1
|
Flores-Campos R, García-Domínguez DJ, Hontecillas-Prieto L, Jiménez-Cortegana C, de la Cruz-Merino L, Sánchez-Margalet V. Flow cytometry analysis of myeloid derived suppressor cells using 6 color labeling. Methods Cell Biol 2024; 190:1-10. [PMID: 39515873 DOI: 10.1016/bs.mcb.2024.08.006] [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] [Indexed: 11/16/2024]
Abstract
Myeloid-derived suppressor cells (MDSCs) encompass a diverse population of immature myeloid cells categorized into granulocytic and monocytic groups. These cells exert immune-suppressive functions within the tumor microenvironment, primarily influenced by cytokines and tumor-associated factors. Research has consistently linked elevated MDSC levels to unfavorable cancer prognosis and poor responses to immunotherapies. Here, we detail the materials, equipment, and methods involved in MDSC analysis in human peripheral blood by flow cytometry, emphasizing the importance of selecting appropriate antibody clones and fluorochromes for precise cell population discrimination. The gating strategy is described, with particular attention to the challenges associated with defining conjugated antibody labeling positive and negative populations.
Collapse
Affiliation(s)
- Rocío Flores-Campos
- Clinical Biochemistry Service, Hospital Universitario Virgen Macarena, University of Seville, Seville, Spain; Department of Clinical Oncology, Hospital Universitario Virgen Macarena, University of Seville, Seville, Spain
| | - Daniel J García-Domínguez
- Department of Medical Biochemistry and Molecular Biology and Immunology, Medical School, Virgen Macarena University Hospital, University of Seville, Seville, Spain; Institute of Biomedicine of Seville (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain
| | - Lourdes Hontecillas-Prieto
- Clinical Biochemistry Service, Hospital Universitario Virgen Macarena, University of Seville, Seville, Spain; Department of Medical Biochemistry and Molecular Biology and Immunology, Medical School, Virgen Macarena University Hospital, University of Seville, Seville, Spain; Institute of Biomedicine of Seville (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain
| | - Carlos Jiménez-Cortegana
- Clinical Biochemistry Service, Hospital Universitario Virgen Macarena, University of Seville, Seville, Spain; Department of Medical Biochemistry and Molecular Biology and Immunology, Medical School, Virgen Macarena University Hospital, University of Seville, Seville, Spain
| | - Luis de la Cruz-Merino
- Department of Clinical Oncology, Hospital Universitario Virgen Macarena, University of Seville, Seville, Spain; Institute of Biomedicine of Seville (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain; Department of Medicine, University of Seville, Seville, Spain.
| | - Víctor Sánchez-Margalet
- Clinical Biochemistry Service, Hospital Universitario Virgen Macarena, University of Seville, Seville, Spain; Department of Medical Biochemistry and Molecular Biology and Immunology, Medical School, Virgen Macarena University Hospital, University of Seville, Seville, Spain; Institute of Biomedicine of Seville (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain.
| |
Collapse
|
2
|
Sarfi S, Azaryan E, Naseri M. Immune System of Dental Pulp in Inflamed and Normal Tissue. DNA Cell Biol 2024; 43:369-386. [PMID: 38959180 DOI: 10.1089/dna.2024.0044] [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] [Indexed: 07/05/2024] Open
Abstract
Teeth are vulnerable to structural compromise, primarily attributed to carious lesions, in which microorganisms originating from the oral cavity deteriorate the mineralized structures of enamel and dentin, subsequently infiltrating the underlying soft connective tissue, known as the dental pulp. Nonetheless, dental pulp possesses the necessary capabilities to detect and defend against bacteria and their by-products, using a variety of intricate defense mechanisms. The pulp houses specialized cells known as odontoblasts, which encounter harmful substances produced by oral bacteria. These cells identify pathogens at an early stage and commence the immune system response. As bacteria approach the pulp, various cell types within the pulp, such as different immune cells, stem cells, fibroblasts, as well as neuronal and vascular networks, contribute a range of defense mechanisms. Therefore, the immune system is present in the healthy pulp to restrain the initial spread of pathogens, and then in the inflamed pulp, it prepares the conditions for necrosis or regeneration, so inflammatory response mechanisms play a critical role in maintaining tissue homeostasis. This review aims to consolidate the existing literature on the immune system in dental pulp, encompassing current knowledge on this topic that explains the diverse mechanisms of recognition and defense against pathogens exhibited by dental pulp cells, elucidates the mechanisms of innate and adaptive immunity in inflamed pulp, and highlights the difference between inflamed and normal pulp tissue.
Collapse
Affiliation(s)
- Sepideh Sarfi
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran
- Department of Immunology, School of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Ehsaneh Azaryan
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohsen Naseri
- Cellular, and Molecular Research Center, Department of Molecular Medicine, Birjand University of Medical Sciences, Birjand, Iran
| |
Collapse
|
3
|
Surendar J, Hackenberg RK, Schmitt-Sánchez F, Ossendorff R, Welle K, Stoffel-Wagner B, Sage PT, Burger C, Wirtz DC, Strauss AC, Schildberg FA. Osteomyelitis is associated with increased anti-inflammatory response and immune exhaustion. Front Immunol 2024; 15:1396592. [PMID: 38736874 PMCID: PMC11082283 DOI: 10.3389/fimmu.2024.1396592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 04/12/2024] [Indexed: 05/14/2024] Open
Abstract
Introduction Osteomyelitis (OMS) is a bone infection causing bone pain and severe complications. A balanced immune response is critical to eradicate infection without harming the host, yet pathogens manipulate immunity to establish a chronic infection. Understanding OMS-driven inflammation is essential for disease management, but comprehensive data on immune profiles and immune cell activation during OMS are lacking. Methods Using high-dimensional flow cytometry, we investigated the detailed innate and adaptive systemic immune cell populations in OMS and age- and sex-matched controls. Results Our study revealed that OMS is associated with increased levels of immune regulatory cells, namely T regulatory cells, B regulatory cells, and T follicular regulatory cells. In addition, the expression of immune activation markers HLA-DR and CD86 was decreased in OMS, while the expression of immune exhaustion markers TIM-3, PD-1, PD-L1, and VISTA was increased. Members of the T follicular helper (Tfh) cell family as well as classical and typical memory B cells were significantly increased in OMS individuals. We also found a strong correlation between memory B cells and Tfh cells. Discussion We conclude that OMS skews the host immune system towards the immunomodulatory arm and that the Tfh memory B cell axis is evident in OMS. Therefore, immune-directed therapies may be a promising alternative for eradication and recurrence of infection in OMS, particularly in individuals and areas where antibiotic resistance is a major concern.
Collapse
Affiliation(s)
- Jayagopi Surendar
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Roslind K. Hackenberg
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
| | - Fabio Schmitt-Sánchez
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Robert Ossendorff
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Kristian Welle
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Birgit Stoffel-Wagner
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Peter T. Sage
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Christof Burger
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Dieter C. Wirtz
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Andreas C. Strauss
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Frank A. Schildberg
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
4
|
Bline KE, Wilt AL, Alexander RN, Andrews AN, Mertz SE, Ye F, Steele LM, Wolfe AL, Mejias A, Ramilo O. Myeloid-derived suppressor cells and T cell populations in children with Multisystem Inflammatory Syndrome. Pediatr Res 2024; 95:1288-1294. [PMID: 38042945 DOI: 10.1038/s41390-023-02919-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 10/31/2023] [Accepted: 11/03/2023] [Indexed: 12/04/2023]
Abstract
BACKGROUND Multisystem inflammatory syndrome in children (MIS-C) represents a hyperinflammatory state that can result in multi-organ dysfunction and death. Myeloid-derived suppressor cells (MDSC) are an immunosuppressive cell population that expands under inflammatory conditions and suppresses T cell function. We hypothesized that MDSC would be increased in children with MIS-C and that MDSC expansion would be associated with T cell lymphopenia. METHODS We conducted a prospective, observational study. Initial blood samples were collected within 48 h of admission. Age-matched healthy controls underwent sampling once. MDSC and T cell populations were identified by flow cytometric methods. RESULTS We enrolled 22 children with MIS-C (12 ICU, 10 ward) and 21 healthy controls (HC). Children with MIS-C demonstrated significantly higher MDSC compared to HC, and MDSC expansion persisted for >3 weeks in the ICU group. Children with MIS-C admitted to the ICU demonstrated significantly lower absolute numbers of T cells and natural killer cells. There were no significant associations between MDSC and cardiac dysfunction, duration of hospitalization, or vasoactive inotrope score. CONCLUSIONS Our study suggests that children critically ill with MIS-C have expansion of MDSC and associated decreased T cell and NK cell populations. Our results did not demonstrate associations between MDSC and clinical outcomes. IMPACT Multisystem inflammatory syndrome in children (MIS-C) is a dysregulated immune response occurring several weeks after SARS-CoV-2 infection that can result in multi-organ dysfunction and death. Children severely ill with MIS-C demonstrated increased myeloid-derived suppressor cells and decreased absolute numbers of CD4+ and CD8 + T cells and NK cells compared to healthy controls. There was no significant association between MDSC numbers and clinical outcomes; including cardiac dysfunction, length of stay, or requirement of vasoactive support, in children with MIS-C.
Collapse
Affiliation(s)
- Katherine E Bline
- Center for Vaccines and Immunity, Nationwide Children's Hospital, Columbus, OH, USA.
- Division of Critical Care Medicine, Nationwide Children's Hospital, Columbus, OH, USA.
| | - Anna L Wilt
- Center for Vaccines and Immunity, Nationwide Children's Hospital, Columbus, OH, USA
| | - Robin N Alexander
- Biostatistics Resource at Nationwide Children's Hospital, Columbus, OH, USA
| | - Angel N Andrews
- Center for Vaccines and Immunity, Nationwide Children's Hospital, Columbus, OH, USA
| | - Sara E Mertz
- Center for Vaccines and Immunity, Nationwide Children's Hospital, Columbus, OH, USA
| | - Fang Ye
- Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA
| | - Lisa M Steele
- Division of Critical Care Medicine, Nationwide Children's Hospital, Columbus, OH, USA
| | - Amber L Wolfe
- Division of Critical Care Medicine, Nationwide Children's Hospital, Columbus, OH, USA
| | - Asuncion Mejias
- Department of Infectious Disease, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Octavio Ramilo
- Department of Infectious Disease, St. Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|
5
|
Wakita H, Lu Y, Li X, Kobayashi T, Hachiya T, Ide H, Horie S. Evaluating Leukocyte Telomere Length and Myeloid-Derived Suppressor Cells as Biomarkers for Prostate Cancer. Cancers (Basel) 2024; 16:1386. [PMID: 38611064 PMCID: PMC11011111 DOI: 10.3390/cancers16071386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 03/27/2024] [Accepted: 03/30/2024] [Indexed: 04/14/2024] Open
Abstract
BACKGROUND Leukocyte telomere length (LTL) and myeloid-derived suppressor cells (MDSC) are associated with aging and the development and progression of cancer. However, the exact nature of this relationship remains unclear. Our study aimed to investigate the potential of LTL and MDSC as diagnostic biomarkers for prostate cancer while also seeking to deepen our understanding of the relationship of these potential biomarkers to each other. METHODS Our study involved patients undergoing a prostate biopsy. We analyzed the relative LTL in genomic DNA obtained from peripheral blood leukocytes as well as the percentage of MDSC and their subtypes in peripheral blood mononuclear cells (PBMC). Our evaluation focused on examining the relationship between LTL and MDSC and pathological diagnoses as well as investigating the correlation between LTL and MDSC levels. RESULTS In our study of 102 participants, 56 were pathologically diagnosed with localized prostate cancer (cancer group), while 46 tested negative (control group). The cancer group exhibited significantly shorter LTL in comparison to the control group (p = 0.024). Additionally, the cancer group showed a tendency towards a higher percentage of monocytic MDSC (M-MDSC), although this difference did not reach statistical significance (p = 0.056). Our multivariate logistic regression analysis revealed that patients with shorter LTL and higher percentages of M-MDSC had a 2.98-fold (95% CI = 1.001-8.869, p = 0.049) and 3.03-fold (95% CI = 1.152-7.977, p = 0.025) increased risk of prostate cancer diagnosis, respectively. There was also a significant negative correlation between LTL and M-MDSC. (r = -0.347, p < 0.001). CONCLUSIONS Our research has established a correlation between LTL and MDSC in patients undergoing biopsy for prostate cancer. Notably, we observed that individuals with localized prostate cancer tend to have shorter LTL and a higher percentage of M-MDSC prior to their diagnosis. These findings suggest that LTL and M-MDSC could potentially serve as adjunctive biomarkers for the early diagnosis of prostate cancer.
