1
|
Shanmugavadivu A, Carter K, Zonouzi AP, Waisman A, Regen T. Protocol for the collection and analysis of the different immune cell subsets in the murine intestinal lamina propria. STAR Protoc 2024; 5:103154. [PMID: 38935510 PMCID: PMC11260864 DOI: 10.1016/j.xpro.2024.103154] [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: 03/14/2024] [Revised: 04/28/2024] [Accepted: 06/05/2024] [Indexed: 06/29/2024] Open
Abstract
The intestinal lamina propria (LP) is a leukocyte-rich cornerstone of the immune system owing to its vital role in immune surveillance and barrier defense against external pathogens. Here, we present a protocol for isolating and analyzing immune cell subsets from the mouse intestinal LP for further downstream applications. Starting from tissue collection and cleaning, epithelium removal, and enzymatic digestion to collection of single cells, we explain each step in detail to maximize the yield of immune cells from the intestinal LP.
Collapse
Affiliation(s)
- Arthi Shanmugavadivu
- Institute of Molecular Medicine, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; Lab of Neuroimmunology, VIB-UAntwerp Center for Molecular Neurology, 2610 Antwerp, Belgium.
| | - Katlynn Carter
- Institute of Molecular Medicine, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Aysan Poursadegh Zonouzi
- Institute of Molecular Medicine, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Ari Waisman
- Institute of Molecular Medicine, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Tommy Regen
- Institute of Molecular Medicine, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany.
| |
Collapse
|
2
|
Lassoued N, Yero A, Jenabian MA, Soret R, Pilon N. Efficient enzyme-free method to assess the development and maturation of the innate and adaptive immune systems in the mouse colon. Sci Rep 2024; 14:11063. [PMID: 38744932 PMCID: PMC11094196 DOI: 10.1038/s41598-024-61834-5] [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: 01/08/2024] [Accepted: 05/10/2024] [Indexed: 05/16/2024] Open
Abstract
Researchers who aim to globally analyze the gastrointestinal immune system via flow cytometry have many protocol options to choose from, with specifics generally tied to gut wall layers of interest. To get a clearer idea of the approach we should use on full-thickness colon samples from mice, we first undertook a systematic comparison of three tissue dissociation techniques: two based on enzymatic cocktails and the other one based on manual crushing. Using flow cytometry panels of general markers of lymphoid and myeloid cells, we found that the presence of cell-surface markers and relative cell population frequencies were more stable with the mechanical method. Both enzymatic approaches were associated with a marked decrease of several cell-surface markers. Using mechanical dissociation, we then developed two minimally overlapping panels, consisting of a total of 26 antibodies, for serial profiling of lymphoid and myeloid lineages from the mouse colon in greater detail. Here, we highlight how we accurately delineate these populations by manual gating, as well as the reproducibility of our panels on mouse spleen and whole blood. As a proof-of-principle of the usefulness of our general approach, we also report segment- and life stage-specific patterns of immune cell profiles in the colon. Overall, our data indicate that mechanical dissociation is more suitable and efficient than enzymatic methods for recovering immune cells from all colon layers at once. Additionally, our panels will provide researchers with a relatively simple tool for detailed immune cell profiling in the murine gastrointestinal tract, regardless of life stage or experimental conditions.
Collapse
Affiliation(s)
- Nejia Lassoued
- Molecular Genetics of Development Laboratory, Department of Biological Sciences, Université du Québec à Montréal, Montreal, QC, Canada
- Centre d'excellence en recherche sur les maladies orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montreal, QC, Canada
| | - Alexis Yero
- Centre d'excellence en recherche sur les maladies orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montreal, QC, Canada
- Human Immuno-Virology Laboratory, Department of Biological Sciences, Université du Québec à Montréal, Montreal, QC, Canada
| | - Mohammad-Ali Jenabian
- Centre d'excellence en recherche sur les maladies orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montreal, QC, Canada
- Human Immuno-Virology Laboratory, Department of Biological Sciences, Université du Québec à Montréal, Montreal, QC, Canada
| | - Rodolphe Soret
- Molecular Genetics of Development Laboratory, Department of Biological Sciences, Université du Québec à Montréal, Montreal, QC, Canada.
- Centre d'excellence en recherche sur les maladies orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montreal, QC, Canada.
| | - Nicolas Pilon
- Molecular Genetics of Development Laboratory, Department of Biological Sciences, Université du Québec à Montréal, Montreal, QC, Canada.
- Centre d'excellence en recherche sur les maladies orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montreal, QC, Canada.
- Department of Pediatrics, Université de Montréal, Montreal, QC, Canada.
| |
Collapse
|
3
|
DaMata JP, Zelkoski AE, Nhan PB, Ennis KHE, Kim JS, Lu Z, Malloy AMW. Dissociation protocols influence the phenotypes of lymphocyte and myeloid cell populations isolated from the neonatal lymph node. Front Immunol 2024; 15:1368118. [PMID: 38756770 PMCID: PMC11097666 DOI: 10.3389/fimmu.2024.1368118] [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: 01/10/2024] [Accepted: 04/18/2024] [Indexed: 05/18/2024] Open
Abstract
Frequencies and phenotypes of immune cells differ between neonates and adults in association with age-specific immune responses. Lymph nodes (LN) are critical tissue sites to quantify and define these differences. Advances in flow cytometry have enabled more multifaceted measurements of complex immune responses. Tissue processing can affect the immune cells under investigation that influence key findings. To understand the impact on immune cells in the LN after processing for single-cell suspension, we compared three dissociation protocols: enzymatic digestion, mechanical dissociation with DNase I treatment, and mechanical dissociation with density gradient separation. We analyzed cell yields, viability, phenotypic and maturation markers of immune cells from the lung-draining LN of neonatal and adult mice two days after intranasal respiratory syncytial virus (RSV) infection. While viability was consistent across age groups, the protocols influenced the yield of subsets defined by important phenotypic and activation markers. Moreover, enzymatic digestion did not show higher overall yields of conventional dendritic cells and macrophages from the LN. Together, our findings show that the three dissociation protocols have similar impacts on the number and viability of cells isolated from the neonatal and adult LN. However, enzymatic digestion impacts the mean fluorescence intensity of key lineage and activation markers that may influence experimental findings.
Collapse
Affiliation(s)
- Jarina P. DaMata
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, Uniformed Services University of Health Sciences (USUHS), Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Amanda E. Zelkoski
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, Uniformed Services University of Health Sciences (USUHS), Bethesda, MD, United States
| | - Paula B. Nhan
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, Uniformed Services University of Health Sciences (USUHS), Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Katherine H. E. Ennis
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, Uniformed Services University of Health Sciences (USUHS), Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Ji Sung Kim
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, Uniformed Services University of Health Sciences (USUHS), Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Zhongyan Lu
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, Uniformed Services University of Health Sciences (USUHS), Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Allison M. W. Malloy
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, Uniformed Services University of Health Sciences (USUHS), Bethesda, MD, United States
| |
Collapse
|
4
|
Burke Schinkel SC, Barros PO, Berthoud T, Byrareddy SN, McGuinty M, Cameron DW, Angel JB. Comparative analysis of human gut- and blood-derived mononuclear cells: contrasts in function and phenotype. Front Immunol 2024; 15:1336480. [PMID: 38444848 PMCID: PMC10912472 DOI: 10.3389/fimmu.2024.1336480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 02/05/2024] [Indexed: 03/07/2024] Open
Abstract
Introduction Alterations in the gut immune system have been implicated in various diseases.The challenge of obtaining gut tissues from healthy individuals, commonly performed via surgical explants, has limited the number of studies describing the phenotype and function of gut-derived immune cells in health. Methods Here, by means of recto-sigmoid colon biopsies obtained during routine care (colon cancer screening in healthy adults), the phenotype and function of immune cells present in the gut were described and compared to those found in blood. Results The proportion of CD4+, CD8+, MAIT, γδ+ T, and NK cells phenotype, expression of integrins, and ability to produce cytokine in response to stimulation with PMA and ionomycin. T cells in the gut were found to predominantly have a memory phenotype as compared to T cells in blood where a naïve phenotype predominates. Recto-sigmoid mononuclear cells also had higher PD-1 and Ki67 expression. Furthermore, integrin expression and cytokine production varied by cell type and location in blood vs. gut. Discussion These findings demonstrate the differences in functionality of these cells when compared to their blood counterparts and validate previous studies on phenotype within gut-derived immune cells in humans (where cells have been obtained through surgical means). This study suggests that recto-sigmoid biopsies collected during colonoscopy can be a reliable yet more accessible sampling method for follow up of alterations of gut derived immune cells in clinical settings.
