1
|
Chen C, Wang H, Xu L, Guo Z, Fu M, Xia H, He Q, Zhang R, He J. MAPK signaling pathway induced LOX-1 + polymorphonuclear myeloid-derived suppressor cells in biliary atresia. Clin Immunol 2024; 268:110355. [PMID: 39237078 DOI: 10.1016/j.clim.2024.110355] [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: 05/27/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/07/2024]
Abstract
Biliary atresia (BA) is a severe pediatric liver disease characterized by progressive bile duct destruction and fibrosis, leading to significant liver damage and frequently necessitating liver transplantation. This study elucidates the role of LOX-1+ polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) in BA pathogenesis and assesses their potential as non-invasive early diagnostic biomarkers. Using flow cytometry, immunofluorescence, and molecular profiling, we analyzed the expression and activity of these cells in peripheral blood and liver tissues from BA patients and controls. Our findings reveal a significant increase in the frequencies and function of LOX-1+PMN-MDSCs in BA patients, along with MAPK signaling pathway upregulation, indicating their involvement in disease mechanisms. Additionally, the frequencies of LOX-1+PMN-MDSC in peripheral blood significantly positively correlate with liver function parameters in BA patients, demonstrating diagnostic performance comparable to traditional serum markers. These findings suggest that LOX-1+PMN-MDSCs contribute to the immunosuppressive environment in BA and could serve as potential diagnostic targets.
Collapse
Affiliation(s)
- Cheng Chen
- Guangdong Provincial Key Laboratory of Research in Structure Birth Defect Disease and Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Hezhen Wang
- Guangdong Provincial Key Laboratory of Research in Structure Birth Defect Disease and Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Lili Xu
- Clinical Laboratory, Guangzhou Red Cross Hospital, Jinan University, Guangzhou 510220, China
| | - Zhipeng Guo
- Guangdong Provincial Key Laboratory of Research in Structure Birth Defect Disease and Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Ming Fu
- Guangdong Provincial Key Laboratory of Research in Structure Birth Defect Disease and Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Huimin Xia
- Guangdong Provincial Key Laboratory of Research in Structure Birth Defect Disease and Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Qiuming He
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center Liuzhou Hospital, Liuzhou 545000, China; Department of Neonatal Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Ruizhong Zhang
- Guangdong Provincial Key Laboratory of Research in Structure Birth Defect Disease and Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China.
| | - Juan He
- Guangdong Provincial Key Laboratory of Research in Structure Birth Defect Disease and Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China.
| |
Collapse
|
2
|
Pateraki P, Latsoudis H, Papadopoulou A, Gontika I, Fragiadaki I, Mavroudi I, Bizymi N, Batsali A, Klontzas ME, Xagorari A, Michalopoulos E, Sotiropoulos D, Yannaki E, Stavropoulos-Giokas C, Papadaki HA. Perspectives for the Use of Umbilical Cord Blood in Transplantation and Beyond: Initiatives for an Advanced and Sustainable Public Banking Program in Greece. J Clin Med 2024; 13:1152. [PMID: 38398465 PMCID: PMC10889829 DOI: 10.3390/jcm13041152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 02/11/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
The umbilical cord blood (UCB) donated in public UCB banks is a source of hematopoietic stem cells (HSC) alternative to bone marrow for allogeneic HSC transplantation (HSCT). However, the high rejection rate of the donated units due to the strict acceptance criteria and the wide application of the haploidentical HSCT have resulted in significant limitation of the use of UCB and difficulties in the economic sustainability of the public UCB banks. There is an ongoing effort within the UCB community to optimize the use of UCB in the field of HSCT and a parallel interest in exploring the use of UCB for applications beyond HSCT i.e., in the fields of cell therapy, regenerative medicine and specialized transfusion medicine. In this report, we describe the mode of operation of the three public UCB banks in Greece as an example of an orchestrated effort to develop a viable UCB banking system by (a) prioritizing the enrichment of the national inventory by high-quality UCB units from populations with rare human leukocyte antigens (HLA), and (b) deploying novel sustainable applications of UCB beyond HSCT, through national and international collaborations. The Greek paradigm of the public UCB network may become an example for countries, particularly with high HLA heterogeneity, with public UCB banks facing sustainability difficulties and adds value to the international efforts aiming to sustainably expand the public UCB banking system.
Collapse
Affiliation(s)
- Patra Pateraki
- Law Directorate of the Health Region of Crete, Ministry of Health, Heraklion, 71500 Heraklion, Greece;
- Public Cord Blood Bank of Crete, Department of Hematology, University Hospital of Heraklion, 71500 Heraklion, Greece; (I.G.); (I.F.); (I.M.); (N.B.); (A.B.)
| | - Helen Latsoudis
- Institute of Computer Sciences, Foundation for Research and Technology–Hellas (FORTH), 70013 Heraklion, Greece;
| | - Anastasia Papadopoulou
- Gene and Cell Therapy Center, Department of Hematology-HCT Unit, George Papanikolaou Hospital, 57010 Thessaloniki, Greece;
| | - Ioanna Gontika
- Public Cord Blood Bank of Crete, Department of Hematology, University Hospital of Heraklion, 71500 Heraklion, Greece; (I.G.); (I.F.); (I.M.); (N.B.); (A.B.)
- Hemopoiesis Research Laboratory, School of Medicine, University of Crete, 71500 Heraklion, Greece;
| | - Irene Fragiadaki
- Public Cord Blood Bank of Crete, Department of Hematology, University Hospital of Heraklion, 71500 Heraklion, Greece; (I.G.); (I.F.); (I.M.); (N.B.); (A.B.)
- Hemopoiesis Research Laboratory, School of Medicine, University of Crete, 71500 Heraklion, Greece;
| | - Irene Mavroudi
- Public Cord Blood Bank of Crete, Department of Hematology, University Hospital of Heraklion, 71500 Heraklion, Greece; (I.G.); (I.F.); (I.M.); (N.B.); (A.B.)
- Hemopoiesis Research Laboratory, School of Medicine, University of Crete, 71500 Heraklion, Greece;
| | - Nikoleta Bizymi
- Public Cord Blood Bank of Crete, Department of Hematology, University Hospital of Heraklion, 71500 Heraklion, Greece; (I.G.); (I.F.); (I.M.); (N.B.); (A.B.)
- Hemopoiesis Research Laboratory, School of Medicine, University of Crete, 71500 Heraklion, Greece;
| | - Aristea Batsali
- Public Cord Blood Bank of Crete, Department of Hematology, University Hospital of Heraklion, 71500 Heraklion, Greece; (I.G.); (I.F.); (I.M.); (N.B.); (A.B.)
- Hemopoiesis Research Laboratory, School of Medicine, University of Crete, 71500 Heraklion, Greece;
| | - Michail E. Klontzas
- Department of Radiology, School of Medicine, University of Crete, 71500 Heraklion, Greece;
- Department of Medical Imaging, University Hospital of Heraklion, 71500 Heraklion, Greece
| | - Angeliki Xagorari
- Public Cord Blood Bank, Department of Hematology, George Papanikolaou Hospital, 57010 Thessaloniki, Greece; (A.X.); (D.S.)
| | - Efstathios Michalopoulos
- Hellenic Cord Blood Bank (HCBB), Biomedical Research Foundation Academy of Athens, 11527 Athens, Greece; (E.M.); (C.S.-G.)
| | - Damianos Sotiropoulos
- Public Cord Blood Bank, Department of Hematology, George Papanikolaou Hospital, 57010 Thessaloniki, Greece; (A.X.); (D.S.)
| | - Evangelia Yannaki
- Hemopoiesis Research Laboratory, School of Medicine, University of Crete, 71500 Heraklion, Greece;
| | - Catherine Stavropoulos-Giokas
- Hellenic Cord Blood Bank (HCBB), Biomedical Research Foundation Academy of Athens, 11527 Athens, Greece; (E.M.); (C.S.-G.)
| | - Helen A. Papadaki
- Public Cord Blood Bank of Crete, Department of Hematology, University Hospital of Heraklion, 71500 Heraklion, Greece; (I.G.); (I.F.); (I.M.); (N.B.); (A.B.)
- Hemopoiesis Research Laboratory, School of Medicine, University of Crete, 71500 Heraklion, Greece;
| |
Collapse
|
3
|
Schwarz J, Rühle J, Stephan K, Dietz S, Geißert J, Schoppmeier U, Frick JS, Hudalla H, Lajqi T, Poets CF, Gille C, Köstlin-Gille N. HIF-1α targeted deletion in myeloid cells decreases MDSC accumulation and alters microbiome in neonatal mice. Eur J Immunol 2023; 53:e2250144. [PMID: 37044112 DOI: 10.1002/eji.202250144] [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: 08/19/2022] [Revised: 04/01/2023] [Accepted: 04/11/2023] [Indexed: 04/14/2023]
Abstract
The newborn's immune system is faced with the challenge of having to learn quickly to fight off infectious agents, but tolerating the colonization of the body surfaces with commensals without reacting with an excessive inflammatory response. Myeloid-derived suppressor cells (MDSC) are innate immune cells with suppressive activity on other immune cells that regulate fetal-maternal tolerance during pregnancy and control intestinal inflammation in neonates. Until now, nothing is known about the role of MDSC in microbiome establishment. One of the transcription factors regulating MDSC homeostasis is the hypoxia-inducible factor 1α (HIF-1α). We investigated the impact of HIF-1α on MDSC accumulation and microbiome establishment during the neonatal period in a mouse model with targeted deletion of HIF-1α in myeloid cells (Hif1a loxP/loxP LysMCre+). We show that in contrast to wildtype mice, where an extensive expansion of MDSC was observed, MDSC expansion in neonatal Hif1a loxP/loxP LysMCre+ mice was dramatically reduced both systemically and locally in the intestine. This was accompanied by an altered microbiome composition and intestinal T-cell homeostasis. Our results point toward a role of MDSC in inflammation regulation in the context of microbiome establishment and thus reveal a new aspect of the biological role of MDSC during the neonatal period.
Collapse
Affiliation(s)
- Julian Schwarz
- Tuebingen University Children's Hospital, Department of Neonatology, Tuebingen, Germany
| | - Jessica Rühle
- Tuebingen University Children's Hospital, Department of Neonatology, Tuebingen, Germany
| | - Kevin Stephan
- Tuebingen University Children's Hospital, Department of Neonatology, Tuebingen, Germany
| | - Stefanie Dietz
- Tuebingen University Children's Hospital, Department of Neonatology, Tuebingen, Germany
- Heidelberg University Children's Hospital, Department of Neonatology, Heidelberg, Germany
| | - Janina Geißert
- Institute for Medical Microbiology and Hygiene, University Hospital Tuebingen, Tuebingen, Germany
- NGS-Competence Center Tuebingen, Institute for Medical Microbiology and Hygiene, University Hospital Tuebingen, Tuebingen, Germany
| | - Ulrich Schoppmeier
- Institute for Medical Microbiology and Hygiene, University Hospital Tuebingen, Tuebingen, Germany
| | - Julia S Frick
- Institute for Medical Microbiology and Hygiene, University Hospital Tuebingen, Tuebingen, Germany
- MVZ Laboratory Ludwigsburg GbR, Germany
| | - Hannes Hudalla
- Heidelberg University Children's Hospital, Department of Neonatology, Heidelberg, Germany
| | - Trim Lajqi
- Heidelberg University Children's Hospital, Department of Neonatology, Heidelberg, Germany
| | - Christian F Poets
- Tuebingen University Children's Hospital, Department of Neonatology, Tuebingen, Germany
| | - Christian Gille
- Heidelberg University Children's Hospital, Department of Neonatology, Heidelberg, Germany
| | - Natascha Köstlin-Gille
- Tuebingen University Children's Hospital, Department of Neonatology, Tuebingen, Germany
- Heidelberg University Children's Hospital, Department of Neonatology, Heidelberg, Germany
| |
Collapse
|
4
|
Ostrand-Rosenberg S, Lamb TJ, Pawelec G. Here, There, and Everywhere: Myeloid-Derived Suppressor Cells in Immunology. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1183-1197. [PMID: 37068300 PMCID: PMC10111205 DOI: 10.4049/jimmunol.2200914] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/06/2023] [Indexed: 04/19/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) were initially identified in humans and mice with cancer where they profoundly suppress T cell- and NK cell-mediated antitumor immunity. Inflammation is a central feature of many pathologies and normal physiological conditions and is the dominant driving force for the accumulation and function of MDSCs. Therefore, MDSCs are present in conditions where inflammation is present. Although MDSCs are detrimental in cancer and conditions where cellular immunity is desirable, they are beneficial in settings where cellular immunity is hyperactive. Because MDSCs can be generated ex vivo, they are being exploited as therapeutic agents to reduce damaging cellular immunity. In this review, we discuss the detrimental and beneficial roles of MDSCs in disease settings such as bacterial, viral, and parasitic infections, sepsis, obesity, trauma, stress, autoimmunity, transplantation and graft-versus-host disease, and normal physiological settings, including pregnancy and neonates as well as aging. The impact of MDSCs on vaccination is also discussed.
Collapse
Affiliation(s)
- Suzanne Ostrand-Rosenberg
- Division of Microbiology and Immunology, Department of Pathology, University of Utah 84112, Salt Lake City, UT
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Tracey J. Lamb
- Division of Microbiology and Immunology, Department of Pathology, University of Utah 84112, Salt Lake City, UT
| | - Graham Pawelec
- Department of Immunology, Interfaculty Institute for Cell Biology, University of Tübingen, Tübingen, Germany, and Health Sciences North Research Institute, Sudbury, ON, Canada
| |
Collapse
|
5
|
Rühle J, Ginzel M, Dietz S, Schwarz J, Lajqi T, Beer-Hammer S, Poets CF, Gille C, Köstlin-Gille N. Depletion of Ly6G-Expressing Neutrophilic Cells Leads to Altered Peripheral T-Cell Homeostasis and Thymic Development in Neonatal Mice. Int J Mol Sci 2023; 24:7763. [PMID: 37175470 PMCID: PMC10178674 DOI: 10.3390/ijms24097763] [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: 03/23/2023] [Revised: 04/14/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Newborns and especially preterm infants are much more susceptible to infections than adults. Due to immature adaptive immunity, especially innate immune cells play an important role in a newborn's infection defense. Neonatal neutrophils exhibit profound differences in their functionality compared to neutrophils of adults. In particular, neonates possess a relevant population of suppressive neutrophils, which not only inhibit but also specifically modulate the function of T-cells. In this study, we investigated whether neonatal neutrophils are already involved in T-cell development in the thymus. For this purpose, we used a newly developed model of antibody-mediated immune cell depletion in which we administered a depleting antibody to pregnant and then lactating dams. Using this method, we were able to sufficiently deplete Ly6G-positive neutrophils in offspring. We demonstrated that the depletion of neutrophils in newborn mice resulted in altered peripheral T-cell homeostasis with a decreased CD4+/CD8+ T-cell ratio and decreased expression of CD62L. Neutrophil depletion even affected T-cell development in the thymus, with increased double positive thymocytes and a decreased CD4+/CD8+ single positive thymocyte ratio. Altogether, we demonstrated a previously unknown mechanism mediating neutrophils' immunomodulatory effects in newborns.
