458:13-20. [PMID: 31136782 DOI: 10.1016/j.canlet.2019.05.021] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 05/10/2019] [Accepted: 05/14/2019] [Indexed: 01/27/2023]
Abstract
Compared to traditional therapies, such as surgery, radio-chemotherapy, or targeted approaches, immunotherapies based on immune checkpoint blockers (ICBs) have revolutionized the treatment of cancer. Although ICBs have yielded long-lasting results and have improved patient survival, this success has been seriously challenged by clinical observations showing that only a small fraction of patients benefit from this revolutionary therapy and no benefit has been found in patients with highly aggressive tumors. Efforts are currently ongoing to identify factors that predict the response to ICB. Among the different predictive markers established so far, the expression levels of immune checkpoint genes have proven to be important biomarkers for informing treatment choices. Therefore, understanding the mechanisms involved in the regulation of immune checkpoints is a key element that will facilitate novel combination approaches and optimize patient outcome. In this review, we discuss the impact of hypoxia and tumor cell plasticity on immune checkpoint gene expression and provide insight into the therapeutic value of the EMT signature and the rationale for novel combination approaches to improve ICB therapy and maximize the benefits for patients with cancer.
Collapse
Affiliation(s)
- Audrey Lequeux
- Laboratory of Experimental Cancer Research, Tumor Microenvironment Group, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg City, Luxembourg
| | - Muhammad Zaeem Noman
- Laboratory of Experimental Cancer Research, Tumor Microenvironment Group, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg City, Luxembourg
| | - Malina Xiao
- Laboratory of Experimental Cancer Research, Tumor Microenvironment Group, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg City, Luxembourg
| | - Delphine Sauvage
- Laboratory of Experimental Cancer Research, Tumor Microenvironment Group, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg City, Luxembourg
| | - Kris Van Moer
- Laboratory of Experimental Cancer Research, Tumor Microenvironment Group, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg City, Luxembourg
| | - Elodie Viry
- Laboratory of Experimental Cancer Research, Tumor Microenvironment Group, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg City, Luxembourg
| | - Irene Bocci
- Laboratory of Experimental Cancer Research, Tumor Microenvironment Group, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg City, Luxembourg
| | - Meriem Hasmim
- Laboratory of Experimental Cancer Research, Tumor Microenvironment Group, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg City, Luxembourg
| | - Manon Bosseler
- Laboratory of Experimental Cancer Research, Tumor Microenvironment Group, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg City, Luxembourg
| | - Guy Berchem
- Laboratory of Experimental Cancer Research, Tumor Microenvironment Group, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg City, Luxembourg; Centre Hospitalier du Luxembourg, Department of Hemato-Oncology, Luxembourg City, Luxembourg
| | - Bassam Janji
- Laboratory of Experimental Cancer Research, Tumor Microenvironment Group, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg City, Luxembourg.
| |
Collapse
|
43
|
Eckert F, Zwirner K, Boeke S, Thorwarth D, Zips D, Huber SM. Rationale for Combining Radiotherapy and Immune Checkpoint Inhibition for Patients With Hypoxic Tumors. Front Immunol 2019; 10:407. [PMID: 30930892 PMCID: PMC6423917 DOI: 10.3389/fimmu.2019.00407] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 02/15/2019] [Indexed: 12/19/2022] Open
Abstract
In order to compensate for the increased oxygen consumption in growing tumors, tumors need angiogenesis and vasculogenesis to increase the supply. Insufficiency in this process or in the microcirculation leads to hypoxic tumor areas with a significantly reduced pO2, which in turn leads to alterations in the biology of cancer cells as well as in the tumor microenvironment. Cancer cells develop more aggressive phenotypes, stem cell features and are more prone to metastasis formation and migration. In addition, intratumoral hypoxia confers therapy resistance, specifically radioresistance. Reactive oxygen species are crucial in fixing DNA breaks after ionizing radiation. Thus, hypoxic tumor cells show a two- to threefold increase in radioresistance. The microenvironment is enriched with chemokines (e.g., SDF-1) and growth factors (e.g., TGFβ) additionally reducing radiosensitivity. During recent years hypoxia has also been identified as a major factor for immune suppression in the tumor microenvironment. Hypoxic tumors show increased numbers of myeloid derived suppressor cells (MDSCs) as well as regulatory T cells (Tregs) and decreased infiltration and activation of cytotoxic T cells. The combination of radiotherapy with immune checkpoint inhibition is on the rise in the treatment of metastatic cancer patients, but is also tested in multiple curative treatment settings. There is a strong rationale for synergistic effects, such as increased T cell infiltration in irradiated tumors and mitigation of radiation-induced immunosuppressive mechanisms such as PD-L1 upregulation by immune checkpoint inhibition. Given the worse prognosis of patients with hypoxic tumors due to local therapy resistance but also increased rate of distant metastases and the strong immune suppression induced by hypoxia, we hypothesize that the subgroup of patients with hypoxic tumors might be of special interest for combining immune checkpoint inhibition with radiotherapy.
