1
|
Gheorghiu M, Trandafir MF, Savu O, Pasarica D, Bleotu C. Unexpectedly High and Difficult-to-Explain Regenerative Capacity in an 82-Year-Old Patient with Insulin-Requiring Type 2 Diabetes and End-Stage Renal Disease. J Clin Med 2025; 14:2556. [PMID: 40283387 PMCID: PMC12027714 DOI: 10.3390/jcm14082556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 04/02/2025] [Accepted: 04/05/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: The case we present is part of a large study that we conducted on hemodialysis patients with type 2 diabetes mellitus (T2DM) and which set the following objectives: studying changes in the intestinal microbiota, innate and acquired immune response capacity, and tissue regeneration. Methods: (1) For the genetic study of the gut microbiota, special techniques that are not based on cultivation were used since most of the species in the intestinal flora are not cultivable. (2) The immunological study had two targets: innate immunity (inflammation) and adaptive immunity (we chose to address the cellular immune response because, unlike the humoral one, it is insufficiently studied in this category of associated pathologies). As markers for innate immunity (inflammation), the following were determined: IL-6, sIL-6R, IL-1β, TNFα, IL-10, and NGAL. TNFβ/LTα was determined as a marker for adaptive immunity (the cellular immune response). (3) The study of tissue regeneration capacity was performed using NT-3 (this is the first study to do so) and VEGFβ (another marker that is scarce in this category of patients) as markers. All the aforementioned compounds were determined from serum samples, utilizing Merck Millipore ELISA kits for IL-6, IL-1β, IL-10, NT-3, and VEGF β, and Elabscience ELISA kits for IL-6R, TNFα, TNFβ, and NGAL. Results: We were very surprised to find unexpected immunological changes and tissue regenerative capacity in one of the patients studied, an 82-year-old female patient diagnosed with insulin-dependent T2DM with multiple complications, including end-stage renal disease (ESRD). The patient showed a huge capacity for tissue regeneration, combined with amplification of immunological capacity, in comparison to patients in the same group (T2DM and ESRD) and to those in the control group (ESRD). Thus, extremely elevated serum concentrations of IL-1β, IL-6, IL-10, and TNF-β, as well as the tissue regeneration indicators NT-3 and VEGFβ, were obtained in comparison to all other members of the patient group. At the same time, serum levels of the soluble IL-6 receptor (sIL6-R) and TNFα were greatly reduced compared to the test group's mean. Conclusions: All the data obtained during our research were corroborated with those from the specialized literature and entitle us to support the hypothesis that the cause of these unexpected behaviors is the genetically conditioned overproduction (possibly acquired post-infection) of IL-6, along with its predominant anti-inflammatory and pro-regenerative signaling through the membrane-bound receptor IL-6R.
Collapse
Affiliation(s)
- Mihaela Gheorghiu
- Pathophysiology and Immunology Department, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (M.-F.T.); (O.S.); (D.P.)
| | - Maria-Florina Trandafir
- Pathophysiology and Immunology Department, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (M.-F.T.); (O.S.); (D.P.)
| | - Octavian Savu
- “N.C. Paulescu” National Institute of Diabetes, Nutrition and Metabolic Diseases, 020475 Bucharest, Romania
- Doctoral School of “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Daniela Pasarica
- Pathophysiology and Immunology Department, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (M.-F.T.); (O.S.); (D.P.)
| | - Coralia Bleotu
- “Stefan S. Nicolau” Institute of Virology, 030304 Bucharest, Romania;
| |
Collapse
|
2
|
Shoemaker RL, Larsen RJ, Larsen PA. Single-domain antibodies and aptamers drive new opportunities for neurodegenerative disease research. Front Immunol 2024; 15:1426656. [PMID: 39238639 PMCID: PMC11374656 DOI: 10.3389/fimmu.2024.1426656] [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: 05/01/2024] [Accepted: 07/22/2024] [Indexed: 09/07/2024] Open
Abstract
Neurodegenerative diseases (NDs) in mammals, such as Alzheimer's disease (AD), Parkinson's disease (PD), and transmissible spongiform encephalopathies (TSEs), are characterized by the accumulation of misfolded proteins in the central nervous system (CNS). Despite the presence of these pathogenic proteins, the immune response in affected individuals remains notably muted. Traditional immunological strategies, particularly those reliant on monoclonal antibodies (mAbs), face challenges related to tissue penetration, blood-brain barrier (BBB) crossing, and maintaining protein stability. This has led to a burgeoning interest in alternative immunotherapeutic avenues. Notably, single-domain antibodies (or nanobodies) and aptamers have emerged as promising candidates, as their reduced size facilitates high affinity antigen binding and they exhibit superior biophysical stability compared to mAbs. Aptamers, synthetic molecules generated from DNA or RNA ligands, present both rapid production times and cost-effective solutions. Both nanobodies and aptamers exhibit inherent qualities suitable for ND research and therapeutic development. Cross-seeding events must be considered in both traditional and small-molecule-based immunodiagnostic and therapeutic approaches, as well as subsequent neurotoxic impacts and complications beyond protein aggregates. This review delineates the challenges traditional immunological methods pose in ND research and underscores the potential of nanobodies and aptamers in advancing next-generation ND diagnostics and therapeutics.
Collapse
Affiliation(s)
- Rachel L Shoemaker
- Minnesota Center for Prion Research and Outreach (MNPRO), University of Minnesota, St. Paul, MN, United States
- Department of Biomedical and Veterinary Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN, United States
| | - Roxanne J Larsen
- Department of Biomedical and Veterinary Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN, United States
- Priogen Corp., St. Paul, MN, United States
| | - Peter A Larsen
- Minnesota Center for Prion Research and Outreach (MNPRO), University of Minnesota, St. Paul, MN, United States
- Department of Biomedical and Veterinary Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN, United States
| |
Collapse
|
3
|
Velikova T, Kaouri IE, Bakopoulou K, Gulinac M, Naydenova K, Dimitrov M, Peruhova M, Lazova S. Mucosal Immunity and Trained Innate Immunity of the Gut. GASTROENTEROLOGY INSIGHTS 2024; 15:661-675. [DOI: 10.3390/gastroent15030048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2024] Open
Abstract
Mucosal immunity and trained innate immunity of the gut play a pivotal role in maintaining intestinal homeostasis and defending against microbial pathogens. This review provides an overview of the mechanisms underlying mucosal immunity and the concept of trained innate immunity in the gut. We discuss the interaction between gut microbiota and the host immune system, highlighting the role of epithelial cells, dendritic cells, and innate lymphoid cells, as well as the novel concept of trained innate immunity and its role in perpetuating or attenuating gut inflammation. We also comment on the current models for investigating mucosal immunity, their limitations, and how they can be overcome. Additionally, we explore the potential therapeutic implications of modulating mucosal immunity and trained innate immunity in gastrointestinal diseases. Only by elucidating the mechanisms underlying mucosal immunity and the concept of trained innate immunity, innovative approaches to modulate immune responses and restore intestinal homeostasis in the context of gastrointestinal disorders could be implemented.
Collapse
Affiliation(s)
- Tsvetelina Velikova
- Medical Faculty, Sofia University “St. Kliment Ohridski”, 1 Kozyak Str., 1407 Sofia, Bulgaria
| | - Issa El Kaouri
- Faculty of Medicine, Medical University of Sofia, Boulevard “Akademik Ivan Evstratiev Geshov” 15, 1431 Sofia, Bulgaria
| | - Konstantina Bakopoulou
- Faculty of Medicine, Medical University of Sofia, Boulevard “Akademik Ivan Evstratiev Geshov” 15, 1431 Sofia, Bulgaria
| | - Milena Gulinac
- Medical Faculty, Sofia University “St. Kliment Ohridski”, 1 Kozyak Str., 1407 Sofia, Bulgaria
- Department of General and Clinical Pathology, Medical University of Plovdiv, Bul. Vasil Aprilov 15A, 4000 Plovdiv, Bulgaria
| | - Kremena Naydenova
- Clinical Center of Allergology, University Hospital Alexandrovska, Medical University of Sofia, Georgi Sofyiski 1 Str., 1431 Sofia, Bulgaria
| | - Martin Dimitrov
- Department of Genetics, Faculty of Biology, Sofia University “St. Kliment Ohridski”, 8 Dragan Tzankov Str., 1164 Sofia, Bulgaria
| | - Milena Peruhova
- Division of Gastroenterology, University Hospital “Heart and Brain”, Zdrave St. 1, 8000 Burgas, Bulgaria
| | - Snezhina Lazova
- Medical Faculty, Sofia University “St. Kliment Ohridski”, 1 Kozyak Str., 1407 Sofia, Bulgaria
- Pediatric Clinic, University Hospital “N. I. Pirogov”, 21 “General Eduard I. Totleben” Blvd, 1606 Sofia, Bulgaria
- Department of Healthcare, Faculty of Public Health “Prof. Tsekomir Vodenicharov, MD, DSc”, Medical University of Sofia, Bialo More 8 Str., 1527 Sofia, Bulgaria
| |
Collapse
|
4
|
Hernández-Barranco A, Santos V, Mazariegos MS, Caleiras E, Nogués L, Mourcin F, Léonard S, Oblet C, Genebrier S, Rossille D, Benguría A, Sanz A, Vázquez E, Dopazo A, Efeyan A, Ortega-Molina A, Cogne M, Tarte K, Peinado H. NGFR regulates stromal cell activation in germinal centers. Cell Rep 2024; 43:113705. [PMID: 38307025 DOI: 10.1016/j.celrep.2024.113705] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 11/30/2023] [Accepted: 01/09/2024] [Indexed: 02/04/2024] Open
Abstract
Nerve growth factor receptor (NGFR) is expressed by follicular dendritic cells (FDCs). However, the role of NGFR in the humoral response is not well defined. Here, we study the effect of Ngfr loss on lymph node organization and function, demonstrating that Ngfr depletion leads to spontaneous germinal center (GC) formation and an expansion of the GC B cell compartment. In accordance with this effect, stromal cells are altered in Ngfr-/- mice with a higher frequency of FDCs, characterized by CD21/35, MAdCAM-1, and VCAM-1 overexpression. GCs are located ectopically in Ngfr-/- mice, with lost polarization together with impaired high-affinity antibody production and an increase in circulating autoantibodies. We observe higher levels of autoantibodies in Bcl2 Tg/Ngfr-/- mice, concomitant with a higher incidence of autoimmunity and lower overall survival. Our work shows that NGFR is involved in maintaining GC structure and function, participating in GC activation, antibody production, and immune tolerance.
Collapse
Affiliation(s)
- Alberto Hernández-Barranco
- Microenvironment and Metastasis Laboratory, Molecular Oncology Program, Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain
| | - Vanesa Santos
- Microenvironment and Metastasis Laboratory, Molecular Oncology Program, Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain
| | - Marina S Mazariegos
- Microenvironment and Metastasis Laboratory, Molecular Oncology Program, Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain; Liver Injury and Inflammation Laboratory, Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University, 28040 Madrid, Spain
| | - Eduardo Caleiras
- Histopathology Unit, Biotechnology Program, Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain
| | - Laura Nogués
- Microenvironment and Metastasis Laboratory, Molecular Oncology Program, Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain
| | - Frédéric Mourcin
- UMR U1236, University Rennes, INSERM, EFS Bretagne, Equipe Labellisée Ligue Contre le Cancer, 35000 Rennes, France
| | - Simon Léonard
- UMR U1236, University Rennes, INSERM, EFS Bretagne, Equipe Labellisée Ligue Contre le Cancer, 35000 Rennes, France
| | - Christelle Oblet
- Immunology Department, Faculty of Medicine, Limoges University, CNRS Umr 7276, Inserm U1262, 87000 Limoges, France
| | - Steve Genebrier
- UMR U1236, University Rennes, INSERM, EFS Bretagne, Equipe Labellisée Ligue Contre le Cancer, 35000 Rennes, France
| | - Delphine Rossille
- UMR U1236, University Rennes, INSERM, EFS Bretagne, Equipe Labellisée Ligue Contre le Cancer, 35000 Rennes, France; SITI Lab, Pôle Biologie, CHU Rennes, 35000 Rennes, France
| | - Alberto Benguría
- Genomic Unit, Spanish National Cardiovascular Research, Carlos III, 28029 Madrid, Spain
| | - Alba Sanz
- Metabolism and Cell Signaling Laboratory, Molecular Oncology Program, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
| | - Enrique Vázquez
- Genomic Unit, Spanish National Cardiovascular Research, Carlos III, 28029 Madrid, Spain
| | - Ana Dopazo
- Genomic Unit, Spanish National Cardiovascular Research, Carlos III, 28029 Madrid, Spain
| | - Alejo Efeyan
- Metabolism and Cell Signaling Laboratory, Molecular Oncology Program, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
| | - Ana Ortega-Molina
- Metabolism and Cell Signaling Laboratory, Molecular Oncology Program, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain; Metabolism in Cancer and Ageing Laboratory, Immune System and Function Department, Centro de Biología Molecular "Severo Ochoa" (CMBSO-CSIC), Madrid 28049, Spain
| | - Michel Cogne
- UMR U1236, University Rennes, INSERM, EFS Bretagne, Equipe Labellisée Ligue Contre le Cancer, 35000 Rennes, France
| | - Karin Tarte
- UMR U1236, University Rennes, INSERM, EFS Bretagne, Equipe Labellisée Ligue Contre le Cancer, 35000 Rennes, France; SITI Lab, Pôle Biologie, CHU Rennes, 35000 Rennes, France
| | - Héctor Peinado
- Microenvironment and Metastasis Laboratory, Molecular Oncology Program, Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain.
| |
Collapse
|
5
|
Liang Y, Fu YX. LIGHTing CAR T in the tumor microenvironment. Mol Ther 2023; 31:2570-2571. [PMID: 37607540 PMCID: PMC10492016 DOI: 10.1016/j.ymthe.2023.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 08/09/2023] [Accepted: 08/10/2023] [Indexed: 08/24/2023] Open
Affiliation(s)
- Yong Liang
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Yang-Xin Fu
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
6
|
Lim VY, Feng X, Miao R, Zehentmeier S, Ewing-Crystal N, Lee M, Tumanov AV, Oh JE, Iwasaki A, Wang A, Choi J, Pereira JP. Mature B cells and mesenchymal stem cells control emergency myelopoiesis. Life Sci Alliance 2023; 6:e202301924. [PMID: 36717247 PMCID: PMC9889502 DOI: 10.26508/lsa.202301924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 01/20/2023] [Accepted: 01/23/2023] [Indexed: 01/31/2023] Open
Abstract
Systemic inflammation halts lymphopoiesis and prioritizes myeloid cell production. How blood cell production switches from homeostasis to emergency myelopoiesis is incompletely understood. Here, we show that lymphotoxin-β receptor (LTβR) signaling in combination with TNF and IL-1 receptor signaling in bone marrow mesenchymal stem cells (MSCs) down-regulates Il7 expression to shut down lymphopoiesis during systemic inflammation. LTβR signaling in MSCs also promoted CCL2 production during systemic inflammation. Pharmacological or genetic blocking of LTβR signaling in MSCs partially enabled lymphopoiesis and reduced monocyte numbers in the spleen during systemic inflammation, which correlated with reduced survival during systemic bacterial and viral infections. Interestingly, lymphotoxin-α1β2 delivered by B-lineage cells, and specifically by mature B cells, contributed to promote Il7 down-regulation and reduce MSC lymphopoietic activity. Our studies revealed an unexpected role of LTβR signaling in MSCs and identified recirculating mature B cells as an important regulator of emergency myelopoiesis.