Collapse
Affiliation(s)
- Jessica Rühle
- Department of Neonatology, University of Tübingen, D-72076 Tübingen, Germany; (J.R.); (S.D.); (J.S.); (C.F.P.)
| | - Marco Ginzel
- Department of Pediatric and Adolescent Surgery, Paracelsus Medical University Hospital, A-5020 Salzburg, Austria;
| | - Stefanie Dietz
- Department of Neonatology, University of Tübingen, D-72076 Tübingen, Germany; (J.R.); (S.D.); (J.S.); (C.F.P.)
- Department of Neonatology, Heidelberg University Children’s Hospital, D-69120 Heidelberg, Germany; (T.L.); (C.G.)
| | - Julian Schwarz
- Department of Neonatology, University of Tübingen, D-72076 Tübingen, Germany; (J.R.); (S.D.); (J.S.); (C.F.P.)
| | - Trim Lajqi
- Department of Neonatology, Heidelberg University Children’s Hospital, D-69120 Heidelberg, Germany; (T.L.); (C.G.)
| | - Sandra Beer-Hammer
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute of Experimental and Clinical Pharmacology and Pharmacogenomic and ICePhA, University of Tübingen, D-72074 Tübingen, Germany;
| | - Christian F. Poets
- Department of Neonatology, University of Tübingen, D-72076 Tübingen, Germany; (J.R.); (S.D.); (J.S.); (C.F.P.)
| | - Christian Gille
- Department of Neonatology, Heidelberg University Children’s Hospital, D-69120 Heidelberg, Germany; (T.L.); (C.G.)
| | - Natascha Köstlin-Gille
- Department of Neonatology, University of Tübingen, D-72076 Tübingen, Germany; (J.R.); (S.D.); (J.S.); (C.F.P.)
- Department of Neonatology, Heidelberg University Children’s Hospital, D-69120 Heidelberg, Germany; (T.L.); (C.G.)
| |
Collapse
|
6
|
Schlegel C, Liu K, Spring B, Dietz S, Poets CF, Hudalla H, Lajqi T, Köstlin-Gille N, Gille C. Decreased expression of hypoxia-inducible factor 1α (HIF-1α) in cord blood monocytes under anoxia. Pediatr Res 2023; 93:870-877. [PMID: 35906309 PMCID: PMC10033401 DOI: 10.1038/s41390-022-02193-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/24/2022] [Accepted: 06/28/2022] [Indexed: 11/08/2022]
Abstract
BACKGROUND Infections are a major cause for morbidity and mortality in neonates; however, the underling mechanisms for increased infection susceptibility are incompletely understood. Hypoxia, which is present in inflamed tissues, has been identified as an important activation signal for innate immune cells in adults and is mainly mediated by hypoxia-inducible factor 1α (HIF-1α). Fetal tissue pO2 physiologically is low but rises immediately after birth. METHODS In this study, the effect of low oxygen partial pressure (pO2) on HIF-1α expression and its targets phagocytosis, reactive oxygen species (ROS) production and vascular endothelial growth factor (VEGF) secretion was compared in vitro between immune cells from adult peripheral blood and cord blood using anoxia, HIF-1α stabilizer desferroxamin (DFO) and E. coli as stimuli. RESULTS We show that anoxia-induced HIF-1α protein accumulation, phagocytosis, ROS-production and VEGF-expression were greatly diminished in cord blood compared to adult cells. E. coli led to HIF-1α gene expression in adult and cord blood immune cells; however, cord blood cells failed to accumulate HIF-1α protein and VEGF upon E. coli stimulation. CONCLUSIONS Taken together, our results show a diminished activation of cord blood immune cells by low pO2, which might contribute to impaired reactivity in the context of infection. IMPACT Neonatal immune cells do not accumulate HIF-1α under low oxygen partial pressure leading to decreased phagocytosis and decreased ROS production. We demonstrate a previously unknown mechanism of reduced activation of neonatal immune cells in the context of an inflammatory response. This could contribute to the increased susceptibility of newborns and preterm infants to infection.
Collapse
Affiliation(s)
- Christiane Schlegel
- Department of Neonatology, Tübingen University Children's Hospital, Tübingen, Germany
| | - Kai Liu
- Department of Neonatology, Tübingen University Children's Hospital, Tübingen, Germany
| | - Bärbel Spring
- Department of Neonatology, Tübingen University Children's Hospital, Tübingen, Germany
| | - Stefanie Dietz
- Department of Neonatology, Tübingen University Children's Hospital, Tübingen, Germany
- Department of Neonatology, Heidelberg University Children's Hospital, Heidelberg, Germany
| | - Christian F Poets
- Department of Neonatology, Tübingen University Children's Hospital, Tübingen, Germany
| | - Hannes Hudalla
- Department of Neonatology, Heidelberg University Children's Hospital, Heidelberg, Germany
| | - Trim Lajqi
- Department of Neonatology, Heidelberg University Children's Hospital, Heidelberg, Germany
| | - Natascha Köstlin-Gille
- Department of Neonatology, Tübingen University Children's Hospital, Tübingen, Germany.
- Department of Neonatology, Heidelberg University Children's Hospital, Heidelberg, Germany.
| | - Christian Gille
- Department of Neonatology, Heidelberg University Children's Hospital, Heidelberg, Germany
| |
Collapse
|
7
|
Polcz VE, Rincon JC, Hawkins RB, Barrios EL, Efron PA, Moldawer LL, Larson SD. TRAINED IMMUNITY: A POTENTIAL APPROACH FOR IMPROVING HOST IMMUNITY IN NEONATAL SEPSIS. Shock 2023; 59:125-134. [PMID: 36383390 PMCID: PMC9957873 DOI: 10.1097/shk.0000000000002054] [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] [Indexed: 11/17/2022]
Abstract
ABSTRACT Sepsis, a dysregulated host immune response to infection, is one of the leading causes of neonatal mortality worldwide. Improved understanding of the perinatal immune system is critical to improve therapies to both term and preterm neonates at increased risk of sepsis. Our narrative outlines the known and unknown aspects of the human immune system through both the immune tolerant in utero period and the rapidly changing antigen-rich period after birth. We will highlight the key differences in innate and adaptive immunity noted through these developmental stages and how the unique immune phenotype in early life contributes to the elevated risk of overwhelming infection and dysregulated immune responses to infection upon exposure to external antigens shortly after birth. Given an initial dependence on neonatal innate immune host responses, we will discuss the concept of innate immune memory, or "trained immunity," and describe several potential immune modulators, which show promise in altering the dysregulated immune response in newborns and improving resilience to sepsis.
Collapse
Affiliation(s)
- Valerie E Polcz
- Department of Surgery and Sepsis and Critical Illness Research Center, University of Florida College of Medicine, Gainesville, Florida
| | | | | | | | | | | | | |
Collapse
|
8
|
Pang B, Hu C, Li H, Nie X, Wang K, Zhou C, Yi H. Myeloidderived suppressor cells: Escorts at the maternal-fetal interface. Front Immunol 2023; 14:1080391. [PMID: 36817414 PMCID: PMC9932974 DOI: 10.3389/fimmu.2023.1080391] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 01/13/2023] [Indexed: 02/05/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a novel heterogenous group of immunosuppressive cells derived from myeloid progenitors. Their role is well known in tumors and autoimmune diseases. In recent years, the role and function of MDSCs during reproduction have attracted increasing attention. Improving the understanding of their strong association with recurrent implantation failure, pathological pregnancy, and neonatal health has become a focus area in research. In this review, we focus on the interaction between MDSCs and other cell types (immune and non-immune cells) from embryo implantation to postpartum. Furthermore, we discuss the molecular mechanisms that could facilitate the therapeutic targeting of MDSCs. Therefore, this review intends to encourage further research in the field of maternal-fetal interface immunity in order to identify probable pathways driving the accumulation of MDSCs and to effectively target their ability to promote embryo implantation, reduce pathological pregnancy, and increase neonatal health.
Collapse
Affiliation(s)
- Bo Pang
- Central Laboratory, First Hospital of Jilin University, Changchun, Jilin, China.,Cardiology Department, First Hospital of Jilin University, Changchun, Jilin, China
| | - Cong Hu
- Central Laboratory, First Hospital of Jilin University, Changchun, Jilin, China.,Reproductive Medicine Center, Prenatal Diagnosis Center, First Hospital of Jilin University, Changchun, Jilin, China
| | - Huimin Li
- Central Laboratory, First Hospital of Jilin University, Changchun, Jilin, China.,Department of Clinical Laboratory, The Second Hospital of Jilin University, Changchun, China
| | - Xinyu Nie
- Central Laboratory, First Hospital of Jilin University, Changchun, Jilin, China.,Reproductive Medicine Center, Prenatal Diagnosis Center, First Hospital of Jilin University, Changchun, Jilin, China
| | - Keqi Wang
- Central Laboratory, First Hospital of Jilin University, Changchun, Jilin, China.,Cardiology Department, First Hospital of Jilin University, Changchun, Jilin, China
| | - Chen Zhou
- General Department, First Hospital of Jilin University, Changchun, Jilin, China
| | - Huanfa Yi
- Central Laboratory, First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
9
|
Rückle X, Rühle J, Judd L, Hebel J, Dietz S, Poets CF, Gille C, Köstlin-Gille N. Different probiotic strains alter human cord blood monocyte responses. Pediatr Res 2022:10.1038/s41390-022-02400-5. [PMID: 36476746 DOI: 10.1038/s41390-022-02400-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 10/24/2022] [Accepted: 11/09/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Probiotics have a protective effect on various diseases. In neonatology, they are predominantly used to prevent necrotising enterocolitis (NEC), a severe inflammatory disease of the neonatal intestine. The mechanisms by which probiotics act are diverse; little is known about their direct effect on neonatal immune cells. METHODS In this study, we investigated the effect of probiotics on the functions of neonatal monocytes in an in vitro model using three different strains (Lactobacillus rhamnosus (LR), Lactobacillus acidophilus (LA) and Bifidobacterium bifidum (BB)) and mononuclear cells isolated from cord blood. RESULTS We show that stimulation with LR induces proinflammatory effects in neonatal monocytes, such as increased expression of surface molecules involved in monocyte activation, increased production of pro-inflammatory and regulatory cytokines and increased production of reactive oxygen species (ROS). Similar effects were observed when monocytes were stimulated simultaneously with LPS. Stimulation with LA and BB alone or in combination also induced cytokine production in monocytes, with BB showing the least effects. CONCLUSIONS Our results suggest that probiotics increase the defence functions of neonatal monocytes and thus possibly favourably influence the newborn's ability to fight infections. IMPACT Probiotics induce a proinflammatory response in neonatal monocytes in vitro. This is a previously unknown mechanism of how probiotics modulate the immune response of newborns. Probiotic application to neonates may increase their ability to fight off infections.
Collapse
Affiliation(s)
- Xenia Rückle
- Department of Neonatology, Tübingen University Children's Hospital, Tübingen, Germany
| | - Jessica Rühle
- Department of Neonatology, Tübingen University Children's Hospital, Tübingen, Germany
| | - Leonie Judd
- Department of Neonatology, Tübingen University Children's Hospital, Tübingen, Germany
| | - Janine Hebel
- Department of Neonatology, Tübingen University Children's Hospital, Tübingen, Germany
| | - Stefanie Dietz
- Department of Neonatology, Tübingen University Children's Hospital, Tübingen, Germany.,Department of Neonatology, Heidelberg University Children's Hospital, Heidelberg, Germany
| | - Christian F Poets
- Department of Neonatology, Tübingen University Children's Hospital, Tübingen, Germany
| | - Christian Gille
- Department of Neonatology, Heidelberg University Children's Hospital, Heidelberg, Germany
| | - Natascha Köstlin-Gille
- Department of Neonatology, Tübingen University Children's Hospital, Tübingen, Germany. .,Department of Neonatology, Heidelberg University Children's Hospital, Heidelberg, Germany.
| |
Collapse
|
10
|
Perego M, Fu S, Cao Y, Kossenkov A, Yao M, Bonner E, Alicea-Torres K, Liu W, Jiang Z, Chen Z, Fuchs SY, Zhou J, Gabrilovich DI. Mechanisms regulating transitory suppressive activity of neutrophils in newborns: PMNs-MDSCs in newborns. J Leukoc Biol 2022; 112:955-968. [PMID: 35726818 PMCID: PMC9794389 DOI: 10.1002/jlb.4hi0921-514rr] [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: 09/27/2021] [Revised: 05/27/2022] [Accepted: 05/28/2022] [Indexed: 12/30/2022] Open
Abstract
Transitory appearance of immune suppressive polymorphonuclear neutrophils (PMNs) defined as myeloid-derived suppressor cells (PMNs-MDSCs) in newborns is important for their protection from inflammation associated with newly established gut microbiota. Here, we report that inhibition of the type I IFN (IFN1) pathway played a major role in regulation of PMNs-MDSCs-suppressive activity during first weeks of life. Expression of the IFN1 receptor IFNAR1 was markedly lower in PMNs-MDSCs. However, in newborn mice, down-regulation of IFNAR1 was not sufficient to render PMNs immune suppressive. That also required the presence of a positive signal from lactoferrin via its receptor low-density lipoprotein receptor-related protein 2. The latter effect was mediated via NF-κB activation, which was tempered by IFN1 in a manner that involved suppressor of cytokine signaling 3. Thus, we discovered a mechanism of tight regulation of immune suppressive PMNs-MDSCs in newborns, which may be used in the development of therapies of neonatal pathologies.
