1
|
Kenswil KJG, Pisterzi P, Feyen J, Ter Borg M, Rombouts E, Braakman E, Raaijmakers MHGP. Immune composition and its association with hematologic recovery after chemotherapeutic injury in acute myeloid leukemia. Exp Hematol 2021; 105:32-38.e2. [PMID: 34800603 DOI: 10.1016/j.exphem.2021.11.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 10/15/2021] [Accepted: 11/10/2021] [Indexed: 11/25/2022]
Abstract
Chemotherapy-induced bone marrow (BM) injury is a significant cause of morbidity and mortality in acute myeloid leukemia (AML). Time to hematologic recovery after standard ("7 + 3") myeloablative chemotherapy can vary considerably among patients, but the factors that drive or predict BM recovery remain incompletely understood. Here, we assessed the composition of innate and adaptive immune subsets in the regenerating BM (day 17) after induction chemotherapy and related it to hematologic recovery in AML. T cells, and in particular the CD4 central memory (CD4CM) T-cell subset, were significantly enriched in the BM after chemotherapy, suggesting the relative chemoresistance of cells providing long-term memory for systemic pathogens. In contrast, B cells and other hematopoietic subsets were depleted. Higher frequencies of the CD4CM T-cell subset were associated with delayed hematopoietic recovery, whereas a high frequency of natural killer (NK) cells was related to faster recovery of neutrophil counts. The NK/CD4CM ratio in the BM after chemotherapy was significantly associated with the time to subsequent neutrophil recovery (Spearman's ρ = -0.723, p < 0.001, false discovery rate <0.01). The data provide novel insights into adaptive immune cell recovery after injury and identify the NK/CD4CM index as a putative predictor of hematopoietic recovery in AML.
Collapse
Affiliation(s)
| | - Paola Pisterzi
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Jacqueline Feyen
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Mariëtte Ter Borg
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Elwin Rombouts
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Eric Braakman
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | | |
Collapse
|
2
|
Innamarato P, Asby S, Morse J, Mackay A, Hall M, Kidd S, Nagle L, Sarnaik AA, Pilon-Thomas S. Intratumoral Activation of 41BB Costimulatory Signals Enhances CD8 T Cell Expansion and Modulates Tumor-Infiltrating Myeloid Cells. THE JOURNAL OF IMMUNOLOGY 2020; 205:2893-2904. [PMID: 33020146 DOI: 10.4049/jimmunol.2000759] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/04/2020] [Indexed: 12/11/2022]
Abstract
The activation of 41BB costimulatory signals by agonistic Abs enhances the expansion and function of tumor-infiltrating lymphocytes (TILs) for treating cancer patients with adoptive cell therapy. However, the impact of 41BB agonism is not limited to enhancing the activity of T cells, and the mechanism by which additional activation of this costimulatory axis in tumor-associated myeloid cells is poorly understood. In this study, we describe that the intratumoral administration of 41BB agonistic Abs led to increases in CD8 T cell infiltration followed by tumor regression in murine models. We found that granulocytes and monocytes rapidly replaced macrophages and dendritic cells in tumors following administration of anti-41BB Abs. Overall, myeloid cells from anti-41BB-treated tumors had an improved capacity to stimulate T cells in comparison with control-treated tumors. In human coculture systems, we demonstrated that the agonism of the 41BB-41BBL axis enhanced costimulatory signals and effector functions among APC and autologous TILs. Overall, these findings suggest that the effect of 41BB agonistic Abs are supported by additional costimulatory signals from tumor-associated myeloid cells,v leading to enhanced TIL expansion and function.