Collapse
Affiliation(s)
- Franziska Eckert
- Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany
- German Cancer Consortium (DKTK) Partnersite Tuebingen, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kerstin Zwirner
- Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany
| | - Simon Boeke
- Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany
- German Cancer Consortium (DKTK) Partnersite Tuebingen, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Section for Biomedical Physics, Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany
| | - Daniela Thorwarth
- German Cancer Consortium (DKTK) Partnersite Tuebingen, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Section for Biomedical Physics, Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany
| | - Daniel Zips
- Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany
- German Cancer Consortium (DKTK) Partnersite Tuebingen, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stephan M. Huber
- Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany
| |
Collapse
|
44
|
Ma Q, Long W, Xing C, Chu J, Luo M, Wang HY, Liu Q, Wang RF. Cancer Stem Cells and Immunosuppressive Microenvironment in Glioma. Front Immunol 2018; 9:2924. [PMID: 30619286 PMCID: PMC6308128 DOI: 10.3389/fimmu.2018.02924] [Citation(s) in RCA: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 11/28/2018] [Indexed: 12/22/2022] Open
Abstract
Glioma is one of the most common malignant tumors of the central nervous system and is characterized by extensive infiltrative growth, neovascularization, and resistance to various combined therapies. In addition to heterogenous populations of tumor cells, the glioma stem cells (GSCs) and other nontumor cells present in the glioma microenvironment serve as critical regulators of tumor progression and recurrence. In this review, we discuss the role of several resident or peripheral factors with distinct tumor-promoting features and their dynamic interactions in the development of glioma. Localized antitumor factors could be silenced or even converted to suppressive phenotypes, due to stemness-related cell reprogramming and immunosuppressive mediators in glioma-derived microenvironment. Furthermore, we summarize the latest knowledge on GSCs and key microenvironment components, and discuss the emerging immunotherapeutic strategies to cure this disease.
Collapse
Affiliation(s)
- Qianquan Ma
- Department of Neurosurgery in Xiangya Hospital, Central South University, Changsha, China.,Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, United States
| | - Wenyong Long
- Department of Neurosurgery in Xiangya Hospital, Central South University, Changsha, China
| | - Changsheng Xing
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, United States
| | - Junjun Chu
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, United States
| | - Mei Luo
- Department of Neurosurgery in Xiangya Hospital, Central South University, Changsha, China.,Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, United States
| | - Helen Y Wang
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, United States
| | - Qing Liu
- Department of Neurosurgery in Xiangya Hospital, Central South University, Changsha, China
| | - Rong-Fu Wang
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, United States.,Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, TX, United States.,Department of Microbiology and Immunology, Weill Cornell Medical College, Cornell University, New York, NY, United States
| |
Collapse
|
45
|
Li Y, Patel SP, Roszik J, Qin Y. Hypoxia-Driven Immunosuppressive Metabolites in the Tumor Microenvironment: New Approaches for Combinational Immunotherapy. Front Immunol 2018; 9:1591. [PMID: 30061885 PMCID: PMC6054965 DOI: 10.3389/fimmu.2018.01591] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 06/27/2018] [Indexed: 12/13/2022] Open
Abstract
Hypoxia is not only a prominent contributor to the heterogeneity of solid tumors but also a crucial stressor in the microenvironment to drive adaptations for tumors to evade immunosurveillance. Herein, we discuss the potential role of hypoxia within the microenvironment contributing to immune resistance and immune suppression of tumor cells. We outline recent discoveries of hypoxia-driven adaptive mechanisms that diminish immune cell response via skewing the expression of important immune checkpoint molecules (e.g., cluster of differentiation 47, programmed death ligand 1, and human leukocyte antigen G), altered metabolism and metabolites, and pH regulation. Importantly, inhibition of hypoxic stress-relevant pathways can collectively enhance T-cell-mediated tumor cell killing. Furthermore, we discuss how manipulation of hypoxia stress may pose a promising new strategy for a combinational therapeutic intervention to enhance immunotherapy of solid tumors.