Collapse
Affiliation(s)
| | - Priscila O Barros
- Chronic Diseases Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Tamara Berthoud
- Chronic Diseases Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Siddappa N Byrareddy
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Michaeline McGuinty
- Department of Medicine, Division of Infectious Diseases, The Ottawa Hospital, University of Ottawa, Ottawa, ON, Canada
| | - D William Cameron
- Department of Medicine, Division of Infectious Diseases, The Ottawa Hospital, University of Ottawa, Ottawa, ON, Canada
| | - Jonathan B Angel
- Chronic Diseases Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Medicine, Division of Infectious Diseases, The Ottawa Hospital, University of Ottawa, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
5
|
The SMML, Schreurs RRCE, Drewniak A, Bakx R, de Meij TGJ, Budding AE, Poort L, Cense HA, Heij HA, van Heurn LWE, Gorter RR, Bunders MJ. Enhanced Th17 responses in the appendix of children with complex compared to simple appendicitis are associated with microbial dysbiosis. Front Immunol 2024; 14:1258363. [PMID: 38239362 PMCID: PMC10794624 DOI: 10.3389/fimmu.2023.1258363] [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: 07/13/2023] [Accepted: 11/17/2023] [Indexed: 01/22/2024] Open
Abstract
Introduction Appendicitis is one of the most common causes of acute abdominal surgery in children. The clinical course of appendicitis ranges from simple to complex appendicitis. The mechanisms underlying the heterogeneity of appendicitis in children remain largely unclear. Dysregulated T cell responses play an important role in several inflammatory diseases of the intestine, but the extend of T cell dysregulation in appendicitis in children is less well known. Methods To characterize appendiceal T cells in simple and complex appendicitis we performed in-depth immunophenotyping of appendiceal-derived T cells by flow cytometry and correlated this to appendiceal-derived microbiota analyses of the same patient. Results Appendix samples of twenty children with appendicitis (n = 8 simple, n = 12 complex) were collected. T cells in complex appendicitis displayed an increased differentiated phenotype compared to simple appendicitis, including a loss of both CD27 and CD28 by CD4+ T cells and to a lesser extent by CD8+ T cells. Frequencies of phenotypic tissue-resident memory CD69+CD4+ T cells and CD69+CD8+ T cells were decreased in children with complex compared to simple appendicitis, indicating disruption of local tissue-resident immune responses. In line with the increased differentiated phenotype, cytokine production of in particular IL-17A by CD4+ T cells was increased in children with complex compared to simple appendicitis. Furthermore, frequencies of IL-17A+ CD4+ T cells correlated with a dysregulation of the appendiceal microbiota in children with complex appendicitis. Conclusion In conclusion, disruption of local T cell responses, and enhanced pro-inflammatory Th17 responses correlating to changes in the appendiceal microbiota were observed in children with complex compared to simple appendicitis. Further studies are needed to decipher the role of a dysregulated network of microbiota and Th17 cells in the development of complex appendicitis in children.
Collapse
Affiliation(s)
- Sarah-May M. L. The
- Department of Paediatric Surgery, Emma Children’s Hospital, Amsterdam University Medical Center (UMC), University of Amsterdam & Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam, Netherlands
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Renée R. C. E. Schreurs
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Department of Paediatrics, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam & Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Agata Drewniak
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Roel Bakx
- Department of Paediatric Surgery, Emma Children’s Hospital, Amsterdam University Medical Center (UMC), University of Amsterdam & Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam, Netherlands
- Amsterdam Gastroenterology and Metabolism Research Institute, Amsterdam, Netherlands
| | - Tim G. J. de Meij
- Amsterdam Reproduction and Development Research Institute, Amsterdam, Netherlands
- Amsterdam Gastroenterology and Metabolism Research Institute, Amsterdam, Netherlands
- Department of Paediatric Gastroenterology, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | | | | | - Huib A. Cense
- Department of Surgery, Red Cross Hospital, Beverwijk, Netherlands
| | - Hugo A. Heij
- Department of Paediatric Surgery, Emma Children’s Hospital, Amsterdam University Medical Center (UMC), University of Amsterdam & Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - L. W. Ernest van Heurn
- Department of Paediatric Surgery, Emma Children’s Hospital, Amsterdam University Medical Center (UMC), University of Amsterdam & Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam, Netherlands
- Amsterdam Gastroenterology and Metabolism Research Institute, Amsterdam, Netherlands
| | - Ramon R. Gorter
- Department of Paediatric Surgery, Emma Children’s Hospital, Amsterdam University Medical Center (UMC), University of Amsterdam & Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam, Netherlands
- Amsterdam Gastroenterology and Metabolism Research Institute, Amsterdam, Netherlands
| | - Madeleine J. Bunders
- Leibniz Institute of Virology, Hamburg, Germany
- Third Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
6
|
Baumdick ME, Niehrs A, Degenhardt F, Schwerk M, Hinrichs O, Jordan-Paiz A, Padoan B, Wegner LHM, Schloer S, Zecher BF, Malsy J, Joshi VR, Illig C, Schröder-Schwarz J, Möller KJ, Martin MP, Yuki Y, Ozawa M, Sauter J, Schmidt AH, Perez D, Giannou AD, Carrington M, Davis RS, Schumacher U, Sauter G, Huber S, Puelles VG, Melling N, Franke A, Altfeld M, Bunders MJ. HLA-DP on Epithelial Cells Enables Tissue Damage by NKp44 + Natural Killer Cells in Ulcerative Colitis. Gastroenterology 2023; 165:946-962.e13. [PMID: 37454979 PMCID: PMC10529779 DOI: 10.1053/j.gastro.2023.06.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 05/25/2023] [Accepted: 06/13/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND & AIMS Ulcerative colitis (UC) is characterized by severe inflammation and destruction of the intestinal epithelium, and is associated with specific risk single nucleotide polymorphisms in HLA class II. Given the recently discovered interactions between subsets of HLA-DP molecules and the activating natural killer (NK) cell receptor NKp44, genetic associations of UC and HLA-DP haplotypes and their functional implications were investigated. METHODS HLA-DP haplotype and UC risk association analyses were performed (UC: n = 13,927; control: n = 26,764). Expression levels of HLA-DP on intestinal epithelial cells (IECs) in individuals with and without UC were quantified. Human intestinal 3-dimensional (3D) organoid cocultures with human NK cells were used to determine functional consequences of interactions between HLA-DP and NKp44. RESULTS These studies identified HLA-DPA1∗01:03-DPB1∗04:01 (HLA-DP401) as a risk haplotype and HLA-DPA1∗01:03-DPB1∗03:01 (HLA-DP301) as a protective haplotype for UC in European populations. HLA-DP expression was significantly higher on IECs of individuals with UC compared with controls. IECs in human intestinal 3D organoids derived from HLA-DP401pos individuals showed significantly stronger binding of NKp44 compared with HLA-DP301pos IECs. HLA-DP401pos IECs in organoids triggered increased degranulation and tumor necrosis factor production by NKp44+ NK cells in cocultures, resulting in enhanced epithelial cell death compared with HLA-DP301pos organoids. Blocking of HLA-DP401-NKp44 interactions (anti-NKp44) abrogated NK cell activity in cocultures. CONCLUSIONS We identified an UC risk HLA-DP haplotype that engages NKp44 and activates NKp44+ NK cells, mediating damage to intestinal epithelial cells in an HLA-DP haplotype-dependent manner. The molecular interaction between NKp44 and HLA-DP401 in UC can be targeted by therapeutic interventions to reduce NKp44+ NK cell-mediated destruction of the intestinal epithelium in UC.
Collapse
Affiliation(s)
- Martin E Baumdick
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, Germany
| | - Annika Niehrs
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, Germany
| | - Frauke Degenhardt
- Institute of Clinical Molecular Biology, Christian-Albrechts-University, Kiel, Germany
| | - Maria Schwerk
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Hamburg Center for Kidney Health, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ole Hinrichs
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, Germany
| | - Ana Jordan-Paiz
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, Germany
| | - Benedetta Padoan
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, Germany
| | - Lucy H M Wegner
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, Germany
| | - Sebastian Schloer
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, Germany; Research Group Regulatory Mechanisms of Inflammation, Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, and Cells in Motion Interfaculty Center, University of Münster, Münster, Germany
| | - Britta F Zecher
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, Germany; I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jakob Malsy
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, Germany; I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; German Center for Infection Research, Hamburg-Lübeck-Borstel-Riems, Germany
| | - Vinita R Joshi
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, Germany
| | - Christin Illig
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, Germany
| | - Jennifer Schröder-Schwarz
- Institute of Anatomy and Experimental Morphology, Center for Experimental Medicine, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kimberly J Möller
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, Germany
| | - Maureen P Martin
- Basic Science Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, Maryland; Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Yuko Yuki
- Basic Science Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, Maryland; Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | | | | | | | - Daniel Perez
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anastasios D Giannou
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Section of Molecular Immunology and Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mary Carrington
- Basic Science Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, Maryland; Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland; Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts
| | - Randall S Davis
- Departments of Medicine, Microbiology, and Biochemistry and Molecular Genetics, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Udo Schumacher
- Institute of Anatomy and Experimental Morphology, Center for Experimental Medicine, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Samuel Huber
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Section of Molecular Immunology and Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Victor G Puelles
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Hamburg Center for Kidney Health, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Nathaniel Melling
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University, Kiel, Germany
| | - Marcus Altfeld
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, Germany
| | - Madeleine J Bunders
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, Germany; III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Section of Regenerative Medicine and Immunology, III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
7
|
Möller KJ, Wegner LHM, Malsy J, Baumdick ME, Borggrewe M, Jordan-Paiz A, Jung JM, Martrus G, Kretschmer P, Sagebiel AF, Schreurs RRCE, Hagen SH, Burmester G, Clauditz TS, Pals ST, Boettcher M, Melling N, Sauter G, Tomuschat C, Königs I, Schumacher U, Altfeld M, Bernink JH, Perez D, Reinshagen K, Bunders MJ. Expanded ILC2s in human infant intestines promote tissue growth. Mucosal Immunol 2023; 16:408-421. [PMID: 37121384 DOI: 10.1016/j.mucimm.2023.04.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 04/14/2023] [Indexed: 05/02/2023]
Abstract
Early life is characterized by extraordinary challenges, including rapid tissue growth and immune adaptation to foreign antigens after birth. During this developmental stage, infants have an increased risk of immune-mediated diseases. Here, we demonstrate that tissue-resident, interleukin (IL)-13- and IL-4-producing group 2 innate lymphoid cells (ILC2s) are enriched in human infant intestines compared to adult intestines. Organoid systems were employed to assess the role of infant intestinal ILC2s in intestinal development and showed that IL-13 and IL-4 increased epithelial cell proliferation and skewed cell differentiation toward secretory cells. IL-13 furthermore upregulated the production of mediators of type-2 immunity by infant intestinal epithelial cells, including vascular endothelial growth factor-A and IL-26, a chemoattractant for eosinophils. In line with these in vitro findings increased numbers of eosinophils were detected in vivo in infant intestines. Taken together, ILC2s are enriched in infant intestines and can support intestinal development while inducing an epithelial secretory response associated with type 2 immune-mediated diseases.