Collapse
Affiliation(s)
- Patrick Innamarato
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612.,Cancer Biology Ph.D. Program, University of South Florida, Tampa, FL 33620; and
| | - Sarah Asby
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612
| | - Jennifer Morse
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612
| | - Amy Mackay
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612
| | - MacLean Hall
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612.,Cancer Biology Ph.D. Program, University of South Florida, Tampa, FL 33620; and
| | - Scott Kidd
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612
| | - Luz Nagle
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612
| | - Amod A Sarnaik
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612.,Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center, Tampa, FL 33612
| | - Shari Pilon-Thomas
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612;
| |
Collapse
|
3
|
Mak A, Dharmadhikari B, Kow NY, Thamboo TP, Tang Q, Wong LW, Sajikumar S, Wong HY, Schwarz H. Deletion of CD137 Ligand Exacerbates Renal and Cutaneous but Alleviates Cerebral Manifestations in Lupus. Front Immunol 2019; 10:1411. [PMID: 31297111 PMCID: PMC6607944 DOI: 10.3389/fimmu.2019.01411] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 06/04/2019] [Indexed: 01/01/2023] Open
Abstract
The CD137—CD137 ligand (CD137L) costimulatory system is a critical immune checkpoint with pathophysiological implications in autoimmunity. In this study, we investigated the role of CD137L-mediated costimulation on renal, cutaneous and cerebral manifestations in lupus and the underlying immunological mechanism. Lupus-prone C57BL/6lpr−/− (B6.lpr) mice were crossed to C57BL/6.CD137L−/− mice to obtain CD137L-deficient B6.lpr [double knock out (DKO)] mice. We investigated the extent of survival, glomerulonephritis, skin lesions, cerebral demyelination, immune deviation and long-term synaptic plasticity among the two mouse groups. Cytokine levels, frequency of splenic leukocyte subsets and phenotypes were compared between DKO, B6.lpr and B6.WT mice. A 22 month observation of 226 DKO and 137 B6.lpr mice demonstrated significantly more frequent proliferative glomerulonephritis, larger skin lesions and shorter survival in DKO than in B6.lpr mice. Conversely, microglial activation and cerebral demyelination were less pronounced while long-term synaptic plasticity, was superior in DKO mice. Splenic Th17 cells were significantly higher in DKO than in B6.lpr and B6.WT mice while Th1 and Th2 cell frequencies were comparable between DKO and B6.lpr mice. IL-10 and IL-17 expression by T cells was not affected but there were fewer IL-10-producing myeloid (CD11b+) cells, and also lower serum IL-10 levels in DKO than in B6.lpr mice. The absence of CD137L causes an immune deviation toward Th17, fewer IL-10-producing CD11b+ cells and reduced serum IL-10 levels which potentially explain the more severe lupus in DKO mice while leading to reduced microglia activation, lesser cerebral damage and less severe neurological deficits.
Collapse
Affiliation(s)
- Anselm Mak
- Department of Physiology, National University of Singapore, Singapore, Singapore.,Department of Medicine, National University of Singapore, Singapore, Singapore.,Division of Rheumatology, University Medicine Cluster, National University Health System, Singapore, Singapore
| | - Bhushan Dharmadhikari
- Department of Physiology, National University of Singapore, Singapore, Singapore.,Immunlogy Programme, National University of Singapore, Singapore, Singapore
| | - Nien Yee Kow
- Department of Medicine, National University of Singapore, Singapore, Singapore
| | | | - Qianqiao Tang
- Department of Physiology, National University of Singapore, Singapore, Singapore.,Immunlogy Programme, National University of Singapore, Singapore, Singapore
| | - Lik Wei Wong
- Department of Physiology, National University of Singapore, Singapore, Singapore
| | - Sreedharan Sajikumar
- Department of Physiology, National University of Singapore, Singapore, Singapore
| | - Hiu Yi Wong
- Department of Physiology, National University of Singapore, Singapore, Singapore.,Immunlogy Programme, National University of Singapore, Singapore, Singapore
| | - Herbert Schwarz
- Department of Physiology, National University of Singapore, Singapore, Singapore.,Immunlogy Programme, National University of Singapore, Singapore, Singapore
| |
Collapse
|
4
|
Beshara R, Sencio V, Soulard D, Barthélémy A, Fontaine J, Pinteau T, Deruyter L, Ismail MB, Paget C, Sirard JC, Trottein F, Faveeuw C. Alteration of Flt3-Ligand-dependent de novo generation of conventional dendritic cells during influenza infection contributes to respiratory bacterial superinfection. PLoS Pathog 2018; 14:e1007360. [PMID: 30372491 PMCID: PMC6224179 DOI: 10.1371/journal.ppat.1007360] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 11/08/2018] [Accepted: 09/27/2018] [Indexed: 01/08/2023] Open
Abstract
Secondary bacterial infections contribute to the excess morbidity and mortality of influenza A virus (IAV) infection. Disruption of lung integrity and impaired antibacterial immunity during IAV infection participate in colonization and dissemination of the bacteria out of the lungs. One key feature of IAV infection is the profound alteration of lung myeloid cells, characterized by the recruitment of deleterious inflammatory monocytes. We herein report that IAV infection causes a transient decrease of lung conventional dendritic cells (cDCs) (both cDC1 and cDC2) peaking at day 7 post-infection. While triggering emergency monopoiesis, IAV transiently altered the differentiation of cDCs in the bone marrow, the cDC1-biaised pre-DCs being particularly affected. The impaired cDC differentiation during IAV infection was independent of type I interferons (IFNs), IFN-γ, TNFα and IL-6 and was not due to an intrinsic dysfunction of cDC precursors. The alteration of cDC differentiation was associated with a drop of local and systemic production of Fms-like tyrosine kinase 3 ligand (Flt3-L), a critical cDC differentiation factor. Overexpression of Flt3-L during IAV infection boosted the cDC progenitors' production in the BM, replenished cDCs in the lungs, decreased inflammatory monocytes' infiltration and lowered lung damages. This was associated with partial protection against secondary pneumococcal infection, as reflected by reduced bacterial dissemination and prolonged survival. These findings highlight the impact of distal viral infection on cDC genesis in the BM and suggest that Flt3-L may have potential applications in the control of secondary infections.