Collapse
Affiliation(s)
- Vivian Y Lim
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Xing Feng
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Runfeng Miao
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Sandra Zehentmeier
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Nathan Ewing-Crystal
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Moonyoung Lee
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, South Korea
| | - Alexei V Tumanov
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Ji Eun Oh
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Akiko Iwasaki
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Andrew Wang
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
- Department of Medicine (Rheumatology), School of Medicine, Yale University, New Haven, CT, USA
| | - Jungmin Choi
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, South Korea
- Department of Genetics, School of Medicine, Yale University, New Haven, CT, USA
| | - João P Pereira
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| |
Collapse
|
7
|
Zhang Y, Feng X, Chen J, Liu J, Wu J, Tan H, Mi Z, Rong P. Controversial role of ILC3s in intestinal diseases: A novelty perspective on immunotherapy. Front Immunol 2023; 14:1134636. [PMID: 37063879 PMCID: PMC10090672 DOI: 10.3389/fimmu.2023.1134636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 03/13/2023] [Indexed: 03/31/2023] Open
Abstract
ILC3s have been identified as crucial immune regulators that play a role in maintaining host homeostasis and modulating the antitumor response. Emerging evidence supports the idea that LTi cells play an important role in initiating lymphoid tissue development, while other ILC3s can promote host defense and orchestrate adaptive immunity, mainly through the secretion of specific cytokines and crosstalk with other immune cells or tissues. Additionally, dysregulation of ILC3-mediated overexpression of cytokines, changes in subset abundance, and conversion toward other ILC subsets are closely linked with the occurrence of tumors and inflammatory diseases. Regulation of ILC3 cytokines, ILC conversion and LTi-induced TLSs may be a novel strategy for treating tumors and intestinal or extraintestinal inflammatory diseases. Herein, we discuss the development of ILCs, the biology of ILC3s, ILC plasticity, the correlation of ILC3s and adaptive immunity, crosstalk with the intestinal microenvironment, controversial roles of ILC3s in intestinal diseases and potential applications for treatment.
Collapse
Affiliation(s)
- Yunshu Zhang
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Xuefei Feng
- Department of Government & Public Administration, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Juan Chen
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiahao Liu
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jianmin Wu
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hongpei Tan
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ze Mi
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- *Correspondence: Ze Mi, ; Pengfei Rong,
| | - Pengfei Rong
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Key Laboratory of Biological Nanotechnology of National Health Commission, Xiangya Hospital, Central South University, Changsha, Hunan, China
- *Correspondence: Ze Mi, ; Pengfei Rong,
| |
Collapse
|
8
|
Zhang J, Cui J, Gao J, Zhang D, Lin D, Lin J. Polysaccharides of Plantago asiatica enhance antitumor activity via regulating macrophages to M1-like phenotype. Biomed Pharmacother 2023; 159:114246. [PMID: 36652734 DOI: 10.1016/j.biopha.2023.114246] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 01/04/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Monocyte-derived macrophages can be polarized into antitumor M1 phenotype, which inhibited the growth of tumors, and immune-suppressive M2 phenotype, which promoted the development and metastasis of tumors. Plantain polysaccharide (PLP), extracted from the Plantago asiatica, has shown its various biological activities. However, the ability of PLP involved in immune regulation was still obscure. Accordingly, we aimed to investigate whether PLP could polarize macrophages and further inhibit 4T1 tumor cells in vivo and in vitro. In this research, in vitro results showed that PLP displayed the potential in polarizing RAW264.7 macrophages into M1 phenotype and indirect inhibiting migratory effect on 4T1 cells. Furthermore, the phagocytosis and the release of reactive oxygen species (ROS) of macrophages were enhanced. In vivo anti-tumor results demonstrated that PLP could effectively inhibit the growth of 4T1 breast tumors by promoting accumulation of macrophages and T cells in the spleen and lymph node. In conclusion, these findings indicated that PLP inhibited the proliferation and progression of breast tumors by accumulating CD4+, CD8+ T cells and M1-like macrophages in lymph node and spleen, and therefore provided an experimental basis for PLP as a potential antitumor adjunctive therapy in preclinical and clinical trials.
Collapse
Affiliation(s)
- Jiatong Zhang
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jingwen Cui
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jiafeng Gao
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Di Zhang
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Degui Lin
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, China.
| | - Jiahao Lin
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, China; Center of Research and Innovation of Chinese Traditional Veterinary Medicine, China Agricultural University, Beijing, China.
| |
Collapse
|
9
|
Kloch A, Mierzejewska EJ, Welc-Falęciak R, Bajer A, Biedrzycka A. Cytokine gene polymorphism and parasite susceptibility in free-living rodents: Importance of non-coding variants. PLoS One 2023; 18:e0258009. [PMID: 36693052 PMCID: PMC9873194 DOI: 10.1371/journal.pone.0258009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 01/06/2023] [Indexed: 01/25/2023] Open
Abstract
Associations between genetic variants and susceptibility to infections have long been studied in free-living hosts so as to infer the contemporary evolutionary forces that shape the genetic polymorphisms of immunity genes. Despite extensive studies of proteins interacting with pathogen-derived ligands, such as MHC (major histocompatilbility complex) or TLR (Toll-like receptors), little is known about the efferent arm of the immune system. Cytokines are signalling molecules that trigger and modulate the immune response, acting as a crucial link between innate and adaptive immunity. In the present study we investigated how genetic variation in cytokines in bank voles Myodes glareolus affects their susceptibility to infection by parasites (nematodes: Aspiculuris tianjensis, Heligmosomum mixtum, Heligmosomoides glareoli) and microparasites (Cryptosporidium sp, Babesia microti, Bartonella sp.). We focused on three cytokines: tumour necrosis factor (TNF), lymphotoxin alpha (LTα), and interferon beta (IFNβ1). Overall, we identified four single nucleotide polymorphisms (SNPs) associated with susceptibility to nematodes: two located in LTα and two in IFNβ1. One of those variants was synonymous, another located in an intron. Each SNP associated with parasite load was located in or next to a codon under selection, three codons displayed signatures of positive selection, and one of purifying selection. Our results indicate that cytokines are prone to parasite-driven selection and that non-coding variants, although commonly disregarded in studies of the genetic background of host-parasite co-evolution, may play a role in susceptibility to infections in wild systems.
Collapse
Affiliation(s)
- Agnieszka Kloch
- Department of Ecology, Faculty of Biology, University of Warsaw, Warszawa, Poland
- * E-mail:
| | - Ewa J. Mierzejewska
- Wild Urban Evolution and Ecology Laboratory, Centre of New Technologies, University of Warsaw, Warszawa, Poland
| | - Renata Welc-Falęciak
- Department of Parasitology, Faculty of Biology, University of Warsaw, Warszawa, Poland
| | - Anna Bajer
- Department of Eco-epidemiology of Parasitic Diseases, Faculty of Biology, University of Warsaw, Warszawa, Poland
| | | |
Collapse
|
10
|
Chen X, Zheng Y, Liu S, Yu W, Liu Z. CD169 + subcapsular sinus macrophage-derived microvesicles are associated with light zone follicular dendritic cells. Eur J Immunol 2022; 52:1581-1594. [PMID: 35907260 PMCID: PMC9804338 DOI: 10.1002/eji.202249879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 07/21/2022] [Accepted: 07/29/2022] [Indexed: 01/05/2023]
Abstract
Follicular dendritic cells (FDCs) are a specialized type of stromal cells that exclusively reside in B-cell follicles. When inflammation occurs, the FDC network is reorganized to support germinal center (GC) polarization into the light zone (LZ) and dark zone (DZ). Despite the indispensable role of FDCs in supporting humoral responses, the FDC regulatory requirements remain incompletely defined. In this study, we unexpectedly observed an accumulation of CD169+ subcapsular sinus macrophage (SSM)-derived microvesicles (MVs) in the B-cell zone, which were tightly associated with the FDC network. Interestingly, a selective deposition of CD169+ MVs was detected in both GC LZ FDCs in secondary follicles and on predetermined LZ FDCs in primary follicles. The ablation of CD169+ MVs, resulting from SSM depletion, resulted in significantly decreased expression of LZ-related genes in FDCs. In addition, we found that CD169+ MVs could colocalize with fluorescently tagged antigen-containing immune complexes (ICs), supporting a possible role of CD169+ MVs in transporting antigens to the FDC network. Thus, our data reveal intimate crosstalk between FDCs and SSMs located outside B-cell follicles via SSM-released MVs, providing a novel perspective on the mechanisms underlying the regulation of FDC maturation and polarization.
Collapse
Affiliation(s)
- Xin Chen
- Shanghai Institute of ImmunologyDepartment of Immunology and Microbiology, State Key Laboratory of Oncogenes and Related GenesShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yuhan Zheng
- Shanghai Institute of ImmunologyDepartment of Immunology and Microbiology, State Key Laboratory of Oncogenes and Related GenesShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Siming Liu
- Shanghai Institute of ImmunologyDepartment of Immunology and Microbiology, State Key Laboratory of Oncogenes and Related GenesShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Wenjing Yu
- Shanghai Institute of ImmunologyDepartment of Immunology and Microbiology, State Key Laboratory of Oncogenes and Related GenesShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Zhiduo Liu
- Shanghai Institute of ImmunologyDepartment of Immunology and Microbiology, State Key Laboratory of Oncogenes and Related GenesShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
11
|
Zehentmeier S, Lim VY, Ma Y, Fossati J, Ito T, Jiang Y, Tumanov AV, Lee HJ, Dillinger L, Kim J, Csomos K, Walter JE, Choi J, Pereira JP. Dysregulated stem cell niches and altered lymphocyte recirculation cause B and T cell lymphopenia in WHIM syndrome. Sci Immunol 2022; 7:eabo3170. [PMID: 36149943 PMCID: PMC9614684 DOI: 10.1126/sciimmunol.abo3170] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Gain-of-function (GOF) mutations in CXCR4 cause WHIM (warts, hypogammaglobulinemia, infections, and myelokathexis) syndrome, characterized by infections, leukocyte retention in bone marrow (BM), and blood leukopenias. B lymphopenia is evident at early progenitor stages, yet why do CXCR4 GOF mutations that cause B (and T) lymphopenia remain obscure? Using a CXCR4 R334X GOF mouse model of WHIM syndrome, we showed that lymphopoiesis is reduced because of a dysregulated mesenchymal stem cell (MSC) transcriptome characterized by a switch from an adipogenic to an osteolineage-prone program with limited lymphopoietic activity. We identify lymphotoxin beta receptor (LTβR) as a critical pathway promoting interleukin-7 (IL-7) down-regulation in MSCs. Blocking LTβR or CXCR4 signaling restored IL-7 production and B cell development in WHIM mice. LTβR blocking also increased production of IL-7 and B cell activating factor (BAFF) in secondary lymphoid organs (SLOs), increasing B and T cell numbers in the periphery. These studies revealed that LTβR signaling in BM MSCs and SLO stromal cells limits the lymphocyte compartment size.
Collapse
Affiliation(s)
- Sandra Zehentmeier
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, 300 Cedar Street, New Haven, CT, USA
| | - Vivian Y Lim
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, 300 Cedar Street, New Haven, CT, USA
| | - Yifan Ma
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, 300 Cedar Street, New Haven, CT, USA
| | - Julia Fossati
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, 300 Cedar Street, New Haven, CT, USA
| | - Takeshi Ito
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, 300 Cedar Street, New Haven, CT, USA
| | - Yawen Jiang
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, 300 Cedar Street, New Haven, CT, USA
| | - Alexei V Tumanov
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Ho-Joon Lee
- Department of Genetics and Yale Center for Genome Analysis, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, USA
| | - Lukas Dillinger
- X4 Pharmaceuticals Inc., Cambridge, MA, USA
- X4 Pharmaceuticals Inc., Vienna, Austria
| | - Jihyun Kim
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Krisztian Csomos
- Division of Allergy and Immunology, Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Jolan E Walter
- Division of Allergy and Immunology, Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
- Division Allergy and Immunology, Johns Hopkins All Children's Hospital, St. Petersburg, FL, USA
| | - Jungmin Choi
- Department of Genetics and Yale Center for Genome Analysis, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, USA
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - João P Pereira
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, 300 Cedar Street, New Haven, CT, USA
| |
Collapse
|
12
|
Wang B, Wang M, Ao D, Wei X. CXCL13-CXCR5 axis: Regulation in inflammatory diseases and cancer. Biochim Biophys Acta Rev Cancer 2022; 1877:188799. [PMID: 36103908 DOI: 10.1016/j.bbcan.2022.188799] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 09/06/2022] [Accepted: 09/06/2022] [Indexed: 01/10/2023]
Abstract
Chemokine C-X-C motif ligand 13 (CXCL13), originally identified as a B-cell chemokine, plays an important role in the immune system. The interaction between CXCL13 and its receptor, the G-protein coupled receptor (GPCR) CXCR5, builds a signaling network that regulates not only normal organisms but also the development of many diseases. However, the precise action mechanism remains unclear. In this review, we discussed the functional mechanisms of the CXCL13-CXCR5 axis under normal conditions, with special focus on its association with diseases. For certain refractory diseases, we emphasize the diagnostic and therapeutic role of CXCL13-CXCR5 axis.
