1
|
Zhang Y, Nomura M, Nishimura K, Zang W, Koike Y, Xiao M, Ito H, Fukumoto M, Tanaka A, Aoyama Y, Saika W, Hasegawa C, Yamazaki H, Takaori-Kondo A, Inoue D. In-depth functional analysis of BRD9 in fetal hematopoiesis reveals context-dependent roles. iScience 2025; 28:112010. [PMID: 40109374 PMCID: PMC11919606 DOI: 10.1016/j.isci.2025.112010] [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: 07/18/2024] [Revised: 10/19/2024] [Accepted: 02/10/2025] [Indexed: 03/22/2025] Open
Abstract
The hierarchical organization of hematopoietic stem cells (HSCs) governing adult hematopoiesis has been extensively investigated. However, the dynamic epigenomic transition from fetal to adult hematopoiesis remains incompletely understood, particularly regarding the involvement of epigenetic factors. In this study, we investigate the roles of BRD9, an essential component of the non-canonical BAF (ncBAF) complex known to govern the fate of adult HSCs, in fetal hematopoiesis. Consistent with observations in adult hematopoiesis, BRD9 loss impairs fetal HSC stemness and disturbs erythroid maturation. Intriguingly, the impact on myeloid lineage was discrepant: BRD9 loss inhibited and promoted myeloid differentiation in fetal and adult models, respectively. Through comprehensive transcriptomic and epigenomic analysis, we elucidate the differential roles of BRD9 in a context- and lineage-dependent manner. Our data uncover how BRD9/ncBAF complex modulates transcription in a stage-specific manner, providing deeper insights into the epigenetic regulation underlying the transition from fetal to adult hematopoiesis.
Collapse
Affiliation(s)
- Yifan Zhang
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
- Department of Hematology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masaki Nomura
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
- Facility for iPS Cell Therapy, CiRA Foundation, Kyoto, Japan
- Department of Cancer Pathology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Japan
| | - Koutarou Nishimura
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
- Department of Cancer Pathology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Japan
| | - Weijia Zang
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
- Department of Hematology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Cancer Pathology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Japan
| | - Yui Koike
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
- Department of Cancer Pathology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Japan
| | - Muran Xiao
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
| | - Hiromi Ito
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
- Department of Cancer Pathology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Japan
| | - Miki Fukumoto
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
| | - Atsushi Tanaka
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
| | - Yumi Aoyama
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
- Department of Hematology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Wataru Saika
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
- Department of Cancer Pathology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Japan
- Department of Hematology, Shiga University of Medical Science, Otsu, Japan
| | - Chihiro Hasegawa
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
- Department of Cancer Pathology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Japan
- Department of Hematology and Oncology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Hiromi Yamazaki
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
- Department of Cancer Pathology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Japan
| | - Akifumi Takaori-Kondo
- Department of Hematology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Daichi Inoue
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
- Department of Hematology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Cancer Pathology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Japan
- Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
| |
Collapse
|
2
|
Aliazis K, Yenyuwadee S, Phikulsod P, Boussiotis VA. Emergency myelopoiesis in solid cancers. Br J Haematol 2024; 205:798-811. [PMID: 39044285 DOI: 10.1111/bjh.19656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 07/09/2024] [Indexed: 07/25/2024]
Abstract
Cells of the innate and adaptive immune systems are the progeny of haematopoietic stem and progenitor cells (HSPCs). During steady-state myelopoiesis, HSPC undergo differentiation and proliferation but are called to respond directly and acutely to various signals that lead to emergency myelopoiesis, including bone marrow ablation, infections, and sterile inflammation. There is extensive evidence that many solid tumours have the potential to secrete classical myelopoiesis-promoting growth factors and other products able to mimic emergency haematopoiesis, and to aberrantly re-direct myeloid cell development into immunosuppressive cells with tumour promoting properties. Here, we summarize the current literature regarding the effects of solid cancers on HSPCs function and discuss how these effects might shape antitumour responses via a mechanism initiated at a site distal from the tumour microenvironment.
