1
|
Green KA, Ma C, Hoffmann FW, Hoffmann PR, Green WR. Depletion of monocytic myeloid-derived suppressor cells in LP-BM5 murine retroviral infection has a positive impact on virus-induced host immunodeficiency. Virology 2024; 600:110247. [PMID: 39307098 PMCID: PMC11560480 DOI: 10.1016/j.virol.2024.110247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 08/29/2024] [Accepted: 09/16/2024] [Indexed: 11/12/2024]
Abstract
We have shown the induction of CD11b+Ly6C+ monocytic myeloid-derived suppressor cells (M-MDSCs) during infection of B6 mice by LP-BM5 immunodeficiency-inducing retrovirus. We published that the molecular mechanisms of these M-MDSCs vary, and depend on the cell type targeted by the suppression -defined by use of biochemical inhibitors, mouse M-MDSCs knock-out strains and blocking antibodies. These M-MDSCs suppressed proliferation and function of T cells, via nitric oxide synthase/nitric oxide; and that of B cells, ∼50% via INOS/NO along with the negative checkpoint regulator VISTA, reactive nitrogen and oxygen species, and other soluble mediators. Here, LP-BM5 infected mice were treated weekly with 5-Fluorouracil (5-FU), resulting in depletion of peripheral blood and splenic M-MDSCs, reduced MDSC activity, and significantly decreased standard disease parameters of: splenomegaly, impaired B-and T-cell ex vivo polyclonal responses, and viral load. In addition, 5-FU treatment significantly increased percentages of CD4+ and CD8+ T cells.
Collapse
Affiliation(s)
- Kathy A Green
- Geisel School of Medicine at Dartmouth, Lebanon, NH, 03756, USA.
| | - Chi Ma
- John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, 96813, USA
| | - Fukun W Hoffmann
- John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, 96813, USA
| | - Peter R Hoffmann
- John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, 96813, USA
| | - William R Green
- Geisel School of Medicine at Dartmouth, Lebanon, NH, 03756, USA
| |
Collapse
|
2
|
Ma C, Hoffmann FW, Shay AE, Koo I, Green KA, Green WR, Hoffmann PR. Upregulated selenoprotein I during lipopolysaccharide-induced B cell activation promotes lipidomic changes and is required for effective differentiation into IgM-secreting plasma B cells. J Leukoc Biol 2024; 116:6-17. [PMID: 38289835 PMCID: PMC11212798 DOI: 10.1093/jleuko/qiae024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/17/2024] [Accepted: 01/19/2024] [Indexed: 02/01/2024] Open
Abstract
The mechanisms driving metabolic reprogramming during B cell activation are unclear, particularly roles for enzymatic pathways involved in lipid remodeling. We found that murine B cell activation with lipopolysaccharide (LPS) led to a 1.6-fold increase in total lipids that included higher levels of phosphatidylethanolamine (PE) and plasmenyl PE. Selenoprotein I (SELENOI) is an ethanolamine phospholipid transferase involved in the synthesis of both PE and plasmenyl PE, and SELENOI expression was also upregulated during activation. Selenoi knockout (KO) B cells exhibited decreased levels of plasmenyl PE, which plays an important antioxidant role. Lipid peroxidation was measured and found to increase ∼2-fold in KO vs. wild-type (WT) B cells. Cell death was not impacted by KO in LPS-treated B cells and proliferation was only slightly reduced, but differentiation into CD138 + Blimp-1+ plasma B cells was decreased ∼2-fold. This led to examination of B cell receptors important for differentiation that recognize the ligand B cell activating factor, and levels of TACI (transmembrane activator, calcium-modulator, and cytophilin ligand interactor) (CD267) were significantly decreased on KO B cells compared with WT control cells. Vaccination with ovalbumin/adjuvant led to decreased ovalbumin-specific immunoglobulin M (IgM) levels in sera of KO mice compared with WT mice. Real-time polymerase chain reaction analyses revealed a decreased switch from surface to secreted IgM in spleens of KO mice induced by vaccination or LP-BM5 retrovirus infection. Overall, these findings detail the lipidomic response of B cells to LPS activation and reveal the importance of upregulated SELENOI for promoting differentiation into IgM-secreting plasma B cells.
Collapse
Affiliation(s)
- Chi Ma
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, 651 Ilalo Street, Honolulu, HI 96813, United States
| | - FuKun W Hoffmann
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, 651 Ilalo Street, Honolulu, HI 96813, United States
| | - Ashley E Shay
- Huck Institutes of the Life Sciences, The Pennsylvania State University, 101 Huck Life Sciences Building, University Park, PA 16802, United States
| | - Imhoi Koo
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, 107 Animal, Veterinary, and Biomedical Sciences Building, University Park, PA 16802, United States
| | - Kathy A Green
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, One Medical Center Drive HB7556, Lebanon, NH 03756, United States
| | - William R Green
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, One Medical Center Drive HB7556, Lebanon, NH 03756, United States
| | - Peter R Hoffmann
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, 651 Ilalo Street, Honolulu, HI 96813, United States
| |
Collapse
|
3
|
Mahanti K, Saha J, Sarkar D, Pramanik A, Roy Chattopadhyay N, Bhattacharyya S. Alteration of functionality and differentiation directed by changing gene expression patterns in myeloid-derived suppressor cells (MDSCs) in tumor microenvironment and bone marrow through early to terminal phase of tumor progression. J Leukoc Biol 2024; 115:958-984. [PMID: 38236200 DOI: 10.1093/jleuko/qiae013] [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: 09/14/2023] [Revised: 12/06/2023] [Accepted: 01/04/2024] [Indexed: 01/19/2024] Open
Abstract
Myeloid-derived suppressor cells are heterogenous immature myeloid lineage cells that can differentiate into neutrophils, monocytes, and dendritic cells as well. These cells have been characterized to have potent immunosuppressive capacity in neoplasia and a neoplastic chronic inflammatory microenvironment. Increased accumulation of myeloid-derived suppressor cells was reported with poor clinical outcomes in patients. They support neoplastic progression by abrogating antitumor immunity through inhibition of lymphocyte functions and directly by facilitating tumor development. Yet the shifting genetic signatures of this myeloid lineage cell toward immunosuppressive functionality in progressive tumor development remain elusive. We have attempted to identify the gene expression profile using lineage-specific markers of these unique myeloid lineage cells in a tumor microenvironment and bone marrow using a liquid transplantable mice tumor model to trace the changing influence of the tumor microenvironment on myeloid-derived suppressor cells. We analyzed the phenotype, functional shift, suppressive activity, differentiation status, and microarray-based gene expression profile of CD11b+Gr1+ lineage-specific cells isolated from the tumor microenvironment and bone marrow of 4 stages of tumor-bearing mice and compared them with control counterparts. Our analysis of differentially expressed genes of myeloid-derived suppressor cells isolated from bone marrow and the tumor microenvironment reveals unique gene expression patterns in the bone marrow and tumor microenvironment-derived myeloid-derived suppressor cells. It also suggests T-cell suppressive activity of myeloid-derived suppressor cells progressively increases toward the mid-to-late phase of the tumor and a significant differentiation bias of tumor site myeloid-derived suppressor cells toward macrophages, even in the presence of differentiating agents, indicating potential molecular characteristics of myeloid-derived suppressor cells in different stages of the tumor that can emerge as an intervention target.
Collapse
Affiliation(s)
- Krishna Mahanti
- Immunobiology and Translational Medicine Laboratory, Department of Zoology, Sidho Kanho Birsha University, Ranchi Road, Saink School, Purulia, West Bengal 723104, India
| | - Jayasree Saha
- Immunobiology and Translational Medicine Laboratory, Department of Zoology, Sidho Kanho Birsha University, Ranchi Road, Saink School, Purulia, West Bengal 723104, India
- Currently, DST-SERB NPDF, School of Bioscience, IIT Kharagpur, Paschim Medinipur, West Bengal 721302, India
| | - Debanjan Sarkar
- Immunobiology and Translational Medicine Laboratory, Department of Zoology, Sidho Kanho Birsha University, Ranchi Road, Saink School, Purulia, West Bengal 723104, India
| | - Anik Pramanik
- Immunobiology and Translational Medicine Laboratory, Department of Zoology, Sidho Kanho Birsha University, Ranchi Road, Saink School, Purulia, West Bengal 723104, India
| | - Nabanita Roy Chattopadhyay
- Department of Biotechnology, Siksha Bhaban, Visva Bharati, Shantiniketan, Birbhum, West Bengal 731235, India
- Currently, Department of Biotechnology, Haldia Institute of Technology, ICARE Complex, Haldia, West Bengal 721657, India
| | - Sankar Bhattacharyya
- Immunobiology and Translational Medicine Laboratory, Department of Zoology, Sidho Kanho Birsha University, Ranchi Road, Saink School, Purulia, West Bengal 723104, India
| |
Collapse
|
4
|
Fraternale A, Green KA, Schiavano GF, Bruschi M, Retini M, Magnani M, Green WR. Inhibition of myeloid-derived suppressor cell (MDSC) activity by redox-modulating agents restores T and B cell proliferative responses in murine AIDS. Int Immunopharmacol 2023; 124:110882. [PMID: 37659111 DOI: 10.1016/j.intimp.2023.110882] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 08/28/2023] [Accepted: 08/28/2023] [Indexed: 09/04/2023]
Abstract
The mechanisms by which myeloid-derived suppressor cells (MDSCs) mediate inhibition prominently include the production of reactive nitrogen species, in particular those generated by inducible nitric oxide synthase (iNOS), and reactive oxygen species. LP-BM5 murine retroviral infection results in a profound immunodeficiency, known as murine AIDS, as well as in increased numbers and activity of monocytic-type MDSCs (M-MDSCs) that suppress both T and B cell responses. While M-MDSCs suppress T cells ex vivo in a fully iNOS/NO-dependent manner, M-MDSC suppression of B cell responses is only partially due to iNOS/NO. This study preliminarily explored the role of two redox-modulating compounds in inhibiting the M-MDSC suppressive activity in LP-BM5 infection. The tested molecules were: I-152 consisting in a conjugate of N-acetyl-cysteine (NAC) and S-acetyl-cysteamine (SMEA) and C4-GSH that is the n-butanoyl glutathione (GSH) derivative. The results show that both molecules, tested in a concentration range between 3 and 20 mM, blocked the M-MDSC suppression of activated B and T cells ex vivo and restored their proliferative capacity in vivo. Ex vivo I-152 blockade of M-MDSC suppressiveness was more significant for T cell (about 70%) while M-MDSC blockade by C4-GSH was preferential for B cell responsiveness (about 60%), which was also confirmed by in vivo investigation. Beyond insights into redox-dependent suppressive effector mechanism(s) of M-MDSCs in LP-BM5 infection, these findings may ultimately be important to identify new immunotherapeutics against infectious diseases.
Collapse
Affiliation(s)
- Alessandra Fraternale
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, Italy.
| | - Kathy A Green
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, NH, United States
| | | | - Michela Bruschi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, Italy
| | - Michele Retini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, Italy
| | - Mauro Magnani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, Italy
| | - William R Green
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, NH, United States
| |
Collapse
|
5
|
Borgna E, Prochetto E, Gamba JC, Marcipar I, Cabrera G. Role of myeloid-derived suppressor cells during Trypanosoma cruzi infection. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 375:117-163. [PMID: 36967151 DOI: 10.1016/bs.ircmb.2022.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
Chagas disease (CD), caused by the protozoan parasite Trypanosoma cruzi, is the third largest parasitic disease burden globally. Currently, more than 6 million people are infected, mainly in Latin America, but international migration has turned CD into an emerging health problem in many nonendemic countries. Despite intense research, a vaccine is still not available. A complex parasite life cycle, together with numerous immune system manipulation strategies, may account for the lack of a prophylactic or therapeutic vaccine. There is substantial experimental evidence supporting that T. cruzi acute infection generates a strong immunosuppression state that involves numerous immune populations with regulatory/suppressive capacity. Myeloid-derived suppressor cells (MDSCs), Foxp3+ regulatory T cells (Tregs), regulatory dendritic cells and B regulatory cells are some of the regulatory populations that have been involved in the acute immune response elicited by the parasite. The fact that, during acute infection, MDSCs increase notably in several organs, such as spleen, liver and heart, together with the observation that depletion of those cells can decrease mouse survival to 0%, strongly suggests that MDSCs play a major role during acute T. cruzi infection. Accumulating evidence gained in different settings supports the capacity of MDSCs to interact with cells from both the effector and the regulatory arms of the immune system, shaping the outcome of the response in a very wide range of scenarios that include pathological and physiological processes. In this sense, the aim of the present review is to describe the main knowledge about MDSCs acquired so far, including several crosstalk with other immune populations, which could be useful to gain insight into their role during T. cruzi infection.