Collapse
Affiliation(s)
- Kimberly J Möller
- Research Department of Virus Immunology, Leibniz Institute of Virology (LIV), Hamburg, Germany
| | - Lucy H M Wegner
- Research Department of Virus Immunology, Leibniz Institute of Virology (LIV), Hamburg, Germany
| | - Jakob Malsy
- Research Department of Virus Immunology, Leibniz Institute of Virology (LIV), Hamburg, Germany; I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; German Center for Infection Research (DZIF), Hamburg-Lübeck-Borstel-Riems, Germany
| | - Martin E Baumdick
- Research Department of Virus Immunology, Leibniz Institute of Virology (LIV), Hamburg, Germany
| | - Malte Borggrewe
- Research Department of Virus Immunology, Leibniz Institute of Virology (LIV), Hamburg, Germany; III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ana Jordan-Paiz
- Research Department of Virus Immunology, Leibniz Institute of Virology (LIV), Hamburg, Germany
| | - Johannes M Jung
- Research Department of Virus Immunology, Leibniz Institute of Virology (LIV), Hamburg, Germany
| | - Glòria Martrus
- Research Department of Virus Immunology, Leibniz Institute of Virology (LIV), Hamburg, Germany
| | - Paul Kretschmer
- Research Department of Virus Immunology, Leibniz Institute of Virology (LIV), Hamburg, Germany
| | - Adrian F Sagebiel
- Research Department of Virus Immunology, Leibniz Institute of Virology (LIV), Hamburg, Germany; Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Renée R C E Schreurs
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, Amsterdam, the Netherlands; Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Sven H Hagen
- Research Department of Virus Immunology, Leibniz Institute of Virology (LIV), Hamburg, Germany
| | - Gunter Burmester
- Department of Pediatric Gastroenterology, Altonaer Children's Hospital, Hamburg, Germany
| | - Till S Clauditz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Steven T Pals
- Department of Pathology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Michael Boettcher
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf/Altonaer Children's Hospital, Hamburg, Germany
| | - Nathaniel Melling
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Tomuschat
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf/Altonaer Children's Hospital, Hamburg, Germany
| | - Ingo Königs
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf/Altonaer Children's Hospital, Hamburg, Germany
| | - Udo Schumacher
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marcus Altfeld
- Research Department of Virus Immunology, Leibniz Institute of Virology (LIV), Hamburg, Germany
| | - Jochem H Bernink
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), Amsterdam, The Netherlands
| | - Daniel Perez
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Asklepios General Hospital-Altona, Hamburg, Germany
| | - Konard Reinshagen
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf/Altonaer Children's Hospital, Hamburg, Germany
| | - Madeleine J Bunders
- Research Department of Virus Immunology, Leibniz Institute of Virology (LIV), Hamburg, Germany; III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
8
|
Jordan-Paiz A, Martrus G, Steinert FL, Kaufmann M, Sagebiel AF, Schreurs RRCE, Rechtien A, Baumdick ME, Jung JM, Möller KJ, Wegner L, Grüttner C, Richert L, Thünauer R, Schroeder-Schwarz J, van Goudoever JB, Geijtenbeek TBH, Altfeld M, Pals ST, Perez D, Klarenbeek PL, Tomuschat C, Sauter G, Königs I, Schumacher U, Friese MA, Melling N, Reinshagen K, Bunders MJ. CXCR5 +PD-1 ++ CD4 + T cells colonize infant intestines early in life and promote B cell maturation. Cell Mol Immunol 2023; 20:201-213. [PMID: 36600048 PMCID: PMC9886971 DOI: 10.1038/s41423-022-00944-4] [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: 01/18/2022] [Accepted: 10/26/2022] [Indexed: 01/06/2023] Open
Abstract
Gastrointestinal infections are a major cause for serious clinical complications in infants. The induction of antibody responses by B cells is critical for protective immunity against infections and requires CXCR5+PD-1++ CD4+ T cells (TFH cells). We investigated the ontogeny of CXCR5+PD-1++ CD4+ T cells in human intestines. While CXCR5+PD-1++ CD4+ T cells were absent in fetal intestines, CXCR5+PD-1++ CD4+ T cells increased after birth and were abundant in infant intestines, resulting in significant higher numbers compared to adults. These findings were supported by scRNAseq analyses, showing increased frequencies of CD4+ T cells with a TFH gene signature in infant intestines compared to blood. Co-cultures of autologous infant intestinal CXCR5+PD-1+/-CD4+ T cells with B cells further demonstrated that infant intestinal TFH cells were able to effectively promote class switching and antibody production by B cells. Taken together, we demonstrate that functional TFH cells are numerous in infant intestines, making them a promising target for oral pediatric vaccine strategies.
Collapse
Affiliation(s)
- Ana Jordan-Paiz
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, 20251, Germany
| | - Glòria Martrus
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, 20251, Germany
| | - Fenja L Steinert
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, 20251, Germany
- University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Max Kaufmann
- Institute of Neuroimmunology and Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, 20251, Germany
| | - Adrian F Sagebiel
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, 20251, Germany
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Renée R C E Schreurs
- Department of Experimental Immunology; Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, 1105 AZ, The Netherlands
- Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Center, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, 1105 AZ, The Netherlands
| | - Anne Rechtien
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, 20251, Germany
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
- Partner Site Hamburg-Lübeck-Borstel-Riems, German Center for Infection Research (DZIF), Hamburg, 20246, Germany
| | - Martin E Baumdick
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, 20251, Germany
| | - Johannes M Jung
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, 20251, Germany
| | - Kimberly J Möller
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, 20251, Germany
- University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Lucy Wegner
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, 20251, Germany
- University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Cordula Grüttner
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, 20251, Germany
| | - Laura Richert
- University of Bordeaux, Institut National de la Santé et de la Recherche Médicale, Bordeaux Population Health Research Center UMR1219 and INRIA SISTM Team, Bordeaux, 33000, France
| | - Roland Thünauer
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, 20251, Germany
| | - Jennifer Schroeder-Schwarz
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Johannes B van Goudoever
- Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Center, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, 1105 AZ, The Netherlands
| | - Teunis B H Geijtenbeek
- Department of Experimental Immunology; Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, 1105 AZ, The Netherlands
| | - Marcus Altfeld
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, 20251, Germany
| | - Steven T Pals
- Department of Pathology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, 1105 AZ, The Netherlands
| | - Daniel Perez
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Paul L Klarenbeek
- Department of Rheumatology and Clinical Immunology and Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, 1007 MB, The Netherlands
- Amsterdam Rheumatology & Immunology Center, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, 1105 AZ, The Netherlands
| | - Christian Tomuschat
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Ingo Königs
- Department of Pediatric Surgery, Altona Children's Hospital, Hamburg, 22763, Germany
| | - Udo Schumacher
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Manuel A Friese
- Institute of Neuroimmunology and Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, 20251, Germany
| | - Nathaniel Melling
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Konrad Reinshagen
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Madeleine J Bunders
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, 20251, Germany.