Collapse
Affiliation(s)
- Ranin Beshara
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale U1019, Lille, France
- Centre Hospitalier Universitaire de Lille, Lille, France
- Institut Pasteur de Lille, Lille, France
- Laboratoire Microbiologie Santé et Environnement (LMSE), Ecole Doctorale des Sciences et de Technologie, Faculté de Santé Publique, Université Libanaise, Tripoli, Lebanon
| | - Valentin Sencio
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale U1019, Lille, France
- Centre Hospitalier Universitaire de Lille, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Daphnée Soulard
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale U1019, Lille, France
- Centre Hospitalier Universitaire de Lille, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Adeline Barthélémy
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale U1019, Lille, France
- Centre Hospitalier Universitaire de Lille, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Josette Fontaine
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale U1019, Lille, France
- Centre Hospitalier Universitaire de Lille, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Thibault Pinteau
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale U1019, Lille, France
- Centre Hospitalier Universitaire de Lille, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Lucie Deruyter
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale U1019, Lille, France
- Centre Hospitalier Universitaire de Lille, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Mohamad Bachar Ismail
- Laboratoire Microbiologie Santé et Environnement (LMSE), Ecole Doctorale des Sciences et de Technologie, Faculté de Santé Publique, Université Libanaise, Tripoli, Lebanon
| | - Christophe Paget
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale U1019, Lille, France
- Centre Hospitalier Universitaire de Lille, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Jean-Claude Sirard
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale U1019, Lille, France
- Centre Hospitalier Universitaire de Lille, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - François Trottein
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale U1019, Lille, France
- Centre Hospitalier Universitaire de Lille, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Christelle Faveeuw
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale U1019, Lille, France
- Centre Hospitalier Universitaire de Lille, Lille, France
- Institut Pasteur de Lille, Lille, France
| |
Collapse
|
5
|
Harfuddin Z, Dharmadhikari B, Wong SC, Duan K, Poidinger M, Kwajah S, Schwarz H. Transcriptional and functional characterization of CD137L-dendritic cells identifies a novel dendritic cell phenotype. Sci Rep 2016; 6:29712. [PMID: 27431276 PMCID: PMC4949477 DOI: 10.1038/srep29712] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 06/17/2016] [Indexed: 12/24/2022] Open
Abstract
The importance of monocyte-derived dendritic cells (DCs) is evidenced by the fact that they are essential for the elimination of pathogens. Although in vitro DCs can be generated by treatment of monocytes with GM-CSF and IL-4, it is unknown what stimuli induce differentiation of DCs in vivo. CD137L-DCs are human monocyte-derived DC that are generated by CD137 ligand (CD137L) signaling. We demonstrate that the gene signature of in vitro generated CD137L-DCs is most similar to those of GM-CSF and IL-4-generated immature DCs and of macrophages. This is reminiscent of in vivo inflammatory DC which also have been reported to share gene signatures with monocyte-derived DCs and macrophages. Performing direct comparison of deposited human gene expression data with a CD137L-DC dataset revealed a significant enrichment of CD137L-DC signature genes in inflammatory in vivo DCs. In addition, surface marker expression and cytokine secretion by CD137L-DCs resemble closely those of inflammatory DCs. Further, CD137L-DCs express high levels of adhesion molecules, display strong attachment, and employ the adhesion molecule ALCAM to stimulate T cell proliferation. This study characterizes the gene expression profile of CD137L-DCs, and identifies significant similarities of CD137L-DCs with in vivo inflammatory monocyte-derived DCs and macrophages.
