1
|
Nath PR, Maclean M, Nagarajan V, Lee JW, Yakin M, Kumar A, Nadali H, Schmidt B, Kaya KD, Kodati S, Young A, Caspi RR, Kuiper JJW, Sen HN. Single-cell profiling identifies a CD8 bright CD244 bright Natural Killer cell subset that reflects disease activity in HLA-A29-positive birdshot chorioretinopathy. Nat Commun 2024; 15:6443. [PMID: 39085199 PMCID: PMC11291632 DOI: 10.1038/s41467-024-50472-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 07/12/2024] [Indexed: 08/02/2024] Open
Abstract
Birdshot chorioretinopathy is an inflammatory eye condition strongly associated with MHC-I allele HLA-A29. The striking association with MHC-I suggests involvement of T cells, whereas natural killer (NK) cell involvement remains largely unstudied. Here we show that HLA-A29-positive birdshot chorioretinopathy patients have a skewed NK cell pool containing expanded CD16 positive NK cells which produce more proinflammatory cytokines. These NK cells contain populations that express CD8A which is involved in MHC-I recognition on target cells, display gene signatures indicative of high cytotoxic activity (GZMB, PRF1 and ISG15), and signaling through NK cell receptor CD244 (SH2D1B). Long-term monitoring of a cohort of birdshot chorioretinopathy patients with active disease identifies a population of CD8bright CD244bright NK cells, which rapidly declines to normal levels upon clinical remission following successful treatment. Collectively, these studies implicate CD8bright CD244bright NK cells in birdshot chorioretinopathy.
Collapse
Affiliation(s)
- Pulak R Nath
- Clinical and Translational Immunology Unit, Laboratory of Immunology, NEI, NIH, Bethesda, USA.
- Lentigen Technology Inc., A Miltenyi Biotec Company, 910 Clopper Road, Gaithersburg, MD, 20878, USA.
| | - Mary Maclean
- Clinical and Translational Immunology Unit, Laboratory of Immunology, NEI, NIH, Bethesda, USA
- Translational Immunology Section, Office of Science and Technology, NIAMS, Bethesda, NIH, USA
| | - Vijay Nagarajan
- Clinical and Translational Immunology Unit, Laboratory of Immunology, NEI, NIH, Bethesda, USA
- Immunoregulation Section, Laboratory of Immunology, NEI, NIH, Bethesda, USA
| | - Jung Wha Lee
- Clinical and Translational Immunology Unit, Laboratory of Immunology, NEI, NIH, Bethesda, USA
| | - Mehmet Yakin
- Clinical and Translational Immunology Unit, Laboratory of Immunology, NEI, NIH, Bethesda, USA
| | - Aman Kumar
- Clinical and Translational Immunology Unit, Laboratory of Immunology, NEI, NIH, Bethesda, USA
| | - Hadi Nadali
- Clinical and Translational Immunology Unit, Laboratory of Immunology, NEI, NIH, Bethesda, USA
| | - Brian Schmidt
- NIH Intramural Sequencing Center, NIH, Rockville, USA
| | - Koray D Kaya
- Medical Genetics and Ophthalmic Genomics Unit, NEI, NIH, Bethesda, USA
| | - Shilpa Kodati
- Clinical and Translational Immunology Unit, Laboratory of Immunology, NEI, NIH, Bethesda, USA
| | - Alice Young
- NIH Intramural Sequencing Center, NIH, Rockville, USA
| | - Rachel R Caspi
- Immunoregulation Section, Laboratory of Immunology, NEI, NIH, Bethesda, USA
| | - Jonas J W Kuiper
- Department of Ophthalmology, University Medical Center Utrecht, University of Utrecht, Utrecht, Netherlands.
| | - H Nida Sen
- Clinical and Translational Immunology Unit, Laboratory of Immunology, NEI, NIH, Bethesda, USA
| |
Collapse
|
2
|
Shi J, Yin W, Chen W. Mathematical models of TCR initial triggering. Front Immunol 2024; 15:1411614. [PMID: 39091495 PMCID: PMC11291225 DOI: 10.3389/fimmu.2024.1411614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 07/05/2024] [Indexed: 08/04/2024] Open
Abstract
T cell receptors (TCRs) play crucial roles in regulating T cell response by rapidly and accurately recognizing foreign and non-self antigens. The process involves multiple molecules and regulatory mechanisms, forming a complex network to achieve effective antigen recognition. Mathematical modeling techniques can help unravel the intricate network of TCR signaling and identify key regulators that govern it. In this review, we introduce and briefly discuss relevant mathematical models of TCR initial triggering, with a focus on kinetic proofreading (KPR) models with different modified structures. We compare the topology structures, biological hypotheses, parameter choices, and simulation performance of each model, and summarize the advantages and limitations of them. Further studies on TCR modeling design, aiming for an optimized balance of specificity and sensitivity, are expected to contribute to the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Jiawei Shi
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory for Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Weiwei Yin
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou, China
| | - Wei Chen
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory for Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
- Department of Cell Biology, School of Medicine, Zhejiang University, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
| |
Collapse
|
3
|
Han XQ, Pan YR, Zhong YQ, Tian TT, Liu X, Zhang XJ, Zhang YA. Identification and functional analyses of CD4-1 + cells in grass carp (Ctenopharyngodon idella). FISH & SHELLFISH IMMUNOLOGY 2024; 150:109649. [PMID: 38797336 DOI: 10.1016/j.fsi.2024.109649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 05/29/2024]
Abstract
In mammals, CD4 is found to be expressed on T cells and innate immune cells, however, teleost cells bearing CD4 have not been well identified and characterized. In this study, we identified two different CD4-1+ cell subsets in grass carp (Ctenopharyngodon idella): CD4-1+ lymphocytes (Lym) and CD4-1+ myeloid cells (Mye), both of which had the highest proportions in the head kidney. The mRNA expression analysis showed that CD4-1, CD4-2, TCRβ, CD3γ/δ, and LCK1 are highly expressed in CD4-1+ Lym and also expressed in CD4-1+ Mye. Furthermore, we found that CD4-1+ Lym have a Lym morphology and highly express T-cell cytokines, suggesting that they are CD4+ T cells equivalent to mammalian Th cells. On the other hand, CD4-1+ Mye were found to have a morphology of macrophage and highly express macrophage marker gene MCSFR, indicating that they are macrophages. In addition, functional analysis revealed that CD4-1+ Mye possess phagocytic ability and great antigen-processing ability. Taken together, our study sheds further light on the composition and function of CD4+ cells in teleost fish.
Collapse
Affiliation(s)
- Xue-Qing Han
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Yi-Ru Pan
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Ya-Qin Zhong
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Tian-Tian Tian
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Xun Liu
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Xu-Jie Zhang
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, College of Fisheries, Huazhong Agricultural University, Wuhan, China; Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Wuhan, China; Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China.
| | - Yong-An Zhang
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, College of Fisheries, Huazhong Agricultural University, Wuhan, China; Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Wuhan, China; Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China.
| |
Collapse
|
4
|
Simonson AW, Zeppa JJ, Bucsan AN, Chao MC, Pokkali S, Hopkins F, Chase MR, Vickers AJ, Sutton MS, Winchell CG, Myers AJ, Ameel CL, Kelly R, Krouse B, Hood LE, Li J, Lehman CC, Kamath M, Tomko J, Rodgers MA, Donlan R, Chishti H, Jacob Borish H, Klein E, Scanga CA, Fortune S, Lin PL, Maiello P, Roederer M, Darrah PA, Seder RA, Flynn JL. CD4 T cells and CD8α+ lymphocytes are necessary for intravenous BCG-induced protection against tuberculosis in macaques. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.14.594183. [PMID: 38798646 PMCID: PMC11118459 DOI: 10.1101/2024.05.14.594183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Tuberculosis (TB) is a major cause of morbidity and mortality worldwide despite widespread intradermal (ID) BCG vaccination in newborns. We previously demonstrated that changing the route and dose of BCG vaccination from 5×105 CFU ID to 5×107 CFU intravenous (IV) resulted in prevention of infection and disease in a rigorous, highly susceptible non-human primate model of TB. Identifying the immune mechanisms of protection for IV BCG will facilitate development of more effective vaccines against TB. Here, we depleted select lymphocyte subsets in IV BCG vaccinated macaques prior to Mtb challenge to determine the cell types necessary for that protection. Depletion of CD4 T cells or all CD8α expressing lymphoycytes (both innate and adaptive) resulted in loss of protection in most macaques, concomitant with increased bacterial burdens (~4-5 log10 thoracic CFU) and dissemination of infection. In contrast, depletion of only adaptive CD8αβ+ T cells did not significantly reduce protection against disease. Our results demonstrate that CD4 T cells and innate CD8α+ lymphocytes are critical for IV BCG-induced protection, supporting investigation of how eliciting these cells and their functions can improve future TB vaccines.
Collapse
Affiliation(s)
- Andrew W. Simonson
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine; Pittsburgh, PA, USA
- Center for Vaccine Research, University of Pittsburgh School of Medicine; Pittsburgh, PA, USA
| | - Joseph J. Zeppa
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine; Pittsburgh, PA, USA
- Center for Vaccine Research, University of Pittsburgh School of Medicine; Pittsburgh, PA, USA
| | - Allison N. Bucsan
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH); Bethesda, MD, USA
| | - Michael C. Chao
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health; Boston, MA, USA
| | - Supriya Pokkali
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH); Bethesda, MD, USA
| | - Forrest Hopkins
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health; Boston, MA, USA
| | - Michael R. Chase
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health; Boston, MA, USA
| | - Andrew J. Vickers
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health; Boston, MA, USA
| | - Matthew S. Sutton
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH); Bethesda, MD, USA
| | - Caylin G. Winchell
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine; Pittsburgh, PA, USA
- Center for Vaccine Research, University of Pittsburgh School of Medicine; Pittsburgh, PA, USA
| | - Amy J. Myers
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine; Pittsburgh, PA, USA
- Center for Vaccine Research, University of Pittsburgh School of Medicine; Pittsburgh, PA, USA
| | - Cassaundra L. Ameel
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine; Pittsburgh, PA, USA
- Center for Vaccine Research, University of Pittsburgh School of Medicine; Pittsburgh, PA, USA
| | - Ryan Kelly
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine; Pittsburgh, PA, USA
- Center for Vaccine Research, University of Pittsburgh School of Medicine; Pittsburgh, PA, USA
| | - Ben Krouse
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine; Pittsburgh, PA, USA
- Center for Vaccine Research, University of Pittsburgh School of Medicine; Pittsburgh, PA, USA
| | - Luke E. Hood
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine; Pittsburgh, PA, USA
- Center for Vaccine Research, University of Pittsburgh School of Medicine; Pittsburgh, PA, USA
| | - Jiaxiang Li
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine; Pittsburgh, PA, USA
- Center for Vaccine Research, University of Pittsburgh School of Medicine; Pittsburgh, PA, USA
| | - Chelsea C. Lehman
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH); Bethesda, MD, USA
| | - Megha Kamath
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH); Bethesda, MD, USA
| | - Jaime Tomko
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine; Pittsburgh, PA, USA
- Center for Vaccine Research, University of Pittsburgh School of Medicine; Pittsburgh, PA, USA
| | - Mark A. Rodgers
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine; Pittsburgh, PA, USA
- Center for Vaccine Research, University of Pittsburgh School of Medicine; Pittsburgh, PA, USA
| | - Rachel Donlan
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine; Pittsburgh, PA, USA
- Center for Vaccine Research, University of Pittsburgh School of Medicine; Pittsburgh, PA, USA
| | - Harris Chishti
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine; Pittsburgh, PA, USA
- Center for Vaccine Research, University of Pittsburgh School of Medicine; Pittsburgh, PA, USA
| | - H. Jacob Borish
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine; Pittsburgh, PA, USA
- Center for Vaccine Research, University of Pittsburgh School of Medicine; Pittsburgh, PA, USA
| | - Edwin Klein
- Division of Animal Laboratory Resources, University of Pittsburgh School of Medicine; Pittsburgh, PA, USA
| | - Charles A. Scanga
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine; Pittsburgh, PA, USA
- Center for Vaccine Research, University of Pittsburgh School of Medicine; Pittsburgh, PA, USA
| | - Sarah Fortune
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health; Boston, MA, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, USA
| | - Philana Ling Lin
- Center for Vaccine Research, University of Pittsburgh School of Medicine; Pittsburgh, PA, USA
- Department of Pediatrics, Children’s Hospital of the University of Pittsburgh of UPMC; Pittsburgh, PA, USA
| | - Pauline Maiello
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine; Pittsburgh, PA, USA
- Center for Vaccine Research, University of Pittsburgh School of Medicine; Pittsburgh, PA, USA
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH); Bethesda, MD, USA
| | - Patricia A. Darrah
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH); Bethesda, MD, USA
| | - Robert A. Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH); Bethesda, MD, USA
| | - JoAnne L. Flynn
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine; Pittsburgh, PA, USA
- Center for Vaccine Research, University of Pittsburgh School of Medicine; Pittsburgh, PA, USA
| |
Collapse
|
5
|
Miceska S, Škof E, Gašljević G, Kloboves-Prevodnik V. Morphological and Immunocytochemical Characterization of Tumor Spheroids in Ascites from High-Grade Serous Carcinoma. Cells 2023; 12:2390. [PMID: 37830603 PMCID: PMC10572258 DOI: 10.3390/cells12192390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/19/2023] [Accepted: 09/28/2023] [Indexed: 10/14/2023] Open
Abstract
Tumor spheroids in the ascites of high-grade serous carcinoma (HGSC) are poorly described. Our objective was to describe their morphological features, cellular composition, PD-1 and PD-L1 expression, and survival correlation of these parameters. The density and size of spheroids were assessed in Giemsa-stained smears; the cell composition of spheroids, including tumor cells, immune cells, capillaries, and myofibroblasts, as well as PD-1 and PD-L1 expression on tumor and immune cells was assessed in immunocytochemically stained cell block sections. Forty-seven patients with primary HGSC and malignant ascites were included. A cut-off value for a spheroid density of 10% was established, which significantly predicted overall survival. However, spheroid size did not correlate with survival outcomes. Spheroids were primarily composed of tumor cells, but the presence of lymphocytes and macrophages was also confirmed. Moreover, capillaries were present in the spheroids of three patients, but the presence of myofibroblasts was not confirmed. PD-1 was expressed on lymphocytes but not on tumor cells. PD-L1 expression was seen on both tumor and immune cells, assessed by 22C3 and SP263 antibody clones but not by the SP142 clone. Our results highlight the potential of routine cytopathological techniques to analyze spheroids in HGSC ascites as a valuable tool to investigate their potential as prognostic markers.