Collapse
Affiliation(s)
- Konstantinos Aliazis
- Department of Hematology-Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Sasitorn Yenyuwadee
- Department of Hematology-Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Department of Dermatology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Ployploen Phikulsod
- Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Vassiliki A Boussiotis
- Department of Hematology-Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
3
|
Görgülügil G, Uğur Kurtoğlu A, Uğur S, Kurtoğlu E. Importance of CD71 + Erythrocyte Cell Levels in Prognosis in Patients With β-Thalassemia. J Clin Lab Anal 2024; 38:e25084. [PMID: 38924171 PMCID: PMC11252824 DOI: 10.1002/jcla.25084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/22/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES CD71+ erythroid cells (CECs) are immature red blood cells (proerythroblasts, erythroblasts, and reticulocytes). CECs play an important role in the development of sepsis and cancer by causing immunosuppression. We examined the CEC levels in the peripheral blood of beta thalassemia (βThal) patients and investigated the relationship between CECs and the clinical status of the patients, especially splenectomy. METHODS Sixty-eight patients with βThal (46 splenectomized and 22 nonsplenectomized) and 15 healthy controls were included in this study. The hemogram parameters, ferritin, and CECs (flow cytometry method) were measured. RESULTS It was observed that the CEC level in the patient group was significantly higher than the control group (p < 0.05). CEC levels were found to be significantly higher in patients with splenectomy than in patients with nonsplenectomy (p < 0.05). CEC levels were higher in patients with nontransfusion-dependent βT (NTD-βThal) than in patients with transfusion-dependent βT (TD-βThal) (p < 0.05). CEC levels were found to be significantly higher in patients with splenectomy than in patients with nonsplenectomy in both TD-βThal and NTD-βThal groups (p < 0.05). There was a moderate-negative correlation was detected between CECs and Hb levels (r = -0.467; p < 0.05). CONCLUSIONS High CEC levels in βThal patients develop as a result of ineffective erythropoiesis. We think that keeping CEC levels under control is important for prognosis, especially in patients with splenectomy.
Collapse
Affiliation(s)
| | | | - Sevcan Uğur
- Department of RheumatologyAntalya City HospitalAntalyaTurkey
| | - Erdal Kurtoğlu
- Department of HematologySağlık Bilimleri UniversityAntalyaTurkey
| |
Collapse
|
4
|
Feng Z, Liu S, Su M, Song C, Lin C, Zhao F, Li Y, Zeng X, Zhu Y, Hou Y, Ren C, Zhang H, Yi P, Ji Y, Wang C, Li H, Ma M, Luo L, Li L. TANGO6 regulates cell proliferation via COPI vesicle-mediated RPB2 nuclear entry. Nat Commun 2024; 15:2371. [PMID: 38490996 PMCID: PMC10943085 DOI: 10.1038/s41467-024-46720-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 03/01/2024] [Indexed: 03/18/2024] Open
Abstract
Coat protein complex I (COPI) vesicles mediate the retrograde transfer of cargo between Golgi cisternae and from the Golgi to the endoplasmic reticulum (ER). However, their roles in the cell cycle and proliferation are unclear. This study shows that TANGO6 associates with COPI vesicles via two transmembrane domains. The TANGO6 N- and C-terminal cytoplasmic fragments capture RNA polymerase II subunit B (RPB) 2 in the cis-Golgi during the G1 phase. COPI-docked TANGO6 carries RPB2 to the ER and then to the nucleus. Functional disruption of TANGO6 hinders the nuclear entry of RPB2, which accumulates in the cytoplasm, causing cell cycle arrest in the G1 phase. The conditional depletion or overexpression of TANGO6 in mouse hematopoietic stem cells results in compromised or expanded hematopoiesis. Our study results demonstrate that COPI vesicle-associated TANGO6 plays a role in the regulation of cell cycle progression by directing the nuclear transfer of RPB2, making it a potential target for promoting or arresting cell expansion.
Collapse
Affiliation(s)
- Zhi Feng
- Research center of Stem cells and Ageing, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, PR China
| | - Shengnan Liu
- Institute of Developmental Biology and Regenerative Medicine, Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Southwest University, Chongqing, 400715, PR China
| | - Ming Su
- Research center of Stem cells and Ageing, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, PR China
| | - Chunyu Song
- Institute of Developmental Biology and Regenerative Medicine, Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Southwest University, Chongqing, 400715, PR China
| | - Chenyu Lin
- Institute of Developmental Biology and Regenerative Medicine, Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Southwest University, Chongqing, 400715, PR China
| | - Fangying Zhao
- Institute of Developmental Biology and Regenerative Medicine, Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Southwest University, Chongqing, 400715, PR China
| | - Yang Li
- Research center of Stem cells and Ageing, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, PR China
| | - Xianyan Zeng
- Institute of Life Sciences, Laboratory of Developmental Biology, Department of Cell Biology and Genetics, Chongqing Medical University, Chongqing, 400016, PR China
| | - Yong Zhu
- Institute of Life Sciences, Laboratory of Developmental Biology, Department of Cell Biology and Genetics, Chongqing Medical University, Chongqing, 400016, PR China
| | - Yu Hou
- Institute of Life Sciences, Laboratory of Developmental Biology, Department of Cell Biology and Genetics, Chongqing Medical University, Chongqing, 400016, PR China
| | - Chunguang Ren
- Institute of Life Sciences, Laboratory of Developmental Biology, Department of Cell Biology and Genetics, Chongqing Medical University, Chongqing, 400016, PR China
| | - Huan Zhang
- Institute of Life Sciences, Laboratory of Developmental Biology, Department of Cell Biology and Genetics, Chongqing Medical University, Chongqing, 400016, PR China
| | - Ping Yi
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, PR China
| | - Yong Ji
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, 211166, PR China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin Medical University, Harbin, 150076, Heilongjiang, PR China
| | - Chao Wang
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, PR China
| | - Hongtao Li
- Institute of Developmental Biology and Regenerative Medicine, Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Southwest University, Chongqing, 400715, PR China
| | - Ming Ma
- Institute of Developmental Biology and Regenerative Medicine, Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Southwest University, Chongqing, 400715, PR China
| | - Lingfei Luo
- Institute of Developmental Biology and Regenerative Medicine, Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Southwest University, Chongqing, 400715, PR China.
