1
|
He S, Huang Q, Cheng J. The unfolding story of dying tumor cells during cancer treatment. Front Immunol 2023; 14:1073561. [PMID: 36993986 PMCID: PMC10040581 DOI: 10.3389/fimmu.2023.1073561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 02/27/2023] [Indexed: 03/15/2023] Open
Abstract
Generally, the demise of cancer cells in different ways enables the body to clear these harmful cells. However, cancer cells obtain unlimited replication and immortality from successful circumvention of cell death via various mechanisms. Some evidence suggests that treatment-induced dying tumor cells even promote cancer progression. Notably, therapeutic interventions to harness the immune system against tumor cells have shown complicated influences in clinics. Herein, there is an urgent need to clarify the underlying mechanisms that influence the outcome and regulation of the immune system during cancer treatment. In this review, we provide an account on the cell death modes and the relationship between dying tumor cells with tumor immune microenvironment during cancer treatment, focusing on immunotherapy, from mechanistic standpoint to emerging limitations and future directions.
Collapse
Affiliation(s)
| | - Qian Huang
- *Correspondence: Jin Cheng, ; Qian Huang,
| | - Jin Cheng
- *Correspondence: Jin Cheng, ; Qian Huang,
| |
Collapse
|
2
|
de Oliveira Cardoso E, Santiago KB, Conti BJ, Conte FL, Tasca KI, Romagnoli GG, de Assis Golim M, Rainho CA, Bastos JK, Sforcin JM. Brazilian green propolis: A novel tool to improve the cytotoxic and immunomodulatory action of docetaxel on MCF-7 breast cancer cells and on women monocyte. Phytother Res 2021; 36:448-461. [PMID: 34862831 DOI: 10.1002/ptr.7345] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 11/02/2021] [Accepted: 11/12/2021] [Indexed: 12/14/2022]
Abstract
Docetaxel (DTX) is used against breast cancer despite its side effects such as toxicity and immunosuppression. Here we investigated the cytotoxic and immunomodulatory effects of the ethanol solution extract of propolis (EEP) in combination with DTX on MCF-7 breast cancer cells and on women's monocyte. The cytotoxic potential of EEP + DTX was assessed by MTT assay and the type of tumor cell death was evaluated by flow cytometry. The effects of EEP + DTX on the migration and invasion of MCF-7 cells were analyzed. Cytokine production by monocytes was assessed by ELISA and the expression of cell surface markers was evaluated by flow cytometry. We also assessed the fungicidal activity of monocytes against Candida albicans and the generation of reactive oxygen species (ROS). Finally, the impact of the supernatants of treated monocytes in the viability, migration, and invasiveness of tumor cells was assessed. EEP enhanced the cytotoxicity of DTX alone against MCF-7 cells by inducing necrosis and inhibiting their migratory ability. EEP + DTX exerted no cytotoxic effects on monocytes and stimulated HLA-DR expression, TNF-α, and IL-6 production, exerted an immunorestorative action in the fungicidal activity, and reduced the oxidative stress. Our findings have practical implications and reveal new insights for complementary medicine.
Collapse
Affiliation(s)
- Eliza de Oliveira Cardoso
- Department of Chemical and Biological Sciences, Institute of Biosciences, São Paulo State University (UNESP), Campus Botucatu, São Paulo, Brazil
| | - Karina Basso Santiago
- Department of Chemical and Biological Sciences, Institute of Biosciences, São Paulo State University (UNESP), Campus Botucatu, São Paulo, Brazil
| | - Bruno José Conti
- Department of Chemical and Biological Sciences, Institute of Biosciences, São Paulo State University (UNESP), Campus Botucatu, São Paulo, Brazil
| | - Fernanda Lopes Conte
- Department of Chemical and Biological Sciences, Institute of Biosciences, São Paulo State University (UNESP), Campus Botucatu, São Paulo, Brazil
| | - Karen Ingrid Tasca
- Department of Chemical and Biological Sciences, Institute of Biosciences, São Paulo State University (UNESP), Campus Botucatu, São Paulo, Brazil
| | | | - Marjorie de Assis Golim
- Botucatu Blood Center, School of Medicine, São Paulo State University (UNESP), Campus Botucatu, São Paulo, Brazil
| | - Cláudia Aparecida Rainho
- Department of Chemical and Biological Sciences, Institute of Biosciences, São Paulo State University (UNESP), Campus Botucatu, São Paulo, Brazil
| | - Jairo Kenupp Bastos
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, Brazil
| | - José Maurício Sforcin
- Department of Chemical and Biological Sciences, Institute of Biosciences, São Paulo State University (UNESP), Campus Botucatu, São Paulo, Brazil
| |
Collapse
|
3
|
Jeny F, Bernaudin JF, Valeyre D, Kambouchner M, Pretolani M, Nunes H, Planès C, Besnard V. Hypoxia Promotes a Mixed Inflammatory-Fibrotic Macrophages Phenotype in Active Sarcoidosis. Front Immunol 2021; 12:719009. [PMID: 34456926 PMCID: PMC8385772 DOI: 10.3389/fimmu.2021.719009] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/26/2021] [Indexed: 11/30/2022] Open
Abstract
Background Macrophages are pivotal cells in sarcoidosis. Monocytes-derived (MD) macrophages have recently been demonstrated to play a major role especially in pulmonary sarcoidosis. From inflammatory tissues to granulomas, they may be exposed to low oxygen tension environments. As hypoxia impact on sarcoidosis immune cells has never been addressed, we designed the present study to investigate MD-macrophages from sarcoidosis patients in this context. We hypothesized that hypoxia may induce functional changes on MD-macrophages which could have a potential impact on the course of sarcoidosis. Methods We studied MD-macrophages, from high active sarcoidosis (AS) (n=26), low active or inactive sarcoidosis (IS) (n=24) and healthy controls (n=34) exposed 24 hours to normoxia (21% O2) or hypoxia (1.5% O2). Different macrophage functions were explored: hypoxia-inducible factor-1α (HIF-1α) and nuclear factor-kappa B (NF-κB) activation, cytokines secretion, phagocytosis, CD80/CD86/HLA-DR expression, profibrotic response. Results We observed that hypoxia, with a significantly more pronounced effect in AS compared with controls and IS, increased the HIF-1α trans-activity, promoted a proinflammatory response (TNFα, IL1ß) without activating NF-κB pathway and a profibrotic response (TGFß1, PDGF-BB) with PAI-1 secretion associated with human lung fibroblast migration inhibition. These results were confirmed by immunodetection of HIF-1α and PAI-1 in granulomas observed in pulmonary biopsies from patients with sarcoidosis. Hypoxia also decreased the expression of CD80/CD86 and HLA-DR on MD-macrophages in the three groups while it did not impair phagocytosis and the expression of CD36 expression on cells in AS and IS at variance with controls. Conclusions Hypoxia had a significant impact on MD-macrophages from sarcoidosis patients, with the strongest effect seen in patients with high active disease. Therefore, hypoxia could play a significant role in sarcoidosis pathogenesis by increasing the macrophage proinflammatory response, maintaining phagocytosis and reducing antigen presentation, leading to a deficient T cell response. In addition, hypoxia could favor fibrosis by promoting profibrotic cytokines response and by sequestering fibroblasts in the vicinity of granulomas.