Collapse
Affiliation(s)
- Simona Miceska
- Department of Cytopathology, Institute of Oncology Ljubljana, Zaloška Cesta 2, 1000 Ljubljana, Slovenia;
- Faculty of Medicine, University of Ljubljana, Korytkova Ulica 2, 1000 Ljubljana, Slovenia
| | - Erik Škof
- Faculty of Medicine, University of Ljubljana, Korytkova Ulica 2, 1000 Ljubljana, Slovenia
- Department of Medical Oncology, Institute of Oncology Ljubljana, Zaloška Cesta 2, 1000 Ljubljana, Slovenia
| | - Gorana Gašljević
- Department of Pathology, Institute of Oncology Ljubljana, Zaloška Cesta 2, 1000 Ljubljana, Slovenia
- Faculty of Medicine, University of Maribor, Taborska Ulica 8, 2000 Maribor, Slovenia
| | - Veronika Kloboves-Prevodnik
- Department of Cytopathology, Institute of Oncology Ljubljana, Zaloška Cesta 2, 1000 Ljubljana, Slovenia;
- Faculty of Medicine, University of Maribor, Taborska Ulica 8, 2000 Maribor, Slovenia
| |
Collapse
|
6
|
Martín-Faivre L, Gaudaire D, Laugier C, Bouraïma-Lelong H, Zientara S, Hans A. Development of a chronic focal equine arteritis virus infection of a male reproductive tract cell line. J Virol Methods 2023; 319:114756. [PMID: 37268046 DOI: 10.1016/j.jviromet.2023.114756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/08/2023] [Accepted: 05/27/2023] [Indexed: 06/04/2023]
Abstract
Equine arteritis virus (EAV) is an Alphaarterivirus (family Arteriviridae, order Nidovirales) that frequently causes an influenza-like illness in adult horses, but can also cause the abortions in mares and death of newborn foals. Once primary infection has been established, EAV can persist in the reproductive tract of some stallions. However, the mechanisms enabling this persistence, which depends on testosterone, remain largely unknown. We aimed to establish an in vitro model of non-cytopathic EAV infection to study viral persistence. In this work, we infected several cell lines originating from the male reproductive tract of different species. EAV infection was fully cytopathic for 92BR (donkey cells) and DDT1 MF-2 (hamster cells) cells, and less cytopathic for PC-3 cells (human cells); ST cells (porcine cells) seemed to eliminate the virus; LNCaP (human cells) and GC-1 spg (murine cells) cells were not permissive to EAV infection; finally, TM3 cells (murine cells) were permissive to EAV infection without any overt cytopathic effects. Infected TM3 cells can be maintained at least 7 days in culture without any subculture. They can also be subcultured over 39 days (subculturing them at 1:2 the first time at 5 dpi and then every 2-3 days), but in this case, the percentage of infected cells remains low. Infected TM3 cells may therefore provide a new model to study the host-pathogen interactions and to help determine the mechanisms involved in EAV persistence in stallion reproductive tract.
Collapse
Affiliation(s)
- Lydie Martín-Faivre
- ANSES Laboratory for Animal Health, Normandy site. PhEED Unit, Goustranville, France.
| | - Delphine Gaudaire
- ANSES Laboratory for Animal Health, Normandy site. PhEED Unit, Goustranville, France
| | - Claire Laugier
- ANSES Laboratory for Animal Health, Normandy site. PhEED Unit, Goustranville, France
| | | | - Stéphan Zientara
- Université Paris-Est, ANSES, Maisons-Alfort Laboratory for Animal Health, UMR Virology ANSES, INRAE, ENVA, Maisons-Alfort, France
| | - Aymeric Hans
- ANSES Laboratory for Animal Health, Normandy site. PhEED Unit, Goustranville, France
| |
Collapse
|
7
|
Antonopoulou T, Kanakousaki E, Dimitropoulos C, Manidakis N, Athanassakis I. Aberrant expression of T cell receptors in monocyte/macrophage RAW 264.7 cells: FCγRII/III compensates the need for CD3. Mol Immunol 2023; 157:167-175. [PMID: 37028131 DOI: 10.1016/j.molimm.2023.03.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 03/19/2023] [Accepted: 03/26/2023] [Indexed: 04/08/2023]
Abstract
Conventionally T-cell receptors (TCRs) have so far been considered as a T-lymphocyte privilege. However, recent findings also place TCR expression in non-lymphoid cells, namely neutrophils, eosinophils and macrophages. In order to examine the ectopic expression of TCR, this study focused on RAW 264.7 cells, which have been broadly used for their macrophage properties. Immunofluorescence staining detected 70% and 40% of the cells to express TCRαβ and TCRγδ respectively, which was also verified by RT-PCR experiments and confocal microscopy analysis. Interestingly, except from the predicted 292 and 288 bp gene products for the α- and γ-chain, additional products at 220 and 550 bp could be detected, respectively. RAW 264.7 cells also expressed the co-stimulatory CD4 and CD8 markers at a percentage of 61% and 14% respectively, which supported the expression of TCRs. However, only low numbers of cells expressed CD3ε and CD3ζ (9% and 7% respectively). Such observations contradicted the existing knowledge, and indicated that TCRs would be supported by other molecules for reaching the membrane and transducing their signal. Such candidate molecules could be the Fcγ receptors (FcγRs). Indeed, the FcγRII/III receptor was found to be expressed in 75% of the cells, which also expressed at a percentage of 25% major histocompatibility complex (MHC) class II molecules. Engagement of the FcγRII/III receptor by a recombinant IgG2aCH2 fragment, except from stimulating the macrophage-dependent properties of the cells, was shown to reduce expression of TCRαβ and γδ indicating that FcγRII/III was indeed used by TCRs for their transport to the cell membrane. In order to examine the ability of RAW 264.7 cells to simultaneously display antigen presenting- and T-cell properties, functional experiments as to antigen-specific antibody and IL-2 production were performed. In in vitro immunization assays in the presence of naïve B cells, RAW264.7 failed to promote antibody production. However, RAW 264.7 cells could compete with antigen-stimulated macrophages but not T cells when applied to a system of in vivo antigen-sensitized cells followed by an in vitro immunization protocol. Interestingly, simultaneous addition of antigen and the IgG2aCH2 fragment to RAW 264.7 cells could promote IL-2 production from the cells, indicating that FcγRII/III activation could also support TCR stimulation. Extrapolating these findings to cells of the myeloid origin, the above results dictate novel regulatory mechanisms towards the alteration of the immune response.
Collapse
|
8
|
Sulzmaier FJ, Kern N, Ahn SJ, Polovina A, Ho J, Hussain A, Cyprus G, Macedo C, Pandit R, Crago W, Rowell E, Timmer JC, Eckelman BP. INBRX-120, a CD8α-targeted detuned IL-2 that selectively expands and activates tumoricidal effector cells for safe and durable in vivo responses. J Immunother Cancer 2023; 11:jitc-2022-006116. [PMID: 36653071 PMCID: PMC9853254 DOI: 10.1136/jitc-2022-006116] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2023] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND As a major driver of lymphocyte proliferation and activation interleukin 2 (IL-2) is a crucial mediator for antitumor responses. Despite promising activity in a subset of patients, wider therapeutic utility of IL-2 (aldesleukin) has been hampered by severe dose-limiting toxicities, the expansion of immunosuppressive regulatory T cells and a poor pharmacokinetic (PK) profile. Recent engineering efforts, including non-α IL-2 variants, have lowered the toxicity profile, but have yet to induce meaningful antitumor activity in a wider patient population. METHODS We engineered INBRX-120, a CD8α-targeted Cisleukin™ molecule consisting of an affinity tuned IL-2 (IL2-x) connected to two high affinity CD8α-specific single domain antibodies via an effector-silenced Fc domain. To show that this large affinity differential enables directed IL-2 cis-signaling exclusively on CD8α-expressing tumoricidal effector cell populations, INBRX-120 effects on target cell expansion, activation and antitumor activity were tested in vitro. In vivo antitumor efficacy was evaluated in syngeneic mouse models alone or in combination with programmed cell death protein-1 (PD-1) blockade. Preclinical safety, as well as pharmacodynamic (PD) and PK profiling was carried out in non-human primates. RESULTS INBRX-120 effectively expanded and enhanced the cytotoxic capacity of CD8 T cells and natural killer cells towards tumor cells without affecting regulatory T cells in vitro and in vivo. In syngeneic mouse models, INBRX-120 surrogate showed safe, potent, and durable antitumor efficacy alone and in combination with PD-1 blockade. In non-human primates, INBRX-120 expanded and activated CD8α-expressing effector cells, showed a favorable PK profile, and was well tolerated up to a dose of 1 mg/kg. CONCLUSIONS Through its unique cis-signaling activity on CD8α-expressing effector cells, INBRX-120 overcomes the major limitations of IL-2-based therapy and effectively harnesses IL-2's potent intrinsic antitumor activity. This novel therapeutic strategy promises safer clinical activity that could induce meaningful antitumor efficacy in a wider set of patients with various cancer indications.
Collapse
Affiliation(s)
| | | | | | | | - Jason Ho
- Inhibrx Inc, La Jolla, California, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Romão PR, Teixeira PC, Schipper L, da Silva I, Santana Filho P, Júnior LCR, Peres A, Gonçalves da Fonseca S, Chagas Monteiro M, Lira FS, Andrey Cipriani Frade M, Comerlato J, Comerlato C, Sant'Anna FH, Bessel M, Abreu CM, Wendland EM, Dorneles GP. Viral load is associated with mitochondrial dysfunction and altered monocyte phenotype in acute severe SARS-CoV-2 infection. Int Immunopharmacol 2022; 108:108697. [PMID: 35405594 PMCID: PMC8920784 DOI: 10.1016/j.intimp.2022.108697] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/21/2022] [Accepted: 03/10/2022] [Indexed: 02/07/2023]
Abstract
Monocytes play a major role in the initial innate immune response to SARS-CoV-2. Although viral load may correlate with several clinical outcomes in COVID-19, much less is known regarding their impact on innate immune phenotype. We evaluated the monocyte phenotype and mitochondrial function in severe COVID-19 patients (n = 22) with different viral burden (determined by the median of viral load of the patients) at hospital admission. Severe COVID-19 patients presented lower frequency of CD14 + CD16- classical monocytes and CD39 expression on CD14 + monocytes, and higher frequency of CD14 + CD16 + intermediate and CD14-CD16 + nonclassical monocytes as compared to healthy controls independently of viral load. COVID-19 patients with high viral load exhibited increased GM-CSF, PGE-2 and lower IFN-α as compared to severe COVID-19 patients with low viral load (p < 0.05). CD14 + monocytes of COVID-19 patients with high viral load presented higher expression of PD-1 but lower HLA-DR on the cell surface than severe COVID-19 patients with low viral load. All COVID-19 patients presented decreased monocyte mitochondria membrane polarization, but high SARS-CoV-2 viral load was associated with increased mitochondrial reactive oxygen species. In this sense, higher viral load induces mitochondrial reactive oxygen species generation associated with exhaustion profile in CD14 + monocytes of severe COVID-19 patients. Altogether, these data shed light on new pathological mechanisms involving SARS-CoV-2 viral load on monocyte activation and mitochondrial function, which were associated with COVID-19 severity.
Collapse
Affiliation(s)
- Pedro Rt Romão
- Laboratory of Cellular and Molecular Immunology, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil; Graduate Program in Biosciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil.
| | - Paula C Teixeira
- Laboratory of Cellular and Molecular Immunology, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil; Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Lucas Schipper
- Laboratory of Cellular and Molecular Immunology, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil; Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Igor da Silva
- Laboratory of Cellular and Molecular Immunology, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Paulo Santana Filho
- Laboratory of Cellular and Molecular Immunology, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Luiz Carlos Rodrigues Júnior
- Graduate Program in Biosciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Alessandra Peres
- Laboratory of Cellular and Molecular Immunology, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil; Graduate Program in Biosciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | | | - Marta Chagas Monteiro
- Graduate Program in Pharmaceutical Science, Health Science Institute, Federal University of Pará/UFPA, Belém, Pará, Brazil
| | - Fabio S Lira
- Exercise and Immunometabolism Research Group, Postgraduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista (UNESP), Presidente Prudente 19060-900, SP, Brazil
| | - Marco Andrey Cipriani Frade
- Dermatology Division, Department of Medical Clinics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | | | | | | | - Marina Bessel
- Hospital Moinhos de Vento, Porto Alegre, Rio Grande do Sul, Brazil
| | - Celina Monteiro Abreu
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Eliana M Wendland
- Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil; Graduate Program in Pediatrics, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil.
| | - Gilson P Dorneles
- Laboratory of Cellular and Molecular Immunology, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil; Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| |
Collapse
|
10
|
Johanna I, Hernández-López P, Heijhuurs S, Scheper W, Bongiovanni L, de Bruin A, Beringer DX, Oostvogels R, Straetemans T, Sebestyen Z, Kuball J. Adding Help to an HLA-A*24:02 Tumor-Reactive γδTCR Increases Tumor Control. Front Immunol 2021; 12:752699. [PMID: 34759930 PMCID: PMC8573335 DOI: 10.3389/fimmu.2021.752699] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/06/2021] [Indexed: 11/13/2022] Open
Abstract
γδT cell receptors (γδTCRs) recognize a broad range of malignantly transformed cells in mainly a major histocompatibility complex (MHC)-independent manner, making them valuable additions to the engineered immune effector cell therapy that currently focuses primarily on αβTCRs and chimeric antigen receptors (CARs). As an exception to the rule, we have previously identified a γδTCR, which exerts antitumor reactivity against HLA-A*24:02-expressing malignant cells, however without the need for defined HLA-restricted peptides, and without exhibiting any sign of off-target toxicity in humanized HLA-A*24:02 transgenic NSG (NSG-A24:02) mouse models. This particular tumor-HLA-A*24:02-specific Vγ5Vδ1TCR required CD8αα co-receptor for its tumor reactive capacity when introduced into αβT cells engineered to express a defined γδTCR (TEG), referred to as TEG011; thus, it was only active in CD8+ TEG011. We subsequently explored the concept of additional redirection of CD4+ T cells through co-expression of the human CD8α gene into CD4+ and CD8+ TEG011 cells, later referred as TEG011_CD8α. Adoptive transfer of TEG011_CD8α cells in humanized HLA-A*24:02 transgenic NSG (NSG-A24:02) mice injected with tumor HLA-A*24:02+ cells showed superior tumor control in comparison to TEG011, and to mock control groups. The total percentage of mice with persisting TEG011_CD8α cells, as well as the total number of TEG011_CD8α cells per mice, was significantly improved over time, mainly due to a dominance of CD4+CD8+ double-positive TEG011_CD8α, which resulted in higher total counts of functional T cells in spleen and bone marrow. We observed that tumor clearance in the bone marrow of TEG011_CD8α-treated mice associated with better human T cell infiltration, which was not observed in the TEG011-treated group. Overall, introduction of transgenic human CD8α receptor on TEG011 improves antitumor reactivity against HLA-A*24:02+ tumor cells and further enhances in vivo tumor control.