| | - Li Li
- Research center of Stem cells and Ageing, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, PR China.
| |
Collapse
|
5
|
Pang XQ, Li X, Zhu WH, Huang RK, Mo ZS, Huang ZX, Zhang Y, Xie DY, Gao ZL. LAG3 + erythroid progenitor cells inhibit HBsAg seroclearance during finite pegylated interferon treatment through LAG3 and TGF-β. Antiviral Res 2023; 213:105592. [PMID: 37004734 DOI: 10.1016/j.antiviral.2023.105592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 03/16/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023]
Abstract
HBsAg seroclearance, the ideal aim of anti-hepatitis B virus (HBV) treatment, cannot be achieved easily. Anemia is another common issue for chronic hepatitis B (CHB) patients, which leads to elevation of erythroid progenitor cells (EPCs) and immune suppression in cancer. This study investigated the role of EPCs in HBsAg seroclearance following pegylated interferon-α (PEG-IFN) treatment. CD45+EPC accumulation in CHB patients and an AAV/HBV mice model was found in the circulation and liver by flow cytometry and immunofluorescence tests. Wright-Giemsa staining showed that these pathological CD45+EPCs presented elevated erythroid cells with relative immature morphologies and atypical cells compared with the control cells. CD45+EPCs were associated with immune tolerance and decreased HBsAg seroclearance during finite PEG-IFN treatment. CD45+EPCs suppressed antigen non-specific T cell activation and HBV-specific CD8+T cells, partially through transforming growth factor β (TGF-β). RNA-seq revealed that CD45+EPCs in patients with CHB presented a distinct gene expression profile compared with CD45-EPCs and CD45+EPCs from cord blood. Notably, CD45+EPCs from patients with CHB expressed high level of Lymphocyte-activation gene 3 (LAG3), an immune checkpoint molecule, and were then defined as LAG3+EPCs. LAG3+EPCs diminished the function of antigen presenting cells through LAG3, which was another mechanism by which LAG3+EPCs' suppressed HBV-specific CD8+T cells. Anti-LAG3 and anti-TGF-β combination treatment decreased serum HBeAg, HBV DNA levels and HBsAg level, as well as HBsAg-expression in hepatocytes during PEG-IFN treatment in the AAV/HBV mice model. Conclusions: LAG3+EPCs inhibited the efficacy of PEG-IFN treatment on HBsAg seroclearance induced by LAG3 and TGF-β. Anti-LAG3, anti-TGF-β and PEG-IFN combination treatment might facilitate HBV clearance.
Collapse
Affiliation(s)
- Xiu-Qing Pang
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, China; Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, China; Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Xing Li
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, China; Department of Medical Oncology, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, China.
| | - Wei-Hang Zhu
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, China; Department of Medical Oncology, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, China
| | - Run-Kai Huang
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, China; Department of Medical Oncology, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, China
| | - Zhi-Shuo Mo
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, China; Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, China; Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Ze-Xuan Huang
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, China
| | - Yuan Zhang
- Department of Obstetrics, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, China
| | - Dong-Ying Xie
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, China; Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, China; Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Zhi-Liang Gao
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, China; Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, China; Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China.
| |
Collapse
|
6
|
Zhang H, Li F, Yang M, Zhang W, He M, Xu H, Wang C, Zhang Y, Wang W, Gao Y, Du X, Li Y. MCL-1 Inhibitor S63845 Distinctively Affects Intramedullary and Extramedullary Hematopoiesis. Pharmaceutics 2023; 15:pharmaceutics15041085. [PMID: 37111571 PMCID: PMC10144179 DOI: 10.3390/pharmaceutics15041085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 03/30/2023] Open
Abstract
Conventional chemotherapy for killing cancer cells using cytotoxic drugs suffers from low selectivity, significant toxicity, and a narrow therapeutic index. Hyper-specific targeted drugs achieve precise destruction of tumors by inhibiting molecular pathways that are critical to tumor growth. Myeloid cell leukemia 1 (MCL-1), an important pro-survival protein in the BCL-2 family, is a promising antitumor target. In this study, we chose to investigate the effects of S63845, a small-molecule inhibitor that targets MCL-1, on the normal hematopoietic system. A mouse model of hematopoietic injury was constructed, and the effects of the inhibitor on the hematopoietic system of mice were evaluated via routine blood tests and flow cytometry. The results showed that S63845 affected the hematopoiesis of various lineages in the early stage of action, causing extramedullary compensatory hematopoiesis in the myeloid and megakaryocytic lineages. The maturation of the erythroid lineage in the intramedullary and extramedullary segments was blocked to varying degrees, and both the intramedullary and extramedullary lymphoid lineages were inhibited. This study provides a complete description of the effects of MCL-1 inhibitor on the intramedullary and extramedullary hematopoietic lineages, which is important for the selection of combinations of antitumor drugs and the prevention of adverse hematopoiesis-related effects.