Collapse
Affiliation(s)
| | - Shuyu Fu
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Yingjiao Cao
- Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Sciences, Tianjin Medical University, Tianjin, China
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | | | - Meng Yao
- Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Sciences, Tianjin Medical University, Tianjin, China
| | - Erin Bonner
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Kevin Alicea-Torres
- The Wistar Institute, Philadelphia, Pennsylvania, USA
- Biology Department, University of Puerto Rico-Humacao, Humacao, Puerto Rico, USA
| | - Wangkai Liu
- Department of Pediatrics, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhilong Jiang
- Department of Respiratory and Critical Care Medicine, Zhongshan Hospital; Shanghai Institute of Respiratory Disease, Fudan University, Shanghai, China
| | - Zhihong Chen
- Department of Respiratory and Critical Care Medicine, Zhongshan Hospital; Shanghai Institute of Respiratory Disease, Fudan University, Shanghai, China
| | - Serge Y Fuchs
- Department of Biomedical Sciences, School of Veterinary Medicine University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jie Zhou
- Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Sciences, Tianjin Medical University, Tianjin, China
| | | |
Collapse
|
11
|
Molnar K, Riedel H, Schwarz J, Dietz S, Spring B, Haag L, Poets CF, Gille C, Köstlin-Gille N. Group B streptococci infection model shows decreased regulatory capacity of cord blood cells. Pediatr Res 2022; 92:1407-1416. [PMID: 35165359 PMCID: PMC9700511 DOI: 10.1038/s41390-021-01880-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/17/2021] [Accepted: 07/27/2021] [Indexed: 11/09/2022]
Abstract
BACKGROUND Sepsis is one of the leading causes of morbidity and mortality in the neonatal period. Compared to adults, neonates are more susceptible to infections, especially to systemic infections with Group B Streptococcus (GBS). Furthermore, neonates show defects in terminating inflammation. The immunological causes for the increased susceptibility to infection and the prolonged inflammatory response are still incompletely understood. METHODS In the present study, we aimed to investigate the reaction of cord blood mononuclear cells (MNC) to stimulation with GBS in comparison to that of MNC from adult blood with focus on the proliferative response in an in vitro infection model with heat-inactivated GBS. RESULTS We demonstrate that after stimulation with GBS the proliferation of T cells from adult blood strongly decreased, while the proliferation of cord blood T cells remained unchanged. This effect could be traced back to a transformation of adult monocytes, but not cord blood monocytes, to a suppressive phenotype with increased expression of the co-inhibitory molecule programmed death ligand 1 (PD-L1). CONCLUSIONS These results point towards an increased inflammatory capacity of neonatal MNC after stimulation with GBS. Targeting the prolonged inflammatory response of neonatal immune cells may be a strategy to prevent complications of neonatal infections. IMPACT Neonatal sepsis often leads to post-inflammatory complications. Causes for sustained inflammation in neonates are incompletely understood. We show that cord blood T cells exhibited increased proliferative capacity after stimulation with group B streptococci (GBS) in comparison to adult T cells. Adult monocytes but not cord blood monocytes acquired suppressive activity and expressed increased levels of PD-L1 after GBS stimulation. Increased proliferative capacity of neonatal T cells and decreased suppressive activity of neonatal monocytes during GBS infection may contribute to prolonged inflammation and development of post-inflammatory diseases in newborns.
Collapse
Affiliation(s)
- Kriszta Molnar
- Department of Neonatology, Tübingen University Children's Hospital, Calwerstrasse 7, 72076, Tübingen, Germany
| | - Hannah Riedel
- Department of Neonatology, Tübingen University Children's Hospital, Calwerstrasse 7, 72076, Tübingen, Germany
| | - Julian Schwarz
- Department of Neonatology, Tübingen University Children's Hospital, Calwerstrasse 7, 72076, Tübingen, Germany
| | - Stefanie Dietz
- Department of Neonatology, Tübingen University Children's Hospital, Calwerstrasse 7, 72076, Tübingen, Germany
| | - Bärbel Spring
- Department of Neonatology, Tübingen University Children's Hospital, Calwerstrasse 7, 72076, Tübingen, Germany
| | - Laura Haag
- Department of Neonatology, Tübingen University Children's Hospital, Calwerstrasse 7, 72076, Tübingen, Germany
| | - Christian F Poets
- Department of Neonatology, Tübingen University Children's Hospital, Calwerstrasse 7, 72076, Tübingen, Germany
| | - Christian Gille
- Department of Neonatology, Heidelberg University Children's Hospital, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany.
| | - Natascha Köstlin-Gille
- Department of Neonatology, Tübingen University Children's Hospital, Calwerstrasse 7, 72076, Tübingen, Germany
| |
Collapse
|
12
|
Feng S, Cui Y, Zhou Y, Shao L, Miao H, Dou J, Liu T, Wang C, Zhang Y. Continuous renal replacement therapy attenuates polymorphonuclear myeloid-derived suppressor cell expansion in pediatric severe sepsis. Front Immunol 2022; 13:990522. [PMID: 36263056 PMCID: PMC9575946 DOI: 10.3389/fimmu.2022.990522] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 09/06/2022] [Indexed: 11/13/2022] Open
Abstract
Background Myeloid-derived suppressor cells (MDSCs) expansion is an important mechanism underlying immunosuppression during sepsis. Though continuous renal replacement therapy (CRRT) may attenuate hyperinflammatory response in sepsis, its role in regulating MDSCs is unknown. The aim of this study was to assess the potential role of CRRT involved in sepsis-induced MDSCs expansion in pediatric sepsis. Method The proportion of polymorphonuclear MDSCs (PMN-MDSCs) was detected before CRRT (pre-CRRT), at 24 hours after CRRT (CRRT 1st day) and on the 7th day after CRRT (CRRT 7th day). The correlation analyses were performed to elucidate the relationship of MDSCs with clinical indexes in sepsis. Results Totally 22 pediatric patients with sepsis were enrolled [median age 44 (IQR15, 83) months]. PMN-MDSCs were expanded in pediatric sepsis compared with healthy controls (4.30% vs. 0.37%, P=0.04). The proportion of PMN-MDSCs showed a decreased tendency on the CRRT 7th day compared with that on the CRRT 1st day in survivors (2.29% vs.5.32%, P = 0.088). There was no significant difference in the proportion of PMN-MDSCs between survivors and non-survivors before CRRT (4.51% vs. 3.33%, P=0.745). The levels of interleukin 6 (IL-6) was decreased on the CRRT 7th day compared with CRRT 1st day in survivors. In the subgroups of patients with significantly decreased IL-6 levels after CRRT, the proportion of PMN-MDSCs on the CRRT 7th day were also significantly decreased compared with that on the CRRT 1st day (2.21% vs. 6.67%, P = 0.033). Conclusion The proportion of PMN-MDSCs was down-regulated on the CRRT 7th day in survivors with sepsis. The reduced PMN-MDSCs expansion may relate to decreased IL-6 level.
Collapse
Affiliation(s)
- Shuyun Feng
- Department of Critical Care Medicine, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yun Cui
- Department of Critical Care Medicine, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Pediatric Critical Care, Shanghai Jiao Tong University, Shanghai, China
| | - Yiping Zhou
- Department of Critical Care Medicine, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Pediatric Critical Care, Shanghai Jiao Tong University, Shanghai, China
| | - Lujing Shao
- Department of Critical Care Medicine, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huijie Miao
- Department of Critical Care Medicine, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiaying Dou
- Department of Critical Care Medicine, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Pediatric Critical Care, Shanghai Jiao Tong University, Shanghai, China
| | - Tiantian Liu
- Department of Critical Care Medicine, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chunxia Wang
- Department of Critical Care Medicine, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Pediatric Critical Care, Shanghai Jiao Tong University, Shanghai, China
- Clinical Research Unit, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Yucai Zhang, ; Chunxia Wang,
| | - Yucai Zhang
- Department of Critical Care Medicine, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Pediatric Critical Care, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Yucai Zhang, ; Chunxia Wang,
| |
Collapse
|
13
|
Targeting tumour-reprogrammed myeloid cells: the new battleground in cancer immunotherapy. Semin Immunopathol 2022; 45:163-186. [PMID: 36161514 PMCID: PMC9513014 DOI: 10.1007/s00281-022-00965-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/13/2022] [Indexed: 11/08/2022]
Abstract
Tumour microenvironment is a complex ecosystem in which myeloid cells are the most abundant immune elements. This cell compartment is composed by different cell types, including neutrophils, macrophages, dendritic cells, and monocytes but also unexpected cell populations with immunosuppressive and pro-tumour roles. Indeed, the release of tumour-derived factors influences physiological haematopoiesis producing unconventional cells with immunosuppressive and tolerogenic functions such as myeloid-derived suppressor cells. These pro-tumour myeloid cell populations not only support immune escape directly but also assist tumour invasion trough non-immunological activities. It is therefore not surprising that these cell subsets considerably impact in tumour progression and cancer therapy resistance, including immunotherapy, and are being investigated as potential targets for developing a new era of cancer therapy. In this review, we discuss emerging strategies able to modulate the functional activity of these tumour-supporting myeloid cells subverting their accumulation, recruitment, survival, and functions. These innovative approaches will help develop innovative, or improve existing, cancer treatments.
Collapse
|
14
|
Munansangu BSM, Kenyon C, Walzl G, Loxton AG, Kotze LA, du Plessis N. Immunometabolism of Myeloid-Derived Suppressor Cells: Implications for Mycobacterium tuberculosis Infection and Insights from Tumor Biology. Int J Mol Sci 2022; 23:3512. [PMID: 35408873 PMCID: PMC8998693 DOI: 10.3390/ijms23073512] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/04/2022] [Accepted: 03/08/2022] [Indexed: 02/04/2023] Open
Abstract
The field of immunometabolism seeks to decipher the complex interplay between the immune system and the associated metabolic pathways. The role of small molecules that can target specific metabolic pathways and subsequently alter the immune landscape provides a desirable platform for new therapeutic interventions. Immunotherapeutic targeting of suppressive cell populations, such as myeloid-derived suppressor cells (MDSC), by small molecules has shown promise in pathologies such as cancer and support testing of similar host-directed therapeutic approaches in MDSC-inducing conditions such as tuberculosis (TB). MDSC exhibit a remarkable ability to suppress T-cell responses in those with TB disease. In tumors, MDSC exhibit considerable plasticity and can undergo metabolic reprogramming from glycolysis to fatty acid oxidation (FAO) and oxidative phosphorylation (OXPHOS) to facilitate their immunosuppressive functions. In this review we look at the role of MDSC during M. tb infection and how their metabolic reprogramming aids in the exacerbation of active disease and highlight the possible MDSC-targeted metabolic pathways utilized during M. tb infection, suggesting ways to manipulate these cells in search of novel insights for anti-TB therapies.
Collapse
Affiliation(s)
| | | | | | | | | | - Nelita du Plessis
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 7505, South Africa; (B.S.M.M.); (C.K.); (G.W.); (A.G.L.); (L.A.K.)
| |
Collapse
|
15
|
Dietz S, Schwarz J, Velic A, González-Menéndez I, Quintanilla-Martinez L, Casadei N, Marmé A, Poets CF, Gille C, Köstlin-Gille N. Human Leucocyte Antigen G and Murine Qa-2 Are Critical for Myeloid Derived Suppressor Cell Expansion and Activation and for Successful Pregnancy Outcome. Front Immunol 2022; 12:787468. [PMID: 35111157 PMCID: PMC8801456 DOI: 10.3389/fimmu.2021.787468] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/27/2021] [Indexed: 12/03/2022] Open
Abstract
During pregnancy, maternal immune system has to balance tightly between protection against pathogens and tolerance towards a semi-allogeneic organism. Dysfunction of this immune adaptation can lead to severe complications such as pregnancy loss, preeclampsia or fetal growth restriction. In the present study we analyzed the impact of the murine MHC class Ib molecule Qa-2 on pregnancy outcome in vivo. We demonstrate that lack of Qa-2 led to intrauterine growth restriction and increased abortion rates especially in late pregnancy accompanied by a disturbed trophoblast invasion and altered spiral artery remodeling as well as protein aggregation in trophoblast cells indicating a preeclampsia-like phenotype. Furthermore, lack of Qa-2 caused imbalanced immunological adaptation to pregnancy with altered immune cell and especially T-cell homeostasis, reduced Treg numbers and decreased accumulation and functional activation of myeloid-derived suppressor cells. Lastly, we show that application of sHLA-G reduced abortion rates in Qa-2 deficient mice by inducing MDSC. Our results highlight the importance of an interaction between HLA-G and MDSC for pregnancy success and the therapeutic potential of HLA-G for treatment of immunological pregnancy complications.
Collapse
Affiliation(s)
- Stefanie Dietz
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany
| | - Julian Schwarz
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany
| | - Ana Velic
- Interfaculty Institute for Cell Biology, Proteome Center Tuebingen (PCT), University of Tuebingen, Tübingen, Germany
| | | | | | - Nicolas Casadei
- Next Generation Sequencing (NGS) Competence Center Tuebingen (NCCT), Tuebingen, Germany; Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany
| | - Alexander Marmé
- Gynecology and Obstetrics Practice, Am Lustnauer Tor, Tuebingen, Germany
| | - Christian F Poets
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany
| | - Christian Gille
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany.,Department of Neonatology, Heidelberg University Children's Hospital, Heidelberg, Germany
| | - Natascha Köstlin-Gille
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany.,Department of Neonatology, Heidelberg University Children's Hospital, Heidelberg, Germany
| |
Collapse
|
16
|
Morales-Mantilla DE, Kain B, Le D, Flores AR, Paust S, King KY. Hematopoietic stem and progenitor cells improve survival from sepsis by boosting immunomodulatory cells. eLife 2022; 11:74561. [PMID: 35166205 PMCID: PMC8846591 DOI: 10.7554/elife.74561] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 01/31/2022] [Indexed: 12/12/2022] Open
Abstract
New therapeutic strategies to reduce sepsis-related mortality are urgently needed, as sepsis accounts for one in five deaths worldwide. Since hematopoietic stem and progenitor cells (HSPCs) are responsible for producing blood and immune cells, including in response to immunological stress, we explored their potential for treating sepsis. In a mouse model of Group A Streptococcus (GAS)-induced sepsis, severe immunological stress was associated with significant depletion of bone marrow HSPCs and mortality within approximately 5–7 days. We hypothesized that the inflammatory environment of GAS infection drives rapid HSPC differentiation and depletion that can be rescued by infusion of donor HSPCs. Indeed, infusion of 10,000 naïve HSPCs into GAS-infected mice resulted in rapid myelopoiesis and a 50–60% increase in overall survival. Surprisingly, mice receiving donor HSPCs displayed a similar pathogen load compared to untreated mice. Flow cytometric analysis revealed a significantly increased number of myeloid-derived suppressor cells in HSPC-infused mice, which correlated with reduced inflammatory cytokine levels and restored HSPC levels. These findings suggest that HSPCs play an essential immunomodulatory role that may translate into new therapeutic strategies for sepsis.