Collapse
Affiliation(s)
- Haruhiko Wakita
- Department of Urology, Graduate School of Medicine, Juntendo University, Tokyo 113-8431, Japan; (H.W.); (Y.L.); (X.L.); (T.K.); (H.I.)
| | - Yan Lu
- Department of Urology, Graduate School of Medicine, Juntendo University, Tokyo 113-8431, Japan; (H.W.); (Y.L.); (X.L.); (T.K.); (H.I.)
| | - Xiaoxu Li
- Department of Urology, Graduate School of Medicine, Juntendo University, Tokyo 113-8431, Japan; (H.W.); (Y.L.); (X.L.); (T.K.); (H.I.)
| | - Takuro Kobayashi
- Department of Urology, Graduate School of Medicine, Juntendo University, Tokyo 113-8431, Japan; (H.W.); (Y.L.); (X.L.); (T.K.); (H.I.)
| | - Tsuyoshi Hachiya
- Department of Advanced Informatics for Genetic Disease, Graduate School of Medicine, Juntendo University, Tokyo 113-8431, Japan;
| | - Hisamitsu Ide
- Department of Urology, Graduate School of Medicine, Juntendo University, Tokyo 113-8431, Japan; (H.W.); (Y.L.); (X.L.); (T.K.); (H.I.)
- Department of Digital Therapeutics, Graduate School of Medicine, Juntendo University, Tokyo 113-8431, Japan
| | - Shigeo Horie
- Department of Urology, Graduate School of Medicine, Juntendo University, Tokyo 113-8431, Japan; (H.W.); (Y.L.); (X.L.); (T.K.); (H.I.)
- Department of Advanced Informatics for Genetic Disease, Graduate School of Medicine, Juntendo University, Tokyo 113-8431, Japan;
- Department of Digital Therapeutics, Graduate School of Medicine, Juntendo University, Tokyo 113-8431, Japan
| |
Collapse
|
6
|
Wang H, Yang J, Li X, Zhao H. Current state of immune checkpoints therapy for glioblastoma. Heliyon 2024; 10:e24729. [PMID: 38298707 PMCID: PMC10828821 DOI: 10.1016/j.heliyon.2024.e24729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 01/12/2024] [Accepted: 01/12/2024] [Indexed: 02/02/2024] Open
Abstract
Glioblastoma (GBM), one of the most aggressive forms of brain cancer, has limited treatment options. Recent years have witnessed the remarkable success of checkpoint inhibitor immunotherapy across various cancer types. Against this backdrop, several clinical trials investigating checkpoint inhibitors for GBM are underway in multiple countries. Furthermore, the integration of immunotherapy with traditional treatment approaches is now emerging as a highly promising strategy. This review summarizes the latest advancements in checkpoint inhibitor immunotherapy for GBM treatment. We provide a concise yet comprehensive overview of current GBM immunotherapy options. Additionally, this review underscores combination strategies and potential biomarkers for predicting response and resistance in GBM immunotherapies.
Collapse
Affiliation(s)
- He Wang
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, Shandong, 266005, China
| | - Jing Yang
- Department of Emergency Surgery, the Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, Shandong, 266005, China
| | - Xiangjun Li
- School of medicine, Department of Breast surgery, the Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shandong, 266000, China
| | - Hai Zhao
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, Shandong, 266005, China
| |
Collapse
|
7
|
Liu T, Rosek A, Gonzalez De Los Santos F, Phan SH. Detection of myeloid-derived suppressor cells by flow cytometry. Methods Cell Biol 2023; 184:1-15. [PMID: 38555150 DOI: 10.1016/bs.mcb.2023.06.006] [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] [Indexed: 04/02/2024]
Abstract
Recently discovered heterogeneous myeloid-derived suppressor cells (MDSCs) are some of the most discussed immunosuppressive cells in contemporary immunology, especially in the tumor microenvironment, and are defined primarily by their T cell immunosuppressive function. The importance of these cells extend to other chronic pathological conditions as well, including chronic infection, inflammation, and tissue remodeling. In many of these conditions, their accumulation/expansion correlates with disease progression, poor prognosis, and reduced survival, which highlights the potential of how these cells may be used in a clinical setting as both prognostic factor and therapeutic target. In healthy individuals, these cells are usually not present in the circulation. Therefore, monitoring this cell population is of potential clinical significance, and utility in basic research. However, these cells have a complex phenotype without one single marker of sufficient specificity for their identification. Flow cytometry is a powerful tool allowing multi-parameter analysis of heterogeneous cell populations, which makes it ideally suitable for the complex phenotypic analysis essential for identification and enumeration of circulating MDSCs. This approach has the potential to provide a novel clinically useful tool for assessment of prognosis and treatment outcomes. The protocol in this chapter describes a flow cytometric analysis to identify and quantify MDSCs from human or mouse whole blood leukocytes and peripheral blood mononuclear cells, as well as a single cell suspension from solid tissue, by using multicolor fluorescence-conjugated antibodies against their surface markers.
Collapse
Affiliation(s)
- Tianju Liu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, United States.
| | - Alyssa Rosek
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, United States
| | | | - Sem H Phan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, United States.
| |
Collapse
|
8
|
Tamadaho RSE, Osei-Mensah J, Arndts K, Debrah LB, Debrah AY, Layland LE, Hoerauf A, Pfarr K, Ritter M. Reduced Type 2 Innate Lymphocyte Cell Frequencies in Patent Wuchereria bancrofti-Infected Individuals. Pathogens 2023; 12:pathogens12050665. [PMID: 37242335 DOI: 10.3390/pathogens12050665] [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: 02/06/2023] [Revised: 04/06/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
Approximately 51 million individuals suffer from lymphatic filariasis (LF) caused mainly by the filarial worm Wuchereria bancrofti. Mass drug administration (MDA) programs led to a significant reduction in the number of infected individuals, but the consequences of the treatment and clearance of infection in regard to host immunity remain uncertain. Thus, this study investigates the composition of myeloid-derived suppressor cells (MDSCs), macrophage subsets and innate lymphoid cells (ILCs), in patent (circulating filarial antigen (CFA)+ microfilariae (MF)+) and latent (CFA+MF-) W. bancrofti-infected individuals, previously W. bancrofti-infected (PI) individuals cured of the infection due to MDA, uninfected controls (endemic normal (EN)) and individuals who suffer from lymphoedema (LE) from the Western Region of Ghana. Frequencies of ILC2 were significantly reduced in W. bancrofti-infected individuals, while the frequencies of MDSCs, M2 macrophages, ILC1 and ILC3 were comparable between the cohorts. Importantly, clearance of infection due to MDA restored the ILC2 frequencies, suggesting that ILC2 subsets might migrate to the site of infection within the lymphatic tissue. In general, the immune cell composition in individuals who cured the infection were comparable to the uninfected individuals, showing that filarial-driven changes of the immune responses require an active infection and are not maintained upon the clearance of the infection.
Collapse
Affiliation(s)
- Ruth S E Tamadaho
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn (UKB), 53127 Bonn, Germany
| | - Jubin Osei-Mensah
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), UPO, PMB, Kumasi 00233, Ghana
- Department of Pathobiology, School of Veterinary Medicine, Kwame Nkrumah University of Science and Technology, UPO, PMB, Kumasi 00233, Ghana
| | - Kathrin Arndts
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn (UKB), 53127 Bonn, Germany
| | - Linda Batsa Debrah
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), UPO, PMB, Kumasi 00233, Ghana
- Department of Clinical Microbiology, School of Medicine and Dentistry, Kwame Nkrumah University of Science and Technology, UPO, PMB, Kumasi 00233, Ghana
| | - Alexander Y Debrah
- Faculty of Allied Health Sciences, Kwame Nkrumah University of Science and Technology, UPO, PMB, Kumasi 00233, Ghana
| | - Laura E Layland
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn (UKB), 53127 Bonn, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, 53127 Bonn, Germany
| | - Achim Hoerauf
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn (UKB), 53127 Bonn, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, 53127 Bonn, Germany
- German-West African Centre for Global Health and Pandemic Prevention (G-WAC), Partner Site Bonn, 53127 Bonn, Germany
| | - Kenneth Pfarr
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn (UKB), 53127 Bonn, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, 53127 Bonn, Germany
| | - Manuel Ritter
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn (UKB), 53127 Bonn, Germany
| |
Collapse
|
9
|
Role of myeloid-derived suppressor cells in tumor recurrence. Cancer Metastasis Rev 2023; 42:113-142. [PMID: 36640224 PMCID: PMC9840433 DOI: 10.1007/s10555-023-10079-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 01/09/2023] [Indexed: 01/15/2023]
Abstract
The establishment of primary tumor cells in distant organs, termed metastasis, is the principal cause of cancer mortality and is a crucial therapeutic target in oncology. Thus, it is critical to establish a better understanding of metastatic progression for the future development of improved therapeutic approaches. Indeed, such development requires insight into the timing of tumor cell dissemination and seeding of distant organs resulting in occult lesions. Following dissemination of tumor cells from the primary tumor, they can reside in niches in distant organs for years or decades, following which they can emerge as an overt metastasis. This timeline of metastatic dormancy is regulated by interactions between the tumor, its microenvironment, angiogenesis, and tumor antigen-specific T-cell responses. An improved understanding of the mechanisms and interactions responsible for immune evasion and tumor cell release from dormancy would help identify and aid in the development of novel targeted therapeutics. One such mediator of dormancy is myeloid derived suppressor cells (MDSC), whose number in the peripheral blood (PB) or infiltrating tumors has been associated with cancer stage, grade, patient survival, and metastasis in a broad range of tumor pathologies. Thus, extensive studies have revealed a role for MDSCs in tumor escape from adoptive and innate immune responses, facilitating tumor progression and metastasis; however, few studies have considered their role in dormancy. We have posited that MDSCs may regulate disseminated tumor cells resulting in resurgence of senescent tumor cells. In this review, we discuss clinical studies that address mechanisms of tumor recurrence including from dormancy, the role of MDSCs in their escape from dormancy during recurrence, the development of occult metastases, and the potential for MDSC inhibition as an approach to prolong the survival of patients with advanced malignancies. We stress that assessing the impact of therapies on MDSCs versus other cellular targets is challenging within the multimodality interventions required clinically.