Collapse
Affiliation(s)
- Inez Johanna
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | | | - Sabine Heijhuurs
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Wouter Scheper
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Laura Bongiovanni
- Department of Biomolecular Health Sciences, Dutch Molecular Pathology Center, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Alain de Bruin
- Department of Biomolecular Health Sciences, Dutch Molecular Pathology Center, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands.,Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Dennis X Beringer
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Rimke Oostvogels
- Department of Hematology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Trudy Straetemans
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands.,Department of Hematology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Zsolt Sebestyen
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Jürgen Kuball
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands.,Department of Hematology, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
11
|
Characterization of Adaptive-like γδ T Cells in Ugandan Infants during Primary Cytomegalovirus Infection. Viruses 2021; 13:v13101987. [PMID: 34696417 PMCID: PMC8537190 DOI: 10.3390/v13101987] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 09/21/2021] [Accepted: 09/27/2021] [Indexed: 12/15/2022] Open
Abstract
Gamma-delta (γδ) T cells are unconventional T cells that help control cytomegalovirus (CMV) infection in adults. γδ T cells develop early in gestation, and a fetal public γδ T cell receptor (TCR) clonotype is detected in congenital CMV infections. However, age-dependent γδ T cell responses to primary CMV infection are not well-understood. Flow cytometry and TCR sequencing was used to comprehensively characterize γδ T cell responses to CMV infection in a cohort of 32 infants followed prospectively from birth. Peripheral blood γδ T cell frequencies increased during infancy, and were higher among CMV-infected infants relative to uninfected. Clustering analyses revealed associations between CMV infection and activation marker expression on adaptive-like Vδ1 and Vδ3, but not innate-like Vγ9Vδ2 γδ T cell subsets. Frequencies of NKG2C+CD57+ γδ T cells were temporally associated with the quantity of CMV shed in saliva by infants with primary infection. The public γδ TCR clonotype was only detected in CMV-infected infants <120 days old and at lower frequencies than previously described in fetal infections. Our findings support the notion that CMV infection drives age-dependent expansions of specific γδ T cell populations, and provide insight for novel strategies to prevent CMV transmission and disease.
Collapse
|
12
|
Kaur G, Wright K, Mital P, Hibler T, Miranda JM, Thompson LA, Halley K, Dufour JM. Neonatal Pig Sertoli Cells Survive Xenotransplantation by Creating an Immune Modulatory Environment Involving CD4 and CD8 Regulatory T Cells. Cell Transplant 2021; 29:963689720947102. [PMID: 32841048 PMCID: PMC7564626 DOI: 10.1177/0963689720947102] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The acute cell-mediated immune response presents a significant barrier to
xenotransplantation. Immune-privileged Sertoli cells (SC) can prolong the
survival of co-transplanted cells including xenogeneic islets, hepatocytes, and
neurons by protecting them from immune rejection. Additionally, SC survive as
allo- and xenografts without the use of any immunosuppressive drugs suggesting
elucidating the survival mechanism(s) of SC could be used to improve survival of
xenografts. In this study, the survival and immune response generated toward
neonatal pig SC (NPSC) or neonatal pig islets (NPI), nonimmune-privileged
controls, was compared after xenotransplantation into naïve Lewis rats without
immune suppression. The NPSC survived throughout the study, while NPI were
rejected within 9 days. Analysis of the grafts revealed that macrophages and T
cells were the main immune cells infiltrating the NPSC and NPI grafts. Further
characterization of the T cells within the grafts indicated that the NPSC grafts
contained significantly more cluster of differentiation 4 (CD4) and cluster of
differentiation 8 (CD8) regulatory T cells (Tregs) at early time points than the
NPI grafts. Additionally, the presence of increased amounts of interleukin 10
(IL-10) and transforming growth factor (TGF) β and decreased levels of tumor
necrosis factor (TNF) α and apoptosis in the NPSC grafts compared to NPI grafts
suggests the presence of regulatory immune cells in the NPSC grafts. The NPSC
expressed several immunoregulatory factors such as TGFβ, thrombospondin-1
(THBS1), indoleamine-pyrrole 2,3-dioxygenase, and galectin-1, which could
promote the recruitment of these regulatory immune cells to the NPSC grafts. In
contrast, NPI grafts had fewer Tregs and increased apoptosis and inflammation
(increased TNFα, decreased IL-10 and TGFβ) suggestive of cytotoxic immune cells
that contribute to their early rejection. Collectively, our data suggest that a
regulatory graft environment with regulatory immune cells including CD4 and
CD8 Tregs in NPSC grafts could be attributed to the prolonged survival of the
NPSC xenografts.
Collapse
Affiliation(s)
- Gurvinder Kaur
- Department of Cell Biology and Biochemistry, 12343Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Department of Medical Education, 12343Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Kandis Wright
- Department of Cell Biology and Biochemistry, 12343Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Payal Mital
- Department of Cell Biology and Biochemistry, 12343Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Taylor Hibler
- Department of Cell Biology and Biochemistry, 12343Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Jonathan M Miranda
- Department of Cell Biology and Biochemistry, 12343Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Lea Ann Thompson
- Department of Cell Biology and Biochemistry, 12343Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Katelyn Halley
- Department of Cell Biology and Biochemistry, 12343Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Jannette M Dufour
- Department of Cell Biology and Biochemistry, 12343Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Department of Medical Education, 12343Texas Tech University Health Sciences Center, Lubbock, TX, USA
| |
Collapse
|
13
|
Characterization of innate and adaptive immune cells involved in the foreign body reaction to polypropylene meshes in the human abdomen. Hernia 2021; 26:309-323. [PMID: 33788008 PMCID: PMC8881270 DOI: 10.1007/s10029-021-02396-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/10/2021] [Indexed: 12/25/2022]
Abstract
Background Polypropylene (PP) mesh is widely used to reinforce tissues. The foreign body reaction (FBR) to the implant is dominated by innate immune cells, especially macrophages. However, considerable numbers of adaptive immune cells, namely T cells, have also been regularly observed, which appear to play a crucial role in the long-term host response. Methods This study investigated the FBR to seven human PP meshes, which were removed from the abdomen for recurrence after a median of one year. Using immunofluorescence microscopy, the FBR was examined for various innate (CD11b+ myeloid, CD68+ macrophages, CD56+ NK) and adaptive immune cells (CD3+ T, CD4+ T-helper, CD8+ cytotoxic, FoxP3+ T-regulatory, CD20+ B) as well as “conventional” immune cells (defined as cells expressing their specific immune cell marker without co-expressing CD68). Results T-helper cells (19%) and regulatory T-cells (25%) were present at comparable rates to macrophages, and clustered significantly toward the mesh fibers. For all cell types the lowest proportions of “conventional” cells (< 60%) were observed at the mesh–tissue interface, but increased considerably at about 50–100 µm, indicating reduced stimulation with rising distance to the mesh fibers. Conclusion Both innate and adaptive immune cells participate in the chronic FBR to PP meshes with T cells and macrophages being the predominant cell types, respectively. In concordance with the previous data, many cells presented a “hybrid” pattern near the mesh fibers. The complexity of the immune reaction seen within the foreign body granuloma may explain why approaches focusing on specific cell types have not been very successful in reducing the chronic FBR. Supplementary Information The online version contains supplementary material available at 10.1007/s10029-021-02396-7.
Collapse
|
14
|
Complete Freund's adjuvant-free experimental autoimmune encephalomyelitis in Dark Agouti rats is a valuable tool for multiple sclerosis studies. J Neuroimmunol 2021; 354:577547. [PMID: 33765502 DOI: 10.1016/j.jneuroim.2021.577547] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 03/02/2021] [Accepted: 03/12/2021] [Indexed: 12/21/2022]
Abstract
Experimental autoimmune encephalomyelitis (EAE) is classically induced with complete Freund's adjuvant (CFA). The immune response against CFA has a confounding influence on the translational capacity of EAE as a multiple sclerosis model. Here, we compare clinical, cellular and molecular properties between syngeneic spinal cord homogenate (SCH)- and SCH + CFA-immunized Dark Agouti rats. EAE signs were observed earlier and the cumulative clinical score was higher without CFA. Also, a higher number of immune cells infiltrates in the spinal cords was noticed at the peak of EAE without CFA. High spinal cord abundance of CD8+CD11bc+MHC class II+ cells was detected in SCH-immunized rats. Myelin basic protein -specific response can be elicited in the cells from the lymph nodes draining the site of SCH immunization. This CFA-free EAE is a reliable multiple sclerosis model.
Collapse
|
15
|
Groth EE, Weber M, Bahmer T, Pedersen F, Kirsten A, Börnigen D, Rabe KF, Watz H, Ammerpohl O, Goldmann T. Exploration of the sputum methylome and omics deconvolution by quadratic programming in molecular profiling of asthma and COPD: the road to sputum omics 2.0. Respir Res 2020; 21:274. [PMID: 33076907 PMCID: PMC7574293 DOI: 10.1186/s12931-020-01544-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 10/11/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND To date, most studies involving high-throughput analyses of sputum in asthma and COPD have focused on identifying transcriptomic signatures of disease. No whole-genome methylation analysis of sputum cells has been performed yet. In this context, the highly variable cellular composition of sputum has potential to confound the molecular analyses. METHODS Whole-genome transcription (Agilent Human 4 × 44 k array) and methylation (Illumina 450 k BeadChip) analyses were performed on sputum samples of 9 asthmatics, 10 healthy and 10 COPD subjects. RNA integrity was checked by capillary electrophoresis and used to correct in silico for bias conferred by RNA degradation during biobank sample storage. Estimates of cell type-specific molecular profiles were derived via regression by quadratic programming based on sputum differential cell counts. All analyses were conducted using the open-source R/Bioconductor software framework. RESULTS A linear regression step was found to perform well in removing RNA degradation-related bias among the main principal components of the gene expression data, increasing the number of genes detectable as differentially expressed in asthma and COPD sputa (compared to controls). We observed a strong influence of the cellular composition on the results of mixed-cell sputum analyses. Exemplarily, upregulated genes derived from mixed-cell data in asthma were dominated by genes predominantly expressed in eosinophils after deconvolution. The deconvolution, however, allowed to perform differential expression and methylation analyses on the level of individual cell types and, though we only analyzed a limited number of biological replicates, was found to provide good estimates compared to previously published data about gene expression in lung eosinophils in asthma. Analysis of the sputum methylome indicated presence of differential methylation in genomic regions of interest, e.g. mapping to a number of human leukocyte antigen (HLA) genes related to both major histocompatibility complex (MHC) class I and II molecules in asthma and COPD macrophages. Furthermore, we found the SMAD3 (SMAD family member 3) gene, among others, to lie within differentially methylated regions which has been previously reported in the context of asthma. CONCLUSIONS In this methodology-oriented study, we show that methylation profiling can be easily integrated into sputum analysis workflows and exhibits a strong potential to contribute to the profiling and understanding of pulmonary inflammation. Wherever RNA degradation is of concern, in silico correction can be effective in improving both sensitivity and specificity of downstream analyses. We suggest that deconvolution methods should be integrated in sputum omics analysis workflows whenever possible in order to facilitate the unbiased discovery and interpretation of molecular patterns of inflammation.
Collapse
Affiliation(s)
- Espen E Groth
- LungenClinic Grosshansdorf, Großhansdorf, Germany. .,Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Großhansdorf, Germany. .,Department of Internal Medicine I, Pneumology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany. .,Department of Oncology, Hematology and BMT with Section Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Melanie Weber
- Program in Applied and Computational Mathematics, Princeton University, Princeton, NJ, USA
| | - Thomas Bahmer
- LungenClinic Grosshansdorf, Großhansdorf, Germany.,Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Großhansdorf, Germany.,Department of Internal Medicine I, Pneumology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Frauke Pedersen
- LungenClinic Grosshansdorf, Großhansdorf, Germany.,Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Großhansdorf, Germany.,Pulmonary Research Institute at LungenClinic Grosshansdorf, Großhansdorf, Germany
| | - Anne Kirsten
- Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Großhansdorf, Germany.,Pulmonary Research Institute at LungenClinic Grosshansdorf, Großhansdorf, Germany
| | - Daniela Börnigen
- Bioinformatics Core Unit, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Klaus F Rabe
- LungenClinic Grosshansdorf, Großhansdorf, Germany.,Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Großhansdorf, Germany
| | - Henrik Watz
- Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Großhansdorf, Germany.,Pulmonary Research Institute at LungenClinic Grosshansdorf, Großhansdorf, Germany
| | - Ole Ammerpohl
- Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Großhansdorf, Germany.,Institute of Human Genetics, University Medical Center Ulm, Ulm, Germany
| | - Torsten Goldmann
- Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Großhansdorf, Germany.,Research Center Borstel, Pathology, Borstel, Germany
| |
Collapse
|
16
|
Mørch AM, Bálint Š, Santos AM, Davis SJ, Dustin ML. Coreceptors and TCR Signaling - the Strong and the Weak of It. Front Cell Dev Biol 2020; 8:597627. [PMID: 33178706 PMCID: PMC7596257 DOI: 10.3389/fcell.2020.597627] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 09/28/2020] [Indexed: 12/02/2022] Open
Abstract
The T-cell coreceptors CD4 and CD8 have well-characterized and essential roles in thymic development, but how they contribute to immune responses in the periphery is unclear. Coreceptors strengthen T-cell responses by many orders of magnitude - beyond a million-fold according to some estimates - but the mechanisms underlying these effects are still debated. T-cell receptor (TCR) triggering is initiated by the binding of the TCR to peptide-loaded major histocompatibility complex (pMHC) molecules on the surfaces of other cells. CD4 and CD8 are the only T-cell proteins that bind to the same pMHC ligand as the TCR, and can directly associate with the TCR-phosphorylating kinase Lck. At least three mechanisms have been proposed to explain how coreceptors so profoundly amplify TCR signaling: (1) the Lck recruitment model and (2) the pseudodimer model, both invoked to explain receptor triggering per se, and (3) two-step coreceptor recruitment to partially triggered TCRs leading to signal amplification. More recently it has been suggested that, in addition to initiating or augmenting TCR signaling, coreceptors effect antigen discrimination. But how can any of this be reconciled with TCR signaling occurring in the absence of CD4 or CD8, and with their interactions with pMHC being among the weakest specific protein-protein interactions ever described? Here, we review each theory of coreceptor function in light of the latest structural, biochemical, and functional data. We conclude that the oldest ideas are probably still the best, i.e., that their weak binding to MHC proteins and efficient association with Lck allow coreceptors to amplify weak incipient triggering of the TCR, without comprising TCR specificity.
Collapse
Affiliation(s)
- Alexander M. Mørch
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Štefan Bálint
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Ana Mafalda Santos
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Simon J. Davis
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Michael L. Dustin
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
17
|
Fonseca S, Pereira V, Lau C, Teixeira MDA, Bini-Antunes M, Lima M. Human Peripheral Blood Gamma Delta T Cells: Report on a Series of Healthy Caucasian Portuguese Adults and Comprehensive Review of the Literature. Cells 2020; 9:cells9030729. [PMID: 32188103 PMCID: PMC7140678 DOI: 10.3390/cells9030729] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/06/2020] [Accepted: 03/13/2020] [Indexed: 12/20/2022] Open
Abstract
Gamma delta T cells (Tc) are divided according to the type of Vδ and Vγ chains they express, with two major γδ Tc subsets being recognized in humans: Vδ2Vγ9 and Vδ1. Despite many studies in pathological conditions, only a few have quantified the γδ Tc subsets in healthy adults, and a comprehensive review of the factors influencing its representation in the blood is missing. Here we quantified the total γδ Tc and the Vδ2/Vγ9 and Vδ1 Tc subsets in the blood from 30 healthy, Caucasian, Portuguese adults, we characterized their immunophenotype by 8-color flow cytometry, focusing in a few relevant Tc markers (CD3/TCR-γδ, CD5, CD8), and costimulatory (CD28), cytotoxic (CD16) and adhesion (CD56) molecules, and we examined the impacts of age and gender. Additionally, we reviewed the literature on the influences of race/ethnicity, age, gender, special periods of life, past infections, diet, medications and concomitant diseases on γδ Tc and their subsets. Given the multitude of factors influencing the γδ Tc repertoire and immunophenotype and the high variation observed, caution should be taken in interpreting “abnormal” γδ Tc values and repertoire deviations, and the clinical significance of small populations of “phenotypically abnormal” γδ Tc in the blood.