Collapse
|
6
|
Stevenson MM, Valanparambil RM, Tam M. Myeloid-Derived Suppressor Cells: The Expanding World of Helminth Modulation of the Immune System. Front Immunol 2022; 13:874308. [PMID: 35757733 PMCID: PMC9229775 DOI: 10.3389/fimmu.2022.874308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 05/02/2022] [Indexed: 01/09/2023] Open
Abstract
Infection with helminths or parasitic worms are highly prevalent worldwide especially in developing regions. Helminths cause chronic infections that are associated with suppression of immune responses to unrelated pathogens, vaccines, and by-stander antigens responsible for dysregulated immune responses as occurs in diseases such as allergies. Helminths use multiple mechanisms to modulate the immune system to evade the highly polarized type 2 immune response required to expel adult worms and for immunity to reinfection. Anthelmintic drugs are efficient in reducing adult worm burdens in helminth-infected individuals, but resistance to these drugs is rapidly increasing and vaccines against these pathogens are not available. Emerging evidence indicate that helminths induce myeloid-derived suppressor cells (MDSC), originally described in tumor-bearing mice and cancer patients. MDSC are a heterogenous population of immature cells that consist of two distinct sub-populations, polymorphonuclear (PMN)-MDSC and monocytic (M)-MDSC based on morphology and phenotype. MDSC suppress the function of T cells and other innate and adaptive immune cells including NK cells and B cells. During cancer or infection with bacteria or viruses, there is marked expansion of MDSC. Furthermore, the frequencies of MDSC correlate inversely with the prognosis and survival of tumor-bearing hosts as well as bacterial and viral burdens, persistence, and outcome in infected hosts. Currently, there is a paucity of data on MDSC and helminth infections. Here, we provide a survey of the evidence accumulated so far that overall support a role for MDSC in modulating immune responses during helminth infections. We review data from studies in various helminths, including those that infect humans. Finally, we summarize the progress to date in understanding the role of MDSC in helminth infections and briefly discuss potential host-directed strategies to target MDSC-mediated suppression of immune responses to helminths in favor of development of immunity to eliminate adult worms and possibly induce protection against reinfection.
Collapse
Affiliation(s)
- Mary M Stevenson
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada.,Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, QC, Canada
| | - Rajesh M Valanparambil
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, QC, Canada
| | - Mifong Tam
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| |
Collapse
|
7
|
Jiménez-Cortegana C, Galassi C, Klapp V, Gabrilovich DI, Galluzzi L. Myeloid-Derived Suppressor Cells and Radiotherapy. Cancer Immunol Res 2022; 10:545-557. [DOI: 10.1158/2326-6066.cir-21-1105] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 01/21/2022] [Accepted: 02/22/2022] [Indexed: 11/16/2022]
Abstract
Abstract
Myeloid-derived suppressor cells (MDSC) are a heterogeneous population of pathologically activated, mostly immature, myeloid cells that exert robust immunosuppressive functions. MDSCs expand during oncogenesis and have been linked to accelerated disease progression and resistance to treatment in both preclinical tumor models and patients with cancer. Thus, MDSCs stand out as promising targets for the development of novel immunotherapeutic regimens with superior efficacy. Here, we summarize accumulating preclinical and clinical evidence indicating that MDSCs also hamper the efficacy of radiotherapy (RT), as we critically discuss the potential of MDSC-targeting strategies as tools to achieve superior immunotherapeutic tumor control by RT in the clinic.
Collapse
Affiliation(s)
- Carlos Jiménez-Cortegana
- Department of Radiation Oncology, Weill Cornell Medical College, New York, New York
- Department of Medical Biochemistry, Molecular Biology and Immunology, Faculty of Medicine, University of Seville, Seville, Spain
| | - Claudia Galassi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, New York
| | - Vanessa Klapp
- Department of Radiation Oncology, Weill Cornell Medical College, New York, New York
| | | | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, New York
- Sandra and Edward Meyer Cancer Center, New York, New York
- Caryl and Israel Englander Institute for Precision Medicine, New York, New York
| |
Collapse
|
8
|
Tamadaho RSE, Ritter M, Wiszniewsky A, Arndts K, Mack M, Hoerauf A, Layland LE. Infection-Derived Monocytic MDSCs Require TGF-β to Suppress Filarial-Specific IFN-γ But Not IL-13 Release by Filarial-Specific CD4+ T Cells In Vitro. FRONTIERS IN TROPICAL DISEASES 2022. [DOI: 10.3389/fitd.2021.707100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Lymphatic filariasis (LF) remains a major health problem with severe economic repercussions in endemic communities of Sub-saharan Africa, South-East Asia and South America. The rodent-specific nematode Litomosoides sigmodontis (Ls) is used to study the immunomodulatory potential of filariae and research has elucidated pathways involving regulatory T cells (Tregs), IL-10 producing cells and alternatively activated macrophages (AAMs) and that CD4+ T cells play a paramount role during infection. Myeloid-derived suppressor cells (MDSCs) have been identified and characterised in man in cancer and other pathologies. The hallmark of MDSC populations is the suppression of T and B cell responses using various mechanisms, which are mostly specific to the pathology or setting. However, until now, it remains unclear whether they play a role in filarial-specific responses. We report here that monocytic MDSCs (Mo-MDSCs, CD11b+Ly6C+Ly6G-) and polymorphonuclear MDSCs (PMN-MDSCs, CD11b+Ly6Cint/loLy6G+) expanded in the thoracic cavity (TC, the site of infection) and correlated positively with filarial life-stages in Ls-infected BALB/c mice. In vitro, only infection-derived Mo-MDSCs showed a suppressive nature by preventing IL-13 and IFN-γ secretion from filarial-specific CD4+ T cells upon co-culture with soluble worm extract. This suppression was not mediated by IL-10, IL-6 or TNF-α, and did not require cell-contact, nitric oxide (NO), IL-4/IL-5 signalling pathways or CCR2. Interestingly, neutralizing TGF-β significantly rescued IFN-γ but not IL-13 production by filarial-specific CD4+ T cells. In comparison to naive cells, PCR array data showed an overall down-regulation of inflammatory pathways in both infection-derived Mo-MDSCs and PMN-MDSCs. In conclusion, these primary data sets show activity and expansion of MDSCs during Ls infection adding this regulatory cell type to the complex milieu of host responses during chronic helminth infections.
Collapse
|
9
|
Arabatzis TJ, Wakley AA, McLane VD, Canonico D, Cao L. Effects of HIV gp120 on Neuroinflammation in Immunodeficient vs. Immunocompetent States. J Neuroimmune Pharmacol 2021; 16:437-453. [PMID: 32627098 PMCID: PMC7785647 DOI: 10.1007/s11481-020-09936-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 06/17/2020] [Indexed: 11/30/2022]
Abstract
HIV affects 37 million people worldwide, 25-69% of which develop HIV-associated neurocognitive disorders (HAND) regardless of antiviral treatment. HIV infection of the brain decreases cognitive function, disrupts/impairs learning and memory, and reduces quality of life for those affected. HIV-induced neuroinflammation has been associated with viral proteins such as gp120 and Tat, which remain elevated in the CNS even in patients with low peripheral viremia counts. In this study, we examined the effects of gp120 on neuroinflammation in immunodeficient vs. immunocompetent states by examining neuroinflammatory markers in gp120tg mice with or without systemic immunodeficiency caused by murine retroviral administration (LP-BM5 murine AIDS). Changes in inflammatory cytokine/chemokine mRNA expression was complex and dependent upon expression of gp120 protein, immunodeficiency status, brain region (hippocampus, frontal lobe, or striatum), and age. Gp120 expression reduced hippocampal synaptophysin expression but did not affect animals' learning/memory on the spontaneous T-maze test in our experimental conditions. Our results emphasize the critical role of the neuroinflammatory micro-environment and the peripheral immune system context in which gp120 acts. Multiple factors, particularly system-level differences in the immune response of different brain regions, need to be considered when developing treatment for HAND. Graphical Abstract.
Collapse
Affiliation(s)
- Taxiarhia J Arabatzis
- Department of Biomedical Sciences, University of New England College of Osteopathic Medicine, 11 Hills Beach Road, Biddeford, ME, 04005, USA
- Department of Biology, University of New England College of Arts and Sciences, Biddeford, ME, USA
| | - Alexa A Wakley
- Department of Biomedical Sciences, University of New England College of Osteopathic Medicine, 11 Hills Beach Road, Biddeford, ME, 04005, USA
| | - Virginia D McLane
- Department of Biomedical Sciences, University of New England College of Osteopathic Medicine, 11 Hills Beach Road, Biddeford, ME, 04005, USA
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, P.O. Box 980613, Richmond, VA, 23298-0613, USA
| | - Dalton Canonico
- Department of Biology, University of New England College of Arts and Sciences, Biddeford, ME, USA
| | - Ling Cao
- Department of Biomedical Sciences, University of New England College of Osteopathic Medicine, 11 Hills Beach Road, Biddeford, ME, 04005, USA.
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA.
| |
Collapse
|
10
|
Boosting GSH Using the Co-Drug Approach: I-152, a Conjugate of N-acetyl-cysteine and β-mercaptoethylamine. Nutrients 2019; 11:nu11061291. [PMID: 31181621 PMCID: PMC6627109 DOI: 10.3390/nu11061291] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 06/04/2019] [Accepted: 06/05/2019] [Indexed: 12/22/2022] Open
Abstract
Glutathione (GSH) has poor pharmacokinetic properties; thus, several derivatives and biosynthetic precursors have been proposed as GSH-boosting drugs. I-152 is a conjugate of N-acetyl-cysteine (NAC) and S-acetyl-β-mercaptoethylamine (SMEA) designed to release the parent drugs (i.e., NAC and β-mercaptoethylamine or cysteamine, MEA). NAC is a precursor of L-cysteine, while MEA is an aminothiol able to increase GSH content; thus, I-152 represents the very first attempt to combine two pro-GSH molecules. In this review, the in-vitro and in-vivo metabolism, pro-GSH activity and antiviral and immunomodulatory properties of I-152 are discussed. Under physiological GSH conditions, low I-152 doses increase cellular GSH content; by contrast, high doses cause GSH depletion but yield a high content of NAC, MEA and I-152, which can be used to resynthesize GSH. Preliminary in-vivo studies suggest that the molecule reaches mouse organs, including the brain, where its metabolites, NAC and MEA, are detected. In cell cultures, I-152 replenishes experimentally depleted GSH levels. Moreover, administration of I-152 to C57BL/6 mice infected with the retroviral complex LP-BM5 is effective in contrasting virus-induced GSH depletion, exerting at the same time antiviral and immunomodulatory functions. I-152 acts as a pro-GSH agent; however, GSH derivatives and NAC cannot completely replicate its effects. The co-delivery of different thiol species may lead to unpredictable outcomes, which warrant further investigation.
Collapse
|
11
|
Dorhoi A, Glaría E, Garcia-Tellez T, Nieuwenhuizen NE, Zelinskyy G, Favier B, Singh A, Ehrchen J, Gujer C, Münz C, Saraiva M, Sohrabi Y, Sousa AE, Delputte P, Müller-Trutwin M, Valledor AF. MDSCs in infectious diseases: regulation, roles, and readjustment. Cancer Immunol Immunother 2019; 68:673-685. [PMID: 30569204 PMCID: PMC11028159 DOI: 10.1007/s00262-018-2277-y] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 10/29/2018] [Indexed: 12/24/2022]
Abstract
Many pathogens, ranging from viruses to multicellular parasites, promote expansion of MDSCs, which are myeloid cells that exhibit immunosuppressive features. The roles of MDSCs in infection depend on the class and virulence mechanisms of the pathogen, the stage of the disease, and the pathology associated with the infection. This work compiles evidence supported by functional assays on the roles of different subsets of MDSCs in acute and chronic infections, including pathogen-associated malignancies, and discusses strategies to modulate MDSC dynamics to benefit the host.
