1
|
Wholey WY, Meyer AR, Yoda ST, Chackerian B, Zikherman J, Cheng W. Minimal determinants for lifelong antiviral antibody responses in BALB/c mice from a single exposure to virus-like immunogens at low doses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.02.20.529089. [PMID: 36865112 PMCID: PMC9979986 DOI: 10.1101/2023.02.20.529089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
However, due to the complex compositions of natural virions, the molecular determinants of Ab durability from viral infection or inactivated viral vaccines have been incompletely understood. Here we used a reductionist system of liposome-based virus-like structures to examine the durability of Abs in primary immune responses in mice. This system allowed us to independently vary fundamental viral attributes and to do so without additional adjuvants to model natural viruses. We show that a single injection of antigens (Ags) orderly displayed on a virion-sized liposome is sufficient to induce a long-lived neutralizing Ab (nAb) response. Introduction of internal nucleic acids dramatically modulates the magnitude of long-term Ab responses without alteration of the long-term kinetic trends. These Abs are characterized by exceptionally slow off-rates of ~0.0005 s-1, which emerged as early as day 5 after injection and these off-rates are comparable to that of affinity-matured monoclonal Abs. A single injection of these structures at doses as low as 100 ng led to lifelong nAb production in BALB/c mice. Thus, a minimal virus-like immunogen can give rise to potent and long-lasting antiviral Abs in a primary response in mice without live infection. This has important implications for understanding both live viral infection and for optimized vaccine design.
Collapse
|
2
|
Bachmann MF, Vogel M, Speiser DE. Successful Allergen-Specific Immunotherapy: Induction of Unresponsiveness by 'Vaccination'. Vaccines (Basel) 2023; 11:1852. [PMID: 38140255 PMCID: PMC10748047 DOI: 10.3390/vaccines11121852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
The mechanisms of action of allergen-specific immunotherapy (AIT) are often referred to as the induction of 'tolerance'. However, immunological 'tolerance' is defined as an alteration in the function or composition of immune cells. For AIT, this is not always the case, because it can also induce allergen-specific IgG antibodies that block allergic responses. To include all possible mechanisms that may mediate successful AIT, it is advantageous to use the scientific term 'unresponsiveness' instead of 'tolerance'. In praxis, the term 'vaccination' is also appropriate, as AIT medications are specialized vaccines.
Collapse
Affiliation(s)
- Martin F. Bachmann
- Department of Biomedical Research (DBMR), University of Bern, 3008 Bern, Switzerland; (M.V.); (D.E.S.)
- Department of Rheumatology and Immunology, University Hospital of Bern, 3010 Bern, Switzerland
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford OX1 2JD, UK
| | - Monique Vogel
- Department of Biomedical Research (DBMR), University of Bern, 3008 Bern, Switzerland; (M.V.); (D.E.S.)
- Department of Rheumatology and Immunology, University Hospital of Bern, 3010 Bern, Switzerland
| | - Daniel E. Speiser
- Department of Biomedical Research (DBMR), University of Bern, 3008 Bern, Switzerland; (M.V.); (D.E.S.)
- Department of Rheumatology and Immunology, University Hospital of Bern, 3010 Bern, Switzerland
- Department of Oncology, Lausanne University Hospital and University of Lausanne, 1066 Lausanne, Switzerland
| |
Collapse
|
3
|
Gaspar-Castillo C, Rodríguez MH, Ortiz-Navarrete V, Alpuche-Aranda CM, Martinez-Barnetche J. Structural and immunological basis of cross-reactivity between dengue and Zika infections: Implications in serosurveillance in endemic regions. Front Microbiol 2023; 14:1107496. [PMID: 37007463 PMCID: PMC10063793 DOI: 10.3389/fmicb.2023.1107496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 02/24/2023] [Indexed: 03/19/2023] Open
Abstract
Dengue and Zika are arthropod-borne viral diseases present in more than 100 countries around the world. In the past decade, Zika emerged causing widespread outbreaks in new regions, where dengue has been endemic-epidemic for a long period. The wide and extensive dissemination of the mosquito vectors, Aedes aegypti, and Ae. albopictus, favor the co-existence of both infections in the same regions. Together with an important proportion of asymptomatic infections, similar clinical manifestations, and a short time window for acute infection confirmatory tests, it is difficult to differentially estimate both dengue and Zika incidence and prevalence. DENV and ZIKV flavivirus share high structural similarity, inducing a cross-reactive immune response that leads to false positives in serological tests particularly in secondary infections. This results in overestimation of recent Zika outbreaks seroprevalence in dengue endemic regions. In this review, we address the biological basis underlying DENV and ZIKV structural homology; the structural and cellular basis of immunological cross reactivity; and the resulting difficulties in measuring dengue and Zika seroprevalence. Finally, we offer a perspective about the need for more research to improve serological tests performance.