Collapse
Affiliation(s)
- Sónia Fonseca
- Laboratory of Cytometry, Unit for Hematology Diagnosis, Department of Hematology, Hospital de Santo António (HSA), Centro Hospitalar Universitário do Porto (CHUP), Unidade Multidisciplinar de Investigação Biomédica, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto (UMIB/ICBAS/UP); 4099-001 Porto Porto, Portugal; (S.F.); (C.L.); (M.d.A.T.)
| | - Vanessa Pereira
- Department of Clinical Pathology, Centro Hospitalar de Vila Nova de Gaia/Espinho (CHVNG/E); 4434-502 Vila Nova de Gaia, Portugal;
| | - Catarina Lau
- Laboratory of Cytometry, Unit for Hematology Diagnosis, Department of Hematology, Hospital de Santo António (HSA), Centro Hospitalar Universitário do Porto (CHUP), Unidade Multidisciplinar de Investigação Biomédica, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto (UMIB/ICBAS/UP); 4099-001 Porto Porto, Portugal; (S.F.); (C.L.); (M.d.A.T.)
| | - Maria dos Anjos Teixeira
- Laboratory of Cytometry, Unit for Hematology Diagnosis, Department of Hematology, Hospital de Santo António (HSA), Centro Hospitalar Universitário do Porto (CHUP), Unidade Multidisciplinar de Investigação Biomédica, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto (UMIB/ICBAS/UP); 4099-001 Porto Porto, Portugal; (S.F.); (C.L.); (M.d.A.T.)
| | - Marika Bini-Antunes
- Laboratory of Immunohematology and Blood Donors Unit, Department of Hematology, Hospital de Santo António (HSA), Centro Hospitalar Universitário do Porto (CHUP), Unidade Multidisciplinar de Investigação Biomédica, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto (UMIB/ICBAS/UP); 4099-001Porto, Portugal;
| | - Margarida Lima
- Laboratory of Cytometry, Unit for Hematology Diagnosis, Department of Hematology, Hospital de Santo António (HSA), Centro Hospitalar Universitário do Porto (CHUP), Unidade Multidisciplinar de Investigação Biomédica, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto (UMIB/ICBAS/UP); 4099-001 Porto Porto, Portugal; (S.F.); (C.L.); (M.d.A.T.)
- Correspondence: ; Tel.: + 351-22-20-77-500
| |
Collapse
|
18
|
Carpenter RS, Jiang RR, Brennan FH, Hall JCE, Gottipati MK, Niewiesk S, Popovich PG. Human immune cells infiltrate the spinal cord and impair recovery after spinal cord injury in humanized mice. Sci Rep 2019; 9:19105. [PMID: 31836828 PMCID: PMC6911055 DOI: 10.1038/s41598-019-55729-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 11/27/2019] [Indexed: 02/07/2023] Open
Abstract
Humanized mice can be used to better understand how the human immune system responds to central nervous system (CNS) injury and inflammation. The optimal parameters for using humanized mice in preclinical CNS injury models need to be established for appropriate use and interpretation. Here, we show that the developmental age of the human immune system significantly affects anatomical and functional outcome measures in a preclinical model of traumatic spinal cord injury (SCI). Specifically, it takes approximately 3-4 months for a stable and functionally competent human immune system to develop in neonatal immune compromised mice after they are engrafted with human umbilical cord blood stem cells. Humanized mice receiving a SCI before or after stable engraftment exhibit significantly different neuroinflammatory profiles. Importantly, the development of a mature human immune system was associated with worse lesion pathology and neurological recovery after SCI. In these mice, human T cells infiltrate the spinal cord lesion and directly contact human macrophages. Together, data in this report establish an optimal experimental framework for using humanized mice to help translate promising preclinical therapies for CNS injury.
Collapse
Affiliation(s)
- Randall S Carpenter
- Neuroscience Graduate Program, The Ohio State University, Columbus, Ohio, USA
- Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, Ohio, USA
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, Ohio, USA
- Department of Neuroscience, The Ohio State University, Columbus, Ohio, USA
| | - Roselyn R Jiang
- Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, Ohio, USA
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, Ohio, USA
- Department of Neuroscience, The Ohio State University, Columbus, Ohio, USA
| | - Faith H Brennan
- Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, Ohio, USA
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, Ohio, USA
- Department of Neuroscience, The Ohio State University, Columbus, Ohio, USA
| | - Jodie C E Hall
- Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, Ohio, USA
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, Ohio, USA
- Department of Neuroscience, The Ohio State University, Columbus, Ohio, USA
| | - Manoj K Gottipati
- Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, Ohio, USA
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, Ohio, USA
- Department of Neuroscience, The Ohio State University, Columbus, Ohio, USA
| | - Stefan Niewiesk
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, USA
| | - Phillip G Popovich
- Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, Ohio, USA.
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, Ohio, USA.
- Department of Neuroscience, The Ohio State University, Columbus, Ohio, USA.
| |
Collapse
|
19
|
Proliferation-competent Tcf1+ CD8 T cells in dysfunctional populations are CD4 T cell help independent. Proc Natl Acad Sci U S A 2019; 116:20070-20076. [PMID: 31530725 PMCID: PMC6778176 DOI: 10.1073/pnas.1902701116] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
T cell maintenance in chronic infection and cancer follows a hierarchical order. Short-lived effector CD8 T cells are constitutively replaced from a proliferation-competent Tcf1-expressing progenitor population. This occurs spontaneously at low levels and increases in magnitude upon blocking PD-1 signaling. We explore how CD4 T cell help controls transition and survival of the progenitors and their progeny by utilizing single-cell RNA sequencing. Unexpectedly, absence of CD4 help caused reductions in cell numbers only among terminally differentiated cells while proliferation-competent progenitor cells remained unaffected with regard to their numbers and their overall phenotype. In fact, upon restoration of a functional CD4 compartment, the progenitors began to regenerate the effector CD8 T cells. Thus, unlike memory T cells for which secondary expansion requires CD4 T cell help, this is not a necessity for proliferation-competent progenitor cells in dysfunctional populations. Our data therefore reveals that proliferation-competent cells in dysfunctional populations show a previously unrecognized uncoupling of CD4 T cell help that is otherwise required by conventional memory T cells.
Collapse
|
20
|
Are rats more human than mice? Immunobiology 2019; 224:172-176. [DOI: 10.1016/j.imbio.2018.09.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 09/12/2018] [Accepted: 09/19/2018] [Indexed: 11/23/2022]
|
21
|
The Differential Effects of Leukocyte-Containing and Pure Platelet-Rich Plasma on Nucleus Pulposus-Derived Mesenchymal Stem Cells: Implications for the Clinical Treatment of Intervertebral Disc Degeneration. Stem Cells Int 2018; 2018:7162084. [PMID: 30425747 PMCID: PMC6218728 DOI: 10.1155/2018/7162084] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 07/30/2018] [Accepted: 08/09/2018] [Indexed: 12/18/2022] Open
Abstract
Background Platelet-rich plasma (PRP) is a promising strategy for intervertebral disc degeneration. However, the potential harmful effects of leukocytes in PRP on nucleus pulposus-derived mesenchymal stem cells (NPMSCs) have seldom been studied. This study aimed at comparatively evaluating effects of pure platelet-rich plasma (P-PRP) and leukocyte-containing platelet-rich plasma (L-PRP) on rabbit NPMSCs in vitro. Methods NPMSCs isolated from rabbit NP tissues were treated with L-PRP or P-PRP in vitro, and then cell proliferation and expression of stem cell markers, proinflammatory cytokines (TNF-α, IL-1β), production of ECM (extracellular matrix-related protein), and NF-κB p65 protein were validated by CCK-8 assay, real-time polymerase chain reaction, enzyme-linked immunosorbent assay, immunofluorescence, and western blot respectively. Results NPMSCs differentiate into nucleus pulposus-like cells after treatment of PRPs (P-PRP and L-PRP), and NPMSCs exhibited maximum proliferation at a 10% PRP dose. L-PRP had observably higher concentration of leukocytes, TNF-α, and IL-1β than P-PRP. Furthermore, compared to P-PRP, L-PRP induced the differentiated NPMSCs to upregulate the expression of TNF-α and IL-1β, enhanced activation of the NF-κB pathway, increased the expression of MMP-1 and MMP-13, and produced less ECM in differentiated NPMSCs. Conclusions Both P-PRP and L-PRP can induce the proliferation and NP-differentiation of NPMSCs. Compared to L-PRP, P-PRP can avoid the activation of the NF-κB pathway, thus reducing the inflammatory and catabolic responses.
Collapse
|
22
|
Schulte-Wrede U, Sörensen T, Grün JR, Häupl T, Hirseland H, Steinbrich-Zöllner M, Wu P, Radbruch A, Poddubnyy D, Sieper J, Syrbe U, Grützkau A. An explorative study on deep profiling of peripheral leukocytes to identify predictors for responsiveness to anti-tumour necrosis factor alpha therapies in ankylosing spondylitis: natural killer cells in focus. Arthritis Res Ther 2018; 20:191. [PMID: 30157966 PMCID: PMC6116509 DOI: 10.1186/s13075-018-1692-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 07/31/2018] [Indexed: 12/17/2022] Open
Abstract
Background Therapeutic targeting of tumour necrosis factor (TNF)-α is highly effective in ankylosing spondylitis (AS) patients. However, since one-third of anti-TNF-treated AS patients do not show an adequate clinical response there is an urgent need for new biomarkers that would aid clinicians in their decision-making to select appropriate therapeutic options. Thus, the aim of this explorative study was to identify cell-based biomarkers in peripheral blood that could be used for a pre-treatment stratification of AS patients. Methods A high-dimensional, multi-parametric flow cytometric approach was applied to identify baseline predictors in 31 AS patients before treatment with the TNF blockers adalimumab (TNF-neutralisation) and etanercept (soluble TNF receptor). Results As the major result, the frequencies of natural killer (NK) cells, and in particular CD8-positive (CD8+) NK cell subsets, were most predictive for therapeutic outcome in AS patients. While an inverse correlation between classical CD56+/CD16+ NK cells and reduction of disease activity was observed, the CD8+ NK cell subset behaved in the opposite direction. At baseline, responders showed significantly increased frequencies of CD8+ NK cells compared with non-responders. Conclusions This is the first study demonstrating that the composition of the NK cell compartment has predictive power for prediction of therapeutic outcome for anti-TNF-α blockers, and we identified CD8+ NK cells as a potential new player in the TNF-α-driven chronic inflammatory immune response of AS. Electronic supplementary material The online version of this article (10.1186/s13075-018-1692-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ursula Schulte-Wrede
- German Rheumatism Research Center Berlin (DRFZ), an Institute of the Leibniz-Association, Immune Monitoring Core Facility, Charitéplatz 1, 10117, Berlin, Germany
| | - Till Sörensen
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Joachim R Grün
- German Rheumatism Research Center Berlin (DRFZ), an Institute of the Leibniz-Association, Immune Monitoring Core Facility, Charitéplatz 1, 10117, Berlin, Germany.,German Rheumatism Research Center Berlin (DRFZ), an Institute of the Leibniz-Association, Bioinformatics Group, Berlin, Germany
| | - Thomas Häupl
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Heike Hirseland
- German Rheumatism Research Center Berlin (DRFZ), an Institute of the Leibniz-Association, Immune Monitoring Core Facility, Charitéplatz 1, 10117, Berlin, Germany
| | - Marta Steinbrich-Zöllner
- Department of Gastroenterology, Infectiology and Rheumatology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Peihua Wu
- Department of Gastroenterology, Infectiology and Rheumatology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Andreas Radbruch
- German Rheumatism Research Center Berlin (DRFZ), an Institute of the Leibniz-Association, Immune Monitoring Core Facility, Charitéplatz 1, 10117, Berlin, Germany.,German Rheumatism Research Center Berlin (DRFZ), an Institute of the Leibniz-Association, Cell Biology Group, Berlin, Germany
| | - Denis Poddubnyy
- Department of Gastroenterology, Infectiology and Rheumatology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,German Rheumatism Research Center Berlin (DRFZ), an Institute of the Leibniz-Association, Epidemiology Unit, Berlin, Germany
| | - Joachim Sieper
- Department of Gastroenterology, Infectiology and Rheumatology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Uta Syrbe
- Department of Gastroenterology, Infectiology and Rheumatology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Andreas Grützkau
- German Rheumatism Research Center Berlin (DRFZ), an Institute of the Leibniz-Association, Immune Monitoring Core Facility, Charitéplatz 1, 10117, Berlin, Germany.
| |
Collapse
|
23
|
Diedrichs-Möhring M, Kaufmann U, Wildner G. The immunopathogenesis of chronic and relapsing autoimmune uveitis – Lessons from experimental rat models. Prog Retin Eye Res 2018; 65:107-126. [DOI: 10.1016/j.preteyeres.2018.02.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 02/19/2018] [Accepted: 02/22/2018] [Indexed: 12/12/2022]
|
24
|
Layton DS, Xiao X, Bentley JD, Lu L, Stewart CR, Bean AGD, Adams TE. Development of an anti-ferret CD4 monoclonal antibody for the characterisation of ferret T lymphocytes. J Immunol Methods 2017; 444:29-35. [PMID: 28216237 PMCID: PMC7094458 DOI: 10.1016/j.jim.2017.02.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 02/01/2017] [Accepted: 02/14/2017] [Indexed: 12/21/2022]
Abstract
The ferret is an established animal model for a number of human respiratory viral infections, such as influenza virus and more recently, Ebola virus. However, a paucity of immunological reagents has hampered the study of cellular immune responses. Here we describe the development and characterisation of a novel monoclonal antibody (mAb) against the ferret CD4 antigen and the characterisation of ferret CD4 T lymphocytes. Recombinant production and purification of the ferret CD4 ectodomain soluble protein allowed hybridoma generation and the generation of a mAb (FeCD4) showing strong binding to ferret CD4 protein and lymphoid cells by flow cytometry. FeCD4 bound to its cognate antigen post-fixation with paraformaldehyde (PFA) which is routinely used to inactivate highly pathogenic viruses. We have also used FeCD4 in conjunction with other immune cell markers to characterise ferret T cells in both primary and secondary lymphoid organs. In summary, we have developed an important reagent for the study of cellular immunological responses in the ferret model of infectious disease.