Collapse
Affiliation(s)
- Yiliang Li
- Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin, China
| | - Sapna Pradyuman Patel
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jason Roszik
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Yong Qin
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
46
|
Celik AA, Simper GS, Huyton T, Blasczyk R, Bade-Döding C. HLA-G mediated immune regulation is impaired by a single amino acid exchange in the alpha 2 domain. Hum Immunol 2018; 79:453-462. [DOI: 10.1016/j.humimm.2018.03.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 03/09/2018] [Accepted: 03/27/2018] [Indexed: 01/04/2023]
|
47
|
Taylor CT, Colgan SP. Regulation of immunity and inflammation by hypoxia in immunological niches. Nat Rev Immunol 2017; 17:774-785. [PMID: 28972206 PMCID: PMC5799081 DOI: 10.1038/nri.2017.103] [Citation(s) in RCA: 425] [Impact Index Per Article: 60.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Immunological niches are focal sites of immune activity that can have varying microenvironmental features. Hypoxia is a feature of physiological and pathological immunological niches. The impact of hypoxia on immunity and inflammation can vary depending on the microenvironment and immune processes occurring in a given niche. In physiological immunological niches, such as the bone marrow, lymphoid tissue, placenta and intestinal mucosa, physiological hypoxia controls innate and adaptive immunity by modulating immune cell proliferation, development and effector function, largely via transcriptional changes driven by hypoxia-inducible factor (HIF). By contrast, in pathological immunological niches, such as tumours and chronically inflamed, infected or ischaemic tissues, pathological hypoxia can drive tissue dysfunction and disease development through immune cell dysregulation. Here, we differentiate between the effects of physiological and pathological hypoxia on immune cells and the consequences for immunity and inflammation in different immunological niches. Furthermore, we discuss the possibility of targeting hypoxia-sensitive pathways in immune cells for the treatment of inflammatory disease.
Collapse
Affiliation(s)
- Cormac T Taylor
- UCD Conway Institute, Systems Biology Ireland and the School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Sean P Colgan
- Department of Medicine and the Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, 80045 Colorado, USA
| |
Collapse
|
48
|
Garziera M, Scarabel L, Toffoli G. Hypoxic Modulation of HLA-G Expression through the Metabolic Sensor HIF-1 in Human Cancer Cells. J Immunol Res 2017; 2017:4587520. [PMID: 28781970 PMCID: PMC5525073 DOI: 10.1155/2017/4587520] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 05/16/2017] [Accepted: 06/11/2017] [Indexed: 12/17/2022] Open
Abstract
The human leukocyte antigen-G (HLA-G) is considered an immune checkpoint molecule involved in tumor immune evasion. Hypoxia and the metabolic sensor hypoxia-inducible factor 1 (HIF-1) are hallmarks of metastasization, angiogenesis, and intense tumor metabolic activity. The purpose of this review was to examine original in vitro studies carried out in human cancer cell lines, which reported data about HLA-G expression and HIF-1 mediated-HLA-G expression in response to hypoxia. The impact of HLA-G genomic variability on the hypoxia responsive elements (HREs) specific for HIF-1 binding was also discussed. Under hypoxia, HLA-G-negative cell lines might transcribe HLA-G without translation of the protein while in contrast, HLA-G-positive cell lines, showed a reduced HLA-G transcriptional activity and protein level. HIF-1 modulation of HLA-G expression induced by hypoxia was demonstrated in different cell lines. HLA-G SNPs rs1632947 and rs41551813 located in distinct HREs demonstrated a prominent role of HIF-1 binding by DNA looping. Our research revealed a fine regulation of HLA-G in hypoxic conditions through HIF-1, depending on the cellular type and HLA-G genomic variability. Specifically, SNPs found in HREs should be considered in future investigations as markers with potential clinical value especially in metastatic malignancies.