Collapse
Affiliation(s)
- Hexiao Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China (Y.G.)
- Tianjin Institutes of Health Science, Tianjin 301600, China
| | - Fei Li
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Ming Yang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China (Y.G.)
- Tianjin Institutes of Health Science, Tianjin 301600, China
| | - Wenshan Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China (Y.G.)
- Tianjin Institutes of Health Science, Tianjin 301600, China
| | - Mei He
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China (Y.G.)
- Tianjin Institutes of Health Science, Tianjin 301600, China
| | - Hui Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China (Y.G.)
- Tianjin Institutes of Health Science, Tianjin 301600, China
| | - Chaoqun Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China (Y.G.)
- Tianjin Institutes of Health Science, Tianjin 301600, China
| | - Yiran Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China (Y.G.)
- Tianjin Institutes of Health Science, Tianjin 301600, China
| | - Wei Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China (Y.G.)
- Tianjin Institutes of Health Science, Tianjin 301600, China
| | - Yingdai Gao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China (Y.G.)
- Tianjin Institutes of Health Science, Tianjin 301600, China
| | - Xue Du
- Department of Gynecology, Tianjin Union Medical Center, Tianjin Medical University, Tianjin 300121, China
- Correspondence: (X.D.); (Y.L.)
| | - Yinghui Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China (Y.G.)
- Tianjin Institutes of Health Science, Tianjin 301600, China
- Correspondence: (X.D.); (Y.L.)
| |
Collapse
|
7
|
Li J, Liu L, Xing J, Chen D, Fang C, Mo F, Gong Y, Tan Z, Liang G, Xiao W, Tang S, Wei H, Zhao S, Xie H, Pan X, Yin X, Huang J. TLR7 modulates extramedullary splenic erythropoiesis in P. yoelii NSM-infected mice through the regulation of iron metabolism of macrophages with IFN-γ. Front Immunol 2023; 14:1123074. [PMID: 37180169 PMCID: PMC10174296 DOI: 10.3389/fimmu.2023.1123074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 04/13/2023] [Indexed: 05/15/2023] Open
Abstract
Splenomegaly is a prominent clinical manifestation of malaria and the causes remain incompletely clear. Anemia is induced in malaria and extramedullary splenic erythropoiesis is compensation for the loss of erythrocytes. However, the regulation of extramedullary splenic erythropoiesis in malaria is unknown. An inflammatory response could facilitate extramedullary splenic erythropoiesis in the settings of infection and inflammation. Here, when mice were infected with rodent parasites, Plasmodium yoelii NSM, TLR7 expression in splenocytes was increased. To explore the roles of TLR7 in splenic erythropoiesis, we infected wild-type and TLR7 -/- C57BL/6 mice with P. yoelii NSM and found that the development of splenic erythroid progenitor cells was impeded in TLR7 -/- mice. Contrarily, the treatment of the TLR7 agonist, R848, promoted extramedullary splenic erythropoiesis in wild-type infected mice, which highlights the implication of TLR7 on splenic erythropoiesis. Then, we found that TLR7 promoted the production of IFN-γ that could enhance phagocytosis of infected erythrocytes by RAW264.7. After phagocytosis of infected erythrocytes, the iron metabolism of RAW264.7 was upregulated, evidenced by higher iron content and expression of Hmox1 and Slc40a1. Additionally, the neutralization of IFN-γ impeded the extramedullary splenic erythropoiesis modestly and reduced the iron accumulation in the spleen of infected mice. In conclusion, TLR7 promoted extramedullary splenic erythropoiesis in P. yoelii NSM-infected mice. TLR7 enhanced the production of IFN-γ, and IFN-γ promoted phagocytosis of infected erythrocytes and the iron metabolism of macrophages in vitro, which may be related to the regulation of extramedullary splenic erythropoiesis by TLR7.