Collapse
Affiliation(s)
- Anca Dorhoi
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer 10, 17493, Greifswald, Insel Riems, Germany.
- Faculty of Mathematics and Natural Sciences, University of Greifswald, Greifswald, Germany.
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany.
| | - Estibaliz Glaría
- Nuclear Receptor Group, Department of Cell Biology, Physiology and Immunology, School of Biology, University of Barcelona, Av. Diagonal, 643, 3rd floor, 08028, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain
| | | | | | - Gennadiy Zelinskyy
- Institute of Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Benoit Favier
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, CEA, Université Paris Sud 11, INSERM U1184, IBJF, Fontenay-aux-Roses, France
| | - Anurag Singh
- University Children's Hospital and Interdisciplinary Center for Infectious Diseases, University of Tübingen, Tübingen, Germany
| | - Jan Ehrchen
- Department of Dermatology, University Hospital Münster, Münster, Germany
| | - Cornelia Gujer
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zurich, Switzerland
| | - Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zurich, Switzerland
| | - Margarida Saraiva
- i3S-Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Yahya Sohrabi
- Molecular and Translational Cardiology, Department of Cardiovascular Medicine, University Hospital Münster, Münster, Germany
- Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Ana E Sousa
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Peter Delputte
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | | | - Annabel F Valledor
- Nuclear Receptor Group, Department of Cell Biology, Physiology and Immunology, School of Biology, University of Barcelona, Av. Diagonal, 643, 3rd floor, 08028, Barcelona, Spain.
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain.
| |
Collapse
|
12
|
Ochando J, Conde P, Utrero-Rico A, Paz-Artal E. Tolerogenic Role of Myeloid Suppressor Cells in Organ Transplantation. Front Immunol 2019; 10:374. [PMID: 30894860 PMCID: PMC6414442 DOI: 10.3389/fimmu.2019.00374] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 02/14/2019] [Indexed: 01/10/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSC) are a heterogeneous population of immature cells of myeloid origin with a specific immune inhibitory function that negatively regulates the adaptive immune response. Since MDSC participate in the promotion of tolerance in the context of organ transplantation, therapeutic strategies that regulate the induction and development of MDSC have been the center of scientist attention. Here we review literature regarding induction of MDSC with demonstrated suppressive function among different types of allografts and their mechanism of action. While manipulation of MDSC represents a potential therapeutic approach for the promotion of donor specific tolerance in solid organ transplantation, further characterization of their specific phenotype, which distinguishes MDSC from non-suppressive myeloid cells, and detailed evaluation of the inhibitory mechanism that determines their suppressive function, is necessary for the realistic application of MDSC as biomarkers in health and disease and their potential use as immune cell therapy in organ transplantation.
Collapse
Affiliation(s)
- Jordi Ochando
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Immunología de Trasplantes, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Madrid, Spain
| | - Patricia Conde
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Immunología de Trasplantes, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Madrid, Spain
| | - Alberto Utrero-Rico
- Grupo de Inmunodeficiencias e Inmunología del Trasplante, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Estela Paz-Artal
- Grupo de Inmunodeficiencias e Inmunología del Trasplante, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain.,School of Medicine, Complutense University, Madrid, Spain
| |
Collapse
|
13
|
Infection of B Cell Follicle-Resident Cells by Friend Retrovirus Occurs during Acute Infection and Is Maintained during Viral Persistence. mBio 2019; 10:mBio.00004-19. [PMID: 30782653 PMCID: PMC6381274 DOI: 10.1128/mbio.00004-19] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
B cell follicles of the spleen and lymph nodes are immune privileged sites and serve as sanctuaries for infected CD4+ cells in HIV infection. It is assumed that CD8+ T cell responses promote the establishment of the reservoir, as B cell follicles do not permit CD8+ T cell entry. Here we analyzed the infected cell population in the Friend retrovirus (FV) infection and investigated whether FV can similarly infect follicular cells. For analysis of FV-infected cells, we constructed a recombinant FV encoding the bright fluorescent protein mWasabi and performed flow cytometry with cells isolated from spleens, lymph nodes and bone marrow of FV-mWasabi-infected mice. Using t-stochastic neighbor embedding for data exploration, we demonstrate how the target cell population changes during the course of infection. While FV was widely distributed in erythrocytes, myeloid cells, B cells, and CD4+ T cells in the acute phase of infection, the bulk viral load in the late phase was carried by macrophages and follicular B and CD4+ T cells, suggesting that FV persists in cells that are protected from CD8+ T cell killing. Importantly, seeding into follicular cells was equally observed in CD8+ T cell-depleted mice and in highly FV-susceptible mice that mount a very weak immune response, demonstrating that infection of follicular cells is not driven by immune pressure. Our data demonstrate that infection of cells in the B cell follicle is a characteristic of the FV infection, making this murine retrovirus an even more valuable model for development of retrovirus immunotherapy approaches.IMPORTANCE Human immunodeficiency virus is notorious for its ability to avoid clearance by therapeutic interventions, which is partly attributed to the establishment of reservoirs in latently infected cells and cells that reside in immunologically privileged B cell follicles. In the work presented here, we show that cells of the B cell follicle are equally infected by a simple mouse gammaretrovirus. Using fluorescently labeled Friend retrovirus, we found that B cells and T cells in the B cell follicle, while not carrying the bulk of the virus load, were indeed infected by Friend virus in the early acute phase of the infection and persisted in the chronic infection. Our results suggest that infection of follicular cells may be a shared property of lymphotropic viruses and propose the FV infection of mice as a useful model to study strategies for follicular reservoir elimination.
Collapse
|
14
|
Ohl K, Tenbrock K. Reactive Oxygen Species as Regulators of MDSC-Mediated Immune Suppression. Front Immunol 2018; 9:2499. [PMID: 30425715 PMCID: PMC6218613 DOI: 10.3389/fimmu.2018.02499] [Citation(s) in RCA: 247] [Impact Index Per Article: 41.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 10/10/2018] [Indexed: 01/10/2023] Open
Abstract
Reactive oxygen species (ROS) molecules are implicated in signal transduction pathways and thereby control a range of biological activities. Immune cells are constantly confronted with ROS molecules under both physiologic and pathogenic conditions. Myeloid-derived suppressor cells (MDSCs) are immunosuppressive, immature myeloid cells and serve as major regulators of pathogenic and inflammatory immune responses. In addition to their own release of ROS, MDSCs often arise in oxidative-stress prone environments such as in tumors or during inflammation and infection. This evidently close relationship between MDSCs and ROS prompted us to summarize what is currently known about ROS signaling within MDSCs and to elucidate how MDSCs use ROS to modulate other immune cells. ROS not only activate anti-oxidative pathways but also induce transcriptional programs that regulate the fate and function of MDSCs. Furthermore, MDSCs release ROS molecules as part of a major mechanism to suppress T cell responses. Targeting redox-regulation of MDSCs thus presents a promising approach to cancer therapy and the role of redox-signaling in MDSCs in other disease states such as infection, inflammation and autoimmunity would appear to be well worth investigating.
Collapse
Affiliation(s)
- Kim Ohl
- Department of Pediatrics, Medical Faculty, RWTH Aachen, Aachen, Germany
| | - Klaus Tenbrock
- Department of Pediatrics, Medical Faculty, RWTH Aachen, Aachen, Germany
| |
Collapse
|
15
|
Payne KK, Aqbi HF, Butler SE, Graham L, Keim RC, Wan W, Idowu MO, Bear HD, Wang XY, Manjili MH. Gr1 -/low CD11b -/low MHCII + myeloid cells boost T cell anti-tumor efficacy. J Leukoc Biol 2018; 104:1215-1228. [PMID: 29985529 PMCID: PMC6258302 DOI: 10.1002/jlb.5a0717-276rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 06/05/2018] [Accepted: 06/06/2018] [Indexed: 01/04/2023] Open
Abstract
Conventional APCs that express MHC class II (MHCII) and co-stimulatory molecules include dendritic cells (DCs) and macrophages. Beyond these conventional APCs, immune stimulatory cells have been more recently shown to extend to a class of atypical APCs, composed of mast cells, basophils, and eosinophils. Here, we describe a unique type of APC, Gr1-/low CD11b-/low cells with a granularity and size characteristic of myeloid cells and with the ability to present Ag for crosspresentation. These cells constitutively express MHCII and the costimulatory molecules, CD80, CD86, and CD40. They do not express pan markers of myeloid DCs (CD11c), plasmacytoid DCs (Ly6C), or macrophages (F4/80), and their frequency is inversely correlated with myeloid-derived suppressor cells (MDSCs) in tumor-bearing mice. Among splenocytes, they are more abundant than DCs and macrophages, and they exhibit antitumor immune stimulatory function at a steady state without further activation, ex vivo. They are also found within the tumor bed where they retain their immune stimulatory function. Our findings suggest the use of these novel APCs in additional preclinical studies to further investigate their utility in APC-based cancer immunotherapies.
Collapse
Affiliation(s)
- Kyle K Payne
- Department of Microbiology & Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA.,Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA.,Department of Immunology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Hussein F Aqbi
- Department of Microbiology & Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA.,Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Savannah E Butler
- Department of Microbiology & Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA.,Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Laura Graham
- Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA.,Department of Surgery, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Rebecca C Keim
- Department of Microbiology & Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA.,Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Wen Wan
- Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA.,Department of Biostatistics, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Michael O Idowu
- Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA.,Department of Pathology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Harry D Bear
- Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA.,Department of Surgery, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Xiang-Yang Wang
- Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA.,Department of Human & Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA.,VCU Institute of Molecular Medicine, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Masoud H Manjili
- Department of Microbiology & Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA.,Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA.,VCU Institute of Molecular Medicine, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| |
Collapse
|
16
|
Özkan B, Lim H, Park SG. Immunomodulatory Function of Myeloid-Derived Suppressor Cells during B Cell-Mediated Immune Responses. Int J Mol Sci 2018; 19:E1468. [PMID: 29762501 PMCID: PMC5983618 DOI: 10.3390/ijms19051468] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 05/03/2018] [Accepted: 05/12/2018] [Indexed: 12/20/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) play roles in immune regulation during neoplastic and non-neoplastic inflammatory responses. This immune regulatory function is directed mainly toward T cells. However, MDSCs also regulate other cell populations, including B cells, during inflammatory responses. Indeed, B cells are essential for antibody-mediated immune responses. MDSCs regulate B cell immune responses directly via expression of effector molecules and indirectly by controlling other immune regulatory cells. B cell-mediated immune responses are a major component of the overall immune response; thus, MDSCs play a prominent role in their regulation. Here, we review the current knowledge about MDSC-mediated regulation of B cell responses.
Collapse
Affiliation(s)
- Bilgenaz Özkan
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Korea.
| | - Heejin Lim
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Korea.
| | - Sung-Gyoo Park
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Korea.
| |
Collapse
|
17
|
Wang Y, Schafer CC, Hough KP, Tousif S, Duncan SR, Kearney JF, Ponnazhagan S, Hsu HC, Deshane JS. Myeloid-Derived Suppressor Cells Impair B Cell Responses in Lung Cancer through IL-7 and STAT5. THE JOURNAL OF IMMUNOLOGY 2018; 201:278-295. [PMID: 29752311 DOI: 10.4049/jimmunol.1701069] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 04/23/2018] [Indexed: 12/29/2022]
Abstract
Myeloid-derived suppressor cells (MDSCs) are known suppressors of antitumor immunity, affecting amino acid metabolism and T cell function in the tumor microenvironment. However, it is unknown whether MDSCs regulate B cell responses during tumor progression. Using a syngeneic mouse model of lung cancer, we show reduction in percentages and absolute numbers of B cell subsets including pro-, pre-, and mature B cells in the bone marrow (BM) of tumor-bearing mice. The kinetics of this impaired B cell response correlated with the progressive infiltration of MDSCs. We identified that IL-7 and downstream STAT5 signaling that play a critical role in B cell development and differentiation were also impaired during tumor progression. Global impairment of B cell function was indicated by reduced serum IgG levels. Importantly, we show that anti-Gr-1 Ab-mediated depletion of MDSCs not only rescued serum IgG and IL-7 levels but also reduced TGF-β1, a known regulator of stromal IL-7, suggesting MDSC-mediated regulation of B cell responses. Furthermore, blockade of IL-7 resulted in reduced phosphorylation of downstream STAT5 and B cell differentiation in tumor-bearing mice and administration of TGF-β-blocking Ab rescued these IL-7-dependent B cell responses. Adoptive transfer of BM-derived MDSCs from tumor-bearing mice into congenic recipients resulted in significant reductions of B cell subsets in the BM and in circulation. MDSCs also suppressed B cell proliferation in vitro in an arginase-dependent manner that required cell-to-cell contact. Our results indicate that tumor-infiltrating MDSCs may suppress humoral immune responses and promote tumor escape from immune surveillance.