Collapse
Affiliation(s)
- Marica Garziera
- Experimental and Clinical Pharmacology Unit, CRO Aviano National Cancer Institute, IRCCS, Via F. Gallini 2, 33081 Aviano, Italy
| | - Lucia Scarabel
- Experimental and Clinical Pharmacology Unit, CRO Aviano National Cancer Institute, IRCCS, Via F. Gallini 2, 33081 Aviano, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, CRO Aviano National Cancer Institute, IRCCS, Via F. Gallini 2, 33081 Aviano, Italy
| |
Collapse
|
49
|
Abstract
In this review we note that the placenta and cancer both develop in microenvironments in which there are gradients of oxygen availability. Whilst fundamentally different in that placental development is organised and physiological whilst cancer is chaotic and pathological, there are similarities in their respective capacities to proliferate, invade adjacent tissues, generate a blood supply and avoid rejection by the immune system. We provide a brief description of the hypoxia-inducible factor (HIF) pathway and indicate the ways by which HIF activity can be regulated to achieve oxygen homeostasis. We then exemplify the potential role of the HIF pathway in contributing to those functions shared between the placenta and cancer through effects on cellular proliferation, cell death, angiogenesis, blood vessel co-option, vascular mimicry, cell adhesion molecules, secretion of matrix metalloproteinases, antigen presentation mechanisms and immunosuppressive factors. We advocate future studies to explore these similarities and differences in the hope of improving our understanding of both systems and hence treatments of placental disorders and cancer.
Collapse
|
50
|
Poras I, Yaghi L, Martelli-Palomino G, Mendes-Junior CT, Muniz YCN, Cagnin NF, Sgorla de Almeida B, Castelli EC, Carosella ED, Donadi EA, Moreau P. Haplotypes of the HLA-G 3' Untranslated Region Respond to Endogenous Factors of HLA-G+ and HLA-G- Cell Lines Differentially. PLoS One 2017; 12:e0169032. [PMID: 28045999 PMCID: PMC5207740 DOI: 10.1371/journal.pone.0169032] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 12/09/2016] [Indexed: 12/11/2022] Open
Abstract
The immune checkpoint HLA-G prevents maternal rejection of the fetus and contributes in cancer invasion and acceptance of allografts. The 5’ and 3’ regulatory regions of the HLA-G gene are polymorphic and balancing selection probably maintains this variability. It is proposed that nucleotide variations may affect the level of HLA-G expression. To investigate this issue we aimed to analyze how haplotypes of the 3’ untranslated region (3’UTR) with highest worldwide frequencies, namely UTR-1, UTR-2, UTR-3, UTR-4, UTR-5, UTR-18 and UTR-7, impact the expression of a luciferase reporter gene in vitro. Experiments performed with the HLA-G positive cell lines JEG-3 (choricarcinoma) and FON (melanoma), and with the HLA-G negative cell lines M8 (melanoma) and U251MG (glioblastoma) showed that the HLA-G 3’UTR polymorphism influences the response to endogenous cellular factors and may vary according to the cell type. UTR-5 and UTR-7 impact the activity of luciferase the most whereas UTR-2, UTR-3, UTR-4, and UTR-18 have intermediate impact, and UTR-1 has the lowest impact. These results corroborate the previous associations between amounts of plasma sHLA-G levels and 3’UTR haplotypes in healthy individuals and reinforce that 3’UTR typing may be a predictor of the genetic predisposition of an individual to express different levels of HLA-G.