Collapse
|
10
|
Shen M, Fan X, Shen Y, Wang X, Wu R, Wang Y, Huang C, Zhao S, Zheng Y, Men R, Luo X, Yang L. Myeloid-derived suppressor cells ameliorate liver mitochondrial damage to protect against autoimmune hepatitis by releasing small extracellular vesicles. Int Immunopharmacol 2023; 114:109540. [PMID: 36516541 DOI: 10.1016/j.intimp.2022.109540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 11/22/2022] [Accepted: 12/01/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Autoimmune hepatitis (AIH) is an inflammatory liver disease that is associated with impaired self-tolerance. Myeloid-derived supprfessor cells (MDSCs) have been considered to exert counterregulatory effects on AIH. However, the specific mechanism underlying these effects is unclear. Herein, we investigated the efficacy and safety of MDSCs in protecting against AIH and explored the underlying mechanism. METHODS Circulating and liver MDSC expression levels in 71 AIH patients and 47 healthy control (HC) individuals were detected by flow cytometry and immunohistochemistry. The adoptive transfer of induced bone marrow-derived MDSCs (BM MDSCs) to AIH mice was used to explore the function of MDSCs. Hepatic injury and mitochondrial damage were evaluated by transaminase levels, histopathology, immunohistochemistry, transmission electron microscopy and western blotting. MDSCs were pretreated with the small extracellular vesicle (sEV) generation inhibitor GW4869 to explore the mechanism. Importantly, sEVs derived from MDSCs and MDSCs-GW4869 were injected into model mice to monitor mitochondrial function and biogenesis. RESULTS Circulating and liver MDSCs were expanded in AIH patients and mouse model. Furthermore, the follow-up data of AIH patients showed that immunosuppressive therapy further promoted the expansion of MDSCs. More importantly, the adoptive transfer of BM MDSCs to AIH mice effectively ameliorated liver injury and regulated the imbalance of the immune microenvironment. Additionally, BM MDSCs reduced liver mitochondrial damage and improved mitochondrial biogenesis. Mechanistically, sEVs derived from BM MDSCs showed the same biological effects as cells, and blocking sEV production weakened the function of BM MDSCs. Finally, multiple long-term administrations of BM MDSCs were proven to be safe in general. CONCLUSION In conclusion, MDSCs ameliorate liver mitochondrial damage to protect against autoimmune hepatitis by releasing small extracellular vesicles.
Collapse
Affiliation(s)
- Mengyi Shen
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-related Molecular Network, Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaoli Fan
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-related Molecular Network, Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yi Shen
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-related Molecular Network, Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaoze Wang
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-related Molecular Network, Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ruiqi Wu
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-related Molecular Network, Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yi Wang
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-related Molecular Network, Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chen Huang
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-related Molecular Network, Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Shenglan Zhao
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-related Molecular Network, Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yanyi Zheng
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-related Molecular Network, Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ruoting Men
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-related Molecular Network, Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xuefeng Luo
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-related Molecular Network, Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Li Yang
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-related Molecular Network, Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
11
|
Immunophenotype and function of circulating myeloid derived suppressor cells in COVID-19 patients. Sci Rep 2022; 12:22570. [PMID: 36581679 PMCID: PMC9799710 DOI: 10.1038/s41598-022-26943-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 12/22/2022] [Indexed: 12/30/2022] Open
Abstract
The pathogenesis of coronavirus disease 2019 (COVID-19) is not fully elucidated. COVID-19 is due to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) which causes severe illness and death in some people by causing immune dysregulation and blood T cell depletion. Increased numbers of myeloid-derived suppressor cells (MDSCs) play a diverse role in the pathogenesis of many infections and cancers but their function in COVID-19 remains unclear. To evaluate the function of MDSCs in relation with the severity of COVID-19. 26 PCR-confirmed COVID-19 patients including 12 moderate and 14 severe patients along with 11 healthy age- and sex-matched controls were enrolled. 10 ml whole blood was harvested for cell isolation, immunophenotyping and stimulation. The immunophenotype of MDSCs by flow cytometry and T cells proliferation in the presence of MDSCs was evaluated. Serum TGF-β was assessed by ELISA. High percentages of M-MDSCs in males and of P-MDSCs in female patients were found in severe and moderate affected patients. Isolated MDSCs of COVID-19 patients suppressed the proliferation and intracellular levels of IFN-γ in T cells despite significant suppression of T regulatory cells but up-regulation of precursor regulatory T cells. Serum analysis shows increased levels of TGF-β in severe patients compared to moderate and control subjects (HC) (P = 0.003, P < 0.0001, respectively). The frequency of MDSCs in blood shows higher frequency among both moderate and severe patients and may be considered as a predictive factor for disease severity. MDSCs may suppress T cell proliferation by releasing TGF-β.
Collapse
|
12
|
Novel Identification of Myeloid-Derived Suppressor Cells in Children With Septic Shock. Pediatr Crit Care Med 2022; 23:e555-e563. [PMID: 36094492 DOI: 10.1097/pcc.0000000000003071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVES Immunoparalysis in children with septic shock is associated with increased risk of nosocomial infections and death. Myeloid-derived suppressor cells (MDSCs) potently suppress T cell function and may perpetuate immunoparalysis. Our goal was to test the hypothesis that children with septic shock would demonstrate increased proportions of MDSCs and impaired immune function compared with healthy controls. DESIGN Prospective observational study. SETTING Fifty-four bed PICU in a quaternary-care children's hospital. PATIENTS Eighteen children with septic shock and thirty age-matched healthy children. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Peripheral blood mononuclear cells (PBMCs) were isolated from whole blood and stained for cell surface markers to identify MDSCs by flow cytometric analysis, including granulocytic and monocytic subsets. Adaptive and innate immune function was measured by ex vivo stimulation of whole blood with phytohemagglutinin-induced interferon (IFN) γ production and lipopolysaccharide (LPS)-induced tumor necrosis factor (TNF)-α production, respectively. Prolonged organ dysfunction (OD) was defined as greater than 7 days. Children with septic shock had a higher percentage of circulating MDSCs, along with lower LPS-induced TNFα and phytohemagglutinin-induced IFNγ production capacities, compared with healthy controls. A cut-off of 25.2% MDSCs of total PBMCs in initial samples was optimal to discriminate children with septic shock who went on to have prolonged OD, area under the curve equal to 0.86. Children with prolonged OD also had decreased TNFα production capacity over time compared with those who recovered more quickly ( p = 0.02). CONCLUSIONS This article is the first to describe increased MDSCs in children with septic shock, along with an association between early increase in MDSCs and adverse OD outcomes in this population. It remains unclear if MDSCs play a causative role in sepsis-induced immune suppression in children. Additional studies are warranted to establish MDSC as a potential therapeutic target.
Collapse
|
13
|
Bonavia AS, Samuelsen A, Luthy J, Halstead ES. Integrated machine learning approaches for flow cytometric quantification of myeloid-derived suppressor cells in acute sepsis. Front Immunol 2022; 13:1007016. [PMID: 36466851 PMCID: PMC9714638 DOI: 10.3389/fimmu.2022.1007016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 11/04/2022] [Indexed: 09/05/2024] Open
Abstract
Highly heterogeneous cell populations require multiple flow cytometric markers for appropriate phenotypic characterization. This exponentially increases the complexity of 2D scatter plot analyses and exacerbates human errors due to variations in manual gating of flow data. We describe a semi-automated workflow, based entirely on the Flowjo Graphical User Interface (GUI), that involves the stepwise integration of several, newly available machine learning tools for the analysis of myeloid-derived suppressor cells (MDSCs) in septic and non-septic critical illness. Supervised clustering of flow cytometric data showed correlation with, but significantly different numbers of, MDSCs as compared with the cell numbers obtained by manual gating. Neither quantification method predicted 30-day clinical outcomes in a cohort of 16 critically ill and septic patients and 5 critically ill and non-septic patients. Machine learning identified a significant decrease in the proportion of PMN-MDSC in critically ill and septic patients as compared with healthy controls. There was no difference between the proportion of these MDSCs in septic and non-septic critical illness.
Collapse
Affiliation(s)
- Anthony S. Bonavia
- Division of Critical Care Medicine, Department of Anesthesiology and Perioperative Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
- Department of Anesthesiology and Perioperative Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Abigail Samuelsen
- Division of Critical Care Medicine, Department of Anesthesiology and Perioperative Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Joshua Luthy
- Product Innovation Division, BD Life Sciences - FlowJo, Ashland, OR, United States
| | - E. Scott Halstead
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| |
Collapse
|
14
|
Lee EJ, Jung S, Park KH, Park SI. Flow cytometry-based immunophenotyping of myeloid-derived suppressor cells in human breast cancer patient blood samples. J Immunol Methods 2022; 510:113348. [PMID: 36058258 DOI: 10.1016/j.jim.2022.113348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/24/2022] [Accepted: 08/29/2022] [Indexed: 12/31/2022]
Abstract
Multi-color flow cytometry is the standard approach for immunophenotyping clinical samples. With the recent advances in cancer immunotherapy, myeloid-derived suppressor cells (MDSC), immature myeloid-lineage cells in cancer patient blood and the tumor microenvironment, are highlighted as an important immune cell population that correlates with prognosis and therapeutic efficacy. In contrast to their clear functions and existence, immunophenotyping of MDSC is not consistent among investigators due to surface antigens overlapping with many normal hematopoietic lineage cell populations. We performed a clinical study and analyzed more than 1000 breast cancer patients blood samples to quantitate MDSC during breast cancer progression. In this methodology manuscript, we described detailed procedures for study design, sample logistics and handling, staining and flow cytometric analysis. This protocol used a 7-color fluorochrome-conjugated antibody panel to analyze polymorphonuclear (PMN)- and monocytic (M)-MDSC subsets simultaneously. The interim analysis results of this study showed that both PMN and M-MDSC populations are increased in patients with bone metastasis compared with patients with visceral organ metastasis. In conclusion, this work provides a versatile, comprehensive, and practical protocol to measure MDSC in patient blood samples.
Collapse
Affiliation(s)
- Eun Jung Lee
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, Republic of Korea; The BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Seungpil Jung
- Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Anam Hospital, Seoul, Republic of Korea
| | - Kyong Hwa Park
- Division of Oncology and Hematology, Department of Internal Medicine, Korea University Anam Hospital, Seoul, Republic of Korea.
| | - Serk In Park
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, Republic of Korea; The BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea; Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
15
|
Shigehiro T, Ueno M, Kijihira M, Takahashi R, Umemura C, Taha EA, Kurosaka C, Asayama M, Murakami H, Satoh A, Nakamura Y, Futami J, Masuda J. Immune State Conversion of the Mesenteric Lymph Node in a Mouse Breast Cancer Model. Int J Mol Sci 2022; 23:ijms231911035. [PMID: 36232335 PMCID: PMC9570492 DOI: 10.3390/ijms231911035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/17/2022] [Accepted: 09/18/2022] [Indexed: 11/16/2022] Open
Abstract
Secondary lymphoid tissues, such as the spleen and lymph nodes (LNs), contribute to breast cancer development and metastasis in both anti- and pro-tumoral directions. Although secondary lymphoid tissues have been extensively studied, very little is known about the immune conversion in mesenteric LNs (mLNs) during breast cancer development. Here, we demonstrate inflammatory immune conversion of mLNs in a metastatic 4T1 breast cancer model. Splenic T cells were significantly decreased and continuously suppressed IFN-γ production during tumor development, while myeloid-derived suppressor cells (MDSCs) were dramatically enriched. However, T cell numbers in the mLN did not decrease, and the MDSCs only moderately increased. T cells in the mLN exhibited conversion from a pro-inflammatory state with high IFN-γ expression to an anti-inflammatory state with high expression of IL-4 and IL-10 in early- to late-stages of breast cancer development. Interestingly, increased migration of CD103+CD11b+ dendritic cells (DCs) into the mLN, along with increased (1→3)-β-D-glucan levels in serum, was observed even in late-stage breast cancer. This suggests that CD103+CD11b+ DCs could prime cancer-reactive T cells. Together, the data indicate that the mLN is an important lymphoid tissue contributing to breast cancer development.