Collapse
Affiliation(s)
- Binhan Wang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Manni Wang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Danyi Ao
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
13
|
LAMP-1 Chimeric to HIV-1 p55Gag in the Immunization of Neonate Mice Induces an Early Germinal Center Formation and AID Expression. Vaccines (Basel) 2022; 10:vaccines10081246. [PMID: 36016134 PMCID: PMC9414238 DOI: 10.3390/vaccines10081246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 11/29/2022] Open
Abstract
Neonates have a limited adaptive response of plasma cells, germinal center (GC) B cells, and T follicular helper cells (TFH). As neonatal vaccination can be an important tool for AIDS prevention, these limitations need to be overcome. Chimeric DNA vaccine encoding p55Gag HIV-1 protein conjugated with lysosomal-associated membrane protein 1 (LAMP-1) has been described as immunogenic in the neonate period. Herein, we investigated the immunologic mechanisms involved in neonatal immunization with a LAMP-1/p55Gag (LAMP/Gag) DNA vaccine in a C57BL/6 mouse background. Neonatal LAMP/Gag vaccination induced strong Gag-specific T-cell response until adulthood and elevated levels of anti-Gag IgG antibodies. We also demonstrated for the first time that the immunogenicity of the neonatal period with LAMP/Gag is due to the induction of high-affinity anti-p24 IgG antibodies and long-term plasma cells. Together with that, there is the generation of early TFH cells and the formation of GC sites with the upregulation of activation-induced cytidine deaminase (AID) enzyme mRNA and protein expression in draining lymph nodes after neonatal LAMP/Gag vaccination. These findings underscore that the LAMP-1 strategy in the chimeric vaccine could be useful to enhance antibody production even in the face of neonatal immaturity, and they contribute to the development of new vaccine approaches for other emerging pathogens at an early stage of life.
Collapse
|
14
|
LTα, TNF, and ILC3 in Peyer's Patch Organogenesis. Cells 2022; 11:cells11121970. [PMID: 35741098 PMCID: PMC9221848 DOI: 10.3390/cells11121970] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 06/11/2022] [Accepted: 06/17/2022] [Indexed: 02/05/2023] Open
Abstract
TNF and LTα are structurally related cytokines of the TNF superfamily. Their genes are located in close proximity to each other and to the Ltb gene within the TNF/LT locus inside MHC. Unlike Ltb, transcription of Tnf and of Lta is tightly controlled, with the Tnf gene being an immediate early gene that is rapidly induced in response to various inflammatory stimuli. Genes of the TNF/LT locus play a crucial role in lymphoid tissue organogenesis, although some aspects of their specific contribution remain controversial. Here, we present new findings and discuss the distinct contribution of TNF produced by ILC3 cells to Peyer’s patch organogenesis.
Collapse
|
15
|
Aldossary AM, Ekweremadu CS, Offe IM, Alfassam HA, Han S, Onyali VC, Ozoude CH, Ayeni EA, Nwagwu CS, Halwani AA, Almozain NH, Tawfik EA. A guide to oral vaccination: Highlighting electrospraying as a promising manufacturing technique toward a successful oral vaccine development. Saudi Pharm J 2022; 30:655-668. [PMID: 35812139 PMCID: PMC9257926 DOI: 10.1016/j.jsps.2022.03.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/10/2022] [Indexed: 11/26/2022] Open
Abstract
Most vaccines approved by regulatory bodies are administered via intramuscular or subcutaneous injections and have shortcomings, such as the risk of needle-associated blood infections, pain and swelling at the injection site. Orally administered vaccines are of interest, as they elicit both systemic and mucosal immunities, in which mucosal immunity would neutralize the mucosa invading pathogen before the onset of an infection. Hence, oral vaccination can eliminate the injection associated adverse effects and enhance the person's compliance. Conventional approaches to manufacturing oral vaccines, such as coacervation, spray drying, and membrane emulsification, tend to alter the structural proteins in vaccines that result from high temperature, organic and toxic solvents during production. Electrohydrodynamic processes, specifically electrospraying, could solve these challenges, as it also modulates antigen release and has a high loading efficiency. This review will highlight the mucosal immunity and biological basis of the gastrointestinal immune system, different oral vaccine delivery approaches, and the application of electrospraying in vaccines development.
Collapse
Key Words
- APCs, Antigen-presenting cells
- BALT, Bronchus-associated lymphoid tissue
- DCs, Dendritic cells
- Electrospraying
- FAE, Follicle-associated epithelium
- GALT, Gut-associated lymphoid tissue
- GIT, Gastro-intestinal tract
- HIV, Human immune virus
- IL, Interleukin
- Ig, Immunoglobulin
- Infectious diseases
- MALT, Mucosa-associated lymphoid tissue
- MLN, Mesenteric lymph nodes
- MNPs, Micro/Nanoparticles
- Mucosal immunity
- Mucosal pathogen
- NALT, Nasopharynx-associated lymphoid tissue
- Oral vaccines
- PLGA, Polylactide-co-glycolide acid
- PP, Peyer’s patches
- Secretory, (SIgA1 and SIgA2)
- TGF-β, Transforming growth factor-β
- TLRs, Toll-like receptors
- WHO, World Health Organization
Collapse
Affiliation(s)
- Ahmad M. Aldossary
- National Center of Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh, Saudi Arabia
| | - Chinedu S.M. Ekweremadu
- Department of Pharmaceutics and Pharmaceutical Technology, Enugu State University of Science and Technology, Agbani, Enugu State, Nigeria
| | - Ifunanya M. Offe
- Department of Biological Sciences, Faculty of Natural Sciences and Environmental Studies, Godfrey Okoye University, Enugu, Nigeria
| | - Haya A. Alfassam
- KACST-BWH Centre of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science and Technology (KACST), Riyadh, Saudi Arabia
| | - Sooyeon Han
- UCL Medical School, University College London, London, United Kingdom
| | - Vivian C. Onyali
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, United State
| | - Chukwuebuka H. Ozoude
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, University of Lagos, College of Medicine Campus, Surulere, Lagos, Nigeria
| | - Emmanuel A. Ayeni
- The Research Unit, New Being Foundation, Abuja, Nigeria
- Department of Pharmacognosy and Drug Development, Faculty of Pharmaceutical Sciences, Ahmadu Bello University, Zaria, Nigeria
| | - Chinekwu S. Nwagwu
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, University of Nigeria Nsukka, Nigeria
| | - Abdulrahman A. Halwani
- Pharmaceutics Department, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
- Regenerative Medicine Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Nada H. Almozain
- Pharmaceutical Services Department, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Essam A. Tawfik
- National Center of Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh, Saudi Arabia
| |
Collapse
|
16
|
Nayar S, Pontarini E, Campos J, Berardicurti O, Smith CG, Asam S, Gardner DH, Colafrancesco S, Lucchesi D, Coleby R, Chung MM, Iannizzotto V, Hunter K, Bowman SJ, Carlesso G, Herbst R, McGettrick HM, Browning J, Buckley CD, Fisher BA, Bombardieri M, Barone F. Immunofibroblasts regulate LTα3 expression in tertiary lymphoid structures in a pathway dependent on ICOS/ICOSL interaction. Commun Biol 2022; 5:413. [PMID: 35508704 PMCID: PMC9068764 DOI: 10.1038/s42003-022-03344-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 04/10/2022] [Indexed: 01/15/2023] Open
Abstract
Immunofibroblasts have been described within tertiary lymphoid structures (TLS) that regulate lymphocyte aggregation at sites of chronic inflammation. Here we report, for the first time, an immunoregulatory property of this population, dependent on inducible T-cell co-stimulator ligand and its ligand (ICOS/ICOS-L). During inflammation, immunofibroblasts, alongside other antigen presenting cells, like dendritic cells (DCs), upregulate ICOSL, binding incoming ICOS + T cells and inducing LTα3 production that, in turn, drives the chemokine production required for TLS assembly via TNFRI/II engagement. Pharmacological or genetic blocking of ICOS/ICOS-L interaction results in defective LTα expression, abrogating both lymphoid chemokine production and TLS formation. These data provide evidence of a previously unknown function for ICOSL-ICOS interaction, unveil a novel immunomodulatory function for immunofibroblasts, and reveal a key regulatory function of LTα3, both as biomarker of TLS establishment and as first driver of TLS formation and maintenance in mice and humans.
Collapse
Affiliation(s)
- Saba Nayar
- Centre for Translational Inflammation Research, Institute of Inflammation and Ageing, College of Medical & Dental Sciences, University of Birmingham Research Laboratories, Queen Elizabeth Hospital, Birmingham, B15 2WB, UK
- National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre and Department of Rheumatology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Birmingham Tissue Analytics, Institute of Translational Medicine, University of Birmingham, Birmingham, UK
| | - Elena Pontarini
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Joana Campos
- Centre for Translational Inflammation Research, Institute of Inflammation and Ageing, College of Medical & Dental Sciences, University of Birmingham Research Laboratories, Queen Elizabeth Hospital, Birmingham, B15 2WB, UK
| | - Onorina Berardicurti
- Rheumatology Unit, Department of Biotechnological and Applied Clinical Science, University of L'Aquila, L'Aquila, Italy
| | - Charlotte G Smith
- Centre for Translational Inflammation Research, Institute of Inflammation and Ageing, College of Medical & Dental Sciences, University of Birmingham Research Laboratories, Queen Elizabeth Hospital, Birmingham, B15 2WB, UK
| | - Saba Asam
- Centre for Translational Inflammation Research, Institute of Inflammation and Ageing, College of Medical & Dental Sciences, University of Birmingham Research Laboratories, Queen Elizabeth Hospital, Birmingham, B15 2WB, UK
| | - David H Gardner
- Centre for Translational Inflammation Research, Institute of Inflammation and Ageing, College of Medical & Dental Sciences, University of Birmingham Research Laboratories, Queen Elizabeth Hospital, Birmingham, B15 2WB, UK
- Birmingham Tissue Analytics, Institute of Translational Medicine, University of Birmingham, Birmingham, UK
| | | | - Davide Lucchesi
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Rachel Coleby
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Ming-May Chung
- Centre for Translational Inflammation Research, Institute of Inflammation and Ageing, College of Medical & Dental Sciences, University of Birmingham Research Laboratories, Queen Elizabeth Hospital, Birmingham, B15 2WB, UK
| | - Valentina Iannizzotto
- Centre for Translational Inflammation Research, Institute of Inflammation and Ageing, College of Medical & Dental Sciences, University of Birmingham Research Laboratories, Queen Elizabeth Hospital, Birmingham, B15 2WB, UK
| | - Kelly Hunter
- Birmingham Tissue Analytics, Institute of Translational Medicine, University of Birmingham, Birmingham, UK
| | - Simon J Bowman
- Centre for Translational Inflammation Research, Institute of Inflammation and Ageing, College of Medical & Dental Sciences, University of Birmingham Research Laboratories, Queen Elizabeth Hospital, Birmingham, B15 2WB, UK
- National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre and Department of Rheumatology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Gianluca Carlesso
- Early Oncology ICA, AstraZeneca, One Medimmune Way, Gaithersburg, MD 20878, MD, USA
| | - Ronald Herbst
- Early Oncology ICA, AstraZeneca, One Medimmune Way, Gaithersburg, MD 20878, MD, USA
| | - Helen M McGettrick
- Centre for Translational Inflammation Research, Institute of Inflammation and Ageing, College of Medical & Dental Sciences, University of Birmingham Research Laboratories, Queen Elizabeth Hospital, Birmingham, B15 2WB, UK
| | - Jeff Browning
- Departments of Microbiology and Rheumatology, Boston University School of Medicine, Boston, MA, USA
| | - Christopher D Buckley
- Centre for Translational Inflammation Research, Institute of Inflammation and Ageing, College of Medical & Dental Sciences, University of Birmingham Research Laboratories, Queen Elizabeth Hospital, Birmingham, B15 2WB, UK
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Benjamin A Fisher
- Centre for Translational Inflammation Research, Institute of Inflammation and Ageing, College of Medical & Dental Sciences, University of Birmingham Research Laboratories, Queen Elizabeth Hospital, Birmingham, B15 2WB, UK
- National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre and Department of Rheumatology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Michele Bombardieri
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Francesca Barone
- Centre for Translational Inflammation Research, Institute of Inflammation and Ageing, College of Medical & Dental Sciences, University of Birmingham Research Laboratories, Queen Elizabeth Hospital, Birmingham, B15 2WB, UK.
- Candel Therapeutics, Needham, Boston, MA, USA.
| |
Collapse
|
17
|
Piacente F, Bottero M, Benzi A, Vigo T, Uccelli A, Bruzzone S, Ferrara G. Neuroprotective Potential of Dendritic Cells and Sirtuins in Multiple Sclerosis. Int J Mol Sci 2022; 23:ijms23084352. [PMID: 35457169 PMCID: PMC9025744 DOI: 10.3390/ijms23084352] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/06/2022] [Accepted: 04/11/2022] [Indexed: 12/04/2022] Open
Abstract
Myeloid cells, including parenchymal microglia, perivascular and meningeal macrophages, and dendritic cells (DCs), are present in the central nervous system (CNS) and establish an intricate relationship with other cells, playing a crucial role both in health and in neurological diseases. In this context, DCs are critical to orchestrating the immune response linking the innate and adaptive immune systems. Under steady-state conditions, DCs patrol the CNS, sampling their local environment and acting as sentinels. During neuroinflammation, the resulting activation of DCs is a critical step that drives the inflammatory response or the resolution of inflammation with the participation of different cell types of the immune system (macrophages, mast cells, T and B lymphocytes), resident cells of the CNS and soluble factors. Although the importance of DCs is clearly recognized, their exact function in CNS disease is still debated. In this review, we will discuss modern concepts of DC biology in steady-state and during autoimmune neuroinflammation. Here, we will also address some key aspects involving DCs in CNS patrolling, highlighting the neuroprotective nature of DCs and emphasizing their therapeutic potential for the treatment of neurological conditions. Recently, inhibition of the NAD+-dependent deac(et)ylase sirtuin 6 was demonstrated to delay the onset of experimental autoimmune encephalomyelitis, by dampening DC trafficking towards inflamed LNs. Thus, a special focus will be dedicated to sirtuins’ role in DCs functions.
Collapse
Affiliation(s)
- Francesco Piacente
- Department of Experimental Medicine (DIMES), University of Genova, Viale Benedetto XV, 1, 16132 Genoa, Italy; (F.P.); (A.B.)
| | - Marta Bottero
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy; (M.B.); (T.V.); (A.U.); (G.F.)
| | - Andrea Benzi
- Department of Experimental Medicine (DIMES), University of Genova, Viale Benedetto XV, 1, 16132 Genoa, Italy; (F.P.); (A.B.)
| | - Tiziana Vigo
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy; (M.B.); (T.V.); (A.U.); (G.F.)
| | - Antonio Uccelli
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy; (M.B.); (T.V.); (A.U.); (G.F.)
| | - Santina Bruzzone
- Department of Experimental Medicine (DIMES), University of Genova, Viale Benedetto XV, 1, 16132 Genoa, Italy; (F.P.); (A.B.)