Collapse
Affiliation(s)
- Daniel E Morales-Mantilla
- Graduate Program in Immunology, Baylor College of Medicine, Houston, United States.,Department of Pediatrics, Division of Infectious Diseases, Baylor College of Medicine, Houston, United States
| | - Bailee Kain
- Department of Pediatrics, Division of Infectious Diseases, Baylor College of Medicine, Houston, United States.,Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, United States
| | - Duy Le
- Graduate Program in Immunology, Baylor College of Medicine, Houston, United States.,Department of Pediatrics, Division of Infectious Diseases, Baylor College of Medicine, Houston, United States
| | - Anthony R Flores
- Division of Infectious Diseases, Department of Pediatrics, UTHSC/McGovern Medical School, Houston, United States
| | - Silke Paust
- The Scripps Research Institute, Department of Immunology and Microbiology, La Jolla, United States
| | - Katherine Y King
- Graduate Program in Immunology, Baylor College of Medicine, Houston, United States.,Department of Pediatrics, Division of Infectious Diseases, Baylor College of Medicine, Houston, United States.,Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, United States
| |
Collapse
|
17
|
Bizymi N, Georgopoulou A, Mastrogamvraki N, Matheakakis A, Gontika I, Fragiadaki I, Mavroudi I, Papadaki HA. Myeloid-Derived Suppressor Cells (MDSC) in the Umbilical Cord Blood: Biological Significance and Possible Therapeutic Applications. J Clin Med 2022; 11:jcm11030727. [PMID: 35160177 PMCID: PMC8836851 DOI: 10.3390/jcm11030727] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/20/2022] [Accepted: 01/25/2022] [Indexed: 02/04/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) represent a heterogeneous population of myeloid cells that suppress immune responses in cancer, infection, and trauma. They mainly act by inhibiting T-cells, natural-killer cells, and dendritic cells, and also by inducing T-regulatory cells, and modulating macrophages. Although they are mostly associated with adverse prognosis of the underlying disease entity, they may display positive effects in specific situations, such as in allogeneic hematopoietic stem cell transplantation (HSCT), where they suppress graft-versus-host disease (GVHD). They also contribute to the feto-maternal tolerance, and in the fetus growth process, whereas several pregnancy complications have been associated with their defects. Human umbilical cord blood (UCB) is a source rich in MDSCs and their myeloid progenitor cells. Recently, a number of studies have investigated the generation, isolation, and expansion of UCB-MDSCs for potential clinical application associated with their immunosuppressive properties, such as GVHD, and autoimmune and inflammatory diseases. Given that a significant proportion of UCB units in cord blood banks are not suitable for clinical use in HSCT, they might be used as a significant source of MDSCs for research and clinical purposes. The current review summarizes the roles of MDSCs in the UCB, as well as their promising applications.
Collapse
Affiliation(s)
- Nikoleta Bizymi
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece; (N.B.); (A.M.); (I.M.)
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
| | - Anthie Georgopoulou
- Public Cord Blood Bank of Crete, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece; (A.G.); (N.M.); (I.G.); (I.F.)
| | - Natalia Mastrogamvraki
- Public Cord Blood Bank of Crete, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece; (A.G.); (N.M.); (I.G.); (I.F.)
| | - Angelos Matheakakis
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece; (N.B.); (A.M.); (I.M.)
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
| | - Ioanna Gontika
- Public Cord Blood Bank of Crete, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece; (A.G.); (N.M.); (I.G.); (I.F.)
| | - Irene Fragiadaki
- Public Cord Blood Bank of Crete, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece; (A.G.); (N.M.); (I.G.); (I.F.)
| | - Irene Mavroudi
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece; (N.B.); (A.M.); (I.M.)
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
- Public Cord Blood Bank of Crete, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece; (A.G.); (N.M.); (I.G.); (I.F.)
| | - Helen A. Papadaki
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece; (N.B.); (A.M.); (I.M.)
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
- Public Cord Blood Bank of Crete, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece; (A.G.); (N.M.); (I.G.); (I.F.)
- Correspondence: ; Tel.: +30-2810394637
| |
Collapse
|
18
|
Bert S, Ward EJ, Nadkarni S. Neutrophils in pregnancy: New insights into innate and adaptive immune regulation. Immunology 2021; 164:665-676. [PMID: 34287859 PMCID: PMC8561097 DOI: 10.1111/imm.13392] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/06/2021] [Accepted: 07/08/2021] [Indexed: 12/16/2022] Open
Abstract
The immunology of pregnancy has been the focus of many studies to better understand how the mother is able to tolerate the presence of a semi-allogeneic fetus. Far from the initial view of pregnancy as a state of immunosuppression, successful fetal development from implantation to birth is now known to be under the control of an intricate balance of immune cells. The balance between pro-inflammatory functions used to promote embryo implantation and placental development and immunosuppressive activity to maintain maternal tolerance of the fetus is an immunological phenotype unique to pregnancy, which is dependent on the time of gestation. Neutrophils are one of a host of innate immune cells detected at the maternal-fetal interface, but very little is known of their function. In this review, we explore the emerging functions of neutrophils during pregnancy and their interactions with and regulation of T cells, a key adaptive immune cell population essential for the establishment of fetal-maternal tolerance.
Collapse
Affiliation(s)
- Serena Bert
- William Harvey Research InstituteBarts and the London School of MedicineQueen Mary UniversityLondonUK
| | - Eleanor J. Ward
- William Harvey Research InstituteBarts and the London School of MedicineQueen Mary UniversityLondonUK
| | - Suchita Nadkarni
- William Harvey Research InstituteBarts and the London School of MedicineQueen Mary UniversityLondonUK
| |
Collapse
|
19
|
|
20
|
The Enigma of Low-Density Granulocytes in Humans: Complexities in the Characterization and Function of LDGs during Disease. Pathogens 2021; 10:pathogens10091091. [PMID: 34578124 PMCID: PMC8470838 DOI: 10.3390/pathogens10091091] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/24/2021] [Accepted: 08/25/2021] [Indexed: 12/17/2022] Open
Abstract
Low-density granulocytes (LDGs) have been characterized as important immune cells during healthy and disease states in humans, including microbial infections, cancer, and autoimmune dysfunction. However, the classification of this cell type is similar to other immune cells (e.g., neutrophils, myeloid-derived suppressor cells) and ambiguous functional standards have rendered LDG identification and isolation daunting. Furthermore, most research involving LDGs has mainly focused on adult cells and subjects, leaving increased uncertainty surrounding younger populations, especially in vulnerable neonatal groups where LDG numbers are elevated. This review aims to bring together the current research in the field of LDG biology in the context of immunity to disease, with a focus on infection. In addition, we propose to highlight the gaps in the field that, if filled, could improve upon isolation techniques and functional characterizations for LDGs separate from neutrophils and myeloid-derived suppressor cells (MDSCs). This will not only enhance understanding of LDGs during disease processes and how they differ from other cell types but will also aid in the interpretation of comparative studies and results with the potential to inform development of novel therapeutics to improve disease states in patients.
Collapse
|
21
|
Vance JK, Rawson TW, Povroznik JM, Brundage KM, Robinson CM. Myeloid-Derived Suppressor Cells Gain Suppressive Function during Neonatal Bacterial Sepsis. Int J Mol Sci 2021; 22:ijms22137047. [PMID: 34208904 PMCID: PMC8268718 DOI: 10.3390/ijms22137047] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 06/27/2021] [Indexed: 11/21/2022] Open
Abstract
Neonates are at an increased risk of an infectious disease. This is consistent with an increased abundance of myeloid-derived suppressor cells (MDSCs) compared with older children and adults. Using a murine model of neonatal bacterial sepsis, we demonstrate that MDSCs modulate their activity during an infection to enhance immune suppressive functions. A gene expression analysis shows that MDSCs increased NOS2, Arg-1 and IL-27p28 expression in vitro and in vivo in response to Escherichia coli O1:K1:H7 and this is regulated at the level of the gene expression. Changes in the effector gene expression are consistent with increased enzymatic activity and cytokine secretion. The neonatal MDSCs express toll-like receptor (TLR) 2, 4 and 5 capable of recognizing pathogen-associated molecular patterns (PAMPS) on E. coli. However, a variable level of effector expression was achieved in response to LPS, peptidoglycan or flagellin. Individual bacterial PAMPs did not stimulate the expression of Arg-l and IL-27p28 equivalently to E. coli. However, the upregulation of NOS2 was achieved in response to LPS, peptidoglycan and flagella. The increased immune suppressive profile translated to an enhanced suppression of CD4+ T cell proliferation. Collectively, these findings increase our understanding of the dynamic nature of MDSC activity and suggest that these cells abundant in early life can acquire activity during an infection that suppresses protective immunity.
Collapse
Affiliation(s)
- Jordan K. Vance
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV 26506, USA; (J.K.V.); (T.W.R.); (J.M.P.); (K.M.B.)
| | - Travis W. Rawson
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV 26506, USA; (J.K.V.); (T.W.R.); (J.M.P.); (K.M.B.)
| | - Jessica M. Povroznik
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV 26506, USA; (J.K.V.); (T.W.R.); (J.M.P.); (K.M.B.)
| | - Kathleen M. Brundage
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV 26506, USA; (J.K.V.); (T.W.R.); (J.M.P.); (K.M.B.)
| | - Cory M. Robinson
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV 26506, USA; (J.K.V.); (T.W.R.); (J.M.P.); (K.M.B.)
- Vaccine Development Center, Department of Microbiology, Immunology, and Cell Biology, West Virginia University Health Sciences Center, Morgantown, WV 26506, USA
- Correspondence:
| |
Collapse
|
22
|
Zhang Y, Wang X, Zhang R, Wang X, Fu H, Yang W. MDSCs interactions with other immune cells and their role in maternal-fetal tolerance. Int Rev Immunol 2021; 41:534-551. [PMID: 34128752 DOI: 10.1080/08830185.2021.1938566] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
MDSCs (myeloid-derived suppressor cells) are a population of immature and heterogeneous bone marrow cells with immunosuppressive functions, and they are mainly divided into two subgroups: granulocytic MDSCs (G-MDSCs) and monocytic MDSCs (M-MDSCs). Immunosuppression is the main and most important function of MDSCs, and they mainly exert an inhibitory effect through endoplasmic reticulum stress and some enzymes related to inhibitors, as well as some cytokines and other factors. In addition, MDSCs also interact with other immune cells, especially NK cells, DCs and Tregs, to participate in immune regulation. A large number of MDSCs are found during normal pregnancy. Combined with their immunosuppressive effects, these results suggest that MDSCs are likely to be closely related to maternal-fetal immune tolerance. This review mainly shows the interaction of MDSCs with other immune cells and the important role of MDSCs in maternal-fetal tolerance. The current research shows that MDSCs are mainly mediated by STAT3, HLA-G, CXCR2, Arg-1 and HIF1-α in immune regulation during pregnancy. Interpreting maternal-fetal tolerance from the perspective of MDSCs provides a special perspective for research on immune regulation and maternal-fetal tolerance of MDSCs to obtain a more comprehensive understanding of immune regulation and immune tolerance.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xiaoya Wang
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Rongchao Zhang
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xi Wang
- Department of Clinical Laboratory, The first Hospital of Jilin University, Changchun, China
| | - Haiying Fu
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Wei Yang
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| |
Collapse
|
23
|
Liu J, Yang Q, Chen Z, Lv S, Tang J, Xing Z, Shi M, Lei A, Xiao G, He Y. TFF3 mediates the NF-κB/COX2 pathway to regulate PMN-MDSCs activation and protect against necrotizing enterocolitis. Eur J Immunol 2021; 51:1110-1125. [PMID: 33547649 DOI: 10.1002/eji.202048768] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 11/27/2020] [Accepted: 02/04/2021] [Indexed: 12/17/2022]
Abstract
Intestinal trefoil factor 3 (TFF3) plays an important role in repairing the intestinal mucosa. However, the detailed mechanism regarding immune regulation by TFF3 is not well defined. Here, we reported that treatment of mouse BM cells and human peripheral blood mononuclear cells from healthy volunteers with TFF3 activated polymorphnuclear myeloid-derived suppressor cells (PMN-MDSCs) in vitro. We also found that prostaglandin E2 is a major TFF3-mediated MDSC target, and that NF-κB/COX2 signaling was involved in this process. Moreover, TFF3 treatment or transfer of TFF3-derived PMN-MDSCs (TFF3-MDSCs) to experimental necrotizing enterocolitis (NEC) mice caused PMN-MDSC accumulation in the lamina propria (LP), which was associated with decreased intestinal inflammation, permeability, bacterial loading, and prolonged survival. Interestingly, no NEC severity remission was observed in Rag1 KO mice that were given TFF3-MDSCs, but coinjection with CD4+ T cells significantly relieved NEC inflammation. Overall, TFF3 mediates the NF-κB/COX2 pathway to regulate PMN-MDSC activation and attenuates NEC in a T-cell-dependent manner, which suggests a novel mechanism in preventing NEC occurrence.