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany.
| |
Collapse
|
9
|
Uniken Venema WTC, Ramírez-Sánchez AD, Bigaeva E, Withoff S, Jonkers I, McIntyre RE, Ghouraba M, Raine T, Weersma RK, Franke L, Festen EAM, van der Wijst MGP. Gut mucosa dissociation protocols influence cell type proportions and single-cell gene expression levels. Sci Rep 2022; 12:9897. [PMID: 35701452 PMCID: PMC9197976 DOI: 10.1038/s41598-022-13812-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 05/27/2022] [Indexed: 01/15/2023] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) has revolutionized the study of the cellular landscape of organs. Most single-cell protocols require fresh material, which limits sample size per experiment, and consequently, introduces batch effects. This is especially true for samples acquired through complex medical procedures, such as intestinal mucosal biopsies. Moreover, the tissue dissociation procedure required for obtaining single cells is a major source of noise; different dissociation procedures applied to different compartments of the tissue induce artificial gene expression differences between cell subsets. To overcome these challenges, we have developed a one-step dissociation protocol and demonstrated its use on cryopreserved gut mucosal biopsies. Using flow cytometry and scRNA-seq analysis, we compared this one-step dissociation protocol with the current gold standard, two-step collagenase digestion, and an adaptation of a recently published alternative, three-step cold-active Bacillus licheniformus protease digestion. Both cell viability and cell type composition were comparable between the one-step and two-step collagenase dissociation, with the former being more time-efficient. The cold protease digestion resulted in equal cell viability, but better preserves the epithelial cell types. Consequently, to analyze the rarer cell types, such as glial cells, larger total biopsy cell numbers are required as input material. The multi-step protocols affected cell types spanning multiple compartments differently. In summary, we show that cryopreserved gut mucosal biopsies can be used to overcome the logistical challenges and batch effects in large scRNA-seq studies. Furthermore, we demonstrate that using cryopreserved biopsies digested using a one-step collagenase protocol enables large-scale scRNA-seq, FACS, organoid generation and intraepithelial lymphocyte expansion.
Collapse
Affiliation(s)
- Werna T C Uniken Venema
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Aarón D Ramírez-Sánchez
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Emilia Bigaeva
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Sebo Withoff
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Iris Jonkers
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | | | | | - Tim Raine
- Department of Gastroenterology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Rinse K Weersma
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Lude Franke
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Eleonora A M Festen
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Monique G P van der Wijst
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
10
|
Marsh SE, Walker AJ, Kamath T, Dissing-Olesen L, Hammond TR, de Soysa TY, Young AMH, Murphy S, Abdulraouf A, Nadaf N, Dufort C, Walker AC, Lucca LE, Kozareva V, Vanderburg C, Hong S, Bulstrode H, Hutchinson PJ, Gaffney DJ, Hafler DA, Franklin RJM, Macosko EZ, Stevens B. Dissection of artifactual and confounding glial signatures by single-cell sequencing of mouse and human brain. Nat Neurosci 2022; 25:306-316. [PMID: 35260865 DOI: 10.1038/s41593-022-01022-8] [Citation(s) in RCA: 166] [Impact Index Per Article: 83.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 01/25/2022] [Indexed: 12/31/2022]
Abstract
A key aspect of nearly all single-cell sequencing experiments is dissociation of intact tissues into single-cell suspensions. While many protocols have been optimized for optimal cell yield, they have often overlooked the effects that dissociation can have on ex vivo gene expression. Here, we demonstrate that use of enzymatic dissociation on brain tissue induces an aberrant ex vivo gene expression signature, most prominently in microglia, which is prevalent in published literature and can substantially confound downstream analyses. To address this issue, we present a rigorously validated protocol that preserves both in vivo transcriptional profiles and cell-type diversity and yield across tissue types and species. We also identify a similar signature in postmortem human brain single-nucleus RNA-sequencing datasets, and show that this signature is induced in freshly isolated human tissue by exposure to elevated temperatures ex vivo. Together, our results provide a methodological solution for preventing artifactual gene expression changes during fresh tissue digestion and a reference for future deeper analysis of the potential confounding states present in postmortem human samples.
Collapse
Affiliation(s)
- Samuel E Marsh
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Alec J Walker
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Tushar Kamath
- Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Lasse Dissing-Olesen
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Timothy R Hammond
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - T Yvanka de Soysa
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Adam M H Young
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Sarah Murphy
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
| | - Abdulraouf Abdulraouf
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Naeem Nadaf
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Connor Dufort
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
| | - Alicia C Walker
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
| | - Liliana E Lucca
- Department of Neurology and Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Velina Kozareva
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Charles Vanderburg
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Soyon Hong
- UK Dementia Research Institute, University College London, London, UK
| | - Harry Bulstrode
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Peter J Hutchinson
- Department of Clinical Neurosciences, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | | | - David A Hafler
- Department of Neurology and Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Robin J M Franklin
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Evan Z Macosko
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Beth Stevens
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
11
|
Saidj T, Baba Amer Y, Plonquet A, Henry A, Souvannanorath S, Relaix F, Beldi-Ferchiou A, Authier FJ. Optimized Flow Cytometry Strategy for Phenotyping Intramuscular Leukocytes: Application to the Evaluation of Myopathological Processes. J Neuropathol Exp Neurol 2022; 81:193-207. [DOI: 10.1093/jnen/nlab136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Abstract
Phenotyping intramuscular immune cells is essential for the characterization of dysimmune/inflammatory myopathies (DIM). Flow cytometry (FC) is the most reliable technique for analyzing leukocyte subpopulations and evaluating their activation levels. We developed a purely mechanical protocol for extracting cells from muscle tissue allowing us to preserve cell surface epitopes and determined its applicability to experimental pathology in mice and myopathological diagnosis in human. Skeletal muscle regeneration in mice was associated with a transient enrichment of macrophages (CD11bhighGr-1+), myeloid dendritic cells (CD3−C8+CD11bhigh), CD8+ T cells (CD3+C8+), and NK cells (CD3− CD11bhighNKp46+). In murine models of inherited muscle dystrophies, leukocytes represented 23%–84% of intramuscular mononuclear cells, with a percentage of CD8+ T cells (4%–17%) mirroring that of all CD45+ cells, while MDCs remained a minority. In human 16 samples (DIM: n = 9; nonimmune conditions: n = 7), DIM was associated with intramuscular recruitment of CD8+ T cells, but not CD4+ T cells and NK cells. FC allowed concomitant quantification of HLA-DR, CD25, CD38, and CD57 activation/differentiation biomarkers and showed increased activation levels of CD4+ and CD8+ T cells in DIM. In conclusion, FC is an appropriate method for quantifying intramuscular leukocyte subpopulations and analyzing their activation states.
Collapse
Affiliation(s)
- Tassadit Saidj
- Université Paris Est Créteil, INSERM, IMRB, Equipe Relaix, Creteil, France
| | - Yasmine Baba Amer
- Université Paris Est Créteil, INSERM, IMRB, Equipe Relaix, Creteil, France
| | - Anne Plonquet
- AP-HP, Hôpitaux Universitaires Henri Mondor, Laboratoire d'immunologie Biologique, Créteil, France
| | - Adeline Henry
- Université Paris Est Créteil, INSERM, IMRB, Plateforme de Cytométrie en flux, Créteil, France
| | - Sarah Souvannanorath
- Département de Pathologie, APHP, Hôpitaux Universitaires Henri Mondor, Centre de Référence des Maladies Rares Neuromusculaire Nord/Est/Ile-de-France, ERN Euro-NMD, Créteil, France
- Université Paris Est Créteil, INSERM, IMRB, Equipe Relaix, Creteil, France
| | - Frederic Relaix
- Université Paris Est Créteil, INSERM, IMRB, Equipe Relaix, Creteil, France
| | - Asma Beldi-Ferchiou
- AP-HP, Hôpitaux Universitaires Henri Mondor, Laboratoire d'immunologie Biologique, Créteil, France
- Université Paris Est Créteil, INSERM, IMRB, Equipe Cohen, Créteil, France
| | - François Jérôme Authier
- Université Paris Est Créteil, INSERM, IMRB, Equipe Relaix, Creteil, France
- Département de Pathologie, APHP, Hôpitaux Universitaires Henri Mondor, Centre de Référence des Maladies Rares Neuromusculaire Nord/Est/Ile-de-France, ERN Euro-NMD, Créteil, France
- Université Paris Est Créteil, INSERM, IMRB, Equipe Relaix, Creteil, France
| |
Collapse
|
12
|
Schreurs RRCE, Baumdick ME, Drewniak A, Bunders MJ. In vitro co-culture of human intestinal organoids and lamina propria-derived CD4 + T cells. STAR Protoc 2021; 2:100519. [PMID: 34036282 PMCID: PMC8138864 DOI: 10.1016/j.xpro.2021.100519] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Crosstalk between immune cells and intestinal stem cells (ISCs) in vivo plays a critical role in tissue homeostasis and inflammation; however, in vitro models based on primary cells recapitulating this interaction were lacking. Here, we provide a detailed protocol for an autologous in vitro long-term 3D co-culture system of human intestinal CD4+ T cells and ISCs to study T cell-intestinal epithelial cell interactions during tissue development and inflammation. For complete details on the use and execution of this protocol, please refer to Schreurs et al. (2019). Reproducible generation and culture of intestinal organoids with intestinal CD4+ T cells Isolation of intestinal crypts and lamina propria-derived lymphocytes In vitro system to model immune-mediated regulation of intestinal growth and inflammation
Collapse
Affiliation(s)
- Renée R C E Schreurs
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Martin E Baumdick
- Research Department Virus Immunology, Leibniz Institute for Experimental Virology, Hamburg 20251, Germany
| | - Agata Drewniak
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands.,Kiadis Pharma B.V., Amsterdam 1105 BV, the Netherlands
| | - Madeleine J Bunders
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands.,Research Department Virus Immunology, Leibniz Institute for Experimental Virology, Hamburg 20251, Germany
| |
Collapse
|
13
|
Jørgensen PB, Fenton TM, Mörbe UM, Riis LB, Jakobsen HL, Nielsen OH, Agace WW. Identification, isolation and analysis of human gut-associated lymphoid tissues. Nat Protoc 2021; 16:2051-2067. [PMID: 33619391 DOI: 10.1038/s41596-020-00482-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 12/09/2020] [Indexed: 02/06/2023]
Abstract
Gut-associated lymphoid tissues (GALTs) comprise key intestinal immune inductive sites, including the Peyer's patches of the small intestine and different types of isolated lymphoid follicle (ILF) found along the length of the gut. Our understanding of human GALT is limited due to a lack of protocols for their isolation. Here we describe a technique that, uniquely among intestinal cell isolation protocols, allows identification and isolation of all human GALT, as well as GALT-free intestinal lamina propria (LP). The technique involves the mechanical separation of intestinal mucosa from the submucosa, allowing the identification and isolation of submucosal ILF (SM-ILF), LP-embedded mucosal ILF (M-ILF) and LP free of contaminating lymphoid tissue. Individual SM-ILF, M-ILF and Peyer's patch follicles can be subsequently digested for downstream cellular and molecular characterization. The technique, which takes 4-10 h, will be useful for researchers interested in intestinal immune development and function in health and disease.