- Correspondence: ; Tel.: +39-(0)10-353-8150
| | - Giovanni Ferrara
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy; (M.B.); (T.V.); (A.U.); (G.F.)
| |
Collapse
|
18
|
Ahmed A, Köhler S, Klotz R, Giese N, Hackert T, Springfeld C, Zörnig I, Jäger D, Halama N. Tertiary lymphoid structures and their association to immune phenotypes and circulatory IL2 levels in pancreatic ductal adenocarcinoma. Oncoimmunology 2022; 11:2027148. [PMID: 35127251 PMCID: PMC8812743 DOI: 10.1080/2162402x.2022.2027148] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 12/17/2021] [Accepted: 12/17/2021] [Indexed: 12/19/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDA) is usually unresponsive to immunotherapeutic approaches. However, tertiary lymphoid structures (TLS) are associated with favorable patient outcomes in PDA. A better understanding of the B cell infiltrate and biological features of TLS formation is needed to further explore their potential and improve patient management. We analyzed tumor tissues (n = 55) and corresponding blood samples (n = 51) from PDA patients by systematical immunohistochemistry and multiplex cytokine measurements. The tissue was compartmentalized in "tumor" and "stroma" and separately examined. Clinical patient information was used to perform survival analyses. We found that the mere number of B cells is not associated with patient survival, but formation of TLS in the peritumoral stroma is a prognostic favorable marker in PDA patients. TLS-positive tissues show a higher density of CD8+ T cells and CD20+ B cells and a higher IL2 level in the peritumoral stroma than tissues without TLS. Compartmental assessment shows that gradients of IL2 expression differ with regard to TLS formation: TLS presence is associated with higher IL2 levels in the stromal than in the tumoral compartment, while no difference is seen in patients without TLS. Focusing on the stroma-to-serum gradient, only patients without TLS show significantly higher IL2 levels in the serum than in stroma. Finally, low circulatory IL2 levels are associated with local TLS formation. Our findings highlight that TLS are prognostic favorable and associated with antitumoral features in the microenvironment of PDA. Also, we propose easily accessible serum IL2 levels as a potential marker for TLS prediction.
Collapse
Affiliation(s)
- Azaz Ahmed
- Medical Oncology and Internal Medicine VI, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, University Heidelberg, Heidelberg, Germany
- Translational Immunotherapy (D240), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sophia Köhler
- Medical Oncology and Internal Medicine VI, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, University Heidelberg, Heidelberg, Germany
| | - Rosa Klotz
- General, Visceral and Transplantation Surgery, University Hospital Heidelberg, University Heidelberg, Heidelberg, Germany
| | - Nathalia Giese
- General, Visceral and Transplantation Surgery, University Hospital Heidelberg, University Heidelberg, Heidelberg, Germany
| | - Thilo Hackert
- General, Visceral and Transplantation Surgery, University Hospital Heidelberg, University Heidelberg, Heidelberg, Germany
| | - Christoph Springfeld
- Medical Oncology and Internal Medicine VI, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, University Heidelberg, Heidelberg, Germany
| | - Inka Zörnig
- Medical Oncology and Internal Medicine VI, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, University Heidelberg, Heidelberg, Germany
| | - Dirk Jäger
- Medical Oncology and Internal Medicine VI, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, University Heidelberg, Heidelberg, Germany
- Applied Tumor Immunity Clinical Cooperation Unit (D120), National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Niels Halama
- Medical Oncology and Internal Medicine VI, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, University Heidelberg, Heidelberg, Germany
- Translational Immunotherapy (D240), German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
19
|
Sun J, Min YQ, Li Y, Sun X, Deng F, Wang H, Ning YJ. Animal Model of Severe Fever With Thrombocytopenia Syndrome Virus Infection. Front Microbiol 2022; 12:797189. [PMID: 35087498 PMCID: PMC8787146 DOI: 10.3389/fmicb.2021.797189] [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] [Received: 10/18/2021] [Accepted: 11/29/2021] [Indexed: 12/30/2022] Open
Abstract
Severe fever with thrombocytopenia syndrome (SFTS), an emerging life-threatening infectious disease caused by SFTS bunyavirus (SFTSV; genus Bandavirus, family Phenuiviridae, order Bunyavirales), has been a significant medical problem. Currently, there are no licensed vaccines or specific therapeutic agents available and the viral pathogenesis remains largely unclear. Developing appropriate animal models capable of recapitulating SFTSV infection in humans is crucial for both the study of the viral pathogenic processes and the development of treatment and prevention strategies. Here, we review the current progress in animal models for SFTSV infection by summarizing susceptibility of various potential animal models to SFTSV challenge and the clinical manifestations and histopathological changes in these models. Together with exemplification of studies on SFTSV molecular mechanisms, vaccine candidates, and antiviral drugs, in which animal infection models are utilized, the strengths and limitations of the existing SFTSV animal models and some important directions for future research are also discussed. Further exploration and optimization of SFTSV animal models and the corresponding experimental methods will be undoubtedly valuable for elucidating the viral infection and pathogenesis and evaluating vaccines and antiviral therapies.
Collapse
Affiliation(s)
- Jiawen Sun
- State Key Laboratory of Virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Yuan-Qin Min
- State Key Laboratory of Virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Yunjie Li
- State Key Laboratory of Virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Xiulian Sun
- State Key Laboratory of Virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Fei Deng
- State Key Laboratory of Virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Hualin Wang
- State Key Laboratory of Virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Yun-Jia Ning
- State Key Laboratory of Virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
20
|
Jiang L, Yilmaz M, Uehara M, Cavazzoni CB, Kasinath V, Zhao J, Naini SM, Li X, Banouni N, Fiorina P, Shin SR, Tullius SG, Bromberg JS, Sage PT, Abdi R. Characterization of Leptin Receptor + Stromal Cells in Lymph Node. Front Immunol 2022; 12:730438. [PMID: 35111151 PMCID: PMC8801441 DOI: 10.3389/fimmu.2021.730438] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 12/29/2021] [Indexed: 11/14/2022] Open
Abstract
Lymph node (LN)-resident stromal cells play an essential role in the proper functioning of LNs. The stromal compartment of the LN undergoes significant compensatory changes to produce a milieu amenable for regulation of the immune response. We have identified a distinct population of leptin receptor-expressing (LepR+) stromal cells, located in the vicinity of the high endothelial venules (HEVs) and lymphatics. These LepR+ stromal cells expressed markers for fibroblastic reticular cells (FRCs), but they lacked markers for follicular dendritic cells (FDCs) and marginal reticular cells (MRCs). Leptin signaling deficiency led to heightened inflammatory responses within the LNs of db/db mice, leakiness of HEVs, and lymphatic fragmentation. Leptin signaling through the JAK/STAT pathway supported LN stromal cell survival and promoted the anti-inflammatory properties of these cells. Conditional knockout of the LepR+ stromal cells in LNs resulted in HEV and extracellular matrix (ECM) abnormalities. Treatment of ob/ob mice with an agonist leptin fusion protein restored the microarchitecture of LNs, reduced intra-LN inflammatory responses, and corrected metabolic abnormalities. Future studies are needed to study the importance of LN stomal cell dysfunction to the pathogenesis of inflammatory responses in type 2 diabetes (T2D) in humans.
Collapse
Affiliation(s)
- Liwei Jiang
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
| | - Mine Yilmaz
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Mayuko Uehara
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Cecilia B. Cavazzoni
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Vivek Kasinath
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Jing Zhao
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Said Movahedi Naini
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Xiaofei Li
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Naima Banouni
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Paolo Fiorina
- Division of Nephrology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Su Ryon Shin
- Biomaterials Innovation Research Center, Division of Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, United States
| | - Stefan G. Tullius
- Division of Transplant Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Jonathan S. Bromberg
- Departments of Surgery and Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Peter T. Sage
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Reza Abdi
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
21
|
El Shikh MEM, El Sayed R, Aly NAR, Prediletto E, Hands R, Fossati-Jimack L, Bombardieri M, Lewis MJ, Pitzalis C. Follicular dendritic cell differentiation is associated with distinct synovial pathotype signatures in rheumatoid arthritis. Front Med (Lausanne) 2022; 9:1013660. [PMID: 36465908 PMCID: PMC9709129 DOI: 10.3389/fmed.2022.1013660] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 10/24/2022] [Indexed: 11/17/2022] Open
Abstract
Follicular dendritic cells (FDCs) fundamentally contribute to the formation of synovial ectopic lymphoid-like structures in rheumatoid arthritis (RA) which is associated with poor clinical prognosis. Despite this critical role, regulation of FDC development in the RA synovium and its correlation with synovial pathotype differentiation remained largely unknown. Here, we demonstrate that CNA.42+ FDCs distinctively express the pericyte/fibroblast-associated markers PDGFR-β, NG2, and Thy-1 in the synovial perivascular space but not in established follicles. In addition, synovial RNA-Seq analysis revealed that expression of the perivascular FDC markers was strongly correlated with PDGF-BB and fibroid synovitis, whereas TNF-α/LT-β was significantly associated with lymphoid synovitis and expression of CR1, CR2, and FcγRIIB characteristic of mature FDCs in lymphoid follicles. Moreover, PDGF-BB induced CNA.42+ FDC differentiation and CXCL13 secretion from NG2+ synovial pericytes, and together with TNF-α/LT-β conversely regulated early and late FDC differentiation genes in unsorted RA synovial fibroblasts (RASF) and this was confirmed in flow sorted stromal cell subsets. Furthermore, RASF TNF-αR expression was upregulated by TNF-α/LT-β and PDGF-BB; and TNF-α/LT-β-activated RASF retained ICs and induced B cell activation in in vitro germinal center reactions typical of FDCs. Additionally, FDCs trapped peptidyl citrulline, and strongly correlated with IL-6 expression, and plasma cell, B cell, and T cell infiltration of the RA synovium. Moreover, synovial FDCs were significantly associated with RA disease activity and radiographic features of tissue damage. To the best of our knowledge, this is the first report describing the reciprocal interaction between PDGF-BB and TNF-α/LT-β in synovial FDC development and evolution of RA histological pathotypes. Selective targeting of this interplay could inhibit FDC differentiation and potentially ameliorate RA in clinically severe and drug-resistant patients.
Collapse
|
22
|
Chen RE, Gorman MJ, Zhu DY, Carreño JM, Yuan D, VanBlargan LA, Burdess S, Lauffenburger DA, Kim W, Turner JS, Droit L, Handley SA, Chahin S, Deepak P, O'Halloran JA, Paley MA, Presti RM, Wu GF, Krammer F, Alter G, Ellebedy AH, Kim AHJ, Diamond MS. Reduced antibody activity against SARS-CoV-2 B.1.617.2 delta virus in serum of mRNA-vaccinated individuals receiving tumor necrosis factor-α inhibitors. MED 2021; 2:1327-1341.e4. [PMID: 34812429 PMCID: PMC8599018 DOI: 10.1016/j.medj.2021.11.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/27/2021] [Accepted: 11/11/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Although vaccines effectively prevent coronavirus disease 2019 (COVID-19) in healthy individuals, they appear to be less immunogenic in individuals with chronic inflammatory disease (CID) or receiving chronic immunosuppression therapy. METHODS Here we assessed a cohort of 77 individuals with CID treated as monotherapy with chronic immunosuppressive drugs for antibody responses in serum against historical and variant severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) viruses after immunization with the BNT162b2 mRNA vaccine. FINDINGS Longitudinal analysis showed the greatest reductions in neutralizing antibodies and Fc effector function capacity in individuals treated with tumor necrosis factor alpha (TNF-α) inhibitors (TNFi), and this pattern appeared to be worse against the B.1.617.2 delta virus. Within 5 months of vaccination, serum neutralizing titers of all TNFi-treated individuals tested fell below the presumed threshold correlate for antibody-mediated protection. However, TNFi-treated individuals receiving a third mRNA vaccine dose boosted their serum neutralizing antibody titers by more than 16-fold. CONCLUSIONS Vaccine boosting or administration of long-acting prophylaxis (e.g., monoclonal antibodies) will likely be required to prevent SARS-CoV-2 infection in this susceptible population. FUNDING This study was supported by grants and contracts from the NIH (R01 AI157155, R01AI151178, and HHSN75N93019C00074; NIAID Centers of Excellence for Influenza Research and Response (CEIRR) contracts HHSN272201400008C and 75N93021C00014; and Collaborative Influenza Vaccine Innovation Centers [CIVIC] contract 75N93019C00051).
Collapse
Affiliation(s)
- Rita E Chen
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Daniel Y Zhu
- Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Juan Manuel Carreño
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dansu Yuan
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Laura A VanBlargan
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Samantha Burdess
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Wooseob Kim
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jackson S Turner
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Lindsay Droit
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Scott A Handley
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Salim Chahin
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Parakkal Deepak
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Jane A O'Halloran
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Michael A Paley
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Rachel M Presti
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO, USA
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, Saint Louis, MO, USA
| | - Gregory F Wu
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Ali H Ellebedy
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
- Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO, USA
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, Saint Louis, MO, USA
| | - Alfred H J Kim
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO, USA
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
- Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO, USA
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
23
|
Duan L, Liu D, Chen H, Mintz MA, Chou MY, Kotov DI, Xu Y, An J, Laidlaw BJ, Cyster JG. Follicular dendritic cells restrict interleukin-4 availability in germinal centers and foster memory B cell generation. Immunity 2021; 54:2256-2272.e6. [PMID: 34555336 PMCID: PMC8516727 DOI: 10.1016/j.immuni.2021.08.028] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 06/02/2021] [Accepted: 08/27/2021] [Indexed: 11/24/2022]
Abstract
B cells within germinal centers (GCs) enter cycles of antibody affinity maturation or exit the GC as memory cells or plasma cells. Here, we examined the contribution of interleukin (IL)-4 on B cell fate decisions in the GC. Single-cell RNA-sequencing identified a subset of light zone GC B cells expressing high IL-4 receptor-a (IL4Ra) and CD23 and lacking a Myc-associated signature. These cells could differentiate into pre-memory cells. B cell-specific deletion of IL4Ra or STAT6 favored the pre-memory cell trajectory, and provision of exogenous IL-4 in a wild-type context reduced pre-memory cell frequencies. IL-4 acted during antigen-specific interactions but also influenced bystander cells. Deletion of IL4Ra from follicular dendritic cells (FDCs) increased the availability of IL-4 in the GC, impaired the selection of affinity-matured B cells, and reduced memory cell generation. We propose that GC FDCs establish a niche that limits bystander IL-4 activity, focusing IL-4 action on B cells undergoing selection and enhancing memory cell differentiation.