Collapse
Affiliation(s)
- Jingping Liu
- Department of Clinical Laboratory, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, P. R. China
| | - Qiong Yang
- Chronic Disease Laboratory, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, P. R. China
| | - Ziyang Chen
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P. R. China
| | - Shuaijun Lv
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P. R. China
| | - Jian Tang
- Department of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, P. R. China
| | - Zhe Xing
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P. R. China
| | - Mengyu Shi
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P. R. China
| | - Aihua Lei
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, P. R. China
| | - Gang Xiao
- Department of Clinical Laboratory, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, P. R. China
| | - Yumei He
- Department of Clinical Laboratory, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, P. R. China.,Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P. R. China.,Guangdong Provincial Key Laboratory of Proteomics, Southern Medical University, Guangzhou, P. R. China
| |
Collapse
|
24
|
Sanchez-Pino MD, Dean MJ, Ochoa AC. Myeloid-derived suppressor cells (MDSC): When good intentions go awry. Cell Immunol 2021; 362:104302. [PMID: 33592540 DOI: 10.1016/j.cellimm.2021.104302] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 01/08/2021] [Accepted: 01/10/2021] [Indexed: 02/07/2023]
Abstract
MDSC are a heterogeneous population of immature myeloid cells that are released by biological stress such as tissue damage and inflammation. Conventionally, MDSC are known for their detrimental role in chronic inflammation and neoplastic conditions. However, their intrinsic functions in immunoregulation, wound healing, and angiogenesis are intended to protect from over-reactive immune responses, maintenance of immunotolerance, tissue repair, and homeostasis. Paradoxically, under certain conditions, MDSC can impair protective immune responses and exacerbate the disease. The transition from protective to harmful MDSC is most likely driven by environmental and epigenetic mechanisms induced by prolonged exposure to unresolved inflammatory triggers. Here, we review several examples of the dual impact of MDSC in conditions such as maternal-fetal tolerance, self-antigens immunotolerance, obesity-associated cancer, sepsis and trauma. Moreover, we also highlighted the evidence indicating that MDSC have a role in COVID-19 pathophysiology. Finally, we have summarized the evidence indicating epigenetic mechanisms associated with MDSC function.
Collapse
Affiliation(s)
- Maria Dulfary Sanchez-Pino
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center School of Medicine, New Orleans, LA 70112, USA; Department of Genetics, Louisiana State University Health Sciences Center School of Medicine, New Orleans, LA 70112, USA.
| | - Matthew J Dean
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center School of Medicine, New Orleans, LA 70112, USA
| | - Augusto C Ochoa
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center School of Medicine, New Orleans, LA 70112, USA; Department of Pediatrics, Louisiana State University Health Sciences Center School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
25
|
Kidzeru E, Gasper MA, Shao D, Edlefsen PT, Lejarcegui N, Havyarimana E, Urdahl K, Gantt S, Horton H, Jaspan H, Gervassi A. Myeloid-derived suppressor cells and their association with vaccine immunogenicity in South African infants. J Leukoc Biol 2021; 110:939-950. [PMID: 33477200 DOI: 10.1002/jlb.5a0420-281r] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 11/23/2020] [Accepted: 12/28/2020] [Indexed: 12/25/2022] Open
Abstract
The role of Myeloid-Derived Suppressor Cells (MDSC) in infant immune ontogeny is unknown. Here, we evaluated MDSC frequency and relationship with infant vaccine responses throughout the first year of life in a prospective cohort study. Ninety-one South African infant-mother pairs were enrolled at delivery, and blood samples were collected at 0, 6, 10, and 14 weeks, 6 months, 9 months, and 1 year. MDSC frequencies were quantified, and immune responses to the childhood vaccines Bacillus Calmette-Guérin (BCG), hepatitis B (HepB), and combination diphtheria, tetanus, and pertussis (dTaP) were measured by Ag-specific CD4+ T cell proliferation and interferon gamma (IFN-γ) production. Vaccine-specific Ab responses to HepB, dTaP, and Haemophilus influenzae type b (Hib) were quantified via Enzyme-Linked Immunosorbent assay (ELISA). MDSC frequency in mother-infant pairs was strongly correlated; the frequency of MDSC decreased in both mothers and infants during the months after delivery/birth; and by 1 year, infant MDSC frequencies rebounded to birth levels. Higher MDSC frequency at vaccination was associated with a lack of subsequent IFN-γ release in response to vaccine Ags, with the exception of BCG. With the exception of a weak, positive correlation between MDSC frequency at 6 weeks (time of initial vaccination) and peak Hepatitis B surface antigen Ab titer, Polymorphonuclear Myeloid-Derived Suppressor Cells (PMN-MDSC) was not correlated with T cell proliferation or Ab responses in this study. The potential for MDSC-mediated suppression of vaccine Ag-specific IFN-γ responses should be explored further, and considered when evaluating candidate infant vaccines.
Collapse
Affiliation(s)
- Elvis Kidzeru
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | | | - Danica Shao
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Paul T Edlefsen
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Nicholas Lejarcegui
- University of Washington Department of Global Health, Seattle, Washington, USA
| | - Enock Havyarimana
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Kevin Urdahl
- Seattle Children's Research Institute, Seattle, Washington, USA
| | - Soren Gantt
- University of British Columbia Department of Pediatrics and BC Children's Hospital Research Institute, Vancouver, Canada
| | - Helen Horton
- University of Washington Department of Global Health, Seattle, Washington, USA.,Jansen Pharmaceuticals, Beerse, Belgium
| | - Heather Jaspan
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,Seattle Children's Research Institute, Seattle, Washington, USA.,University of Washington Department of Global Health, Seattle, Washington, USA
| | - Ana Gervassi
- Seattle Children's Research Institute, Seattle, Washington, USA
| |
Collapse
|
26
|
Dietz S, Schwarz J, Rühle J, Schaller M, Fehrenbacher B, Marmé A, Schmid E, Peter A, Poets CF, Gille C, Köstlin-Gille N. Extracellular vesicles released by myeloid-derived suppressor cells from pregnant women modulate adaptive immune responses. Cell Immunol 2020; 361:104276. [PMID: 33517124 DOI: 10.1016/j.cellimm.2020.104276] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 12/18/2020] [Accepted: 12/22/2020] [Indexed: 12/19/2022]
Abstract
Immunological pregnancy complications are a main challenge in reproductive medicine. Mechanisms regulating the adaptation of the maternal immune system to pregnancy are incompletely understood and therapeutic options limited. Myeloid derived suppressor cells (MDSC) are immune-modulatory cells expanding during healthy pregnancy and seem to play a crucial role for maternal-fetal tolerance. Recent studies showed that exosomes produced by MDSC have immune-modulatory effects corresponding to their parental cells under different pathological conditions. Here, we investigated immunological effects of exosomes of GR-MDSC during pregnancy. Isolated GR-MDSC exosomes from peripheral blood of pregnant women were tested for functionality in different in vitro assays. We show that GR-MDSC exosomes exhibited profound immune-modulatory effects such as suppression of T-cell proliferation, T helper 2 (Th2)-cell polarization, induction of regulatory T-cells and inhibition of lymphocyte cytotoxicity. Our results confirm that MDSC-derived exosomes functionally correspond to their parental cells and identify them as an interesting therapeutic target for immunological pregnancy complications.
Collapse
Affiliation(s)
- Stefanie Dietz
- Tuebingen University Children's Hospital, Department of Neonatology, Tuebingen, Germany
| | - Julian Schwarz
- Tuebingen University Children's Hospital, Department of Neonatology, Tuebingen, Germany
| | - Jessica Rühle
- Tuebingen University Children's Hospital, Department of Neonatology, Tuebingen, Germany
| | - Martin Schaller
- Department of Dermatology, University of Tuebingen, Tuebingen, Germany
| | | | | | - Evi Schmid
- Department of Pediatric Surgery & Pediatric Urology, University of Tuebingen, Germany
| | - Andreas Peter
- German Centre for Diabetes Research (DZD), Tuebingen, Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tuebingen, Tuebingen, Germany; Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, Germany
| | - Christian F Poets
- Tuebingen University Children's Hospital, Department of Neonatology, Tuebingen, Germany
| | - Christian Gille
- Tuebingen University Children's Hospital, Department of Neonatology, Tuebingen, Germany.
| | | |
Collapse
|
27
|
Köstlin-Gille N, Gille C. Myeloid-Derived Suppressor Cells in Pregnancy and the Neonatal Period. Front Immunol 2020; 11:584712. [PMID: 33162999 PMCID: PMC7581934 DOI: 10.3389/fimmu.2020.584712] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 09/21/2020] [Indexed: 12/14/2022] Open
Abstract
During pregnancy, the immune systems of mother and offspring are challenged by their close adjacency to balance tolerance and rejection. After birth the neonate has to continue this balance towards its new environment by tolerating commensals while rejecting pathogens and towards its developing tissues to avoid inflammatory damage while overcoming immunosuppression. Our group was the first to link immunosuppressive features of myeloid derived suppressor cells (MDSC) to materno-fetal tolerance, neonatal susceptibility to infection and inflammation control. Here we summarize recent advances in this dynamic field.
Collapse
Affiliation(s)
| | - Christian Gille
- Department of Neonatology, University Children's Hospital Tuebingen, Tübingen, Germany
| |
Collapse
|
28
|
Köstlin-Gille N, Flaig LA, Ginzel M, Arand J, Poets CF, Gille C. Granulocytic Myeloid-Derived Suppressor Cells in Breast Milk (BM-MDSC) Correlate with Gestational Age and Postnatal Age and Are Influenced by Infant's Sex. Nutrients 2020; 12:nu12092571. [PMID: 32854237 PMCID: PMC7551580 DOI: 10.3390/nu12092571] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/18/2020] [Accepted: 08/24/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Infections are the main cause of death in preterm infants. Causative agents often descend from the intestinal flora of the infected neonate, indicating insufficient protection by the mucosal barrier. Breast milk (BM) contains different subsets of immune cells. We recently showed that BM contains significant numbers of myeloid-derived suppressor cells (MDSC)-immune cells that actively suppress pro-inflammatory immune responses-and hypothesized that the transfer of BM-MDSC may modulate the mucosal immunity of the newborn. METHODS Percentages of MDSC in the BM from mothers of 86 preterm infants between 23 + 0 and 36 + 6 weeks of gestation during their first five postnatal weeks were analyzed by flow cytometry and correlated with maternal and infant characteristics. RESULTS Percentages of BM-MDSC positively correlated with gestational age and postnatal age. The expression of activation markers on BM-MDSC did not change with gestational age, but it decreased with postnatal age. Mothers who received antepartum tocolytics had lower percentages of BM-MDSC, and infant's sex strongly influenced percentages of BM-MDSC. CONCLUSION Our results point toward a role of BM-MDSC for immune regulation in the neonatal gut, making them a potential target of immune-based therapies shortly after birth.
Collapse
|
29
|
Kobbe P, Bläsius FM, Lichte P, Oberbeck R, Hildebrand F. Neuroendocrine Modulation of the Immune Response after Trauma and Sepsis: Does It Influence Outcome? J Clin Med 2020; 9:jcm9072287. [PMID: 32708472 PMCID: PMC7408630 DOI: 10.3390/jcm9072287] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/13/2020] [Accepted: 07/16/2020] [Indexed: 12/29/2022] Open
Abstract
Although the treatment of multiple-injured patients has been improved during the last decades, sepsis and multiple organ failure (MOF) still remain the major cause of death. Following trauma, profound alterations of a large number of physiological systems can be observed that may potentially contribute to the development of sepsis and MOF. This includes alterations of the neuroendocrine and the immune system. A large number of studies focused on posttraumatic changes of the immune system, but the cause of posttraumatic immune disturbance remains to be established. However, an increasing number of data indicate that the bidirectional interaction between the neuroendocrine and the immune system may be an important mechanism involved in the development of sepsis and MOF. The aim of this article is to highlight the current knowledge of the neuroendocrine modulation of the immune system during trauma and sepsis.
Collapse
Affiliation(s)
- Philipp Kobbe
- Deparment of Trauma and Reconstructive Surgery, University Hospital RWTH Aachen, D-52074 Aachen, Germany; (P.K.); (F.M.B.); (P.L.)
| | - Felix M. Bläsius
- Deparment of Trauma and Reconstructive Surgery, University Hospital RWTH Aachen, D-52074 Aachen, Germany; (P.K.); (F.M.B.); (P.L.)
| | - Philipp Lichte
- Deparment of Trauma and Reconstructive Surgery, University Hospital RWTH Aachen, D-52074 Aachen, Germany; (P.K.); (F.M.B.); (P.L.)
| | - Reiner Oberbeck
- Deparment of Trauma and Hand Surgery, Wald-Klinikum, 07548 Gera, Germany;
| | - Frank Hildebrand
- Deparment of Trauma and Reconstructive Surgery, University Hospital RWTH Aachen, D-52074 Aachen, Germany; (P.K.); (F.M.B.); (P.L.)
- Correspondence: ; Tel.: +49-241-89350
| |
Collapse
|
30
|
Zheng ZM, Yang HL, Lai ZZ, Wang CJ, Yang SL, Li MQ, Shao J. Myeloid-derived suppressor cells in obstetrical and gynecological diseases. Am J Reprod Immunol 2020; 84:e13266. [PMID: 32418253 DOI: 10.1111/aji.13266] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/08/2020] [Accepted: 05/11/2020] [Indexed: 12/15/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous group of myeloid-origin cells which have immunosuppressive activities in several conditions, such as cancer and inflammation. Recent research has also associated MDSCs with numerous obstetrical and gynecological diseases. During pregnancy, MDSCs accumulate to ensure maternal-fetal immune tolerance, whereas they are decreased in patients who suffer from early miscarriage or pre-eclampsia. While the etiology of endometriosis is still unknown, abnormal accumulation of MDSCs in the peripheral blood and peritoneal fluid, alongside an increased level of reactive oxygen species (ROS), has been observed in these patients, which is central to the cellular immune regulations by MDSCs. Additionally, the regulation of MDSCs observed in tumours is also applicable to gynecologic neoplasms, including ovarian cancer and cervical cancer. More recently, emerging evidence has shown that there are high levels of MDSCs in premature ovarian failure (POF) and in vitro fertilization (IVF), but the underlying mechanisms are unknown. In this review, the generation and mechanisms of MDSCs are summarized. In particular, the modulation of these cells in immune-related obstetrical and gynecological diseases is discussed, including potential treatment options targeting MDSCs.