Collapse
Affiliation(s)
- Tsukasa Shigehiro
- Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba 278-0022, Japan
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
- Correspondence: (T.S.); (J.M.); Tel.: +81-47-121-4060 (T.S.); +81-86-251-8003 (J.M.)
| | - Maho Ueno
- Department of Applied Chemistry and Biotechnology, Faculty of Engineering, Okayama University, Okayama 700-8530, Japan
| | - Mayumi Kijihira
- Department of Applied Chemistry and Biotechnology, Faculty of Engineering, Okayama University, Okayama 700-8530, Japan
| | - Ryotaro Takahashi
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan
| | - Chiho Umemura
- Division of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-8530, Japan
| | - Eman A. Taha
- Division of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-8530, Japan
| | - Chisaki Kurosaka
- Department of Applied Chemistry and Biotechnology, Faculty of Engineering, Okayama University, Okayama 700-8530, Japan
| | - Megumi Asayama
- Department of Applied Chemistry and Biotechnology, Faculty of Engineering, Okayama University, Okayama 700-8530, Japan
| | - Hiroshi Murakami
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan
- Division of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-8530, Japan
| | - Ayano Satoh
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan
- Division of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-8530, Japan
| | - Yoshimasa Nakamura
- Graduate School of Environmental and Life Science, Okayama University, Okayama 700-8530, Japan
| | - Junichiro Futami
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan
- Division of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-8530, Japan
| | - Junko Masuda
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan
- Division of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-8530, Japan
- Department of Pharmacology, Tokyo Women’s Medical University, Shinjuku, Tokyo 162-8666, Japan
- Correspondence: (T.S.); (J.M.); Tel.: +81-47-121-4060 (T.S.); +81-86-251-8003 (J.M.)
| |
Collapse
|
16
|
van der Pan K, de Bruin-Versteeg S, Damasceno D, Hernández-Delgado A, van der Sluijs-Gelling AJ, van den Bossche WBL, de Laat IF, Díez P, Naber BAE, Diks AM, Berkowska MA, de Mooij B, Groenland RJ, de Bie FJ, Khatri I, Kassem S, de Jager AL, Louis A, Almeida J, van Gaans-van den Brink JAM, Barkoff AM, He Q, Ferwerda G, Versteegen P, Berbers GAM, Orfao A, van Dongen JJM, Teodosio C. Development of a standardized and validated flow cytometry approach for monitoring of innate myeloid immune cells in human blood. Front Immunol 2022; 13:935879. [PMID: 36189252 PMCID: PMC9519388 DOI: 10.3389/fimmu.2022.935879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 08/18/2022] [Indexed: 11/13/2022] Open
Abstract
Innate myeloid cell (IMC) populations form an essential part of innate immunity. Flow cytometric (FCM) monitoring of IMCs in peripheral blood (PB) has great clinical potential for disease monitoring due to their role in maintenance of tissue homeostasis and ability to sense micro-environmental changes, such as inflammatory processes and tissue damage. However, the lack of standardized and validated approaches has hampered broad clinical implementation. For accurate identification and separation of IMC populations, 62 antibodies against 44 different proteins were evaluated. In multiple rounds of EuroFlow-based design-testing-evaluation-redesign, finally 16 antibodies were selected for their non-redundancy and separation power. Accordingly, two antibody combinations were designed for fast, sensitive, and reproducible FCM monitoring of IMC populations in PB in clinical settings (11-color; 13 antibodies) and translational research (14-color; 16 antibodies). Performance of pre-analytical and analytical variables among different instruments, together with optimized post-analytical data analysis and reference values were assessed. Overall, 265 blood samples were used for design and validation of the antibody combinations and in vitro functional assays, as well as for assessing the impact of sample preparation procedures and conditions. The two (11- and 14-color) antibody combinations allowed for robust and sensitive detection of 19 and 23 IMC populations, respectively. Highly reproducible identification and enumeration of IMC populations was achieved, independently of anticoagulant, type of FCM instrument and center, particularly when database/software-guided automated (vs. manual “expert-based”) gating was used. Whereas no significant changes were observed in identification of IMC populations for up to 24h delayed sample processing, a significant impact was observed in their absolute counts after >12h delay. Therefore, accurate identification and quantitation of IMC populations requires sample processing on the same day. Significantly different counts were observed in PB for multiple IMC populations according to age and sex. Consequently, PB samples from 116 healthy donors (8-69 years) were used for collecting age and sex related reference values for all IMC populations. In summary, the two antibody combinations and FCM approach allow for rapid, standardized, automated and reproducible identification of 19 and 23 IMC populations in PB, suited for monitoring of innate immune responses in clinical and translational research settings.
Collapse
Affiliation(s)
- Kyra van der Pan
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Daniela Damasceno
- Translational and Clinical Research Program, Cancer Research Center (IBMCC; University of Salamanca - CSIC), Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca (Universidad de Salamanca, and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Alejandro Hernández-Delgado
- Translational and Clinical Research Program, Cancer Research Center (IBMCC; University of Salamanca - CSIC), Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca (Universidad de Salamanca, and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | | | - Wouter B. L. van den Bossche
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
- Department of Immunology, Department of Neurosurgery, Brain Tumor Center, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Inge F. de Laat
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Paula Díez
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Annieck M. Diks
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Bas de Mooij
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Rick J. Groenland
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Fenna J. de Bie
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Indu Khatri
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Sara Kassem
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Anniek L. de Jager
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Alesha Louis
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Julia Almeida
- Translational and Clinical Research Program, Cancer Research Center (IBMCC; University of Salamanca - CSIC), Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca (Universidad de Salamanca, and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | | | - Alex-Mikael Barkoff
- Institute of Biomedicine, Research Center for Infections and Immunity, University of Turku (UTU), Turku, Finland
| | - Qiushui He
- Institute of Biomedicine, Research Center for Infections and Immunity, University of Turku (UTU), Turku, Finland
| | - Gerben Ferwerda
- Section of Paediatric Infectious Diseases, Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
| | - Pauline Versteegen
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Guy A. M. Berbers
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Alberto Orfao
- Translational and Clinical Research Program, Cancer Research Center (IBMCC; University of Salamanca - CSIC), Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca (Universidad de Salamanca, and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Jacques J. M. van Dongen
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
- Translational and Clinical Research Program, Cancer Research Center (IBMCC; University of Salamanca - CSIC), Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca (Universidad de Salamanca, and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- *Correspondence: Jacques J. M. van Dongen,
| | - Cristina Teodosio
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
- Translational and Clinical Research Program, Cancer Research Center (IBMCC; University of Salamanca - CSIC), Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca (Universidad de Salamanca, and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| |
Collapse
|
17
|
Bizymi N, Matthaiou AM, Matheakakis A, Voulgari I, Aresti N, Zavitsanou K, Karasachinidis A, Mavroudi I, Pontikoglou C, Papadaki HA. New Perspectives on Myeloid-Derived Suppressor Cells and Their Emerging Role in Haematology. J Clin Med 2022; 11:jcm11185326. [PMID: 36142973 PMCID: PMC9504532 DOI: 10.3390/jcm11185326] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/05/2022] [Accepted: 09/08/2022] [Indexed: 12/03/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are immature cells of myeloid origin that have gained researchers’ attention, as they constitute promising biomarkers and targets for novel therapeutic strategies (i.e., blockage of development, differentiation, depletion, and deactivation) in several conditions, including neoplastic, autoimmune, infective, and inflammatory diseases, as well as pregnancy, obesity, and graft rejection. They are characterised in humans by the typical immunophenotype of CD11b+CD33+HLA-DR–/low and immune-modulating properties leading to decreased T-cell proliferation, induction of T-regulatory cells (T-regs), hindering of natural killer (NK) cell functionality, and macrophage M2-polarisation. The research in the field is challenging, as there are still difficulties in defining cell-surface markers and gating strategies that uniquely identify the different populations of MDSCs, and the currently available functional assays are highly demanding. There is evidence that MDSCs display altered frequency and/or functionality and could be targeted in immune-mediated and malignant haematologic diseases, although there is a large variability of techniques and results between different laboratories. This review presents the current literature concerning MDSCs in a clinical point of view in an attempt to trigger future investigation by serving as a guide to the clinical haematologist in order to apply them in the context of precision medicine as well as the researcher in the field of experimental haematology.
Collapse
Affiliation(s)
- Nikoleta Bizymi
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
- Laboratory of Molecular and Cellular Pneumonology, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
| | - Andreas M. Matthaiou
- Laboratory of Molecular and Cellular Pneumonology, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
- Respiratory Physiology Laboratory, Medical School, University of Cyprus, 2029 Nicosia, Cyprus
| | - Angelos Matheakakis
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
| | - Ioanna Voulgari
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
| | - Nikoletta Aresti
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
| | - Konstantina Zavitsanou
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
| | - Anastasios Karasachinidis
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
| | - Irene Mavroudi
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
| | - Charalampos Pontikoglou
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
| | - Helen A. Papadaki
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
- Correspondence: ; Tel.: +30-2810394637
| |
Collapse
|
18
|
Costa A, Thirant C, Kramdi A, Pierre-Eugène C, Louis-Brennetot C, Blanchard O, Surdez D, Gruel N, Lapouble E, Pierron G, Sitbon D, Brisse H, Gauthier A, Fréneaux P, Bohec M, Raynal V, Baulande S, Leclere R, Champenois G, Nicolas A, Meseure D, Bellini A, Marabelle A, Geoerger B, Mechta-Grigoriou F, Schleiermacher G, Menger L, Delattre O, Janoueix-Lerosey I. Single-cell transcriptomics reveals shared immunosuppressive landscapes of mouse and human neuroblastoma. J Immunother Cancer 2022; 10:jitc-2022-004807. [PMID: 36054452 PMCID: PMC9362821 DOI: 10.1136/jitc-2022-004807] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2022] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND High-risk neuroblastoma is a pediatric cancer with still a dismal prognosis, despite multimodal and intensive therapies. Tumor microenvironment represents a key component of the tumor ecosystem the complexity of which has to be accurately understood to define selective targeting opportunities, including immune-based therapies. METHODS We combined various approaches including single-cell transcriptomics to dissect the tumor microenvironment of both a transgenic mouse neuroblastoma model and a cohort of 10 biopsies from neuroblastoma patients, either at diagnosis or at relapse. Features of related cells were validated by multicolor flow cytometry and functional assays. RESULTS We show that the immune microenvironment of MYCN-driven mouse neuroblastoma is characterized by a low content of T cells, several phenotypes of macrophages and a population of cells expressing signatures of myeloid-derived suppressor cells (MDSCs) that are molecularly distinct from the various macrophage subsets. We document two cancer-associated fibroblasts (CAFs) subsets, one of which corresponding to CAF-S1, known to have immunosuppressive functions. Our data unravel a complex content in myeloid cells in patient tumors and further document a striking correspondence of the microenvironment populations between both mouse and human tumors. We show that mouse intratumor T cells exhibit increased expression of inhibitory receptors at the protein level. Consistently, T cells from patients are characterized by features of exhaustion, expressing inhibitory receptors and showing low expression of effector cytokines. We further functionally demonstrate that MDSCs isolated from mouse neuroblastoma have immunosuppressive properties, impairing the proliferation of T lymphocytes. CONCLUSIONS Our study demonstrates that neuroblastoma tumors have an immunocompromised microenvironment characterized by dysfunctional T cells and accumulation of immunosuppressive cells. Our work provides a new and precious data resource to better understand the neuroblastoma ecosystem and suggest novel therapeutic strategies, targeting both tumor cells and components of the microenvironment.
