1
|
Spencer J, Dionisi C. Immature B cell homing shapes human lymphoid tissue structure and function. J Exp Med 2024; 221:e20240085. [PMID: 39093311 PMCID: PMC11296955 DOI: 10.1084/jem.20240085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/18/2024] [Accepted: 07/23/2024] [Indexed: 08/04/2024] Open
Abstract
Shortly after the emergence of newly formed human B cells from bone marrow as transitional cells, they diverge along two developmental pathways that can be distinguished by the level of IgM they express and migratory biases. Here, we propose that differential tissue homing of immature B cell subsets contributes to human lymphoid tissue structure and function.
Collapse
Affiliation(s)
- Jo Spencer
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Chiara Dionisi
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, UK
| |
Collapse
|
2
|
Viallard JF, Parrens M, Blanco P, Moreau JF, Oksenhendler E, Fieschi C. Influence of Splenomegaly and Splenectomy on the Immune Cell Profile of Patients with Common Variable Immunodeficiency Disease. J Clin Immunol 2024; 44:46. [PMID: 38231432 DOI: 10.1007/s10875-023-01648-8] [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: 07/31/2023] [Accepted: 12/19/2023] [Indexed: 01/18/2024]
Abstract
PURPOSE About 25% of patients with common variable immunodeficiency disease (CVID) have splenomegaly, necessitating sometimes splenectomy whom consequences on the immunological profile of CVID patients have never been studied. We analyzed 11 CVID patients' comprehensive blood immune cell phenotypes pre- and post-splenectomy. METHODS Flow cytometry analyses of immune cell populations. RESULTS Among 89 CVID cohort patients, 41 with splenomegaly, splenomegaly was strongly associated with granulomatous disease, autoimmune disorders, lymphoid hyperplasia, and/or portal hypertension. CVID patients with splenomegaly have significant peripheral lymphopenia (p = 0.001), and significantly fewer peripheral class-switched memory B cells (smBs) (p = 0.001), CD4+ T lymphocytes (p = 0.001), NK (p = 0.0001) and dendritic cells (p ≤ 0.01), and significantly more circulating CD4+ and CD8+ (p = 0.00001) T cell subset activation (p = 0.00005), than CVID patients without splenomegaly. Examination of splenectomy impact on circulating lymphocyte subset distributions demonstrated the drastically enhanced total circulating lymphocyte count post-splenectomy, predominantly B lymphocytes and CD8+ T cells. However, splenectomy did not change B cell distribution, with smBs remaining persistently low, in contrast to complete inversion of the circulating T cell composition, with reversal of the CD4+/CD8+ ratio suggesting that amplification of the CD8+ T cell compartment is a CVID characteristic in patients with splenomegaly. Our results highlight this CD8+ amplification in CVID-splenomegaly patients that might be explained by a homing effect to the spleen and/or possible chronic virus replication, which in turn could induce T cell expansions. CONCLUSION Splenectomizing CVID patients with splenomegaly restores the absolute circulating lymphocyte count, suggesting that the decreased T cell count in the presence of splenomegaly cannot be used as an exclusive criterion for combined immunodeficiency.
Collapse
Affiliation(s)
- Jean-François Viallard
- Department of Internal Medicine and Infectious Diseases, University of Bordeaux, Haut-Lévêque Hospital, 5, Avenue Magellan, 33604, Pessac, France.
| | - Marie Parrens
- Pathology Department, University of Bordeaux, Haut-Lévêque Hospital, 33604, Pessac, France
| | | | | | - Eric Oksenhendler
- Clinical Immunology Department, Saint-Louis Hospital, Paris-Diderot University, 75475, Paris, France
| | - Claire Fieschi
- Clinical Immunology Department, Saint-Louis Hospital, Paris-Diderot University, 75475, Paris, France
| |
Collapse
|
3
|
Park S, Kim D, Jung H, Choi IP, Kwon HJ, Lee Y. Contribution of HSP90 Cleavage to the Cytotoxic Effect of Suberoylanilide Hydroxamic Acid In Vivo and the Involvement of TXNIP in HSP90 Cleavage. Biomol Ther (Seoul) 2024; 32:115-122. [PMID: 38148557 PMCID: PMC10762275 DOI: 10.4062/biomolther.2023.104] [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: 05/30/2023] [Revised: 07/11/2023] [Accepted: 07/21/2023] [Indexed: 12/28/2023] Open
Abstract
Heat shock protein (HSP) 90 is expressed in most living organisms, and several client proteins of HSP90 are necessary for cancer cell survival and growth. Previously, we found that HSP90 was cleaved by histone deacetylase (HDAC) inhibitors and proteasome inhibitors, and the cleavage of HSP90 contributes to their cytotoxicity in K562 leukemia cells. In this study, we first established mouse xenograft models with K562 cells expressing the wild-type or cleavage-resistant mutant HSP90β and found that the suppression of tumor growth by the HDAC inhibitor suberoylanilide hydroxamic acid (SAHA) was interrupted by the mutation inhibiting the HSP90 cleavage in vivo. Next, we investigated the possible function of thioredoxin interacting protein (TXNIP) in the HSP90 cleavage induced by SAHA. TXNIP is a negative regulator for thioredoxin, an antioxidant protein. SAHA transcriptionally induced the expression of TXNIP in K562 cells. HSP90 cleavage was induced by SAHA also in the thymocytes of normal mice and suppressed by an anti-oxidant and pan-caspase inhibitor. When the thymocytes from the TXNIP knockout mice and their wild-type littermate control mice were treated with SAHA, the HSP90 cleavage was detected in the thymocytes of the littermate controls but suppressed in those of the TXNIP knockout mice suggesting the requirement of TXNIP for HSP90 cleavage. We additionally found that HSP90 cleavage was induced by actinomycin D, β-mercaptoethanol, and p38 MAPK inhibitor PD169316 suggesting its prevalence. Taken together, we suggest that HSP90 cleavage occurs also in vivo and contributes to the anti-cancer activity of various drugs in a TXNIP-dependent manner.
Collapse
Affiliation(s)
- Sangkyu Park
- Biotechnology Research Institute, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Dongbum Kim
- Institute of Medical Science, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Haiyoung Jung
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
- Department of Functional Genomics, Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - In Pyo Choi
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Hyung-Joo Kwon
- Institute of Medical Science, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Younghee Lee
- Biotechnology Research Institute, Chungbuk National University, Cheongju 28644, Republic of Korea
- Department of Biochemistry, College of Natural Sciences, Chungbuk National University, Cheongju 28644, Republic of Korea
| |
Collapse
|
4
|
Ding W, Xu D, Li F, Huang C, Song T, Zhong N, Lai K, Deng Z. Intrapulmonary IFN-γ instillation causes chronic lymphocytic inflammation in the spleen and lung through the CXCR3 pathway. Int Immunopharmacol 2023; 122:110675. [PMID: 37481849 DOI: 10.1016/j.intimp.2023.110675] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 07/05/2023] [Accepted: 07/17/2023] [Indexed: 07/25/2023]
Abstract
Some patients with chronic refractory cough have high levels of pulmonary IFN-γ and IFN-γ-producing T lymphocytes. Pulmonary IFN-γ administration causes acute airway lymphocytic inflammation and cough hypersensitivity by increasing the number of pulmonary IFN-γ-producing T lymphocytes, but these lymphocytes may be recruited from other organs. Intraperitoneal IFN-γ injection can increase the spleen weight of mice. It remains elusive whether pulmonary IFN-γ can induce chronic airway lymphocytic inflammation and cough hypersensitivity by stimulating the proliferation of IFN-γ -producing T lymphocytes in the spleen. Here, we found that pulmonary IFN-γ administration induced chronic airway inflammation and chronic cough hypersensitivity with an increased number of IFN-γ-producing T lymphocytes in the spleen, blood and lung. Pulmonary IFN-γ administration also increased 1) the proliferation of spleen lymphocytes in vivo and 2) the IP-10 level and CXCR3+ T lymphocyte numbers in the spleen and lung of mice. IP-10 could promote the proliferation of spleen lymphocytes in vitro but not blood lymphocytes or lung-resident lymphocytes. AMG487, a potent inhibitor of binding between IP-10 and CXCR3, could block pulmonary IFN-γ instillation-induced chronic airway lymphocytic inflammation and the proliferation of IFN-γ-producing T lymphocytes in mouse spleens. In conclusion, intrapulmonary IFN-γ instillation may induce the proliferation of splenic IFN-γ-producing T lymphocytes through IP-10 and the CXCR3 pathway. The IFN-γ-producing T lymphocytes in blood, partly released from the mouse spleen, may be partly attracted to the lung by pulmonary IP-10 through the CXCR3 pathway. IFN-γ-producing T lymphocytes and IFN-γ in the lung may cause chronic airway lymphocytic inflammation and chronic cough hypersensitivity.