Collapse
Affiliation(s)
- Lihui Duan
- Department of Microbiology and Immunology and Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Dan Liu
- Department of Microbiology and Immunology and Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Hsin Chen
- Department of Microbiology and Immunology and Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Michelle A Mintz
- Department of Microbiology and Immunology and Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Marissa Y Chou
- Department of Microbiology and Immunology and Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Dmitri I Kotov
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, and Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Ying Xu
- Department of Microbiology and Immunology and Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jinping An
- Department of Microbiology and Immunology and Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Brian J Laidlaw
- Department of Microbiology and Immunology and Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jason G Cyster
- Department of Microbiology and Immunology and Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
24
|
The gut microbiota induces Peyer's-patch-dependent secretion of maternal IgA into milk. Cell Rep 2021; 36:109655. [PMID: 34496253 DOI: 10.1016/j.celrep.2021.109655] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 03/03/2021] [Accepted: 08/12/2021] [Indexed: 12/21/2022] Open
Abstract
The evolutionary strategy of transferring maternal antibodies via milk profoundly impacts the survival, lifelong health, and wellbeing of all neonates, including a pronounced impact on human breastfeeding success and infant development. While there has been increased recognition that interorgan connectivity influences the quality of a mother's milk, potentially to personalize it for her offspring, the underlying bases for these processes are incompletely resolved. Here, we define an essential role of Peyer's patches (PPs) for the generation of plasma cells that secrete maternal immunoglobulin A (IgA) into milk. Our metagenomic analysis reveals that the presence of certain residential microorganisms in the gastrointestinal (GI) tract, such as Bacteroides acidifaciens and Prevotella buccalis, is indispensable for the programming of maternal IgA synthesis prior to lactational transfer. Our data provide important insights into how the microbiome of the maternal GI environment, specifically through PPs, can be communicated to the next generation via milk.
Collapse
|
25
|
CXCL13 Signaling in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1302:71-90. [PMID: 34286442 DOI: 10.1007/978-3-030-62658-7_6] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Chemokines have emerged as important players in tumorigenic process. An extensive body of literature generated over the last two or three decades strongly implicate abnormally activated or functionally disrupted chemokine signaling in liaising most-if not all-hallmark processes of cancer. It is well-known that chemokine signaling networks within the tumor microenvironment are highly versatile and context-dependent: exert both pro-tumoral and antitumoral activities. The C-X-C motif chemokine ligand 13 (CXCL13), and its cognate receptor CXCR5, represents an emerging example of chemokine signaling axes, which express the ability to modulate tumor growth and progression in either way. Collateral evidence indicate that CXCL13-CXCR5 axis may directly modulate tumor growth by inducing proliferation of cancer cells, as well as promoting invasive phenotypes and preventing their apoptosis. In addition, CXCL13-CXCR5 axis may also indirectly modulate tumor growth by regulating noncancerous cells, particularly the immune cells, within the tumor microenvironment. Here, we review the role of CXCL13, together with CXCR5, in the human tumor microenvironment. We first elaborate their patterns of expression, regulation, and biological functions in normal physiology. We then consider how their aberrant activity, as a result of differential overexpression or co-expression, may directly or indirectly modulate the growth of tumors through effects on both cancerous and noncancerous cells.
Collapse
|
26
|
Shou Y, Koroleva E, Spencer CM, Shein SA, Korchagina AA, Yusoof KA, Parthasarathy R, Leadbetter EA, Akopian AN, Muñoz AR, Tumanov AV. Redefining the Role of Lymphotoxin Beta Receptor in the Maintenance of Lymphoid Organs and Immune Cell Homeostasis in Adulthood. Front Immunol 2021; 12:712632. [PMID: 34335629 PMCID: PMC8320848 DOI: 10.3389/fimmu.2021.712632] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 06/29/2021] [Indexed: 02/04/2023] Open
Abstract
Lymphotoxin beta receptor (LTβR) is a promising therapeutic target in autoimmune and infectious diseases as well as cancer. Mice with genetic inactivation of LTβR display multiple defects in development and organization of lymphoid organs, mucosal immune responses, IgA production and an autoimmune phenotype. As these defects are imprinted in embryogenesis and neonate stages, the impact of LTβR signaling in adulthood remains unclear. Here, to overcome developmental defects, we generated mice with inducible ubiquitous genetic inactivation of LTβR in adult mice (iLTβRΔ/Δ mice) and redefined the role of LTβR signaling in organization of lymphoid organs, immune response to mucosal bacterial pathogen, IgA production and autoimmunity. In spleen, postnatal LTβR signaling is required for development of B cell follicles, follicular dendritic cells (FDCs), recruitment of neutrophils and maintenance of the marginal zone. Lymph nodes of iLTβRΔ/Δ mice were reduced in size, lacked FDCs, and had disorganized subcapsular sinus macrophages. Peyer`s patches were smaller in size and numbers, and displayed reduced FDCs. The number of isolated lymphoid follicles in small intestine and colon were also reduced. In contrast to LTβR-/- mice, iLTβRΔ/Δ mice displayed normal thymus structure and did not develop signs of systemic inflammation and autoimmunity. Further, our results suggest that LTβR signaling in adulthood is required for homeostasis of neutrophils, NK, and iNKT cells, but is dispensable for the maintenance of polyclonal IgA production. However, iLTβRΔ/Δ mice exhibited an increased sensitivity to C. rodentium infection and failed to develop pathogen-specific IgA responses. Collectively, our study uncovers new insights of LTβR signaling in adulthood for the maintenance of lymphoid organs, neutrophils, NK and iNKT cells, and IgA production in response to mucosal bacterial pathogen.
Collapse
Affiliation(s)
- Yajun Shou
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States,Department of Gastroenterology, Second Xiangya Hospital of Central South University, Changsha, China
| | - Ekaterina Koroleva
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | | | - Sergey A. Shein
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Anna A. Korchagina
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Kizil A. Yusoof
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Raksha Parthasarathy
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Elizabeth A. Leadbetter
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Armen N. Akopian
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Amanda R. Muñoz
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Alexei V. Tumanov
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States,*Correspondence: Alexei V. Tumanov,
| |
Collapse
|
27
|
Rana M, La Bella A, Lederman R, Volpe BT, Sherry B, Diamond B. Follicular dendritic cell dysfunction contributes to impaired antigen-specific humoral responses in sepsis-surviving mice. J Clin Invest 2021; 131:146776. [PMID: 33956665 PMCID: PMC8203464 DOI: 10.1172/jci146776] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 04/29/2021] [Indexed: 12/29/2022] Open
Abstract
Sepsis survivors exhibit impaired responsiveness to antigen (Ag) challenge associated with increased mortality from infection. The contribution of follicular dendritic cells (FDCs) in the impaired humoral response in sepsis-surviving mice is investigated in this study. We demonstrated that mice subjected to sepsis from cecal ligation and puncture (CLP mice) have reduced NP-specific high-affinity class-switched Ig antibodies (Abs) compared with sham-operated control mice following immunization with the T cell-dependent Ag, NP-CGG. NP-specific germinal center (GC) B cells in CLP mice exhibited reduced TNF-α and AID mRNA expression compared with sham-operated mice. CLP mice showed a reduction in FDC clusters, a reduced binding of immune complexes on FDCs, and reduced mRNA expression of CR2, ICAM-1, VCAM-1, FcγRIIB, TNFR1, IKK2, and LTβR compared with sham-operated mice. Adoptive transfer studies showed that there was no B cell-intrinsic defect. In summary, our data suggest that the reduced Ag-specific Ab response in CLP mice is secondary to a disruption in FDC and GC B cell function.
Collapse
Affiliation(s)
- Minakshi Rana
- Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases
| | - Andrea La Bella
- Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases
| | - Rivka Lederman
- Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases
| | | | - Barbara Sherry
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, New York, New York, USA
| | - Betty Diamond
- Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases
| |
Collapse
|
28
|
Lütge M, Pikor NB, Ludewig B. Differentiation and activation of fibroblastic reticular cells. Immunol Rev 2021; 302:32-46. [PMID: 34046914 PMCID: PMC8361914 DOI: 10.1111/imr.12981] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/17/2021] [Accepted: 04/30/2021] [Indexed: 12/29/2022]
Abstract
Secondary lymphoid organs (SLO) are underpinned by fibroblastic reticular cells (FRC) that form dedicated microenvironmental niches to secure induction and regulation of innate and adaptive immunity. Distinct FRC subsets are strategically positioned in SLOs to provide niche factors and govern efficient immune cell interaction. In recent years, the use of specialized mouse models in combination with single-cell transcriptomics has facilitated the elaboration of the molecular FRC landscape at an unprecedented resolution. While single-cell RNA-sequencing has advanced the resolution of FRC subset characterization and function, the high dimensionality of the generated data necessitates careful analysis and validation. Here, we reviewed novel findings from high-resolution transcriptomic analyses that refine our understanding of FRC differentiation and activation processes in the context of infection and inflammation. We further discuss concepts, strategies, and limitations for the analysis of single-cell transcriptome data from FRCs and the wide-ranging implications for our understanding of stromal cell biology.
Collapse
Affiliation(s)
- Mechthild Lütge
- Institute of Immunobiology, Medical Research Center, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Natalia B Pikor
- Institute of Immunobiology, Medical Research Center, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Burkhard Ludewig
- Institute of Immunobiology, Medical Research Center, Kantonsspital St. Gallen, St. Gallen, Switzerland.,Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| |
Collapse
|
29
|
Sarmiento Varón L, De Rosa J, Rodriguez R, Fernández PM, Billordo LA, Baz P, Beccaglia G, Spada N, Mendoza FT, Barberis CM, Vay C, Arabolaza ME, Paoli B, Arana EI. Role of Tonsillar Chronic Inflammation and Commensal Bacteria in the Pathogenesis of Pediatric OSA. Front Immunol 2021; 12:648064. [PMID: 33995367 PMCID: PMC8116894 DOI: 10.3389/fimmu.2021.648064] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 04/06/2021] [Indexed: 01/01/2023] Open
Abstract
Immune responses at the boundary between the host and the world beyond are complex and mucosal tissue homeostasis relies on them. Obstructive sleep apnea (OSA) is a syndrome suffered by children with hypertrophied tonsils. We have previously demonstrated that these tonsils present a defective regulatory B cell (Breg) compartment. Here, we extend those findings by uncovering the crucial role of resident pro-inflammatory B and T cells in sustaining tonsillar hypertrophy and hyperplasia by producing TNFα and IL17, respectively, in ex vivo cultures. Additionally, we detected prominent levels of expression of CD1d by tonsillar stratified as well as reticular epithelium, which have not previously been reported. Furthermore, we evidenced the hypertrophy of germinal centers (GC) and the general hyperplasia of B lymphocytes within the tissue and the lumen of the crypts. Of note, such B cells resulted mainly (IgG/IgM)+ cells, with some IgA+ cells located marginally in the follicles. Finally, by combining bacterial culture from the tonsillar core and subsequent identification of the respective isolates, we determined the most prevalent species within the cohort of OSA patients. Although the isolated species are considered normal oropharyngeal commensals in children, we confirmed their capacity to breach the epithelial barrier. Our work sheds light on the pathological mechanism underlying OSA, highlighting the relevance taken by the host immune system when defining infection versus colonization, and opening alternatives of treatment.
Collapse
Affiliation(s)
- Lindybeth Sarmiento Varón
- Institute of Immunology, Genetics and Metabolism (INIGEM), Clinical Hospital 'José de San Martín', University of Buenos Aires (UBA), National Council for Scientific and Technological Research (CONICET), Buenos Aires, Argentina
| | - Javier De Rosa
- Institute of Immunology, Genetics and Metabolism (INIGEM), Clinical Hospital 'José de San Martín', University of Buenos Aires (UBA), National Council for Scientific and Technological Research (CONICET), Buenos Aires, Argentina
| | - Raquel Rodriguez
- Institute of Immunology, Genetics and Metabolism (INIGEM), Clinical Hospital 'José de San Martín', University of Buenos Aires (UBA), National Council for Scientific and Technological Research (CONICET), Buenos Aires, Argentina.,Allergy and Immunology Division, Clinical Hospital 'José de San Martín', UBA, Buenos Aires, Argentina
| | - Pablo M Fernández
- Institute of Immunology, Genetics and Metabolism (INIGEM), Clinical Hospital 'José de San Martín', University of Buenos Aires (UBA), National Council for Scientific and Technological Research (CONICET), Buenos Aires, Argentina.,Department of Immunology, School of Medicine, UBA, Buenos Aires, Argentina
| | - L Ariel Billordo
- Institute of Immunology, Genetics and Metabolism (INIGEM), Clinical Hospital 'José de San Martín', University of Buenos Aires (UBA), National Council for Scientific and Technological Research (CONICET), Buenos Aires, Argentina
| | - Plácida Baz
- Institute of Immunology, Genetics and Metabolism (INIGEM), Clinical Hospital 'José de San Martín', University of Buenos Aires (UBA), National Council for Scientific and Technological Research (CONICET), Buenos Aires, Argentina
| | - Gladys Beccaglia
- Department of Pathology, Clinical Hospital 'José de San Martín', Buenos Aires, Argentina
| | - Nicolás Spada
- Department of Pathology, Clinical Hospital 'José de San Martín', Buenos Aires, Argentina
| | - F Tatiana Mendoza
- Department of Clinical Biochemistry and Bacteriology, School of Pharmacy and Biochemistry, Clinical Hospital 'Jose de San Martín', UBA, Buenos Aires, Argentina
| | - Claudia M Barberis
- Department of Clinical Biochemistry and Bacteriology, School of Pharmacy and Biochemistry, Clinical Hospital 'Jose de San Martín', UBA, Buenos Aires, Argentina
| | - Carlos Vay
- Department of Clinical Biochemistry and Bacteriology, School of Pharmacy and Biochemistry, Clinical Hospital 'Jose de San Martín', UBA, Buenos Aires, Argentina
| | - M Elena Arabolaza
- Pediatric Otolaryngology Division, Clinical Hospital 'José de San Martín', Buenos Aires, Argentina
| | - Bibiana Paoli
- Pediatric Otolaryngology Division, Clinical Hospital 'José de San Martín', Buenos Aires, Argentina
| | - Eloísa I Arana
- Institute of Immunology, Genetics and Metabolism (INIGEM), Clinical Hospital 'José de San Martín', University of Buenos Aires (UBA), National Council for Scientific and Technological Research (CONICET), Buenos Aires, Argentina.,Department of Immunology, School of Medicine, UBA, Buenos Aires, Argentina
| |
Collapse
|
30
|
Novel Concepts: Langerhans Cells in the Tumour Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1273:147-158. [PMID: 33119880 DOI: 10.1007/978-3-030-49270-0_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Langerhans cells (LCs) are immune cells that reside in the stratified epithelium of the skin and mucosal membranes. They play a range of roles in the skin, including antigen presentation and maintenance of peripheral tolerance. Reports of LC numbers have been variable in different cancer types, with the majority of studies indicating a reduction in their number. Changes in the cytokine profile and other secreted molecules, downregulation of surface molecules on cells and hypoxia all contribute to the regulation of LCs in the tumour microenvironment. Functionally, LCs have been reported to regulate immunity and carcinogenesis in different cancer types. An improved understanding of the function and biology of LCs in tumours is essential knowledge that underpins the development of new cancer immunotherapies.