Collapse
Affiliation(s)
- Florence Jeny
- INSERM UMR 1272, Sorbonne Paris-Nord University, Bobigny, France
- AP-HP, Pulmonology Department, Avicenne Hospital, Bobigny, France
| | - Jean-François Bernaudin
- INSERM UMR 1272, Sorbonne Paris-Nord University, Bobigny, France
- Faculty of Medicine, Sorbonne University, Paris, France
| | - Dominique Valeyre
- INSERM UMR 1272, Sorbonne Paris-Nord University, Bobigny, France
- AP-HP, Pulmonology Department, Avicenne Hospital, Bobigny, France
| | - Marianne Kambouchner
- INSERM UMR 1272, Sorbonne Paris-Nord University, Bobigny, France
- AP-HP, Pathology Department, Avicenne Hospital, Bobigny, France
| | - Marina Pretolani
- Inserm UMR1152, Physiopathology and Epidemiology of Respiratory Diseases, Paris, France
- Faculty of Medicine, Bichat Hospital, Paris University, Paris, France
- Laboratory of Excellence, INFLAMEX, Paris University, DHU FIRE, Paris, France
| | - Hilario Nunes
- INSERM UMR 1272, Sorbonne Paris-Nord University, Bobigny, France
- AP-HP, Pulmonology Department, Avicenne Hospital, Bobigny, France
| | - Carole Planès
- INSERM UMR 1272, Sorbonne Paris-Nord University, Bobigny, France
- AP-HP, Physiology Department, Avicenne Hospital, Bobigny, France
| | - Valérie Besnard
- INSERM UMR 1272, Sorbonne Paris-Nord University, Bobigny, France
| |
Collapse
|
4
|
Riva A, Palma E, Devshi D, Corrigall D, Adams H, Heaton N, Menon K, Preziosi M, Zamalloa A, Miquel R, Ryan JM, Wright G, Fairclough S, Evans A, Shawcross D, Schierwagen R, Klein S, Uschner FE, Praktiknjo M, Katzarov K, Hadzhiolova T, Pavlova S, Simonova M, Trebicka J, Williams R, Chokshi S. Soluble TIM3 and Its Ligands Galectin-9 and CEACAM1 Are in Disequilibrium During Alcohol-Related Liver Disease and Promote Impairment of Anti-bacterial Immunity. Front Physiol 2021; 12:632502. [PMID: 33776793 PMCID: PMC7987668 DOI: 10.3389/fphys.2021.632502] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 02/10/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND AIMS Immunoregulatory checkpoint receptors (CR) contribute to the profound immunoparesis observed in alcohol-related liver disease (ALD) and in vitro neutralization of inhibitory-CRs TIM3/PD1 on anti-bacterial T-cells can rescue innate and adaptive anti-bacterial immunity. Recently described soluble-CR forms can modulate immunity in inflammatory conditions, but the contributions of soluble-TIM3 and soluble-PD1 and other soluble-CRs to immune derangements in ALD remain unclear. METHODS In Alcoholic Hepatitis (AH; n = 19), alcohol-related cirrhosis (ARC; n = 53) and healthy control (HC; n = 27) subjects, we measured by Luminex technology (i) plasma levels of 16 soluble-CRs, 12 pro/anti-inflammatory cytokines and markers of gut bacterial translocation; (ii) pre-hepatic, post-hepatic and non-hepatic soluble-CR plasma levels in ARC patients undergoing TIPS; (iii) soluble-CRs production from ethanol-treated immunocompetent precision cut human liver slices (PCLS); (iv) whole-blood soluble-CR expression upon bacterial challenge. By FACS, we assessed the relationship between soluble-TIM3 and membrane-TIM3 and rescue of immunity in bacterial-challenged PBMCs. RESULTS Soluble-TIM3 was the dominant plasma soluble-CR in ALD vs. HC (p = 0.00002) and multivariate analysis identified it as the main driver of differences between groups. Soluble-CRs were strongly correlated with pro-inflammatory cytokines, gut bacterial translocation markers and clinical indices of disease severity. Ethanol exposure or bacterial challenge did not induce soluble-TIM3 production from PCLS nor from whole-blood. Bacterial challenge prompted membrane-TIM3 hyperexpression on PBMCs from ALD patient's vs. HC (p < 0.002) and was inversely correlated with plasma soluble-TIM3 levels in matched patients. TIM3 ligands soluble-Galectin-9 and soluble-CEACAM1 were elevated in ALD plasma (AH > ARC; p < 0.002). In vitro neutralization of Galectin-9 and soluble-CEACAM1 improved the defective anti-bacterial and anti-inflammatory cytokine production from E. coli-challenged PBMCs in ALD patients. CONCLUSIONS Alcohol-related liver disease patients exhibit supra-physiological plasma levels of soluble-TIM3, particularly those with greater disease severity. This is also associated with increased levels of soluble TIM3-ligands and membrane-TIM3 expression on immune cells. Soluble-TIM3 can block the TIM3-ligand synapse and improve anti-bacterial immunity; however, the increased levels of soluble TIM3-binding ligands in patients with ALD negate any potential immunostimulatory effects. We believe that anti-TIM3 neutralizing antibodies currently in Phase I clinical trials or soluble-TIM3 should be investigated further for their ability to enhance anti-bacterial immunity. These agents could potentially represent an innovative immune-based supportive approach to rescue anti-bacterial defenses in ALD patients.
Collapse
Affiliation(s)
- Antonio Riva
- Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
- Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| | - Elena Palma
- Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
- Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| | - Dhruti Devshi
- Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
- Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| | - Douglas Corrigall
- Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
- Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
- Department of Gastroenterology, Basildon University Hospital, Basildon, United Kingdom
| | - Huyen Adams
- Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
- Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
- Department of Gastroenterology, Royal Berkshire Hospital, Reading, United Kingdom
| | - Nigel Heaton
- Institute of Liver Studies, King’s College London, London, United Kingdom
| | - Krishna Menon
- Institute of Liver Studies, King’s College London, London, United Kingdom
| | - Melissa Preziosi
- Institute of Liver Studies, King’s College London, London, United Kingdom
| | - Ane Zamalloa
- Institute of Liver Studies, King’s College London, London, United Kingdom
| | - Rosa Miquel
- Liver Histopathology Laboratory, Institute of Liver Studies, King’s College Hospital, London, United Kingdom
| | - Jennifer M. Ryan
- Gastrointestinal and Liver Services, Royal Free Hospital, London, United Kingdom
| | - Gavin Wright
- Department of Gastroenterology, Basildon University Hospital, Basildon, United Kingdom
| | - Sarah Fairclough
- Department of Gastroenterology, Basildon University Hospital, Basildon, United Kingdom
| | - Alexander Evans
- Department of Gastroenterology, Royal Berkshire Hospital, Reading, United Kingdom
| | - Debbie Shawcross
- Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| | - Robert Schierwagen
- Translational Hepatology, Department of Internal Medicine I, University Hospital Frankfurt, Frankfurt, Germany
| | - Sabine Klein
- Translational Hepatology, Department of Internal Medicine I, University Hospital Frankfurt, Frankfurt, Germany
| | - Frank E. Uschner
- Translational Hepatology, Department of Internal Medicine I, University Hospital Frankfurt, Frankfurt, Germany
| | | | - Krum Katzarov
- Department of Gastroenterology, Hepatobiliary Surgery and Transplantology, Military Medical Academy, Sofia, Bulgaria
| | - Tanya Hadzhiolova
- Department of Gastroenterology, Hepatobiliary Surgery and Transplantology, Military Medical Academy, Sofia, Bulgaria
| | - Slava Pavlova
- Department of Gastroenterology, Hepatobiliary Surgery and Transplantology, Military Medical Academy, Sofia, Bulgaria
| | - Marieta Simonova
- Department of Gastroenterology, Hepatobiliary Surgery and Transplantology, Military Medical Academy, Sofia, Bulgaria
| | - Jonel Trebicka
- Translational Hepatology, Department of Internal Medicine I, University Hospital Frankfurt, Frankfurt, Germany
- European Foundation for the Study of Chronic Liver Failure (EF-CLIF), Barcelona, Spain
| | - Roger Williams
- Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
- Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| | - Shilpa Chokshi
- Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
- Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| |
Collapse
|
5
|
Domagala M, Laplagne C, Leveque E, Laurent C, Fournié JJ, Espinosa E, Poupot M. Cancer Cells Resistance Shaping by Tumor Infiltrating Myeloid Cells. Cancers (Basel) 2021; 13:E165. [PMID: 33418996 PMCID: PMC7825276 DOI: 10.3390/cancers13020165] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/23/2020] [Accepted: 12/30/2020] [Indexed: 12/11/2022] Open
Abstract
Interactions between malignant cells and neighboring stromal and immune cells profoundly shape cancer progression. New forms of therapies targeting these cells have revolutionized the treatment of cancer. However, in order to specifically address each population, it was essential to identify and understand their individual roles in interaction between malignant cells, and the formation of the tumor microenvironment (TME). In this review, we focus on the myeloid cell compartment, a prominent, and heterogeneous group populating TME, which can initially exert an anti-tumoral effect, but with time actively participate in disease progression. Macrophages, dendritic cells, neutrophils, myeloid-derived suppressor cells, mast cells, eosinophils, and basophils act alone or in concert to shape tumor cells resistance through cellular interaction and/or release of soluble factors favoring survival, proliferation, and migration of tumor cells, but also immune-escape and therapy resistance.
Collapse
Affiliation(s)
- Marcin Domagala
- Centre de Recherches en Cancérologie de Toulouse, Inserm UMR1037, 31037 Toulouse, France; (M.D.); (C.L.); (E.L.); (C.L.); (J.-J.F.); (E.E.)
- Université Toulouse III Paul-Sabatier, 31400 Toulouse, France
- ERL 5294 CNRS, 31037 Toulouse, France
| | - Chloé Laplagne
- Centre de Recherches en Cancérologie de Toulouse, Inserm UMR1037, 31037 Toulouse, France; (M.D.); (C.L.); (E.L.); (C.L.); (J.-J.F.); (E.E.)
- Université Toulouse III Paul-Sabatier, 31400 Toulouse, France
- ERL 5294 CNRS, 31037 Toulouse, France
| | - Edouard Leveque
- Centre de Recherches en Cancérologie de Toulouse, Inserm UMR1037, 31037 Toulouse, France; (M.D.); (C.L.); (E.L.); (C.L.); (J.-J.F.); (E.E.)
- Université Toulouse III Paul-Sabatier, 31400 Toulouse, France
- ERL 5294 CNRS, 31037 Toulouse, France
| | - Camille Laurent
- Centre de Recherches en Cancérologie de Toulouse, Inserm UMR1037, 31037 Toulouse, France; (M.D.); (C.L.); (E.L.); (C.L.); (J.-J.F.); (E.E.)
- Université Toulouse III Paul-Sabatier, 31400 Toulouse, France
- ERL 5294 CNRS, 31037 Toulouse, France
- IUCT-O, 31000 Toulouse, France
| | - Jean-Jacques Fournié
- Centre de Recherches en Cancérologie de Toulouse, Inserm UMR1037, 31037 Toulouse, France; (M.D.); (C.L.); (E.L.); (C.L.); (J.-J.F.); (E.E.)
- Université Toulouse III Paul-Sabatier, 31400 Toulouse, France
- ERL 5294 CNRS, 31037 Toulouse, France
| | - Eric Espinosa
- Centre de Recherches en Cancérologie de Toulouse, Inserm UMR1037, 31037 Toulouse, France; (M.D.); (C.L.); (E.L.); (C.L.); (J.-J.F.); (E.E.)
- Université Toulouse III Paul-Sabatier, 31400 Toulouse, France
- ERL 5294 CNRS, 31037 Toulouse, France
| | - Mary Poupot
- Centre de Recherches en Cancérologie de Toulouse, Inserm UMR1037, 31037 Toulouse, France; (M.D.); (C.L.); (E.L.); (C.L.); (J.-J.F.); (E.E.)