Collapse
Affiliation(s)
- Wenbin Ding
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Dongting Xu
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Fengying Li
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chuqin Huang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Tongtong Song
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Nanshan Zhong
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Kefang Lai
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Zheng Deng
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
5
|
Atmakusuma TD, Girson R, Koesnoe S. Correlations between Iron Load and CD4 in Adult Transfusion-Dependent Beta Thalassemia. Anemia 2021; 2021:5549503. [PMID: 34239727 PMCID: PMC8233081 DOI: 10.1155/2021/5549503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 06/04/2021] [Accepted: 06/09/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Thalassemia is a hereditary disease, and severe anemia is the main phenotype of major thalassemia. Furthermore, the most important method in the management of this disease is red blood cell transfusion. Regular transfusions administered 1 or 2 times every month improve prognosis and survival. However, there is higher risk of infections and iron overload, especially in transfusion-dependent thalassemia (TDT). Infections are the second leading cause of death in adult TDT, after heart failure. Higher risk of infection is also influenced by multiple blood transfusions which causes alteration in immune response due to alloimmunization, transfusion-related infections, and iron overload. Meanwhile, iron overload in TDT alters both innate and specific immune responses. Furthermore, previous studies have shown the correlation between ferritin with CD4, but this has not been carried out in Indonesia. Therefore, this study aims to determine the correlations between iron overload (serum ferritin and transferrin saturation) and specific immune cells (CD4). METHODS This is a cross-sectional study, and a total number of 64 subjects were examined consecutively. Chest X-ray and blood sera were obtained. The total number of subjects was 64. The seromarkers HBsAg, anti-HCV, and anti-HIV were tested using the ELISA method. Serum ferritin and transferrin saturation was tested using ECLIA, and lymphocyte subsets were analyzed using flowcytometry. Meanwhile, the correlation between variables was determined using Spearman's test. RESULTS The results showed that 4.9% subjects were HBsAg positive, 10.7% were anti-HCV positive, and none were anti-HIV positive. There were 4 subjects with lung tuberculosis based on the 41 chest X-ray. Meanwhile, there was a weak negative and insignificant correlation between serum ferritin with CD4 (p=0.75; r = -0.04) and a weak positive and insignificant correlation between transferrin saturation with CD4 (p=0.133; r = 0.19). CONCLUSION There were no correlations between iron overload (ferritin) and cellular immunity (CD4) in adult transfusion-dependent thalassemia.
Collapse
Affiliation(s)
- Tubagus Djumhana Atmakusuma
- Division of Hematology-Medical Oncology, Department of Internal Medicine Dr. Cipto Mangunkusumo General Hospital, Faculty of Medicine Universitas Indonesia, Kota Depok, Indonesia
| | - Ralph Girson
- Division of Hematology-Medical Oncology, Department of Internal Medicine Dr. Cipto Mangunkusumo General Hospital, Faculty of Medicine Universitas Indonesia, Kota Depok, Indonesia
| | - Sukamto Koesnoe
- Division of Allergy-Immunology, Department of Internal Medicine. Dr. Cipto Mangunkusumo General Hospital, Faculty of Medicine Universitas Indonesia, Kota Depok, Indonesia
| |
Collapse
|
6
|
Tombácz I, Laczkó D, Shahnawaz H, Muramatsu H, Natesan A, Yadegari A, Papp TE, Alameh MG, Shuvaev V, Mui BL, Tam YK, Muzykantov V, Pardi N, Weissman D, Parhiz H. Highly efficient CD4+ T cell targeting and genetic recombination using engineered CD4+ cell-homing mRNA-LNP. Mol Ther 2021; 29:3293-3304. [PMID: 34091054 PMCID: PMC8571164 DOI: 10.1016/j.ymthe.2021.06.004] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 04/30/2021] [Accepted: 06/01/2021] [Indexed: 11/23/2022] Open
Abstract
Nucleoside-modified messenger RNA (mRNA)-lipid nanoparticles (LNPs) are the basis for the first two EUA (Emergency Use Authorization) COVID-19 vaccines. The use of nucleoside-modified mRNA as a pharmacological agent opens immense opportunities for therapeutic, prophylactic and diagnostic molecular interventions. In particular, mRNA-based drugs may specifically modulate immune cells, such as T lymphocytes, for immunotherapy of oncologic, infectious and other conditions. The key challenge, however, is that T cells are notoriously resistant to transfection by exogenous mRNA. Here, we report that conjugating CD4 antibody to LNPs enables specific targeting and mRNA interventions to CD4+ cells, including T cells. After systemic injection in mice, CD4-targeted radiolabeled mRNA-LNPs accumulated in spleen, providing ∼30-fold higher signal of reporter mRNA in T cells isolated from spleen as compared with non-targeted mRNA-LNPs. Intravenous injection of CD4-targeted LNPs loaded with Cre recombinase-encoding mRNA provided specific dose-dependent loxP-mediated genetic recombination, resulting in reporter gene expression in about 60% and 40% of CD4+ T cells in spleen and lymph nodes, respectively. T cell phenotyping showed uniform transfection of T cell subpopulations, with no variability in uptake of CD4-targeted mRNA-LNPs in naive, central memory, and effector cells. The specific and efficient targeting and transfection of mRNA to T cells established in this study provides a platform technology for immunotherapy of devastating conditions and HIV cure.
Collapse
Affiliation(s)
- István Tombácz
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Dorottya Laczkó
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hamna Shahnawaz
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hiromi Muramatsu
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ambika Natesan
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Amir Yadegari
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tyler E Papp
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mohamad-Gabriel Alameh
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Vladimir Shuvaev
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | | | - Ying K Tam
- Acuitas Therapeutics, Vancouver, BC V6T 1Z3, Canada
| | - Vladimir Muzykantov
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Norbert Pardi
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Drew Weissman
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hamideh Parhiz
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
7
|
Yang F, Nielsen SCA, Hoh RA, Röltgen K, Wirz OF, Haraguchi E, Jean GH, Lee JY, Pham TD, Jackson KJL, Roskin KM, Liu Y, Nguyen K, Ohgami RS, Osborne EM, Nadeau KC, Niemann CU, Parsonnet J, Boyd SD. Shared B cell memory to coronaviruses and other pathogens varies in human age groups and tissues. Science 2021; 372:738-741. [PMID: 33846272 PMCID: PMC8139427 DOI: 10.1126/science.abf6648] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 04/07/2021] [Indexed: 12/12/2022]
Abstract
It remains unclear whether B cell repertoires against coronaviruses and other pathogens differ between adults and children and how important these distinctions are. Yang et al. analyzed blood samples from young children and adults, as well as tissues from deceased organ donors, characterizing the B cell receptor (BCR) repertoires specific to six common pathogens and two viruses that they had not seen before: Ebola virus and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Children had higher frequencies of B cells with convergent BCR heavy chains against previously encountered pathogens and higher frequencies of class-switched convergent B cell clones against SARS-CoV-2 and related coronaviruses. These findings suggest that encounters with coronaviruses in early life may produce cross-reactive memory B cell populations that contribute to divergent COVID-19 susceptibilities. Science, this issue p. 738 Vaccination and infection promote the formation, tissue distribution, and clonal evolution of B cells, which encode humoral immune memory. We evaluated pediatric and adult blood and deceased adult organ donor tissues to determine convergent antigen-specific antibody genes of similar sequences shared between individuals. B cell memory varied for different pathogens. Polysaccharide antigenspecific clones were not exclusive to the spleen. Adults had higher clone frequencies and greater class switching in lymphoid tissues than blood, while pediatric blood had abundant class-switched convergent clones. Consistent with reported serology, prepandemic children had class-switched convergent clones to severe acute respiratory syndrome coronavirus 2 with weak cross-reactivity to other coronaviruses, while adult blood or tissues showed few such clones. These results highlight the prominence of early childhood B cell clonal expansions and cross-reactivity for future responses to novel pathogens.
Collapse
Affiliation(s)
- Fan Yang
- Department of Pathology, Stanford University, Stanford, CA 94305, USA.
| | | | - Ramona A Hoh
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Katharina Röltgen
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | | | - Emily Haraguchi
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Grace H Jean
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Ji-Yeun Lee
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Tho D Pham
- Department of Pathology, Stanford University, Stanford, CA 94305, USA.,Stanford Blood Center, Stanford University, Stanford, CA 94305, USA
| | | | - Krishna M Roskin
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH 45267, USA.,Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.,Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Yi Liu
- Calico Life Sciences, South San Francisco, CA 94080, USA
| | - Khoa Nguyen
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Robert S Ohgami
- Department of Pathology, University of California, San Francisco, CA 94143, USA
| | - Eleanor M Osborne
- Sarah Cannon Cancer Center, Tennessee Oncology, Smyrna, TN 37167, USA
| | - Kari C Nadeau
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University, Stanford, CA 94305, USA.,Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, CA 94305, USA
| | - Claus U Niemann
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94143, USA.,Department of Surgery, Division of Transplantation, University of California, San Francisco, CA 94143, USA
| | - Julie Parsonnet
- Department of Medicine, Stanford University, Stanford, CA 94305, USA.,Epidemiology and Population Health, Stanford University, Stanford, CA 94305, USA
| | - Scott D Boyd
- Department of Pathology, Stanford University, Stanford, CA 94305, USA. .,Sean N. Parker Center for Allergy and Asthma Research, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
8
|
Gualdrón-López M, Díaz-Varela M, Toda H, Aparici-Herraiz I, Pedró-Cos L, Lauzurica R, Lacerda MVG, Fernández-Sanmartín MA, Fernandez-Becerra C, Del Portillo HA. Multiparameter Flow Cytometry Analysis of the Human Spleen Applied to Studies of Plasma-Derived EVs From Plasmodium vivax Patients. Front Cell Infect Microbiol 2021; 11:596104. [PMID: 33732657 PMCID: PMC7957050 DOI: 10.3389/fcimb.2021.596104] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 01/05/2021] [Indexed: 11/13/2022] Open
Abstract
The spleen is a secondary lymphoid organ with multiple functions including the removal of senescent red blood cells and the coordination of immune responses against blood-borne pathogens, such as malaria parasites. Despite the major role of the spleen, the study of its function in humans is limited by ethical implications to access human tissues. Here, we employed multiparameter flow cytometry combined with cell purification techniques to determine human spleen cell populations from transplantation donors. Spleen immuno-phenotyping showed that CD45+ cells included B (30%), CD4+ T (16%), CD8+ T (10%), NK (6%) and NKT (2%) lymphocytes. Myeloid cells comprised neutrophils (16%), monocytes (2%) and DCs (0.3%). Erythrocytes represented 70%, reticulocytes 0.7% and hematopoietic stem cells 0.02%. Extracellular vesicles (EVs) are membrane-bound nanoparticles involved in intercellular communication and secreted by almost all cell types. EVs play several roles in malaria that range from modulation of immune responses to vascular alterations. To investigate interactions of plasma-derived EVs from Plasmodium vivax infected patients (PvEVs) with human spleen cells, we used size-exclusion chromatography (SEC) to separate EVs from the bulk of soluble plasma proteins and stained isolated EVs with fluorescent lipophilic dyes. The integrated cellular analysis of the human spleen and the methodology employed here allowed in vitro interaction studies of human spleen cells and EVs that showed an increased proportion of T cells (CD4+ 3 fold and CD8+ 4 fold), monocytes (1.51 fold), B cells (2.3 fold) and erythrocytes (3 fold) interacting with PvEVs as compared to plasma-derived EVs from healthy volunteers (hEVs). Future functional studies of these interactions can contribute to unveil pathophysiological processes involving the spleen in vivax malaria.