Collapse
Affiliation(s)
- Yong Wang
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Cara C Schafer
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Kenneth P Hough
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Sultan Tousif
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Steven R Duncan
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - John F Kearney
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294; and
| | | | - Hui-Chen Hsu
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Jessy S Deshane
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294;
| |
Collapse
|
18
|
Ben-Meir K, Twaik N, Baniyash M. Plasticity and biological diversity of myeloid derived suppressor cells. Curr Opin Immunol 2018; 51:154-161. [PMID: 29614426 DOI: 10.1016/j.coi.2018.03.015] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 01/27/2018] [Accepted: 03/13/2018] [Indexed: 10/17/2022]
Abstract
Myeloid derived suppressor cells (MDSCs) are immature myeloid cells characterized by diverse phenotypes and functions. They impair effector functions of immune cells and promote tumor growth, angiogenesis, and tissue damage. In pathologies characterized by chronic inflammation, MDSCs are arrested in their immature state and migrate from the bone marrow to the periphery and to the site of inflammation, where they mediate immunosuppression. When reaching new environments, which exhibit a different array of cytokines, chemokines, and pro-inflammatory mediators, MDSCs sense and adapt to the altered micro-environment by virtue of acquiring different suppressive features/functions that involve changing their cell fate, surface receptors, metabolism and intracellular as well as secreted molecules. This review summarizes some of the latest publications highlighting various layers of MDSC plasticity in relation to different pathologies. We discuss treatments capitalizing on MDSC plasticity aimed at combating MDSCs or manipulating their suppressive activity for improved therapy.
Collapse
Affiliation(s)
- Kerem Ben-Meir
- The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, The Hebrew University, POB 12272, Jerusalem 91120, Israel
| | - Nira Twaik
- The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, The Hebrew University, POB 12272, Jerusalem 91120, Israel
| | - Michal Baniyash
- The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, The Hebrew University, POB 12272, Jerusalem 91120, Israel.
| |
Collapse
|
19
|
Bian Z, Abdelaal AM, Shi L, Liang H, Xiong L, Kidder K, Venkataramani M, Culpepper C, Zen K, Liu Y. Arginase-1 is neither constitutively expressed in nor required for myeloid-derived suppressor cell-mediated inhibition of T-cell proliferation. Eur J Immunol 2018; 48:1046-1058. [PMID: 29488625 DOI: 10.1002/eji.201747355] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 01/23/2018] [Accepted: 02/20/2018] [Indexed: 11/06/2022]
Abstract
Although previous reports suggest that tumor-induced myeloid-derived suppressor cells (MDSC) inhibit T cells by L-arginine depletion through arginase-1 activity, we herein show that arginase-1 is neither inherently expressed in MDSC nor required for MDSC-mediated inhibition. Employing Percoll density gradients, large expansions of MDSC in the bone marrow of tumor-bearing mice were isolated and demonstrated potent inhibition in T-cell proliferation activated by TCR-ligation, Concanavalin A, PMA plus ionomycin, or IL-2. Despite demonstrating characteristic immunosuppressive capacity, these MDSC exhibit no arginase-1 expression and/or exert their inhibitory effects independent of arginase-1 activity. However, arginase-1 expression in MDSC can be induced by exposure to TCR-activated T cells or their culture medium, but not T cells activated by other means or growing tumor cells. Further investigation reveals multiple cytokines secreted by TCR-activated T cells as orchestrating two signaling-relay axes, IL-6-to-IL-4 and GM-CSF/IL-4-to-IL-10, leading to arginase-1 expression in MDSC. Specifically, IL-6 signaling increases IL-4R, enabling IL-4 to induce arginase-1 expression; similarly, GM-CSF in concert with IL-4 induces IL-10R, allowing IL-10-mediated induction. Surprisingly, our study indicates that induction of arginase-1 expression is not conducive to the critical MDSC-mediated inhibition toward T cells, which is rather dependent on direct cell contacts undiminished by PD-L1 blockade or SIRPα deficiency.
Collapse
Affiliation(s)
- Zhen Bian
- Program of Immunology and Molecular Cellular Biology, Department of Biology, Center for Diagnostics and Therapeutics, Center of Inflammation, Immunity and Infection, Georgia State University, Atlanta, GA, USA
| | - Ahmed Mansour Abdelaal
- Program of Immunology and Molecular Cellular Biology, Department of Biology, Center for Diagnostics and Therapeutics, Center of Inflammation, Immunity and Infection, Georgia State University, Atlanta, GA, USA
| | - Lei Shi
- Program of Immunology and Molecular Cellular Biology, Department of Biology, Center for Diagnostics and Therapeutics, Center of Inflammation, Immunity and Infection, Georgia State University, Atlanta, GA, USA
| | - Hongwei Liang
- Program of Immunology and Molecular Cellular Biology, Department of Biology, Center for Diagnostics and Therapeutics, Center of Inflammation, Immunity and Infection, Georgia State University, Atlanta, GA, USA
| | - Lanqiao Xiong
- Program of Immunology and Molecular Cellular Biology, Department of Biology, Center for Diagnostics and Therapeutics, Center of Inflammation, Immunity and Infection, Georgia State University, Atlanta, GA, USA
| | - Koby Kidder
- Program of Immunology and Molecular Cellular Biology, Department of Biology, Center for Diagnostics and Therapeutics, Center of Inflammation, Immunity and Infection, Georgia State University, Atlanta, GA, USA
| | - Mahathi Venkataramani
- Program of Immunology and Molecular Cellular Biology, Department of Biology, Center for Diagnostics and Therapeutics, Center of Inflammation, Immunity and Infection, Georgia State University, Atlanta, GA, USA
| | - Courtney Culpepper
- Program of Immunology and Molecular Cellular Biology, Department of Biology, Center for Diagnostics and Therapeutics, Center of Inflammation, Immunity and Infection, Georgia State University, Atlanta, GA, USA
| | - Ke Zen
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Advanced Institute for Life Sciences, Nanjing University, Nanjing, Jiangsu, China
| | - Yuan Liu
- Program of Immunology and Molecular Cellular Biology, Department of Biology, Center for Diagnostics and Therapeutics, Center of Inflammation, Immunity and Infection, Georgia State University, Atlanta, GA, USA
| |
Collapse
|
20
|
Shen M, Wang J, Yu W, Zhang C, Liu M, Wang K, Yang L, Wei F, Wang SE, Sun Q, Ren X. A novel MDSC-induced PD-1 -PD-L1 + B-cell subset in breast tumor microenvironment possesses immuno-suppressive properties. Oncoimmunology 2018; 7:e1413520. [PMID: 29632731 DOI: 10.1080/2162402x.2017.1413520] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 11/30/2017] [Accepted: 11/30/2017] [Indexed: 10/18/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous group of myeloid cells that suppress T-cell activity in a tumor microenvironment. However, the suppressive function of MDSCs on B cells and its underlying mechanism remain unclear. Here, we show that in 4T1 breast cancer mice, a significantly increased number of MDSCs, in parallel with splenic B cells, are accumulated when compared to normal mice. In the presence of MDSCs, the surface molecules of B cells are remolded, with checkpoint-related molecules such as PD-1 and PD-L1 changing prominently. MDSCs also emerge as vital regulators in B-cell immune functions such as proliferation, apoptosis and the abilities to secrete antibodies and cytokines. Our study further identifies that MDSCs can transform normal B cells to a subtype of immuno- regulatory B cells (Bregs) which inhibit T-cell response. Furthermore, we identified a novel kind of Bregs with a specific phenotype PD-1-PD-L1+CD19+, which exert the greatest suppressive effects on T cells in comparison with the previously reported Bregs characterized as CD1d+CD5+CD19+, CD5+CD19+ and Interleukin (IL)-10-secreting B cells. Our results highlight that MDSCs regulate B-cell response and may serve as a therapeutic approach in anti-tumor treatment. Investigation of this new Breg subtype extends our understanding of regulation of T-cell response and sheds new light on anti-tumor immunity and immune therapy.
Collapse
Affiliation(s)
- Meng Shen
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Jian Wang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Wenwen Yu
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Chen Zhang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Min Liu
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Kaiyuan Wang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Lili Yang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Feng Wei
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Shizhen Emily Wang
- Department of Pathology, University of California, San Diego, California, USA
| | - Qian Sun
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Xiubao Ren
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| |
Collapse
|
21
|
Zhang W, Li J, Qi G, Tu G, Yang C, Xu M. Myeloid-derived suppressor cells in transplantation: the dawn of cell therapy. J Transl Med 2018; 16:19. [PMID: 29378596 PMCID: PMC5789705 DOI: 10.1186/s12967-018-1395-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Accepted: 01/22/2018] [Indexed: 02/07/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a series of innate cells that play a significant role in inhibiting T cell-related responses. This heterogeneous population of immature cells is involved in tumor immunity. Recently, the function and importance of MDSCs in transplantation have garnered the attention of scientists and have become an important focus of transplantation immunology research because MDSCs play a key role in establishing immune tolerance in transplantation. In this review, we summarize recent studies of MDSCs in different types of transplantation. We also focus on the influence of immunosuppressive drugs on MDSCs as well as future obstacles and research directions in this field.
Collapse
Affiliation(s)
- Weitao Zhang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai Key Laboratory of Organ Transplantation, 180 Fenglin Road, Shanghai, 200032 China
| | - Jiawei Li
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai Key Laboratory of Organ Transplantation, 180 Fenglin Road, Shanghai, 200032 China
| | - Guisheng Qi
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai Key Laboratory of Organ Transplantation, 180 Fenglin Road, Shanghai, 200032 China
| | - Guowei Tu
- Department of Intensive Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Cheng Yang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai Key Laboratory of Organ Transplantation, 180 Fenglin Road, Shanghai, 200032 China
| | - Ming Xu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai Key Laboratory of Organ Transplantation, 180 Fenglin Road, Shanghai, 200032 China
| |
Collapse
|
22
|
Dorhoi A, Du Plessis N. Monocytic Myeloid-Derived Suppressor Cells in Chronic Infections. Front Immunol 2018; 8:1895. [PMID: 29354120 PMCID: PMC5758551 DOI: 10.3389/fimmu.2017.01895] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 12/11/2017] [Indexed: 01/04/2023] Open
Abstract
Heterogeneous populations of myeloid regulatory cells (MRC), including monocytes, macrophages, dendritic cells, and neutrophils, are found in cancer and infectious diseases. The inflammatory environment in solid tumors as well as infectious foci with persistent pathogens promotes the development and recruitment of MRC. These cells help to resolve inflammation and establish host immune homeostasis by restricting T lymphocyte function, inducing regulatory T cells and releasing immune suppressive cytokines and enzyme products. Monocytic MRC, also termed monocytic myeloid-derived suppressor cells (M-MDSC), are bona fide phagocytes, capable of pathogen internalization and persistence, while exerting localized suppressive activity. Here, we summarize molecular pathways controlling M-MDSC genesis and functions in microbial-induced non-resolved inflammation and immunopathology. We focus on the roles of M-MDSC in infections, including opportunistic extracellular bacteria and fungi as well as persistent intracellular pathogens, such as mycobacteria and certain viruses. Better understanding of M-MDSC biology in chronic infections and their role in antimicrobial immunity, will advance development of novel, more effective and broad-range anti-infective therapies.