Collapse
Affiliation(s)
- Daniel S Layton
- CSIRO Health and Biosecurity, Australian Animal Health Laboratory, Geelong, Victoria, Australia.
| | - Xiaowen Xiao
- CSIRO Manufacturing, Parkville, Victoria, Australia
| | | | - Louis Lu
- CSIRO Manufacturing, Parkville, Victoria, Australia
| | - Cameron R Stewart
- CSIRO Health and Biosecurity, Australian Animal Health Laboratory, Geelong, Victoria, Australia
| | - Andrew G D Bean
- CSIRO Health and Biosecurity, Australian Animal Health Laboratory, Geelong, Victoria, Australia
| | | |
Collapse
|
25
|
Carmona SJ, Teichmann SA, Ferreira L, Macaulay IC, Stubbington MJT, Cvejic A, Gfeller D. Single-cell transcriptome analysis of fish immune cells provides insight into the evolution of vertebrate immune cell types. Genome Res 2017; 27:451-461. [PMID: 28087841 PMCID: PMC5340972 DOI: 10.1101/gr.207704.116] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 01/12/2017] [Indexed: 12/30/2022]
Abstract
The immune system of vertebrate species consists of many different cell types that have distinct functional roles and are subject to different evolutionary pressures. Here, we first analyzed conservation of genes specific for all major immune cell types in human and mouse. Our results revealed higher gene turnover and faster evolution of trans-membrane proteins in NK cells compared with other immune cell types, and especially T cells, but similar conservation of nuclear and cytoplasmic protein coding genes. To validate these findings in a distant vertebrate species, we used single-cell RNA sequencing of lck:GFP cells in zebrafish and obtained the first transcriptome of specific immune cell types in a nonmammalian species. Unsupervised clustering and single-cell TCR locus reconstruction identified three cell populations, T cells, a novel type of NK-like cells, and a smaller population of myeloid-like cells. Differential expression analysis uncovered new immune-cell-specific genes, including novel immunoglobulin-like receptors, and neofunctionalization of recently duplicated paralogs. Evolutionary analyses confirmed the higher gene turnover of trans-membrane proteins in NK cells compared with T cells in fish species, suggesting that this is a general property of immune cell types across all vertebrates.
Collapse
Affiliation(s)
- Santiago J Carmona
- Ludwig Center for Cancer Research, University of Lausanne, 1066 Epalinges, Switzerland
- Swiss Institute of Bioinformatics (SIB), 1015 Lausanne, Switzerland
| | - Sarah A Teichmann
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, United Kingdom
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge CB10 1SA, United Kingdom
| | - Lauren Ferreira
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge CB10 1SA, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge CB2 0XY, United Kingdom
- Wellcome Trust-Medical Research Council, Cambridge Stem Cell Institute, Cambridge CB2 1QR, United Kingdom
| | - Iain C Macaulay
- Sanger Institute-EBI Single-Cell Genomics Centre, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1HH, United Kingdom
| | - Michael J T Stubbington
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, United Kingdom
| | - Ana Cvejic
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge CB10 1SA, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge CB2 0XY, United Kingdom
- Wellcome Trust-Medical Research Council, Cambridge Stem Cell Institute, Cambridge CB2 1QR, United Kingdom
| | - David Gfeller
- Ludwig Center for Cancer Research, University of Lausanne, 1066 Epalinges, Switzerland
- Swiss Institute of Bioinformatics (SIB), 1015 Lausanne, Switzerland
| |
Collapse
|
26
|
Boieri M, Ulvmoen A, Sudworth A, Lendrem C, Collin M, Dickinson AM, Kveberg L, Inngjerdingen M. IL-12, IL-15, and IL-18 pre-activated NK cells target resistant T cell acute lymphoblastic leukemia and delay leukemia development in vivo. Oncoimmunology 2017; 6:e1274478. [PMID: 28405496 DOI: 10.1080/2162402x.2016.1274478] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 12/15/2016] [Accepted: 12/15/2016] [Indexed: 12/17/2022] Open
Abstract
NK cells have shown promise in therapy of hematological cancers, in particular against acute myeloid leukemia. In contrast, the more NK cell-resistant acute lymphoblastic leukemia (ALL) is difficult to treat with NK-cell-based therapies, and we hypothesized that pre-activation of NK cells could overcome this resistance. We show in pediatric and adult patients with T-cell ALL (T-ALL) perturbed NK cell effector functions at diagnosis. Using an in vivo rat model for T-ALL, Roser leukemia (RL), suppressed NK cell effector functions were observed. NK cells from T-ALL patients had reduced expression of the activating receptors NKp46 and DNAM-1, but not NKG2D. In contrast to T-ALL patients, NKG2D but not NKp46 was downregulated on NK cells during rat RL. Decreased frequencies of terminally differentiated NKG2A+CD57-CD56dim NK cells in human T-ALL was paralleled in the rat by reduced frequencies of bone marrow NK cells expressing the maturation marker CD11b, possibly indicating impairment of differentiation during leukemia. RL was highly resistant to autologous NK cells, but this resistance was overcome upon pre-activation of NK cells with IL-12, IL-15, and IL-18, with concomitant upregulation of activation markers and activating receptors. Importantly, adoptive transfers of IL-12, IL-15, and IL-18 pre-activated NK cells significantly slowed progression of RL in vivo. The data thus shows that T-ALL blasts normally resistant to NK cells may be targeted by cytokine pre-activated autologous NK cells, and this approach could have potential implications for immunotherapeutic protocols using NK cells to more efficiently target leukemia.
Collapse
Affiliation(s)
- Margherita Boieri
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Aina Ulvmoen
- Department of Immunology, Oslo University Hospital , Oslo, Norway
| | - Amanda Sudworth
- Department of Immunology, Oslo University Hospital , Oslo, Norway
| | - Clare Lendrem
- Institute of Cellular Medicine, Medical School, Newcastle University , Newcastle-upon-Tyne, UK
| | - Matthew Collin
- Institute of Cellular Medicine, Medical School, Newcastle University , Newcastle-upon-Tyne, UK
| | - Anne M Dickinson
- Institute of Cellular Medicine, Medical School, Newcastle University , Newcastle-upon-Tyne, UK
| | - Lise Kveberg
- Department of Immunology, Institute of Clinical Medicine, University of Oslo , Oslo, Norway
| | | |
Collapse
|
27
|
Vasconcelos DM, Gonçalves RM, Almeida CR, Pereira IO, Oliveira MI, Neves N, Silva AM, Ribeiro AC, Cunha C, Almeida AR, Ribeiro CC, Gil AM, Seebach E, Kynast KL, Richter W, Lamghari M, Santos SG, Barbosa MA. Fibrinogen scaffolds with immunomodulatory properties promote in vivo bone regeneration. Biomaterials 2016; 111:163-178. [DOI: 10.1016/j.biomaterials.2016.10.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 09/30/2016] [Accepted: 10/01/2016] [Indexed: 01/27/2023]
|
28
|
Kadivar M, Petersson J, Svensson L, Marsal J. CD8αβ+ γδ T Cells: A Novel T Cell Subset with a Potential Role in Inflammatory Bowel Disease. THE JOURNAL OF IMMUNOLOGY 2016; 197:4584-4592. [PMID: 27849165 DOI: 10.4049/jimmunol.1601146] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 10/13/2016] [Indexed: 01/27/2023]
Abstract
γδ T cells have been attributed a wide variety of functions, which in some cases may appear as contradictory. To better understand the enigmatic biology of γδ T cells it is crucial to define the constituting subpopulations. γδ T cells have previously been categorized into two subpopulations: CD8αα+ and CD8- cells. In this study we have defined and characterized a novel subset of human γδ T-cells expressing CD8αβ. These CD8αβ+ γδ T cells differed from the previously described γδ T cell subsets in several aspects, including the degree of enrichment within the gut mucosa, the activation status in blood, the type of TCRδ variant used in blood, and small but significant differences in their response to IL-2 stimulation. Furthermore, the novel subset expressed cytotoxic mediators and CD69, and produced IFN-γ and TNF-α. In patients with active inflammatory bowel disease the mucosal frequencies of CD8αβ+ γδ T cells were significantly lower as compared with healthy controls, correlated negatively with the degree of disease activity, and increased to normal levels as a result of anti-TNF-α therapy. In conclusion, our results demonstrate that CD8αβ+ γδ T cells constitute a novel lymphocyte subset, which is strongly enriched within the gut and may play an important role in gut homeostasis and mucosal healing in inflammatory bowel disease.
Collapse
Affiliation(s)
| | - Julia Petersson
- Immunology Section, Lund University, S-22184 Lund, Sweden; and
| | - Lena Svensson
- Immunology Section, Lund University, S-22184 Lund, Sweden; and
| | - Jan Marsal
- Immunology Section, Lund University, S-22184 Lund, Sweden; and .,Department of Gastroenterology, Skåne University Hospital, S-22185 Lund, Sweden
| |
Collapse
|
29
|
Dee CT, Nagaraju RT, Athanasiadis EI, Gray C, Fernandez Del Ama L, Johnston SA, Secombes CJ, Cvejic A, Hurlstone AFL. CD4-Transgenic Zebrafish Reveal Tissue-Resident Th2- and Regulatory T Cell-like Populations and Diverse Mononuclear Phagocytes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 197:3520-3530. [PMID: 27694495 PMCID: PMC5073357 DOI: 10.4049/jimmunol.1600959] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 09/07/2016] [Indexed: 12/20/2022]
Abstract
CD4+ T cells are at the nexus of the innate and adaptive arms of the immune system. However, little is known about the evolutionary history of CD4+ T cells, and it is unclear whether their differentiation into specialized subsets is conserved in early vertebrates. In this study, we have created transgenic zebrafish with vibrantly labeled CD4+ cells allowing us to scrutinize the development and specialization of teleost CD4+ leukocytes in vivo. We provide further evidence that CD4+ macrophages have an ancient origin and had already emerged in bony fish. We demonstrate the utility of this zebrafish resource for interrogating the complex behavior of immune cells at cellular resolution by the imaging of intimate contacts between teleost CD4+ T cells and mononuclear phagocytes. Most importantly, we reveal the conserved subspecialization of teleost CD4+ T cells in vivo. We demonstrate that the ancient and specialized tissues of the gills contain a resident population of il-4/13b-expressing Th2-like cells, which do not coexpress il-4/13a Additionally, we identify a contrasting population of regulatory T cell-like cells resident in the zebrafish gut mucosa, in marked similarity to that found in the intestine of mammals. Finally, we show that, as in mammals, zebrafish CD4+ T cells will infiltrate melanoma tumors and obtain a phenotype consistent with a type 2 immune microenvironment. We anticipate that this unique resource will prove invaluable for future investigation of T cell function in biomedical research, the development of vaccination and health management in aquaculture, and for further research into the evolution of adaptive immunity.
Collapse
Affiliation(s)
- Christopher T Dee
- Faculty of Life Sciences, The University of Manchester, Manchester M13 9PT, United Kingdom
| | - Raghavendar T Nagaraju
- Faculty of Life Sciences, The University of Manchester, Manchester M13 9PT, United Kingdom
| | - Emmanouil I Athanasiadis
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1HH, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge CB2 0PT, United Kingdom
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, Cambridge CB2 1QR, United Kingdom
| | - Caroline Gray
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield S10 2RX, United Kingdom
- Bateson Centre, University of Sheffield, Sheffield S10 2TN, United Kingdom; and
| | | | - Simon A Johnston
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield S10 2RX, United Kingdom
- Bateson Centre, University of Sheffield, Sheffield S10 2TN, United Kingdom; and
| | - Christopher J Secombes
- Scottish Fish Immunology Research Centre, School of Biological Sciences, University of Aberdeen, Aberdeen AB24 3UU, United Kingdom
| | - Ana Cvejic
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1HH, United Kingdom;
- Department of Haematology, University of Cambridge, Cambridge CB2 0PT, United Kingdom
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, Cambridge CB2 1QR, United Kingdom
| | - Adam F L Hurlstone
- Faculty of Life Sciences, The University of Manchester, Manchester M13 9PT, United Kingdom;
| |
Collapse
|
30
|
Day K, Waite LL, Alonso A, Irvin MR, Zhi D, Thibeault KS, Aslibekyan S, Hidalgo B, Borecki IB, Ordovas JM, Arnett DK, Tiwari HK, Absher DM. Heritable DNA Methylation in CD4+ Cells among Complex Families Displays Genetic and Non-Genetic Effects. PLoS One 2016; 11:e0165488. [PMID: 27792787 PMCID: PMC5085095 DOI: 10.1371/journal.pone.0165488] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 10/12/2016] [Indexed: 12/20/2022] Open
Abstract
DNA methylation at CpG sites is both heritable and influenced by environment, but the relative contributions of each to DNA methylation levels are unclear. We conducted a heritability analysis of CpG methylation in human CD4+ cells across 975 individuals from 163 families in the Genetics of Lipid-lowering Drugs and Diet Network (GOLDN). Based on a broad-sense heritability (H2) value threshold of 0.4, we identified 20,575 highly heritable CpGs among the 174,445 most variable autosomal CpGs (SD > 0.02). Tests for associations of heritable CpGs with genotype at 2,145,360 SNPs using 717 of 975 individuals showed that ~74% were cis-meQTLs (< 1 Mb away from the CpG), 6% of CpGs exhibited trans-meQTL associations (>1 Mb away from the CpG or located on a different chromosome), and 20% of CpGs showed no strong significant associations with genotype (based on a p-value threshold of 1e-7). Genes proximal to the genotype independent heritable CpGs were enriched for functional terms related to regulation of T cell activation. These CpGs were also among those that distinguished T cells from other blood cell lineages. Compared to genes proximal to meQTL-associated heritable CpGs, genotype independent heritable CpGs were moderately enriched in the same genomic regions that escape erasure during primordial germ cell development and could carry potential for generational transmission.
Collapse
Affiliation(s)
- Kenneth Day
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama, United States of America
| | - Lindsay L. Waite
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama, United States of America
- University of Alabama at Birmingham, School of Public Health, Department of Biostatistics, Birmingham, Alabama, United States of America
| | - Arnald Alonso
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama, United States of America
| | - Marguerite R. Irvin
- University of Alabama at Birmingham, School of Public Health, Department of Epidemiology, Birmingham, Alabama, United States of America
| | - Degui Zhi
- University of Alabama at Birmingham, School of Public Health, Department of Biostatistics, Birmingham, Alabama, United States of America
| | - Krista S. Thibeault
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama, United States of America
| | - Stella Aslibekyan
- University of Alabama at Birmingham, School of Public Health, Department of Epidemiology, Birmingham, Alabama, United States of America
| | - Bertha Hidalgo
- University of Alabama at Birmingham, School of Public Health, Department of Biostatistics, Birmingham, Alabama, United States of America
| | - Ingrid B. Borecki
- Washington University, Division of Statistical Genomics, St. Louis, Missouri, United States of America
| | - Jose M. Ordovas
- Tufts University, Jean Mayer USDA Human Nutrition Research Center on Aging, Boston, Massachusetts, United States of America
- IMDEA-Food, Madrid, Spain
| | - Donna K. Arnett
- University of Alabama at Birmingham, School of Public Health, Department of Epidemiology, Birmingham, Alabama, United States of America
| | - Hemant K. Tiwari
- University of Alabama at Birmingham, School of Public Health, Department of Biostatistics, Birmingham, Alabama, United States of America
| | - Devin M. Absher
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama, United States of America
- * E-mail:
| |
Collapse
|
31
|
Chen S, Sugiyama K, Inamura M, Tanaka S, Onda K, Yin H, Hirano T. Effects of insulin on pharmacodynamics of immunosuppressive drugs against mitogen-activated human peripheral blood mononuclear cells. Immunopharmacol Immunotoxicol 2016; 38:372-8. [PMID: 27443993 DOI: 10.1080/08923973.2016.1214143] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
CONTEXT Diabetes mellitus is one of the most common causes of chronic renal failure. Immunosuppressive efficacies of glucocorticoids, calcineurin inhibitors, and mycophenolic acid are possibly affected by insulin after renal transplantation in these patients. OBJECTIVES We investigated the effects of insulin on responses of mitogen-activated human peripheral blood mononuclear cells (PBMCs) to several immunosuppressive drugs. MATERIALS AND METHODS Antiproliferative efficacies of prednisolone, hydrocortisone, cyclosporine, tacrolimus, and mycophenolic acid against concanavalin A-stimulated PBMCs were evaluated in the presence of physiological (5 μunits/mL) and super physiological (50 μunits/mL) concentrations of insulin. Insulin-receptor expressions on PBMCs were evaluated by flow cytometry. RESULTS Insulin itself had no effects on the mitogen-induced proliferation of PBMCs. The IC50 values of cyclosporine against the mitogen-activated PBMCs in the presence of 5 or 50 μunits/mL insulin were significantly higher than those of cyclosporine without insulin (p < 0.05). The IC50 values of mycophenolic acid significantly increased by 50 μunits/mL insulin (p < 0.01). Insulin receptors were detected on the mitogen-activated CD4(+)/CD14(+ )cells in PBMCs. DISCUSSION AND CONCLUSIONS These results indicate that insulin at even physiological concentration attenuates suppressive efficacies of several immunosuppressive drugs against mitogen-activated proliferation of human PBMCs, possibly via insulin receptors. Insulin used in dialysis patients accompanying diabetes mellitus is suggested to attenuate efficacies of immunosuppressive drugs after renal transplantation.