Collapse
Affiliation(s)
- Melisa Gualdrón-López
- ISGlobal, Hospital Clinic - Universitat de Barcelona, Barcelona, Spain.,IGTP: Germans Trias i Pujol Research Institute, Barcelona, Spain
| | | | - Haruka Toda
- ISGlobal, Hospital Clinic - Universitat de Barcelona, Barcelona, Spain
| | | | - Laura Pedró-Cos
- IGTP: Germans Trias i Pujol Research Institute, Barcelona, Spain
| | - Ricardo Lauzurica
- Nephrology Service, Germans Trias i Pujol University Hospital, Badalona, Spain
| | - Marcus V G Lacerda
- Fundaçao de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil.,Instituto Leônidas & Maria Deane (ILMD), Fiocruz, Manaus, Brazil
| | | | - Carmen Fernandez-Becerra
- ISGlobal, Hospital Clinic - Universitat de Barcelona, Barcelona, Spain.,IGTP: Germans Trias i Pujol Research Institute, Barcelona, Spain
| | - Hernando A Del Portillo
- ISGlobal, Hospital Clinic - Universitat de Barcelona, Barcelona, Spain.,IGTP: Germans Trias i Pujol Research Institute, Barcelona, Spain.,ICREA: Catalan Institution for Research and Advanced Studies, Barcelona, Spain
| |
Collapse
|
9
|
Matas‐Céspedes A, Brown L, Mahbubani KT, Bareham B, Higgins J, Curran M, de Haan L, Lapointe J, Stebbings R, Saeb‐Parsy K. Use of human splenocytes in an innovative humanised mouse model for prediction of immunotherapy-induced cytokine release syndrome. Clin Transl Immunology 2020; 9:e1202. [PMID: 33173582 PMCID: PMC7641894 DOI: 10.1002/cti2.1202] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/09/2020] [Accepted: 10/06/2020] [Indexed: 01/17/2023] Open
Abstract
OBJECTIVES Humanised mice have emerged as valuable models for pre-clinical testing of the safety and efficacy of immunotherapies. Given the variety of models available, selection of the most appropriate humanised mouse model is critical in study design. Here, we aimed to develop a model for predicting cytokine release syndrome (CRS) while minimising graft-versus-host disease (GvHD). METHODS To overcome donor-induced variation, we directly compared the in vitro and in vivo immune phenotype of immunodeficient NSG mice reconstituted with human bone marrow (BM) CD34+ haematopoietic stem cells (HSCs), peripheral blood mononuclear cells (PBMCs) or spleen mononuclear cells (SPMCs) from the same human donors. SPMC engraftment in NSG-dKO mice, which lack MHC class I and II, was also evaluated as a strategy to limit GvHD. Another group of mice was engrafted with umbilical cord blood (UCB) CD34+ HSCs. Induction of CRS in vivo was investigated upon administration of the anti-CD3 monoclonal antibody OKT3. RESULTS PBMC- and SPMC-reconstituted NSG mice showed short-term survival, with engrafted human T cells exhibiting mostly an effector memory phenotype. Survival in SPMC-reconstituted NSG-dKO mice was significantly longer. Conversely, both BM and UCB-HSC models showed longer survival, without demonstrable GvHD and a more naïve T-cell phenotype. PBMC- and SPMC-reconstituted mice, but not BM-HSC or UCB-HSC mice, experienced severe clinical signs of CRS upon administration of OKT3. CONCLUSION PBMC- and SPMC-reconstituted NSG mice better predict OKT3-mediated CRS. The SPMC model allows generation of large experimental groups, and the use of NSG-dKO mice mitigates the limitation of early GvHD.
Collapse
Affiliation(s)
- Alba Matas‐Céspedes
- Clinical Pharmacology and Safety SciencesR&DAstraZenecaCambridgeUK
- Department of SurgeryUniversity of Cambridge and NIHR Cambridge Biomedical CampusCambridgeUK
| | - Lee Brown
- Clinical Pharmacology and Safety SciencesR&DAstraZenecaCambridgeUK
| | - Krishnaa T Mahbubani
- Department of SurgeryUniversity of Cambridge and NIHR Cambridge Biomedical CampusCambridgeUK
| | - Bethany Bareham
- Department of SurgeryUniversity of Cambridge and NIHR Cambridge Biomedical CampusCambridgeUK
| | - Jackie Higgins
- Department of SurgeryUniversity of Cambridge and NIHR Cambridge Biomedical CampusCambridgeUK
| | - Michelle Curran
- Clinical Pharmacology and Safety SciencesR&DAstraZenecaCambridgeUK
- Department of SurgeryUniversity of Cambridge and NIHR Cambridge Biomedical CampusCambridgeUK
| | - Lolke de Haan
- Clinical Pharmacology and Safety SciencesR&DAstraZenecaCambridgeUK
- Present address:
ADC TherapeuticsLondonUK
| | | | | | - Kourosh Saeb‐Parsy
- Department of SurgeryUniversity of Cambridge and NIHR Cambridge Biomedical CampusCambridgeUK
| |
Collapse
|
10
|
Perez MD, Seu L, Lowman KE, Moylan DC, Tidwell C, Samuel S, Duverger A, Wagner FH, Carlin E, Sharma V, Pope B, Raman C, Erdmann N, Locke J, Hu H, Sabbaj S, Kutsch O. The tetraspanin CD151 marks a unique population of activated human T cells. Sci Rep 2020; 10:15748. [PMID: 32978478 PMCID: PMC7519159 DOI: 10.1038/s41598-020-72719-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 09/04/2020] [Indexed: 02/06/2023] Open
Abstract
Tetraspanins are a family of proteins with an array of functions that are well studied in cancer biology, but their importance in immunology is underappreciated. Here we establish the tetraspanin CD151 as a unique marker of T-cell activation and, in extension, an indicator of elevated, systemic T-cell activity. Baseline CD151 expression found on a subset of T-cells was indicative of increased activation of the MAPK pathway. Following TCR/CD3 activation, CD151 expression was upregulated on the overall T-cell population, a quintessential feature of an activation marker. CD151+ T-cell frequencies in the spleen, an organ with increased immune activity, were twice as high as in paired peripheral blood samples. This CD151+ T-cell frequency increase was not paralleled by an increase of CD25 or CD38, demonstrating that CD151 expression is regulated independently of other T-cell activation markers. CD151+ T-cells were also more likely to express preformed granzyme B, suggesting that CD151+ T cells are pro-inflammatory. To this end, HIV-1 patients on antiretroviral therapy who are reported to exhibit chronically elevated levels of immune activity, had significantly higher CD4+CD151+ T-cell frequencies than healthy controls, raising the possibility that proinflammatory CD151+ T cells could contribute to the premature immunological aging phenotype observed in these patients.
Collapse
Affiliation(s)
- Mildred D Perez
- Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lillian Seu
- Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kelsey E Lowman
- Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - David C Moylan
- Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Christopher Tidwell
- Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Shekwonya Samuel
- Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Alexandra Duverger
- Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Frederic H Wagner
- Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Eric Carlin
- Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Vishal Sharma
- Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Brandon Pope
- Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Chander Raman
- Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nathan Erdmann
- Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jayme Locke
- Department of Surgery, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hui Hu
- Department of Microbiology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Steffanie Sabbaj
- Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Olaf Kutsch
- Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
11
|
Abstract
PURPOSE OF REVIEW Immunotherapy strategies alternative to current antiretroviral therapies will need to address viral diversity while increasing the immune system's ability to efficiently target the latent virus reservoir. Antibody-based molecules can be designed based on broadly neutralizing and non-neutralizing antibodies that target free virions and infected cells. These multispecific molecules, either by IgG-like or non-IgG-like in structure, aim to target several independent HIV-1 epitopes and/or engage effector cells to eliminate the replicating virus and infected cells. This detailed review is intended to stimulate discussion on future requirements for novel immunotherapeutic molecules. RECENT FINDINGS Bispecific and trispecific antibodies are engineered as a single molecules to target two or more independent epitopes on the HIV-1 envelope (Env). These antibody-based molecules have increased avidity for Env, leading to improved neutralization potency and breadth compared with single parental antibodies. Furthermore, bispecific and trispecific antibodies that engage cellular receptors with one arm of the molecule help concentrate inhibitory molecules to the sites of potential infection and facilitate engagement of immune effector cells and Env-expressing target cells for their elimination. SUMMARY Recently engineered antibody-based molecules of different sizes and structures show promise in vitro or in vivo and are encouraging candidates for HIV treatment.