- Université Toulouse III Paul-Sabatier, 31400 Toulouse, France
- ERL 5294 CNRS, 31037 Toulouse, France
| |
Collapse
|
6
|
Lin H, Ji DK, Lucherelli MA, Reina G, Ippolito S, Samorì P, Bianco A. Comparative Effects of Graphene and Molybdenum Disulfide on Human Macrophage Toxicity. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2002194. [PMID: 32743979 DOI: 10.1002/smll.202002194] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 06/24/2020] [Indexed: 06/11/2023]
Abstract
Graphene and other 2D materials, such as molybdenum disulfide, have been increasingly used in electronics, composites, and biomedicine. In particular, MoS2 and graphene hybrids have attracted a great interest for applications in the biomedical research, therefore stimulating a pertinent investigation on their safety in immune cells like macrophages, which commonly engulf these materials. In this study, M1 and M2 macrophage viability and activation are mainly found to be unaffected by few-layer graphene (FLG) and MoS2 at doses up to 50 µg mL-1 . The uptake of both materials is confirmed by transmission electron microscopy, inductively coupled plasma mass spectrometry, and inductively coupled plasma atomic emission spectroscopy. Notably, both 2D materials increase the secretion of inflammatory cytokines in M1 macrophages. At the highest dose, FLG decreases CD206 expression while MoS2 decreases CD80 expression. CathB and CathL gene expressions are dose-dependently increased by both materials. Despite a minimal impact on the autophagic pathway, FLG is found to increase the expression of Atg5 and autophagic flux, as observed by Western blotting of LC3-II, in M1 macrophages. Overall, FLG and MoS2 are of little toxicity in human macrophages even though they are found to trigger cell stress and inflammatory responses.
Collapse
Affiliation(s)
- Hazel Lin
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry UPR 3572, University of Strasbourg, ISIS, Strasbourg, 67000, France
| | - Ding-Kun Ji
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry UPR 3572, University of Strasbourg, ISIS, Strasbourg, 67000, France
| | - Matteo Andrea Lucherelli
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry UPR 3572, University of Strasbourg, ISIS, Strasbourg, 67000, France
| | - Giacomo Reina
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry UPR 3572, University of Strasbourg, ISIS, Strasbourg, 67000, France
| | | | - Paolo Samorì
- CNRS, Université de Strasbourg, ISIS, Strasbourg, 67000, France
| | - Alberto Bianco
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry UPR 3572, University of Strasbourg, ISIS, Strasbourg, 67000, France
- CNRS, Université de Strasbourg, ISIS, Strasbourg, 67000, France
| |
Collapse
|
7
|
Farina AR, Cappabianca L, Sebastiano M, Zelli V, Guadagni S, Mackay AR. Hypoxia-induced alternative splicing: the 11th Hallmark of Cancer. J Exp Clin Cancer Res 2020; 39:110. [PMID: 32536347 PMCID: PMC7294618 DOI: 10.1186/s13046-020-01616-9] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 06/03/2020] [Indexed: 12/16/2022] Open
Abstract
Hypoxia-induced alternative splicing is a potent driving force in tumour pathogenesis and progression. In this review, we update currents concepts of hypoxia-induced alternative splicing and how it influences tumour biology. Following brief descriptions of tumour-associated hypoxia and the pre-mRNA splicing process, we review the many ways hypoxia regulates alternative splicing and how hypoxia-induced alternative splicing impacts each individual hallmark of cancer. Hypoxia-induced alternative splicing integrates chemical and cellular tumour microenvironments, underpins continuous adaptation of the tumour cellular microenvironment responsible for metastatic progression and plays clear roles in oncogene activation and autonomous tumour growth, tumor suppressor inactivation, tumour cell immortalization, angiogenesis, tumour cell evasion of programmed cell death and the anti-tumour immune response, a tumour-promoting inflammatory response, adaptive metabolic re-programming, epithelial to mesenchymal transition, invasion and genetic instability, all of which combine to promote metastatic disease. The impressive number of hypoxia-induced alternative spliced protein isoforms that characterize tumour progression, classifies hypoxia-induced alternative splicing as the 11th hallmark of cancer, and offers a fertile source of potential diagnostic/prognostic markers and therapeutic targets.
Collapse
Affiliation(s)
- Antonietta Rosella Farina
- Department of Applied Clinical and Biotechnological Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Lucia Cappabianca
- Department of Applied Clinical and Biotechnological Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Michela Sebastiano
- Department of Applied Clinical and Biotechnological Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Veronica Zelli
- Department of Applied Clinical and Biotechnological Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Stefano Guadagni
- Department of Applied Clinical and Biotechnological Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Andrew Reay Mackay
- Department of Applied Clinical and Biotechnological Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| |
Collapse
|
8
|
Abstract
Myeloid-derived suppressor cells (MDSCs) have a strong immunosuppressive character that allows them to regulate immune responses and hinder overt inflammatory responses. In cancer, this leads to tumor immune evasion and disease progression. MDSCs come in at least two forms: monocytic (Mo-MDSCs) and granulocytic (G-MDSCs). The classical definition of MDSCs as immature myeloid cells blocked from differentiating has been challenged by recent studies suggesting that Mo-MDSCs and G-MDSCs may represent monocytes and granulocytes that have acquired immunosuppressive properties. The molecular mechanism behind their generation and their true origins are now widely debated. In this review we discuss the different proposed mechanisms of the generation of both types of MDSCs, with a special focus on human MDSCs in cancer.
Collapse
|
9
|
Jin SH, Ahn KJ, An S. Importance of the immune response to Mycobacterium leprae in the skin. BIOMEDICAL DERMATOLOGY 2018. [DOI: 10.1186/s41702-017-0012-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
10
|
Zheng X, Zhang Y, Liu Y, Fang L, Li L, Sun J, Pan Z, Xin W, Huang P. HIF‐2α activated lncRNA NEAT1 promotes hepatocellular carcinoma cell invasion and metastasis by affecting the epithelial‐mesenchymal transition. J Cell Biochem 2017; 119:3247-3256. [DOI: 10.1002/jcb.26481] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 10/31/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Xiaowei Zheng
- Department of PharmacyZhejiang Cancer HospitalHangzhouZhejiangPeople's Republic of China
| | - Yiwen Zhang
- Department of PharmacyZhejiang Cancer HospitalHangzhouZhejiangPeople's Republic of China
| | - Yujia Liu
- Department of PharmacyZhejiang Cancer HospitalHangzhouZhejiangPeople's Republic of China
| | - Luo Fang
- Department of PharmacyZhejiang Cancer HospitalHangzhouZhejiangPeople's Republic of China
| | - Li Li
- Department of PharmacyThe First People's Hospital of Chun'an CountyZhejiangPeople's Republic of China
| | - Jiao Sun
- Department of PharmacyZhejiang Cancer HospitalHangzhouZhejiangPeople's Republic of China
| | - Zongfu Pan
- Department of PharmacyZhejiang Cancer HospitalHangzhouZhejiangPeople's Republic of China
| | - Wenxiu Xin
- Department of PharmacyZhejiang Cancer HospitalHangzhouZhejiangPeople's Republic of China
| | - Ping Huang
- Department of PharmacyZhejiang Cancer HospitalHangzhouZhejiangPeople's Republic of China
| |
Collapse
|
11
|
Tono T, Aihara S, Hoshiyama T, Arinuma Y, Nagai T, Hirohata S. Effects of CTLA4-Ig on human monocytes. Inflamm Regen 2017; 37:24. [PMID: 29259723 PMCID: PMC5725919 DOI: 10.1186/s41232-017-0054-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 10/04/2017] [Indexed: 11/18/2022] Open
Abstract
Background Abatacept, a CTLA4-Ig fusion protein attenuates T cell activation by inhibiting the CD80/86-CD28 costimulatory pathway that is required for the proper T cell activation and thus displays beneficial effects in the treatment of rheumatoid arthritis (RA). Although some studies have disclosed the in vitro effects of this biological agent on the immune-competent cells, the precise mechanisms of action in RA still remain unclear. The current studies were therefore undertaken to explore the effects of abatacept on monocytes in detail. Methods Monocytes from healthy donors were cultured in the presence of staphylococcal enterotoxin B (SEB) with pharmacologically attainable concentrations of abatacept or control IgG-Fc. The expression of CD80 and CD86 and the induction of apoptosis of monocytes were measured by flow cytometry. The expression of CD80 and CD86 messenger RNA (mRNA) was determined by quantitative RT-PCR. Results Abatacept promoted apoptosis of SEB-stimulated monocytes. The induction of apoptosis of monocytes by these biological agents was reversed by the addition of IgG, but not IgG-F(ab′)2 fragments. Furthermore, abatacept significantly suppressed the expression of CD80, but not that of CD86 at protein levels. Finally, abatacept significantly suppressed the expression of mRNA for CD80 of monocytes stimulated with SEB, but not that of CD86. Conclusions These results demonstrate that one of the mechanisms of action of abatacept involves the induction of apoptosis of monocytes, which involves interaction with Fc receptor on monocytes. Moreover, the data also demonstrate that abatacept selectively suppresses the expression of CD80 at mRNA levels.
Collapse
Affiliation(s)
- Toshihiro Tono
- Department of Rheumatology and Infectious Diseases, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0374 Japan
| | - Satoko Aihara
- Department of Rheumatology and Infectious Diseases, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0374 Japan
| | - Takayuki Hoshiyama
- Department of Rheumatology and Infectious Diseases, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0374 Japan
| | - Yoshiyuki Arinuma
- Department of Rheumatology and Infectious Diseases, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0374 Japan
| | - Tatsuo Nagai
- Department of Rheumatology and Infectious Diseases, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0374 Japan
| | - Shunsei Hirohata
- Department of Rheumatology and Infectious Diseases, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0374 Japan
| |
Collapse
|
12
|
Xu B, Gao Y, Zhan S, Ge W. Quantitative proteomic profiling for clarification of the crucial roles of lysosomes in microbial infections. Mol Immunol 2017; 87:122-131. [PMID: 28433889 DOI: 10.1016/j.molimm.2017.04.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 03/22/2017] [Accepted: 04/04/2017] [Indexed: 02/05/2023]
Abstract
Lysosomes play vital roles in both innate and adaptive immunity. It is widely accepted that lysosomes do not function exclusively as a digestive organelle. It is also involved in the process of immune cells against pathogens. However, the changes in the lysosomal proteome caused by infection with various microbes are still largely unknown, and our understanding of the proteome of the purified lysosome is another obstacle that needs to be resolved. Here, we performed a proteomic study on lysosomes enriched from THP1 cells after infection with Listeria monocytogenes (L.m), Herpes Simplex Virus 1 (HSV-1) and Vesicular Stomatitis Virus (VSV). In combination with the gene ontology (GO) analysis, we identified 284 lysosomal-related proteins from a total of 4560 proteins. We also constructed the protein-protein interaction networks for the differentially expressed proteins and revealed the core lysosomal proteins, including SRC in the L. m treated group, SRC, GLB1, HEXA and HEXB in the HSV-1 treated group and GLB1, CTSA, CTSB, HEXA and HEXB in the VSV treated group, which are involved in responding to diverse microbial infections. This study not only reveals variable lysosome responses depending on the bacterial or virus infection, but also provides the evidence based on which we propose a novel approach to proteome research for investigation of the function of the enriched organelles.