Collapse
Affiliation(s)
- Anca Dorhoi
- Institute of Immunology, Bundesforschungsinstitut für Tiergesundheit, Friedrich-Loeffler-Institut (FLI), Insel Riems, Germany.,Faculty of Mathematics and Natural Sciences, University of Greifswald, Greifswald, Germany.,Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Nelita Du Plessis
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, SAMRC Centre for Tuberculosis Research, DST and NRF Centre of Excellence for Biomedical TB Research, Stellenbosch University, Tygerberg, South Africa
| |
Collapse
|
23
|
Drabczyk-Pluta M, Werner T, Hoffmann D, Leng Q, Chen L, Dittmer U, Zelinskyy G. Granulocytic myeloid-derived suppressor cells suppress virus-specific CD8 + T cell responses during acute Friend retrovirus infection. Retrovirology 2017; 14:42. [PMID: 28835242 PMCID: PMC5569525 DOI: 10.1186/s12977-017-0364-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 08/11/2017] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Myeloid-derived suppressor cells (MDSCs) can suppress T cell responses in several different diseases. Previously these suppressive cells were observed to expand in HIV patients and in a mouse retrovirus model, yet their suppressive effect on virus-specific CD8+ T cells in vitro and in vivo has not been characterized thus far. RESULTS We used the Friend retrovirus (FV) model to demonstrate that MDSCs expand and become activated during the late phase of acute FV infection. Only the subpopulation of granulocytic MDSCs (gMDSCs) but not monocytic MDSC suppressed virus-specific CD8+ T cell proliferation and function in vitro. gMDSCs expressed arginase 1, high levels of the inhibitory ligand PD-L1 and the ATP dephosphorylating enzyme CD39 on the cell surface upon infection. All three molecules were involved in the suppressive effect of the gMDSCs in vitro. MDSC depletion experiments in FV-infected mice revealed that they restrict virus-specific CD8+ T cell responses and thus affect the immune control of chronic retroviruses in vivo. CONCLUSIONS Our study demonstrates that MDSCs become activated and expand during the acute phase of retrovirus infection. Their suppressive activity on virus-specific CD8+ T cells may contribute to T cell dysfunction and the development of chronic infection.
Collapse
Affiliation(s)
- Malgorzata Drabczyk-Pluta
- Institute of Virology, University Hospital Essen, University of Duisburg-Essen, Hufelandstraße 55, 45147 Essen, Germany
| | - Tanja Werner
- Institute of Virology, University Hospital Essen, University of Duisburg-Essen, Hufelandstraße 55, 45147 Essen, Germany
| | - Daniel Hoffmann
- Research Group Bioinformatics, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | - Qibin Leng
- Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Lieping Chen
- Department of Immunobiology, Yale School of Medicine, Yale University, New Haven, CT USA
| | - Ulf Dittmer
- Institute of Virology, University Hospital Essen, University of Duisburg-Essen, Hufelandstraße 55, 45147 Essen, Germany
| | - Gennadiy Zelinskyy
- Institute of Virology, University Hospital Essen, University of Duisburg-Essen, Hufelandstraße 55, 45147 Essen, Germany
| |
Collapse
|
24
|
Myeloid-derived suppressor cells modulate B-cell responses. Immunol Lett 2017; 188:108-115. [PMID: 28687234 DOI: 10.1016/j.imlet.2017.07.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 06/27/2017] [Accepted: 07/03/2017] [Indexed: 02/07/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) are key regulators of adaptive immunity by suppressing T-cell functions. However, their potential action on or interaction with B cells remained poorly understood. Here we demonstrate that human polymorphonuclear MDSCs differentially modulate B-cell function by suppressing B-cell proliferation and antibody production. We further demonstrate that this MDSC-mediated effect is cell contact dependent and involves established mediators such as arginase-1, nitric oxide (NO), reactive oxygen species (ROS) as well as B-cell death. Collectively, our studies provide novel evidence that human MDSCs modulate B cells, which could have future implications for immunotherapy approaches.
Collapse
|
25
|
Tamadaho RSE, Hoerauf A, Layland LE. Immunomodulatory effects of myeloid-derived suppressor cells in diseases: Role in cancer and infections. Immunobiology 2017; 223:432-442. [PMID: 29246400 DOI: 10.1016/j.imbio.2017.07.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 06/05/2017] [Accepted: 07/02/2017] [Indexed: 01/05/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) are heterogeneous cells capable of abrogating T and B cells responses and have been identified in numerous cancers. As with other regulatory cell populations, they aim to maintain balance between host-defence-associated inflammation and ensuing tissue pathology. MDSC accumulation and/or activation involve several growth factors and cytokines including Granulocyte Macrophage-Colony Stimulating Factor (GM-CSF) and Interleukin (IL)-6 and suppression has been linked to receptors such as IL-4Rα. Other immune pathways, such as Toll-like receptors (TLRs) have also been shown to interfere in MDSC activity adding to the complexity in clarifying their pathways. Monocytic- (Mo-MDSCs) and polymorphonuclear- (PMN-MDSCs) cells are two subsets of MDSCs that have been well characterized and have been shown to function through different mechanisms although both appear to require nitric oxide. In human and murine model settings, MDSCs have been shown to have inhibitory effects on T cell responses during bacterial, parasitic and viral pathologies and an increase of MDSC numbers has been associated with pathological conditions. Interestingly, the environment impacts on MDSC activity and regulatory T cells (Tregs), mast cells and a few cells that may help MDSC in order to regulate immune responses. Since the majority of pioneering data on MDSCs has stemmed from research on malignancies, this review will summarize MDSC biology and function in cancer and highlight current knowledge about these cells during infectious pathologies as well.
Collapse
Affiliation(s)
- Ruth S E Tamadaho
- Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital of Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - Achim Hoerauf
- Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital of Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany; German Centre for Infection Research (DZIF), Partner Site, Bonn-Cologne, Bonn, Germany
| | - Laura E Layland
- Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital of Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany; German Centre for Infection Research (DZIF), Partner Site, Bonn-Cologne, Bonn, Germany.
| |
Collapse
|
26
|
O'Connor MA, Rastad JL, Green WR. The Role of Myeloid-Derived Suppressor Cells in Viral Infection. Viral Immunol 2017; 30:82-97. [PMID: 28051364 DOI: 10.1089/vim.2016.0125] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are heterogeneous immature myeloid cells that are well described as potent immune regulatory cells during human cancer and murine tumor models. Reports of MDSCs during viral infections remain limited, and their association with immunomodulation of viral diseases is still being defined. Here, we provide an overview of MDSCs or MDSC-like cells identified during viral infections, including murine viral models and human viral diseases. Understanding the similarities and/or differences of virally induced versus tumor-derived MDSCs will be important for designing future immunotherapeutic approaches.
Collapse
Affiliation(s)
- Megan A O'Connor
- 1 Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth , Lebanon , New Hampshire
| | - Jessica L Rastad
- 1 Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth , Lebanon , New Hampshire
| | - William R Green
- 1 Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth , Lebanon , New Hampshire.,2 Norris Cotton Cancer Center , Geisel School of Medicine at Dartmouth, Lebanon , New Hampshire
| |
Collapse
|
27
|
Rastad JL, Green WR. Myeloid-derived suppressor cells in murine AIDS inhibit B-cell responses in part via soluble mediators including reactive oxygen and nitrogen species, and TGF-β. Virology 2016; 499:9-22. [PMID: 27632561 DOI: 10.1016/j.virol.2016.08.031] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 08/28/2016] [Accepted: 08/29/2016] [Indexed: 12/17/2022]
Abstract
Monocytic myeloid-derived suppressor cells (M-MDSCs) were increased during LP-BM5 retroviral infection, and were capable of suppressing not only T-cell, but also B-cell responses. In addition to previously demonstrating iNOS- and VISTA-dependent M-MDSC mechanisms, in this paper, we detail how M-MDSCs utilized soluble mediators, including the reactive oxygen and nitrogen species superoxide, peroxynitrite, and nitric oxide, and TGF-β, to suppress B cells in a predominantly contact-independent manner. Suppression was independent of cysteine-depletion and hydrogen peroxide production. When two major mechanisms of suppression (iNOS and VISTA) were eliminated in double knockout mice, M-MDSCs from LP-BM5-infected mice were able to compensate using other, soluble mechanisms in order to maintain suppression of B cells. The IL-10 producing regulatory B-cell compartment was among the targets of M-MDSC-mediated suppression.
Collapse
Affiliation(s)
- Jessica L Rastad
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, United States
| | - William R Green
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, United States; Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, United States.
| |
Collapse
|
28
|
Glutathione Depletion Is Linked with Th2 Polarization in Mice with a Retrovirus-Induced Immunodeficiency Syndrome, Murine AIDS: Role of Proglutathione Molecules as Immunotherapeutics. J Virol 2016; 90:7118-7130. [PMID: 27226373 DOI: 10.1128/jvi.00603-16] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 05/19/2016] [Indexed: 12/24/2022] Open
Abstract
UNLABELLED Injection of the LP-BM5 murine leukemia virus into mice causes murine AIDS, a disease characterized by many dysfunctions of immunocompetent cells. To establish whether the disease is characterized by glutathione imbalance, reduced glutathione (GSH) and cysteine were quantified in different organs. A marked redox imbalance, consisting of GSH and/or cysteine depletion, was found in the lymphoid organs, such as the spleen and lymph nodes. Moreover, a significant decrease in cysteine and GSH levels in the pancreas and brain, respectively, was measured at 5 weeks postinfection. The Th2 immune response was predominant at all times investigated, as revealed by the expression of Th1/Th2 cytokines. Furthermore, investigation of the activation status of peritoneal macrophages showed that the expression of genetic markers of alternative activation, namely, Fizz1, Ym1, and Arginase1, was induced. Conversely, expression of inducible nitric oxide synthase, a marker of classical activation of macrophages, was detected only when Th1 cytokines were expressed at high levels. In vitro studies revealed that during the very early phases of infection, GSH depletion and the downregulation of interleukin-12 (IL-12) p40 mRNA were correlated with the dose of LP-BM5 used to infect the macrophages. Treatment of LP-BM5-infected mice with N-(N-acetyl-l-cysteinyl)-S-acetylcysteamine (I-152), an N-acetyl-cysteine supplier, restored GSH/cysteine levels in the organs, reduced the expression of alternatively activated macrophage markers, and increased the level of gamma interferon production, while it decreased the levels of Th2 cytokines, such as IL-4 and IL-5. Our findings thus establish a link between GSH deficiency and Th1/Th2 disequilibrium in LP-BM5 infection and indicate that I-152 can be used to restore the GSH level and a balanced Th1/Th2 response in infected mice. IMPORTANCE The first report of an association between Th2 polarization and alteration of the redox state in LP-BM5 infection is presented. Moreover, it provides evidence that LP-BM5 infection causes a decrease in the thiol content of peritoneal macrophages, which can influence IL-12 production. The restoration of GSH levels by GSH-replenishing molecules can represent a new therapeutic avenue to fight this retroviral infection, as it reestablishes the Th1/Th2 balance. Immunotherapy based on the use of pro-GSH molecules would permit LP-BM5 infection and probably all those viral infections characterized by GSH deficiency and a Th1/Th2 imbalance to be more effectively combated.