Collapse
Affiliation(s)
- Marina Tuyishime
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Guido Ferrari
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
12
|
Newman L, Jasim DA, Prestat E, Lozano N, de Lazaro I, Nam Y, Assas BM, Pennock J, Haigh SJ, Bussy C, Kostarelos K. Splenic Capture and In Vivo Intracellular Biodegradation of Biological-Grade Graphene Oxide Sheets. ACS NANO 2020; 14:10168-10186. [PMID: 32658456 PMCID: PMC7458483 DOI: 10.1021/acsnano.0c03438] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 07/13/2020] [Indexed: 05/20/2023]
Abstract
Carbon nanomaterials, including 2D graphene-based materials, have shown promising applicability to drug delivery, tissue engineering, diagnostics, and various other biomedical areas. However, to exploit the benefits of these materials in some of the areas mentioned, it is necessary to understand their possible toxicological implications and long-term fate in vivo. We previously demonstrated that following intravenous administration, 2D graphene oxide (GO) nanosheets were largely excreted via the kidneys; however, a small but significant portion of the material was sequestered in the spleen. Herein, we interrogate the potential consequences of this accumulation and the fate of the spleen-residing GO over a period of nine months. We show that our thoroughly characterized GO materials are not associated with any detectable pathological consequences in the spleen. Using confocal Raman mapping of tissue sections, we determine the sub-organ biodistribution of GO at various time points after administration. The cells largely responsible for taking up the material are confirmed using immunohistochemistry coupled with Raman spectroscopy, and transmission electron microscopy (TEM). This combination of techniques identified cells of the splenic marginal zone as the main site of GO bioaccumulation. In addition, through analyses using both bright-field TEM coupled with electron diffraction and Raman spectroscopy, we reveal direct evidence of in vivo intracellular biodegradation of GO sheets with ultrastructural precision. This work offers critical information about biological processing and degradation of thin GO sheets by normal mammalian tissue, indicating that further development and exploitation of GO in biomedicine would be possible.
Collapse
Affiliation(s)
- Leon Newman
- Nanomedicine
Lab, National Graphene Institute and Faculty of Biology, Medicine
& Health, The University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Dhifaf A. Jasim
- Nanomedicine
Lab, National Graphene Institute and Faculty of Biology, Medicine
& Health, The University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Eric Prestat
- Department
of Materials, School of Natural Sciences, The University of Manchester, Manchester, M13 9PL, United Kingdom
| | - Neus Lozano
- Nanomedicine
Lab, National Graphene Institute and Faculty of Biology, Medicine
& Health, The University of Manchester, Manchester, M13 9PT, United Kingdom
- Catalan
Institute of Nanoscience and Nanotechnology (ICN2), Barcelona, 08193, Spain
| | - Irene de Lazaro
- Nanomedicine
Lab, National Graphene Institute and Faculty of Biology, Medicine
& Health, The University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Yein Nam
- Nanomedicine
Lab, National Graphene Institute and Faculty of Biology, Medicine
& Health, The University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Bakri M. Assas
- Lydia
Becker Institute of Immunology and Inflammation, and Division of Infection,
Immunity and Respiratory Medicine, School of Biological Sciences,
Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, United Kingdom
- Department
of Immunology, Faculty of Applied Sciences, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Joanne Pennock
- Lydia
Becker Institute of Immunology and Inflammation, and Division of Infection,
Immunity and Respiratory Medicine, School of Biological Sciences,
Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Sarah J. Haigh
- Department
of Materials, School of Natural Sciences, The University of Manchester, Manchester, M13 9PL, United Kingdom
| | - Cyrill Bussy
- Nanomedicine
Lab, National Graphene Institute and Faculty of Biology, Medicine
& Health, The University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Kostas Kostarelos
- Nanomedicine
Lab, National Graphene Institute and Faculty of Biology, Medicine
& Health, The University of Manchester, Manchester, M13 9PT, United Kingdom
- Catalan
Institute of Nanoscience and Nanotechnology (ICN2), Barcelona, 08193, Spain
| |
Collapse
|
13
|
Gee K, Isani MA, Fode A, Maselli KM, Zuber SM, Fowler KL, Squillaro AI, Nucho LMA, Grikscheit TC. Spleen Organoid Units Generate Functional Human and Mouse Tissue-Engineered Spleen in a Murine Model. Tissue Eng Part A 2020; 26:411-418. [DOI: 10.1089/ten.tea.2019.0178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Kristin Gee
- Division of Pediatric Surgery and Developmental Biology and Regenerative Medicine, Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
| | - Mubina A. Isani
- Division of Pediatric Surgery and Developmental Biology and Regenerative Medicine, Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
| | - Alexa Fode
- Division of Pediatric Surgery and Developmental Biology and Regenerative Medicine, Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
| | - Kathryn M. Maselli
- Division of Pediatric Surgery and Developmental Biology and Regenerative Medicine, Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
| | - Samuel M. Zuber
- Division of Pediatric Surgery and Developmental Biology and Regenerative Medicine, Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
| | - Kathryn L. Fowler
- Division of Pediatric Surgery and Developmental Biology and Regenerative Medicine, Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
| | - Anthony I. Squillaro
- Division of Pediatric Surgery and Developmental Biology and Regenerative Medicine, Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
| | - Laura-Marie A. Nucho
- Division of Pediatric Surgery and Developmental Biology and Regenerative Medicine, Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
| | - Tracy C. Grikscheit
- Division of Pediatric Surgery and Developmental Biology and Regenerative Medicine, Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California
| |
Collapse
|
14
|
Zhou B, Zhao Z, Zhang X, Deng W, Li Y. Effect of Allogenic Bone Marrow Mesenchymal Stem Cell Transplantation on T Cells of Old Mice. Cell Reprogram 2019; 22:30-35. [PMID: 31895587 DOI: 10.1089/cell.2019.0055] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
To evaluate age-related changes in T cells and stem cell-related genes in mice and the changes in T cells and stem cell-related genes after allogenic bone marrow mesenchymal stem cell (BMSC) transplantation, BALB/c mice were divided into young (2 months, n = 5) and old (20 months, n = 5) groups and 1 × 107 BMSCs from 3-week-old C57BL/6J mice were injected into the old mice (n = 5). T lymphocytes including CD3+, CD8+, CD8+CD28+, and CD8+CD44lowCD62LhighSca-1+ stem cell-like memory T cells from spleens were analyzed by flow cytometry. mRNA transcriptions of the tumor suppressor p16INK4A and the senescence inhibiting AUF 1 and stem cell-related ADAM12, GIL3, c-MYC, NANOG, Wnt, HOX11, Sox2, Oct3/4, and KLF4 genes were analyzed by quantitative reverse transcription-polymerase chain reaction for comparison between young and old mice and old mice after BMSC application. Stem cell-related genes were reduced transcribed in old mice, an action that could be partly or completely reversed for some genes by BMSC injections. The proportion of CD8+CD28+ T cells in the spleens of old mice was significantly reduced (p < 0.01), indicating advanced proliferative T cell senescence. The CD8+CD44lowCD62Lhigh cell fraction was significantly reduced and that of CD8+CD44lowCD62LhighSca-1+ increased in splenic CD8+ cells of old mice, both actions of which were reversed by BMSC injections. p16INK4A transcription was enhanced and AUF1 transcription was reduced in old mice, the latter effect partly reversed by BMSC injections. BMSC injections led to recovery of stem cell-related gene activation or BMSC stem cell-related gene expression tolerance in spleens of old mice.
Collapse
Affiliation(s)
- Biyu Zhou
- Department of Plastic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhenmin Zhao
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China
| | - Xiangyu Zhang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenbin Deng
- Department of Orthopedics, Yang Si Hospital, Shanghai, China
| | - Yan Li
- International Medical Plastic and Cosmetic Centre, China Meitan General Hospital, Beijing, China
| |
Collapse
|
15
|
Min B, Park M, Jeon K, Park JS, Seo H, Jeong S, Kang YK. Age-associated bimodal transcriptional drift reduces intergenic disparities in transcription. Aging (Albany NY) 2019; 10:789-807. [PMID: 29706608 PMCID: PMC5940109 DOI: 10.18632/aging.101428] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 04/20/2018] [Indexed: 02/03/2023]
Abstract
This study addressed the question of how well the quantitative transcriptome structure established in early life is maintained and how consistently it appears with increasing age, and if there is age-associated alteration of gene expression (A3GE), how much influence the Huntington’s disease (HD) genotype exerts on it. We examined 285 exonic sequences of 175 genes using targeted PCR sequencing in skeletal muscle, brain, and splenic CD4+ T cells of wild-type and HD mice. In contrast to the muscle and brain, T cells exhibited large A3GE, suggesting a strong contribution to functional decline of the organism. This A3GE was markedly intensified in age-matched HD T cells, which exhibited accelerated aging as determined by reduced telomere length. Regression analysis suggested that gene expression levels change at a rate of approximately 3% per month with age. We found a bimodal relationship in A3GE in T cells in that weakly expressed genes in young mice were increasingly transcribed in older animals whereas highly expressed genes in the young were decreasingly expressed with age. This bimodal transcriptional drift in the T cell transcriptome data causes the differences in transcription rate between genes to progressively reduce with age.