Collapse
Affiliation(s)
- Shuiling Chen
- a Department of Clinical Pharmacology, School of Pharmacy , Tokyo University of Pharmacy and Life Sciences , Tokyo , Japan ;,b Cardiovascular Department , Xiyuan Hospital, China Academy of Chinese Medical Sciences , Beijing , P.R. China
| | - Kentaro Sugiyama
- a Department of Clinical Pharmacology, School of Pharmacy , Tokyo University of Pharmacy and Life Sciences , Tokyo , Japan
| | - Mariko Inamura
- a Department of Clinical Pharmacology, School of Pharmacy , Tokyo University of Pharmacy and Life Sciences , Tokyo , Japan
| | - Sachiko Tanaka
- a Department of Clinical Pharmacology, School of Pharmacy , Tokyo University of Pharmacy and Life Sciences , Tokyo , Japan
| | - Kenji Onda
- a Department of Clinical Pharmacology, School of Pharmacy , Tokyo University of Pharmacy and Life Sciences , Tokyo , Japan
| | - Huijun Yin
- b Cardiovascular Department , Xiyuan Hospital, China Academy of Chinese Medical Sciences , Beijing , P.R. China
| | - Toshihiko Hirano
- a Department of Clinical Pharmacology, School of Pharmacy , Tokyo University of Pharmacy and Life Sciences , Tokyo , Japan
| |
Collapse
|
32
|
Greeson JM, Gettes DR, Spitsin S, Dubé B, Benton TD, Lynch KG, Douglas SD, Evans DL. The Selective Serotonin Reuptake Inhibitor Citalopram Decreases Human Immunodeficiency Virus Receptor and Coreceptor Expression in Immune Cells. Biol Psychiatry 2016; 80:33-9. [PMID: 26725193 PMCID: PMC4862937 DOI: 10.1016/j.biopsych.2015.11.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 10/02/2015] [Accepted: 11/02/2015] [Indexed: 11/15/2022]
Abstract
BACKGROUND This study investigated whether the selective serotonin reuptake inhibitor (SSRI) citalopram downregulates the expression of the human immunodeficiency virus (HIV) receptor cluster of differentiation 4 (CD4) and coreceptors chemokine receptor type 5 and chemokine-related receptor type 4 (CCR5 and CXCR4) on peripheral blood mononuclear cells (PBMCs) and macrophages ex vivo as a potential mechanism of reducing susceptibility to HIV infection. METHODS The sample included 150 participants 18-58 years old (59% women, 65% African American, 61% with depression). Monocyte-depleted PBMCs were treated with phytohemagglutinin for 72 hours and then cultured in the presence of interleukin-2 with vehicle control or the SSRI (10(-6) mol/L) for 2 hours. To generate monocyte-derived macrophages, monocytes were cultured for 7 days, after which either vehicle control or SSRI (10(-6) mol/L) was added for 2 hours. RNA was collected from both cell types, and messenger RNA expression of CD4, CCR5, and CXCR4 was measured by real-time polymerase chain reaction. RESULTS In PBMCs, SSRI treatment decreased expression of CD4 (p = .009), CCR5 (p = .008), and CXCR4 (p < .0001). In monocyte-derived macrophages, SSRI treatment decreased expression of CD4 (p < .0001) and CXCR4 (p = .0003), but not CCR5 (p = .71). The suppressive effects of the SSRI on receptor expression did not differ as a function of depression diagnosis or depressive symptom severity. CONCLUSIONS Treatment with the SSRI at a physiologic dose decreased CD4, CCR5, and CXCR4 expression on PBMCs and macrophages ex vivo. These findings suggest that SSRI treatment, independent of depression status, downregulates HIV receptor and coreceptor expression and may reduce susceptibility of immune cells to HIV infection and decrease inflammation. If clinical trials confirm the present findings, ultimately there may be a role for using SSRI treatment adjunctively in HIV and acquired immunodeficiency syndrome.
Collapse
Affiliation(s)
- Jeffrey M. Greeson
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - David R. Gettes
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Sergei Spitsin
- Division of Allergy and Immunology, The Children’s Hospital of Philadelphia Research Institute, Philadelphia, PA 19104
| | - Benoit Dubé
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Tami D. Benton
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA,Department of Child and Adolescent Psychiatry and Behavioral Science, The Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Kevin G. Lynch
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Steven D. Douglas
- Division of Allergy and Immunology, The Children’s Hospital of Philadelphia Research Institute, Philadelphia, PA 19104,Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Dwight L. Evans
- Departments of Psychiatry, Medicine, and Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA,Corresponding Author: Dwight L. Evans, M.D., University of Pennsylvania Perelman School of Medicine, Department of Psychiatry, 305 Blockley Hall, 423 Guardian Drive, Philadelphia, PA 19104, TEL: 215-662-2899,
| |
Collapse
|
33
|
Takizawa F, Magadan S, Parra D, Xu Z, Korytář T, Boudinot P, Sunyer JO. Novel Teleost CD4-Bearing Cell Populations Provide Insights into the Evolutionary Origins and Primordial Roles of CD4+ Lymphocytes and CD4+ Macrophages. THE JOURNAL OF IMMUNOLOGY 2016; 196:4522-35. [PMID: 27183628 DOI: 10.4049/jimmunol.1600222] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 03/28/2016] [Indexed: 12/24/2022]
Abstract
Tetrapods contain a single CD4 coreceptor with four Ig domains that likely arose from a primordial two-domain ancestor. Notably, teleost fish contain two CD4 genes. Like tetrapod CD4, CD4-1 of rainbow trout includes four Ig domains, whereas CD4-2 contains only two. Because CD4-2 is reminiscent of the prototypic two-domain CD4 coreceptor, we hypothesized that by characterizing the cell types bearing CD4-1 and CD4-2, we would shed light into the evolution and primordial roles of CD4-bearing cells. Using newly established mAbs against CD4-1 and CD4-2, we identified two bona-fide CD4(+) T cell populations: a predominant lymphocyte population coexpressing surface CD4-1 and CD4-2 (CD4 double-positive [DP]), and a minor subset expressing only CD4-2 (CD4-2 single-positive [SP]). Although both subsets produced equivalent levels of Th1, Th17, and regulatory T cell cytokines upon bacterial infection, CD4-2 SP lymphocytes were less proliferative and displayed a more restricted TCRβ repertoire. These data suggest that CD4-2 SP cells represent a functionally distinct population and may embody a vestigial CD4(+) T cell subset, the roles of which reflect those of primeval CD4(+) T cells. Importantly, we also describe the first CD4(+) monocyte/macrophage population in a nonmammalian species. Of all myeloid subsets, we found the CD4(+) population to be the most phagocytic, whereas CD4(+) lymphocytes lacked this capacity. This study fills in an important gap in the knowledge of teleost CD4-bearing leukocytes, thus revealing critical insights into the evolutionary origins and primordial roles of CD4(+) lymphocytes and CD4(+) monocytes/macrophages.
Collapse
Affiliation(s)
- Fumio Takizawa
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Susana Magadan
- Virologie et Immunologie Moléculaires, INRA, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - David Parra
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Universitat Autònoma de Barcelona, Barcelona 08193, Spain; and
| | - Zhen Xu
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104; Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agriculture University, Wuhan, Hubei 430070, China
| | - Tomáš Korytář
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Pierre Boudinot
- Virologie et Immunologie Moléculaires, INRA, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - J Oriol Sunyer
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104;
| |
Collapse
|
34
|
Abstract
In vivo depletion of T lymphocytes is a means of studying the role of specific T cell populations during defined phases of in vivo immune responses. In this unit, a protocol is provided for injecting monoclonal antibodies (mAbs) into wild-type adult mice. Depletion of the appropriate subset of cells is verified by flow cytometry analysis of lymph node and spleen cell suspensions in pilot experiments. Once conditions have been established, depleted mice can be used to study the impact of T cell subsets on a variety of in vivo immune responses. The depleted condition may be maintained by repeated injections of the monoclonal antibody, or reversed by normal thymopoiesis following discontinuation of antibody administration.
Collapse
Affiliation(s)
- Karen Laky
- National Institutes of Health, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland
| | | |
Collapse
|
35
|
Ivanov S, Scallan JP, Kim KW, Werth K, Johnson MW, Saunders BT, Wang PL, Kuan EL, Straub AC, Ouhachi M, Weinstein EG, Williams JW, Briseño C, Colonna M, Isakson BE, Gautier EL, Förster R, Davis MJ, Zinselmeyer BH, Randolph GJ. CCR7 and IRF4-dependent dendritic cells regulate lymphatic collecting vessel permeability. J Clin Invest 2016; 126:1581-91. [PMID: 26999610 PMCID: PMC4811132 DOI: 10.1172/jci84518] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 02/09/2016] [Indexed: 12/18/2022] Open
Abstract
Lymphatic collecting vessels direct lymph into and from lymph nodes (LNs) and can become hyperpermeable as the result of a previous infection. Enhanced permeability has been implicated in compromised immunity due to reduced flow of lymph and immune cells to LNs, which are the primary site of antigen presentation to T cells. Presently, very little is known about the molecular signals that affect lymphatic collecting vessel permeability. Here, we have shown that lymphatic collecting vessel permeability is controlled by CCR7 and that the chronic hyperpermeability of collecting vessels observed in Ccr7-/- mice is followed by vessel fibrosis. Reexpression of CCR7 in DCs, however, was sufficient to reverse the development of such fibrosis. IFN regulatory factor 4-positive (IRF4+) DCs constitutively interacted with collecting lymphatics, and selective ablation of this DC subset in Cd11c-Cre Irf4fl/fl mice also rendered lymphatic collecting vessels hyperpermeable and fibrotic. Together, our data reveal that CCR7 plays multifaceted roles in regulating collecting vessel permeability and fibrosis, with one of the key players being IRF4-dependent DCs.
Collapse
Affiliation(s)
- Stoyan Ivanov
- Department of Pathology and Immunology, Washington University
School of Medicine, St. Louis, Missouri, USA
| | - Joshua P. Scallan
- Department of Medical Pharmacology and Physiology, University of
Missouri, Columbia, Missouri, USA
| | - Ki-Wook Kim
- Department of Pathology and Immunology, Washington University
School of Medicine, St. Louis, Missouri, USA
| | - Kathrin Werth
- Institute of Immunology, Hannover Medical School, Hannover,
Germany
| | - Michael W. Johnson
- Department of Pathology and Immunology, Washington University
School of Medicine, St. Louis, Missouri, USA
- Division of Biostatistics, Washington University School of
Medicine, St. Louis, Missouri, USA
| | - Brian T. Saunders
- Department of Pathology and Immunology, Washington University
School of Medicine, St. Louis, Missouri, USA
| | - Peter L. Wang
- Department of Pathology and Immunology, Washington University
School of Medicine, St. Louis, Missouri, USA
| | - Emma L. Kuan
- Immunology Institute, Icahn School of Medicine at Mount Sinai,
New York, New York, USA
| | - Adam C. Straub
- Robert M. Berne Cardiovascular Research Center, University of
Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Melissa Ouhachi
- INSERM UMRS 1166, Sorbonne Universités, University Pierre and
Marie Curie (UPMC), University of Paris 06, Pitié-Salpêtrière Hospital, Paris, France
| | - Erica G. Weinstein
- Immunology Institute, Icahn School of Medicine at Mount Sinai,
New York, New York, USA
| | - Jesse W. Williams
- Department of Pathology and Immunology, Washington University
School of Medicine, St. Louis, Missouri, USA
| | - Carlos Briseño
- Department of Pathology and Immunology, Washington University
School of Medicine, St. Louis, Missouri, USA
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University
School of Medicine, St. Louis, Missouri, USA
| | - Brant E. Isakson
- Robert M. Berne Cardiovascular Research Center, University of
Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Emmanuel L. Gautier
- Department of Pathology and Immunology, Washington University
School of Medicine, St. Louis, Missouri, USA
- INSERM UMRS 1166, Sorbonne Universités, University Pierre and
Marie Curie (UPMC), University of Paris 06, Pitié-Salpêtrière Hospital, Paris, France
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, Hannover,
Germany
| | - Michael J. Davis
- Department of Medical Pharmacology and Physiology, University of
Missouri, Columbia, Missouri, USA
| | - Bernd H. Zinselmeyer
- Department of Pathology and Immunology, Washington University
School of Medicine, St. Louis, Missouri, USA
| | - Gwendalyn J. Randolph
- Department of Pathology and Immunology, Washington University
School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
36
|
Hewavisenti RV, Morris KM, O'Meally D, Cheng Y, Papenfuss AT, Belov K. The identification of immune genes in the milk transcriptome of the Tasmanian devil (Sarcophilus harrisii). PeerJ 2016; 4:e1569. [PMID: 26793426 PMCID: PMC4715465 DOI: 10.7717/peerj.1569] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 12/15/2015] [Indexed: 01/25/2023] Open
Abstract
Tasmanian devil (Sarcophilus harrisii) pouch young, like other marsupials, are born underdeveloped and immunologically naïve, and are unable to mount an adaptive immune response. The mother’s milk provides nutrients for growth and development as well as providing passive immunity. To better understand immune response in this endangered species, we set out to characterise the genes involved in passive immunity by sequencing and annotating the transcriptome of a devil milk sample collected during mid-lactation. At mid-lactation we expect the young to have heightened immune responses, as they have emerged from the pouch, encountering new pathogens. A total of 233,660 transcripts were identified, including approximately 17,827 unique protein-coding genes and 846 immune genes. The most highly expressed transcripts were dominated by milk protein genes such as those encoding early lactation protein, late lactation proteins, α-lactalbumin, α-casein and β-casein. There were numerous highly expressed immune genes including lysozyme, whey acidic protein, ferritin and major histocompatibility complex I and II. Genes encoding immunoglobulins, antimicrobial peptides, chemokines and immune cell receptors were also identified. The array of immune genes identified in this study reflects the importance of the milk in providing immune protection to Tasmanian devil young and provides the first insight into Tasmanian devil milk.