Collapse
Affiliation(s)
- Carlos Gaspar-Castillo
- Center for Infectious Diseases Research, National Institute of Public Health, Cuernavaca, Mexico
| | - Mario H. Rodríguez
- Center for Infectious Diseases Research, National Institute of Public Health, Cuernavaca, Mexico
| | - Vianney Ortiz-Navarrete
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute, Mexico City, Mexico
| | - Celia M. Alpuche-Aranda
- Center for Infectious Diseases Research, National Institute of Public Health, Cuernavaca, Mexico
- Celia M. Alpuche-Aranda,
| | - Jesus Martinez-Barnetche
- Center for Infectious Diseases Research, National Institute of Public Health, Cuernavaca, Mexico
- *Correspondence: Jesus Martinez-Barnetche,
| |
Collapse
|
4
|
Martinez-Barnetche J, Godoy-Lozano EE, Saint Remy-Hernández S, Pacheco-Olvera DL, Téllez-Sosa J, Valdovinos-Torres H, Pastelin-Palacios R, Mena H, Zambrano L, López-Macías C. Characterization of immunoglobulin loci in the gigantic genome of Ambystoma mexicanum. Front Immunol 2023; 14:1039274. [PMID: 36776846 PMCID: PMC9911811 DOI: 10.3389/fimmu.2023.1039274] [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: 09/08/2022] [Accepted: 01/06/2023] [Indexed: 01/28/2023] Open
Abstract
Background The axolotl, Ambystoma mexicanum is a unique biological model for complete tissue regeneration. Is a neotenic endangered species and is highly susceptible to environmental stress, including infectious disease. In contrast to other amphibians, the axolotl is particularly vulnerable to certain viral infections. Like other salamanders, the axolotl genome is one of the largest (32 Gb) and the impact of genome size on Ig loci architecture is unknown. To better understand the immune response in axolotl, we aimed to characterize the immunoglobulin loci of A. mexicanum and compare it with other model vertebrates. Methods The most recently published genome sequence of A. mexicanum (V6) was used for alignment-based annotation and manual curation using previously described axolotl Ig sequences or reference sequences from other vertebrates. Gene models were further curated using A. mexicanum spleen RNA-seq data. Human, Xenopus tropicalis, Danio rerio (zebrafish), and eight tetrapod reference genomes were used for comparison. Results Canonical A. mexicanum heavy chain (IGH), lambda (IGL), sigma (IGS), and the putative surrogate light chain (SLC) loci were identified. No kappa locus was found. More than half of the IGHV genes and the IGHF gene are pseudogenes and there is no clan I IGHV genes. Although the IGH locus size is proportional to genome size, we found local size restriction in the IGHM gene and the V gene intergenic distances. In addition, there were V genes with abnormally large V-intron sizes, which correlated with loss of gene functionality. Conclusion The A. mexicanum immunoglobulin loci share the same general genome architecture as most studied tetrapods. Consistent with its large genome, Ig loci are larger; however, local size restrictions indicate evolutionary constraints likely to be imposed by high transcriptional demand of certain Ig genes, as well as the V(D)J recombination over very long genomic distance ranges. The A. mexicanum has undergone an extensive process of Ig gene loss which partially explains a reduced potential repertoire diversity that may contribute to its impaired antibody response.
Collapse
Affiliation(s)
- Jesús Martinez-Barnetche
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Morelos, Mexico,*Correspondence: Jesús Martinez-Barnetche, ; Constantino López-Macías,
| | | | - Stephanie Saint Remy-Hernández
- Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México City, Mexico,Unidad de Investigación Médica en Inmunoquímica, UMAE Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, México City, Mexico
| | - Diana Laura Pacheco-Olvera
- Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México City, Mexico,Unidad de Investigación Médica en Inmunoquímica, UMAE Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, México City, Mexico
| | - Juan Téllez-Sosa
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Morelos, Mexico
| | - Humberto Valdovinos-Torres
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Morelos, Mexico
| | | | - Horacio Mena
- Laboratorio de Restauración Ecológica, Instituto de Biología. Universidad Nacional Autónoma de México, México City, Mexico
| | - Luis Zambrano
- Laboratorio de Restauración Ecológica, Instituto de Biología. Universidad Nacional Autónoma de México, México City, Mexico
| | - Constantino López-Macías
- Unidad de Investigación Médica en Inmunoquímica, UMAE Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, México City, Mexico,*Correspondence: Jesús Martinez-Barnetche, ; Constantino López-Macías,
| |
Collapse
|
5
|
Abstract
COVID-19 illness is associated with diverse neurological manifestations. Its exceptionally high prevalence results from unprecedented genetic diversity, genomic recombination, and superspreading. With each new mutation and variant, there are foreseeable risks of rising fatality and novel neurological motor complications in childhood and adult cases. This chapter provides an extensive review of COVID-19 neurological illness, notably the motor manifestations. Innovative treatments have been developed to stem the spread of infectious contagious illness, and attenuate the resultant cytokine storm and other postinfectious immune aspects responsible for postacute COVID-19 syndrome due to the multiplier effect of infection, immunity, and inflammation, termed I3.