Collapse
Affiliation(s)
- Byungkuk Min
- Development and Differentiation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Yuseong-gu, Daejeon 34141, South Korea
| | - Myungsun Park
- Development and Differentiation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Yuseong-gu, Daejeon 34141, South Korea
| | - Kyuheum Jeon
- Development and Differentiation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Yuseong-gu, Daejeon 34141, South Korea
| | - Jung Sun Park
- Development and Differentiation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Yuseong-gu, Daejeon 34141, South Korea
| | - Hyemyung Seo
- Department of Molecular and Life Sciences, Hanyang University, Sangnok-gu, Ansan, Gyeonggi-do 15588, South Korea
| | - Sangkyun Jeong
- Mibyeong Research Center, Korea Institute of Oriental Medicine (KIOM), Daejeon 305-811, Korea
| | - Yong-Kook Kang
- Development and Differentiation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Yuseong-gu, Daejeon 34141, South Korea
| |
Collapse
|
16
|
Characterization of immune cell subtypes in three commonly used mouse strains reveals gender and strain-specific variations. J Transl Med 2019; 99:93-106. [PMID: 30353130 PMCID: PMC6524955 DOI: 10.1038/s41374-018-0137-1] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 08/22/2018] [Accepted: 09/12/2018] [Indexed: 12/21/2022] Open
Abstract
The lack of consensus on bone marrow (BM) and splenic immune cell profiles in preclinical mouse strains complicates comparative analysis across different studies. Although studies have documented relative distribution of immune cells from peripheral blood in mice, similar studies for BM and spleen from naïve mice are lacking. In an effort to establish strain- and gender-specific benchmarks for distribution of various immune cell subtypes in these organs, we performed immunophenotypic analysis of BM cells and splenocytes from both genders of three commonly used murine strains (C57BL/6NCr, 129/SvHsd, and BALB/cAnNCr). Total neutrophils and splenic macrophages were significantly higher in C57BL/6NCr, whereas total B cells were lower. Within C57BL/6NCr female mice, BM B cells were elevated with respect to the males whereas splenic mDCs and splenic neutrophils were reduced. Within BALB/cAnNCr male mice, BM CD4+ Tregs were elevated with respect to the other strains. Furthermore, in male BALB/cAnNCr mice, NK cells were elevated with respect to the other strains in both BM and spleen. Splenic CD4+ Tregs and splenic CD8+ T cells were reduced in male BALB/c mice in comparison to female mice. Bone marrow CD4+ T cells and mDCs were significantly increased in 129/SvHsd whereas splenic CD8+ T cells were reduced. In general, males exhibited higher immature myeloid cells, macrophages, and NK cells. To our knowledge, this study provides a first attempt to systematically establish organ-specific benchmarks on immune cells in studies involving these mouse strains.
Collapse
|
17
|
Zhang J, Kaiser MG, Deist MS, Gallardo RA, Bunn DA, Kelly TR, Dekkers JCM, Zhou H, Lamont SJ. Transcriptome Analysis in Spleen Reveals Differential Regulation of Response to Newcastle Disease Virus in Two Chicken Lines. Sci Rep 2018; 8:1278. [PMID: 29352240 PMCID: PMC5775430 DOI: 10.1038/s41598-018-19754-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 01/05/2018] [Indexed: 01/13/2023] Open
Abstract
Enhancing genetic resistance of chickens to Newcastle Disease Virus (NDV) provides a promising way to improve poultry health, and to alleviate poverty and food insecurity in developing countries. In this study, two inbred chicken lines with different responses to NDV, Fayoumi and Leghorn, were challenged with LaSota NDV strain at 21 days of age. Through transcriptome analysis, gene expression in spleen at 2 and 6 days post-inoculation was compared between NDV-infected and control groups, as well as between chicken lines. At a false discovery rate <0.05, Fayoumi chickens, which are relatively more resistant to NDV, showed fewer differentially expressed genes (DEGs) than Leghorn chickens. Several interferon-stimulated genes were identified as important DEGs regulating immune response to NDV in chicken. Pathways predicted by IPA analysis, such as "EIF-signaling", "actin cytoskeleton organization nitric oxide production" and "coagulation system" may contribute to resistance to NDV in Fayoumi chickens. The identified DEGs and predicted pathways may contribute to differential responses to NDV between the two chicken lines and provide potential targets for breeding chickens that are more resistant to NDV.
Collapse
Affiliation(s)
- Jibin Zhang
- Department of Animal Science, Iowa State University, 806 Stange Rd, 2255 Kildee Hall, Ames, IA, 50011, USA
| | - Michael G Kaiser
- Department of Animal Science, Iowa State University, 806 Stange Rd, 2255 Kildee Hall, Ames, IA, 50011, USA
| | - Melissa S Deist
- Department of Animal Science, Iowa State University, 806 Stange Rd, 2255 Kildee Hall, Ames, IA, 50011, USA
| | - Rodrigo A Gallardo
- Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - David A Bunn
- Department of Animal Science, University of California, Davis, CA, 95616, USA
| | - Terra R Kelly
- One Health Institute, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Jack C M Dekkers
- Department of Animal Science, Iowa State University, 806 Stange Rd, 2255 Kildee Hall, Ames, IA, 50011, USA
| | - Huaijun Zhou
- Department of Animal Science, University of California, Davis, CA, 95616, USA
| | - Susan J Lamont
- Department of Animal Science, Iowa State University, 806 Stange Rd, 2255 Kildee Hall, Ames, IA, 50011, USA.
| |
Collapse
|
18
|
Gerzanich V, Makar TK, Guda PR, Kwon MS, Stokum JA, Woo SK, Ivanova S, Ivanov A, Mehta RI, Morris AB, Bryan J, Bever CT, Simard JM. Salutary effects of glibenclamide during the chronic phase of murine experimental autoimmune encephalomyelitis. J Neuroinflammation 2017; 14:177. [PMID: 28865458 PMCID: PMC5581426 DOI: 10.1186/s12974-017-0953-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Accepted: 08/27/2017] [Indexed: 01/03/2023] Open
Abstract
Background In multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE), inflammation is perpetuated by both infiltrating leukocytes and astrocytes. Recent work implicated SUR1-TRPM4 channels, expressed mostly by astrocytes, in murine EAE. We tested the hypothesis that pharmacological inhibition of SUR1 during the chronic phase of EAE would be beneficial. Methods EAE was induced in mice using myelin oligodendrocyte glycoprotein (MOG) 35–55. Glibenclamide (10 μg/day) was administered beginning 12 or 24 days later. The effects of treatment were determined by clinical scoring and tissue examination. Drug within EAE lesions was identified using bodipy-glibenclamide. The role of SUR1-TRPM4 in primary astrocytes was characterized using patch clamp and qPCR. Demyelinating lesions from MS patients were studied by immunolabeling and immunoFRET. Results Administering glibenclamide beginning 24 days after MOG35–55 immunization, well after clinical symptoms had plateaued, improved clinical scores, reduced myelin loss, inflammation (CD45, CD20, CD3, p65), and reactive astrocytosis, improved macrophage phenotype (CD163), and decreased expression of tumor necrosis factor (TNF), B-cell activating factor (BAFF), chemokine (C-C motif) ligand 2 (CCL2) and nitric oxide synthase 2 (NOS2) in lumbar spinal cord white matter. Glibenclamide accumulated within EAE lesions, and had no effect on leukocyte sequestration. In primary astrocyte cultures, activation by TNF plus IFNγ induced de novo expression of SUR1-TRPM4 channels and upregulated Tnf, Baff, Ccl2, and Nos2 mRNA, with glibenclamide blockade of SUR1-TRPM4 reducing these mRNA increases. In demyelinating lesions from MS patients, astrocytes co-expressed SUR1-TRPM4 and BAFF, CCL2, and NOS2. Conclusions SUR1-TRPM4 may be a druggable target for disease modification in MS.
Collapse
Affiliation(s)
- Volodymyr Gerzanich
- Department of Neurosurgery, University of Maryland School of Medicine, 22 S. Greene St., Suite S12D, Baltimore, MD, 21201-1595, USA
| | - Tapas K Makar
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.,Research Service and MS Center of Excellence, Veterans Affairs Maryland Health Care System, Baltimore, MD, 21201, USA
| | - Poornachander Reddy Guda
- Research Service and MS Center of Excellence, Veterans Affairs Maryland Health Care System, Baltimore, MD, 21201, USA
| | - Min Seong Kwon
- Department of Neurosurgery, University of Maryland School of Medicine, 22 S. Greene St., Suite S12D, Baltimore, MD, 21201-1595, USA
| | - Jesse A Stokum
- Department of Neurosurgery, University of Maryland School of Medicine, 22 S. Greene St., Suite S12D, Baltimore, MD, 21201-1595, USA
| | - Seung Kyoon Woo
- Department of Neurosurgery, University of Maryland School of Medicine, 22 S. Greene St., Suite S12D, Baltimore, MD, 21201-1595, USA
| | - Svetlana Ivanova
- Department of Neurosurgery, University of Maryland School of Medicine, 22 S. Greene St., Suite S12D, Baltimore, MD, 21201-1595, USA
| | - Alexander Ivanov
- Department of Neurosurgery, University of Maryland School of Medicine, 22 S. Greene St., Suite S12D, Baltimore, MD, 21201-1595, USA
| | - Rupal I Mehta
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Alexandra Brooke Morris
- Research Service and MS Center of Excellence, Veterans Affairs Maryland Health Care System, Baltimore, MD, 21201, USA
| | - Joseph Bryan
- Pacific Northwest Diabetes Research Institute, 720 Broadway, Seattle, WA, 98122, USA
| | - Christopher T Bever
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.,Research Service and MS Center of Excellence, Veterans Affairs Maryland Health Care System, Baltimore, MD, 21201, USA
| | - J Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, 22 S. Greene St., Suite S12D, Baltimore, MD, 21201-1595, USA. .,Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA. .,Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA. .,Neurosurgical Service, Veterans Affairs Maryland Health Care System, Baltimore, MD, 21201, USA.