Collapse
|
31
|
Furukawa M, Ito S, Suzuki S, Fuchimoto D, Onishi A, Niimi K, Usami K, Wu G, Bazer FW, Ogasawara K, Watanabe K, Aso H, Nochi T. Organogenesis of Ileal Peyer's Patches Is Initiated Prenatally and Accelerated Postnatally With Comprehensive Proliferation of B Cells in Pigs. Front Immunol 2020; 11:604674. [PMID: 33424851 PMCID: PMC7793923 DOI: 10.3389/fimmu.2020.604674] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/05/2020] [Indexed: 11/21/2022] Open
Abstract
Morphogenesis and differentiation of organs is required for subsequent functional maturation. The morphological features of Peyer's patches vary among species. In pigs, they develop extensively in the ileum as ileal Peyer's patches (IPPs). However, the role of IPPs in the porcine immune system remains to be elucidated because of a lack of complete understanding of IPP organogenesis. Results of the present study revealed that development of porcine IPPs is initiated prenatally between embryonic days 76 and 91. The process of IPP organogenesis is concomitant with increased transcriptional patterns of CXCL13 and CCL19. IPPs undergo further development postnatally by forming central, marginal, and subepithelial zones. Importantly, a large number of proliferating B cells and apoptotic cells are found in porcine IPPs postnatally, but not prenatally. The expression level of IgM in proliferating B cells depends on the zone in which distinct B cells are separately localized after birth. Specifically, IgM+ cells are predominantly found in the central zone, whereas IgM-/low cells are abundant in the marginal zone. Importantly, the cellular feature of IPPs differs from that of mesenteric lymph nodes (MLNs) where such distinct zones are not formed both prenatally and postnatally. Our findings suggest that IPPs (not MLNs) in postnatal pigs are involved in complementing functions of the primary lymphoid tissue that promotes the differentiation and maturation of B cells.
Collapse
Affiliation(s)
- Mutsumi Furukawa
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Shun Ito
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Shunichi Suzuki
- Division of Animal Science, Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Tsukuba, Japan
| | - Daiichiro Fuchimoto
- Division of Animal Science, Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Tsukuba, Japan
| | - Akira Onishi
- Department of Animal Science and Resources, Nihon University College of Bioresource Sciences, Fujisawa, Japan
| | - Kanae Niimi
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Katsuki Usami
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Guoyao Wu
- Department of Animal Science, Texas A&M University, College Station, TX, United States
| | - Fuller W Bazer
- Department of Animal Science, Texas A&M University, College Station, TX, United States
| | - Kouetsu Ogasawara
- Department of Immunobiology, Tohoku University Institute of Development, Aging and Cancer, Sendai, Japan
| | - Kouichi Watanabe
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Hisashi Aso
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Tomonori Nochi
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.,International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
32
|
Wang X, Wu Z, Qiu W, Chen P, Xu X, Han W. Programming CAR T cells to enhance anti-tumor efficacy through remodeling of the immune system. Front Med 2020; 14:726-745. [PMID: 32794014 DOI: 10.1007/s11684-020-0746-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Accepted: 12/18/2019] [Indexed: 12/11/2022]
Abstract
Chimeric antigen receptor (CAR) T cells have been indicated effective in treating B cell acute lymphoblastic leukemia and non-Hodgkin lymphoma and have shown encouraging results in preclinical and clinical studies. However, CAR T cells have achieved minimal success against solid malignancies because of the additional obstacles of their insufficient migration into tumors and poor amplification and persistence, in addition to antigen-negative relapse and an immunosuppressive microenvironment. Various preclinical studies are exploring strategies to overcome the above challenges. Mobilization of endogenous immune cells is also necessary for CAR T cells to obtain their optimal therapeutic effect given the importance of the innate immune responses in the elimination of malignant tumors. In this review, we focus on the recent advances in the engineering of CAR T cell therapies to restore the immune response in solid malignancies, especially with CAR T cells acting as cellular carriers to deliver immunomodulators to tumors to mobilize the endogenous immune response. We also explored the sensitizing effects of conventional treatment approaches, such as chemotherapy and radiotherapy, on CAR T cell therapy. Finally, we discuss the combination of CAR T cells with biomaterials or oncolytic viruses to enhance the anti-tumor outcomes of CAR T cell therapies in solid tumors.
Collapse
Affiliation(s)
- Xiaohui Wang
- College of Biotechnology, Southwest University, Chongqing, 400715, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Stem Cell & Regenerative Medicine, Daping Hospital and Research Institute of Surgery, Chongqing, 400042, China
- Molecular & Immunological Department, Bio-therapeutic Department, Chinese PLA General Hospital, Beijing, 100853, China
| | - Zhiqiang Wu
- Molecular & Immunological Department, Bio-therapeutic Department, Chinese PLA General Hospital, Beijing, 100853, China
| | - Wei Qiu
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Stem Cell & Regenerative Medicine, Daping Hospital and Research Institute of Surgery, Chongqing, 400042, China
| | - Ping Chen
- College of Biotechnology, Southwest University, Chongqing, 400715, China
| | - Xiang Xu
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Stem Cell & Regenerative Medicine, Daping Hospital and Research Institute of Surgery, Chongqing, 400042, China.
| | - Weidong Han
- Molecular & Immunological Department, Bio-therapeutic Department, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
33
|
Ramírez-Otarola N, Maldonado K, Cavieres G, Bozinovic F, Sabat P. Nutritional ecology and ecological immunology in degus: Does early nutrition affect the postnatal development of the immune function? JOURNAL OF EXPERIMENTAL ZOOLOGY PART 2020; 335:239-249. [PMID: 33184965 DOI: 10.1002/jez.2429] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/19/2020] [Accepted: 10/29/2020] [Indexed: 11/10/2022]
Abstract
Environmental conditions experienced by developing animals have an impact on the development and maturity of the immune system. Specifically, the diet experienced during early development influences the maintenance and function of the immune system in young and adult animals. It is well known that exposure to low-protein diets during early development are related to an attenuation of immunocompetence in adulthood. While this functional linkage has been widely studied in altricial models' mammals, it has been little explored how the nutritional history modulates the immune function in precocial animals. We evaluated the effect of dietary protein consumed during early development on the immune function and the oxidative costs in the precocial Caviomorph rodent Octodon degus, or degu. We evaluated components of the acute phase response (APR) and oxidative parameters before and after immune challenge. We found that after the immune challenge, the juveniles on the low-protein dietary treatment exhibited an attenuation of body temperature but showed higher levels of lipid peroxidation than juvenile degus on the high-protein diet. We did not find a significant effect of the interaction between diet and immune challenge on body mass, levels of inflammatory proteins, nor in the total antioxidant capacity. Our results suggest that some components of the immune function and the oxidative status in the degu can be modulated by diet during development. However, the modulation would depend on the immune variables analyzed, and the characteristics of the immune system of precocial rodents.
Collapse
Affiliation(s)
- Natalia Ramírez-Otarola
- Escuela de Medicina Veterinaria, Facultad de Ciencias, Universidad Mayor, Huechuraba, Santiago, Chile.,Departamento de Ciencias Ecológicas, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Karin Maldonado
- Departamento de Ciencias Ecológicas, Facultad de Ciencias, Universidad de Chile, Santiago, Chile.,Departamento de Ciencias, Facultad de Artes Liberales, Universidad Adolfo Ibañez, Santiago, Chile
| | - Grisel Cavieres
- Center of Applied Ecology and Sustainability (CAPES), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Francisco Bozinovic
- Center of Applied Ecology and Sustainability (CAPES), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo Sabat
- Departamento de Ciencias Ecológicas, Facultad de Ciencias, Universidad de Chile, Santiago, Chile.,Center of Applied Ecology and Sustainability (CAPES), Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
34
|
Wang Y, Jiang M, Zhu J, Qu J, Qin K, Zhao D, Wang L, Dong L, Zhang X. The safety and efficacy of lenvatinib combined with immune checkpoint inhibitors therapy for advanced hepatocellular carcinoma. Biomed Pharmacother 2020; 132:110797. [PMID: 33068935 DOI: 10.1016/j.biopha.2020.110797] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/11/2020] [Accepted: 09/17/2020] [Indexed: 12/17/2022] Open
Abstract
China has one of the highest incidence rates of hepatocellular carcinoma (HCC) in the world. As most patients are diagnosed with advanced or unretractable HCC, systematic therapy is still the main treatment method for HCC. Currently, tyrosine kinase inhibitors (TKIs) and Immune checkpoint inhibitors (ICIs) are both the chief systematic therapy. And some studies have shown that the combination of TKIs and ICIs is more effective than monotherapy. The purpose of this review is to outline the rationale for the combination between lenvatinib and anti-PD-1(programmed cell death 1) and clinical trials to support this "golden combination". We also discuss the commonly treatment-emergent adverse events (AEs) and solutions for the patients with HCC who received the combination between lenvatinib and anti-PD-1 antibodies. Finally, we focus on the novel approaches, future perspectives and potential challenges about the combination of TKIs and ICIs.
Collapse
Affiliation(s)
- Yun Wang
- Hospital of Qingdao University, Qingdao, China
| | - Man Jiang
- Hospital of Qingdao University, Qingdao, China
| | | | - Jialin Qu
- Hospital of Qingdao University, Qingdao, China
| | - Kang Qin
- Hospital of Qingdao University, Qingdao, China
| | - Deze Zhao
- Hospital of Qingdao University, Qingdao, China
| | - Li Wang
- Hospital of Qingdao University, Qingdao, China
| | - Lina Dong
- Hospital of Qingdao University, Qingdao, China
| | | |
Collapse
|
35
|
Morsink MAJ, Willemen NGA, Leijten J, Bansal R, Shin SR. Immune Organs and Immune Cells on a Chip: An Overview of Biomedical Applications. MICROMACHINES 2020; 11:mi11090849. [PMID: 32932680 PMCID: PMC7570325 DOI: 10.3390/mi11090849] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 12/27/2022]
Abstract
Understanding the immune system is of great importance for the development of drugs and the design of medical implants. Traditionally, two-dimensional static cultures have been used to investigate the immune system in vitro, while animal models have been used to study the immune system’s function and behavior in vivo. However, these conventional models do not fully emulate the complexity of the human immune system or the human in vivo microenvironment. Consequently, many promising preclinical findings have not been reproduced in human clinical trials. Organ-on-a-chip platforms can provide a solution to bridge this gap by offering human micro-(patho)physiological systems in which the immune system can be studied. This review provides an overview of the existing immune-organs-on-a-chip platforms, with a special emphasis on interorgan communication. In addition, future challenges to develop a comprehensive immune system-on-chip model are discussed.
Collapse
Affiliation(s)
- Margaretha A. J. Morsink
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, MA 02139, USA; (M.A.J.M.); (N.G.A.W.)
- Department of Developmental BioEngineering, Faculty of Science and Technology, Technical Medical Centre, University of Twente, Drienerlolaan 5, 7522 NB Enschede, The Netherlands;
- Translational Liver Research, Department of Medical Cell BioPhysics, Technical Medical Centre, Faculty of Science and Technology, University of Twente Drienerlolaan 5, 7522 NB Enschede, The Netherlands;
| | - Niels G. A. Willemen
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, MA 02139, USA; (M.A.J.M.); (N.G.A.W.)
- Department of Developmental BioEngineering, Faculty of Science and Technology, Technical Medical Centre, University of Twente, Drienerlolaan 5, 7522 NB Enschede, The Netherlands;
| | - Jeroen Leijten
- Department of Developmental BioEngineering, Faculty of Science and Technology, Technical Medical Centre, University of Twente, Drienerlolaan 5, 7522 NB Enschede, The Netherlands;
| | - Ruchi Bansal
- Translational Liver Research, Department of Medical Cell BioPhysics, Technical Medical Centre, Faculty of Science and Technology, University of Twente Drienerlolaan 5, 7522 NB Enschede, The Netherlands;
| | - Su Ryon Shin
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, MA 02139, USA; (M.A.J.M.); (N.G.A.W.)
- Correspondence: ; Tel.: +1-617-768-8320
| |
Collapse
|
36
|
Li C, Lam E, Perez-Shibayama C, Ward LA, Zhang J, Lee D, Nguyen A, Ahmed M, Brownlie E, Korneev KV, Rojas O, Sun T, Navarre W, He HH, Liao S, Martin A, Ludewig B, Gommerman JL. Early-life programming of mesenteric lymph node stromal cell identity by the lymphotoxin pathway regulates adult mucosal immunity. Sci Immunol 2020; 4:4/42/eaax1027. [PMID: 31862865 DOI: 10.1126/sciimmunol.aax1027] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 11/19/2019] [Indexed: 12/14/2022]
Abstract
Redundant mechanisms support immunoglobulin A (IgA) responses to intestinal antigens. These include multiple priming sites [mesenteric lymph nodes (MLNs), Peyer's patches, and isolated lymphoid follicles] and various cytokines that promote class switch to IgA, even in the absence of T cells. Despite these backup mechanisms, vaccination against enteric pathogens such as rotavirus has limited success in some populations. Genetic and environmental signals experienced during early life are known to influence mucosal immunity, yet the mechanisms for how these exposures operate remain unclear. Here, we used rotavirus infection to follow antigen-specific IgA responses through time and in different gut compartments. Using genetic and pharmacological approaches, we tested the role of the lymphotoxin (LT) pathway-known to support IgA responses-at different developmental stages. We found that LT-β receptor (LTβR) signaling in early life programs intestinal IgA responses in adulthood by affecting antibody class switch recombination to IgA and subsequent generation of IgA antibody-secreting cells within an intact MLN. In addition, early-life LTβR signaling dictates the phenotype and function of MLN stromal cells to support IgA responses in the adult. Collectively, our studies uncover new mechanistic insights into how early-life LTβR signaling affects mucosal immune responses during adulthood.