Collapse
Affiliation(s)
- Isabelle Poras
- Commissariat à l’Energie Atomique et aux Energies Alternatives, Institut des Maladies Emergentes et des Thérapies Innovantes, Service de Recherches en Hémato-Immunologie, Hôpital Saint-Louis, Paris, France
- Université Paris-Diderot, Sorbonne Paris-Cité, UMR_E5, Institut Universitaire d’Hématologie, Hôpital Saint-Louis, Paris, France
| | - Layale Yaghi
- Commissariat à l’Energie Atomique et aux Energies Alternatives, Institut des Maladies Emergentes et des Thérapies Innovantes, Service de Recherches en Hémato-Immunologie, Hôpital Saint-Louis, Paris, France
- Université Paris-Diderot, Sorbonne Paris-Cité, UMR_E5, Institut Universitaire d’Hématologie, Hôpital Saint-Louis, Paris, France
- Lebanese University, School of Medicine, Hadath, Lebanon
| | - Gustavo Martelli-Palomino
- Commissariat à l’Energie Atomique et aux Energies Alternatives, Institut des Maladies Emergentes et des Thérapies Innovantes, Service de Recherches en Hémato-Immunologie, Hôpital Saint-Louis, Paris, France
- Université Paris-Diderot, Sorbonne Paris-Cité, UMR_E5, Institut Universitaire d’Hématologie, Hôpital Saint-Louis, Paris, France
- Programa de Pós-graduação em Ciências da Saúde, Centro de Ciências da Saúde. Universidade Federal do Rio Grande do Norte, Natal, Rio Grande do Norte, Brasil
| | - Celso T. Mendes-Junior
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Yara Costa Netto Muniz
- Commissariat à l’Energie Atomique et aux Energies Alternatives, Institut des Maladies Emergentes et des Thérapies Innovantes, Service de Recherches en Hémato-Immunologie, Hôpital Saint-Louis, Paris, France
- Université Paris-Diderot, Sorbonne Paris-Cité, UMR_E5, Institut Universitaire d’Hématologie, Hôpital Saint-Louis, Paris, France
- Departamento de Biologia Celular, Embriologia e Genética, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianopolis, Santa Catarina, Brasil
| | - Natalia F. Cagnin
- Commissariat à l’Energie Atomique et aux Energies Alternatives, Institut des Maladies Emergentes et des Thérapies Innovantes, Service de Recherches en Hémato-Immunologie, Hôpital Saint-Louis, Paris, France
- Université Paris-Diderot, Sorbonne Paris-Cité, UMR_E5, Institut Universitaire d’Hématologie, Hôpital Saint-Louis, Paris, France
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Bibiana Sgorla de Almeida
- Commissariat à l’Energie Atomique et aux Energies Alternatives, Institut des Maladies Emergentes et des Thérapies Innovantes, Service de Recherches en Hémato-Immunologie, Hôpital Saint-Louis, Paris, France
- Université Paris-Diderot, Sorbonne Paris-Cité, UMR_E5, Institut Universitaire d’Hématologie, Hôpital Saint-Louis, Paris, France
- Departamento de Biologia Celular, Embriologia e Genética, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianopolis, Santa Catarina, Brasil
- Divisão de Imunologia Clínica, Departamento de Clínica Médica, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brasil
| | - Erick C. Castelli
- Department of Pathology, School of Medicine, Universidade Estadual Paulista, Unesp, Botucatu, São Paulo, Brazil
| | - Edgardo D. Carosella
- Commissariat à l’Energie Atomique et aux Energies Alternatives, Institut des Maladies Emergentes et des Thérapies Innovantes, Service de Recherches en Hémato-Immunologie, Hôpital Saint-Louis, Paris, France
- Université Paris-Diderot, Sorbonne Paris-Cité, UMR_E5, Institut Universitaire d’Hématologie, Hôpital Saint-Louis, Paris, France
| | - Eduardo A. Donadi
- Divisão de Imunologia Clínica, Departamento de Clínica Médica, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brasil
| | - Philippe Moreau
- Commissariat à l’Energie Atomique et aux Energies Alternatives, Institut des Maladies Emergentes et des Thérapies Innovantes, Service de Recherches en Hémato-Immunologie, Hôpital Saint-Louis, Paris, France
- Université Paris-Diderot, Sorbonne Paris-Cité, UMR_E5, Institut Universitaire d’Hématologie, Hôpital Saint-Louis, Paris, France
- * E-mail:
| |
Collapse
|