Collapse
|
29
|
Foks AC, Van Puijvelde GHM, Wolbert J, Kröner MJ, Frodermann V, Van Der Heijden T, Van Santbrink PJ, Boon L, Bot I, Kuiper J. CD11b+Gr-1+ myeloid-derived suppressor cells reduce atherosclerotic lesion development in LDLr deficient mice. Cardiovasc Res 2016; 111:252-61. [PMID: 27234908 DOI: 10.1093/cvr/cvw114] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 05/16/2016] [Indexed: 01/25/2023] Open
Abstract
AIMS Myeloid-derived suppressor cells (MDSCs) form a heterogeneous population of cells composed of early myeloid progenitor cells and immature myeloid cells, which strongly suppress pro-inflammatory immune cells in inflammatory diseases. Currently, it is unknown whether MDSCs contribute to atherosclerosis, a chronic inflammatory disease in which accumulation of lipoproteins in the arterial wall activates the immune system causing abnormal vascular remodelling and vessel occlusion. Here, we investigated whether and how MDSCs contribute to the development of atherosclerosis. METHODS AND RESULTS We show that MDSCs arise in the bone marrow of LDLr(-/-) mice during atherosclerosis and strongly suppress proliferation of T cells. Adoptive transfer of MDSCs into both female and male LDLr(-/-) mice fed a Western-type diet (WTD) ameliorates atherosclerosis with 35%. We observed a 54% reduction in adventitial T cells, and more specifically, MDSCs suppress Th1 and Th17 cells. In addition, treatment with MDSCs reduces circulating pro-atherogenic B2 cells. We found two subsets of MDSCs in the bone marrow of hypercholesterolemic mice, monocytic and granulocytic MDSCs (mo- and gr-MDSCs, respectively), of which the percentage of mo-MDSCs significantly increased during WTD feeding. Moreover, mo-MDSCs completely abolished splenocyte proliferation, whereas gr-MDSCs were unable to suppress proliferation. Mechanistically, we show that MDSCs from atherosclerotic mice suppress T cells in an IFN-γ- and nitric oxide-dependent manner, which is associated with the action of mo-MDSCs. CONCLUSION This study demonstrates that MDSCs develop during atherosclerosis and reduce atherosclerosis via suppression of pro-inflammatory immune responses.
Collapse
Affiliation(s)
- Amanda C Foks
- Division of Biopharmaceutics, Leiden University, Gorlaeus Laboratories, Einsteinweg 55, Leiden 2333 CC, The Netherlands
| | - Gijs H M Van Puijvelde
- Division of Biopharmaceutics, Leiden University, Gorlaeus Laboratories, Einsteinweg 55, Leiden 2333 CC, The Netherlands
| | - Jolien Wolbert
- Division of Biopharmaceutics, Leiden University, Gorlaeus Laboratories, Einsteinweg 55, Leiden 2333 CC, The Netherlands
| | - Mara J Kröner
- Division of Biopharmaceutics, Leiden University, Gorlaeus Laboratories, Einsteinweg 55, Leiden 2333 CC, The Netherlands
| | - Vanessa Frodermann
- Division of Biopharmaceutics, Leiden University, Gorlaeus Laboratories, Einsteinweg 55, Leiden 2333 CC, The Netherlands
| | - Thomas Van Der Heijden
- Division of Biopharmaceutics, Leiden University, Gorlaeus Laboratories, Einsteinweg 55, Leiden 2333 CC, The Netherlands
| | - Peter J Van Santbrink
- Division of Biopharmaceutics, Leiden University, Gorlaeus Laboratories, Einsteinweg 55, Leiden 2333 CC, The Netherlands
| | | | - Ilze Bot
- Division of Biopharmaceutics, Leiden University, Gorlaeus Laboratories, Einsteinweg 55, Leiden 2333 CC, The Netherlands
| | - Johan Kuiper
- Division of Biopharmaceutics, Leiden University, Gorlaeus Laboratories, Einsteinweg 55, Leiden 2333 CC, The Netherlands
| |
Collapse
|
30
|
Xu X, Meng Q, Erben U, Wang P, Glauben R, Kühl AA, Wu H, Ma CW, Hu M, Wang Y, Sun W, Jia J, Wu X, Chen W, Siegmund B, Qin Z. Myeloid-derived suppressor cells promote B-cell production of IgA in a TNFR2-dependent manner. Cell Mol Immunol 2016; 14:597-606. [PMID: 27133471 DOI: 10.1038/cmi.2015.103] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 11/20/2015] [Accepted: 11/20/2015] [Indexed: 12/31/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are well known for their capacity to suppress antitumor T-cell responses, but their effects on B-cell function and antibody production remain unclear. Here, we found that MDSCs that accumulated around the germinal center in the spleen of tumor-bearing mice co-located with B cells. In the presence of MDSCs, the antibody reaction to a surrogate antigen was significantly enhanced in mice, especially the immunoglobulin (Ig)A subtype. Co-culture with MDSCs promoted both proliferation and differentiation of B cells into IgA-producing plasma cells in vitro. Interestingly, the cross talk between MDSCs and B cells required cell-cell contact. MDSCs from tumor necrosis factor receptor (TNFR) 2-/- mice, but not from TNFR1-/- mice, failed to promote B-cell responses. Further investigation suggested that interleukin-10 and transforming growth factor-β1 were crucial for the MDSC-mediated promotion of IgA responses. These results demonstrate a novel mechanism of MDSC-mediated immune regulation during tumor growth.
Collapse
Affiliation(s)
- Xia Xu
- Key Laboratory of Protein and Peptide Pharmaceuticals, Chinese Academy of Sciences-University of Tokyo Joint Laboratory of Structural Virology and Immunology, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Qinghong Meng
- Key Laboratory of Protein and Peptide Pharmaceuticals, Chinese Academy of Sciences-University of Tokyo Joint Laboratory of Structural Virology and Immunology, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ulrike Erben
- Medical Department for Gastroenterology, Infectious Diseases and Rheumatology/Research Center ImmunoSciences, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, Berlin 12200, Germany
| | - Peigang Wang
- Key Laboratory of Protein and Peptide Pharmaceuticals, Chinese Academy of Sciences-University of Tokyo Joint Laboratory of Structural Virology and Immunology, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Rainer Glauben
- Medical Department for Gastroenterology, Infectious Diseases and Rheumatology/Research Center ImmunoSciences, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, Berlin 12200, Germany
| | - Anja A Kühl
- Medical Department for Gastroenterology, Infectious Diseases and Rheumatology/Research Center ImmunoSciences, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, Berlin 12200, Germany
| | - Hao Wu
- Key Laboratory of Protein and Peptide Pharmaceuticals, Chinese Academy of Sciences-University of Tokyo Joint Laboratory of Structural Virology and Immunology, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Chung Wah Ma
- Infinitus Chinese Herbal Immunity Research Centre, Guangzhou 510665, China
| | - Minghua Hu
- Infinitus Chinese Herbal Immunity Research Centre, Guangzhou 510665, China
| | - Yuanyuan Wang
- Infinitus Chinese Herbal Immunity Research Centre, Guangzhou 510665, China
| | - Wei Sun
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine Berlin-Buch, Berlin 13125, Germany
| | - Junying Jia
- Key Laboratory of Protein and Peptide Pharmaceuticals, Chinese Academy of Sciences-University of Tokyo Joint Laboratory of Structural Virology and Immunology, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Xinyi Wu
- Key Laboratory of Protein and Peptide Pharmaceuticals, Chinese Academy of Sciences-University of Tokyo Joint Laboratory of Structural Virology and Immunology, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Wei Chen
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine Berlin-Buch, Berlin 13125, Germany
| | - Britta Siegmund
- Medical Department for Gastroenterology, Infectious Diseases and Rheumatology/Research Center ImmunoSciences, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, Berlin 12200, Germany
| | - Zhihai Qin
- Key Laboratory of Protein and Peptide Pharmaceuticals, Chinese Academy of Sciences-University of Tokyo Joint Laboratory of Structural Virology and Immunology, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
31
|
Kennedy DE, Witte PL, Knight KL. Bone marrow fat and the decline of B lymphopoiesis in rabbits. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 58:30-9. [PMID: 26577994 PMCID: PMC4775299 DOI: 10.1016/j.dci.2015.11.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 11/05/2015] [Indexed: 05/03/2023]
Abstract
B lymphopoiesis is necessary to generate a diverse pool of naïve B cells that are able to respond to a broad spectrum of antigens during immune responses to pathogens and to vaccination. Rabbits have been utilized for many years to generate high affinity monoclonal and polyclonal antibodies. Specific antibodies generated in rabbits have greatly advanced scientific discoveries, but the unique qualities of rabbit B cell development have been underappreciated. Unlike in humans and mice, where B lymphopoiesis declines in mid to late life, B lymphopoiesis in rabbits arrests early in life, between 2 and 4 months of age. This review focuses on the early loss of B cell development in rabbits and the contribution of the bone marrow microenvironment to this process. We also propose directions for future research in this area, and discuss how the rabbit can be used as a model to understand the decline of B lymphopoiesis that occurs in humans late in life. Such studies will be important for developing therapeutics targeted to prevent and/or reverse declining B lymphopoiesis in the elderly, as well as boosting immunity and antibody responses after infection or vaccination.
Collapse
Affiliation(s)
- Domenick E Kennedy
- Loyola University Chicago, Department of Microbiology and Immunology, USA
| | - Pamela L Witte
- Loyola University Chicago, Department of Microbiology and Immunology, USA
| | - Katherine L Knight
- Loyola University Chicago, Department of Microbiology and Immunology, USA.
| |
Collapse
|
32
|
Wang JZ, Zhang YH, Guo XH, Zhang HY, Zhang Y. The double-edge role of B cells in mediating antitumor T-cell immunity: Pharmacological strategies for cancer immunotherapy. Int Immunopharmacol 2016; 36:73-85. [PMID: 27111515 DOI: 10.1016/j.intimp.2016.04.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Revised: 04/10/2016] [Accepted: 04/13/2016] [Indexed: 12/22/2022]
Abstract
Emerging evidence reveals the controversial role of B cells in antitumor immunity, but the underlying mechanisms have to be explored. Three latest articles published in the issue 521 of Nature in 2015 reconfirmed the puzzling topic and put forward some explanations of how B cells regulate antitumor T-cell responses both positively and negatively. This paper attempts to demonstrate that different B-cell subpopulations have distinct immunological properties and that they are involved in either antitumor responses or immunosuppression. Recent studies supporting the positive and negative roles of B cells in tumor development were summarized comprehensively. Several specific B-cell subpopulations, such as IgG(+), IgA(+), IL-10(+), and regulatory B cells, were described in detail. The mechanisms underlying the controversial B-cell effects were mainly attributed to different B-cell subpopulations, different B-cell-derived cytokines, direct B cell-T cell interaction, different cancer categories, and different malignant stages, and the immunological interaction between B cells and T cells is mediated by dendritic cells. Promising B-cell-based antitumor strategies were proposed and novel B-cell regulators were summarized to present interesting therapeutic targets. Future investigations are needed to make sure that B-cell-based pharmacological strategies benefit cancer immunotherapy substantially.