Collapse
Affiliation(s)
- Jiajie Li
- Department of Infectious Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lin Liu
- Department of Infectious Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Junmin Xing
- Sino-French Hoffmann Institute, Department of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Dianhui Chen
- Department of Infectious Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chao Fang
- Sino-French Hoffmann Institute, Department of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Feng Mo
- Department of Infectious Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yumei Gong
- Sino-French Hoffmann Institute, Department of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Zhengrong Tan
- Sino-French Hoffmann Institute, Department of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Guikuan Liang
- Sino-French Hoffmann Institute, Department of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Wei Xiao
- Sino-French Hoffmann Institute, Department of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Shanni Tang
- Department of Infectious Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Haixia Wei
- Sino-French Hoffmann Institute, Department of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Shan Zhao
- Sino-French Hoffmann Institute, Department of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Hongyan Xie
- Sino-French Hoffmann Institute, Department of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Xingfei Pan
- Department of Infectious Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- *Correspondence: Xingfei Pan, ; Xiaomao Yin, ; Jun Huang,
| | - Xiaomao Yin
- Department of Laboratory Medicine, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
- *Correspondence: Xingfei Pan, ; Xiaomao Yin, ; Jun Huang,
| | - Jun Huang
- Sino-French Hoffmann Institute, Department of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
- Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Laboratory Medicine, Lecong Hospital, Foshan, China
- *Correspondence: Xingfei Pan, ; Xiaomao Yin, ; Jun Huang,
| |
Collapse
|
8
|
Zhang L, Patel S, Soulakova JN, Caldwell CC, St Pierre Schneider B. Mild hypobaric hypoxia influences splenic proliferation during the later phase of stress erythropoiesis. Exp Biol Med (Maywood) 2021; 247:509-518. [PMID: 34904451 DOI: 10.1177/15353702211060775] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Tissue trauma and hemorrhagic shock are common battlefield injuries that can induce hypoxia, inflammation, and/or anemia. Inflammation and hypoxia can initiate adaptive mechanisms, such as stress erythropoiesis in the spleen, to produce red blood cells and restore the oxygen supply. In a military context, mild hypobaric hypoxia-part of the environmental milieu during aeromedical evacuation or en route care-may influence adaptive mechanisms, such as stress erythropoiesis, and host defense. In the present study, healthy (control), muscle trauma, and polytrauma (muscle trauma and hemorrhagic shock) mice were exposed to normobaric normoxia or hypobaric hypoxia for ∼17.5 h to test the hypothesis that hypobaric hypoxia exposure influences splenic erythropoiesis and splenic inflammation after polytrauma. This hypothesis was partially supported. The polytrauma + hypobaric hypoxia group exhibited more splenic neutrophils, fewer total spleen cells, and fewer splenic proliferating cells than the polytrauma+normobaric normoxia group; however, no splenic erythroid cell differences were detected between the two polytrauma groups. We also compared splenic erythropoiesis and myeloid cell numbers among control, muscle trauma, and polytrauma groups. More reticulocytes at 1.7 days (40 h) post-trauma (dpt) and neutrophils at 4 dpt were produced in the muscle trauma mice than corresponding control mice. In contrast to muscle trauma, polytrauma led to a reduced red blood cell count and elevated serum erythropoietin levels at 1.7 dpt. There were more erythroid subsets and apoptotic reticulocytes in the polytrauma mice than muscle trauma mice at 4 and 8 dpt. At 14 dpt, the red blood cell count of the polytrauma + normobaric normoxia mice was 12% lower than that of the control + normobaric normoxia mice; however, no difference was observed between polytrauma + hypobaric hypoxia and control + hypobaric hypoxia mice. Our findings suggest muscle trauma alone induces stress erythropoiesis; in a polytrauma model, hypobaric hypoxia exposure may result in the dysregulation of splenic cells, requiring a treatment plan to ensure adequate immune functioning.
Collapse
Affiliation(s)
- Liyuan Zhang
- School of Nursing, University of Nevada, Las Vegas, NV 89154, USA
| | - Shailey Patel
- School of Nursing, University of Nevada, Las Vegas, NV 89154, USA
| | - Julia N Soulakova
- Department of Population Health Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Charles C Caldwell
- College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | | |
Collapse
|
9
|
Mashhouri S, Koleva P, Huynh M, Okoye I, Shahbaz S, Elahi S. Sex Matters: Physiological Abundance of Immuno-Regulatory CD71+ Erythroid Cells Impair Immunity in Females. Front Immunol 2021; 12:705197. [PMID: 34367164 PMCID: PMC8334724 DOI: 10.3389/fimmu.2021.705197] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 07/05/2021] [Indexed: 11/13/2022] Open
Abstract
Mature erythrocytes are the major metabolic regulators by transporting oxygen throughout the body. However, their precursors and progenitors defined as CD71+ Erythroid Cells (CECs) exhibit a wide range of immunomodulatory properties. Here, we uncover pronounced sexual dimorphism in CECs. We found female but not male mice, both BALB/c and C57BL/6, and human females were enriched with CECs. CECs, mainly their progenitors defined as CD45+CECs expressed higher levels of reactive oxygen species (ROS), PDL-1, VISTA, Arginase II and Arginase I compared to their CD45- counterparts. Consequently, CECs by the depletion of L-arginine suppress T cell activation and proliferation. Expansion of CECs in anemic mice and also post-menstrual cycle in women can result in L-arginine depletion in different microenvironments in vivo (e.g. spleen) resulting in T cell suppression. As proof of concept, we found that anemic female mice and mice adoptively transferred with CECs from anemic mice became more susceptible to Bordetella pertussis infection. These observations highlight the role of sex and anemia-mediated immune suppression in females. Notably, enriched CD45+CECs may explain their higher immunosuppressive properties in female BALB/c mice. Finally, we observed significantly more splenic central macrophages in female mice, which can explain greater extramedullary erythropoiesis and subsequently abundance of CECs in the periphery. Thus, sex-specific differences frequency in the frequency of CECs might be imprinted by differential erythropoiesis niches and hormone-dependent manner.