Collapse
Affiliation(s)
- Ana Costa
- Inserm U830, Equipe Labellisée LNCC, Diversity and Plasticity of Childhood Tumors Lab, PSL Research University, Institut Curie Research Centre, Paris, France.,SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France
| | - Cécile Thirant
- Inserm U830, Equipe Labellisée LNCC, Diversity and Plasticity of Childhood Tumors Lab, PSL Research University, Institut Curie Research Centre, Paris, France.,SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France
| | - Amira Kramdi
- Inserm U830, Equipe Labellisée LNCC, Diversity and Plasticity of Childhood Tumors Lab, PSL Research University, Institut Curie Research Centre, Paris, France.,SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France
| | - Cécile Pierre-Eugène
- Inserm U830, Equipe Labellisée LNCC, Diversity and Plasticity of Childhood Tumors Lab, PSL Research University, Institut Curie Research Centre, Paris, France.,SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France
| | - Caroline Louis-Brennetot
- Inserm U830, Equipe Labellisée LNCC, Diversity and Plasticity of Childhood Tumors Lab, PSL Research University, Institut Curie Research Centre, Paris, France.,SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France
| | - Orphée Blanchard
- Inserm U830, Equipe Labellisée LNCC, Diversity and Plasticity of Childhood Tumors Lab, PSL Research University, Institut Curie Research Centre, Paris, France.,SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France
| | - Didier Surdez
- Inserm U830, Equipe Labellisée LNCC, Diversity and Plasticity of Childhood Tumors Lab, PSL Research University, Institut Curie Research Centre, Paris, France.,SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France
| | - Nadege Gruel
- Inserm U830, Equipe Labellisée LNCC, Diversity and Plasticity of Childhood Tumors Lab, PSL Research University, Institut Curie Research Centre, Paris, France.,Department of Translational Research, Institut Curie, Paris, France
| | - Eve Lapouble
- Unité de Génétique Somatique, Institut Curie, Paris, France
| | - Gaëlle Pierron
- Unité de Génétique Somatique, Institut Curie, Paris, France
| | - Deborah Sitbon
- Unité de Génétique Somatique, Institut Curie, Paris, France
| | - Hervé Brisse
- Department of Imaging, PSL Research University, Institut Curie, Paris, France
| | | | - Paul Fréneaux
- Department of Biopathology, Institut Curie, Paris, France
| | - Mylène Bohec
- Genomics of Excellence (ICGex) Platform, Institut Curie, Paris, France
| | - Virginie Raynal
- Inserm U830, Equipe Labellisée LNCC, Diversity and Plasticity of Childhood Tumors Lab, PSL Research University, Institut Curie Research Centre, Paris, France.,Genomics of Excellence (ICGex) Platform, Institut Curie, Paris, France
| | - Sylvain Baulande
- Genomics of Excellence (ICGex) Platform, Institut Curie, Paris, France
| | - Renaud Leclere
- Department of Biopathology, Institut Curie, Paris, France
| | | | - Andre Nicolas
- Department of Biopathology, Institut Curie, Paris, France
| | - Didier Meseure
- Department of Biopathology, Institut Curie, Paris, France
| | - Angela Bellini
- SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France.,Department of Translational Research, Institut Curie, Paris, France.,Laboratory Recherche Translationnelle en Oncologie Pédiatrique (RTOP), Laboratoire "Gilles Thomas", Institut Curie, Paris, France
| | - Aurelien Marabelle
- Inserm U1015 & CIC1428, Université Paris Saclay, Gustave Roussy, Villejuif, France
| | - Birgit Geoerger
- Inserm U1015, Department of Pediatric and Adolescent Oncology, Université Paris-Saclay, Gustave Roussy, Villejuif, France
| | - Fatima Mechta-Grigoriou
- Inserm U830, Equipe labelisée LNCC, Stress and Cancer Laboratory, PSL Research University, Institut Curie Research Centre, Paris, France
| | - Gudrun Schleiermacher
- SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France.,Department of Translational Research, Institut Curie, Paris, France.,Laboratory Recherche Translationnelle en Oncologie Pédiatrique (RTOP), Laboratoire "Gilles Thomas", Institut Curie, Paris, France
| | - Laurie Menger
- Inserm U932, PSL Research University, Institut Curie, Paris, France
| | - Olivier Delattre
- Inserm U830, Equipe Labellisée LNCC, Diversity and Plasticity of Childhood Tumors Lab, PSL Research University, Institut Curie Research Centre, Paris, France.,SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France
| | - Isabelle Janoueix-Lerosey
- Inserm U830, Equipe Labellisée LNCC, Diversity and Plasticity of Childhood Tumors Lab, PSL Research University, Institut Curie Research Centre, Paris, France .,SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France
| |
Collapse
|
19
|
Immunosuppressive cells in cancer: mechanisms and potential therapeutic targets. J Hematol Oncol 2022; 15:61. [PMID: 35585567 PMCID: PMC9118588 DOI: 10.1186/s13045-022-01282-8] [Citation(s) in RCA: 186] [Impact Index Per Article: 93.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/03/2022] [Indexed: 02/08/2023] Open
Abstract
Immunotherapies like the adoptive transfer of gene-engineered T cells and immune checkpoint inhibitors are novel therapeutic modalities for advanced cancers. However, some patients are refractory or resistant to these therapies, and the mechanisms underlying tumor immune resistance have not been fully elucidated. Immunosuppressive cells such as myeloid-derived suppressive cells, tumor-associated macrophages, tumor-associated neutrophils, regulatory T cells (Tregs), and tumor-associated dendritic cells are critical factors correlated with immune resistance. In addition, cytokines and factors secreted by tumor cells or these immunosuppressive cells also mediate the tumor progression and immune escape of cancers. Thus, targeting these immunosuppressive cells and the related signals is the promising therapy to improve the efficacy of immunotherapies and reverse the immune resistance. However, even with certain success in preclinical studies or in some specific types of cancer, large perspectives are unknown for these immunosuppressive cells, and the related therapies have undesirable outcomes for clinical patients. In this review, we comprehensively summarized the phenotype, function, and potential therapeutic targets of these immunosuppressive cells in the tumor microenvironment.
Collapse
|
20
|
Schrijver IT, Karakike E, Théroude C, Baumgartner P, Harari A, Giamarellos-Bourboulis EJ, Calandra T, Roger T. High levels of monocytic myeloid-derived suppressor cells are associated with favorable outcome in patients with pneumonia and sepsis with multi-organ failure. Intensive Care Med Exp 2022; 10:5. [PMID: 35146585 PMCID: PMC8831012 DOI: 10.1186/s40635-022-00431-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 01/26/2022] [Indexed: 12/29/2022] Open
Abstract
Background Myeloid-derived suppressor cells (MDSCs) are immature myeloid cells with immunosuppressive functions sub-classified into monocytic and polymorphonuclear MDSCs (M-MDSCs and PMN-MDSCs). Clinical studies reported increased levels of MDSCs that were associated with poor outcome in sepsis patients. Since sepsis patients exhibit signs of inflammation and immunosuppression, MDSCs may provide benefit by dampening deleterious inflammation in some patients. To test this hypothesis, we measured MDSCs in critically ill sepsis patients with pneumonia and multi-organ dysfunctions and a high likelihood of death. Methods This was a prospective multicenter observational cohort study performed in eight ICUs in Athens and Thessaloniki, Greece, enrolling critically ill patients with pneumonia and sepsis with multi-organ dysfunctions. A flow cytometry approach using blood collected at study inclusion in tubes containing lyophilized antibodies combined to unsupervised clustering was developed to quantify M-MDSCs and PMN-MDSCs. Results Forty-eight patients were included, of whom 34 died within 90 days. At study inclusion, M-MDSCs and PMN-MDSCs were increased in sepsis patients when compared to healthy subjects (3.07% vs 0.96% and 22% vs 2.1% of leukocytes, respectively; p < 10–4). Increased PMN-MDSCs were associated with secondary infections (p = 0.024) and new sepsis episodes (p = 0.036). M-MDSCs were more abundant in survivors than in patients who died within 28 days (p = 0.028). Stratification of patients according to M-MDSC levels revealed that high levels of M-MDSC were associated with reduced 90-day mortality (high vs low M-MDSCs: 47% vs 84% mortality, p = 0.003, hazard ratio [HR] = 3.2, 95% CI 1.4–7.2). Combining high M-MDSC levels with low Acute Physiology and Chronic Health Evaluation (APACHE) II score improved patient stratification (M-MDSCshigh/APACHE IIlow vs M-MDSCslow/APACHE IIlow: 20% vs 80% 90-day mortality, p = 0.0096, HR = 7.2, 95% CI 1.6–32). In multivariate analyses high M-MDSCs remained correlated with improved survival in patients with low APACHE II score (p = 0.05, HR = 5.26, 95% CI 1.0–27.8). Conclusion This is the first study to associate high levels of M-MDSCs with improved survival in sepsis patients. Supplementary Information The online version contains supplementary material available at 10.1186/s40635-022-00431-0.
Collapse
Affiliation(s)
- Irene T Schrijver
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, CLED.04.407, Chemin des Boveresses 155, 1066, Epalinges, Switzerland
| | - Eleni Karakike
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Charlotte Théroude
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, CLED.04.407, Chemin des Boveresses 155, 1066, Epalinges, Switzerland
| | - Pétra Baumgartner
- Department of Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Alexandre Harari
- Department of Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | | | - Thierry Calandra
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, CLED.04.407, Chemin des Boveresses 155, 1066, Epalinges, Switzerland
| | - Thierry Roger
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, CLED.04.407, Chemin des Boveresses 155, 1066, Epalinges, Switzerland.
| |
Collapse
|
21
|
Soler DC, Kerstetter-Fogle A, Young AB, Rayman P, Finke JH, Debanne SM, Cooper KD, Barnholtz-Sloan J, Sloan AE, McCormick TS. Healthy myeloid-derived suppressor cells express the surface ectoenzyme Vanin-2 (VNN2). Mol Immunol 2022; 142:1-10. [PMID: 34953280 PMCID: PMC8800381 DOI: 10.1016/j.molimm.2021.12.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 11/18/2021] [Accepted: 12/09/2021] [Indexed: 02/03/2023]
Abstract
Study of human monocytic Myeloid-Derived Suppressor cells Mo-MDSC (CD14+ HLA-DRneg/low) has been hampered by the lack of positive cell-surface markers. In order to identify positive markers for Mo-MDSC, we performed microarray analysis comparing Mo-MDSC cells from healthy subjects versus CD14+ HLA-DRhigh monocytes. We have identified the surface ectoenzyme Vanin-2(VNN2) protein as a novel biomarker highly-enriched in healthy subjects Mo-MDSC. Indeed, healthy subjects Mo-MDSC cells expressed 68 % VNN2, whereas only 9% VNN2 expression was observed on CD14+ HLA-DRhigh cells (n = 4 p < 0.01). The top 10 percent positive VNN2 monocytes expressed CD33 and CD11b while being negative for HLA-DR, CD3, CD15, CD19 and CD56, consistent with a Mo-MDSC phenotype. CD14+VNN2high monocytes were able to inhibit CD8 T cell proliferation comparably to traditional Mo-MDSC at 51 % and 48 % respectively. However, VNN2 expression on CD14+ monocytes from glioma patients was inversely correlated to their grade. CD14+VNN2high monocytes thus appear to mark a monocytic population similar to Mo-MDSC only in healthy subjects, which may be useful for tumor diagnoses.