Collapse
Affiliation(s)
| | - Katrina M Morris
- Faculty of Veterinary Science, University of Sydney , Sydney , Australia
| | - Denis O'Meally
- Faculty of Veterinary Science, University of Sydney , Sydney , Australia
| | - Yuanyuan Cheng
- Faculty of Veterinary Science, University of Sydney , Sydney , Australia
| | - Anthony T Papenfuss
- Bioinformatics Division, The Walter and Eliza Hall Institute for Medical Research, Parkville, Australia; Bioinformatics and Cancer Genomics, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - Katherine Belov
- Faculty of Veterinary Science, University of Sydney , Sydney , Australia
| |
Collapse
|
37
|
Huang Y, Yang Z, Huang C, McGowan J, Casper T, Sun D, Born WK, O'Brien RL. γδ T Cell-Dependent Regulatory T Cells Prevent the Development of Autoimmune Keratitis. THE JOURNAL OF IMMUNOLOGY 2015; 195:5572-81. [PMID: 26566677 DOI: 10.4049/jimmunol.1501604] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 10/14/2015] [Indexed: 12/12/2022]
Abstract
To prevent potentially damaging inflammatory responses, the eye actively promotes local immune tolerance via a variety of mechanisms. Owing to trauma, infection, or other ongoing autoimmunity, these mechanisms sometimes fail, and an autoimmune disorder may develop in the eye. In mice of the C57BL/10 (B10) background, autoimmune keratitis often develops spontaneously, particularly in the females. Its incidence is greatly elevated in the absence of γδ T cells, such that ∼80% of female B10.TCRδ(-/-) mice develop keratitis by 18 wk of age. In this article, we show that CD8(+) αβ T cells are the drivers of this disease, because adoptive transfer of CD8(+), but not CD4(+), T cells to keratitis-resistant B10.TCRβ/δ(-/-) hosts induced a high incidence of keratitis. This finding was unexpected because in other autoimmune diseases, more often CD4(+) αβ T cells, or both CD4(+) and CD8(+) αβ T cells, mediate the disease. Compared with wild-type B10 mice, B10.TCRδ(-/-) mice also show increased percentages of peripheral memory phenotype CD8(+) αβ T cells, along with an elevated frequency of CD8(+) αβ T cells biased to produce inflammatory cytokines. In addition, B10.TCRδ-/- mice have fewer peripheral CD4(+) CD25(+) Foxp3(+) αβ regulatory T cells (Tregs), which express lower levels of receptors needed for Treg development and function. Together, these observations suggest that in B10 background mice, γδ T cells are required to generate adequate numbers of CD4(+) CD25(+) Foxp3(+) Tregs, and that in B10.TCRδ(-/-) mice a Treg deficiency allows dysregulated effector or memory CD8(+) αβ T cells to infiltrate the cornea and provoke an autoimmune attack.
Collapse
Affiliation(s)
- Yafei Huang
- Department of Biomedical Research, National Jewish Health, Denver, CO 80206; Department of Immunology and Microbiology, University of Colorado Denver School of Medicine, Aurora, CO 80045; Joint Laboratory for Stem Cell Engineering and Technology Transfer, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City 430030, People's Republic of China
| | - Zhifang Yang
- Department of Biomedical Research, National Jewish Health, Denver, CO 80206; Department of Immunology and Microbiology, University of Colorado Denver School of Medicine, Aurora, CO 80045; Department of Breast and Thyroid Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City 430030, People's Republic of China; and
| | - Chunjian Huang
- Department of Biomedical Research, National Jewish Health, Denver, CO 80206
| | - Jessica McGowan
- Department of Biomedical Research, National Jewish Health, Denver, CO 80206
| | - Tamara Casper
- Department of Biomedical Research, National Jewish Health, Denver, CO 80206
| | - Deming Sun
- Doheny Eye Institute, Department of Ophthalmology, University of Southern California, Los Angeles, CA 90033
| | - Willi K Born
- Department of Biomedical Research, National Jewish Health, Denver, CO 80206; Department of Immunology and Microbiology, University of Colorado Denver School of Medicine, Aurora, CO 80045
| | - Rebecca L O'Brien
- Department of Biomedical Research, National Jewish Health, Denver, CO 80206; Department of Immunology and Microbiology, University of Colorado Denver School of Medicine, Aurora, CO 80045;
| |
Collapse
|
38
|
Zhang ZM, Shi R, Chen H, Zhang Z. Lesional accumulation of CD8+ cells in sciatic nerves of experimental autoimmune neuritis rats. Neurol Sci 2015; 37:199-203. [DOI: 10.1007/s10072-015-2384-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 09/16/2015] [Indexed: 12/12/2022]
|
39
|
Roy R, Haase T, Ma N, Bader A, Becker M, Seifert M, Choi YH, Falk V, Stamm C. Decellularized amniotic membrane attenuates postinfarct left ventricular remodeling. J Surg Res 2015; 200:409-19. [PMID: 26421709 DOI: 10.1016/j.jss.2015.08.022] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 07/24/2015] [Accepted: 08/14/2015] [Indexed: 10/23/2022]
Abstract
BACKGROUND Placenta and amnion have been suggested as sources of juvenile cells and tissues for use in surgical regenerative medicine. We previously determined the impact of amniotic epithelial cells induced to undergo epithelial-to-mesenchymal transition (EMT) on myocardial remodeling processes and now evaluated the effects of naïve and processed amniotic membrane (AM) on postischemic left ventricular (LV) geometry and function. METHODS Human AM was used in unmodified form (AM), after EMT induction by transforming growth factor β (EMT-AM), and after decellularization (Decell-AM). After characterization by histology, electron microscopy, splenocyte proliferation assay, and cytokine release, myocardial infarction was induced in 6-8-week old male BALB/c mice by permanent left anterior descending coronary occlusion, and AM patches were sutured to the anterior LV surface (n = 10 per group). Infarcted hearts without AM or sham-operated mice were used as controls (n = 10 each). After 4 weeks, LV pressure-volume curves were recorded using a conductance catheter before the animals were sacrificed and the hearts analyzed by histology. RESULTS TGF-ß treatment induced EMT-like changes in amniotic epithelial cells but increased AM xenoreactivity in vitro (splenocyte proliferation) and in vivo (CD4+ cell invasion). Moreover, in vitro interleukin-6 release from AM and from cardiac fibroblasts co-incubated with AM was 300- or 100-fold higher than that of interleukin-10, whereas Decell-AM did not release any cytokines. AM- and Decell-AM-treated hearts had smaller infarct size and greater infarct scar thickness than infarct control hearts, but there was no difference in myocardial capillary density or the number of TUNEL positive apoptotic cells. LV contractile function was better in the AM and EMT-AM groups than in infarcted control hearts, but dP/dt max, dP/dt min, stroke work, and cardiac output were best preserved in mice treated with Decell-AM. Volume-based parameters (LV end-systolic and end-diastolic volume as well as LV ejection fraction) did not differ between AM and Decell-AM. CONCLUSIONS Decellularized AM supports postinfarct ventricular dynamics independent of the actual regeneration processes. As a cell-free approach to support the infarcted heart, this concept warrants further investigation.
Collapse
Affiliation(s)
- Rajika Roy
- Berlin-Brandenburg Center for Regenerative Therapies, Berlin, Germany
| | - Tobias Haase
- Berlin-Brandenburg Center for Regenerative Therapies, Berlin, Germany
| | - Nan Ma
- Berlin-Brandenburg Center for Regenerative Therapies, Berlin, Germany
| | - Andreas Bader
- Berlin-Brandenburg Center for Regenerative Therapies, Berlin, Germany
| | - Matthias Becker
- Berlin-Brandenburg Center for Regenerative Therapies, Berlin, Germany
| | - Martina Seifert
- Berlin-Brandenburg Center for Regenerative Therapies, Berlin, Germany
| | | | | | - Christof Stamm
- Berlin-Brandenburg Center for Regenerative Therapies, Berlin, Germany; Deutsches Herzzentrum Berlin, Berlin, Germany.
| |
Collapse
|
40
|
Remmele CW, Luther CH, Balkenhol J, Dandekar T, Müller T, Dittrich MT. Integrated inference and evaluation of host-fungi interaction networks. Front Microbiol 2015; 6:764. [PMID: 26300851 PMCID: PMC4523839 DOI: 10.3389/fmicb.2015.00764] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 07/13/2015] [Indexed: 12/18/2022] Open
Abstract
Fungal microorganisms frequently lead to life-threatening infections. Within this group of pathogens, the commensal Candida albicans and the filamentous fungus Aspergillus fumigatus are by far the most important causes of invasive mycoses in Europe. A key capability for host invasion and immune response evasion are specific molecular interactions between the fungal pathogen and its human host. Experimentally validated knowledge about these crucial interactions is rare in literature and even specialized host–pathogen databases mainly focus on bacterial and viral interactions whereas information on fungi is still sparse. To establish large-scale host–fungi interaction networks on a systems biology scale, we develop an extended inference approach based on protein orthology and data on gene functions. Using human and yeast intraspecies networks as template, we derive a large network of pathogen–host interactions (PHI). Rigorous filtering and refinement steps based on cellular localization and pathogenicity information of predicted interactors yield a primary scaffold of fungi–human and fungi–mouse interaction networks. Specific enrichment of known pathogenicity-relevant genes indicates the biological relevance of the predicted PHI. A detailed inspection of functionally relevant subnetworks reveals novel host–fungal interaction candidates such as the Candida virulence factor PLB1 and the anti-fungal host protein APP. Our results demonstrate the applicability of interolog-based prediction methods for host–fungi interactions and underline the importance of filtering and refinement steps to attain biologically more relevant interactions. This integrated network framework can serve as a basis for future analyses of high-throughput host–fungi transcriptome and proteome data.
Collapse
Affiliation(s)
| | | | | | - Thomas Dandekar
- Department of Bioinformatics, University of Würzburg Würzburg, Germany
| | - Tobias Müller
- Department of Bioinformatics, University of Würzburg Würzburg, Germany
| | - Marcus T Dittrich
- Department of Bioinformatics, University of Würzburg Würzburg, Germany ; Department of Human Genetics, University of Würzburg Würzburg, Germany
| |
Collapse
|
41
|
Wobser M, Roth S, Reinartz T, Rosenwald A, Goebeler M, Geissinger E. CD68 expression is a discriminative feature of indolent cutaneous CD8-positive lymphoid proliferation and distinguishes this lymphoma subtype from other CD8-positive cutaneous lymphomas. Br J Dermatol 2015; 172:1573-1580. [PMID: 25524664 DOI: 10.1111/bjd.13628] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/10/2014] [Indexed: 12/16/2022]
Abstract
BACKGROUND Indolent cutaneous CD8+ lymphoid proliferation is a recently described rare entity among cutaneous T-cell lymphomas that typically presents with solitary skin lesions at acral sites. Separation from otherwise aggressive T-cell lymphomas bearing a cytotoxic CD8+ phenotype is fundamental to avoid unnecessary harmful treatment. However, up to now, no reliable discriminative marker has been identified. OBJECTIVES Motivated by these diagnostic quandaries, we have analyzed a large series of archived formalin-fixed paraffin-embedded (FFPE) specimens of atypical CD8+ cutaneous infiltrates with clear-cut diagnosis and clinical follow-up (n = 44) including five cases of indolent CD8+ lymphoid proliferation by using immunohistochemistry with the aim of obtaining markers predictive of subtype assignment. RESULTS We identified exclusive expression of CD68 by lymphoma cells within the subgroup of indolent CD8+ lymphoid proliferation (5/5 cases). Specific CD68 expression in this entity was confirmed by the application of several monoclonal antibodies (KP1, PG-M1, KiM1P) against the CD68 molecule available for FFPE tissue. In contrast, none of the infiltrates of the other CD8+ cutaneous lymphoma entities stained positive for CD68 (0/39). CONCLUSIONS Based on these observations, we suggest CD68 as a new discriminative marker which is helpful in distinguishing indolent CD8+ lymphoid proliferation from other CD8+ cutaneous lymphomas in ambiguous cases.
Collapse
Affiliation(s)
- M Wobser
- Department of Dermatology, Comprehensive Cancer Center Mainfranken, University Hospital Wuerzburg, Josef-Schneider-Str. 2, Wuerzburg, 97080, Germany
| | - S Roth
- Institute of Pathology, Comprehensive Cancer Center Mainfranken, University of Wuerzburg, Wuerzburg, Germany
| | - T Reinartz
- Department of Dermatology, Comprehensive Cancer Center Mainfranken, University Hospital Wuerzburg, Josef-Schneider-Str. 2, Wuerzburg, 97080, Germany
| | - A Rosenwald
- Institute of Pathology, Comprehensive Cancer Center Mainfranken, University of Wuerzburg, Wuerzburg, Germany
| | - M Goebeler
- Department of Dermatology, Comprehensive Cancer Center Mainfranken, University Hospital Wuerzburg, Josef-Schneider-Str. 2, Wuerzburg, 97080, Germany
| | - E Geissinger
- Institute of Pathology, Comprehensive Cancer Center Mainfranken, University of Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
42
|
Short-term high-fat diet increases macrophage markers in skeletal muscle accompanied by impaired insulin signalling in healthy male subjects. Clin Sci (Lond) 2014; 128:143-51. [PMID: 25148551 DOI: 10.1042/cs20140179] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Macrophage markers in skeletal muscle of obese subjects are elevated and inversely relate to insulin sensitivity. The present study aimed to investigate whether short-term high-fat high-calorie (HFHC) diet already increases macrophage markers and affects glucose metabolism in skeletal muscle of healthy lean subjects. Muscle biopsies were obtained from 24 healthy lean young men before and after a 5-day HFHC-diet. mRNA expression levels of relevant genes in muscle and glucose, insulin, C-peptide and cholesteryl ester transfer protein (CETP) levels in plasma were measured. In addition, we assessed hepatic triacylglycerol ('triglyceride') (HTG) content by magnetic resonance spectroscopy and subcutaneous white adipose tissue (sWAT) biopsies were analysed histologically from a subset of subjects (n=8). A 5-day HFHC-diet markedly increased skeletal muscle mRNA expression of the general macrophage markers CD68 (3.7-fold, P<0.01) and CD14 (3.2-fold, P<0.01), as well as the M1 macrophage markers MARCO (11.2-fold, P<0.05), CD11c (1.8-fold, P<0.05) and MRC1 (1.7-fold, P<0.05). This was accompanied by down-regulation of SLC2A4 and GYS1 mRNA expression, and elevated plasma glucose (+4%, P<0.001) and insulin (+55%, P<0.001) levels together with homoeostasis model assessment of insulin resistance (HOMA-IR) (+48%, P<0.001), suggesting development of insulin resistance (IR). Furthermore, the HFHC-diet markedly increased HTG (+118%, P<0.001) and plasma CETP levels (+21%, P<0.001), a marker of liver macrophage content, whereas sWAT macrophage content remained unchanged. In conclusion, short-term HFHC-diet increases expression of macrophage markers in skeletal muscle of healthy men accompanied by reduced markers of insulin signalling and development of IR. Therefore, recruitment of macrophages into muscle may be an early event in development of IR in response to short-term HFHC-feeding.