Collapse
Affiliation(s)
- Peter B Jørgensen
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Thomas M Fenton
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Urs M Mörbe
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Lene B Riis
- Department of Pathology, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| | - Henrik L Jakobsen
- Department of Gastroenterology, Surgical Section, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| | - Ole H Nielsen
- Department of Gastroenterology, Medical Section, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| | - William W Agace
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark. .,Immunology Section, Lund University, Lund, Sweden.
| |
Collapse
|
14
|
Schreurs RRCE, Sagebiel AF, Steinert FL, Highton AJ, Klarenbeek PL, Drewniak A, Bakx R, The SML, Ribeiro CMS, Perez D, Reinshagen K, Geijtenbeek TBH, van Goudoever JB, Bunders MJ. Intestinal CD8 + T cell responses are abundantly induced early in human development but show impaired cytotoxic effector capacities. Mucosal Immunol 2021; 14:605-614. [PMID: 33772147 PMCID: PMC8075922 DOI: 10.1038/s41385-021-00382-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 09/30/2020] [Accepted: 11/02/2020] [Indexed: 02/04/2023]
Abstract
Gastrointestinal viral infections are a major global cause of disease and mortality in infants. Cytotoxic CD8+ T cells are critical to achieve viral control. However, studies investigating the development of CD8+ T cell immunity in human tissues early in life are lacking. Here, we investigated the maturation of the CD8+ T cell compartment in human fetal, infant and adult intestinal tissues. CD8+ T cells exhibiting a memory phenotype were already detected in fetal intestines and increased after birth. Infant intestines preferentially harbored effector CCR7-CD45RA-CD127-KLRG1+/- CD8+ T cells compared to tissue-resident memory CD69+CD103+CD8+ T cells detected in adults. Functional cytotoxic capacity, including cytokine and granzyme B production of infant intestinal effector CD8+ T cells was, however, markedly reduced compared to adult intestinal CD8+ T cells. This was in line with the high expression of the inhibitory molecule PD-1 by infant intestinal effector CD8+ T cells. Taken together, we demonstrate that intestinal CD8+ T cell responses are induced early in human development, however exhibit a reduced functionality. The impaired CD8+ T cell functionality early in life contributes to tolerance during foreign antigen exposure after birth, however functions as an immune correlate for the increased susceptibility to gastrointestinal viral infections in infancy.
Collapse
Affiliation(s)
- R. R. C. E. Schreurs
- grid.7177.60000000084992262Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center (AUMC), University of Amsterdam (UvA), Amsterdam, The Netherlands ,grid.7177.60000000084992262Department of Pediatrics, Emma Children’s Hospital, AUMC, UvA, Amsterdam, The Netherlands
| | - A. F. Sagebiel
- grid.418481.00000 0001 0665 103XHeinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - F. L. Steinert
- grid.418481.00000 0001 0665 103XHeinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - A. J. Highton
- grid.418481.00000 0001 0665 103XHeinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - P. L. Klarenbeek
- grid.7177.60000000084992262Department of Clinical Immunology and Rheumatology and Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, AUMC, UvA, Amsterdam, The Netherlands ,grid.16872.3a0000 0004 0435 165XAmsterdam Rheumatology & Immunology Center, AUMC, UvA, Amsterdam, The Netherlands
| | - A. Drewniak
- grid.7177.60000000084992262Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center (AUMC), University of Amsterdam (UvA), Amsterdam, The Netherlands ,grid.467476.00000 0004 0483 1848Kiadis Pharma B.V., Amsterdam, The Netherlands
| | - R. Bakx
- Department of Pediatric Surgery, Pediatric Surgery Center of Amsterdam, AUMC, Amsterdam, The Netherlands
| | - S. M. L. The
- grid.7177.60000000084992262Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center (AUMC), University of Amsterdam (UvA), Amsterdam, The Netherlands ,Department of Pediatric Surgery, Pediatric Surgery Center of Amsterdam, AUMC, Amsterdam, The Netherlands
| | - C. M. S. Ribeiro
- grid.7177.60000000084992262Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center (AUMC), University of Amsterdam (UvA), Amsterdam, The Netherlands
| | - D. Perez
- grid.13648.380000 0001 2180 3484Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - K. Reinshagen
- grid.13648.380000 0001 2180 3484Department of Pediatric Surgery, UKE, Hamburg, Germany
| | - T. B. H. Geijtenbeek
- grid.7177.60000000084992262Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center (AUMC), University of Amsterdam (UvA), Amsterdam, The Netherlands
| | - J. B. van Goudoever
- grid.7177.60000000084992262Department of Pediatrics, Emma Children’s Hospital, AUMC, UvA, Amsterdam, The Netherlands ,grid.12380.380000 0004 1754 9227Department of Pediatrics, Emma Children’s Hospital, AUMC, Vrije Universiteit, Amsterdam, The Netherlands
| | - M. J. Bunders
- grid.7177.60000000084992262Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center (AUMC), University of Amsterdam (UvA), Amsterdam, The Netherlands ,grid.7177.60000000084992262Department of Pediatrics, Emma Children’s Hospital, AUMC, UvA, Amsterdam, The Netherlands ,grid.418481.00000 0001 0665 103XHeinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| |
Collapse
|
15
|
García-Rodríguez I, Sridhar A, Pajkrt D, Wolthers KC. Put Some Guts into It: Intestinal Organoid Models to Study Viral Infection. Viruses 2020; 12:v12111288. [PMID: 33187072 PMCID: PMC7697248 DOI: 10.3390/v12111288] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/30/2020] [Accepted: 11/09/2020] [Indexed: 02/06/2023] Open
Abstract
The knowledge about enteric viral infection has vastly increased over the last eight years due to the development of intestinal organoids and enteroids that suppose a step forward from conventional studies using cell lines. Intestinal organoids and enteroids are three-dimensional (3D) models that closely mimic intestinal cellular heterogeneity and organization. The barrier function within these models has been adapted to facilitate viral studies. In this review, several adaptations (such as organoid-derived two-dimensional (2D) monolayers) and original intestinal 3D models are discussed. The specific advantages and applications, as well as improvements of each model are analyzed and an insight into the possible path for the field is given.
Collapse
Affiliation(s)
- Inés García-Rodríguez
- OrganoVIR Lab, Department of Medical Microbiology, Amsterdam UMC, Academic Medical Center, University of Amsterdam, 1100 AZ Amsterdam, The Netherlands; (I.G.-R.); (A.S.)
- Department of Pediatrics Infectious Diseases, Emma Children’s Hospital, Amsterdam UMC, Academic Medical Center, University of Amsterdam, 1100 AZ Amsterdam, The Netherlands;
| | - Adithya Sridhar
- OrganoVIR Lab, Department of Medical Microbiology, Amsterdam UMC, Academic Medical Center, University of Amsterdam, 1100 AZ Amsterdam, The Netherlands; (I.G.-R.); (A.S.)