Collapse
Affiliation(s)
- David C. Soler
- The Department of Neurosurgery, Cleveland Clinic Foundation, Cleveland, OH 44195.,Brain Tumor and Neuro-Oncology Center, and the Center of Excellence for Translational Neuro-Oncology, Cleveland Clinic Foundation, Cleveland, OH 44195.,University Hospitals-Seidman Center and the Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - Amber Kerstetter-Fogle
- The Department of Neurosurgery, Cleveland Clinic Foundation, Cleveland, OH 44195.,Brain Tumor and Neuro-Oncology Center, and the Center of Excellence for Translational Neuro-Oncology, Cleveland Clinic Foundation, Cleveland, OH 44195.,University Hospitals-Seidman Center and the Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - Andrew B. Young
- Department of Dermatology, University Hospitals-Cleveland Medical Center and the Case Western University School of Medicine, 11100 Euclid Avenue, Cleveland, OH, 44106 USA.,The Murdough Family Center for Psoriasis, University Hospitals-Cleveland Medical Center and the Case Western University School of Medicine, 11100 Euclid Avenue, Cleveland, OH, 44106 USA
| | - Pat Rayman
- Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - James H. Finke
- Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - Sarah M. Debanne
- Epidemiology and Biostatistics, University Hospitals-Cleveland Medical Center and the Case Western University School of Medicine, 11100 Euclid Avenue, Cleveland, OH, 44106 USA
| | - Kevin D. Cooper
- Department of Dermatology, University Hospitals-Cleveland Medical Center and the Case Western University School of Medicine, 11100 Euclid Avenue, Cleveland, OH, 44106 USA.,The Murdough Family Center for Psoriasis, University Hospitals-Cleveland Medical Center and the Case Western University School of Medicine, 11100 Euclid Avenue, Cleveland, OH, 44106 USA
| | - Jill Barnholtz-Sloan
- The Department of Neurosurgery, Cleveland Clinic Foundation, Cleveland, OH 44195.,Brain Tumor and Neuro-Oncology Center, and the Center of Excellence for Translational Neuro-Oncology, Cleveland Clinic Foundation, Cleveland, OH 44195.,University Hospitals-Seidman Center and the Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland Clinic Foundation, Cleveland, OH 44195.,Epidemiology and Biostatistics, University Hospitals-Cleveland Medical Center and the Case Western University School of Medicine, 11100 Euclid Avenue, Cleveland, OH, 44106 USA
| | - Andrew E. Sloan
- The Department of Neurosurgery, Cleveland Clinic Foundation, Cleveland, OH 44195.,Brain Tumor and Neuro-Oncology Center, and the Center of Excellence for Translational Neuro-Oncology, Cleveland Clinic Foundation, Cleveland, OH 44195.,University Hospitals-Seidman Center and the Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - Thomas S. McCormick
- Department of Dermatology, University Hospitals-Cleveland Medical Center and the Case Western University School of Medicine, 11100 Euclid Avenue, Cleveland, OH, 44106 USA.,The Murdough Family Center for Psoriasis, University Hospitals-Cleveland Medical Center and the Case Western University School of Medicine, 11100 Euclid Avenue, Cleveland, OH, 44106 USA
| |
Collapse
|
22
|
Bline K, Andrews A, Moore-Clingenpeel M, Mertz S, Ye F, Best V, Sayegh R, Tomatis-Souverbielle C, Quintero AM, Maynard Z, Glowinski R, Mejias A, Ramilo O. Myeloid-Derived Suppressor Cells and Clinical Outcomes in Children With COVID-19. Front Pediatr 2022; 10:893045. [PMID: 35733812 PMCID: PMC9207271 DOI: 10.3389/fped.2022.893045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 04/29/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Although children with COVID-19 account for fewer hospitalizations than adults, many develop severe disease requiring intensive care treatment. Critical illness due to COVID-19 has been associated with lymphopenia and functional immune suppression. Myeloid-derived suppressor cells (MDSCs) potently suppress T cells and are significantly increased in adults with severe COVID-19. The role of MDSCs in the immune response of children with COVID-19 is unknown. AIMS We hypothesized that children with severe COVID-19 will have expansion of MDSC populations compared to those with milder disease, and that higher proportions of MDSCs will correlate with clinical outcomes. METHODS We conducted a prospective, observational study on a convenience sample of children hospitalized with PCR-confirmed COVID-19 and pre-pandemic, uninfected healthy controls (HC). Blood samples were obtained within 48 h of admission and analyzed for MDSCs, T cells, and natural killer (NK) cells by flow cytometry. Demographic information and clinical outcomes were obtained from the electronic medical record and a dedicated survey built for this study. RESULTS Fifty children admitted to the hospital were enrolled; 28 diagnosed with symptomatic COVID-19 (10 requiring ICU admission) and 22 detected by universal screening (6 requiring ICU admission). We found that children with severe COVID-19 had a significantly higher percentage of MDSCs than those admitted to the ward and uninfected healthy controls. Increased percentages of MDSCs in peripheral blood mononuclear cells (PBMC) were associated with CD4+ T cell lymphopenia. MDSC expansion was associated with longer hospitalizations and need for respiratory support in children admitted with acute COVID-19. CONCLUSION These findings suggest that MDSCs are part of the dysregulated immune responses observed in children with severe COVID-19 and may play a role in disease pathogenesis. Future mechanistic studies are required to further understand the function of MDSCs in the setting of SARS-CoV-2 infection in children.
Collapse
Affiliation(s)
- Katherine Bline
- Center for Vaccines and Immunity, Nationwide Children's Hospital, Columbus, OH, United States.,Division of Critical Care Medicine, Nationwide Children's Hospital, Columbus, OH, United States
| | - Angel Andrews
- Center for Vaccines and Immunity, Nationwide Children's Hospital, Columbus, OH, United States
| | | | - Sara Mertz
- Center for Vaccines and Immunity, Nationwide Children's Hospital, Columbus, OH, United States
| | - Fang Ye
- Center for Vaccines and Immunity, Nationwide Children's Hospital, Columbus, OH, United States
| | - Victoria Best
- Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, United States
| | - Rouba Sayegh
- Division of Infectious Disease, Nationwide Children's Hospital, Columbus, OH, United States
| | - Cristina Tomatis-Souverbielle
- Center for Vaccines and Immunity, Nationwide Children's Hospital, Columbus, OH, United States.,Division of Infectious Disease, Nationwide Children's Hospital, Columbus, OH, United States
| | - Ana M Quintero
- Division of Infectious Disease, Nationwide Children's Hospital, Columbus, OH, United States
| | - Zachary Maynard
- Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, United States
| | - Rebecca Glowinski
- Center for Vaccines and Immunity, Nationwide Children's Hospital, Columbus, OH, United States
| | - Asuncion Mejias
- Center for Vaccines and Immunity, Nationwide Children's Hospital, Columbus, OH, United States.,Division of Infectious Disease, Nationwide Children's Hospital, Columbus, OH, United States
| | - Octavio Ramilo
- Center for Vaccines and Immunity, Nationwide Children's Hospital, Columbus, OH, United States.,Division of Infectious Disease, Nationwide Children's Hospital, Columbus, OH, United States
| |
Collapse
|
23
|
Treatment with ribociclib shows favourable immunomodulatory effects in patients with hormone receptor-positive breast cancer-findings from the RIBECCA trial. Eur J Cancer 2021; 162:45-55. [PMID: 34953442 DOI: 10.1016/j.ejca.2021.11.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 11/22/2021] [Indexed: 01/15/2023]
Abstract
BACKGROUND Inhibitors of the cyclin-dependent kinases 4 and 6 (CDK4/6i) have significantly improved clinical outcomes in patients with advanced hormone receptor-positive (HR+) breast cancer and have demonstrated favourable antitumour immune responses in preclinical studies. METHODS Here, we investigated peripheral immune responses to ribociclib in patients with metastatic HR+ breast cancer as a preplanned exploratory subanalysis of the RIBECCA trial (NCT03096847). Peripheral blood mononuclear cells were subjected to immune cell profiling, gene expression analysis of immune-related signatures, and deep T cell receptor profiling before treatment started and after 12 weeks of treatment with ribociclib. RESULTS Gene expression analysis revealed an upregulation of signatures associated with an activated adaptive immune system and a decrease in immunosuppressive cytokine signalling during treatment with ribociclib. Profiling of peripheral immune cell subpopulations showed a decrease in Treg cell frequencies, which was associated with treatment response. Furthermore, induction of CD4+ naive T cells could be seen, whereas effector and memory T cell populations remained largely unchanged. Correspondingly, T cell repertoire diversity remained mostly unchanged during treatment, although an increase in clonality could be observed in single patients. CONCLUSIONS We show that treatment with ribociclib has significant effects on the peripheral innate and adaptive immune response in patients with HR+ breast cancer. Our data suggest that these effects lead to an activation of an already existing immune response rather than a de novo induction and make a strong case for future combination strategies of CDK4/6i with immunotherapies to enhance the adaptive immune response in HR+ breast cancer.
Collapse
|
24
|
Wittmann N, Behrendt AK, Mishra N, Bossaller L, Meyer-Bahlburg A. Instructions for Flow Cytometric Detection of ASC Specks as a Readout of Inflammasome Activation in Human Blood. Cells 2021; 10:2880. [PMID: 34831104 PMCID: PMC8616555 DOI: 10.3390/cells10112880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/14/2021] [Accepted: 10/21/2021] [Indexed: 02/06/2023] Open
Abstract
Inflammasome activation is linked to the aggregation of the adaptor protein ASC into a multiprotein complex, known as the ASC speck. Redistribution of cytosolic ASC to this complex has been widely used as a readout for inflammasome activation and precedes the downstream proteolytic release of the proinflammatory cytokines, IL-1β and IL-18. Although inflammasomes are important for many diseases such as periodic fever syndromes, COVID-19, gout, sepsis, atherosclerosis and Alzheimer's disease, only a little knowledge exists on the precise and cell type specific occurrence of inflammasome activation in patient samples ex vivo. In this report, we provide detailed information about the optimal conditions to reliably identify inflammasome activated monocytes by ASC speck formation using a modified flow cytometric method introduced by Sester et al. in 2015. Since no protocol for optimal sample processing exists, we tested human blood samples for various conditions including anticoagulant, time and temperature, the effect of one freeze-thaw cycle for PBMC storage, and the fast generation of a positive control. We believe that this flow cytometric protocol will help researchers to perform high quality translational research in multicenter studies, and therefore provide a basis for investigating the role of the inflammasome in the pathogenesis of various diseases.
Collapse
Affiliation(s)
- Nico Wittmann
- Pediatric Rheumatology, Department Pediatric and Adolescent Medicine, University Medicine, University of Greifswald, 17489 Greifswald, Germany; (N.W.); (A.-K.B.)
| | - Ann-Kathrin Behrendt
- Pediatric Rheumatology, Department Pediatric and Adolescent Medicine, University Medicine, University of Greifswald, 17489 Greifswald, Germany; (N.W.); (A.-K.B.)
| | - Neha Mishra
- Section of Rheumatology, Department of Medicine A, University Medicine, University of Greifswald, 17489 Greifswald, Germany;
| | - Lukas Bossaller
- Section of Rheumatology, Department of Medicine A, University Medicine, University of Greifswald, 17489 Greifswald, Germany;
| | - Almut Meyer-Bahlburg
- Pediatric Rheumatology, Department Pediatric and Adolescent Medicine, University Medicine, University of Greifswald, 17489 Greifswald, Germany; (N.W.); (A.-K.B.)
| |
Collapse
|
25
|
Vanhaver C, van der Bruggen P, Bruger AM. MDSC in Mice and Men: Mechanisms of Immunosuppression in Cancer. J Clin Med 2021; 10:jcm10132872. [PMID: 34203451 PMCID: PMC8268873 DOI: 10.3390/jcm10132872] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/16/2021] [Accepted: 06/18/2021] [Indexed: 12/12/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) expand during pathological conditions in both humans and mice and their presence is linked to poor clinical outcomes for cancer patients. Studying MDSC immunosuppression is restricted by MDSCs’ rarity, short lifespan, heterogeneity, poor viability after freezing and the lack of MDSC-specific markers. In this review, we will compare identification and isolation strategies for human and murine MDSCs. We will also assess what direct and indirect immunosuppressive mechanisms have been attributed to MDSCs. While some immunosuppressive mechanisms are well-documented in mice, e.g., generation of ROS, direct evidence is still lacking in humans. In future, bulk or single-cell genomics could elucidate which phenotypic and functional phenotypes MDSCs adopt in particular microenvironments and help to identify potential targets for therapy.