Collapse
Affiliation(s)
- Siavash Mashhouri
- School of Dentistry, Division of Foundational Sciences, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Petya Koleva
- School of Dentistry, Division of Foundational Sciences, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Mai Huynh
- School of Dentistry, Division of Foundational Sciences, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Isobel Okoye
- School of Dentistry, Division of Foundational Sciences, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Shima Shahbaz
- School of Dentistry, Division of Foundational Sciences, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Shokrollah Elahi
- School of Dentistry, Division of Foundational Sciences, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
10
|
Jonsson NN, Cox DK, Piper EK, Valdivieso EFM, Constantinoiu C, Jackson LA, Stear MJ, Ross EM, Tabor AE. Allelic Variation in Protein Tyrosine Phosphatase Receptor Type-C in Cattle Influences Erythrocyte, Leukocyte and Humoral Responses to Infestation With the Cattle Tick Rhipicephalus australis. Front Immunol 2021; 12:675979. [PMID: 34305905 PMCID: PMC8300432 DOI: 10.3389/fimmu.2021.675979] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 06/11/2021] [Indexed: 11/13/2022] Open
Abstract
The protein tyrosine phosphatase receptor type-C (PTPRC) gene encodes the common leukocyte antigen (CD45) receptor. CD45 affects cell adhesion, migration, cytokine signalling, cell development, and activation state. Four families of the gene have been identified in cattle: a taurine group (Family 1), two indicine groups (Families 2 and 4) and an African “taurindicine” group (Family 3). Host resistance in cattle to infestation with ticks is moderately heritable and primarily manifests as prevention of attachment and feeding by larvae. This study was conducted to describe the effects of PTPRC genotype on immune-response phenotypes in cattle that display a variable immune responsiveness to ticks. Thirty tick-naïve Santa-Gertrudis cattle (a stabilized composite of 5/8 taurine and 3/8 indicine) were artificially infested with ticks weekly for 13 weeks and ranked according to their tick counts. Blood samples were taken from control and tick-challenged cattle immediately before, then at 21 d after infestation and each subsequent week for 9 weeks. Assays included erythrocyte profiles, white blood cell counts, the percentage of cellular subsets comprising the peripheral blood mononuclear cell (PBMC) population, and the ability of PBMC to recognize and proliferate in response to stimulation with tick antigens in vitro. The cattle were PTPRC genotyped using a RFLP assay that differentiated Family 1 and 3 together (220 bp), from Family 2 (462 bp), and from Family 4 (486 bp). The PTPRC allele frequencies were Family 1/3 = 0.34; Family 2 = 0.47; Family 4 = 0.19. There was no significant association between PTPRC genotype and tick count. Each copy of the Family 1/3 allele significantly decreased total leucocyte count (WCC) and CD8+ cells. Increasing dosage of Family 2 alleles significantly increased red blood cell count (RCC), haematocrit (PCV), and haemoglobin (Hb) concentration in blood. Increasing dosage of the Family 4 allele was associated with increased WCC, reduced RCC, reduced PCV and reduced Hb. Homozygote Family 1/3 animals had consistently lower IgG1 in response to tick Ag than homozygote Family 2 animals. The PTPRC genotype influences the bovine immune response to ticks but was not associated with the observed variation in resistance to tick infestation in this study.