Collapse
|
43
|
Gale G, Ostman S, Saalman R, Telemo E, Jontell M, Hasséus B. Immunophenotype in orofacial granulomatosis with and without Crohn's disease. Med Oral Patol Oral Cir Bucal 2014; 19:e584-591. [PMID: 25350593 PMCID: PMC4259375 DOI: 10.4317/medoral.20187] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 08/22/2014] [Indexed: 01/29/2023] Open
Abstract
Objectives: The aim of this investigation was to characterise and compare the inflammatory infiltrates in patients with orofacial granulomatosis solely (OFG-S) and OFG with coexisting Crohn’s disease (OFG+CD).
Study Design: Biopsy specimens with granulomas were obtained from patients with OFG-S (n=11) and OFG+CD (n=11) and immunostained with antibodies against CD1a, CD3, CD4, CD8, CD11c, CD20, CD68 and mast cell tryptase, followed by quantitative analysis.
Results: Analyses of the connective tissue revealed a significantly higher number of CD3-expressing T cells and CD11c-expressing dendritic cells in the connective tissue of patients with OFG-S compared to patients with OFG+CD. Mast cells displayed a high level of activation, although no significant difference was detected when comparing the two groups.
Conclusions: The results show a different composition of the inflammatory infiltrate in patients with OFG-S compared to patients with OFG+CD. The present observations support that partly divergent immune mechanisms are involved in these two different subcategories of OFG.
Key words:Granulomas, autoimmunity, T cells, B cells, dendritic cells, children, adults.
Collapse
Affiliation(s)
- Gita Gale
- Department of Oral Medicine and Pathology, Institute of Odontology The Sahlgrenska Academy, University of Gothenburg, Box 450, SE405 30 Gothenburg, Sweden,
| | | | | | | | | | | |
Collapse
|
44
|
Liu C, Guo Q, Li J, Wang S, Wang Y, Li B, Yang H. Identification of rabbit annulus fibrosus-derived stem cells. PLoS One 2014; 9:e108239. [PMID: 25259600 PMCID: PMC4178129 DOI: 10.1371/journal.pone.0108239] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 08/26/2014] [Indexed: 12/20/2022] Open
Abstract
Annulus fibrosus (AF) injuries can lead to substantial deterioration of intervertebral disc (IVD) which characterizes degenerative disc disease (DDD). However, treatments for AF repair/regeneration remain challenging due to the intrinsic heterogeneity of AF tissue at cellular, biochemical, and biomechanical levels. In this study, we isolated and characterized a sub-population of cells from rabbit AF tissue which formed colonies in vitro and could self-renew. These cells showed gene expression of typical surface antigen molecules characterizing mesenchymal stem cells (MSCs), including CD29, CD44, and CD166. Meanwhile, they did not express negative markers of MSCs such as CD4, CD8, and CD14. They also expressed Oct-4, nucleostemin, and SSEA-4 proteins. Upon induced differentiation they showed typical osteogenesis, chondrogenesis, and adipogenesis potential. Together, these AF-derived colony-forming cells possessed clonogenicity, self-renewal, and multi-potential differentiation capability, the three criteria characterizing MSCs. Such AF-derived stem cells may potentially be an ideal candidate for DDD treatments using cell therapies or tissue engineering approaches.
Collapse
Affiliation(s)
- Chen Liu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Orthopedic Institute, Soochow University, Suzhou, Jiangsu, China
| | - Qianping Guo
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Orthopedic Institute, Soochow University, Suzhou, Jiangsu, China
| | - Jun Li
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Shenghao Wang
- Orthopedic Institute, Soochow University, Suzhou, Jiangsu, China
| | - Yibin Wang
- Orthopedic Institute, Soochow University, Suzhou, Jiangsu, China
| | - Bin Li
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Orthopedic Institute, Soochow University, Suzhou, Jiangsu, China
- * E-mail: (BL); (HY)
| | - Huilin Yang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Orthopedic Institute, Soochow University, Suzhou, Jiangsu, China
- * E-mail: (BL); (HY)
| |
Collapse
|
45
|
Raeven RHM, Brummelman J, Pennings JLA, Nijst OEM, Kuipers B, Blok LER, Helm K, van Riet E, Jiskoot W, van Els CACM, Han WGH, Kersten GFA, Metz B. Molecular signatures of the evolving immune response in mice following a Bordetella pertussis infection. PLoS One 2014; 9:e104548. [PMID: 25137043 PMCID: PMC4138111 DOI: 10.1371/journal.pone.0104548] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 07/14/2014] [Indexed: 12/21/2022] Open
Abstract
Worldwide resurgence of pertussis necessitates the need for improvement of pertussis vaccines and vaccination strategies. Since natural infections induce a longer-lasting immunity than vaccinations, detailed knowledge of the immune responses following natural infection can provide important clues for such improvement. The purpose was to elucidate the kinetics of the protective immune response evolving after experimental Bordetella pertussis (B. pertussis) infection in mice. Data were collected from (i) individual analyses, i.e. microarray, flow cytometry, multiplex immunoassays, and bacterial clearance; (ii) twelve time points during the infection; and (iii) different tissues involved in the immune responses, i.e. lungs, spleen and blood. Combined data revealed detailed insight in molecular and cellular sequence of events connecting different phases (innate, bridging and adaptive) of the immune response following the infection. We detected a prolonged acute phase response, broad pathogen recognition, and early gene signatures of subsequent T-cell recruitment in the lungs. Activation of particular transcription factors and specific cell markers provided insight into the time course of the transition from innate towards adaptive immune responses, which resulted in a broad spectrum of systemic antibody subclasses and splenic Th1/Th17 memory cells against B. pertussis. In addition, signatures preceding the local generation of Th1 and Th17 cells as well as IgA in the lungs, considered key elements in protection against B. pertussis, were established. In conclusion, molecular and cellular immunological processes in response to live B. pertussis infection were unraveled, which may provide guidance in selecting new vaccine candidates that should evoke local and prolonged protective immune responses.
Collapse
Affiliation(s)
- René H. M. Raeven
- Intravacc, Bilthoven, The Netherlands
- Division of Drug Delivery Technology, Leiden Academic Centre for Drug Research, Leiden, The Netherlands
| | - Jolanda Brummelman
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Jeroen L. A. Pennings
- Centre for Health Protection (GZB), National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | | | - Betsy Kuipers
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | | | - Kina Helm
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | | | - Wim Jiskoot
- Division of Drug Delivery Technology, Leiden Academic Centre for Drug Research, Leiden, The Netherlands
| | - Cecile A. C. M. van Els
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Wanda G. H. Han
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Gideon F. A. Kersten
- Intravacc, Bilthoven, The Netherlands
- Division of Drug Delivery Technology, Leiden Academic Centre for Drug Research, Leiden, The Netherlands
| | | |
Collapse
|
46
|
High frequencies of polyfunctional CD8+ NK cells in chronic HIV-1 infection are associated with slower disease progression. J Virol 2014; 88:12397-408. [PMID: 25122796 DOI: 10.1128/jvi.01420-14] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
UNLABELLED Natural killer (NK) cells are effector and regulatory innate immune cells and play a critical role in the first line of defense against various viral infections. Although previous reports have indicated the vital contributions of NK cells to HIV-1 immune control, nongenetic NK cell parameters directly associated with slower disease progression have not been defined yet. In a longitudinal, retrospective study of 117 untreated HIV-infected subjects, we show that higher frequencies as well as the absolute numbers of CD8(+) CD3(-) lymphocytes are linked to delayed HIV-1 disease progression. We show that the majority of these cells are well-described blood NK cells. In a subsequent cross-sectional study, we demonstrate a significant loss of CD8(+) NK cells in untreated HIV-infected individuals, which correlated with HIV loads and inversely correlated with CD4(+) T cell counts. CD8(+) NK cells had modestly higher frequencies of CD57-expressing cells than CD8(-) cells, but CD8(+) and CD8(-) NK cells showed no differences in the expression of a number of activating and inhibiting NK cell receptors. However, CD8(+) NK cells exhibited a more functional profile, as detected by cytokine production and degranulation. IMPORTANCE We demonstrate that the frequency of highly functional CD8(+) NK cells is inversely associated with HIV-related disease markers and linked with delayed disease progression. These results thus indicate that CD8(+) NK cells represent a novel NK cell-derived, innate immune correlate with an improved clinical outcome in HIV infection.
Collapse
|
47
|
Mulder R, Banete A, Basta S. Spleen-derived macrophages are readily polarized into classically activated (M1) or alternatively activated (M2) states. Immunobiology 2014; 219:737-45. [PMID: 24954891 DOI: 10.1016/j.imbio.2014.05.005] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 04/25/2014] [Accepted: 05/29/2014] [Indexed: 10/25/2022]
Abstract
Bone marrow derived macrophages (BM-MΦ) that differentiate from precursor cells can be polarized into classically activated pro-inflammatory (M1) or alternatively activated (M2) states depending upon the cytokine microenvironment. We questioned whether tissue MΦ, such as spleen-derived MΦ (Sp-MΦ), have the ability to differentiate into M1 or M2 cells. We show in response to activation with IFN-gamma (IFN-γ) and lipopolysaccharide (LPS), that the Sp-MΦ readily acquired an M1 status indicated by up-regulation of iNOS mRNA, nitric oxide (NO) production, and the co-stimulatory molecule CD86. Conversely, Sp-MΦ exposed to IL-4 exhibited increased levels of mannose receptor (CD 206), arginase-1 (Arg)-1 mRNA expression, and significant urea production typical of M2 cells. At this stage of differentiation, the M2 Sp-MΦ were more efficient at phagocytosis of cell-associated antigens than their M1 counterparts. This polarization was not indefinite as the cells could revert back to their original state upon the removal of stimuli and exhibited flexibility to convert from M2 to M1. Remarkably, both M1 and M2 Sp-MΦ induced more CD4 expression on their cells surface after stimulation. We also demonstrate that adherent macrophages cultured for a short term in IL-4 enhances ARG-1 and YM-1 mRNA along with increasing urea producing capacity: traits indicative of an M2 phenotype. Moreover, in response to in vivo virus infection, the adherent macrophages obtained from spleens rapidly express iNOS. These results provide new evidence for the polarization capabilities of Sp-MΦ when exposed to pro-inflammatory or anti-inflammatory cytokines.
Collapse
Affiliation(s)
- Rylend Mulder
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Andra Banete
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Sameh Basta
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada.
| |
Collapse
|
48
|
Möller K, Boltze J, Pösel C, Seeger J, Stahl T, Wagner DC. Sterile inflammation after permanent distal MCA occlusion in hypertensive rats. J Cereb Blood Flow Metab 2014; 34:307-15. [PMID: 24220169 PMCID: PMC3915208 DOI: 10.1038/jcbfm.2013.199] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 10/18/2013] [Accepted: 10/21/2013] [Indexed: 12/30/2022]
Abstract
The pathophysiology of stroke is governed by immune reactions within and remote from the injured brain. Hypertension, a major cause and comorbidity of stroke, entails systemic vascular inflammation and may influence poststroke immune responses. This aspect is, however, underestimated in previous studies. Here we aimed to delineate the sequence of cellular inflammation after stroke in spontaneously hypertensive (SH) rats. Spontaneously hypertensive rats were subjected to permanent middle cerebral artery occlusion and killed after 1 or 4 days. Immune cells of the peripheral blood and those which have infiltrated the injured brain were identified and quantified by flow cytometry. The spatial distribution of myeloid cells and T lymphocytes, and the infarct volume were assessed by histology. We observed a concerted infiltration of immune cells into the ischemic brain of SH rats. At day 1, primarily neutrophils, monocytes, macrophages, and myeloid dendritic cells entered the brain, whereas the situation at day 4 was dominated by microglia, macrophages, lymphatic dendritic cells, and T cells. Postischemic inflammation did not cause secondary tissue damage during the subacute stage of experimental stroke in SH rats. Considering the intrinsic vascular pathology of SH rats, our study validates this strain for further translational research in poststroke inflammation.
Collapse
Affiliation(s)
- Karoline Möller
- 1] Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany [2] Institute of Veterinary Anatomy, University of Leipzig, Leipzig, Germany
| | - Johannes Boltze
- 1] Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany [2] Translational Centre for Regenerative Medicine, University of Leipzig, Leipzig, Germany [3] Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Claudia Pösel
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Johannes Seeger
- Institute of Veterinary Anatomy, University of Leipzig, Leipzig, Germany
| | | | - Daniel-Christoph Wagner
- 1] Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany [2] Translational Centre for Regenerative Medicine, University of Leipzig, Leipzig, Germany
| |
Collapse
|
49
|
Quantification of entry phenotypes of macrophage-tropic HIV-1 across a wide range of CD4 densities. J Virol 2013; 88:1858-69. [PMID: 24307580 DOI: 10.1128/jvi.02477-13] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Defining a macrophage-tropic phenotype for HIV-1 to assess a role in pathogenesis is complicated by the fact that HIV-1 isolates vary continuously in their ability to enter monocyte-derived macrophages (MDMs) in vitro, and MDMs vary in their ability to support HIV-1 entry. To overcome these limitations, we identified consistent differences in entry phenotypes between five paired blood-derived, T cell-tropic HIV-1 env genes, four of which are CCR5-using (R5) and one of which is CXCR4-using (X4), and cerebrospinal fluid (CSF)-derived, R5 macrophage-tropic env genes. We performed entry assays using the CD4- and CCR5-inducible Affinofile cell line, expressing a range of CD4 levels that approximates the range from MDMs to CD4(+) T cells. The macrophage-tropic viruses were significantly better at infecting cells expressing low levels of CD4 than the T cell-tropic viruses from the same subjects, with the titration of CD4 providing a distinctive and quantitative phenotype. This difference in CD4 utilization was not due to macrophage-tropic viruses being CD4 independent. Furthermore, macrophage-tropic viruses did not differ from paired T cell-tropic viruses in their ability to use low levels of CCR5 (tpaired = -1.39; P = 0.24) or their use of an alternative conformation of CCR5. We also infected MDMs with a panel of viruses and observed that infectivity of each virus differed across four donors and between three preparations from a single donor. We concluded that the evolutionary transition from replication in T cells to that in macrophages involves a phenotypic transition to acquire the ability to infect cells expressing low levels of CD4 and that this phenotype is more reliably measured in Affinofile cells than in macrophages. IMPORTANCE HIV-1 typically infects memory T cells by using CD4 and CCR5 to enter cells. The virus evolves to infect new cell types by changing the coreceptor from CCR5 to CXCR4 to infect naive T cells or adapting to the use of low levels of CD4 to infect macrophages. However, defining the phenotype of macrophage tropism has been difficult due to inherent variability in the use of macrophages generated in culture to support entry of HIV-1. We describe the use of Affinofile cells with inducible and variable levels of CD4 to identify a signature phenotype for macrophage-tropic HIV-1. The ability to define HIV-1 variants that have evolved an entry phenotype that allows more efficient entry into cells with low levels of CD4 sets the stage for a clearer placement of these variants in HIV-associated pathogenesis.
Collapse
|
50
|
Elevated CD3+ and CD8+ tumor-infiltrating immune cells correlate with prolonged survival in glioblastoma patients despite integrated immunosuppressive mechanisms in the tumor microenvironment and at the systemic level. J Neuroimmunol 2013; 264:71-83. [DOI: 10.1016/j.jneuroim.2013.08.013] [Citation(s) in RCA: 249] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 08/12/2013] [Accepted: 08/22/2013] [Indexed: 01/16/2023]
|