- Department of Pediatrics Infectious Diseases, Emma Children’s Hospital, Amsterdam UMC, Academic Medical Center, University of Amsterdam, 1100 AZ Amsterdam, The Netherlands;
| | - Dasja Pajkrt
- Department of Pediatrics Infectious Diseases, Emma Children’s Hospital, Amsterdam UMC, Academic Medical Center, University of Amsterdam, 1100 AZ Amsterdam, The Netherlands;
| | - Katja C. Wolthers
- OrganoVIR Lab, Department of Medical Microbiology, Amsterdam UMC, Academic Medical Center, University of Amsterdam, 1100 AZ Amsterdam, The Netherlands; (I.G.-R.); (A.S.)
- Correspondence:
| |
Collapse
|
16
|
Drug efflux transporters and metabolic enzymes in human circulating and testicular T-cell subsets: relevance to HIV pharmacotherapy. AIDS 2020; 34:1439-1449. [PMID: 32310902 DOI: 10.1097/qad.0000000000002548] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES ATP-binding cassette (ABC) drug efflux transporters and drug metabolic enzymes could reduce antiretroviral concentrations in HIV target cells. The testis has been demonstrated to be a sanctuary site, displaying suboptimal antiretroviral concentrations and persistent HIV infection. Therefore, we compared the expression and function of ABC transporters and metabolic enzymes in CD4 and CD8 T cells isolated from human testis and peripheral blood mononuclear cells (PBMCs), and assessed their expression in circulating naive and memory CD4 T-cell phenotypes. DESIGN Testicular tissue and blood were collected from 15 uninfected donors undergoing gender affirmation surgery. Testicular interstitial cells were isolated by enzymatic digestion, whereas PBMCs were isolated from blood by density gradient centrifugation. The expression and/or function of ABC transporters and metabolic enzymes were examined in blood and testicular T-cell subsets by flow cytometry. RESULTS ABC transporters (P-gp, BCRP, MRP1) and metabolic enzymes (CYP3A4, UGT1A1) were expressed in testicular and circulating CD4 and CD8 T cells, as well as in circulating naive, central, transitional, and effector memory T-cell phenotypes. MRP1 demonstrated lower frequencies in T cells from testis compared with PBMCs, as well as in circulating naive T cells compared with the memory T-cell phenotypes. Functional activity of P-gp and BCRP was detected in T-cell subsets from testis and PBMCs. CONCLUSION Our findings demonstrate for the first time that antiretroviral drug efflux transporters and metabolic enzymes are functionally expressed in T-cell subsets infiltrating the human testis. These transporters and enzymes can reduce antiretroviral intracellular concentrations, potentially contributing to residual HIV replication in the testis, and negatively impact HIV cure strategies.
Collapse
|
17
|
Gómez-Massa E, Lasa-Lázaro M, Gil-Etayo FJ, Ulloa-Márquez E, Justo I, Loinaz C, Calvo-Pulido J, Paz-Artal E, Talayero P. Donor helper innate lymphoid cells are replaced earlier than lineage positive cells and persist long-term in human intestinal grafts - a descriptive study. Transpl Int 2020; 33:1016-1029. [PMID: 32246810 DOI: 10.1111/tri.13609] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/04/2020] [Accepted: 03/25/2020] [Indexed: 12/11/2022]
Abstract
Intestinal grafts carry large donor lymphoid load that is replaced by recipient cells. The dynamics of this process may influence the tolerance, rejection or graft-versus-host disease. We analysed distribution and turnover of T and B (Lin+) lymphocytes, natural killer (NK) and helper innate lymphoid cells (hILC) in intestinal epithelium (IEp) and lamina propia (LP) from a long-term cohort of eight intestinal recipients and from a single patient monitored deeply during the first 8 months post-transplant (posTx). Long-term intestinal grafts showed significantly higher %hILC than native bowels in IEp and LP until 10 years posTx and recovery to normal levels was observed afterwards. We also observed an imbalance between hILC subsets in IEp [increase of type 1 (ILC1) and decrease in type 3 (ILC3) innate lymphoid cells] that persisted along posTx time even when %hILC was similar to native bowels. Regarding hILC origin, we still detected the presence of donor cells at 13 years posTx. However, this chimerism was significantly lower than in Lin+ and NK populations. According to these findings, observation from the patient monitored in early posTx period showed that recipient hILC repopulate earlier and faster than Lin+ cells, with increase in ILC1 related to rejection and infection episodes.
Collapse
Affiliation(s)
- Elena Gómez-Massa
- Department of Immunology, University Hospital 12 de Octubre, Madrid, Spain.,Imas12 Research Institute, University Hospital 12 de Octubre, Madrid, Spain
| | - María Lasa-Lázaro
- Department of Immunology, University Hospital 12 de Octubre, Madrid, Spain.,Imas12 Research Institute, University Hospital 12 de Octubre, Madrid, Spain
| | | | | | - Iago Justo
- HPB Surgery and Abdominal Transplantation Unit, General Surgery Service, University Hospital 12 de Octubre, Madrid, Spain
| | - Carmelo Loinaz
- HPB Surgery and Abdominal Transplantation Unit, General Surgery Service, University Hospital 12 de Octubre, Madrid, Spain
| | - Jorge Calvo-Pulido
- HPB Surgery and Abdominal Transplantation Unit, General Surgery Service, University Hospital 12 de Octubre, Madrid, Spain
| | - Estela Paz-Artal
- Department of Immunology, University Hospital 12 de Octubre, Madrid, Spain.,Imas12 Research Institute, University Hospital 12 de Octubre, Madrid, Spain.,School of Medicine, Complutense University, Madrid, Spain.,Section of Immunology, San Pablo CEU University, Madrid, Spain
| | - Paloma Talayero
- Department of Immunology, University Hospital 12 de Octubre, Madrid, Spain.,Imas12 Research Institute, University Hospital 12 de Octubre, Madrid, Spain
| |
Collapse
|
18
|
Stoner KA, Beamer MA, Avolia HA, Meyn LA, Hillier SL, Achilles SL. Optimization of processing female genital tissue samples for lymphocyte analysis by flow cytometry. Am J Reprod Immunol 2020; 83:e13227. [PMID: 31991032 DOI: 10.1111/aji.13227] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 01/10/2020] [Accepted: 01/21/2020] [Indexed: 11/27/2022] Open
Abstract
PROBLEM A variety of methods have been used to process cervical cytobrush and genital tissue for flow cytometric evaluation of immune cell populations. We sought to optimize genital tract specimen processing and to determine if blood could be used as a model for assessment of tissue processing methods. METHOD OF STUDY Cervical cytobrushes, PBMCs, and genital tissue samples (cervical and endometrial biopsies) were subjected to varying processing conditions to characterize the effects on cell yields, lymphocyte viability, and surface receptors. We exposed PBMC and tissue specimens to varied collagenase types, concentrations, and exposure durations and cytobrushes to immediate vs delayed processing with/without vortexing. RESULTS PBMCs and tissues exposed to varying enzymatic digestion conditions demonstrated stability of some cell surface receptors, including CD3+ , CD4+ , and CD8+ , while others, including CCR6+ , were cleaved when exposed to any concentration of collagenase B, or ≥0.25 mg/mL of collagenase D. We observed increased CD69 expression (marker of cell activation) after exposure to collagenase B. Neither a 2-hour delay in cytobrush processing nor vortexing at a setting of 50% for 30 seconds had significant impacts on viability or quantities of genital immune cells of interest. CONCLUSION Although tissue digestion with collagenase D was sufficient to recover and analyze cells from endometrial biopsy specimens, cervical biopsy specimens required a limited exposure to collagenase B at 1 mg/mL to optimize cell yield and viability for cytometric analysis. PBMCs can be used as a model to assess the impact of tissue processing on co-receptor expression and to optimize methods prior to study implementation.
Collapse
Affiliation(s)
- Kevin A Stoner
- Magee-Womens Research and Foundation, Pittsburgh, PA, USA
| | - May A Beamer
- Magee-Womens Research and Foundation, Pittsburgh, PA, USA
| | | | - Leslie A Meyn
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sharon L Hillier
- Magee-Womens Research and Foundation, Pittsburgh, PA, USA.,Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sharon L Achilles
- Magee-Womens Research and Foundation, Pittsburgh, PA, USA.,Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
19
|
Nijmeijer BM, Sarrami‐Forooshani R, Steba GS, Schreurs RRCE, Koekkoek SM, Molenkamp R, Schinkel J, Reiss P, Siegenbeek van Heukelom ML, van der Valk M, Ribeiro CMS, Geijtenbeek TBH. HIV-1 exposure and immune activation enhance sexual transmission of Hepatitis C virus by primary Langerhans cells. J Int AIDS Soc 2019; 22:e25268. [PMID: 30932366 PMCID: PMC6442005 DOI: 10.1002/jia2.25268] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 03/05/2019] [Indexed: 01/31/2023] Open
Abstract
INTRODUCTION The significant rise in incidence of Hepatitis C virus (HCV) infection among men-who-have-sex-with-men (MSM) living with HIV-1 suggests that HCV under specific circumstances is transmitted via sexual contact. During sexual transmission HCV has to cross the epithelial barrier to either directly enter the blood stream or indirectly via mucosal immune cells. However, the mechanisms of sexual transmission of HCV remain unclear. We investigated the role of Langerhans cells (LCs) in HCV susceptibility during sexual contact as LCs are among the first cells in mucosal tissues to encounter invading viruses. METHODS We investigated the phenotype of primary LCs in anal biopsies from MSM living with HIV-1. To investigate the role of primary LCs in HCV infection and transmission, we have used both isolated primary skin LCs and the ex vivo tissue transmission model. RESULTS Our data identified an important role for mucosal LCs in facilitating HCV transmission after HIV-1 exposure or immune activation. LCs were detected within mucosal anal tissues obtained from HIV-1 positive MSM biopsies. In order to perform functional studies, we used primary LCs from skin, which have a similar phenotype as mucosal LCs. Immature LCs were neither infected nor transmitted HCV to hepatocytes. Notably, exposure to HIV-1 significantly increased HCV transmission by LCs in the ex vivo transmission model. HIV-1 replication was crucial for the increased HCV transmission as HIV-1 inhibitors significantly reduced HIV-1-induced HCV transmission. Moreover, tissue immune activation of LCs also increased HCV transmission to target cells. CONCLUSIONS Thus, our data strongly indicate that HIV-1 or immune activation in MSM leads to capture of HCV by mucosal LCs, which might facilitate transmission to other cells or allow entry of HCV into the blood. This novel transmission mechanism by LCs also implicates that the activation state of LCs is an important cellular determinant for HCV susceptibility after sexual contact.