Collapse
Affiliation(s)
- Zi-Meng Zheng
- Insitute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China.,NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Hui-Li Yang
- Insitute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China.,NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Zhen-Zhen Lai
- Insitute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China.,NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Cheng-Jie Wang
- Insitute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Shao-Liang Yang
- Insitute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China.,NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Ming-Qing Li
- Insitute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China.,NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Jun Shao
- Insitute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| |
Collapse
|
31
|
Weinhage T, Kölsche T, Rieger-Fackeldey E, Schmitz R, Antoni AC, Ahlmann M, Foell D, Wittkowski H. Cord Blood Low-Density Granulocytes Correspond to an Immature Granulocytic Subset with Low Expression of S100A12. THE JOURNAL OF IMMUNOLOGY 2020; 205:56-66. [PMID: 32444390 DOI: 10.4049/jimmunol.1901308] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 04/22/2020] [Indexed: 12/22/2022]
Abstract
Although substantial progress has been achieved concerning neonatal sepsis, its lethality remains considerably high, and further insights into peculiarities and malfunctions of neonatal immunity are needed. This study aims to contribute to a better understanding of the role of human neonatal granulocyte subpopulations and calgranulin C (S100A12). For this purpose, we gathered 136 human cord blood (CB) samples. CD66b+ CB low-density granulocytes (LDG) and CB normal-density granulocytes were isolated and functionally and phenotypically compared with healthy adult control granulocytes. We could identify CB-LDG as CD66bbright CD64high CD16low CD35low CD10low S100A12med-low and, based on these markers, recovered in whole CB stainings. Consistent with flow cytometric findings, microscopic imaging supported an immature phenotype of CB-LDG with decreased S100A12 expression. In CB serum of healthy neonates, S100A12 was found to be higher in female newborns when compared with males. Additionally, S100A12 levels correlated positively with gestational age independently from sex. We could solidify functional deficits of CB-LDG concerning phagocytosis and generation of neutrophil extracellular traps. Our study reveals that previously described suppressive effects of CB-LDG on CD4+ T cell proliferation are exclusively due to phagocytosis of stimulation beads used in cocultures and absent when using soluble or coated Abs. In conclusion, we characterize CB-LDG as immature neutrophils with functional deficits and decreased expression and storage of S100A12. Concerning their cross-talk with the adaptive immunity, we found no direct inhibitory effect of LDG. Neonatal LDG may thus represent a distinct population that differs from LDG populations found in adults.
Collapse
Affiliation(s)
- Toni Weinhage
- Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster, 48149 Münster, Germany
| | - Tristan Kölsche
- Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster, 48149 Münster, Germany
| | - Esther Rieger-Fackeldey
- Department of Pediatrics, Technical University of Munich, 80804 Munich, Germany.,Department of General Pediatrics, University Children's Hospital Münster, 48149 Münster, Germany
| | - Ralf Schmitz
- Department of Gynecology and Obstetrics, University Hospital Münster, 48149 Münster, Germany
| | - Anne-Charlotte Antoni
- Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster, 48149 Münster, Germany
| | - Martina Ahlmann
- Department of Pediatric Oncology and Hematology, University Hospital Münster, 48149 Münster, Germany; and
| | - Dirk Foell
- Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster, 48149 Münster, Germany.,Interdisciplinary Centre of Clinical Research, University of Münster, 48149 Münster, Germany
| | - Helmut Wittkowski
- Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster, 48149 Münster, Germany;
| |
Collapse
|
32
|
Viemann D. S100-Alarmins Are Essential Pilots of Postnatal Innate Immune Adaptation. Front Immunol 2020; 11:688. [PMID: 32425933 PMCID: PMC7203218 DOI: 10.3389/fimmu.2020.00688] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/26/2020] [Indexed: 12/14/2022] Open
Abstract
The restricted capacity of newborn infants to mount inflammatory responses toward microbial challenges has traditionally been linked to the high risk of septic diseases during the neonatal period. In recent years, substantial evidence has been provided that this characteristic of the neonatal immune system is actually a meaningful physiologic state that is based on specific transiently active cellular and molecular mechanisms and required for a favorable course of postnatal immune adaptation. The identification of physiologically high amounts of S100-alarmins in neonates has been one of the crucial pieces in the puzzle that contributed to the change of concept. In this context, innate immune immaturity could be redefined and assigned to the epigenetic silence of adult-like cell-autonomous regulation at the beginning of life. S100-alarmins represent an alternative age-specific mechanism of immune regulation that protects neonates from hyperinflammatory immune responses. Here, we summarize how infants are provided with S100-alarmins and why these allow an uneventful clash between the innate immune system and the extrauterine world. The mode of action of S100-alarmins is highlighted including their tuning functions at multiple levels for establishing a state of homeostasis with the environment in the newborn individual.
Collapse
Affiliation(s)
- Dorothee Viemann
- Department of Pediatric Pneumology, Allergy and Neonatology, Hannover Medical School, Hanover, Germany.,Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hanover, Germany.,PRIMAL Consortium, Hanover, Germany
| |
Collapse
|
33
|
Cao P, Sun Z, Feng C, Zhang J, Zhang F, Wang W, Zhao Y. Myeloid-derived suppressor cells in transplantation tolerance induction. Int Immunopharmacol 2020; 83:106421. [PMID: 32217462 DOI: 10.1016/j.intimp.2020.106421] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 02/29/2020] [Accepted: 03/16/2020] [Indexed: 02/06/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) are a group of heterogeneous cells derived from bone marrow. These cells are developed from immature myeloid cells and have strong negative immunomodulatory effects. In the context of pathology (such as tumor, autoimmune disease, trauma, and burns), MDSCs accumulate around tumor and inflammatory tissues, where their main role is to inhibit the function of effector T cells and promote the recruitment of regulatory T cells. MDSCs can be used in organ transplantation to regulate the immune responses that participate in rejection of the transplanted organ. This effect is achieved by increasing the production of MDSCs in vivo or transfusion of MDSCs induced in vitro to establish immune tolerance and prolong the survival of the graft. In this review, we discuss the efficacy of MDSCs in a variety of transplantation studies as well as the induction of immune tolerance to prevent transplant rejection through the use of common clinical immunosuppressants combined with MDSCs.
Collapse
Affiliation(s)
- Peng Cao
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Zejia Sun
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Chang Feng
- Transplantation Biology Research Division, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing 100101, China
| | - Jiandong Zhang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Feilong Zhang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Wei Wang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China.
| | - Yong Zhao
- Transplantation Biology Research Division, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
34
|
A perfect storm: fetal inflammation and the developing immune system. Pediatr Res 2020; 87:319-326. [PMID: 31537013 PMCID: PMC7875080 DOI: 10.1038/s41390-019-0582-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 08/20/2019] [Accepted: 08/23/2019] [Indexed: 12/17/2022]
Abstract
Histologic chorioamnionitis is an inflammatory disorder of the placenta that commonly precedes preterm delivery. Preterm birth related to chorioamnionitis and fetal inflammation has been associated with a risk for serious inflammatory complications in infancy. In addition, preterm infants exposed to chorioamnionitis may be more susceptible to infection in the neonatal intensive care unit and possibly later in life. A significant body of work has established an association between chorioamnionitis and inflammatory processes. However, the potential consequences of this inflammation on postnatal immunity are less understood. In this review, we will discuss current knowledge regarding the effects of fetal exposure to inflammation on postnatal immune responses.
Collapse
|
35
|
Dietz S, Schwarz J, Vogelmann M, Spring B, Molnár K, Orlikowsky TW, Wiese F, Holzer U, Poets CF, Gille C, Köstlin-Gille N. Cord blood granulocytic myeloid-derived suppressor cells impair monocyte T cell stimulatory capacity and response to bacterial stimulation. Pediatr Res 2019; 86:608-615. [PMID: 31349362 DOI: 10.1038/s41390-019-0504-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Revised: 05/20/2019] [Accepted: 07/15/2019] [Indexed: 11/09/2022]
Abstract
BACKGROUND Neonatal sepsis is a leading cause of perinatal morbidity and mortality. In comparison to adults, neonates exhibit a higher susceptibility to infections. Myeloid-derived suppressor cells (MDSCs) are myeloid cells with suppressive activity on other immune cells accumulating during foetal life and controlling inflammation in neonates. Most studies investigating the mechanisms for MDSC-mediated immune suppression have been focused on T-cells. Thus far, little is known about the role of MDSC for monocyte function. METHODS The impact of human cord blood MDSCs (CB-MDSCs) on monocytes was investigated in an in vitro model. CB-MDSCs were co-cultured with peripheral blood mononuclear cells and monocytes were analysed for expression of surface markers, T cell stimulatory and phagocytic capacity, as well as the production of intracellular cytokines by flow cytometry. RESULTS CB-MDSCs increased the expression of co-inhibitory molecules and decreased the expression of major histocompatibility complex class II molecules on monocytes, leading to an impaired T-cell stimulatory capacity. Upon bacterial stimulation, expression of phagocytosis receptors, phagocytosis rates and production of tumor necrosis factor-α by monocytes was diminished by CB-MDSCs. CONCLUSION We show that CB-MDSCs profoundly modulate monocyte functions, thereby indirectly impairing T-cell activation. Further research is needed to figure out if MDSCs could be a therapeutic target for inflammatory diseases in neonates like neonatal sepsis.
Collapse
Affiliation(s)
- Stefanie Dietz
- Department of Neonatology, Tübingen University Children's Hospital, Tübingen, Germany
| | - Julian Schwarz
- Department of Neonatology, Tübingen University Children's Hospital, Tübingen, Germany
| | - Margit Vogelmann
- Department of Neonatology, Tübingen University Children's Hospital, Tübingen, Germany
| | - Bärbel Spring
- Department of Neonatology, Tübingen University Children's Hospital, Tübingen, Germany
| | - Kriszta Molnár
- Department of Neonatology, Tübingen University Children's Hospital, Tübingen, Germany
| | | | - Franziska Wiese
- Department of Hematology and Oncology, Tübingen University Children's Hospital, Tuebingen, Germany
| | - Ursula Holzer
- Department of Hematology and Oncology, Tübingen University Children's Hospital, Tuebingen, Germany
| | - Christian F Poets
- Department of Neonatology, Tübingen University Children's Hospital, Tübingen, Germany
| | - Christian Gille
- Department of Neonatology, Tübingen University Children's Hospital, Tübingen, Germany.
| | | |
Collapse
|
36
|
Liu Y, Perego M, Xiao Q, He Y, Fu S, He J, Liu W, Li X, Tang Y, Li X, Yuan W, Zhou W, Wu F, Jia C, Cui Q, Worthen GS, Jensen EA, Gabrilovich DI, Zhou J. Lactoferrin-induced myeloid-derived suppressor cell therapy attenuates pathologic inflammatory conditions in newborn mice. J Clin Invest 2019; 129:4261-4275. [PMID: 31483289 PMCID: PMC6763238 DOI: 10.1172/jci128164] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 07/09/2019] [Indexed: 12/13/2022] Open
Abstract
Inflammation plays a critical role in the development of severe neonatal morbidities. Myeloid-derived suppressor cells (MDSCs) were recently implicated in the regulation of immune responses in newborns. Here, we report that the presence of MDSCs and their functional activity in infants are closely associated with the maturity of newborns and the presence of lactoferrin (LF) in serum. Low amounts of MDSCs at birth predicted the development of severe pathology in preterm infants - necrotizing enterocolitis (NEC). In vitro treatment of newborn neutrophils and monocytes with LF converted these cells to MDSCs via the LRP2 receptor and activation of the NF-κB transcription factor. Decrease in the expression of LRP2 was responsible for the loss of sensitivity of adult myeloid cells to LF. LF-induced MDSCs (LF-MDSCs) were effective in the treatment of newborn mice with NEC, acting by blocking inflammation, resulting in increased survival. LF-MDSCs were more effective than treatment with LF protein alone. In addition to affecting NEC, LF-MDSCs demonstrated potent ability to control ovalbumin-induced (OVA-induced) lung inflammation, dextran sulfate sodium-induced (DSS-induced) colitis, and concanavalin A-induced (ConA-induced) hepatitis. These results suggest that cell therapy with LF-MDSCs may provide potent therapeutic benefits in infants with various pathological conditions associated with dysregulated inflammation.
Collapse
MESH Headings
- Adult
- Animals
- Animals, Newborn
- Cell- and Tissue-Based Therapy/methods
- Disease Models, Animal
- Enterocolitis, Necrotizing/immunology
- Enterocolitis, Necrotizing/pathology
- Enterocolitis, Necrotizing/therapy
- Female
- Humans
- In Vitro Techniques
- Infant, Newborn
- Infant, Premature
- Inflammation/immunology
- Inflammation/pathology
- Inflammation/therapy
- Lactoferrin/immunology
- Lactoferrin/pharmacology
- Low Density Lipoprotein Receptor-Related Protein-2/immunology
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Transgenic
- Myeloid-Derived Suppressor Cells/drug effects
- Myeloid-Derived Suppressor Cells/immunology
- Myeloid-Derived Suppressor Cells/transplantation
- NF-kappa B/immunology
Collapse
Affiliation(s)
- Yufeng Liu
- Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Sciences, Tianjin Medical University, Tianjin, China
| | | | - Qiang Xiao
- Institute of Human Virology, Zhongshan School of Medicine
| | - Yumei He
- Institute of Human Virology, Zhongshan School of Medicine
| | - Shuyu Fu
- Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Juan He
- Institute of Human Virology, Zhongshan School of Medicine
| | | | - Xing Li
- Third Affiliated Hospital, Sun Yat-sen University (SYSU), Guangzhou, China
| | | | | | - Weiming Yuan
- Guangzhou Women and Children’s Medical Centre, Guangzhou, China
| | - Wei Zhou
- Guangzhou Women and Children’s Medical Centre, Guangzhou, China
| | - Fan Wu
- Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Chunhong Jia
- Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qiliang Cui
- Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - George S. Worthen
- Children’s Hospital of Philadelphia (CHOP), Philadelphia, Pennsylvania, USA
| | - Erik A. Jensen
- Children’s Hospital of Philadelphia (CHOP), Philadelphia, Pennsylvania, USA
| | | | - Jie Zhou
- Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Sciences, Tianjin Medical University, Tianjin, China
| |
Collapse
|
37
|
Ahmadi M, Mohammadi M, Ali-Hassanzadeh M, Zare M, Gharesi-Fard B. MDSCs in pregnancy: Critical players for a balanced immune system at the feto-maternal interface. Cell Immunol 2019; 346:103990. [PMID: 31703912 DOI: 10.1016/j.cellimm.2019.103990] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/08/2019] [Accepted: 09/23/2019] [Indexed: 02/06/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) have emerged as a new immune regulator at the feto-maternal interface. Although the phenotypes and functions of these cells were primarily studied in pathological conditions such as cancers and infections, new evidence has underscored their beneficial roles in homeostasis and physiological circumstances such as normal pregnancy. In this regard, studies have shown an increased number of MDSCs, particularly granulocytic MDSCs, at the feto-maternal interface. These cells participate in maintaining immunological tolerance between mother and semi-allograft fetus through various mechanisms. They further seem to play critical roles in placentation and fetus growth process. The absence or dysregulation of MDSCs during pregnancy have been reported in several pregnancy complications. These cells are also abundant in the cord blood of neonates so as to balance the immune responses and prevent aggressive inflammatory responses. The current review summarizes and organizes detailed data on MDSCs and their roles during pregnancy.