Collapse
Affiliation(s)
- Christophe Vanhaver
- De Duve Institute, Université Catholique de Louvain, Avenue Hippocrate 74, 1200 Brussels, Belgium;
- Correspondence: (C.V.); (A.M.B.)
| | - Pierre van der Bruggen
- De Duve Institute, Université Catholique de Louvain, Avenue Hippocrate 74, 1200 Brussels, Belgium;
- WELBIO, Avenue Hippocrate 74, 1200 Brussels, Belgium
| | - Annika M. Bruger
- De Duve Institute, Université Catholique de Louvain, Avenue Hippocrate 74, 1200 Brussels, Belgium;
- Correspondence: (C.V.); (A.M.B.)
| |
Collapse
|
26
|
Mortezaee K. Myeloid-derived suppressor cells in cancer immunotherapy-clinical perspectives. Life Sci 2021; 277:119627. [PMID: 34004256 DOI: 10.1016/j.lfs.2021.119627] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 05/11/2021] [Accepted: 05/13/2021] [Indexed: 12/19/2022]
Abstract
Myeloid-derived suppressor cells (MDSCs) are heterogeneous and poorly mature cells of innate immunity that their population is increased substantially in cancer patients. MDSCs represent three subsets including CD14+ monocytic (M), CD15+ granulocytic (G) and Lin- early precursor (e) cells. MDSCs release a number of factors that direct several tumorigenic-related events including immune evasion, angiogenesis and metastasis. Assessment of MDSCs can provide valuable information from cancer immunity state, and it can be an indicator of tumor prognosis. The cells can be targeted in combination with current immunotherapeutic schedules, and the outcomes were promising. The focus of this review is to provide an overview of MDSCs, their involvement in tumor-related immunosuppression, and their impact on cancer immunotherapy. Then, strategies are proposed to boost the power of immune system against MDSCs.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| |
Collapse
|
27
|
Pawelec G, Picard E, Bueno V, Verschoor CP, Ostrand-Rosenberg S. MDSCs, ageing and inflammageing. Cell Immunol 2021; 362:104297. [PMID: 33550187 DOI: 10.1016/j.cellimm.2021.104297] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/30/2020] [Accepted: 01/16/2021] [Indexed: 12/20/2022]
Abstract
The challenge of distinguishing between changes attributable to ageing and those attributable to pathology is even greater for the immune system than for many other organs, and this is especially true for myeloid-derived suppressor cells (MDSCs). Hematopoiesis is different in older adults with a bias towards myelopoiesis, and older adults also manifest "inflammageing" exacerbated by disease and contributing to MDSC induction. Hence, at least in humans, one can only investigate MDSCs in the context of ageing and disease states, and not in the context of ageing processes per se. This contribution provides a brief overview of the literature on MDSCs and ageing in humans.
Collapse
Affiliation(s)
- Graham Pawelec
- Department of Immunology, University of Tübingen, Tübingen, Germany; Health Sciences North Research Institute, Sudbury, Ontario, Canada.
| | - Emilie Picard
- Health Sciences North Research Institute, Sudbury, Ontario, Canada
| | - Valquiria Bueno
- Department of Microbiology, Immunology and Parasitology, UNIFESP Federal University of São Paulo, São Paulo, SP, Brazil
| | - Chris P Verschoor
- Health Sciences North Research Institute, Sudbury, Ontario, Canada; Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Suzanne Ostrand-Rosenberg
- Department of Pathology and Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
28
|
Role of myeloid-derived suppressor cells in metastasis. Cancer Metastasis Rev 2021; 40:391-411. [PMID: 33411082 DOI: 10.1007/s10555-020-09947-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 12/02/2020] [Indexed: 02/06/2023]
Abstract
The spread of primary tumor cells to distant organs, termed metastasis, is the principal cause of cancer mortality and is a critical therapeutic target in oncology. Thus, a better understanding of metastatic progression is critical for improved therapeutic approaches requiring insight into the timing of tumor cell dissemination and seeding of distant organs, which can lead to the formation of occult lesions. However, due to limitations in imaging techniques, primary tumors can only be detected when they reach a relatively large size (e.g., > 1 cm3), which, based on our understanding of tumor evolution, is 10 to 20 years (30 doubling times) following tumor initiation. Recent insights into the timing of metastasis are based on the genomic profiling of paired primary tumors and metastases, suggesting that tumor cell seeding of secondary sites occurs early during tumor progression and years prior to diagnosis. Following seeding, tumor cells may remain in a dormant state as single cells or micrometastases before emerging as overt lesions. This timeline and the role of metastatic dormancy are regulated by interactions between the tumor, its microenvironment, and tumor-specific T cell responses. An improved understanding of the mechanisms and interactions responsible for immune evasion and tumor cell release from dormancy would support the development of novel targeted therapeutics. We posit herein that the immunosuppressive mechanisms mediated by myeloid-derived suppressor cells (MDSCs) are a major contributor to tumor progression, and that these mechanisms promote tumor cell escape from dormancy. Thus, while extensive studies have demonstrated a role for MDSCs in the escape from adoptive and innate immune responses (T-, natural killer (NK)-, and B cell responses), facilitating tumor progression and metastasis, few studies have considered their role in dormancy. In this review, we discuss the role of MDSC expansion, driven by tumor burden, and its role in escape from dormancy, resulting in occult metastases, and the potential for MDSC inhibition as an approach to prolong the survival of patients with advanced malignancies.
Collapse
|
29
|
Alberca RW, Andrade MMDS, Branco ACCC, Pietrobon AJ, Pereira NZ, Fernandes IG, Oliveira LDM, Teixeira FME, Beserra DR, de Oliveira EA, Gozzi-Silva SC, Ramos YÁL, de Brito CA, Arnone M, Orfali RL, Aoki V, Duarte AJDS, Sato MN. Frequencies of CD33+CD11b+HLA-DR-CD14-CD66b+ and CD33+CD11b+HLA-DR-CD14+CD66b- Cells in Peripheral Blood as Severity Immune Biomarkers in COVID-19. Front Med (Lausanne) 2020; 7:580677. [PMID: 33178720 PMCID: PMC7592395 DOI: 10.3389/fmed.2020.580677] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 09/08/2020] [Indexed: 12/18/2022] Open
Abstract
Common clinical features of patients with Coronavirus disease-2019 (COVID-19) vary from fever, to acute severe respiratory distress syndrome. Several laboratory parameters are reported as indicators of COVID-19 severity. We hereby describe the possible novel severity biomarkers for COVID-19, CD11b+CD33+HLA-DR-CD14+ cells and CD11b+CD33+HLA-DR-CD66b+ cells.
Collapse
Affiliation(s)
- Ricardo Wesley Alberca
- Laboratorio de Dermatologia e Imunodeficiencias (LIM-56), Departamento de Dermatologia, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Milena Mary de Souza Andrade
- Laboratorio de Dermatologia e Imunodeficiencias (LIM-56), Departamento de Dermatologia, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Anna Cláudia Calvielli Castelo Branco
- Laboratorio de Dermatologia e Imunodeficiencias (LIM-56), Departamento de Dermatologia, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil.,Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Anna Julia Pietrobon
- Laboratorio de Dermatologia e Imunodeficiencias (LIM-56), Departamento de Dermatologia, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil.,Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Nátalli Zanete Pereira
- Laboratorio de Dermatologia e Imunodeficiencias (LIM-56), Departamento de Dermatologia, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Iara Grigoletto Fernandes
- Laboratorio de Dermatologia e Imunodeficiencias (LIM-56), Departamento de Dermatologia, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Luana de Mendonça Oliveira
- Laboratorio de Dermatologia e Imunodeficiencias (LIM-56), Departamento de Dermatologia, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil.,Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Franciane Mouradian Emidio Teixeira
- Laboratorio de Dermatologia e Imunodeficiencias (LIM-56), Departamento de Dermatologia, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil.,Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Danielle Rosa Beserra
- Laboratorio de Dermatologia e Imunodeficiencias (LIM-56), Departamento de Dermatologia, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Emily Araujo de Oliveira
- Laboratorio de Dermatologia e Imunodeficiencias (LIM-56), Departamento de Dermatologia, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Sarah Cristina Gozzi-Silva
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Yasmim Álefe Leuzzi Ramos
- Laboratorio de Dermatologia e Imunodeficiencias (LIM-56), Departamento de Dermatologia, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Cyro Alves de Brito
- Technical Division of Medical Biology, Immunology Center, Adolfo Lutz Institute, São Paulo, Brazil
| | - Marcelo Arnone
- Departamento de Dermatologia, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Raquel Leao Orfali
- Laboratorio de Dermatologia e Imunodeficiencias (LIM-56), Departamento de Dermatologia, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Valeria Aoki
- Laboratorio de Dermatologia e Imunodeficiencias (LIM-56), Departamento de Dermatologia, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | | | - Maria Notomi Sato
- Laboratorio de Dermatologia e Imunodeficiencias (LIM-56), Departamento de Dermatologia, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
30
|
Guven DC, Sahin TK, Dizdar O, Kilickap S. Predictive biomarkers for immunotherapy efficacy in non-small-cell lung cancer: current status and future perspectives. Biomark Med 2020; 14:1383-1392. [PMID: 33064030 DOI: 10.2217/bmm-2020-0310] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 07/07/2020] [Indexed: 02/07/2023] Open
Abstract
In recent years, immune checkpoint inhibitors have rapidly changed treatment paradigms and have been pivotal for the treatment of advanced NSCLC patients. However, many patients don't respond to immunotherapy, and toxicities are a concern. Mounting evidence suggests that PD-L1 expression and tumor mutational burden are useful biomarkers in NSCLC and widely used in clinical practice. Given various limitations of PD-L1 and tumor mutational burden, many candidate biomarkers have emerged. From these biomarkers, peripheral blood-based biomarkers are promising options for the prediction of immunotherapy efficacy with ease of access, repeatability and low cost. This review provides an overview of recent developments on the biomarkers in immunotherapy efficacy together with comments on future perspectives.
Collapse
Affiliation(s)
- Deniz Can Guven
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Taha K Sahin
- Department of Internal Medicine, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Omer Dizdar
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Saadettin Kilickap
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| |
Collapse
|
31
|
Cassetta L, Bruderek K, Skrzeczynska-Moncznik J, Osiecka O, Hu X, Rundgren IM, Lin A, Santegoets K, Horzum U, Godinho-Santos A, Zelinskyy G, Garcia-Tellez T, Bjelica S, Taciak B, Kittang AO, Höing B, Lang S, Dixon M, Müller V, Utikal JS, Karakoç D, Yilmaz KB, Górka E, Bodnar L, Anastasiou OE, Bourgeois C, Badura R, Kapinska-Mrowiecka M, Gotic M, Ter Laan M, Kers-Rebel E, Król M, Santibañez JF, Müller-Trutwin M, Dittmer U, de Sousa AE, Esendağlı G, Adema G, Loré K, Ersvær E, Umansky V, Pollard JW, Cichy J, Brandau S. Differential expansion of circulating human MDSC subsets in patients with cancer, infection and inflammation. J Immunother Cancer 2020; 8:jitc-2020-001223. [PMID: 32907925 PMCID: PMC7481096 DOI: 10.1136/jitc-2020-001223] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2020] [Indexed: 01/25/2023] Open
Abstract
Background Myeloid-derived suppressor cells (MDSC) are a functional myeloid cell subset that includes myeloid cells with immune suppressive properties. The presence of MDSC has been reported in the peripheral blood of patients with several malignant and non-malignant diseases. So far, direct comparison of MDSC across different diseases and Centers is hindered by technical pitfalls and a lack of standardized methodology. To overcome this issue, we formed a network through the COST Action Mye-EUNITER (www.mye-euniter.eu) with the goal to standardize and facilitate the comparative analysis of human circulating MDSC in cancer, inflammation and infection. In this manuscript, we present the results of the multicenter study Mye-EUNITER MDSC Monitoring Initiative, that involved 13 laboratories and compared circulating MDSC subsets across multiple diseases, using a common protocol for the isolation, identification and characterization of these cells. Methods We developed, tested, executed and optimized a standard operating procedure for the isolation and immunophenotyping of MDSC using blood from healthy donors. We applied this procedure to the blood of almost 400 patients and controls with different solid tumors and non-malignant diseases. The latter included viral infections such as HIV and hepatitis B virus, but also psoriasis and cardiovascular disorders. Results We observed that the frequency of MDSC in healthy donors varied substantially between centers and was influenced by technical aspects such as the anticoagulant and separation method used. Expansion of polymorphonuclear (PMN)-MDSC exceeded the expansion of monocytic MDSC (M-MDSC) in five out of six solid tumors. PMN-MDSC expansion was more pronounced in cancer compared with infection and inflammation. Programmed death-ligand 1 was primarily expressed in M-MDSC and e-MDSC and was not upregulated as a consequence of disease. LOX-1 expression was confined to PMN-MDSC. Conclusions This study provides improved technical protocols and workflows for the multi-center analysis of circulating human MDSC subsets. Application of these workflows revealed a predominant expansion of PMN-MDSC in solid tumors that exceeds expansion in chronic infection and inflammation.