Collapse
Affiliation(s)
- Jing-Zhang Wang
- Department of Medical Technology, College of Medicine, Affiliated Hospital, Hebei University of Engineering, Handan 056002, PR China.
| | - Yu-Hua Zhang
- Department of Library, Hebei University of Engineering, Handan 056038, PR China
| | - Xin-Hua Guo
- Department of Medicine, College of Medicine, Hebei University of Engineering, Handan 056002, PR China
| | - Hong-Yan Zhang
- Department of Medical Technology, College of Medicine, Affiliated Hospital, Hebei University of Engineering, Handan 056002, PR China
| | - Yuan Zhang
- Department of Medical Technology, College of Medicine, Affiliated Hospital, Hebei University of Engineering, Handan 056002, PR China
| |
Collapse
|
33
|
Kennedy DE, Witte PL, Knight KL. Withdrawn: Bone marrow fat and the decline of B lymphopoiesis in rabbits. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2015:S0145-305X(15)30071-9. [PMID: 26550685 DOI: 10.1016/j.dci.2015.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 11/03/2015] [Indexed: 06/05/2023]
Affiliation(s)
- Domenick E Kennedy
- Loyola University Chicago, Department of Microbiology and Immunology, USA
| | - Pamela L Witte
- Loyola University Chicago, Department of Microbiology and Immunology, USA
| | - Katherine L Knight
- Loyola University Chicago, Department of Microbiology and Immunology, USA.
| |
Collapse
|
34
|
Zhang H, Li ZL, Ye SB, Ouyang LY, Chen YS, He J, Huang HQ, Zeng YX, Zhang XS, Li J. Myeloid-derived suppressor cells inhibit T cell proliferation in human extranodal NK/T cell lymphoma: a novel prognostic indicator. Cancer Immunol Immunother 2015; 64:1587-99. [PMID: 26497849 PMCID: PMC4643115 DOI: 10.1007/s00262-015-1765-6] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 10/04/2015] [Indexed: 01/04/2023]
Abstract
The expansion of myeloid-derived suppressor cells (MDSCs) and its correlation with advanced disease stage have been shown in solid cancers. Here, we investigated the functional features and clinical significance of MDSCs in extranodal NK/T cell lymphoma (ENKL). A higher percentage of circulating HLA-DR−CD33+CD11b+ MDSCs was observed in ENKL patients than in healthy controls (P < 0.05, n = 32) by flow cytometry analysis. These MDSCs from ENKL patients (ENKL-MDSCs) consisted of CD14+ monocytic (Mo-MDSCs, >60 %) and CD15+ granulocytic (PMN-MDSCs, <20 %) MDSCs. Furthermore, these ENKL-MDSCs expressed higher levels of Arg-1, iNOS and IL-17 compared to the levels of MDSCs from healthy donors, and they expressed moderate levels of TGFβ and IL-10 but lower levels of CD66b. The ENKL-MDSCs strongly suppressed the anti-CD3-induced allogeneic and autologous CD4 T cell proliferation (P < 0.05), but they only slightly suppressed CD8 T cell proliferation (P > 0.05). Interestingly, ENKL-MDSCs inhibited the secretion of IFNγ but promoted IL-10, IL-17 and TGFβ secretion as well as Foxp3 expression in T cells. The administration of inhibitors of iNOS, Arg-1 and ROS significantly reversed the suppression of anti-CD3-induced T cell proliferation by MDSCs (P < 0.05). Importantly, based on multivariate Cox regression analysis, the HLA-DR−CD33+CD11b+ cells and CD14+ Mo-MDSCs were independent predictors for disease-free survival (DFS, P = 0.013 and 0.016) and overall survival (OS, P = 0.017 and 0.027). Overall, our results identified for the first time that ENKL-MDSCs (mainly Mo-MDSCs) have a prognostic value for patients and a suppressive function on T cell proliferation.
Collapse
Affiliation(s)
- Han Zhang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
- Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Ze-Lei Li
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
- Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Shu-Biao Ye
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
- Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Li-Ying Ouyang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
- Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
- Intensive Care Unit Department, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Yu-Shan Chen
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
- Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
- Department of Radiotherapy, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Jia He
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
- Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Hui-Qiang Huang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
- Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Yi-Xin Zeng
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
- Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Xiao-Shi Zhang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China.
- Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China.
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China.
| | - Jiang Li
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China.
- Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China.
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China.
| |
Collapse
|
35
|
Zhao Y, Wu T, Shao S, Shi B, Zhao Y. Phenotype, development, and biological function of myeloid-derived suppressor cells. Oncoimmunology 2015; 5:e1004983. [PMID: 27057424 DOI: 10.1080/2162402x.2015.1004983] [Citation(s) in RCA: 148] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Revised: 12/24/2014] [Accepted: 12/28/2014] [Indexed: 10/22/2022] Open
Abstract
CD11b+Gr-1+ myeloid-derived suppressor cells (MDSCs) are an important population of innate regulatory cells mainly comprising monocytic MDSCs (M-MDSCs) with a phenotype of CD11b+Ly6G-Ly6Chigh and granulocytic MDSCs (G-MDSCs) with a phenotype of CD11b+Ly6G+Ly6Clow in mice. They play crucial roles in the pathogenesis of cancers, chronic infections, autoimmune diseases, and transplantation. Various extracellular factors such as lipopolysaccharide (LPS), macrophage colony-stimulating factor (M-CSF), granulocyte macrophage colony-stimulating factor (GM-CSF), stem cell factor (SCF), interleukin (IL)-6, interferon gamma (IFNγ), IL-1β, vascular endothelial growth factor (VEGF), Hsp72, IL-13, C5a, and prostaglandin E2 (PGE2) can induce MDSC differentiation, whereas IL-4 and all-trans-retinoic acid can inhibit this process. For the intracellular signals, signal transducer and activator of transcription (STAT) family members, C/EBPβ and cyclooxigenase-2 (COX-2) promote MDSC function, whereas interferon regulatory factor-8 (IRF-8) and Smad3 downregulate MDSC activity. The immunosuppressive function of MDSCs is mediated through various effector molecules, primarily cellular metabolism-related molecules such as nitric oxide (NO), arginase, reactive oxygen species (ROS), transforming growth factor β (TGFβ), IL-10, indoleamine 2,3-dioxygenase (IDO), heme oxygenase-1 (HO-1), carbon monoxide (CO), and PGE2. In this article, we will summarize the molecules involved in the induction and function of MDSCs as well as the regulatory pathways of MDSCs.
Collapse
Affiliation(s)
- Yang Zhao
- Transplantation Biology Research Division; State Key Laboratory of Biomembrane and Membrane Biotechnology; Institute of Zoology; Chinese Academy of Sciences ; Beijing, China
| | - Tingting Wu
- Transplantation Biology Research Division; State Key Laboratory of Biomembrane and Membrane Biotechnology; Institute of Zoology; Chinese Academy of Sciences ; Beijing, China
| | - Steven Shao
- Transplantation Biology Research Division; State Key Laboratory of Biomembrane and Membrane Biotechnology; Institute of Zoology; Chinese Academy of Sciences ; Beijing, China
| | - Bingyi Shi
- Organ Transplantation Center of People's Liberation Army; 309 Hospital of Chinese People's Liberation Army ; Beijing, China
| | - Yong Zhao
- Transplantation Biology Research Division; State Key Laboratory of Biomembrane and Membrane Biotechnology; Institute of Zoology; Chinese Academy of Sciences ; Beijing, China
| |
Collapse
|
36
|
Kennedy DE, Knight KL. Inhibition of B Lymphopoiesis by Adipocytes and IL-1-Producing Myeloid-Derived Suppressor Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2015; 195:2666-74. [PMID: 26268654 PMCID: PMC4561202 DOI: 10.4049/jimmunol.1500957] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 07/20/2015] [Indexed: 12/12/2022]
Abstract
B lymphopoiesis declines with age, and this decline correlates with increased adipose tissue in the bone marrow (BM). Also, adipocyte-derived factors are known to inhibit B lymphopoiesis. Using cocultures of mouse BM cells with OP9 stromal cells, we found that adipocyte-conditioned medium induces the generation of CD11b(+)Gr1(+) myeloid cells, which inhibit B cell development in vitro. Adipocyte-conditioned medium-induced CD11b(+)Gr1(+) cells express Arg1 (arginase) and Nos2 (inducible NO synthase) and suppress CD4(+) T cell proliferation, indicating that these cells are myeloid-derived suppressor cells (MDSCs). Blocking arginase and inducible NO synthase did not restore B lymphopoiesis, indicating that inhibition is not mediated by these molecules. Transwell and conditioned-medium experiments showed that MDSCs inhibit B lymphopoiesis via soluble factors, and by cytokine array we identified IL-1 as an important factor. Addition of anti-IL-1 Abs restored B lymphopoiesis in BM cultures containing MDSCs, showing that MDSC inhibition of B lymphopoiesis is mediated by IL-1. By treating hematopoietic precursors with IL-1, we found that multipotent progenitors are targets of IL-1. This study uncovers a novel function for MDSCs to inhibit B lymphopoiesis through IL-1. We suggest that inflammaging contributes to a decline of B lymphopoiesis in aged individuals, and furthermore, that MDSCs and IL-1 provide therapeutic targets for restoration of B lymphopoiesis in aged and obese individuals.
Collapse
Affiliation(s)
- Domenick E Kennedy
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, IL 60153
| | - Katherine L Knight
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, IL 60153
| |
Collapse
|
37
|
Myeloid-derived suppressor cells in B cell malignancies. Tumour Biol 2015; 36:7339-53. [DOI: 10.1007/s13277-015-4004-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 08/25/2015] [Indexed: 02/07/2023] Open
|
38
|
O'Connor MA, Fu WW, Green KA, Green WR. Subpopulations of M-MDSCs from mice infected by an immunodeficiency-causing retrovirus and their differential suppression of T- vs B-cell responses. Virology 2015; 485:263-73. [PMID: 26318248 DOI: 10.1016/j.virol.2015.07.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 07/27/2015] [Accepted: 07/31/2015] [Indexed: 01/05/2023]
Abstract
Monocytic (CD11b(+)Ly6G(±/Lo)Ly6C(+)) myeloid derived suppressor cells (M-MDSCs) expand following murine retroviral LP-BM5 infection and suppress ex vivo polyclonal T-cell and B-cell responses. M-MDSCs 3 weeks post LP-BM5 infection have decreased suppression of T-cell, but not B-cell, responses and alterations in the degree of iNOS/NO dependence of suppression. M-MDSCs from LP-BM5 infected mice were sorted into four quadrant populations (Ly6C/CD11b density): all quadrants suppressed B-cell responses, but only M-MDSCs expressing the highest levels of Ly6C and CD11b (Q2) significantly suppressed T-cell responses. Further subdivision of this Q2 population revealed the Ly6C(+/Hi) M-MDSC subpopulation as the most suppressive, inhibiting T- and B-cell responses in a full, or partially, iNOS/NO-dependent manner, respectively. In contrast, the lower/moderate levels of suppression by the Ly6C(+/Lo) and Ly6C(+/Mid) M-MDSC Q2 subpopulations, whether versus T- and/or B-cells, displayed little/no iNOS dependency for suppression. These results highlight differential phenotypic and functional immunosuppressive M-MDSC subsets in a retroviral immunodeficiency model.
Collapse
Affiliation(s)
- Megan A O'Connor
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Whitney W Fu
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Kathy A Green
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - William R Green
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA; Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA.
| |
Collapse
|
39
|
O'Connor MA, Vella JL, Green WR. Reciprocal relationship of T regulatory cells and monocytic myeloid-derived suppressor cells in LP-BM5 murine retrovirus-induced immunodeficiency. J Gen Virol 2015; 97:509-522. [PMID: 26253145 DOI: 10.1099/jgv.0.000260] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Immunomodulatory cellular subsets, including myeloid-derived suppressor cells (MDSCs) and T regulatory cells (Tregs), contribute to the immunosuppressive tumour microenvironment and are targets of immunotherapy, but their role in retroviral-associated immunosuppression is less well understood. Due to known crosstalk between Tregs and MDSCs in the tumour microenvironment, and also their hypothesized involvement during human immunodeficiency virus/simian immunodeficiency virus infection, studying the interplay between these immune cells during LP-BM5 retrovirus-induced murine AIDS is of interest. IL-10-producing FoxP3+ Tregs expanded after LP-BM5 infection. Following in vivo adoptive transfer of natural Treg (nTreg)-depleted CD4+T-cells, and subsequent LP-BM5 retroviral infection, enriched monocytic MDSCs (M-MDSCs) from these nTreg-depleted mice displayed altered phenotypic subsets. In addition, M-MDSCs from LP-BM5-infected nTreg-depleted mice exhibited increased suppression of T-cell, but not B-cell, responses, compared with M-MDSCs derived from non-depleted LP-BM5-infected controls. Additionally, LP-BM5-induced M-MDSCs modulated the production of IL-10 by FoxP3+ Tregs in vitro. These collective data highlight in vitro and for the first time, to the best of our knowledge, in vivo reciprocal modulation between retroviral-induced M-MDSCs and Tregs, and may provide insight into the immunotherapeutic targeting of such regulatory cells during retroviral infection.