Collapse
Affiliation(s)
- Moslem Ahmadi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mobin Mohammadi
- Cancer and Immunology Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran; Department of Immunology and Hematology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Mohammad Ali-Hassanzadeh
- Department of Immunology, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran; Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Zare
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Behrouz Gharesi-Fard
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran; Infertility Research Center, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
38
|
Silvestre-Roig C, Fridlender ZG, Glogauer M, Scapini P. Neutrophil Diversity in Health and Disease. Trends Immunol 2019; 40:565-583. [PMID: 31160207 PMCID: PMC7185435 DOI: 10.1016/j.it.2019.04.012] [Citation(s) in RCA: 340] [Impact Index Per Article: 56.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/26/2019] [Accepted: 04/28/2019] [Indexed: 12/17/2022]
Abstract
New evidence has challenged the outdated dogma that neutrophils are a homogeneous population of short-lived cells. Although neutrophil subpopulations with distinct functions have been reported under homeostatic and pathological conditions, a full understanding of neutrophil heterogeneity and plasticity is currently lacking. We review here current knowledge of neutrophil heterogeneity and diversity, highlighting the need for deep genomic, phenotypic, and functional profiling of the identified neutrophil subpopulations to determine whether these cells truly represent bona fide novel neutrophil subsets. We suggest that progress in understanding neutrophil heterogeneity will allow the identification of clinically relevant neutrophil subpopulations that may be used in the diagnosis of specific diseases and lead to the development of new therapeutic approaches.
Collapse
Affiliation(s)
- Carlos Silvestre-Roig
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
| | - Zvi G Fridlender
- Institute of Pulmonary Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Michael Glogauer
- Faculty of Dentistry, University of Toronto, and Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Patrizia Scapini
- Department of Medicine, Section of General Pathology, School of Medicine, University of Verona, Verona, Italy.
| |
Collapse
|
39
|
Pawelec G, Verschoor CP, Ostrand-Rosenberg S. Myeloid-Derived Suppressor Cells: Not Only in Tumor Immunity. Front Immunol 2019; 10:1099. [PMID: 31156644 PMCID: PMC6529572 DOI: 10.3389/fimmu.2019.01099] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 04/30/2019] [Indexed: 12/15/2022] Open
Abstract
Since the realization that immature myeloid cells are powerful modulators of the immune response, many studies on “myeloid-derived suppressor cells” (MDSCs) have documented their ability to promote tumor progression in melanoma and other cancers. Whether MDSCs are induced solely pathologically in tumorigenesis, or whether they also represent physiological immune control mechanisms, is not well-understood, but is particularly important in the light of ongoing attempts to block their activities in order to enhance anti-tumor immunity. Here, we briefly review studies which explore (1) how best to identify MDSCs in the context of cancer and how this compares to other conditions in humans; (2) what the suppressive mechanisms of MDSCs are and how to target them pharmacologically; (3) whether levels of MDSCs with various phenotypes are informative for clinical outcome not only in cancer but also other diseases, and (4) whether MDSCs are only found under pathological conditions or whether they also represent a physiological regulatory mechanism for the feedback control of immunity. Studies unequivocally document that MDSCs strongly influence cancer outcomes, but are less informative regarding their relevance to infection, autoimmunity, transplantation and aging, especially in humans. So far, the results of clinical interventions to reverse their negative effects in cancer have been disappointing; thus, developing differential approaches to modulate MSDCs in cancer and other diseases without unduly comprising any normal physiological function requires further exploration.
Collapse
Affiliation(s)
- Graham Pawelec
- Department of Immunology, University of Tübingen, Tübingen, Germany.,Health Sciences North Research Institute, Sudbury, ON, Canada
| | - Chris P Verschoor
- Health Sciences North Research Institute, Sudbury, ON, Canada.,Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, ON, Canada
| | - Suzanne Ostrand-Rosenberg
- Department of Pathology and Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
40
|
Schrijver IT, Théroude C, Roger T. Myeloid-Derived Suppressor Cells in Sepsis. Front Immunol 2019; 10:327. [PMID: 30873175 PMCID: PMC6400980 DOI: 10.3389/fimmu.2019.00327] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 02/08/2019] [Indexed: 12/16/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are immature myeloid cells characterized by their immunosuppressive functions. MDSCs expand during chronic and acute inflammatory conditions, the best described being cancer. Recent studies uncovered an important role of MDSCs in the pathogenesis of infectious diseases along with sepsis. Here we discuss the mechanisms underlying the expansion and immunosuppressive functions of MDSCs, and the results of preclinical and clinical studies linking MDSCs to sepsis pathogenesis. Strikingly, all clinical studies to date suggest that high proportions of blood MDSCs are associated with clinical worsening, the incidence of nosocomial infections and/or mortality. Hence, MDSCs are attractive biomarkers and therapeutic targets for sepsis, especially because these cells are barely detectable in healthy subjects. Blocking MDSC-mediated immunosuppression and trafficking or depleting MDSCs might all improve sepsis outcome. While some key aspects of MDSCs biology need in depth investigations, exploring these avenues may participate to pave the way toward the implementation of personalized medicine and precision immunotherapy for patients suffering from sepsis.
Collapse
Affiliation(s)
- Irene T Schrijver
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital, Epalinges, Switzerland
| | - Charlotte Théroude
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital, Epalinges, Switzerland
| | - Thierry Roger
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital, Epalinges, Switzerland
| |
Collapse
|
41
|
Gleave Parson M, Grimmett J, Vance JK, Witt MR, Seman BG, Rawson TW, Lyda L, Labuda C, Jung JY, Bradford SD, Robinson CM. Murine myeloid-derived suppressor cells are a source of elevated levels of interleukin-27 in early life and compromise control of bacterial infection. Immunol Cell Biol 2019; 97:445-456. [PMID: 30575117 DOI: 10.1111/imcb.12224] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 12/17/2018] [Accepted: 12/17/2018] [Indexed: 12/17/2022]
Abstract
Microbial infections early in life remain a major cause of infant mortality worldwide. This is consistent with immune deficiencies in this population. Interleukin (IL)-27 is suppressive toward a variety of immune cell types, and we have shown that the production of IL-27 is elevated in humans and mice early in life. We hypothesize that elevated levels of IL-27 oppose protective responses to infection during the neonatal period. In this study, we extended previous findings in neonatal mice to identify a population of IL-27 producers that express Gr-1 and were further identified as myeloid-derived suppressor cells (MDSCs) based on the expression of surface markers and functional studies. In neonates, MDSCs are more abundant and contribute to the elevated pool of IL-27 in this population. Although the ability of MDSCs to regulate T lymphocyte activation has been well-studied, sparingly few studies have investigated the influence of MDSCs on innate immune function during bacterial infection. We demonstrate that macrophages are impaired in their ability to control growth of Escherichia coli when cocultured with MDSCs. This bacterium is a significant concern for neonates as a common cause of bacterial sepsis and meningitis. The suppressive effect of MDSCs on macrophage function is mediated by IL-27; inclusion of a reagent to neutralize IL-27 promotes improved control of bacterial growth. Taken together, these results suggest that the increased abundance of MDSCs may contribute to early life susceptibility to infection and further highlight production of IL-27 as a novel MDSC mechanism to suppress immunity.
Collapse
Affiliation(s)
- Madeline Gleave Parson
- Biomedical Sciences Department, West Virginia School of Osteopathic Medicine, Lewisburg, WV, USA
| | - Juanita Grimmett
- Biomedical Sciences Department, West Virginia School of Osteopathic Medicine, Lewisburg, WV, USA
| | - Jordan K Vance
- Department of Microbiology, Immunology, & Cell Biology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Michelle R Witt
- Department of Microbiology, Immunology, & Cell Biology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Brittany G Seman
- Department of Microbiology, Immunology, & Cell Biology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Travis W Rawson
- Department of Microbiology, Immunology, & Cell Biology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Logan Lyda
- Biomedical Sciences Department, West Virginia School of Osteopathic Medicine, Lewisburg, WV, USA
| | - Christopher Labuda
- Biomedical Sciences Department, West Virginia School of Osteopathic Medicine, Lewisburg, WV, USA
| | - Joo-Yong Jung
- Department of Biology, Briar Cliff University, Sioux City, IA, USA
| | - Shelby D Bradford
- Department of Microbiology, Immunology, & Cell Biology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Cory M Robinson
- Department of Microbiology, Immunology, & Cell Biology, West Virginia University School of Medicine, Morgantown, WV, USA
| |
Collapse
|
42
|
Köstlin-Gille N, Dietz S, Schwarz J, Spring B, Pauluschke-Fröhlich J, Poets CF, Gille C. HIF-1α-Deficiency in Myeloid Cells Leads to a Disturbed Accumulation of Myeloid Derived Suppressor Cells (MDSC) During Pregnancy and to an Increased Abortion Rate in Mice. Front Immunol 2019; 10:161. [PMID: 30804946 PMCID: PMC6370686 DOI: 10.3389/fimmu.2019.00161] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 01/18/2019] [Indexed: 12/11/2022] Open
Abstract
Abortions are the most important reason for unintentional childlessness. During pregnancy, maternal immune cells are in close contact to cells of the semi-allogeneic fetus. Dysregulation of the maternal immune system leading to defective adaptation to pregnancy often plays a role in pathogenesis of abortions. Myeloid-derived suppressor cells (MDSC) are myeloid cells that suppress functions of other immune cells, especially T-cells, thereby negatively affecting diseases such as cancer, sepsis or trauma. They seem, however, also necessary for maintenance of maternal-fetal tolerance. Mechanisms regulating MDSC expansion and function during pregnancy are only incompletely understood. In tumor environment, hypoxia is crucial for MDSC accumulation and activation. Hypoxia is also important for early placenta and embryo development. Effects of hypoxia are mediated through hypoxia-inducible factor 1α (HIF-1α). In the present study we aimed to examine the role of HIF-1α in myeloid cells for MDSC accumulation and MDSC function during pregnancy and for pregnancy outcome. We therefore used a mouse model with targeted deletion of HIF-1α in myeloid cells (myeloid HIF-KO) and analyzed blood, spleens and uteri of pregnant mice at gestational day E 10.5 in comparison to non-pregnant animals and wildtype (WT) animals. Further we analyzed pregnancy success by determining rates of failed implantation and abortion in WT and myeloid HIF-KO animals. We found that myeloid HIF-KO in mice led to an abrogated MDSC accumulation in the pregnant uterus and to impaired suppressive activity of MDSC. While expression of chemokine receptors and integrins on MDSC was not affected by HIF-1α, myeloid HIF-KO led to increased apoptosis rates of MDSC in the uterus. Myeloid-HIF-KO resulted in increased proportions of non-pregnant animals after positive vaginal plug and increased abortion rates, suggesting that activation of HIF-1α dependent pathways in MDSC are important for maintenance of pregnancy.
Collapse
Affiliation(s)
| | - Stefanie Dietz
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany
| | - Julian Schwarz
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany
| | - Bärbel Spring
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany
| | | | - Christian F Poets
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany
| | - Christian Gille
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany
| |
Collapse
|
43
|
Cassetta L, Baekkevold ES, Brandau S, Bujko A, Cassatella MA, Dorhoi A, Krieg C, Lin A, Loré K, Marini O, Pollard JW, Roussel M, Scapini P, Umansky V, Adema GJ. Deciphering myeloid-derived suppressor cells: isolation and markers in humans, mice and non-human primates. Cancer Immunol Immunother 2019; 68:687-697. [PMID: 30684003 PMCID: PMC6447515 DOI: 10.1007/s00262-019-02302-2] [Citation(s) in RCA: 168] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 01/11/2019] [Indexed: 12/12/2022]
Abstract
In cancer, infection and inflammation, the immune system's function can be dysregulated. Instead of fighting disease, immune cells may increase pathology and suppress host-protective immune responses. Myeloid cells show high plasticity and adapt to changing conditions and pathological challenges. Despite their relevance in disease pathophysiology, the identity, heterogeneity and biology of myeloid cells is still poorly understood. We will focus on phenotypical and functional markers of one of the key myeloid regulatory subtypes, the myeloid derived suppressor cells (MDSC), in humans, mice and non-human primates. Technical issues regarding the isolation of the cells from tissues and blood, timing and sample handling of MDSC will be detailed. Localization of MDSC in a tissue context is of crucial importance and immunohistochemistry approaches for this purpose are discussed. A minimal antibody panel for MDSC research is provided as part of the Mye-EUNITER COST action. Strategies for the identification of additional markers applying state of the art technologies such as mass cytometry will be highlighted. Such marker sets can be used to study MDSC phenotypes across tissues, diseases as well as species and will be crucial to accelerate MDSC research in health and disease.