Collapse
Affiliation(s)
- Benhong Xu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, National Key Laboratory of Medical Molecular Biology & Department of Immunology, No 5 Dongdan Santiao, Dongcheng District, Beijing 100005, China
| | - Yanpan Gao
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, National Key Laboratory of Medical Molecular Biology & Department of Immunology, No 5 Dongdan Santiao, Dongcheng District, Beijing 100005, China
| | - Shaohua Zhan
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, National Key Laboratory of Medical Molecular Biology & Department of Immunology, No 5 Dongdan Santiao, Dongcheng District, Beijing 100005, China
| | - Wei Ge
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, National Key Laboratory of Medical Molecular Biology & Department of Immunology, No 5 Dongdan Santiao, Dongcheng District, Beijing 100005, China.
| |
Collapse
|
13
|
Baghdadi HH. Targeting Cancer Cells using 3-bromopyruvate for Selective Cancer Treatment. SAUDI JOURNAL OF MEDICINE & MEDICAL SCIENCES 2016; 5:9-19. [PMID: 30787746 PMCID: PMC6298280 DOI: 10.4103/1658-631x.194253] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Cancer treatment deserves more research efforts despite intensive conventional treatment modalities for many types of malignancies. Metastasis and resistance to chemotherapy and radiotherapy receive a lot of global research efforts. The current advances in cancer biology may improve targeting the critical metabolic differences that distinguish cancer cells from normal cells. Cancer cells are highly glycolytic for energy production, exhibit the Warburg effect, establish aggressive acidic microenvironment, maintain cancer stem cells, exhibit resistance to chemotherapy, have low antioxidant systems but different ΔΨm (delta psi, mitochondrial transmembrane potential), express P-glycoprotein for multidrug resistance, upregulate glucose transporters and monocarboxylate transporters and are under high steady-state reactive oxygen species conditions. Normal cells differ in all these aspects. Lactate produced through the Warburg effect helps cancer metastasis. Targeting glycolysis reactions for energy production in cancer cells seems promising in decreasing the proliferation and metastasis of cancer cells. 3-bromopyruvate makes use of cancer biology in treating cancer cells, cancer stem cells and preventing metastasis in human cancer as discussed in this review. Updated advances are analyzed here, which include research analysis of background, experience, readings in the field of cancer biology, oncology and biochemistry.
Collapse
Affiliation(s)
- Hussam H Baghdadi
- Department of Clinical Biochemistry and Molecular Medicine, Taibah University, Al-Madinah Al-Munawwarah, Saudi Arabia
| |
Collapse
|
14
|
Holmannova D, Kolackova M, Kunes P, Krejsek J, Mandak J, Andrys C. Impact of cardiac surgery on the expression of CD40, CD80, CD86 and HLA-DR on B cells and monocytes. Perfusion 2015; 31:391-400. [PMID: 26503949 DOI: 10.1177/0267659115612905] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE We measured and compared changes in the percentage of cells expressing CD80, CD86, CD40, HLA-DR and the expression of these molecules on B cells and monocytes of patients who underwent either on-pump, mini on-pump or off-pump cardiac surgery. METHODS Blood samples from patients who underwent either on-pump, mini on-pump or off-pump cardiac surgery were collected before surgery, instantly after surgery and on the 1(st), 3(rd) and 7(th) days after surgery. Surface expression of CD80, CD86, CD40 and HLA-DR molecules was determined by flow cytometry. RESULTS Our results show that all three surgical techniques altered the expression of these molecules, as well as the percentage relative number of specific cell populations. We identified statistically significant differences when comparing different surgical techniques. On-pump surgery revealed a more pronounced impact on the phenotype of immune system cells than the other techniques. Therefore, it is likely that the function of immune cells is changed the most by on-pump surgery. We found a lower decrease in the number of CD80(+) monocytes and a lower drop in the CD40 expression on monocytes in off-pump patients in comparison with on-pump patients. CONCLUSION All the types of cardiac surgical techniques, off-pump, on-pump and modified mini-invasive on-pump, are associated with changes in CD80, CD86, CD40 and HLA-DR expression. We found several significant differences in the expression of the selected molecules when we compared all three groups of patients.
Collapse
Affiliation(s)
- Drahomira Holmannova
- Department of Clinical Immunology and Allergology, Charles University in Prague, Faculty of Medicine and University Hospital in Hradec Kralove, Czech Republic
| | - Martina Kolackova
- Department of Clinical Immunology and Allergology, Charles University in Prague, Faculty of Medicine and University Hospital in Hradec Kralove, Czech Republic
| | - Pavel Kunes
- Department of Cardiac Surgery, Charles University in Prague, Faculty of Medicine and University Hospital in Hradec Kralove, Czech Republic
| | - Jan Krejsek
- Department of Clinical Immunology and Allergology, Charles University in Prague, Faculty of Medicine and University Hospital in Hradec Kralove, Czech Republic
| | - Jiri Mandak
- Department of Cardiac Surgery, Charles University in Prague, Faculty of Medicine and University Hospital in Hradec Kralove, Czech Republic
| | - Ctirad Andrys
- Department of Clinical Immunology and Allergology, Charles University in Prague, Faculty of Medicine and University Hospital in Hradec Kralove, Czech Republic
| |
Collapse
|
15
|
Lyrio EC, Campos-Souza IC, Corrêa LC, Lechuga GC, Verícimo M, Castro HC, Bourguignon SC, Côrte-Real S, Ratcliffe N, Declercq W, Santos DO. Interaction ofMycobacterium lepraewith the HaCaT human keratinocyte cell line: new frontiers in the cellular immunology of leprosy. Exp Dermatol 2015; 24:536-42. [DOI: 10.1111/exd.12714] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2015] [Indexed: 01/12/2023]
Affiliation(s)
- Eloah C.D. Lyrio
- Laboratório de Biopatógenos e Ativação Celular (LaBiopAc); Departamento de Biologia Celular e Molecular; Universidade Federal Fluminense (UFF); Niterói RJ Brazil
- Programa de Pós-graduação em Ciências e Biotecnologia; UFF; Niterói RJ Brazil
| | - Ivy C. Campos-Souza
- Laboratório de Biopatógenos e Ativação Celular (LaBiopAc); Departamento de Biologia Celular e Molecular; Universidade Federal Fluminense (UFF); Niterói RJ Brazil
| | - Luiz C.D. Corrêa
- Laboratório de Biopatógenos e Ativação Celular (LaBiopAc); Departamento de Biologia Celular e Molecular; Universidade Federal Fluminense (UFF); Niterói RJ Brazil
- Programa de Pós-graduação em Ciências e Biotecnologia; UFF; Niterói RJ Brazil
| | | | - Maurício Verícimo
- Programa de Pós-graduação em Patologia; Hospital Universitário; Niterói RJ Brazil
| | - Helena C. Castro
- Programa de Pós-graduação em Ciências e Biotecnologia; UFF; Niterói RJ Brazil
- Programa de Pós-graduação em Patologia; Hospital Universitário; Niterói RJ Brazil
| | | | | | - Norman Ratcliffe
- Programa de Pós-graduação em Ciências e Biotecnologia; UFF; Niterói RJ Brazil
- College of Science; Swansea University; Wales UK
| | - Wim Declercq
- Inflammation Research Center; University of Ghent-VIB; Ghent Belgium
| | - Dilvani O. Santos
- Laboratório de Biopatógenos e Ativação Celular (LaBiopAc); Departamento de Biologia Celular e Molecular; Universidade Federal Fluminense (UFF); Niterói RJ Brazil
- Programa de Pós-graduação em Ciências e Biotecnologia; UFF; Niterói RJ Brazil
| |
Collapse
|
16
|
Jeon YK, Park SG, Choi IW, Lee SW, Lee SM, Choi I. Cancer cell-associated cytoplasmic B7–H4 is induced by hypoxia through hypoxia-inducible factor-1α and promotes cancer cell proliferation. Biochem Biophys Res Commun 2015; 459:277-283. [DOI: 10.1016/j.bbrc.2015.02.098] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 02/17/2015] [Indexed: 02/07/2023]
|
17
|
Bhandari T, Nizet V. Hypoxia-Inducible Factor (HIF) as a Pharmacological Target for Prevention and Treatment of Infectious Diseases. Infect Dis Ther 2014; 3:159-74. [PMID: 25134687 PMCID: PMC4269623 DOI: 10.1007/s40121-014-0030-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Indexed: 02/07/2023] Open
Abstract
In the present era of ever-increasing antibiotic resistance and increasingly complex and immunosuppressed patient populations, physicians and scientists are seeking novel approaches to battle difficult infectious disease conditions. Development of a serious infection implies a failure of innate immune capabilities in the patient, and one may consider whether pharmacological strategies exist to correct and enhance innate immune cell function. Hypoxia-inducible factor-1 (HIF-1), the central regulator of the cellular response to hypoxic stress, has recently been recognized to control the activation state and key microbicidal functions of immune cells. HIF-1 boosting drugs are in clinical development for anemia and other indications, and could be repositioned as infectious disease therapeutics. With equal attention to opportunities and complexities, we review our current understanding of HIF-1 regulation of microbial host-pathogen interactions with an eye toward future drug development.