Collapse
Affiliation(s)
- Zulkarnain Harfuddin
- Department of Physiology, National University of Singapore, Singapore.,NUS Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore.,NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore
| | - Bhushan Dharmadhikari
- Department of Physiology, National University of Singapore, Singapore.,NUS Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore
| | - Siew Cheng Wong
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore
| | - Kaibo Duan
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore
| | - Michael Poidinger
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore
| | - Shaqireen Kwajah
- Department of Physiology, National University of Singapore, Singapore.,NUS Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore
| | - Herbert Schwarz
- Department of Physiology, National University of Singapore, Singapore.,NUS Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore.,NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore
| |
Collapse
|
6
|
Lakritz JR, Robinson JA, Polydefkis MJ, Miller AD, Burdo TH. Loss of intraepidermal nerve fiber density during SIV peripheral neuropathy is mediated by monocyte activation and elevated monocyte chemotactic proteins. J Neuroinflammation 2015; 12:237. [PMID: 26683323 PMCID: PMC4683776 DOI: 10.1186/s12974-015-0456-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 12/11/2015] [Indexed: 12/25/2022] Open
Abstract
Background Peripheral neuropathy (PN) continues to be a major complication of human immunodeficiency virus (HIV) infection despite successful anti-retroviral therapy. Human HIV-PN can be recapitulated in a CD8-depleted, simian immunodeficiency virus (SIV)-infected rhesus macaque animal model, characterized by a loss of intraepidermal nerve fiber density (IENFD) and damage to the dorsal root ganglia (DRG). Increased monocyte traffic to the DRG has previously been associated with severe DRG pathology, as well as a loss in IENFD. Here, we sought to characterize the molecular signals associated with monocyte activation and trafficking to the DRGs. Methods Eleven SIV-infected CD8-depleted rhesus macaques were compared to four uninfected control animals. sCD14, sCD163, sCD137, regulated on activation normal T cell expressed and secreted (RANTES), and monocyte chemoattractant protein 1 (MCP-1) were measured in plasma and the latter three proteins were also quantified in DRG tissue lysates. All SIV-infected animals received serial skin biopsies to measure IENFD loss as well as BrdU inoculations to measure monocyte turnover during the course of infection. The number of BrdU+ and CD14+ CD16+ peripheral blood monocytes was determined by flow cytometry. The number of MAC387+, CCR2+, CCR5+, and CD137+ cells in DRG tissue was quantified by immunohistochemistry. Results sCD14, sCD163, MCP-1, and sCD137 increased significantly in plasma from pre-infection to necropsy. Plasma sCD163 and RANTES inversely correlated with IENFD. Additionally, sCD137 in DRG tissue lysate was elevated with severe DRG pathology and associated with the recruitment of MAC387+ cells to DRG. Elevated numbers of CCR5+ and CCR2+ satellite cells in the DRG were found, suggesting a chemotactic role of their ligands, RANTES, and MCP-1 in recruiting monocytes to the tissue. Conclusions We characterized the role of systemic (plasma) and tissue-specific (DRG) monocyte activation and associated cytokines in the pathogenesis of SIV-PN. We identified sCD163 and RANTES as potential biomarkers for HIV-PN, as these were associated with a loss of IENFD. Additionally, we identified CD137 signaling to play a role in MAC387+ cell traffic to DRG and possibly contribute to severe pathology. These studies highlight the role of monocyte activation and traffic in the pathogenesis of SIV-PN, while identifying specific signaling proteins for future pharmacological blockade.