Collapse
Affiliation(s)
- Megan A O'Connor
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Jennifer L Vella
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - William R Green
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA.,Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| |
Collapse
|
40
|
Selective Involvement of the Checkpoint Regulator VISTA in Suppression of B-Cell, but Not T-Cell, Responsiveness by Monocytic Myeloid-Derived Suppressor Cells from Mice Infected with an Immunodeficiency-Causing Retrovirus. J Virol 2015; 89:9693-8. [PMID: 26157131 DOI: 10.1128/jvi.00888-15] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 06/30/2015] [Indexed: 12/20/2022] Open
Abstract
Inhibition of T-cell responses in tumor microenvironments by myeloid-derived suppressor cells (MDSCs) is widely accepted. We demonstrated augmentation of monocytic MDSCs whose suppression of not only T-cell, but also B-cell, responsiveness paralleled the immunodeficiency during LP-BM5 retrovirus infection. MDSCs inhibited T cells by inducible nitric oxide synthase (iNOS)/nitric oxide (NO), but uniquely, inhibition of B cells was ~50% dependent each on iNOS/NO and the MDSC-expressed negative-checkpoint regulator VISTA. Blockade with a combination of iNOS/NO and VISTA caused additive or synergistic abrogation of MDSC-mediated suppression of B-cell responsiveness.
Collapse
|
41
|
Wang J, McGuire TR, Britton HC, Schwarz JK, Loberiza FR, Meza JL, Talmadge JE. Lenalidomide and cyclophosphamide immunoregulation in patients with metastatic, castration-resistant prostate cancer. Clin Exp Metastasis 2015; 32:111-24. [DOI: 10.1007/s10585-015-9696-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 01/13/2015] [Indexed: 11/29/2022]
|
42
|
Myeloid-derived suppressor cells impair alveolar macrophages through PD-1 receptor ligation during Pneumocystis pneumonia. Infect Immun 2014; 83:572-82. [PMID: 25404033 DOI: 10.1128/iai.02686-14] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) were recently found to accumulate in the lungs during Pneumocystis pneumonia (PcP). Adoptive transfer of these cells caused lung damage in recipient mice, suggesting that MDSC accumulation is a mechanism of pathogenesis in PcP. In this study, the phagocytic activity of alveolar macrophages (AMs) was found to decrease by 40% when they were incubated with MDSCs from Pneumocystis-infected mice compared to those incubated with Gr-1(+) cells from the bone marrow of uninfected mice. The expression of the PU.1 gene in AMs incubated with MDSCs also was decreased. This PU.1 downregulation was due mainly to decreased histone 3 acetylation and increased DNA methylation caused by MDSCs. MDSCs were found to express high levels of PD-L1, and alveolar macrophages (AMs) were found to express high levels of PD-1 during PcP. Furthermore, PD-1 expression in AMs from uninfected mice was increased by 18-fold when they were incubated with MDSCs compared to those incubated with Gr-1(+) cells from the bone marrow of uninfected mice. The adverse effects of MDSCs on AMs were diminished when the MDSCs were pretreated with anti-PD-L1 antibody, suggesting that MDSCs disable AMs through PD-1/PD-L1 ligation during PcP.
Collapse
|
43
|
Burrack KS, Morrison TE. The role of myeloid cell activation and arginine metabolism in the pathogenesis of virus-induced diseases. Front Immunol 2014; 5:428. [PMID: 25250029 PMCID: PMC4157561 DOI: 10.3389/fimmu.2014.00428] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 08/22/2014] [Indexed: 12/25/2022] Open
Abstract
When an antiviral immune response is generated, a balance must be reached between two opposing pathways: the production of proinflammatory and cytotoxic effectors that drive a robust antiviral immune response to control the infection and regulators that function to limit or blunt an excessive immune response to minimize immune-mediated pathology and repair tissue damage. Myeloid cells, including monocytes and macrophages, play an important role in this balance, particularly through the activities of the arginine-hydrolyzing enzymes nitric oxide synthase 2 (Nos2; iNOS) and arginase 1 (Arg1). Nitric oxide (NO) production by iNOS is an important proinflammatory mediator, whereas Arg1-expressing macrophages contribute to the resolution of inflammation and wound repair. In the context of viral infections, expression of these enzymes can result in a variety of outcomes for the host. NO has direct antiviral properties against some viruses, whereas during other virus infections NO can mediate immunopathology and/or inhibit the antiviral immune response to promote chronic infection. Arg1 activity not only has important wound healing functions but can also inhibit the antiviral immune response during some viral infections. Thus, depending on the specific virus and the tissue(s) involved, the activity of both of these arginine-hydrolyzing enzymes can either exacerbate or limit the severity of virus-induced disease. In this review, we will discuss a variety of viral infections, including HIV, SARS-CoV, LCMV, HCV, RSV, and others, where myeloid cells influence the control and clearance of the virus from the host, as well as the severity and resolution of tissue damage, via the activities of iNOS and/or Arg1. Clearly, monocyte/macrophage activation and arginine metabolism will continue to be important areas of investigation in the context of viral infections.
Collapse
Affiliation(s)
- Kristina S Burrack
- Department of Immunology and Microbiology, University of Colorado School of Medicine , Aurora, CO , USA
| | - Thomas E Morrison
- Department of Immunology and Microbiology, University of Colorado School of Medicine , Aurora, CO , USA
| |
Collapse
|
44
|
Thorn M, Point GR, Burga RA, Nguyen CT, Joseph Espat N, Katz SC. Liver metastases induce reversible hepatic B cell dysfunction mediated by Gr-1+CD11b+ myeloid cells. J Leukoc Biol 2014; 96:883-94. [PMID: 25085111 DOI: 10.1189/jlb.3a0114-012rr] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
LM escape immune surveillance, in part, as a result of the expansion of CD11b+MC, which alter the intrahepatic microenvironment to promote tumor tolerance. HBC make up a significant proportion of liver lymphocytes and appear to delay tumor progression; however, their significance in the setting of LM is poorly defined. Therefore, we characterized HBC and HBC/CD11b+MC interactions using a murine model of LM. Tumor-bearing livers showed a trend toward elevated absolute numbers of CD19+ HBC. A significant increase in the frequency of IgM(lo)IgD(hi) mature HBC was observed in mice with LM compared with normal mice. HBC derived from tumor-bearing mice demonstrated increased proliferation in response to TLR and BCR stimulation ex vivo compared with HBC from normal livers. HBC from tumor-bearing livers exhibited significant down-regulation of CD80 and were impaired in inducing CD4(+) T cell proliferation ex vivo. We implicated hepatic CD11b+MC as mediators of CD80 down-modulation on HBC ex vivo via a CD11b-dependent mechanism that required cell-to-cell contact and STAT3 activity. Therefore, CD11b+MC may compromise the ability of HBC to promote T cell activation in the setting of LM as a result of diminished expression of CD80. Cross-talk between CD11b+MC and HBC may be an important component of LM-induced immunosuppression.
Collapse
Affiliation(s)
- Mitchell Thorn
- Roger Williams Medical Center, Department of Surgery, Providence, Rhode Island, USA; and Boston University School of Medicine, Boston, Massachusetts, USA
| | - Gary R Point
- Roger Williams Medical Center, Department of Surgery, Providence, Rhode Island, USA; and
| | - Rachel A Burga
- Roger Williams Medical Center, Department of Surgery, Providence, Rhode Island, USA; and
| | - Cang T Nguyen
- Roger Williams Medical Center, Department of Surgery, Providence, Rhode Island, USA; and
| | - N Joseph Espat
- Roger Williams Medical Center, Department of Surgery, Providence, Rhode Island, USA; and Boston University School of Medicine, Boston, Massachusetts, USA
| | - Steven C Katz
- Roger Williams Medical Center, Department of Surgery, Providence, Rhode Island, USA; and Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
45
|
Li Y, Tu Z, Qian S, Fung JJ, Markowitz SD, Kusner LL, Kaminski HJ, Lu L, Lin F. Myeloid-derived suppressor cells as a potential therapy for experimental autoimmune myasthenia gravis. THE JOURNAL OF IMMUNOLOGY 2014; 193:2127-34. [PMID: 25057008 DOI: 10.4049/jimmunol.1400857] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
We recently demonstrated that hepatic stellate cells induce the differentiation of myeloid-derived suppressor cells (MDSCs) from myeloid progenitors. In this study, we found that adoptive transfer of these MDSCs effectively reversed disease progression in experimental autoimmune myasthenia gravis (EAMG), a T cell-dependent and B cell-mediated model for myasthenia gravis. In addition to ameliorated disease severity, MDSC-treated EAMG mice showed suppressed acetylcholine receptor (AChR)-specific T cell responses, decreased levels of serum anti-AChR IgGs, and reduced complement activation at the neuromuscular junctions. Incubating MDSCs with B cells activated by anti-IgM or anti-CD40 Abs inhibited the proliferation of these in vitro-activated B cells. Administering MDSCs into mice immunized with a T cell-independent Ag inhibited the Ag-specific Ab production in vivo. MDSCs directly inhibit B cells through multiple mechanisms, including PGE2, inducible NO synthase, and arginase. Interestingly, MDSC treatment in EAMG mice does not appear to significantly inhibit their immune response to a nonrelevant Ag, OVA. These results demonstrated that hepatic stellate cell-induced MDSCs concurrently suppress both T and B cell autoimmunity, leading to effective treatment of established EAMG, and that the MDSCs inhibit AChR-specific immune responses at least partially in an Ag-specific manner. These data suggest that MDSCs could be further developed as a novel approach to treating myasthenia gravis and, even more broadly, other diseases in which T and B cells are involved in pathogenesis.
Collapse
Affiliation(s)
- Yan Li
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Zhidan Tu
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106
| | - Shiguang Qian
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - John J Fung
- Department of Surgery, Cleveland Clinic, Cleveland, OH 44195
| | - Sanford D Markowitz
- Department of Hematology and Oncology, Case Western Reserve University, Cleveland, OH 44106
| | - Linda L Kusner
- Department of Pharmacology and Physiology, George Washington University, Washington, DC 20037; and
| | - Henry J Kaminski
- Department of Pharmacology and Physiology, George Washington University, Washington, DC 20037; and Department of Neurology, George Washington University, Washington, DC 20037
| | - Lina Lu
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Feng Lin
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195; Department of Pathology, Case Western Reserve University, Cleveland, OH 44106;
| |
Collapse
|
46
|
O'Connor MA, Green WR. The role of indoleamine 2,3-dioxygenase in LP-BPM5 murine retroviral disease progression. Virol J 2013; 10:154. [PMID: 23680027 PMCID: PMC3751850 DOI: 10.1186/1743-422x-10-154] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Accepted: 05/06/2013] [Indexed: 11/10/2022] Open
Abstract
Background Indoleamine 2,3-dioxygenase (IDO) is an immunomodulatory intracellular enzyme involved in tryptophan degradation. IDO is induced during cancer and microbial infections by cytokines, ligation of co-stimulatory molecules and/or activation of pattern recognition receptors, ultimately leading to modulation of the immune response. LP-BM5 murine retroviral infection induces murine AIDS (MAIDS), which is characterized by profound and broad immunosuppression of T- and B-cell responses. Our lab has previously described multiple mechanisms regulating the development of immunodeficiency of LP-BM5-induced disease, including Programmed Death 1 (PD-1), IL-10, and T-regulatory (Treg) cells. Immunosuppressive roles of IDO have been demonstrated in other retroviral models, suggesting a possible role for IDO during LP-BM5-induced retroviral disease progression and/or development of viral load. Methods Mice deficient in IDO (B6.IDO−/−) and wildtype C57BL/6 (B6) mice were infected with LP-BM5 murine retrovirus. MAIDS and LP-BM5 viral load were assessed at termination. Results As expected, IDO was un-inducible in B6.IDO−/− during LP-BM5 infection. B6.IDO−/− mice infected with LP-BM5 retrovirus succumbed to MAIDS as indicated by splenomegaly, serum hyper IgG2a and IgM, decreased responsiveness to B- and T-cell mitogens, conversion of a proportion of CD4+ T cells from Thy1.2+ to Thy1.2-, and increased percentages of CD11b+Gr-1+ cells. LP-BM5 infected B6.IDO−/− mice also demonstrated the development of roughly equivalent disease kinetics as compared to infected B6 mice. Splenic viral loads of B6 and B6.IDO−/− mice were also equivalent after infection as measured by LP-BM5-specific Def Gag and Eco Gag viral mRNA, determined by qRT-PCR. Conclusions Collectively, these results demonstrate IDO neither plays an essential role, nor is required, in LP-BM5-induced disease progression or LP-BM5 viral load.
Collapse
Affiliation(s)
- Megan A O'Connor
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire 03756, USA
| | | |
Collapse
|