Collapse
Affiliation(s)
- Nicholas N Jonsson
- Institute of Biodiversity Animal Health and Comparative Medicine, University of Glasgow, Glasgow, United Kingdom
| | - David K Cox
- Institute of Biodiversity Animal Health and Comparative Medicine, University of Glasgow, Glasgow, United Kingdom
| | - Emily K Piper
- School of Veterinary Science, The University of Queensland, Brisbane, QLD, Australia
| | - Emily F Mantilla Valdivieso
- Centre for Animal Science, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, Brisbane, QLD, Australia
| | - Constantin Constantinoiu
- College of Public Health, Biomedical and Veterinary Sciences, James Cook University, Townsville, QLD, Australia
| | - Louise A Jackson
- Biosecurity Sciences Laboratory, Biosecurity Queensland, Department of Agriculture and Fisheries, Queensland Government, Brisbane, QLD, Australia
| | - Michael J Stear
- Department of Animal, Plant and Soil Sciences, La Trobe University, Melbourne, VIC, Australia
| | - Elizabeth M Ross
- Centre for Animal Science, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, Brisbane, QLD, Australia
| | - Ala E Tabor
- Centre for Animal Science, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
11
|
Shahbaz S, Xu L, Osman M, Sligl W, Shields J, Joyce M, Tyrrell DL, Oyegbami O, Elahi S. Erythroid precursors and progenitors suppress adaptive immunity and get invaded by SARS-CoV-2. Stem Cell Reports 2021; 16:1165-1181. [PMID: 33979601 PMCID: PMC8111797 DOI: 10.1016/j.stemcr.2021.04.001] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 01/08/2023] Open
Abstract
SARS-CoV-2 infection is associated with lower blood oxygen levels, even in patients without hypoxia requiring hospitalization. This discordance illustrates the need for a more unifying explanation as to whether SARS-CoV-2 directly or indirectly affects erythropoiesis. Here, we show significantly enriched CD71+ erythroid precursors/progenitors in the blood circulation of COVID-19 patients. We found that these cells have distinctive immunosuppressive properties. In agreement, we observed a strong negative correlation between the frequency of these cells with T and B cell proportions in COVID-19 patients. The expansion of these CD71+ erythroid precursors/progenitors was negatively correlated with the hemoglobin levels. A subpopulation of abundant erythroid cells, CD45+ CD71+ cells, co-express ACE2, TMPRSS2, CD147, and CD26, and these can be infected with SARS-CoV-2. In turn, pre-treatment of erythroid cells with dexamethasone significantly diminished ACE2/TMPRSS2 expression and subsequently reduced their infectivity with SARS-CoV-2. This provides a novel insight into the impact of SARS-CoV-2 on erythropoiesis and hypoxia seen in COVID-19 patients.
Collapse
Affiliation(s)
- Shima Shahbaz
- School of Dentistry, Division of Foundational Sciences, University of Alberta, Edmonton, T6G2E1, AB, Canada
| | - Lai Xu
- School of Dentistry, Division of Foundational Sciences, University of Alberta, Edmonton, T6G2E1, AB, Canada
| | - Mohammed Osman
- Department of Medicine, University of Alberta, Edmonton, T6G2E1, AB, Canada
| | - Wendy Sligl
- Department of Medicine, University of Alberta, Edmonton, T6G2E1, AB, Canada; Department of Critical Care Medicine, University of Alberta, Edmonton, T6G2E1, AB, Canada; Division of Infectious Diseases, University of Alberta, Edmonton, T6G2E1, AB, Canada
| | - Justin Shields
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, T6G2E1, AB, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, T6G2E1, AB, Canada
| | - Michael Joyce
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, T6G2E1, AB, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, T6G2E1, AB, Canada
| | - D Lorne Tyrrell
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, T6G2E1, AB, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, T6G2E1, AB, Canada
| | - Olaide Oyegbami
- School of Dentistry, Division of Foundational Sciences, University of Alberta, Edmonton, T6G2E1, AB, Canada
| | - Shokrollah Elahi
- School of Dentistry, Division of Foundational Sciences, University of Alberta, Edmonton, T6G2E1, AB, Canada; Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, T6G2E1, AB, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, T6G2E1, AB, Canada; Department of Medical Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, T6G2E1, AB, Canada.
| |
Collapse
|
12
|
Shim YA, Weliwitigoda A, Campbell T, Dosanjh M, Johnson P. Splenic erythroid progenitors decrease TNF-α production by macrophages and reduce systemic inflammation in a mouse model of T cell-induced colitis. Eur J Immunol 2020; 51:567-579. [PMID: 33180325 DOI: 10.1002/eji.202048687] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 09/13/2020] [Accepted: 11/11/2020] [Indexed: 12/12/2022]
Abstract
In inflammatory bowel disease (IBD), inflammation can occur beyond the intestine and spread systemically causing complications such as arthritis, cachexia, and anemia. Here, we determine the impact of CD45, a pan-leukocyte marker and tyrosine phosphatase, on IBD. Using a mouse model of T cell transfer colitis, CD25- CD45RBhigh CD4+ T cells were transferred into Rag1-deficient mice (RAGKO) and CD45-deficient RAGKO mice (CD45RAGKO). Weight loss and systemic wasting syndrome were delayed in CD45RAGKO mice compared to RAGKO mice, despite equivalent inflammation in the colon. CD45RAGKO mice had reduced serum levels of TNF-α, and reduced TNF-α production by splenic myeloid cells. CD45RAGKO mice also had increased numbers of erythroid progenitors in the spleen, which had previously been shown to be immunosuppressive. Adoptive transfer of these erythroid progenitors into RAGKO mice reduced their weight loss and TNF-α expression by splenic red pulp macrophages. In vitro, erythroid cells suppressed TNF-α expression in red pulp macrophages in a phagocytosis-dependent manner. These findings show a novel role for erythroid progenitors in suppressing the pro-inflammatory function of splenic macrophages and cachexia associated with IBD.