Collapse
Affiliation(s)
- Jessica R Lakritz
- Department of Biology, Boston College, Chestnut Hill, MA, 02467, USA
| | - Jake A Robinson
- Department of Biology, Boston College, Chestnut Hill, MA, 02467, USA
| | | | - Andrew D Miller
- Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, NY, 14853, USA
| | - Tricia H Burdo
- Department of Biology, Boston College, Chestnut Hill, MA, 02467, USA.
| |
Collapse
|
7
|
Dharmadhikari B, Wu M, Abdullah NS, Rajendran S, Ishak ND, Nickles E, Harfuddin Z, Schwarz H. CD137 and CD137L signals are main drivers of type 1, cell-mediated immune responses. Oncoimmunology 2015; 5:e1113367. [PMID: 27141396 DOI: 10.1080/2162402x.2015.1113367] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 10/22/2015] [Accepted: 10/22/2015] [Indexed: 10/22/2022] Open
Abstract
CD137 is expressed on activated T cells and NK cells, among others, and is a potent co-stimulator of antitumor immune responses. CD137 ligand (CD137L) is expressed by antigen presenting cells (APC), and CD137L reverse signaling into APC enhances their activity. CD137-CD137L interactions as main driver of type 1, cell-mediated immune responses explains the puzzling observation that CD137 agonists which enhance antitumor immune responses also ameliorate autoimmune diseases. Upon co-stimulation by CD137, Th1 CD4+ T cells together with Tc1 CD8+ T cells and NK cells inhibit other T cell subsets, thereby promoting antitumor responses and mitigating non-type 1 auto-immune diseases.
Collapse
Affiliation(s)
- Bhushan Dharmadhikari
- Department of Physiology, and Immunology Programme, National University of Singapore , Singapore
| | - Meihui Wu
- Department of Physiology, and Immunology Programme, National University of Singapore , Singapore
| | - Nur Sharalyn Abdullah
- Department of Physiology, and Immunology Programme, National University of Singapore , Singapore
| | - Sakthi Rajendran
- Department of Physiology, and Immunology Programme, National University of Singapore , Singapore
| | - Nur Diana Ishak
- Department of Physiology, and Immunology Programme, National University of Singapore , Singapore
| | - Emily Nickles
- Department of Physiology, and Immunology Programme, National University of Singapore , Singapore
| | - Zulkarnain Harfuddin
- Department of Physiology, and Immunology Programme, National University of Singapore, Singapore; NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore
| | - Herbert Schwarz
- Department of Physiology, and Immunology Programme, National University of Singapore, Singapore; NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore
| |
Collapse
|
8
|
Kwon B. Is CD137 Ligand (CD137L) Signaling a Fine Tuner of Immune Responses? Immune Netw 2015; 15:121-4. [PMID: 26140043 PMCID: PMC4486774 DOI: 10.4110/in.2015.15.3.121] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 05/15/2015] [Accepted: 06/04/2015] [Indexed: 12/01/2022] Open
Abstract
Now, it has been being accepted that reverse signaling through CD137 ligand (CD137L) plays an important role in vivo during hematopoiesis and in immune regulation. However, due to technical difficulty in dissecting both directional signaling events simultaneously in vivo, most biological activities caused by CD137-CD137L interactions are considered as results from signaling events of the CD137 receptor. To make the story more complex, CD137(-/-) and CD137L(-/-) mice have increased or decreased immune responses in a context-dependent manner. In this Mini review, I will try to provide a plausible explanation for how CD137L signaling is controlled during immune responses.