| |
Collapse
|
19
|
Kieffer C, Ladinsky MS, Ninh A, Galimidi RP, Bjorkman PJ. Longitudinal imaging of HIV-1 spread in humanized mice with parallel 3D immunofluorescence and electron tomography. eLife 2017; 6. [PMID: 28198699 PMCID: PMC5338924 DOI: 10.7554/elife.23282] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 02/13/2017] [Indexed: 02/06/2023] Open
Abstract
Dissemination of HIV-1 throughout lymphoid tissues leads to systemic virus spread following infection. We combined tissue clearing, 3D-immunofluorescence, and electron tomography (ET) to longitudinally assess early HIV-1 spread in lymphoid tissues in humanized mice. Immunofluorescence revealed peak infection density in gut at 10–12 days post-infection when blood viral loads were low. Human CD4+ T-cells and HIV-1–infected cells localized predominantly to crypts and the lower third of intestinal villi. Free virions and infected cells were not readily detectable by ET at 5-days post-infection, whereas HIV-1–infected cells surrounded by pools of free virions were present in ~10% of intestinal crypts by 10–12 days. ET of spleen revealed thousands of virions released by individual cells and discreet cytoplasmic densities near sites of prolific virus production. These studies highlight the importance of multiscale imaging of HIV-1–infected tissues and are adaptable to other animal models and human patient samples. DOI:http://dx.doi.org/10.7554/eLife.23282.001
Collapse
Affiliation(s)
- Collin Kieffer
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Mark S Ladinsky
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Allen Ninh
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Rachel P Galimidi
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Pamela J Bjorkman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| |
Collapse
|
20
|
Fenton JI, Gurzell EA, Davidson EA, Harris WS. Red blood cell PUFAs reflect the phospholipid PUFA composition of major organs. Prostaglandins Leukot Essent Fatty Acids 2016; 112:12-23. [PMID: 27637336 DOI: 10.1016/j.plefa.2016.06.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 06/26/2016] [Accepted: 06/28/2016] [Indexed: 02/02/2023]
Abstract
Numerous clinical trials examining the use of omega-3 long chain polyunsaturated fatty acids (n-3 LCPUFAs) on various health outcomes have been conducted, and fish oil remains one of the most widely used nutritional supplements. More recently, studies have begun to utilize the omega-3 index, defined as the sum of EPA+DHA in red blood cells (RBCs), as both a biomarker of n-3 LCPUFA exposure and a potential risk factor for coronary heart disease (CHD). Considerably less research evaluates whether RBC phospholipid fatty acids reflect the phospholipid fatty acid composition of other tissues across increasing intakes of n-3 LCPUFAs. We fed mice diets containing increasing amounts of EPA+DHA, equivalent to current recommendations by the American Heart Association on a percent of energy basis, and analyzed the phospholipid fatty acid composition of various tissues in relation to RBCs. We observed that RBCs, heart, muscle, spleen, lung, and adipose tissues all respond to dietary supplementation with EPA+DHA with increasing n-3 LCPUFA and decreasing n-6 LCPUFA levels. Furthermore, the n-3 LCPUFA profiles of all measured tissues had strong (r>0.7) and significant (p<0.001) correlations to RBCs. Interestingly, we also observed changes in saturated fatty acid (SFA) and monounsaturated fatty acid (MUFA) levels across various tissues in response to increased EPA+DHA intakes despite there being no change in dietary SFA and MUFA. Specifically, there were increases in RBC SFA and spleen MUFA and decreases in heart MUFA. These demonstrate that the RBC, including the omega-3 index, may serve as a marker for the relative levels of n-3 and n-6 LCPUFAs in phospholipids of certain tissues.
Collapse
Affiliation(s)
- Jenifer I Fenton
- Department of Food Science and Human Nutrition, Michigan State University, United States; College of Osteopathic Medicine, East Lansing, MI, United States.
| | - Eric A Gurzell
- Department of Food Science and Human Nutrition, Michigan State University, United States
| | - Emily A Davidson
- Department of Food Science and Human Nutrition, Michigan State University, United States
| | - William S Harris
- Sanford School of Medicine, The University of South Dakota, and OmegaQuant, LLC, Sioux Falls, SD, United States
| |
Collapse
|
21
|
Thema N, Pretorius A, Tshilwane SI, Liebenberg J, Steyn H, Van Kleef M. Cellular immune responses induced <i>in vitro</i> by <i>Ehrlichia ruminantium</i> secreted proteins and identification of vaccine candidate peptides. ACTA ACUST UNITED AC 2016; 83:e1-e11. [PMID: 27608502 PMCID: PMC6238801 DOI: 10.4102/ojvr.v83i1.1170] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 04/12/2016] [Accepted: 04/14/2016] [Indexed: 01/01/2023]
Abstract
Secreted proteins are reported to induce cell-mediated immunity characterised by the production of interferon-gamma (IFN)-γ. In this study three open reading frames (ORFs) (Erum8060, Erum7760, Erum5000) encoding secreted proteins were selected from the Ehrlichia ruminantium (Welgevonden) genome sequence using bioinformatics tools to determine whether they induce a cellular immune response in vitro with mononuclear cells from needle and tick infected animals. The whole recombinant protein of the three ORFs as well as four adjacent fragments of the Erum5000 protein (Erum5000A, Erum5000B, Erum5000C, Erum5000D) were successfully expressed in a bacterial expression system which was confirmed by immunoblots using anti-His antibodies and sheep sera. These recombinant proteins were assayed with immune sheep and cattle peripheral blood mononuclear cells (PBMCs), spleen and lymph node (LN) cells to determine whether they induce recall cellular immune responses in vitro. Significant proliferative responses and IFN-γ production were evident for all recombinant proteins, especially Erum5000A, in both ruminant species tested. Thus overlapping peptides spanning Erum5000A were synthesised and peptides that induce proliferation of memory CD4+ and CD8+ T cells and production of IFN-γ were identified. These results illustrate that a Th1 type immune response was elicited and these recombinant proteins and peptides may therefore be promising candidates for development of a heartwater vaccine.
Collapse
Affiliation(s)
- Nontobeko Thema
- New Generation Vaccines Programme, Agricultural Research Council-Onderstepoort Veterinary Institute; Department of Veterinary Tropical Diseases, University of Pretoria.
| | | | | | | | | | | |
Collapse
|
22
|
Mangar C, Armitage CW, Timms P, Corcoran LM, Beagley KW. Characterisation of CD4 T cells in healthy and diseased koalas (Phascolarctos cinereus) using cell-type-specific monoclonal antibodies. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 60:80-90. [PMID: 26905635 DOI: 10.1016/j.dci.2016.02.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 02/16/2016] [Accepted: 02/16/2016] [Indexed: 06/05/2023]
Abstract
The koala (Phascolarctos cinereus) is an arboreal herbivorous marsupial that is an Australian icon. Koalas in many parts of Australia are under multiple threats including habitat destruction, dog attacks, vehicular accidents, and infectious diseases such as Chlamydia spp. and the koala retrovirus (KoRV), which may contribute to the incidence of lymphoma and leukaemia in this species. Due to a lack of koala-specific immune reagents and assays there is currently no way to adequately analyse the immune response in healthy, diseased or vaccinated animals. This paper reports the production and characterisation of the first anti-koala CD4 monoclonal antibody (mAb). The koala CD4 gene was identified and used to develop recombinant proteins for mAb production. Fluorochrome-conjugated anti-CD4 mAb was used to measure the levels of CD4(+) lymphocytes collected from koala spleens (41.1%, range 20-45.1%) lymph nodes (36.3%, range 19-55.9%) and peripheral blood (23.8%, range 17.3-35%) by flow cytometry. Biotin-conjugated anti-CD4 mAb was used for western blot to determine an approximate size of 52 kDa for the koala CD4 molecule and used in immunohistochemistry to identify CD4(+) cells in the paracortical region and germinal centres of spleen and lymph nodes. Using the anti-CD4 mab we showed that CD4 cells from vaccinated, but not control, koalas proliferated following in vitro stimulation with UV-inactivated Chlamydia pecorum and recombinant chlamydial antigens. Since CD4(+) T cells have been shown to play a pivotal role in clearing chlamydial infection in both human and mouse infections, using this novel antibody will help determine the role CD4(+) T cells play in protection against chlamydial infection in koalas and also enhance our knowledge of how KoRV affects the koala immune system.
Collapse
Affiliation(s)
- Chandan Mangar
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), P.O Box 4059, Kelvin Grove, Queensland, Australia
| | - Charles W Armitage
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), P.O Box 4059, Kelvin Grove, Queensland, Australia
| | - Peter Timms
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), P.O Box 4059, Kelvin Grove, Queensland, Australia; Faculty of Science, Health, Education and Engineering, University of Sunshine Coast (USC), P.O Box, 4556, Sippy Downs, Queensland, Australia
| | - Lynn M Corcoran
- The Walter Eliza Hall Institute of Medical Research, 1G Royal Parade, 3052, The University of Melbourne, Parkville, Victoria, Australia
| | - Kenneth W Beagley
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), P.O Box 4059, Kelvin Grove, Queensland, Australia.