Collapse
Affiliation(s)
- Yaein Amy Shim
- Department of Microbiology and Immunology, University of British Columbia, British Columbia, Canada
| | - Asanga Weliwitigoda
- Department of Microbiology and Immunology, University of British Columbia, British Columbia, Canada.,Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Teresa Campbell
- Department of Microbiology and Immunology, University of British Columbia, British Columbia, Canada.,Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Manisha Dosanjh
- Department of Microbiology and Immunology, University of British Columbia, British Columbia, Canada
| | - Pauline Johnson
- Department of Microbiology and Immunology, University of British Columbia, British Columbia, Canada
| |
Collapse
|
13
|
Elahi S, Vega-López MA, Herman-Miguel V, Ramírez-Estudillo C, Mancilla-Ramírez J, Motyka B, West L, Oyegbami O. CD71 + Erythroid Cells in Human Neonates Exhibit Immunosuppressive Properties and Compromise Immune Response Against Systemic Infection in Neonatal Mice. Front Immunol 2020; 11:597433. [PMID: 33329589 PMCID: PMC7732591 DOI: 10.3389/fimmu.2020.597433] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 10/29/2020] [Indexed: 01/08/2023] Open
Abstract
Newborns are highly susceptible to infectious diseases. The underlying mechanism of neonatal infection susceptibility has generally been related to their under-developed immune system. Nevertheless, this notion has recently been challenged by the discovery of the physiological abundance of immunosuppressive erythroid precursors CD71+ erythroid cells (CECs) in newborn mice and human cord blood. Here, as proof of concept, we show that these cells are also abundant in the peripheral blood of human newborns. Although their frequency appears to be more variable compared to their counterparts in mice, they rapidly decline by 4 weeks of age. However, their proportion remains significantly higher in infants up to six months of age compared to older infants. We found CD45 expressing CECs, as erythroid progenitors, were the prominent source of reactive oxygen species (ROS) production in both humans and mice. Interestingly, a higher proportion of CD45+CECs was observed in the spleen versus bone marrow of neonatal mice, which was associated with a higher ROS production by splenic CECs compared to their siblings in the bone marrow. CECs from human newborns suppressed cytokine production by CD14 monocytes and T cells, which was partially abrogated by apocynin in vitro. Moreover, the depletion of CECs in neonatal mice increased the number of activated effector immune cells in their spleen and liver, which rendered them more resistant to Listeria monocytogenes infection. This was evident by a significant reduction in the bacteria load in the spleen, liver and brain of treated-mice compared to the control group, which enhanced their survival rate. Our finding highlights the immunoregulatory processes mediated by CECs in newborns. Thus, such tightly regulated immune system in newborns/infants may explain one potential mechanism for the asymptomatic or mild COVID-19 infection in this population.
Collapse
Affiliation(s)
- Shokrollah Elahi
- School of Dentistry, University of Alberta, Edmonton, AB, Canada.,Department of Medical Microbiology and Immunology, University of Alberta, Edmonton AB, Canada.,Department of Oncology, University of Alberta, Edmonton, AB, Canada.,Faculty of Medicine and Dentistry, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| | - Marco Antonio Vega-López
- Dept. Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, Mexico
| | - Vladimir Herman-Miguel
- Dept. Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, Mexico
| | - Carmen Ramírez-Estudillo
- Dept. Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, Mexico
| | - Javier Mancilla-Ramírez
- Escuela Superior deMedicina, Instituto Politecnico Nacional, Hospital de la Mujer, Secretaria de Salud, Mexico City, Mexico
| | - Bruce Motyka
- Alberta Transplant Institute and the Canadian Donation and Transplantation Research Program, Edmonton, AB, Canada.,Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Lori West
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton AB, Canada.,Alberta Transplant Institute and the Canadian Donation and Transplantation Research Program, Edmonton, AB, Canada.,Department of Pediatrics, University of Alberta, Edmonton, AB, Canada.,Department of Surgery, University of Alberta, Edmonton, AB, Canada.,Department of Laboratory Medicine & Pathology, University of Alberta, Edmonton, AB, Canada
| | - Olaide Oyegbami
- School of Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|