Collapse
Affiliation(s)
- Bernadien M Nijmeijer
- Department of Experimental ImmunologyAmsterdam Infection and Immunity InstituteAmsterdam University Medical CentersUniversity of AmsterdamAmsterdamThe Netherlands
| | - Ramin Sarrami‐Forooshani
- Department of Experimental ImmunologyAmsterdam Infection and Immunity InstituteAmsterdam University Medical CentersUniversity of AmsterdamAmsterdamThe Netherlands
| | - Gaby S Steba
- Department of Medical MicrobiologyClinical Virology LaboratoryAmsterdam University Medical CentersUniversity of AmsterdamAmsterdamThe Netherlands
| | - Renée RCE Schreurs
- Department of Experimental ImmunologyAmsterdam Infection and Immunity InstituteAmsterdam University Medical CentersUniversity of AmsterdamAmsterdamThe Netherlands
| | - Sylvie M Koekkoek
- Department of Medical MicrobiologyClinical Virology LaboratoryAmsterdam University Medical CentersUniversity of AmsterdamAmsterdamThe Netherlands
| | - Richard Molenkamp
- Department of Medical MicrobiologyClinical Virology LaboratoryAmsterdam University Medical CentersUniversity of AmsterdamAmsterdamThe Netherlands
| | - Janke Schinkel
- Department of Medical MicrobiologyClinical Virology LaboratoryAmsterdam University Medical CentersUniversity of AmsterdamAmsterdamThe Netherlands
| | - Peter Reiss
- Department of Global HealthAmsterdam University Medical Centers, and Amsterdam Institute for Global Health and DevelopmentAmsterdam University Medical Centers HIV Monitoring FoundationAmsterdamThe Netherlands
- Division of Infectious DiseasesDepartment of Internal MedicineAmsterdam Infection and Immunity InstituteAmsterdam University Medical CentersUniversity of AmsterdamAmsterdamThe Netherlands
| | - Matthijs L Siegenbeek van Heukelom
- Division of Infectious DiseasesDepartment of Internal MedicineAmsterdam Infection and Immunity InstituteAmsterdam University Medical CentersUniversity of AmsterdamAmsterdamThe Netherlands
- Department of DermatologyAmsterdam University Medical CentersUniversity of AmsterdamAmsterdamThe Netherlands
| | - Marc van der Valk
- Division of Infectious DiseasesDepartment of Internal MedicineAmsterdam Infection and Immunity InstituteAmsterdam University Medical CentersUniversity of AmsterdamAmsterdamThe Netherlands
| | - Carla MS Ribeiro
- Department of Experimental ImmunologyAmsterdam Infection and Immunity InstituteAmsterdam University Medical CentersUniversity of AmsterdamAmsterdamThe Netherlands
| | - Teunis BH Geijtenbeek
- Department of Experimental ImmunologyAmsterdam Infection and Immunity InstituteAmsterdam University Medical CentersUniversity of AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
20
|
Schreurs RRCE, Baumdick ME, Sagebiel AF, Kaufmann M, Mokry M, Klarenbeek PL, Schaltenberg N, Steinert FL, van Rijn JM, Drewniak A, The SMML, Bakx R, Derikx JPM, de Vries N, Corpeleijn WE, Pals ST, Gagliani N, Friese MA, Middendorp S, Nieuwenhuis EES, Reinshagen K, Geijtenbeek TBH, van Goudoever JB, Bunders MJ. Human Fetal TNF-α-Cytokine-Producing CD4 + Effector Memory T Cells Promote Intestinal Development and Mediate Inflammation Early in Life. Immunity 2019; 50:462-476.e8. [PMID: 30770246 DOI: 10.1016/j.immuni.2018.12.010] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 10/01/2018] [Accepted: 12/05/2018] [Indexed: 11/29/2022]
Abstract
Although the fetal immune system is considered tolerogenic, preterm infants can suffer from severe intestinal inflammation, including necrotizing enterocolitis (NEC). Here, we demonstrate that human fetal intestines predominantly contain tumor necrosis factor-α (TNF-α)+CD4+CD69+ T effector memory (Tem) cells. Single-cell RNA sequencing of fetal intestinal CD4+ T cells showed a T helper 1 phenotype and expression of genes mediating epithelial growth and cell cycling. Organoid co-cultures revealed a dose-dependent, TNF-α-mediated effect of fetal intestinal CD4+ T cells on intestinal stem cell (ISC) development, in which low T cell numbers supported epithelial development, whereas high numbers abrogated ISC proliferation. CD4+ Tem cell frequencies were higher in inflamed intestines from preterm infants with NEC than in healthy infant intestines and showed enhanced TNF signaling. These findings reveal a distinct population of TNF-α-producing CD4+ T cells that promote mucosal development in fetal intestines but can also mediate inflammation upon preterm birth.
Collapse
Affiliation(s)
- Renée R C E Schreurs
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands; Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Martin E Baumdick
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg 20251, Germany
| | - Adrian F Sagebiel
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg 20251, Germany
| | - Max Kaufmann
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg 20251, Germany
| | - Michal Mokry
- Division of Pediatrics, Department of Pediatric Gastroenterology, Wilhelmina Children's Hospital, Utrecht University Medical Center, Utrecht University, Utrecht 3584 EA, the Netherlands; Regenerative Medicine Center Utrecht, Utrecht University Medical Center, University of Utrecht, Utrecht 3584 CT, the Netherlands
| | - Paul L Klarenbeek
- Department of Clinical Immunology and Rheumatology and Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands; Amsterdam Rheumatology & Immunology Center, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Nicola Schaltenberg
- Department of General, Visceral, and Thoracic Surgery and I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany; Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Fenja L Steinert
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg 20251, Germany
| | - Jorik M van Rijn
- Division of Pediatrics, Department of Pediatric Gastroenterology, Wilhelmina Children's Hospital, Utrecht University Medical Center, Utrecht University, Utrecht 3584 EA, the Netherlands; Regenerative Medicine Center Utrecht, Utrecht University Medical Center, University of Utrecht, Utrecht 3584 CT, the Netherlands
| | - Agata Drewniak
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands; Kiadis Pharma B.V., Amsterdam 1105 BV, the Netherlands
| | - Sarah-May M L The
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands; Department of Pediatric Surgery, Pediatric Surgery Center of Amsterdam, Amsterdam University Medical Center, Amsterdam 1105 AZ, the Netherlands
| | - Roel Bakx
- Department of Pediatric Surgery, Pediatric Surgery Center of Amsterdam, Amsterdam University Medical Center, Amsterdam 1105 AZ, the Netherlands
| | - Joep P M Derikx
- Department of Pediatric Surgery, Pediatric Surgery Center of Amsterdam, Amsterdam University Medical Center, Amsterdam 1105 AZ, the Netherlands
| | - Niek de Vries
- Department of Clinical Immunology and Rheumatology and Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands; Amsterdam Rheumatology & Immunology Center, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Willemijn E Corpeleijn
- Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Steven T Pals
- Department of Pathology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Nicola Gagliani
- Department of General, Visceral, and Thoracic Surgery and I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany; Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institute, Stockholm 17176, Sweden
| | - Manuel A Friese
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg 20251, Germany
| | - Sabine Middendorp
- Division of Pediatrics, Department of Pediatric Gastroenterology, Wilhelmina Children's Hospital, Utrecht University Medical Center, Utrecht University, Utrecht 3584 EA, the Netherlands; Regenerative Medicine Center Utrecht, Utrecht University Medical Center, University of Utrecht, Utrecht 3584 CT, the Netherlands
| | - Edward E S Nieuwenhuis
- Division of Pediatrics, Department of Pediatric Gastroenterology, Wilhelmina Children's Hospital, Utrecht University Medical Center, Utrecht University, Utrecht 3584 EA, the Netherlands; Regenerative Medicine Center Utrecht, Utrecht University Medical Center, University of Utrecht, Utrecht 3584 CT, the Netherlands
| | - Konrad Reinshagen
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Teunis B H Geijtenbeek
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Johannes B van Goudoever
- Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands; Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam 1081 HV, the Netherlands
| | - Madeleine J Bunders
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands; Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands; Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg 20251, Germany.