Collapse
Affiliation(s)
- Byungsuk Kwon
- School of Biological Sciences, University of Ulsan, Ulsan 680-749, Korea
| |
Collapse
|
9
|
Tu TH, Kim CS, Nam-Goong IS, Nam CW, Kim YI, Goto T, Kawada T, Park T, Yoon Park JH, Ryoo ZY, Park JW, Choi HS, Yu R. 4-1BBL signaling promotes cell proliferation through reprogramming of glucose metabolism in monocytes/macrophages. FEBS J 2015; 282:1468-80. [DOI: 10.1111/febs.13236] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 02/11/2015] [Accepted: 02/13/2015] [Indexed: 02/06/2023]
Affiliation(s)
- Thai H. Tu
- Department of Food Science and Nutrition; University of Ulsan; South Korea
| | - Chu-Sook Kim
- Department of Food Science and Nutrition; University of Ulsan; South Korea
| | - Il S. Nam-Goong
- Department of Internal Medicine; Ulsan University Hospital; University of Ulsan College of Medicine; South Korea
| | - Chang W. Nam
- Department of Surgery; Ulsan University Hospital; University of Ulsan College of Medicine; South Korea
| | - Young-Il Kim
- Graduate School of Agriculture; Kyoto University; Uji Japan
| | - Tsuyoshi Goto
- Graduate School of Agriculture; Kyoto University; Uji Japan
| | - Teruo Kawada
- Graduate School of Agriculture; Kyoto University; Uji Japan
| | - Taesun Park
- Department of Food and Nutrition; Yonsei University; Seoul South Korea
| | - Jung H. Yoon Park
- Department of Food Science and Nutrition and Research Institute for Bioscience & Biotechnology; Hallym University; Chuncheon South Korea
| | - Zae Y. Ryoo
- School of Life Science and Biotechnology; Kyungpook National University; Daegu South Korea
| | - Jeong W. Park
- Department of Biological Sciences; University of Ulsan; South Korea
| | - Hye-Seon Choi
- Department of Biological Sciences; University of Ulsan; South Korea
| | - Rina Yu
- Department of Food Science and Nutrition; University of Ulsan; South Korea
| |
Collapse
|
10
|
Libregts SFWM, Nolte MA. Parallels between immune driven-hematopoiesis and T cell activation: 3 signals that relay inflammatory stress to the bone marrow. Exp Cell Res 2014; 329:239-47. [PMID: 25246130 DOI: 10.1016/j.yexcr.2014.09.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Revised: 09/08/2014] [Accepted: 09/11/2014] [Indexed: 12/27/2022]
Abstract
Quiescence, self-renewal, lineage commitment and differentiation of hematopoietic stem cells (HSCs) towards fully mature blood cells are a complex process that involves both intrinsic and extrinsic signals. During steady-state conditions, most hematopoietic signals are provided by various resident cells inside the bone marrow (BM), which establish the HSC micro-environment. However, upon infection, the hematopoietic process is also affected by pathogens and activated immune cells, which illustrates an effective feedback mechanism to hematopoietic stem and progenitor cells (HSPCs) via immune-mediated signals. Here, we review the impact of pathogen-associated molecular patterns (PAMPs), damage-associated molecular patterns (DAMPs), costimulatory molecules and pro-inflammatory cytokines on the quiescence, proliferation and differentiation of HSCs and more committed progenitors. As modulation of HSPC function via these immune-mediated signals holds an interesting parallel with the "three-signal-model" described for the activation and differentiation of naïve T-cells, we propose a novel "three-signal" concept for immune-driven hematopoiesis. In this model, the recognition of PAMPs and DAMPs will activate HSCs and induce proliferation, while costimulatory molecules and pro-inflammatory cytokines confer a second and third signal, respectively, which further regulate expansion, lineage commitment and differentiation of HSPCs. We review the impact of inflammatory stress on hematopoiesis along these three signals and we discuss whether they act independently from each other or that concurrence of these signals is important for an adequate response of HSPCs upon infection.
Collapse
Affiliation(s)
- Sten F W M Libregts
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands
| | - Martijn A Nolte
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands.
| |
Collapse
|
11
|
Tang Q, Jiang D, Harfuddin Z, Cheng K, Moh MC, Schwarz H. Regulation of myelopoiesis by CD137L signaling. Int Rev Immunol 2014; 33:454-69. [PMID: 24941289 DOI: 10.3109/08830185.2014.921163] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
CD137 ligand (CD137L) has emerged as a powerful regulator of myelopoiesis that links emergency situations, such as infections, to the generation of additional myeloid cells, and to their activation and maturation. CD137L is expressed on the cell surface of hematopoietic stem and progenitor cells (HSPC) and antigen presenting cells (APC) as a transmembrane protein. The signaling of CD137L into HSPC induces their proliferation and differentiation to monocytes and macrophages, and in monocytes CD137L signaling induces differentiation to potent dendritic cells (DC). CD137L signaling is initiated by CD137 which is expressed by T cells, once they become activated. Some of these activated, CD137-expressing T cells migrate from the site of infection to the bone marrow where they interact with HSPC to induce myelopoiesis, or they induce monocyte to DC differentiation locally at the site of infection. Therapeutically, induction of CD137L signaling can be utilized to reinitiate myeloid differentiation in acute myeloid leukemia cells, and to generate potent DC for immunotherapy.