Collapse
Affiliation(s)
- Tamara Bhandari
- Center for Immunity, Infection and Inflammation, Department of Pediatrics and Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, USA
| | - Victor Nizet
- Center for Immunity, Infection and Inflammation, Department of Pediatrics and Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, USA.
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, USA.
- Center for Immunity, Infection and Inflammation, Medical Sciences Research 4113, University of California, San Diego, 9500 Gilman Drive, MC 0760, La Jolla, CA, 92093-0760, USA.
| |
Collapse
|
18
|
Harris AJ, Thompson AR, Whyte MK, Walmsley SR. HIF-mediated innate immune responses: cell signaling and therapeutic implications. HYPOXIA 2014; 2:47-58. [PMID: 27774466 PMCID: PMC5045056 DOI: 10.2147/hp.s50269] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Leukocytes recruited to infected, damaged, or inflamed tissues during an immune response must adapt to oxygen levels much lower than those in the circulation. Hypoxia inducible factors (HIFs) are key mediators of cellular responses to hypoxia and, as in other cell types, HIFs are critical for the upregulation of glycolysis, which enables innate immune cells to produce adenosine triphosphate anaerobically. An increasing body of evidence demonstrates that hypoxia also regulates many other innate immunological functions, including cell migration, apoptosis, phagocytosis of pathogens, antigen presentation and production of cytokines, chemokines, and angiogenic and antimicrobial factors. Many of these functions are mediated by HIFs, which are not only stabilized posttranslationally by hypoxia, but also transcriptionally upregulated by inflammatory signals. Here, we review the role of HIFs in the responses of innate immune cells to hypoxia, both in vitro and in vivo, with a particular focus on myeloid cells, on which the majority of studies have so far been carried out.
Collapse
Affiliation(s)
- Alison J Harris
- Academic Unit of Respiratory Medicine, Department of Infection and Immunity, University of Sheffield, Sheffield, UK
| | - Aa Roger Thompson
- Academic Unit of Respiratory Medicine, Department of Infection and Immunity, University of Sheffield, Sheffield, UK
| | - Moira Kb Whyte
- Academic Unit of Respiratory Medicine, Department of Infection and Immunity, University of Sheffield, Sheffield, UK
| | - Sarah R Walmsley
- Academic Unit of Respiratory Medicine, Department of Infection and Immunity, University of Sheffield, Sheffield, UK
| |
Collapse
|
19
|
Giorgio S. Macrophages: plastic solutions to environmental heterogeneity. Inflamm Res 2013; 62:835-43. [DOI: 10.1007/s00011-013-0647-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 07/05/2013] [Indexed: 12/14/2022] Open
|
20
|
Ghio AJ, Dailey LA, Soukup JM, Stonehuerner J, Richards JH, Devlin RB. Growth of human bronchial epithelial cells at an air-liquid interface alters the response to particle exposure. Part Fibre Toxicol 2013; 10:25. [PMID: 23800224 PMCID: PMC3750262 DOI: 10.1186/1743-8977-10-25] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 05/15/2013] [Indexed: 11/16/2022] Open
Abstract
Background We tested the hypothesis that normal human bronchial epithelial (NHBE) cells 1) grown submerged in media and 2) allowed to differentiate at air-liquid interface (ALI) demonstrate disparities in the response to particle exposure. Results Following exposure of submerged NHBE cells to ambient air pollution particle collected in Chapel Hill, NC, RNA for IL-8, IL-6, heme oxygenase 1 (HOX1) and cyclooxygenase 2 (COX2) increased. The same cells allowed to differentiate over 3, 10, and 21 days at ALI demonstrated no such changes following particle exposure. Similarly, BEAS-2B cells grown submerged in media demonstrated a significant increase in IL-8 and HOX1 RNA after exposure to NIST 1648 particle relative to the same cells exposed after growth at ALI. Subsequently, it was not possible to attribute the observed decreases in the response of NHBE cells to differentiation alone since BEAS-2B cells, which do not differentiate, showed similar changes when grown at ALI. With no exposure to particles, differentiation of NHBE cells at ALI over 3 to 21 days demonstrated significant decrements in baseline levels of RNA for the same proteins (i.e. IL-8, IL-6, HOX1, and COX2). With no exposure to particles, BEAS-2B cells grown at ALI showed comparable changes in RNA for IL-8 and HOX1. After the same particle exposure, NHBE cells grown at ALI on a transwell in 95% N2-5% CO2 and exposed to NIST 1648 particle demonstrated significantly greater changes in IL-8 and HOX1 relative to cells grown in 95% air-5% CO2. Conclusions We conclude that growth of NHBE cells at ALI is associated with a diminished biological effect following particle exposure relative to cells submerged in media. This decreased response showed an association with increased oxygen availability.
Collapse
|
21
|
Nickel D, Busch M, Mayer D, Hagemann B, Knoll V, Stenger S. Hypoxia Triggers the Expression of Human β Defensin 2 and Antimicrobial Activity againstMycobacterium tuberculosisin Human Macrophages. THE JOURNAL OF IMMUNOLOGY 2012; 188:4001-7. [DOI: 10.4049/jimmunol.1100976] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
22
|
Rahat MA, Bitterman H, Lahat N. Molecular mechanisms regulating macrophage response to hypoxia. Front Immunol 2011; 2:45. [PMID: 22566835 PMCID: PMC3342364 DOI: 10.3389/fimmu.2011.00045] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 08/29/2011] [Indexed: 12/24/2022] Open
Abstract
Monocytes and Macrophages (Mo/Mɸ) exhibit great plasticity, as they can shift between different modes of activation and, driven by their immediate microenvironment, perform divergent functions. These include, among others, patrolling their surroundings and maintaining homeostasis (resident Mo/Mɸ), combating invading pathogens and tumor cells (classically activated or M1 Mo/Mɸ), orchestrating wound healing (alternatively activated or M2 Mo/Mɸ), and restoring homeostasis after an inflammatory response (resolution Mɸ). Hypoxia is an important factor in the Mɸ microenvironment, is prevalent in many physiological and pathological conditions, and is interdependent with the inflammatory response. Although Mo/Mɸ have been studied in hypoxia, the mechanisms by which hypoxia influences the different modes of their activation, and how it regulates the shift between them, remain unclear. Here we review the current knowledge about the molecular mechanisms that mediate this hypoxic regulation of Mɸ activation. Much is known about the hypoxic transcriptional regulatory network, which includes the master regulators hypoxia-induced factor-1 and NF-κB, as well as other transcription factors (e.g., AP-1, Erg-1), but we also highlight the role of post-transcriptional and post-translational mechanisms. These mechanisms mediate hypoxic induction of Mɸ pro-angiogenic mediators, suppress M1 Mɸ by post-transcriptionally inhibiting pro-inflammatory mediators, and help shift the classically activated Mɸ into an activation state which approximate the alternatively activated or resolution Mɸ.
Collapse
Affiliation(s)
- Michal A Rahat
- Immunology Research Unit, Carmel Medical Center, The Ruth and Bruce Rappaport Faculty of Medicine, Technion Haifa, Israel.
| | | | | |
Collapse
|
23
|
Lahat N, Bitterman H, Weiss-Cerem L, Rahat MA. Hypoxia increases membranal and secreted HLA-DR in endothelial cells, rendering them T-cell activators. Transpl Int 2011; 24:1018-26. [PMID: 21806687 DOI: 10.1111/j.1432-2277.2011.01304.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Transplantation involves preoperative ischemic periods that contribute to endothelial cell (EC) dysfunction and T-cell activation, leading to graft rejection. As hypoxia is a major constituent of ischemia, we evaluated its effect on the ability of ECs to express HLA-DR, which is required for presentation of antigens to T cells, and by itself serves as an important target for allogeneic T cells. Primary human umbilical vein ECs (HUVEC) and the human endothelial cell line EaHy926 were incubated in normoxia or hypoxia (PO(2) < 0.3%). Hypoxia increased the membranal expression (by 4-6 fold, P < 0.01) and secretion (by sixfold, P < 0.05) of HLA-DR protein, without influencing the accumulation of its mRNA. Alternative splicing, attenuated trafficking, or shedding from the plasma membrane were not observed, but the lysosomal inhibitor bafilomycin A1 reduced HLA-DR secretion. Hypoxia-induced endothelial HLA-DR elevated and diminished the secretion of IL-2 and IL-10, respectively, from co-cultured allogeneic CD4(+) T cells in a HLA-DR-dependent manner, as demonstrated by the use of monoclonal anti-HLA-DR. Our results indicate a yet not fully understood post-translational mechanism(s), which elevate both membranal and soluble HLA-DR expression. This elevation is involved in allogeneic T-cell activation, highlighting the pivotal role of ECs in ischemia/hypoxia-associated injury and graft rejection.