Collapse
Affiliation(s)
- Luca Cassetta
- MRC Centre for Reproductive Health, The University of Edinburgh The Queen's Medical Research Institute, Edinburgh, Edinburgh, UK
| | - Kirsten Bruderek
- Department of Otorhinolaryngology, University Hospital Essen, Essen, Germany
| | - Joanna Skrzeczynska-Moncznik
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Małopolska, Poland
| | - Oktawia Osiecka
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Małopolska, Poland
| | - Xiaoying Hu
- Clinical Cooperation Unit Dermato-Oncology, DKFZ, Heidelberg, Baden-Württemberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Centre Mannheim, Mannheim, Baden-Württemberg, Germany
| | - Ida Marie Rundgren
- Department of Biomedical Laboratory Scientist Education and Chemical Engineering, Faculty of Engineering and Natural Sciences, Western Norway University of Applied Sciences, Bergen, Hordaland, Norway
| | - Ang Lin
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institute, Stockholm, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institute, Stockholm, Stockholm, Sweden
| | - Kim Santegoets
- Medical Center, Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University, Nijmegen, Gelderland, The Netherlands
| | - Utku Horzum
- Department of Basic Oncology, Cancer Institute, Hacettepe University, Ankara, Ankara, Turkey
| | - Ana Godinho-Santos
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, University of Lisbon, Lisboa, Lisboa, Portugal
| | - Gennadiy Zelinskyy
- Institute for Virology, University Hospital Essen, Essen, Nordrhein-Westfalen, Germany
| | - Thalia Garcia-Tellez
- HIV Inflammation and Persistence, Pasteur Institute, Paris, Île-de-France, France
| | - Sunčica Bjelica
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, Beograd, Beograd, Serbia
| | - Bartłomiej Taciak
- Department of Cancer Biology, Institute of Biology, Warsaw University of Life Sciences, Warszawa, Poland.,Cellis AG, Zurich, Switzerland
| | | | - Benedikt Höing
- Department of Otorhinolaryngology, University Hospital Essen, Essen, Germany
| | - Stephan Lang
- Department of Otorhinolaryngology, University Hospital Essen, Essen, Germany
| | - Michael Dixon
- Edinburgh Breast Unit and Breast Cancer Now Research Unit, The University of Edinburgh, Edinburgh, Edinburgh, UK
| | - Verena Müller
- Clinical Cooperation Unit Dermato-Oncology, DKFZ, Heidelberg, Baden-Württemberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Centre Mannheim, Mannheim, Baden-Württemberg, Germany
| | - Jochen Sven Utikal
- Department of Dermatology, Venereology and Allergology, University Medical Centre Mannheim, Mannheim, Baden-Württemberg, Germany.,Clinical Cooperation Unit Dermato-Oncology, German Cancer Research Centre, Heidelberg, Baden-Württemberg, Germany
| | - Derya Karakoç
- Department of Medical and Surgical Research, Institute of Health Sciences, Hacettepe University, Ankara, Ankara, Turkey.,Department of General Surgery, Faculty of Medicine, Hacettepe University, Ankara, Ankara, Turkey
| | - Kerim Bora Yilmaz
- Department of Medical and Surgical Research, Institute of Health Sciences, Hacettepe University, Ankara, Ankara, Turkey.,Department of General Surgery, Gulhane Egitim ve Arastirma Hastanesi, Ankara, Ankara, Turkey
| | - Emilia Górka
- Department of Cancer Biology, Institute of Biology, Warsaw University of Life Sciences, Warszawa, Poland.,Cellis AG, Zurich, Switzerland
| | - Lubomir Bodnar
- Department of Oncology and Immunooncology, Hospital Ministry of the Interior and Administration & Warmia and Masuria Oncology Centre, Olsztyn, Poland.,Department of Oncology, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | | | - Christine Bourgeois
- Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, CEA, Université Paris-Sud, Saint-Aubin, Île-de-France, France
| | - Robert Badura
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, University of Lisbon, Lisboa, Lisboa, Portugal.,Serviço de Doenças Infecciosas, Northern Lisbon University Hospital Centre, Lisboa, Lisboa, Portugal
| | | | - Mirjana Gotic
- Clinic of Hematology, Clinical Center of Serbia, Beograd, Beograd, Serbia
| | - Mark Ter Laan
- Medical Center, Department of Neurosurgery, Radboud University, Nijmegen, Gelderland, The Netherlands
| | - Esther Kers-Rebel
- Medical Center, Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University, Nijmegen, Gelderland, The Netherlands
| | - Magdalena Król
- Department of Cancer Biology, Institute of Biology, Warsaw University of Life Sciences, Warszawa, Poland.,Cellis AG, Zurich, Switzerland
| | - Juan Francisco Santibañez
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, Beograd, Beograd, Serbia.,Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O'Higgins, Santiago, Chile
| | | | - Ulf Dittmer
- Institute for Virology, University Hospital Essen, Essen, Nordrhein-Westfalen, Germany
| | - Ana Espada de Sousa
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, University of Lisbon, Lisboa, Lisboa, Portugal
| | - Güneş Esendağlı
- Department of Basic Oncology, Cancer Institute, Hacettepe University, Ankara, Ankara, Turkey.,Department of Medical and Surgical Research, Institute of Health Sciences, Hacettepe University, Ankara, Ankara, Turkey
| | - Gosse Adema
- Department of Radiation Oncology, Radboud University Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Karin Loré
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institute, Stockholm, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institute, Stockholm, Stockholm, Sweden
| | - Elisabeth Ersvær
- Department of Biomedical Laboratory Scientist Education and Chemical Engineering, Faculty of Engineering and Natural Sciences, Western Norway University of Applied Sciences, Bergen, Hordaland, Norway
| | - Viktor Umansky
- Department of Dermatology, Venereology and Allergology, University Medical Centre Mannheim, Mannheim, Baden-Württemberg, Germany.,Clinical Cooperation Unit Dermato-Oncology, German Cancer Research Centre, Heidelberg, Baden-Württemberg, Germany
| | - Jeffrey W Pollard
- MRC Centre for Reproductive Health, The University of Edinburgh The Queen's Medical Research Institute, Edinburgh, Edinburgh, UK
| | - Joanna Cichy
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Małopolska, Poland
| | - Sven Brandau
- Department of Otorhinolaryngology, University Hospital Essen, Essen, Germany .,German Cancer Consortium, Partner Site Essen-Düsseldorf, Germany
| |
Collapse
|
32
|
Yaseen MM, Abuharfeil NM, Darmani H, Daoud A. Recent advances in myeloid-derived suppressor cell biology. Front Med 2020; 15:232-251. [PMID: 32876877 DOI: 10.1007/s11684-020-0797-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 04/17/2020] [Indexed: 12/17/2022]
Abstract
In recent years, studying the role of myeloid-derived suppressor cells (MDSCs) in many pathological inflammatory conditions has become a very active research area. Although the role of MDSCs in cancer is relatively well established, their role in non-cancerous pathological conditions remains in its infancy resulting in much confusion. Our objectives in this review are to address some recent advances in MDSC research in order to minimize such confusion and to provide an insight into their function in the context of other diseases. The following topics will be specifically focused upon: (1) definition and characterization of MDSCs; (2) whether all MDSC populations consist of immature cells; (3) technical issues in MDSC isolation, estimation and characterization; (4) the origin of MDSCs and their anatomical distribution in health and disease; (5) mediators of MDSC expansion and accumulation; (6) factors that determine the expansion of one MDSC population over the other; (7) the Yin and Yang roles of MDSCs. Moreover, the functions of MDSCs will be addressed throughout the text.
Collapse
Affiliation(s)
- Mahmoud Mohammad Yaseen
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid, 22110, Jordan.
| | - Nizar Mohammad Abuharfeil
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Homa Darmani
- Department of Applied Biology, Faculty of Science and Arts, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Ammar Daoud
- Department of Internal Medicine, Faculty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
| |
Collapse
|
33
|
Market M, Tennakoon G, Ng J, Scaffidi M, de Souza CT, Kennedy MA, Auer RC. A Method of Assessment of Human Natural Killer Cell Phenotype and Function in Whole Blood. Front Immunol 2020; 11:963. [PMID: 32508837 PMCID: PMC7251181 DOI: 10.3389/fimmu.2020.00963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 04/23/2020] [Indexed: 11/13/2022] Open
Abstract
The majority of data on human Natural Killer (NK) cell phenotype and function has been generated using cryopreserved peripheral blood mononuclear cells (PBMCs). However, cryopreservation can have adverse effects on PBMCs. In contrast, investigating immune cells in whole blood can reduce the time, volume of blood required, and potential artefacts associated with manipulation of the cells. Whole blood collected from healthy donors and cancer patients was processed by three separate protocols that can be used independently or in parallel to assess extracellular receptors, intracellular signaling protein phosphorylation, and intracellular and extracellular cytokine production in human NK cells. To assess extracellular receptor expression, 200 μL of whole blood was incubated with an extracellular staining (ECS) mix and cells were subsequently fixed and RBCs lysed prior to analysis. The phosphorylation status of signaling proteins was assessed in 500 μL of whole blood following co-incubation with interleukin (IL)-2/12 and an ECS mix for 20 min prior to cell fixation, RBC lysis, and subsequent permeabilization for staining with an intracellular staining (ICS) mix. Cytokine production (IFNγ) was similarly assessed by incubating 1 mL of whole blood with PMA-ionomycin or IL-2/12 prior to incubation with ECS and subsequent ICS antibodies. In addition, plasma was collected from stimulated samples prior to ECS for quantification of secreted IFNγ by ELISA. Results were consistent, despite inherent inter-patient variability. Although we did not investigate an exhaustive list of targets, this approach enabled quantification of representative ECS surface markers including activating (NKG2D and DNAM-1) and inhibitory (NKG2A, PD-1, TIGIT, and TIM-3) receptors, cytokine receptors (CD25, CD122, CD132, and CD212) and ICS markers associated with NK cell activation following stimulation, including signaling protein phosphorylation (p-STAT4, p-STAT5, p-p38 MAPK, p-S6) and IFNγ in both healthy donors and cancer patients. In addition, we compared extracellular receptor expression using whole blood vs. cryopreserved PBMCs and observed a significant difference in the expression of almost all receptors. The methods presented permit a relatively rapid parallel assessment of immune cell receptor expression, signaling protein activity, and cytokine production in a minimal volume of whole blood from both healthy donors and cancer patients.
Collapse
Affiliation(s)
| | - Gayashan Tennakoon
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Juliana Ng
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | | | | | - Michael A Kennedy
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Rebecca C Auer
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Department of Surgery, University of Ottawa, The Ottawa Hospital, Ottawa, ON, Canada
| |
Collapse
|