Collapse
Affiliation(s)
- Luca Cassetta
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, EH16 4TJ, Edinburgh, UK.
| | - Espen S Baekkevold
- Centre for Immune Regulation, Department of Pathology, University of Oslo, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Sven Brandau
- West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Anna Bujko
- Centre for Immune Regulation, Department of Pathology, University of Oslo, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Marco A Cassatella
- Division of General Pathology, Department of Medicine, University of Verona, Verona, Italy
| | - Anca Dorhoi
- Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, 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
| | - Carsten Krieg
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, USA
| | - Ang Lin
- Department of Medicine Solna, Immunology and Allergy Unit, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Karin Loré
- Department of Medicine Solna, Immunology and Allergy Unit, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Olivia Marini
- Division of General Pathology, Department of Medicine, University of Verona, Verona, Italy
| | - Jeffrey W Pollard
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, EH16 4TJ, Edinburgh, UK
| | - Mikael Roussel
- Centre Hospitalier Universitaire, Pôle Biologie, INSERM, UMR U1236, Université Rennes 1, EFS Bretagne, Rennes, France
| | - Patrizia Scapini
- Division of General Pathology, Department of Medicine, University of Verona, Verona, Italy
| | - Viktor Umansky
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| | - Gosse J Adema
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 28, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|
44
|
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immunosuppressive cells of the myeloid lineage upregulated by mediators of inflammation, such as IL-2, granulocyte colony-stimulating factor, and S100A8/A9. These cells have been studied extensively by tumor biologists. Because of their robust immunosuppressive potential, MDSCs have stirred recent interest among transplant immunologists as well. MDSCs inhibit T-cell responses through, among other mechanisms, the activity of arginase-1 and inducible nitric oxide synthase, and the expansion of T regulatory cells. In the context of transplantation, MDSCs have been studied in several animal models, and to a lesser degree in humans. Here, we will review the immunosuppressive qualities of this important cell type and discuss the relevant studies of MDSCs in transplantation. It may be possible to exploit the immunosuppressive capacity of MDSCs for the benefit of transplant patients.
Collapse
|
45
|
Costa S, Bevilacqua D, Cassatella MA, Scapini P. Recent advances on the crosstalk between neutrophils and B or T lymphocytes. Immunology 2018; 156:23-32. [PMID: 30259972 DOI: 10.1111/imm.13005] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 09/12/2018] [Accepted: 09/21/2018] [Indexed: 12/21/2022] Open
Abstract
An increasing body of literature supports a role for neutrophils as players in the orchestration of adaptive immunity. During acute and chronic inflammatory conditions, neutrophils rapidly migrate not only to sites of inflammation, but also to draining lymph nodes and spleen, where they engage bidirectional interactions with B- and T-lymphocyte subsets. Accordingly, a relevant role of neutrophils in modulating B-cell responses under homeostatic conditions has recently emerged. Moreover, specialized immunoregulatory properties towards B or T cells acquired by distinct neutrophil populations, originating under pathological conditions, have been consistently described. In this article, we summarize the most recent data from human studies and murine models on the ability of neutrophils to modulate adaptive immune responses under physiological and pathological conditions and the mechanisms behind these processes.
Collapse
Affiliation(s)
- Sara Costa
- Department of Medicine, Section of General Pathology, School of Medicine, University of Verona, Verona, Italy
| | - Dalila Bevilacqua
- Department of Medicine, Section of General Pathology, School of Medicine, University of Verona, Verona, Italy
| | - Marco A Cassatella
- Department of Medicine, Section of General Pathology, School of Medicine, University of Verona, Verona, Italy
| | - Patrizia Scapini
- Department of Medicine, Section of General Pathology, School of Medicine, University of Verona, Verona, Italy
| |
Collapse
|
46
|
Myeloid-Derived Suppressor Cells and Pulmonary Hypertension. Int J Mol Sci 2018; 19:ijms19082277. [PMID: 30081463 PMCID: PMC6121540 DOI: 10.3390/ijms19082277] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 07/31/2018] [Accepted: 08/01/2018] [Indexed: 01/04/2023] Open
Abstract
Myeloid–derived suppressor cells (MDSCs) comprised a heterogeneous subset of bone marrow–derived myeloid cells, best studied in cancer research, that are increasingly implicated in the pathogenesis of pulmonary vascular remodeling and the development of pulmonary hypertension. Stem cell transplantation represents one extreme interventional strategy for ablating the myeloid compartment but poses a number of translational challenges. There remains an outstanding need for additional therapeutic targets to impact MDSC function, including the potential to alter interactions with innate and adaptive immune subsets, or alternatively, alter trafficking receptors, metabolic pathways, and transcription factor signaling with readily available and safe drugs. In this review, we summarize the current literature on the role of myeloid cells in the development of pulmonary hypertension, first in pulmonary circulation changes associated with myelodysplastic syndromes, and then by examining intrinsic myeloid cell changes that contribute to disease progression in pulmonary hypertension. We then outline several tractable targets and pathways relevant to pulmonary hypertension via MDSC regulation. Identifying these MDSC-regulated effectors is part of an ongoing effort to impact the field of pulmonary hypertension research through identification of myeloid compartment-specific therapeutic applications in the treatment of pulmonary vasculopathies.
Collapse
|
47
|
Lim JY, Ryu DB, Park MY, Lee SE, Park G, Kim TG, Min CK. Ex Vivo Generated Human Cord Blood Myeloid-Derived Suppressor Cells Attenuate Murine Chronic Graft-versus-Host Diseases. Biol Blood Marrow Transplant 2018; 24:2381-2396. [PMID: 30006304 DOI: 10.1016/j.bbmt.2018.07.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 07/03/2018] [Indexed: 01/24/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature myeloid cells with anti-inflammatory activity, and expanded murine MDSCs are capable of attenuating preclinical acute graft-versus-host disease (aGVHD) severity. Two murine cGVHD models were used to evaluate the effectiveness of ex vivo cultured human cord blood (hCB) MDSCs in chronic GVHD (cGVHD). First, GVHD recipients surviving in a classic C57BL/6 into MHC-mismatched BALB/c aGVHD model developed cGVHD. Second, donor pretreatment with granulocyte colony-stimulating factor (G-CSF) induced cGVHD. hCB-MDSCs (1 × 106) were intravenously injected to determine their preventive effects (on days 5, 7, 10, and 21) or therapeutic effects (on days 21, 28, and 35). In the first model the onset of clinical cutaneous cGVHD was significantly delayed in preventive hCB-MDSCs-treated allogeneic recipients. Pathologic scoring of target organs confirmed these clinical results. Importantly, thymic tissues of GVHD mice treated with hCB-MDSCs were less severely damaged, showing higher numbers of double (CD4 and CD8) positive T cells with reduced expansion of donor-type CD4 and CD8 T cells. Moreover, late infusion of hCB-MDSCs controlled the severity of established cGVHD that had occurred in control recipients. In the second model, cGVHD induced by G-CSF-mobilized stem cell graft was associated with promotion of Th 17 and Th 2 differentiation. hCB-MDSCs attenuated clinical and pathologic cGVHD severity. Increased production of IL-17 and more infiltration of T cells and macrophages in cGVHD mice were markedly reduced after hCB-MDSCs treatment. Importantly, Foxp3+ regulatory T cells and IFN-γ-producing T cells were expanded, whereas IL-17- and IL-4-producing T cells were decreased in allogeneic recipients of hCB-MDSCs. Taken together, these results showed that hCB-MDSCs have preclinical capability of attenuating cGVHD by preserving thymus function and regulating Th 17 signaling, suggesting a possible therapeutic strategy for clinical application.
Collapse
Affiliation(s)
- Ji-Young Lim
- Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Da-Bin Ryu
- Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Mi-Young Park
- Catholic Hematopoietic Stem Cell Bank, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sung-Eun Lee
- Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Gyeongsin Park
- Department of Pathology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Tai-Gyu Kim
- Catholic Hematopoietic Stem Cell Bank, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Chang-Ki Min
- Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea.
| |
Collapse
|
48
|
Köstlin N, Schoetensack C, Schwarz J, Spring B, Marmé A, Goelz R, Brodbeck G, Poets CF, Gille C. Granulocytic Myeloid-Derived Suppressor Cells (GR-MDSC) in Breast Milk (BM); GR-MDSC Accumulate in Human BM and Modulate T-Cell and Monocyte Function. Front Immunol 2018; 9:1098. [PMID: 29868036 PMCID: PMC5966528 DOI: 10.3389/fimmu.2018.01098] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 05/02/2018] [Indexed: 12/12/2022] Open
Abstract
Nosocomial bacterial infections (NBI) and necrotizing enterocolitis (NEC) are among the main reasons for death in preterm infants. Both are often caused by bacteria coming from the infected infant’s gut and feeding with breast milk (BM) seems beneficial in their pathogenesis. However, mechanisms causing the protective effect of BM are only incompletely understood. Myeloid-derived suppressor cells (MDSC) are myeloid cells with suppressive activity on other immune cells, recently described to play a role in mediating maternal–fetal tolerance during pregnancy and immune adaptation in newborns. Until now, nothing is known about occurrence and function of MDSC in BM. We analyzed MDSC in BM and peripheral blood of breastfeeding mothers and found that granulocytic MDSC, but not monocytic MDSC, accumulate in BM, exhibit an activated phenotype and increased suppressive activity and modulate TLR-expression on monocytes. Furthermore, we found that the lactotrophic hormones prolactin and oxytocin do not induce MDSC from peripheral blood. This is the first study to describe MDSC with immune-modulatory properties in human BM. Our results point toward a role for MDSC in local immune modulation in the gut possibly protecting infants from NBI and NEC.
Collapse
Affiliation(s)
- Natascha Köstlin
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany
| | - Carolin Schoetensack
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany
| | - Julian Schwarz
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany
| | - Bärbel Spring
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany
| | - Alexander Marmé
- Private Practice in Gynecology and Obstetrics, Tuebingen, Germany
| | - Rangmar Goelz
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany
| | - Gerhard Brodbeck
- Department of Hematology, Tuebingen University Children's Hospital, Tuebingen, Germany
| | - Christian F Poets
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany
| | - Christian Gille
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany
| |
Collapse
|
49
|
Zhang T, Zhou J, Man GCW, Leung KT, Liang B, Xiao B, Ma X, Huang S, Huang H, Hegde VL, Zhong Y, Li Y, Kong GWS, Yiu AKW, Kwong J, Ng PC, Lessey BA, Nagarkatti PS, Nagarkatti M, Wang CC. MDSCs drive the process of endometriosis by enhancing angiogenesis and are a new potential therapeutic target. Eur J Immunol 2018; 48:1059-1073. [PMID: 29460338 DOI: 10.1002/eji.201747417] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/20/2018] [Accepted: 02/14/2018] [Indexed: 12/21/2022]
Abstract
Endometriosis affects women of reproductive age via unclear immunological mechanism(s). Myeloid-derived suppressor cells (MDSCs) are a heterogeneous group of myeloid cells with potent immunosuppressive and angiogenic properties. Here, we found MDSCs significantly increased in the peripheral blood of patients with endometriosis and in the peritoneal cavity of a mouse model of surgically induced endometriosis. Majority of MDSCs were granulocytic, produced ROS, and arginase, and suppressed T-cell proliferation. Depletion of MDSCs by antiGr-1 antibody dramatically suppressed development of endometrial lesions in mice. The chemokines CXCL1, 2, and 5 were expressed at sites of lesion while MDSCs expressed CXCR-2. These CXC-chemokines promoted MDSC migration toward endometriotic implants both in vitro and in vivo. Also, CXCR2-deficient mice show significantly decreased MDSC induction, endometrial lesions, and angiogenesis. Importantly, adoptive transfer of MDSCs into CXCR2-KO mice restored endometriotic growth and angiogenesis. Together, this study demonstrates that MDSCs play a role in the pathogenesis of endometriosis and identifies a novel CXC-chemokine and receptor for the recruitment of MDSCs, thereby providing a potential target for endometriosis treatment.
Collapse
Affiliation(s)
- Tao Zhang
- Department of Obstetrics and Gynaecology, the Chinese University of Hong Kong, Hong Kong
| | - Juhua Zhou
- Institute for Tumor Immunology, School of Life Sciences, Ludong University, Yantai, Shandong, China
| | - Gene Chi Wai Man
- Department of Obstetrics and Gynaecology, the Chinese University of Hong Kong, Hong Kong
| | - Kam Tong Leung
- Department of Paediatrics, the Chinese University of Hong Kong, Hong Kong
| | - Bo Liang
- Department of Obstetrics and Gynaecology, the Chinese University of Hong Kong, Hong Kong
| | - Bo Xiao
- Institute for Tumor Immunology, School of Life Sciences, Ludong University, Yantai, Shandong, China
| | - Xinting Ma
- Institute for Tumor Immunology, School of Life Sciences, Ludong University, Yantai, Shandong, China
| | - Shaoyan Huang
- Department of Oncology, Yantai Mountain Hospital, Yantai, Shandong, China
| | | | - Venkatesh L Hegde
- Department of Obstetrics and Gynecology, Greenville Health Systems, Greenville, South Carolina, SC, USA
| | - Yin Zhong
- Department of Obstetrics and Gynecology, Greenville Health Systems, Greenville, South Carolina, SC, USA
| | - Yanmin Li
- Institute for Tumor Immunology, School of Life Sciences, Ludong University, Yantai, Shandong, China
| | - Grace Wing Shan Kong
- Department of Obstetrics and Gynaecology, the Chinese University of Hong Kong, Hong Kong
| | - Alice Ka Wah Yiu
- Department of Obstetrics and Gynaecology, the Chinese University of Hong Kong, Hong Kong
| | - Joseph Kwong
- Department of Obstetrics and Gynaecology, the Chinese University of Hong Kong, Hong Kong
| | - Pak Cheung Ng
- Department of Paediatrics, the Chinese University of Hong Kong, Hong Kong
| | - Bruce A Lessey
- Department of Obstetrics and Gynecology, Greenville Health Systems, Greenville, South Carolina, SC, USA
| | - Prakash S Nagarkatti
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC, USA
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC, USA
| | - Chi Chiu Wang
- Department of Obstetrics and Gynaecology, the Chinese University of Hong Kong, Hong Kong.,Li Ka Shing Institute of Health Sciences, the Chinese University of Hong Kong, Hong Kong.,School of Biomedical Sciences, the Chinese University of Hong Kong, Hong Kong
| |
Collapse
|
50
|
Myeloid-derived suppressor cells coming of age. Nat Immunol 2018; 19:108-119. [PMID: 29348500 DOI: 10.1038/s41590-017-0022-x] [Citation(s) in RCA: 1334] [Impact Index Per Article: 190.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 11/07/2017] [Indexed: 02/07/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of cells generated during a large array of pathologic conditions ranging from cancer to obesity. These cells represent a pathologic state of activation of monocytes and relatively immature neutrophils. MDSCs are characterized by a distinct set of genomic and biochemical features, and can, on the basis of recent findings, be distinguished by specific surface molecules. The salient feature of these cells is their ability to inhibit T cell function and thus contribute to the pathogenesis of various diseases. In this Review, we discuss the origin and nature of these cells; their distinctive features; and their biological roles in cancer, infectious diseases, autoimmunity, obesity and pregnancy.
Collapse
|