| |
Collapse
|
23
|
Abd-Elhakim YM, Mohamed AAR, Mohamed WA. Hemato-immunologic impact of subchronic exposure to melamine and/or formaldehyde in mice. J Immunotoxicol 2016; 13:713-22. [DOI: 10.3109/1547691x.2016.1170742] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
| | | | - Wafaa A. Mohamed
- Department of Clinical Pathology, Zagazig University, Zagazig, Egypt
| |
Collapse
|
24
|
Clonal evolution of CD8+ T cell responses against latent viruses: relationship among phenotype, localization, and function. J Virol 2014; 89:568-80. [PMID: 25339770 DOI: 10.1128/jvi.02003-14] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Human cytomegalovirus (hCMV) infection is characterized by a vast expansion of resting effector-type virus-specific T cells in the circulation. In mice, interleukin-7 receptor α (IL-7Rα)-expressing cells contain the precursors for long-lived antigen-experienced CD8(+) T cells, but it is unclear if similar mechanisms operate to maintain these pools in humans. Here, we studied whether IL-7Rα-expressing cells obtained from peripheral blood (PB) or lymph nodes (LNs) sustain the circulating effector-type hCMV-specific pool. Using flow cytometry and functional assays, we found that the IL-7Rα(+) hCMV-specific T cell population comprises cells that have a memory phenotype and lack effector features. We used next-generation sequencing of the T cell receptor to compare the clonal repertoires of IL-7Rα(+) and IL-7Rα(-) subsets. We observed limited overlap of clones between these subsets during acute infection and after 1 year. When we compared the hCMV-specific repertoire between PB and paired LNs, we found many identical clones but also clones that were exclusively found in either compartment. New clones that were found in PB during antigenic recall were only rarely identical to the unique LN clones. Thus, although PB IL-7Rα-expressing and LN hCMV-specific CD8(+) T cells show typical traits of memory-type cells, these populations do not seem to contain the precursors for the novel hCMV-specific CD8(+) T cell pool during latency or upon antigen recall. IL-7Rα(+) PB and LN hCMV-specific memory cells form separate virus-specific compartments, and precursors for these novel PB hCMV-specific CD8(+) effector-type T cells are possibly located in other secondary lymphoid tissues or are being recruited from the naive CD8(+) T cell pool. IMPORTANCE Insight into the self-renewal properties of long-lived memory CD8(+) T cells and their location is crucial for the development of both passive and active vaccination strategies. Human CMV infection is characterized by a vast expansion of resting effector-type cells. It is, however, not known how this population is maintained. We here investigated two possible compartments for effector-type cell precursors: circulating acute-phase IL-7Rα-expressing hCMV-specific CD8(+) T cells and lymph node (LN)-residing hCMV-specific (central) memory cells. We show that new clones that appear after primary hCMV infection or during hCMV reactivation seldom originate from either compartment. Thus, although identical clones may be maintained by either memory population, the precursors of the novel clones are probably located in other (secondary) lymphoid tissues or are recruited from the naive CD8(+) T cell pool.
Collapse
|
25
|
Marchese M, Cowan D, Head E, Ma D, Karimi K, Ashthorpe V, Kapadia M, Zhao H, Davis P, Sakic B. Autoimmune manifestations in the 3xTg-AD model of Alzheimer's disease. J Alzheimers Dis 2014; 39:191-210. [PMID: 24150111 DOI: 10.3233/jad-131490] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Immune system activation is frequently reported in patients with Alzheimer's disease (AD). However, it remains unknown whether this is a cause, a consequence, or an epiphenomenon of brain degeneration. OBJECTIVE The present study examines whether immunological abnormalities occur in a well-established murine AD model and if so, how they relate temporally to behavioral deficits and neuropathology. METHODS A broad battery of tests was employed to assess behavioral performance and autoimmune/inflammatory markers in 3xTg-AD (AD) mice and wild type controls from 1.5 to 12 months of age. RESULTS Aged AD mice displayed severe manifestations of systemic autoimmune/inflammatory dise6ase, as evidenced by splenomegaly, hepatomegaly, elevated serum levels of anti-nuclear/anti-dsDNA antibodies, low hematocrit, and increased number of double-negative T splenocytes. However, anxiety-related behavior and altered spleen function were evident as early as 2 months of age, thus preceding typical AD-like brain pathology. Moreover, AD mice showed altered olfaction and impaired "cognitive" flexibility in the first 6 months of life, suggesting mild cognitive impairment-like manifestations before general learning/memory impairments emerged at an older age. Interestingly, all of these features were present in 3xTg-AD mice prior to significant amyloid-β or tau pathology. CONCLUSION The results indicate that behavioral deficits in AD mice develop in parallel with systemic autoimmune/inflammatory disease. These changes antedate AD-like neuropathology, thus supporting a causal link between autoimmunity and aberrant behavior. Consequently, 3xTg-AD mice may be a useful model in elucidating the role of immune system in the etiology of AD.
Collapse
Affiliation(s)
- Monica Marchese
- Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, ON, Canada
| | - David Cowan
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Elizabeth Head
- Department of Molecular & Biomedical Pharmacology, Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Donglai Ma
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Khalil Karimi
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | | | - Minesh Kapadia
- Department of Psychiatry & Behavioral Neurosciences, McMaster University, Hamilton, ON, Canada
| | - Hui Zhao
- Department of Psychiatry & Behavioral Neurosciences, McMaster University, Hamilton, ON, Canada
| | - Paulina Davis
- Department of Molecular & Biomedical Pharmacology, Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Boris Sakic
- Department of Psychiatry & Behavioral Neurosciences, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
26
|
Terahara K, Ishige M, Ikeno S, Mitsuki YY, Okada S, Kobayashi K, Tsunetsugu-Yokota Y. Expansion of activated memory CD4+ T cells affects infectivity of CCR5-tropic HIV-1 in humanized NOD/SCID/JAK3null mice. PLoS One 2013; 8:e53495. [PMID: 23301078 PMCID: PMC3534664 DOI: 10.1371/journal.pone.0053495] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 11/29/2012] [Indexed: 12/18/2022] Open
Abstract
Humanized mice reconstituted with human hematopoietic cells have been developed as an experimental animal model for human immunodeficiency virus type 1 (HIV-1) infection. Myeloablative irradiation is usually performed to augment the engraftment of donor hematopoietic stem cells (HSCs) in recipient mice; however, some mouse strains are susceptible to irradiation, making longitudinal analysis difficult. We previously attempted to construct humanized NOD/SCID/JAK3null (hNOJ) mice, which were not irradiated prior to human HSC transplantation. We found that, over time, many of the reconstituted CD4+ T cells expanded with an activated effector memory phenotype. Therefore, the present study used hNOJ mice that were irradiated (hNOJ (IR+)) or not (hNOJ (IR−)) prior to human HSC transplantation to examine whether the development and cellularity of the reconstituted CD4+ T cells were influenced by the degree of chimerism, and whether they affected HIV-1 infectivity. Indeed, hNOJ (IR+) mice showed a greater degree of chimerism than hNOJ (IR−) mice. However, the conversion of CD4+ T cells to an activated effector memory phenotype, with a high percentage of cells showing Ki-67 expression, occurred in both hNOJ (IR+) and hNOJ (IR−) mice, probably as a result of lymphopenia-induced homeostatic expansion. Furthermore, when hNOJ (IR+) and hNOJ (IR−) mice, which were selected as naïve- and memory CD4+ T cell subset-rich groups, respectively, were infected with CCR5-tropic HIV-1 in vivo, virus replication (as assessed by the plasma viral load) was delayed; however, the titer subsequently reached a 1-log higher level in memory-rich hNOJ (IR−) mice than in naïve-rich hNOJ (IR+) mice, indicating that virus infectivity in hNOJ mice was affected by the different status of the reconstituted CD4+ T cells. Therefore, the hNOJ mouse model should be used selectively, i.e., according to the specific experimental objectives, to gain an appropriate understanding of HIV-1 infection/pathogenesis.
Collapse
Affiliation(s)
- Kazutaka Terahara
- Department of Immunology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Masayuki Ishige
- Department of Immunology, National Institute of Infectious Diseases, Tokyo, Japan
- Division of Hematopoiesis, Center for AIDS Research, Kumamoto University, Kumamoto, Japan
| | - Shota Ikeno
- Department of Immunology, National Institute of Infectious Diseases, Tokyo, Japan
- Laboratory of Viral Infection II, Kitasato Institute for Life Science, Kitasato University, Tokyo, Japan
| | - Yu-ya Mitsuki
- Department of Immunology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Seiji Okada
- Division of Hematopoiesis, Center for AIDS Research, Kumamoto University, Kumamoto, Japan
| | - Kazuo Kobayashi
- Department of Immunology, National Institute of Infectious Diseases, Tokyo, Japan
| | | |
Collapse
|
27
|
SIGN-R1, a C-type lectin, enhances apoptotic cell clearance through the complement deposition pathway by interacting with C1q in the spleen. Cell Death Differ 2012; 20:535-45. [PMID: 23238564 DOI: 10.1038/cdd.2012.160] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Complements, such as C1q and C3, and macrophages in the splenic marginal zone (MZMs) play pivotal roles in the efficient uptake and processing of circulating apoptotic cells. SIGN-R1, a C-type lectin that is highly expressed in a subpopulation of MZMs, regulates the complement fixation pathway by interacting with C1q, to fight blood-borne Streptococcus pneumoniae. Therefore, we examined whether the SIGN-R1-mediated classical complement pathway plays a role in apoptotic cell clearance and immune tolerance. SIGN-R1 first-bound apoptotic cells and this binding was significantly enhanced in the presence of C1q. SIGN-R1-C1q complex then immediately mediated C3 deposition on circulating apoptotic cells in the MZ, leading to the efficient clearance of them. SIGN-R1-mediated C3 deposition was completely abolished in the spleen of SIGN-R1 knockout (KO) mice. Given that SIGN-R1 is not expressed in the liver, we were struck by the finding that C3-deposited apoptotic cells were still found in the liver of wild-type mice, and dramatically reduced in the SIGN-R1 KO liver. In particular, SIGN-R1 deficiency caused delayed clearance of apoptotic cells and aberrant secretion of cytokines, such as TNF-α, IL-6, and TGF-β in the spleen as well as in the liver. In addition, anti-double- and single-stranded DNA antibody level was significantly increased in SIGN-R1-depleted mice compared with control mice. These findings suggest a novel mechanism of apoptotic cell clearance which is initiated by SIGN-R1 in the MZ and identify an integrated role of SIGN-R1 in the systemic clearance of apoptotic cells, linking the recognition of apoptotic cells, the opsonization of complements, and the induction of immune tolerance.