| |
Collapse
|
21
|
Krabbendam L, Nagasawa M, Spits H, Bal SM. Isolation of Human Innate Lymphoid Cells. ACTA ACUST UNITED AC 2018; 122:e55. [DOI: 10.1002/cpim.55] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Lisette Krabbendam
- Department of Experimental Immunology, Academic Medical Center Amsterdam The Netherlands
| | - Maho Nagasawa
- Department of Experimental Immunology, Academic Medical Center Amsterdam The Netherlands
| | - Hergen Spits
- Department of Experimental Immunology, Academic Medical Center Amsterdam The Netherlands
| | - Suzanne M. Bal
- Department of Experimental Immunology, Academic Medical Center Amsterdam The Netherlands
| |
Collapse
|
22
|
Happle C, Meyer-Decking L, Dreier A, Wetzke M, Gläsener S, Grychtol R, Braubach P, Jablonka A, Haid S, Jirmo AC, Habener A, Skuljec J, Hansen G. Improved protocol for simultaneous analysis of leukocyte subsets and epithelial cells from murine and human lung. Exp Lung Res 2018; 44:127-136. [PMID: 29677457 DOI: 10.1080/01902148.2018.1432721] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
PURPOSE To study and isolate lung cells by flow cytometry, enzymatic digestion and generation of single cell suspensions is required. This significantly influences expression of cellular epitopes and protocols need to be adapted for the best isolation and subsequent analysis of specific cellular subsets. MATERIALS AND METHODS We optimized protocols for the simultaneous isolation and characterization of specific human and murine lung cell types. For alveolar epithelial cells (AEC), a primarily dispase based digestion method and for leukocytes, a primarily collagenase based technique was adapted. Protocols were applied in parallel in either single experimental mice or human lung specimens. RESULTS Optimized dispase/DNase digestion yielded a high percentage of Epcam+CD45-CD31- AEC as assessed by flow cytometry. Epcam+CD45-CD3-CD11b-CD11c-CD16/32-CD19-CD31-F4/80- AEC were readily sortable with high purity and typical morphology and function upon in vitro stimulation with lipopolysaccharide or respiratory-syncytial-virus (RSV) infection. To analyze lung leukocytes, specimens were digested with an adapted collagenase/DNase protocol yielding high percentages of viable leukocytes with typical morphology, function, and preserved subset specific leukocyte markers. Both protocols could be applied simultaneously in a single experimental mouse post mortem. Application of both digestion methods in primary human lung specimens yielded similar results with high proportions of Epcam+CD45- human AEC after dispase/DNase digestion and preservation of human T cell epitopes after collagenase/DNase digestion. CONCLUSION The here described protocols were optimized for the simple and efficient isolation of murine and human lung cells. In contrast to previously described techniques, they permit simultaneous in-depth characterization of pulmonary epithelial cells and leukocyte subsets such as T helper, cytotoxic T, and B cells from one sample. As such, they may help to comprehensively and sustainably characterize murine and human lung specimens and facilitate studies on the role of lung immune cells in different respiratory pathologies.
Collapse
Affiliation(s)
- Christine Happle
- a Department of Pediatrics, Allergology, and Neonatology , Hannover Medical School , Hannover , Germany.,b German Center for Lung Research , Biomedical Research in End Stage and Obstructive Lung Disease/ BREATH Hannover , Germany
| | - Lena Meyer-Decking
- a Department of Pediatrics, Allergology, and Neonatology , Hannover Medical School , Hannover , Germany
| | - Anika Dreier
- a Department of Pediatrics, Allergology, and Neonatology , Hannover Medical School , Hannover , Germany.,b German Center for Lung Research , Biomedical Research in End Stage and Obstructive Lung Disease/ BREATH Hannover , Germany
| | - Martin Wetzke
- a Department of Pediatrics, Allergology, and Neonatology , Hannover Medical School , Hannover , Germany
| | - Stephanie Gläsener
- b German Center for Lung Research , Biomedical Research in End Stage and Obstructive Lung Disease/ BREATH Hannover , Germany
| | - Ruth Grychtol
- a Department of Pediatrics, Allergology, and Neonatology , Hannover Medical School , Hannover , Germany.,b German Center for Lung Research , Biomedical Research in End Stage and Obstructive Lung Disease/ BREATH Hannover , Germany
| | - Peter Braubach
- b German Center for Lung Research , Biomedical Research in End Stage and Obstructive Lung Disease/ BREATH Hannover , Germany.,c Department of Pathology , Hannover Medical School , Hannover , Germany
| | - Alexandra Jablonka
- d Department of Clinical Immunology and Rheumatology , Hannover Medical School , Hannover , Germany.,e German Center for Infection Research (DZIF) , Standort Hannover-Braunschweig , Germany
| | - Sibylle Haid
- f Experimental Virology and TWINCORE , a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research , Hannover , Germany
| | - Adan Chari Jirmo
- a Department of Pediatrics, Allergology, and Neonatology , Hannover Medical School , Hannover , Germany.,b German Center for Lung Research , Biomedical Research in End Stage and Obstructive Lung Disease/ BREATH Hannover , Germany
| | - Anika Habener
- a Department of Pediatrics, Allergology, and Neonatology , Hannover Medical School , Hannover , Germany.,b German Center for Lung Research , Biomedical Research in End Stage and Obstructive Lung Disease/ BREATH Hannover , Germany
| | - Jelena Skuljec
- a Department of Pediatrics, Allergology, and Neonatology , Hannover Medical School , Hannover , Germany
| | - Gesine Hansen
- a Department of Pediatrics, Allergology, and Neonatology , Hannover Medical School , Hannover , Germany.,b German Center for Lung Research , Biomedical Research in End Stage and Obstructive Lung Disease/ BREATH Hannover , Germany
| |
Collapse
|
23
|
Trapecar M, Khan S, Roan NR, Chen TH, Telwatte S, Deswal M, Pao M, Somsouk M, Deeks SG, Hunt PW, Yukl S, Sanjabi S. An Optimized and Validated Method for Isolation and Characterization of Lymphocytes from HIV+ Human Gut Biopsies. AIDS Res Hum Retroviruses 2017; 33:S31-S39. [PMID: 28882052 DOI: 10.1089/aid.2017.0208] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The gastrointestinal (GI) tract harbors most of the body's immune cells and is also a major HIV reservoir in ART-treated patients. To achieve a cure, most HIV-infected cells must be identified and eliminated. While obtaining gut biopsies is a relatively noninvasive method of sampling relevant tissue for monitoring HIV activity, immune cell isolation from these limited tissue samples has proven to be challenging. Enzymatic tissue digestion is required for maximal immune cell isolation from gut biopsies. However, these enzymatic digestions can also be detrimental for preservation of cellular surface markers that are required for accurate identification of various subsets of leukocytes. In this study, we describe an optimized protocol for isolation of lymphocytes from human gut biopsies. We also discuss our validation results, which show that compared with several other collagenase preparations, the use of CSLPA maintains high lymphocyte recovery while preserving the integrity of most cellular surface antigens that we tested. Importantly, chemokine receptors that are used to characterize various subsets of T cells, which are notorious for being digested during a typical enzymatic tissue digestion, are highly preserved using this protocol.
Collapse
Affiliation(s)
- Martin Trapecar
- Gladstone Institute of Virology and Immunology, Gladstone Institutes, San Francisco, California
| | - Shahzada Khan
- Gladstone Institute of Virology and Immunology, Gladstone Institutes, San Francisco, California
| | - Nadia R. Roan
- Gladstone Institute of Virology and Immunology, Gladstone Institutes, San Francisco, California
- Department of Urology, University of California, San Francisco, San Francisco, California
| | - Tsui-Hua Chen
- San Francisco VA Health Care System and University of California, San Francisco (UCSF), San Francisco, California
| | - Sushama Telwatte
- San Francisco VA Health Care System and University of California, San Francisco (UCSF), San Francisco, California
| | - Monika Deswal
- Positive Health Program, Department of Medicine, University of California, San Francisco, San Francisco, California
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Montha Pao
- Positive Health Program, Department of Medicine, University of California, San Francisco, San Francisco, California
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Ma Somsouk
- Division of Gastroenterology, Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Steven G. Deeks
- Positive Health Program, Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Peter W. Hunt
- Division of Experimental Medicine, University of California, San Francisco, San Francisco, California
| | - Steven Yukl
- San Francisco VA Health Care System and University of California, San Francisco (UCSF), San Francisco, California
| | - Shomyseh Sanjabi
- Gladstone Institute of Virology and Immunology, Gladstone Institutes, San Francisco, California
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, California
| |
Collapse
|