Collapse
Affiliation(s)
- Conglei Li
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Evelyn Lam
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | | | - Lesley A Ward
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Jianbo Zhang
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Dennis Lee
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Albert Nguyen
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Musaddeque Ahmed
- Department of Medical Biophysics, University of Toronto, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Emma Brownlie
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Kirill V Korneev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences and Department of Immunology, Biological Faculty, Lomonosov Moscow State University, Moscow, Russia
| | - Olga Rojas
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Tian Sun
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - William Navarre
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Housheng Hansen He
- Department of Medical Biophysics, University of Toronto, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Shan Liao
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Alberto Martin
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | | |
Collapse
|
37
|
Gracia G, Cao E, Feeney OM, Johnston APR, Porter CJH, Trevaskis NL. High-Density Lipoprotein Composition Influences Lymphatic Transport after Subcutaneous Administration. Mol Pharm 2020; 17:2938-2951. [DOI: 10.1021/acs.molpharmaceut.0c00348] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Gracia Gracia
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne 3052, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne 3052, Australia
| | - Enyuan Cao
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne 3052, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne 3052, Australia
| | - Orlagh M. Feeney
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne 3052, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne 3052, Australia
| | - Angus P. R. Johnston
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne 3052, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne 3052, Australia
| | - Christopher J. H. Porter
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne 3052, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne 3052, Australia
| | - Natalie L. Trevaskis
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne 3052, Australia
| |
Collapse
|
38
|
Sahputra R, Ruckerl D, Couper KN, Muller W, Else KJ. The Essential Role Played by B Cells in Supporting Protective Immunity Against Trichuris muris Infection Is by Controlling the Th1/Th2 Balance in the Mesenteric Lymph Nodes and Depends on Host Genetic Background. Front Immunol 2019; 10:2842. [PMID: 31921120 PMCID: PMC6915098 DOI: 10.3389/fimmu.2019.02842] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 11/19/2019] [Indexed: 12/16/2022] Open
Abstract
How B cells contribute to protective immunity against parasitic nematodes remains unclear, with their importance as accessory cells underexplored. In this study, anti-CD20 monoclonal antibody (α-CD20 mAb)-mediated depletion of B cells from C57BL/6 mice revealed an important role for B cells in supporting Th2 immune responses and thus expulsion of Trichuris muris (T. muris). C57BL/6 mice normally mount mixed Th1/Th2 immune responses to T. muris and expel the parasite by the third week post infection. However, B cell-depleted C57BL/6 had significantly reduced Th2-type cytokines post infection and failed to expel the parasite. IFN-γ production in the MLN of C57BL/6 mice receiving α-CD20 mAb treatment was not affected, collectively resulting in an overall change in Th1/Th2 balance in favor of Th1. Further, the expression of IFN-γ and IFN-γ-induced genes at the effector site, the gut, was significantly increased in the absence of B cells. Interestingly, and in complete contrast, BALB/c mice, which mount strongly polarized Th2 immune responses, rather than mixed Th1/Th2 immune responses, were still able to expel T. muris in the absence of B cells. We thus hypothesized that the B cell plays a critical role in enabling strong Th2 responses in the context of mixed Th1/Th2 settings, with the role becoming redundant in highly Th2 polarized environments. In support of this, neutralization of IFN-γ in B cell depleted C57BL/6 restored resistance against T. muris infection. Thus, our data suggest an important role of B cells in supporting Th2-type immune responses in mixed IFN-γ-rich Th1/Th2 settings.
Collapse
Affiliation(s)
- Rinal Sahputra
- Division of Infection, Immunity and Respiratory Medicine, Lydia Becker Institute for Immunology, The University of Manchester, Manchester, United Kingdom
| | | | | | | | - Kathryn J. Else
- Division of Infection, Immunity and Respiratory Medicine, Lydia Becker Institute for Immunology, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
39
|
Norouzian M, Mehdipour F, Balouchi Anaraki S, Ashraf MJ, Khademi B, Ghaderi A. Atypical Memory and Regulatory B Cell Subsets in Tumor Draining Lymph Nodes of Head and Neck Squamous Cell Carcinoma Correlate with Good Prognostic Factors. Head Neck Pathol 2019; 14:645-656. [PMID: 31691165 PMCID: PMC7413970 DOI: 10.1007/s12105-019-01095-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Accepted: 10/30/2019] [Indexed: 12/29/2022]
Abstract
Research on the role of B cells in the development and modulation of antitumor immunity has increased in recent years; however, knowledge about B cell phenotype and function in tumor draining lymph nodes (TDLNs) is still incomplete. This study aimed to investigate changes in the phenotypic profile of B cells in TDLNs of head and neck squamous cell carcinoma (HNSCC) during disease progression. Mononuclear cells were isolated from TDLNs and stained with antibodies for CD19 and other B cell-related markers and analyzed by flow cytometry. CD19+ B cells comprised 38.6 ± 8.9% of lymphocytes in TDLNs of HNSCC. Comparison of metastatic and non-metastatic LNs disclosed no significant differences in the frequencies of B cell subsets including antigen-experienced, naïve, switched, unswitched, atypical memory, marginal zone-like B cells, and B cells with regulatory phenotypes. The percentage of atypical memory (CD27-IgM-IgD-) B cells was significantly higher in patients with tongue SCC with no involved LNs (p = 0.033) and correlated inversely with the number of involved LNs. The frequency of CD24hiCD38hi B cells was significantly higher in non-metastatic LNs of patients with grade I compared to grade II (p = 0.016), and the percentage of CD5+ B cells decreased as tumors progressed from stage III to IV (p = 0.008). Our data show that in TDLNs of HNSCC, the frequency of B cells with atypical memory and regulatory phenotypes was significantly associated with good prognostic factors; however, their function remains to be investigated.
Collapse
Affiliation(s)
- Marzieh Norouzian
- grid.412571.40000 0000 8819 4698Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fereshteh Mehdipour
- grid.412571.40000 0000 8819 4698Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, P.O. Box: 71345-3119, Shiraz, Iran
| | - Sima Balouchi Anaraki
- grid.412571.40000 0000 8819 4698Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Javad Ashraf
- grid.412571.40000 0000 8819 4698Department of Oral Pathology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bijan Khademi
- grid.412571.40000 0000 8819 4698Department of Otolaryngology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abbas Ghaderi
- grid.412571.40000 0000 8819 4698Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran ,grid.412571.40000 0000 8819 4698Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, P.O. Box: 71345-3119, Shiraz, Iran
| |
Collapse
|
40
|
Friedrich SK, Lang PA, Friebus-Kardash J, Duhan V, Bezgovsek J, Lang KS. Mechanisms of lymphatic system-specific viral replication and its potential role in autoimmune disease. Clin Exp Immunol 2019; 195:64-73. [PMID: 30444956 DOI: 10.1111/cei.13241] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/13/2018] [Indexed: 12/15/2022] Open
Abstract
Viral infections can be fatal because of the direct cytopathic effects of the virus or the induction of a strong, uncontrolled inflammatory response. Virus and host intrinsic characteristics strongly modulate the outcome of viral infections. Recently we determined the circumstances under which enhanced replication of virus within the lymphoid tissue is beneficial for the outcome of a disease. This enforced viral replication promotes anti-viral immune activation and, counterintuitively, accelerates virus control. In this review we summarize the mechanisms that contribute to enforced viral replication. Antigen-presenting cells and CD169+ macrophages exhibit enforced viral replication after infection with the model viruses lymphocytic choriomeningitis virus (LCMV) and vesicular stomatitis virus (VSV). Ubiquitin-specific peptidase 18 (Usp18), an endogenous type I interferon blocker in CD169+ macrophages, has been identified as a proviral gene, as are B cell activating factor (BAFF) and carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1). Lymphotoxins (LT) strongly enhance viral replication in the spleen and lymph nodes. All these factors modulate splenic architecture and thereby promote the development of CD169+ macrophages. Tumor necrosis factor alpha (TNF-α) and nuclear factor kappa-light-chain-enhancer of activated B cell signaling (NF-κB) have been found to promote the survival of infected CD169+ macrophages, thereby similarly promoting enforced viral replication. Association of autoimmune disease with infections is evident from (1) autoimmune phenomena described during a chronic virus infection; (2) onset of autoimmune disease simultaneous to viral infections; and (3) experimental evidence. Involvement of virus infection during onset of type I diabetes is strongly evident. Epstein-Bar virus (EBV) infection was discussed to be involved in the pathogenesis of systemic lupus erythematosus. In conclusion, several mechanisms promote viral replication in secondary lymphatic organs. Identifying such factors in humans is a challenge for future studies.
Collapse
Affiliation(s)
- S-K Friedrich
- University of Duisburg-Essen, Institute of Immunology, Medical Faculty, Essen, Germany
| | - P A Lang
- Heinrich-Heine-University, Insitute of Molecular Medicine II, Düsseldorf, Germany
| | - J Friebus-Kardash
- University of Duisburg-Essen, Institute of Immunology, Medical Faculty, Essen, Germany
| | - V Duhan
- University of Duisburg-Essen, Institute of Immunology, Medical Faculty, Essen, Germany
| | - J Bezgovsek
- University of Duisburg-Essen, Institute of Immunology, Medical Faculty, Essen, Germany
| | - K S Lang
- University of Duisburg-Essen, Institute of Immunology, Medical Faculty, Essen, Germany
| |
Collapse
|
41
|
Gonzalez Badillo FE, Zisi Tegou F, Abreu MM, Masina R, Sha D, Najjar M, Wright SH, Bayer AL, Korpos É, Pugliese A, Molano RD, Tomei AA. CCL21 Expression in β-Cells Induces Antigen-Expressing Stromal Cell Networks in the Pancreas and Prevents Autoimmune Diabetes in Mice. Diabetes 2019; 68:1990-2003. [PMID: 31371518 PMCID: PMC6754241 DOI: 10.2337/db19-0239] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 07/21/2019] [Indexed: 12/31/2022]
Abstract
Tumors induce tolerance toward their antigens by producing the chemokine CCL21, leading to the formation of tertiary lymphoid organs (TLOs). Ins2-CCL21 transgenic, nonobese diabetic (NOD) mice express CCL21 in pancreatic β-cells and do not develop autoimmune diabetes. We investigated by which mechanisms CCL21 expression prevented diabetes. Ins2-CCL21 mice develop TLOs by 4 weeks of age, consisting of naive CD4+ T cells compartmentalized within networks of CD45-gp38+CD31- fibroblastic reticular cell (FRC)-like cells. Importantly, 12-week-old Ins2-CCL21 TLOs contained FRC-like cells with higher contractility, regulatory, and anti-inflammatory properties and enhanced expression of β-cell autoantigens compared with nontransgenic NOD TLOs found in inflamed islets. Consistently, transgenic mice harbored fewer autoreactive T cells and a higher proportion of regulatory T cells in the islets. Using adoptive transfer and islet transplantation models, we demonstrate that TLO formation in Ins2-CCL21 transgenic islets is critical for the regulation of autoimmunity, and although the effect is systemic, the induction is mediated locally likely by lymphocyte trafficking through TLOs. Overall, our findings suggest that CCL21 promotes TLOs that differ from inflammatory TLOs found in type 1 diabetic islets in that they resemble lymph nodes, contain FRC-like cells expressing β-cell autoantigens, and are able to induce systemic and antigen-specific tolerance leading to diabetes prevention.
Collapse
Affiliation(s)
- Freddy E Gonzalez Badillo
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
- Department of Biomedical Engineering, University of Miami, Miami, FL
| | - Flavia Zisi Tegou
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
- Department of Biomedical Engineering, University of Miami, Miami, FL
| | - Maria M Abreu
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Riccardo Masina
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Divya Sha
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Mejdi Najjar
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Shane H Wright
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Allison L Bayer
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL
| | - Éva Korpos
- Institute of Physiological Chemistry and Pathobiochemistry and Cells in Motion, Cluster of Excellence, University of Muenster, Muenster, Germany
| | - Alberto Pugliese
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL
| | - R Damaris Molano
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Alice A Tomei
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
- Department of Biomedical Engineering, University of Miami, Miami, FL
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL
| |
Collapse
|
42
|
Platt JL, Cascalho M. Non-canonical B cell functions in transplantation. Hum Immunol 2019; 80:363-377. [PMID: 30980861 PMCID: PMC6544480 DOI: 10.1016/j.humimm.2019.04.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 04/04/2019] [Accepted: 04/09/2019] [Indexed: 12/18/2022]
Abstract
B cells are differentiated to recognize antigen and respond by producing antibodies. These activities, governed by recognition of ancillary signals, defend the individual against microorganisms and the products of microorganisms and constitute the canonical function of B cells. Despite the unique differentiation (e.g. recombination and mutation of immunoglobulin gene segments) toward this canonical function, B cells can provide other, "non-canonical" functions, such as facilitating of lymphoid organogenesis and remodeling and fashioning T cell repertoires and modifying T cell responses. Some non-canonical functions are exerted by antibodies, but most are mediated by other products and/or direct actions of B cells. The diverse set of non-canonical functions makes the B cell as much as any cell a central organizer of innate and adaptive immunity. However, the diverse products and actions also confound efforts to weigh the importance of individual non-canonical B cell functions. Here we shall describe the non-canonical functions of B cells and offer our perspective on how those functions converge in the development and governance of immunity, particularly immunity to transplants, and hurdles to advancing understanding of B cell functions in transplantation.