Collapse
|
28
|
Motkowski R, Michalkiewicz J, Mikoluc B, Smolka-Afifi D, Pietrucha B, Kubiszewska I, Piotrowska-Jastrzebska J, Bernatowska E. Peripheral blood T lymphocyte subsets in children with congenital asplenia. Hum Immunol 2012; 73:1091-7. [PMID: 22902394 DOI: 10.1016/j.humimm.2012.08.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Revised: 07/30/2012] [Accepted: 08/09/2012] [Indexed: 11/30/2022]
Abstract
The aim of the current study was to examine whether a congenital lack of the spleen changes distribution, state of activation and function of peripheral lymphocyte T subsets. Seven children with congenital asplenia (CA) aged 1.5-17 years and seven age-matched controls were tested. By triple-color flow cytometry we examined: (1) the expression of CD3(+), CD4(+), CD8(+), CD19(+), and CD56(+) on lymphocytes; (2) the distribution of CD45RA(+) and CD45RO(+) in CD4(+) and CD8(+); (3) the expression of CD27(+) in the CD4(+) and CD8(+) T-cell-bearing CD45RA(+), CD45RO(+), or CD45RB(+). Lymphocyte proliferative responses and cytokines production (IFN-gamma, IL-6, TNF-alfa, and IL-10) in anti-CD3-induced peripheral blood mononuclear cells were tested. The results indicate (1) a normal distribution of the basic lymphocyte subsets, (2) low CD3(+)/CD8(+) percentage but expressing CD8(+high) and non-significantly elevated CD4(+)/CD8(+) ratio, (3) CD45RA(+high) and CD27(+high) in the CD4(+) and CD8(+) T cell, and (4) CD45RB(+high) in the CD4(+) and CD45RO(+high) in the CD8(+). The distribution of CD27(+) in the CD45RA(+) and CD45RO(+) CD4(+) T cells remained unchanged. However, the percentage of CD8(+)/CD45RO(+)/CD27(+) T cells tended to be elevated. Altogether, these data indicate that CA is connected with (1) the presence CD4(+) T cells expressing the "naive" phenotype (CD45RA(+high) RB(+high) and CD27(+high)), (2) high numbers of activated CD8(+) T cells shifted toward the memory phenotype (CD45RO(+high)) but still showing high CD27(+) expression, which may indicate failure in T CD8(+) cytotoxic effectors differentiation, and (3) a tendency to the rather pro-inflammatory status of cells, low IL-10 expression, and suboptimal lymphocytes responses to mitogenic stimulation.
Collapse
Affiliation(s)
- Radoslaw Motkowski
- Department of Pediatrics and Developmental Disorders of Children and Adolescents, Medical University of Bialystok, Waszyngtona Str. 17, 15-224 Bialystok, Poland.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Properties of end-stage human T cells defined by CD45RA re-expression. Curr Opin Immunol 2012; 24:476-81. [PMID: 22554789 DOI: 10.1016/j.coi.2012.04.001] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Accepted: 04/08/2012] [Indexed: 01/10/2023]
Abstract
Persistent viral infections, inflammatory syndromes and ageing all induce the accumulation of highly differentiated CD45RA re-expressing memory T cells. These cells increase during ageing, especially in individuals who are infected with cytomegalovirus (CMV). These cells have decreased proliferative capacity, increased activation of senescence signalling pathways and greater susceptibility to apoptosis in vitro. However these cells are capable of multiple effector functions and thus bear all the hallmarks of short-lived effector T cells. This indicates that senescence signalling may govern the unique characteristics of effector T cells. In this article, we address the functional and migratory properties of these T cells and mechanisms that are involved in their generation. Finally we assess the potential for manipulation of their activity and whether this may improve immune function during ageing.
Collapse
|
30
|
Takayanagi Y, Osawa S, Ikuma M, Takagaki K, Zhang J, Hamaya Y, Yamada T, Sugimoto M, Furuta T, Miyajima H, Sugimoto K. Norepinephrine suppresses IFN-γ and TNF-α production by murine intestinal intraepithelial lymphocytes via the β₁ adrenoceptor. J Neuroimmunol 2012; 245:66-74. [PMID: 22398028 DOI: 10.1016/j.jneuroim.2012.02.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 01/16/2012] [Accepted: 02/10/2012] [Indexed: 11/28/2022]
Abstract
We examined whether norepinephrine (NE) had direct effects on cytokine production by murine intestinal intraepithelial lymphocytes (IELs), compared with splenocytes. CD3⁺ IELs and CD3⁺ splenocytes expressed α(1B), α(1D), α(2C), β₁, β₂, and β₃ adrenoceptors (ARs). NE significantly suppressed IFN-γ and TNF-α production by IELs and splenocytes ex vivo. The suppressive effects of NE in IELs were reversed by β₁ AR antagonist CGP-20712A, whereas those in splenocytes were reversed by β₂ AR antagonist ICI118,551. In IELs, β₁ AR agonist xamoterol mimicked the suppressive effects of NE. These results indicated NE regulates intestinal mucosal immune responses mediated by IELs via β₁ AR.
Collapse
Affiliation(s)
- Yasuhiro Takayanagi
- First Department of Medicine, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
de Porto APNA, Lammers AJJ, Bennink RJ, ten Berge IJM, Speelman P, Hoekstra JBL. Assessment of splenic function. Eur J Clin Microbiol Infect Dis 2010; 29:1465-73. [PMID: 20853172 PMCID: PMC2995208 DOI: 10.1007/s10096-010-1049-1] [Citation(s) in RCA: 116] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2010] [Accepted: 08/17/2010] [Indexed: 12/12/2022]
Abstract
Hyposplenic patients are at risk of overwhelming post-splenectomy infection (OPSI), which carries mortality of up to 70%. Therefore, preventive measures are warranted. However, patients with diminished splenic function are difficult to identify. In this review we discuss immunological, haematological and scintigraphic parameters that can be used to measure splenic function. IgM memory B cells are a potential parameter for assessing splenic function; however, more studies are necessary for its validation. Detection of Howell-Jolly bodies does not reflect splenic function accurately, whereas determining the percentage of pitted erythrocytes is a well-evaluated method and seems a good first-line investigation for assessing splenic function. When assessing spleen function, (99m)Tc-labelled, heat-altered, autologous erythrocyte scintigraphy with multimodality single photon emission computed tomography (SPECT)-CT technology is the best approach, as all facets of splenic function are evaluated. In conclusion, although scintigraphic methods are most reliable, they are not suitable for screening large populations. We therefore recommend using the percentage of pitted erythrocytes, albeit suboptimal, as a first-line investigation and subsequently confirming abnormal readings by means of scintigraphy. More studies evaluating the value of potentially new markers are needed.
Collapse
Affiliation(s)
- A P N A de Porto
- Department of Infectious Diseases, Tropical Medicine and AIDS, G2-105, Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands.
| | | | | | | | | | | |
Collapse
|
32
|
Selective accumulation of virus-specific CD8+ T cells with unique homing phenotype within the human bone marrow. Blood 2008; 112:3293-302. [PMID: 18635810 DOI: 10.1182/blood-2008-02-138040] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The bone marrow plays a unique role within the immune system. We compared the phenotype and function of virus-specific CD8(+) T cells from matched samples of human peripheral blood and bone marrow. Analysis of virus-specific memory CD8(+) T cells showed widely divergent partition of antigen-specific populations between blood and bone marrow. T cells specific for Epstein-Barr virus (EBV) lytic antigens were enriched 3-fold in marrow compared with blood, whereas the response to EBV latent epitopes was equivalent between the 2 compartments. No difference in EBV viral load or expression of the EBV lytic protein was observed between blood and bone marrow. In direct contrast, although cytomegalo-virus (CMV)-specific T cells were the largest virus-specific population within peripheral blood, they were reduced by 60% within marrow. Bone marrow T cells were found to exhibit a unique CCR5(+)CXCR6(+)CXCR3(-) homing phenotype which has not been observed on T cells from other secondary lymphoid organs or peripheral organs. Expression of CCR5 and CXCR6 was higher on EBV-specific T cells within peripheral blood compared with CMV-specific populations. These observations identify a novel bone marrow homing phenotype for CD8(+) memory T cells, which necessitates a reevaluation of the magnitude of antigen-specific populations within the lymphoid system.
Collapse
|
33
|
Kaplan B, Gangemi A, Thielke J, Oberholzer J, Sankary H, Benedetti E. Successful rescue of refractory, severe antibody mediated rejection with splenectomy. Transplantation 2007; 83:99-100. [PMID: 17220802 DOI: 10.1097/01.tp.0000243739.31440.2b] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Antibody-mediated rejection (AMR) commonly occurs after transplantation of ABO-incompatible and sensitized renal transplant. Treatment regimens commonly include a combination of plasmapheresis (PL) and intravenous immunoglobulin (IVIG). However, some cases of AMR remain refractory to treatment. We report a case series of four patients with AMR refractory to standard therapy (ST) who resolved after splenectomy. Four living donor kidney transplant recipients were diagnosed with AMR. Two patients were ABO incompatible, one was cross-match positive and one had no obvious predisposing factors. After failure of therapy with corticosteroids, PL, IVIG, Thymoglobulin, and Rituximab (three patients) or Campath (one patient), AMR was treated with laparoscopic splenectomy. After an average of 11 days of ST, laparoscopic splenectomy was performed for rescue. The urinary output improved immediately in all patients, serum creatinine levels decreased within 48 hr, and ABO titers fell in the ABO-incompatible patient and the cross-match became negative in the two sensitized patients. Splenectomy may play a role in the treatment of AMR refractory to ST.
Collapse
Affiliation(s)
- Bruce Kaplan
- Division of Transplant Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA.
| | | | | | | | | | | |
Collapse
|