Collapse
|
12
|
Tang Q, Koh LK, Jiang D, Schwarz H. CD137 ligand reverse signaling skews hematopoiesis towards myelopoiesis during aging. Aging (Albany NY) 2014; 5:643-52. [PMID: 23945137 PMCID: PMC3808697 DOI: 10.18632/aging.100588] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
CD137 is a costimulatory molecule expressed on activated T cells. Its ligand, CD137L, is expressed on the surface of hematopoietic progenitor cells, and upon binding to CD137 induces reverse signaling into hematopoietic progenitor cells promoting their activation, proliferation and myeloid differentiation. Since aging is associated with an increasing number of myeloid cells we investigated the role of CD137 and CD137L on myelopoiesis during aging. Comparing 3 and 12 months old WT, CD137−/− and CD137L−/− mice we found significantly more granulocytes and monocytes in the bone marrow of older WT mice, while this age-dependent increase was absent in CD137−/− and CD137L−/− mice. Instead, the bone marrow of 12 months old CD137−/− and CD137L−/− mice was characterized by an accumulation of hematopoietic progenitor cells, suggesting that the differentiation of hematopoietic progenitor cells became arrested in the absence of CD137L signaling. CD137L signaling is initiated by activated CD137-expressing, CD4+ T cells. These data identify a novel molecular mechanisms underlying immune aging by demonstrating that CD137-expressing CD4+ T cells in the bone marrow engage CD137L on hematopoietic progenitor cells, and that this CD137L signaling biases hematopoiesis towards myelopoiesis during aging.
Collapse
|
13
|
Cheng K, Wong SC, Linn YC, Ho LP, Chng WJ, Schwarz H. CD137 ligand signalling induces differentiation of primary acute myeloid leukaemia cells. Br J Haematol 2014; 165:134-44. [PMID: 24428589 DOI: 10.1111/bjh.12732] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 11/08/2013] [Indexed: 02/06/2023]
Abstract
CD137 ligand (CD137L), a member of the tumour necrosis factor family, is expressed as a cell surface molecule. Engagement of CD137L on haematopoietic progenitor cells induces monocytic differentiation, and in peripheral monocytes CD137L signalling promotes differentiation to mature dendritic cells. We hypothesized that CD137L signalling would also induce differentiation in transformed myeloid cells. Here we show that recombinant CD137 protein, which crosslinks CD137L and initiates reverse CD137L signalling in myeloid cells, induces morphological changes (adherence, spreading), loss of progenitor markers (CD117), expression of maturation markers (CD11b, CD13) and secretion of cytokines that are indicative of myeloid differentiation. Under the influence of CD137L signalling, acute myeloid leukaemia (AML) cells acquired expression of co-stimulatory molecules (CD80, CD86, CD40), the dendritic cell marker CD83 and dendritic cell activities, enabling them to stimulate T cells. CD137L signalling induced differentiation in 71% (15 of 21) of AML samples, irrespective of French-American-British classification and CD137L expression level. However, the type of response varied with the AML subtype and patient sample. In summary, this study demonstrated that CD137L signalling induced differentiation in malignant cells of AML patients, and suggests that it may be worthwhile to investigate treatment with recombinant CD137 protein as a potential novel therapeutic approach for AML.