Collapse
Affiliation(s)
- Nitza Lahat
- Immunology Research Unit, Carmel Medical Center, The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | | | | | | |
Collapse
|
24
|
Staples KJ, Sotoodehnejadnematalahi F, Pearson H, Frankenberger M, Francescut L, Ziegler-Heitbrock L, Burke B. Monocyte-derived macrophages matured under prolonged hypoxia transcriptionally up-regulate HIF-1α mRNA. Immunobiology 2010; 216:832-9. [PMID: 21281980 DOI: 10.1016/j.imbio.2010.12.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2010] [Accepted: 12/10/2010] [Indexed: 10/18/2022]
Abstract
This study tested the hypothesis that prolonged severe hypoxia during monocyte to macrophage differentiation results in macrophages with a pattern of gene expression and phenotype distinct from those maturing in normal oxygen levels. Macrophages accumulate in hypoxic and anoxic areas within pathological sites such as tumours, wounds, and arthritic joints, and have been proposed as vehicles for gene therapy delivery to such tissues. Several non-pathological tissues are also hypoxic. We therefore argue that differentiation from monocyte to macrophage in hypoxic conditions is a common occurrence. However, the effect of long term severe hypoxia on monocyte to macrophage differentiation has not been studied. Here, using primary human peripheral blood monocytes, we show that maturation for 5 days in 0.2% oxygen results in decreased phagocytosis, and decreased CD40 and CD206 expression. Chronic hypoxia induced much higher mRNA levels of the pro-angiogenic cytokine, VEGF, in adherence-purified macrophages (27-fold), CD14-magnetic bead purified monocytes (90-fold), and PBMC (104-fold) compared to acute (24h) hypoxia (11, 17 and 9-fold, respectively). This suggests that macrophages may play an even greater role in angiogenesis than previously appreciated. Furthermore, chronic hypoxia resulted in up-regulation of HIF-1α mRNA, in all monocyte-derived macrophage types studied. Actinomycin D experiments indicate that the increases in HIF-1α mRNA were not due to increased mRNA stability. To our knowledge this is the first study demonstrating up-regulation of HIF-1α mRNA by hypoxia in macrophages. Taken together, the data support the hypothesis that hypoxia affects monocyte to macrophage maturation, resulting in a distinct gene expression pattern and phenotype.
Collapse
Affiliation(s)
- Karl J Staples
- Department of Infection, Immunity & Inflammation, University of Leicester, Leicester, UK
| | | | | | | | | | | | | |
Collapse
|
25
|
Zuidema MY, Zhang C. Ischemia/reperfusion injury: The role of immune cells. World J Cardiol 2010; 2:325-32. [PMID: 21160610 PMCID: PMC2999044 DOI: 10.4330/wjc.v2.i10.325] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Revised: 08/19/2010] [Accepted: 08/26/2010] [Indexed: 02/06/2023] Open
Abstract
Ischemia/reperfusion (I/R) injury is an inflammatory condition that is characterized by innate immunity and an adaptive immune response. This review is focused on the acute inflammatory response in I/R injury, and also the adaptive immunological mechanisms in chronic ischemic disease that lead to increased vulnerability during acute events, in relation to the cell types that have been shown to mediate innate immunity to an adaptive immune response in I/R, specifically myocardial infarction. Novel aspects are also highlighted in respect to the mechanisms within the cardiovascular system and cardiovascular risk factors that may be involved in the inflammatory response accompanying myocardial infarction. Experimental myocardial I/R has suggested that immune cells may mediate reperfusion injury. Specifically, monocytes, macrophages, T-cells, mast cells, platelets and endothelial cells are discussed with reference to the complement cascade, toll-like receptors, cytokines, oxidative stress, renin-angiotensin system, and in reference to the microvascular system in the signaling mechanisms of I/R. Finally, the findings of the data summarized in this review are most important for possible translation into clinical cardiology practice and possible avenues for drug development.
Collapse
Affiliation(s)
- Mozow Y Zuidema
- Mozow Y Zuidema, Cuihua Zhang, Division of Cardiovascular Disease, Dalton Cardiovascular Research Center, Department of Internal Medicine, Medical Pharmacology and Physiology and Nutritional Science, Dalton Cardiovascular Research Center, University of Missouri, Columbia, School of Medicine, Columbia, MO 65212, United States
| | | |
Collapse
|
26
|
Abstract
The abnormal decrease or the lack of oxygen supply to cells and tissues is called hypoxia. This condition is commonly seen in various diseases such as rheumatoid arthritis and atherosclerosis, also in solid cancers. Pre-clinical and clinical studies have shown that hypoxic cancers are extremely aggressive, resistant to standard therapies (chemotherapy and radiotherapy), and thus very difficult to eradicate. Hypoxia affects both the tumor and the immune cells via various pathways. This review summarizes the most common effects of hypoxia on immune cells that play a key role in the anti-tumor response, the limitation of current therapies, and the potential solutions that were developed for hypoxic malignancies.
Collapse
Affiliation(s)
- Patricia Yotnda
- Center for Cell and Gene Therapy, Baylor College of Medicine, One Baylor Plaza, 77030, Houston, TX, USA.
| | | | | |
Collapse
|
27
|
Abstract
Oxygen is one of the most commonly used therapeutic agents. Injudicious use of oxygen at high partial pressures (hyperoxia) for unproven indications, its known toxic potential, and the acknowledged roles of reactive oxygen species in tissue injury led to skepticism regarding its use. A large body of data indicates that hyperoxia exerts an extensive profile of physiologic and pharmacologic effects that improve tissue oxygenation, exert anti-inflammatory and antibacterial effects, and augment tissue repair mechanisms. These data set the rationale for the use of hyperoxia in a list of clinical conditions characterized by tissue hypoxia, infection, and consequential impaired tissue repair. Data on regional hemodynamic effects of hyperoxia and recent compelling evidence on its anti-inflammatory actions incited a surge of interest in the potential therapeutic effects of hyperoxia in myocardial revascularization and protection, in traumatic and nontraumatic ischemicanoxic brain insults, and in prevention of surgical site infections and in alleviation of septic and nonseptic local and systemic inflammatory responses. Although the margin of safety between effective and potentially toxic doses of oxygen is relatively narrow, the ability to carefully control its dose, meticulous adherence to currently accepted therapeutic protocols, and individually tailored treatment regimens make it a cost-effective safe drug.
Collapse
Affiliation(s)
- Haim Bitterman
- Department of Internal Medicine, Carmel Medical Center, The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
28
|
Bosco MC, Puppo M, Blengio F, Fraone T, Cappello P, Giovarelli M, Varesio L. Monocytes and dendritic cells in a hypoxic environment: Spotlights on chemotaxis and migration. Immunobiology 2008; 213:733-49. [DOI: 10.1016/j.imbio.2008.07.031] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2008] [Accepted: 07/23/2008] [Indexed: 01/20/2023]
|
29
|
Tsoi EM, Kovalenko RI, Kuz’min DA. Comparative-physiological study of leukocyte participation in initiation of lipid peroxidation during nitrite intoxication in rats and muskrats. J EVOL BIOCHEM PHYS+ 2008. [DOI: 10.1134/s002209300804008x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
30
|
Adenosine A2A receptor antagonists: blockade of adenosinergic effects and T regulatory cells. Br J Pharmacol 2008; 153 Suppl 1:S457-64. [PMID: 18311159 DOI: 10.1038/bjp.2008.23] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The intensity and duration of host responses are determined by protective mechanisms that control tissue injury by dampening down inflammation. Adenosine generation and consequent effects, mediated via A2A adenosine receptors (A2AR) on effector cells, play a critical role in the pathophysiological modulation of these responses in vivo. Adenosine is both released by hypoxic cells/tissues and is also generated from extracellular nucleotides by ecto-enzymes e.g. CD39 (ENTPD1) and CD73 that are expressed by the vasculature and immune cells, in particular by T regulatory cell. In general, these adenosinergic mechanisms minimize the extent of collateral damage to host tissues during the course of inflammatory reactions. However, induction of suppressive pathways might also cause escape of pathogens and permit dissemination. In addition, adenosinergic responses may inhibit immune responses while enhancing vascular angiogenic responses to malignant cells that promote tumor growth. Novel drugs that block A2AR-adenosinergic effects and/or adenosine generation have the potential to boost pathogen destruction and to selectively destroy malignant tissues. In the latter instance, future treatment modalities might include novel 'anti-adenosinergic' approaches that augment immune clearance of malignant cells and block permissive angiogenesis. This review addresses several possible pharmacological modalities to block adenosinergic pathways and speculates on their future application together with impacts on human disease.
Collapse
|
31
|
Lahat N, Rahat MA, Kinarty A, Weiss-Cerem L, Pinchevski S, Bitterman H. Hypoxia enhances lysosomal TNF-alpha degradation in mouse peritoneal macrophages. Am J Physiol Cell Physiol 2008; 295:C2-12. [PMID: 18434619 DOI: 10.1152/ajpcell.00572.2007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Infection, simulated by lipopolysaccharide (LPS), is a potent stimulator of tumor necrosis factor-alpha (TNF-alpha) production, and hypoxia often synergizes with LPS to induce higher levels of the secreted cytokine. However, we show that in primary mouse peritoneal macrophages and in three mouse peritoneal macrophage cell lines (RAW 264.7, J774A.1, and PMJ-2R), hypoxia (O(2) < 0.3%) reduces the secretion of LPS-induced TNF-alpha (P < 0.01). In RAW 264.7 cells this reduction was not regulated transcriptionally as TNF-alpha mRNA levels remained unchanged. Rather, hypoxia and LPS reduced the intracellular levels of TNF-alpha by twofold (P < 0.01) by enhancing its degradation in the lysosomes and inhibiting its secretion via secretory lysosomes, as shown by confocal microscopy and verified by the use of the lysosome inhibitor Bafilomycin A1. In addition, although hypoxia did not change the accumulation of the soluble receptor TNF-RII, it increased its binding to the secreted TNF-alpha by twofold (P < 0.05). We suggest that these two posttranslational regulatory checkpoints coexist in hypoxia and may partially explain the reduced secretion and diminished biological activity of TNF-alpha in hypoxic peritoneal macrophages.