Collapse
Affiliation(s)
- Jeffrey L Platt
- Departments of Surgery and of Microbiology & Immunology, University of Michigan, Ann Arbor, MI, United States.
| | - Marilia Cascalho
- Departments of Surgery and of Microbiology & Immunology, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
43
|
Wang Z, Wang W, Chai Q, Zhu M. Langerhans Cells Control Lymphatic Vessel Function during Inflammation via LIGHT-LTβR Signaling. THE JOURNAL OF IMMUNOLOGY 2019; 202:2999-3007. [PMID: 30952816 DOI: 10.4049/jimmunol.1801578] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 03/12/2019] [Indexed: 01/03/2023]
Abstract
The lymphatic vasculature is an important route for dendritic cell (DC) or tumor cell migration from peripheral tissues to draining lymph nodes (DLNs). However, the underlying molecular and cellular mechanisms remain poorly understood. In this study, using conventional bone marrow chimeric mice and additional UVB radiation, we found that deficiency of LIGHT but not lymphotoxin (LT) α1β2, likely on radioresistant Langerhans cells (LCs), resulted in impaired skin DC migration to DLNs during LPS-induced inflammation. In addition, LT β receptor (LTβR), but not herpes virus entry mediator, was found to be the receptor of LIGHT controlling DC migration. Furthermore, conditional deficiency of LTβR in Tie2 cre or Lyve1 cre mice, but not in LTβR-deficient bone marrow chimeric mice, impaired DC migration, suggesting an important role of LTβR in radioresistant lymphatic endothelial cells (LECs), although the role of LTβR in blood endothelial cells remains intriguing. Mechanistically, the gene expression of both CCL21 and CCL19 was found to be reduced in skin LECs isolated from LC-LIGHT-conditionally deficient or Lyve1 cre Ltbr fl/fl mice compared with their controls upon LPS stimulation. Soluble recombinant LIGHT was able to upregulate CCL21 and CCL19 gene expression on SVEC4-10 endothelial cells. Doxycycline, an inhibitor of soluble LIGHT release in the inflamed skin, impaired skin CCL21 and CCL19 expression and DC migration. In addition, melanoma cell metastasis to DLNs was also inhibited in LC-LIGHT-conditionally deficient or Lyve1 cre Ltbr fl/fl mice. Together, our data suggest, to our knowledge, a previously unrecognized scenario in which LCs activate LECs via the LIGHT-LTβR signaling axis to promote DC migration or tumor cell metastasis.
Collapse
Affiliation(s)
- Zhongnan Wang
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; and.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenjun Wang
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; and.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qian Chai
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; and
| | - Mingzhao Zhu
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; and .,College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
44
|
Dostert C, Grusdat M, Letellier E, Brenner D. The TNF Family of Ligands and Receptors: Communication Modules in the Immune System and Beyond. Physiol Rev 2019; 99:115-160. [DOI: 10.1152/physrev.00045.2017] [Citation(s) in RCA: 175] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The tumor necrosis factor (TNF) and TNF receptor (TNFR) superfamilies (TNFSF/TNFRSF) include 19 ligands and 29 receptors that play important roles in the modulation of cellular functions. The communication pathways mediated by TNFSF/TNFRSF are essential for numerous developmental, homeostatic, and stimulus-responsive processes in vivo. TNFSF/TNFRSF members regulate cellular differentiation, survival, and programmed death, but their most critical functions pertain to the immune system. Both innate and adaptive immune cells are controlled by TNFSF/TNFRSF members in a manner that is crucial for the coordination of various mechanisms driving either co-stimulation or co-inhibition of the immune response. Dysregulation of these same signaling pathways has been implicated in inflammatory and autoimmune diseases, highlighting the importance of their tight regulation. Investigation of the control of TNFSF/TNFRSF activities has led to the development of therapeutics with the potential to reduce chronic inflammation or promote anti-tumor immunity. The study of TNFSF/TNFRSF proteins has exploded over the last 30 yr, but there remains a need to better understand the fundamental mechanisms underlying the molecular pathways they mediate to design more effective anti-inflammatory and anti-cancer therapies.
Collapse
Affiliation(s)
- Catherine Dostert
- Department of Infection and Immunity, Experimental and Molecular Immunology, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark; and Life Sciences Research Unit, Molecular Disease Mechanisms Group, University of Luxembourg, Belvaux, Luxembourg
| | - Melanie Grusdat
- Department of Infection and Immunity, Experimental and Molecular Immunology, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark; and Life Sciences Research Unit, Molecular Disease Mechanisms Group, University of Luxembourg, Belvaux, Luxembourg
| | - Elisabeth Letellier
- Department of Infection and Immunity, Experimental and Molecular Immunology, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark; and Life Sciences Research Unit, Molecular Disease Mechanisms Group, University of Luxembourg, Belvaux, Luxembourg
| | - Dirk Brenner
- Department of Infection and Immunity, Experimental and Molecular Immunology, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark; and Life Sciences Research Unit, Molecular Disease Mechanisms Group, University of Luxembourg, Belvaux, Luxembourg
| |
Collapse
|
45
|
|
46
|
Yang K, Liang Y, Sun Z, Liu L, Liao J, Xu H, Zhu M, Fu YX, Peng H. T cell-derived lymphotoxin limits Th1 response during HSV-1 infection. Sci Rep 2018; 8:17727. [PMID: 30531962 PMCID: PMC6286317 DOI: 10.1038/s41598-018-36012-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 10/30/2018] [Indexed: 11/09/2022] Open
Abstract
Though lymphotoxin (LT) is highly expressed by type I helper T (Th1) cells, its contribution to CD4+ T cell differentiation during infections and diseases remains a mystery. In HSV-1 infection, we observed that LTβR signaling is required to limit the Th1 response. Using bone marrow chimeric mice, mixed-T-cell chimeric mice, and LTβR in vivo blockades, we unexpectedly observed that LT, especially T cell-derived LT, played an indispensable role in limiting the Th1 response. The LTβR-Ig blockade promoted the Th1 response by increasing infiltration of monocytes and monocyte-derived DCs and up-regulating IL-12 secretion in the lymphoid environment. Our findings identified a novel role for T cell-derived LT in manipulating Th1 differentiation.
Collapse
Affiliation(s)
- Kaiting Yang
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yong Liang
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zhichen Sun
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Longchao Liu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Jing Liao
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Hairong Xu
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Mingzhao Zhu
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yang-Xin Fu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| | - Hua Peng
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
47
|
Hermida MDR, de Melo CVB, Lima IDS, Oliveira GGDS, Dos-Santos WLC. Histological Disorganization of Spleen Compartments and Severe Visceral Leishmaniasis. Front Cell Infect Microbiol 2018; 8:394. [PMID: 30483481 PMCID: PMC6243053 DOI: 10.3389/fcimb.2018.00394] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 10/19/2018] [Indexed: 01/10/2023] Open
Abstract
The spleen is a secondary lymphoid organ responsible for immune surveillance against blood-circulating pathogens. Absence of the spleen is associated with increased susceptibility to systemic spread and fatal infection by different pathogens. Severe forms of visceral leishmaniasis are associated with disorganization of spleen compartments where cell interactions essential for splenic immunological function take place. White pulp atrophies, secondary lymphoid follicles and marginal zones vanish, and the boundaries separating white and red pulp blur. Leukocyte populations are reduced or disappear or are replaced by plasma cells. In this paper, we review the published data on spleen disorganization in severe forms of visceral leishmaniasis and propose a histological classification to help the exchange of information among research groups.
Collapse
|
48
|
Alsufyani F, Mattoo H, Zhou D, Cariappa A, Van Buren D, Hock H, Avruch J, Pillai S. The Mst1 Kinase Is Required for Follicular B Cell Homing and B-1 B Cell Development. Front Immunol 2018; 9:2393. [PMID: 30386341 PMCID: PMC6199389 DOI: 10.3389/fimmu.2018.02393] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 09/27/2018] [Indexed: 11/22/2022] Open
Abstract
The Mst1 and 2 cytosolic serine/threonine protein kinases are the mammalian orthologs of the Drosophila Hippo protein. Mst1 has been shown previously to participate in T-cell and B-cell trafficking and the migration of lymphocytes into secondary lymphoid organs in a cell intrinsic manner. We show here that the absence of Mst1 alone only modestly impacts B cell homing to lymph nodes. The absence of both Mst1 and 2 in hematopoietic cells results in relatively normal B cell development in the bone marrow and does not impact migration of immature B cells to the spleen. However, follicular B cells lacking both Mst1 and Mst2 mature in the splenic white pulp but are unable to recirculate to lymph nodes or to the bone marrow. These cells also cannot traffic efficiently to the splenic red pulp. The inability of late transitional and follicular B cells lacking Mst 1 and 2 to migrate to the red pulp explains their failure to differentiate into marginal zone B cell precursors and marginal zone B cells. Mst1 and Mst2 are therefore required for follicular B cells to acquire the ability to recirculate and also to migrate to the splenic red pulp in order to generate marginal zone B cells. In addition B-1 a B cell development is defective in the absence of Mst1.
Collapse
Affiliation(s)
- Faisal Alsufyani
- Ragon Institute of MGH, MIT and Harvard, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Hamid Mattoo
- Ragon Institute of MGH, MIT and Harvard, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Dawang Zhou
- Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Annaiah Cariappa
- Ragon Institute of MGH, MIT and Harvard, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Denille Van Buren
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Hanno Hock
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Joseph Avruch
- Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Shiv Pillai
- Ragon Institute of MGH, MIT and Harvard, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
49
|
Yang K, Liang Y, Sun Z, Xue D, Xu H, Zhu M, Fu YX, Peng H. T Cell-Derived Lymphotoxin Is Essential for the Anti-Herpes Simplex Virus 1 Humoral Immune Response. J Virol 2018; 92:e00428-18. [PMID: 29743364 PMCID: PMC6026738 DOI: 10.1128/jvi.00428-18] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 05/03/2018] [Indexed: 11/20/2022] Open
Abstract
B cell-derived lymphotoxin (LT) is required for the development of follicular dendritic cell clusters for the formation of primary and secondary lymphoid follicles, but the role of T cell-derived LT in antibody response has not been well demonstrated. We observed that lymphotoxin β-receptor (LTβR) signaling is essential for optimal humoral immune response and protection against an acute herpes simplex virus 1 (HSV-1) infection. Blocking the LTβR pathway caused poor maintenance of germinal center B (GC-B) cells and follicular helper T (Tfh) cells. Using bone marrow chimeric mice and adoptive transplantation, we determined that T cell-derived LT played an indispensable role in the humoral immune response to HSV-1. Upregulation of gamma interferon by the LTβR-Ig blockade impairs the sustainability of Tfh-like cells, leading to an impaired humoral immune response. Our findings have identified a novel role of T cell-derived LT in the humoral immune response against HSV-1 infection.IMPORTANCE Immunocompromised people are susceptible to HSV-1 infection and lethal recurrence, which could be inhibited by anti-HSV-1 humoral immune response in the host. This study sought to explore the role of T cell-derived LT in the anti-HSV-1 humoral immune response using LT-LTβR signaling-deficient mice and the LTβR-Ig blockade. The data indicate that the T cell-derived LT may play an essential role in sustaining Tfh-like cells and ensure Tfh-like cells' migration into primary or secondary follicles for further maturation. This study provides insights for vaccine development against infectious diseases.
Collapse
Affiliation(s)
- Kaiting Yang
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yong Liang
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Zhichen Sun
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Diyuan Xue
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Hairong Xu
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Mingzhao Zhu
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Yang-Xin Fu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Hua Peng
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
50
|
Zhang Y, Kim TJ, Wroblewska JA, Tesic V, Upadhyay V, Weichselbaum RR, Tumanov AV, Tang H, Guo X, Tang H, Fu YX. Type 3 innate lymphoid cell-derived lymphotoxin prevents microbiota-dependent inflammation. Cell Mol Immunol 2018; 15:697-709. [PMID: 28579615 PMCID: PMC6123485 DOI: 10.1038/cmi.2017.25] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Revised: 03/15/2017] [Accepted: 03/16/2017] [Indexed: 12/25/2022] Open
Abstract
Splenomegaly is a well-known phenomenon typically associated with inflammation. However, the underlying cause of this phenotype has not been well characterized. Furthermore, the splenomegaly phenotype seen in lymphotoxin (LT) signaling-deficient mice is characterized by increased numbers of splenocytes and splenic neutrophils. Splenomegaly, as well as the related phenotype of increased lymphocyte counts in non-lymphoid tissues, is thought to result from the absence of secondary lymphoid tissues in LT-deficient mice. We now present evidence that mice deficient in LTα1β2 or LTβR develop splenomegaly and increased numbers of lymphocytes in non-lymphoid tissues in a microbiota-dependent manner. Antibiotic administration to LTα1β2- or LTβR-deficient mice reduces splenomegaly. Furthermore, re-derived germ-free Ltbr-/- mice do not exhibit splenomegaly or increased inflammation in non-lymphoid tissues compared to specific pathogen-free Ltbr-/- mice. By using various LTβ- and LTβR-conditional knockout mice, we demonstrate that retinoic acid-related orphan receptor γT-positive type 3 innate lymphoid cells provide the required active LT signaling to prevent the development of splenomegaly. Thus, this study demonstrates the importance of LT-mediated immune responses for the prevention of splenomegaly and systemic inflammation induced by microbiota.
Collapse
MESH Headings
- Animals
- Immunity, Innate
- Inflammation/genetics
- Inflammation/immunology
- Inflammation/pathology
- Lymphocytes/immunology
- Lymphocytes/pathology
- Lymphotoxin alpha1, beta2 Heterotrimer/genetics
- Lymphotoxin alpha1, beta2 Heterotrimer/immunology
- Lymphotoxin beta Receptor/genetics
- Lymphotoxin beta Receptor/immunology
- Mice
- Mice, Knockout
- Microbiota/immunology
- Receptors, Antigen, T-Cell, gamma-delta/genetics
- Receptors, Antigen, T-Cell, gamma-delta/immunology
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Pathology, University of Chicago, 60637, Chicago, USA, IL
| | - Tae-Jin Kim
- Department of Pathology, University of Chicago, 60637, Chicago, USA, IL
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, 02841, Seoul, Korea
| | - Joanna A Wroblewska
- Committee on Immunology, Department of Pathology, University of Chicago, 60637, Chicago, IL, USA
| | - Vera Tesic
- Department of Pathology, University of Chicago, 60637, Chicago, USA, IL
| | - Vaibhav Upadhyay
- Committee on Immunology, Department of Pathology, University of Chicago, 60637, Chicago, IL, USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology and The Ludwig Center for Metastasis Research, University of Chicago, 60637, Chicago, IL, USA
| | - Alexei V Tumanov
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center San Antonio, 78229, San Antonio, TX, USA
| | - Hong Tang
- Chinese Academy of Sciences Key Laboratory for Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, 100101, Beijing, China
| | - Xiaohuan Guo
- Institute of Immunology, Tsinghua University School of Medicine, 100084, Beijing, China
| | - Haidong Tang
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Yang-Xin Fu
- Department of Pathology, University of Chicago, 60637, Chicago, USA, IL.
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|