Collapse
Affiliation(s)
- Kin Cheng
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore; Immunology Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore
| | | | | | | | | | | |
Collapse
|
14
|
Harfuddin Z, Kwajah S, Chong Nyi Sim A, Macary PA, Schwarz H. CD137L-stimulated dendritic cells are more potent than conventional dendritic cells at eliciting cytotoxic T-cell responses. Oncoimmunology 2013; 2:e26859. [PMID: 24482752 DOI: 10.4161/onci.26859] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Revised: 10/15/2013] [Accepted: 10/17/2013] [Indexed: 01/06/2023] Open
Abstract
Dendritic cells (DCs) are highly potent initiators of adaptive immune responses and, as such, represent promising tools for immunotherapeutic applications. Despite their potential, the current efficacy of DC-based immunotherapies is poor. CD137 ligand (CD137L) signaling has been used to derive a novel type of DCs from human peripheral blood monocytes, termed CD137L-DCs. Here, we report that CD137L-DCs induce more potent cytotoxic T-cell responses than classical DCs (cDCs). Furthermore, in exploring several DC maturation factors for their ability to enhance the potency of CD137L-DCs, we found the combination of interferon γ (IFNγ) and the mixed Toll-like receptor (TLR)7/8 agonist R848, to display the highest efficacy in potentiating the T-cell co-stimulatory activity of CD137L-DCs. Of particular importance, CD137L-DCs were found to be more efficient than cDCs in activating autologous T cells targeting the cytomegalovirus (CMV)-derived protein pp65. Specifically, CD137L-DC-stimulated T cells were found to secrete higher levels of IFNγ and killed 2-3 times more HLA-matched, pp65-pulsed target cells than T cells activated by cDCs. Finally, in addition to stimulating CD8+ T cells, CD137L-DCs efficiently activated CD4+ T cells. Taken together, these findings demonstrate the superior potency of CD137L-stimulated DCs in activating CMV-specific, autologous T cells, and encourage the further development of CD137L-DCs for antitumor immunotherapy.
Collapse
Affiliation(s)
- Zulkarnain Harfuddin
- Department of Physiology; National University of Singapore; Singapore ; Immunology Programme; National University of Singapore; Singapore ; NUS Graduate School for Integrative Sciences and Engineering; National University of Singapore; Singapore
| | - Shaqireen Kwajah
- Department of Physiology; National University of Singapore; Singapore
| | - Adrian Chong Nyi Sim
- Department of Microbiology; National University of Singapore; Singapore ; Immunology Programme; National University of Singapore; Singapore
| | - Paul Anthony Macary
- Department of Microbiology; National University of Singapore; Singapore ; Immunology Programme; National University of Singapore; Singapore ; NUS Graduate School for Integrative Sciences and Engineering; National University of Singapore; Singapore
| | - Herbert Schwarz
- Department of Physiology; National University of Singapore; Singapore ; Immunology Programme; National University of Singapore; Singapore ; NUS Graduate School for Integrative Sciences and Engineering; National University of Singapore; Singapore
| |
Collapse
|
15
|
Martínez Gómez JM, Chen L, Schwarz H, Karrasch T. CD137 facilitates the resolution of acute DSS-induced colonic inflammation in mice. PLoS One 2013; 8:e73277. [PMID: 24023849 PMCID: PMC3762711 DOI: 10.1371/journal.pone.0073277] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Accepted: 07/18/2013] [Indexed: 12/27/2022] Open
Abstract
Background CD137 and its ligand (CD137L) are potent immunoregulatory molecules that influence activation, proliferation, differentiation and cell death of leukocytes. Expression of CD137 is upregulated in the lamina propria cells of Crohn’s disease patients. Here, the role of CD137 in acute Dextran-Sodium-Sulfate (DSS)-induced colitis in mice was examined. Methods We induced acute large bowel inflammation (colitis) via DSS administration in CD137−/− and wild-type (WT) mice. Colitis severity was evaluated by clinical parameters (weight loss), cytokine secretion in colon segment cultures, and scoring of histological inflammatory parameters. Additionally, populations of lamina propria mononuclear cells (LPMNC) and intraepithelial lymphocytes (IEL) were characterized by flow cytometry. In a subset of mice, resolution of intestinal inflammation was evaluated 3 and 7 days after withdrawal of DSS. Results We found that both CD137−/− and WT mice demonstrated a similar degree of inflammation after 5 days of DSS exposure. However, the resolution of colonic inflammation was impaired in the absence of CD137. This was accompanied by a higher histological score of inflammation, and increased release of the pro-inflammatory mediators granulocyte macrophage colony-stimulating factor (GM-CSF), CXCL1, IL-17 and IFN-γ. Further, there were significantly more neutrophils among the LPMNC of CD137−/− mice, and reduced numbers of macrophages among the IEL. Conclusion We conclude that CD137 plays an essential role in the resolution of acute DSS-induced intestinal inflammation in mice.
Collapse
Affiliation(s)
- Julia M. Martínez Gómez
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Lieping Chen
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Herbert Schwarz
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- * E-mail: (HS); (TK)
| | - Thomas Karrasch
- Department of Internal Medicine I, University of Regensburg, Regensburg, Germany
- * E-mail: (HS); (TK)
| |
Collapse
|