Collapse
Affiliation(s)
- Nitza Lahat
- Immunology Research Unit, Carmel Medical Center, Haifa, Israel
| | | | | | | | | | | |
Collapse
|
32
|
Vuk-Pavlovic S. Rebuilding immunity in cancer patients. Blood Cells Mol Dis 2007; 40:94-100. [PMID: 17827037 PMCID: PMC2225479 DOI: 10.1016/j.bcmd.2007.06.025] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2007] [Revised: 06/30/2007] [Accepted: 06/30/2007] [Indexed: 10/22/2022]
Abstract
Rebuilding and maintaining immunity are paramount to the success of cancer immunotherapy and hematopoietic stem cell transplantation. If immune surveillance indeed can protect from cancer, the very manifestation of malignancy means that the disease has prevailed over immunity. Yet, often, tumor-specific T cells can be found in cancer patients irrespective of vaccination. Interestingly, patients suffering from malignancy often harbor unexpectedly high levels of immature CD14(+)HLA-DR(-) monocytes, although the abundance of CD4(+) cells, CD8(+) cells and CD4(+)CD25(high) cells may be normal. It is plausible that in cancer such cells suppress T cell function, analogous to CD14(+)HLA-DR(-) cells in sepsis and major trauma, in addition to their likely failure to re-present tumor-associated antigens once dendritic cells have initiated the T cell response. Recent evidence indicates that tumor-borne adenosine, lactate and hypoxia in the tumor environment may modulate tumor-specific immunity to a significant extent, but their effects on myeloid cell function are unclear. Thus, understanding and controlling these factors may appreciably impact the success of rebuilding and maintaining immunity in cancer patients.
Collapse
Affiliation(s)
- Stanimir Vuk-Pavlovic
- Department of Internal Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota, USA.
| |
Collapse
|
33
|
From "Hellstrom Paradox" to anti-adenosinergic cancer immunotherapy. Purinergic Signal 2007; 3:129-34. [PMID: 18404426 PMCID: PMC2096757 DOI: 10.1007/s11302-006-9044-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2006] [Accepted: 02/12/2006] [Indexed: 02/06/2023] Open
Abstract
Cancer therapy by endogenous or adoptively transferred anti-tumor T cells is considered complementary to conventional cancer treatment by surgery, radiotherapy or chemotherapy. However, the scope of promising immunotherapeutic protocols is currently limited because tumors can create a ‘hostile–immunosuppressive microenvironment that prevents their destruction by anti-tumor T cells. There is a possibility to develop better and more effective immunotherapies by inactivating mechanisms that inhibit anti-tumor T cells in the tumor microenvironment and thereby protect cancerous tissues from immune damage. This may be now possible because of the recent demonstration that genetic deletion of immunosuppressive A2A and A2B adenosine receptors (A2AR and A2BR) or their pharmacological inactivation can prevent the inhibition of anti-tumor T cells by the hypoxic tumor microenvironment and as a result facilitate full tumor rejection [Ohta A, Gorelik E, Prasad SJ et al (2006) Proc Natl Acad Sci USA 103(35):13132–3137]. This approach is based on in vivo genetic evidence that A2AR play a critical role in the protection of normal tissues from overactive immune cells in acutely inflamed and hypoxic areas. The observations of much improved T-cell-mediated rejection of tumors in mice with inactivated A2AR strongly suggest that A2AR also protects hypoxic cancerous tissues and that A2AR should be inactivated in order to improve tumor rejection by anti-tumor T cells.
Collapse
|
34
|
Gottfried E, Kunz-Schughart LA, Ebner S, Mueller-Klieser W, Hoves S, Andreesen R, Mackensen A, Kreutz M. Tumor-derived lactic acid modulates dendritic cell activation and antigen expression. Blood 2005; 107:2013-21. [PMID: 16278308 DOI: 10.1182/blood-2005-05-1795] [Citation(s) in RCA: 486] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The tumor milieu can influence dendritic cell (DC) differentiation. We analyzed DC differentiation in a 3-dimensional tumor model and propose a new mechanism of DC modulation by the tumor environment. Monocytes were cultured in the presence of IL-4 and GM-CSF within multicellular tumor spheroids (MCTSs) generated from different tumor cell lines. Monocytes invaded the MCTSs and differentiated into tumor-associated dendritic cells (TADCs). The antigen expression was altered on TADCs independent of the culture conditions (immature/mature DCs, Langerhans cells) and IL-12 secretion was reduced. Supernatants of MCTSs could partially transfer the suppressive effect. Conditioned media from urothelial carcinoma cell lines contained high levels of M-CSF and IL-6, both cytokines known to modulate DC differentiation. In contrast, melanoma and prostate carcinoma MCTS cocultures produced little M-CSF and IL-6, but high levels of lactic acid. Indeed, addition of lactic acid during DC differentiation in vitro induced a phenotype comparable with TADCs generated within melanoma and prostate carcinoma MCTSs. Blocking of lactic acid production in melanoma MCTS cocultures reverted the TADC phenotype to normal. We therefore conclude that tumor-derived lactic acid is an important factor modulating the DC phenotype in the tumor environment, which may critically contribute to tumor escape mechanisms.
Collapse
Affiliation(s)
- Eva Gottfried
- Department of Hematology and Oncology, Institute of Pathology, University of Regensburg, Franz-Josef Strauss Allee 11, 93042 Regensburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Ben-Yosef Y, Miller A, Shapiro S, Lahat N. Hypoxia of endothelial cells leads to MMP-2-dependent survival and death. Am J Physiol Cell Physiol 2005; 289:C1321-31. [PMID: 16210427 DOI: 10.1152/ajpcell.00079.2005] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Exposure of endothelial cells (ECs) to hypoxia has separately been shown to induce their angiogenesis or death. Matrix metalloproteinase (MMP)-2 is associated with EC angiogenesis, although recent studies also implicate this molecule in EC death. We studied the effect of hypoxia in the absence or presence of TNF-alpha (characteristic of the inflammatory microenvironment accompanying hypoxia) on MMP-2 expression and its role in angiogenesis (proliferation, migration, and tube formation) and in the death of primary human umbilical vein endothelial cells (HUVECs). Hypoxia alone (24-48 h in 0.3% O(2) in the hypoxic chamber) and furthermore, when combined with TNF-alpha, significantly enhanced MMP-2 expression and activity. Hypoxia also led to a reduction in membrane type 1 MMP (MT1-MMP) and tissue inhibitor of metalloproteinase-2 mRNA and protein while enhancing the expression of alpha(v)beta(3) integrin and the cytoskeletal protein phosphopaxillin. Moreover, hypoxia led to colocalization of alpha(v)beta(3) and MMP-2, but not MT1-MMP, with phosphopaxillin in ECs. These results suggest MT1-MMP-independent activation of MMP-2 during hypoxia and support interactions between the ECM, integrins, and the cytoskeleton in hypoxia-induced MMP-2-related functions. Hypoxia enhanced EC migration in an MMP-2-dependent manner while leading to a reduction of cell number via their apoptosis, which was also dependent on MMP-2. In addition, hypoxia caused an aberrant tubelike formation on Matrigel that appeared to be unaffected by MMP-2. The hypoxia-induced, MMP-2-dependent migration of ECs is in accordance with the proangiogenic role ascribed to MMP-2, while the involvement of this protease in the hypoxia-related death of ECs supports an additional apoptotic role for this protease. Hence, in the hypoxic microenvironment, MMP-2 appears to have a dual autocrine role in determining the fate of ECs.
Collapse
Affiliation(s)
- Yaara Ben-Yosef
- Immunology Research Unit, Carmel Medical Center, 7 Michal St., Haifa 34362, Israel
| | | | | | | |
Collapse
|
36
|
Murdoch C, Muthana M, Lewis CE. Hypoxia Regulates Macrophage Functions in Inflammation. THE JOURNAL OF IMMUNOLOGY 2005; 175:6257-63. [PMID: 16272275 DOI: 10.4049/jimmunol.175.10.6257] [Citation(s) in RCA: 346] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The presence of areas of hypoxia is a prominent feature of various inflamed, diseased tissues, including malignant tumors, atherosclerotic plaques, myocardial infarcts, the synovia of joints with rheumatoid arthritis, healing wounds, and sites of bacterial infection. These areas form when the blood supply is occluded and/or unable to keep pace with the growth and/or infiltration of inflammatory cells in a given area. Macrophages are present in all tissues of the body where they normally assist in guarding against invading pathogens and regulate normal cell turnover and tissue remodeling. However, they are also known to accumulate in large numbers in such ischemic/hypoxic sites. Recent studies show that macrophages then respond rapidly to the hypoxia present by altering their expression of a wide array of genes. In the present study, we outline and compare the phenotypic responses of macrophages to hypoxia in different diseased states and the implications of these for their progression and treatment.
Collapse
Affiliation(s)
- Craig Murdoch
- Tumor Targeting Group, Academic Unit of Pathology, Division of Genomic Medicine, The Henry Wellcome Laboratories for Medical Research, University of Sheffield Medical School, Sheffield S10 2RX
| | | | | |
Collapse
|
37
|
Lewis C, Murdoch C. Macrophage responses to hypoxia: implications for tumor progression and anti-cancer therapies. THE AMERICAN JOURNAL OF PATHOLOGY 2005; 167:627-35. [PMID: 16127144 PMCID: PMC1698733 DOI: 10.1016/s0002-9440(10)62038-x] [Citation(s) in RCA: 312] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/19/2005] [Indexed: 01/22/2023]
Abstract
The presence of multiple areas of hypoxia (low oxygen tension) is a hallmark feature of human and experimental tumors. Monocytes are continually recruited into tumors, differentiate into tumor-associated macrophages (TAMs), and then accumulate in these hypoxic areas. A number of recent studies have shown that macrophages respond to the levels of hypoxia found in tumors by up-regulating such transcription factors as hypoxia-inducible factors 1 and 2, which in turn activate a broad array of mitogenic, pro-invasive, pro-angiogenic, and pro-metastatic genes. This could explain why high numbers of TAMs correlate with poor prognosis in various forms of cancer. In this review, we assess the evidence for hypoxia activating a distinct, pro-tumor phenotype in macrophages and the possible effect of this on the growth, invasion, angiogenesis, and immune evasion of tumors. We also discuss current attempts to selectively target TAMs for destruction or to use them to deliver gene therapy specifically to hypoxic tumor sites.
Collapse
Affiliation(s)
- Claire Lewis
- Tumor Targeting Group, Academic Unit of Pathology, Division of Genomic Medicine, Sir Henry Wellcome Laboratories for Medical Research, University of Sheffield Medical School, Sheffield S10 2RX, UK.
| | | |
Collapse
|
38
|
Luyendyk JP, Shaw PJ, Green CD, Maddox JF, Ganey PE, Roth RA. Coagulation-Mediated Hypoxia and Neutrophil-Dependent Hepatic Injury in Rats Given Lipopolysaccharide and Ranitidine. J Pharmacol Exp Ther 2005; 314:1023-31. [PMID: 15933155 DOI: 10.1124/jpet.105.087981] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Idiosyncrasy-like liver injury occurs in rats cotreated with nonhepatotoxic doses of ranitidine (RAN) and bacterial lipopolysaccharide (LPS). Hepatocellular oncotic necrosis is accompanied by neutrophil (PMN) accumulation and fibrin deposition in LPS/RAN-treated rats, but the contribution of PMNs to injury has not been shown. We tested the hypothesis that PMNs are critical mediators of LPS/RAN-induced liver injury and explored the potential for interaction between PMNs and hemostasis-induced hypoxia. Rats were given either LPS (44.4 x 10(6) endotoxin units/kg) or its vehicle and then RAN (30 mg/kg) or its vehicle 2 h later. They were killed 3 or 6 h after RAN treatment, and hepatocellular injury was estimated from serum alanine aminotransferase activity and liver histopathology. Plasma PMN chemokine concentration and the number of PMNs in liver increased after LPS treatment at 3 h and were not markedly altered by RAN cotreatment. Depletion of circulating PMNs attenuated hepatic PMN accumulation and liver injury and had no effect on coagulation system activation. Anticoagulation with heparin attenuated liver fibrin deposition and injury in LPS/RAN-treated rats; however, heparin had little effect on liver PMN accumulation or plasma chemokine concentration. Liver hypoxia occurred in LPS/RAN-cotreated rats and was significantly reduced by heparin. In vitro, hypoxia enhanced the killing of rat hepatocytes by PMN elastase and shortened its onset, indicating a synergistic interaction between PMNs and hypoxia. The results suggest that PMNs are involved in the hepatocellular injury caused by LPS/RAN-cotreatment and that hemostasis increases sensitivity to PMN-induced hepatocellular injury by causing liver hypoxia.
Collapse
Affiliation(s)
- James P Luyendyk
- Department of Pharmacology and Toxicology, National Food Safety and Toxicology Center, Michigan State University, East Lansing, 48824, USA
| | | | | | | | | | | |
Collapse
|
39
|
Thiel M, Chouker A, Ohta A, Jackson E, Caldwell C, Smith P, Lukashev D, Bittmann I, Sitkovsky MV. Oxygenation inhibits the physiological tissue-protecting mechanism and thereby exacerbates acute inflammatory lung injury. PLoS Biol 2005; 3:e174. [PMID: 15857155 PMCID: PMC1088279 DOI: 10.1371/journal.pbio.0030174] [Citation(s) in RCA: 224] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2004] [Accepted: 03/15/2005] [Indexed: 11/19/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) usually requires symptomatic supportive therapy by intubation and mechanical ventilation with the supplemental use of high oxygen concentrations. Although oxygen therapy represents a life-saving measure, the recent discovery of a critical tissue-protecting mechanism predicts that administration of oxygen to ARDS patients with uncontrolled pulmonary inflammation also may have dangerous side effects. Oxygenation may weaken the local tissue hypoxia-driven and adenosine A2A receptor (A2AR)-mediated anti-inflammatory mechanism and thereby further exacerbate lung injury. Here we report experiments with wild-type and adenosine A2AR-deficient mice that confirm the predicted effects of oxygen. These results also suggest the possibility of iatrogenic exacerbation of acute lung injury upon oxygen administration due to the oxygenation-associated elimination of A2AR-mediated lung tissue-protecting pathway. We show that this potential complication of clinically widely used oxygenation procedures could be completely prevented by intratracheal injection of a selective A2AR agonist to compensate for the oxygenation-related loss of the lung tissue-protecting endogenous adenosine. The identification of a major iatrogenic complication of oxygen therapy in conditions of acute lung inflammation attracts attention to the need for clinical and epidemiological studies of ARDS patients who require oxygen therapy. It is proposed that oxygen therapy in patients with ARDS and other causes of lung inflammation should be combined with anti-inflammatory measures, e.g., with inhalative application of A2AR agonists. The reported observations may also answer the long-standing question as to why the lungs are the most susceptible to inflammatory injury and why lung failure usually precedes multiple organ failure.
Collapse
Affiliation(s)
- Manfred Thiel
- 1Laboratory of Immunology, National Institute of Allergy and Infectious DiseasesNational Institutes of Health, Bethesda, MarylandUnited States of America
- 2Clinic of AnaesthesiologyUniversity of MunichGermany
| | - Alexander Chouker
- 1Laboratory of Immunology, National Institute of Allergy and Infectious DiseasesNational Institutes of Health, Bethesda, MarylandUnited States of America
- 2Clinic of AnaesthesiologyUniversity of MunichGermany
| | - Akio Ohta
- 1Laboratory of Immunology, National Institute of Allergy and Infectious DiseasesNational Institutes of Health, Bethesda, MarylandUnited States of America
- 3New England Inflammation and Tissue Protection Institute, Northeastern UniversityBoston, MassachusettsUnited States of America
| | - Edward Jackson
- 4Pharmacology/Medicine Pittsburgh, University of Pittsburgh School of MedicinePennsylvaniaUnited States of America
| | - Charles Caldwell
- 1Laboratory of Immunology, National Institute of Allergy and Infectious DiseasesNational Institutes of Health, Bethesda, MarylandUnited States of America
| | - Patrick Smith
- 1Laboratory of Immunology, National Institute of Allergy and Infectious DiseasesNational Institutes of Health, Bethesda, MarylandUnited States of America
| | - Dmitry Lukashev
- 1Laboratory of Immunology, National Institute of Allergy and Infectious DiseasesNational Institutes of Health, Bethesda, MarylandUnited States of America
- 3New England Inflammation and Tissue Protection Institute, Northeastern UniversityBoston, MassachusettsUnited States of America
| | - Iris Bittmann
- 5Pathology, Klinikum GrosshadernUniversity of MunichGermany
| | - Michail V Sitkovsky
- 1Laboratory of Immunology, National Institute of Allergy and Infectious DiseasesNational Institutes of Health, Bethesda, MarylandUnited States of America
- 3New England Inflammation and Tissue Protection Institute, Northeastern UniversityBoston, MassachusettsUnited States of America
| |
Collapse
|
40
|
|
41
|
Vengellur A, Woods BG, Ryan HE, Johnson RS, Lapres JJ. Gene expression profiling of the hypoxia signaling pathway in hypoxia-inducible factor 1alpha null mouse embryonic fibroblasts. Gene Expr 2003; 11:181-97. [PMID: 14686790 PMCID: PMC5991159 DOI: 10.3727/000000003108749062] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2003] [Indexed: 12/25/2022]
Abstract
Hypoxia is defined as a deficiency of oxygen reaching the tissues of the body, and it plays a critical role in development and pathological conditions, such as cancer. Once tumors outgrow their blood supply, their central portion becomes hypoxic and the tumor stimulates angiogenesis through the activation of the hypoxia-inducible factors (HIFs). HIFs are transcription factors that are regulated in an oxygen-dependent manner by a group of prolyl hydroxylases (known as PHDs or HPHs). Our understanding of hypoxia signaling is limited by our incomplete knowledge of HIF target genes. cDNA microarrays and a cell line lacking a principal HIF protein, HIF1alpha, were used to identify a more complete set of hypoxia-regulated genes. The microarrays identified a group of 286 clones that were significantly influenced by hypoxia and 54 of these were coordinately regulated by cobalt chloride. The expression profile of HIF1alpha -/- cells also identified a group of downregulated genes encoding enzymes involved in protecting cells from oxidative stress, offering an explanation for the increased sensitivity of HIF1alpha -/- cells to agents that promote this type of response. The microarray studies confirmed the hypoxia-induced expression of the HIF regulating prolyl hydroxylase, PHD2. An analysis of the members of the PHD family revealed that they are differentially regulated by cobalt chloride and hypoxia. These results suggest that HIF1alpha is the predominant isoform in fibroblasts and that it regulates a wide battery of genes critical for normal cellular function and survival under various stresses.
Collapse
Affiliation(s)
- Ajith Vengellur
- *Department of Biochemistry and Molecular Biology and The National Food Safety and Toxicology Center, Michigan State University, East Lansing, MI 48824
| | - Barbara G. Woods
- *Department of Biochemistry and Molecular Biology and The National Food Safety and Toxicology Center, Michigan State University, East Lansing, MI 48824
| | - Heather E. Ryan
- †Molecular Biology Section, Division of Biological Sciences, University of California, San Diego, CA 92093
| | - Randall S. Johnson
- †Molecular Biology Section, Division of Biological Sciences, University of California, San Diego, CA 92093
| | - John J. Lapres
- *Department of Biochemistry and Molecular Biology and The National Food Safety and Toxicology Center, Michigan State University, East Lansing, MI 48824
- Address correspondence to John J. LaPres, 402 Biochemistry Building, Michigan State University, East Lansing, MI 48824-1319. Tel: (517) 432-9282; Fax: (517) 353-9334; E-mail:
| |
Collapse
|