Collapse
Affiliation(s)
- David S Younger
- Department of Clinical Medicine and Neuroscience, CUNY School of Medicine, New York, NY, United States; Department of Medicine, Section of Internal Medicine and Neurology, White Plains Hospital, White Plains, NY, United States.
| |
Collapse
|
6
|
Zhang Z, Gao Y, Li L, Luo J, Gao R. Deficiency of C-reactive protein or human C-reactive protein transgenic treatment aggravates influenza A infection in mice. Front Immunol 2022; 13:1028458. [PMID: 36275680 PMCID: PMC9584053 DOI: 10.3389/fimmu.2022.1028458] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 09/20/2022] [Indexed: 12/15/2022] Open
Abstract
C-reactive protein (CRP) has been shown to be a potential candidate target in the immunotherapy of severe influenza A infection. However, it is unclear on the pathogenesis associated with CRP in influenza infections. Here, we used influenza A H1N1 CA04 to infect human CRP transgenic mice (KI), CRP knockout mice (KO), and wild-type mice (WT), respectively, and compared the viral pathogenicity and associated immune response in those mice. The results showed that CA04 infection resulted in 100%, 80%, and 60% death in KO, KI, and WT mice, respectively. Compared to WT mice, CA04 infection resulted in higher TCID50 in lungs on day 3 after infection but lowered HI antibody titers in sera of survivors on day 21 after infection in KI mice. ELISA assay showed that IFN-γ concentration was significantly increased in sera of WT, KI, or KO mice on day 7 after infection, and IL-17 was remarkably increased in sera of WT mice but decreased in sera of KI mice while no significant change in sera of KO mice on day 3 or 7 after infection. Quantitative RT-PCR showed that the relative expression levels of immune checkpoint CTLA-4, LAIR-1, GITR, BTLA, TIM-3, or PD-1 mRNA in the lung presented decreased levels on day 3 or 7 after infection in KI or KO mice. The correlation analysis showed that mRNA expression levels of the 6 molecules positively correlated with viral TICD50 in WT mice but negatively correlated with viral TCID50 in KI or KO mice. However, only LAIR-1 presented a significant correlation in each lung tissue of WT, KI, or KO mice with CA07 infection statistically. IHC results showed that LAIR-1 positive cells could be found in WT, KO, or KI mice lung tissues with CA04 infection, and the positive cells were mainly distributed in an inflammatory dense area. Our results suggested that deficiency of CRP or human CRP transgenic treatment aggravates influenza A virus infection in mice. CRP is a double sword in immune regulation of influenza infection in which IL-17 and immune checkpoint may be involved.
Collapse
Affiliation(s)
- Zhuohan Zhang
- National Health Commission of People's Republic of China (NHC) Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- National Health Commission of People's Republic of China (NHC) Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yongjun Gao
- National Health Commission of People's Republic of China (NHC) Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- National Health Commission of People's Republic of China (NHC) Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Li Li
- National Health Commission of People's Republic of China (NHC) Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- National Health Commission of People's Republic of China (NHC) Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Junhao Luo
- National Health Commission of People's Republic of China (NHC) Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- National Health Commission of People's Republic of China (NHC) Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Rongbao Gao
- National Health Commission of People's Republic of China (NHC) Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- National Health Commission of People's Republic of China (NHC) Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- *Correspondence: Rongbao Gao,
| |
Collapse
|
7
|
Medetgul-Ernar K, Davis MM. Standing on the shoulders of mice. Immunity 2022; 55:1343-1353. [PMID: 35947979 PMCID: PMC10035762 DOI: 10.1016/j.immuni.2022.07.008] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 07/12/2022] [Accepted: 07/15/2022] [Indexed: 01/13/2023]
Abstract
While inbred mice have informed most of what we know about the immune system in the modern era, they have clear limitations with respect to their ability to be informative regarding genetic heterogeneity or microbial influences. They have also not been very predictive as models of human disease or vaccination results. Although there are concerted attempts to compensate for these flaws, the rapid rise of human studies, driven by both technical and conceptual advances, promises to fill in these gaps, as well as provide direct information about human diseases and vaccination responses. Work on human immunity has already provided important additional perspectives on basic immunology such as the importance of clonal deletion to self-tolerance, and while many challenges remain, it seems inevitable that "the human model" will continue to inform us about the immune system and even allow for the discovery of new mechanisms.
Collapse
Affiliation(s)
- Kwat Medetgul-Ernar
- Immunology Program, Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Mark M Davis
- Howard Hughes Medical Institute, Institute for Immunity, Transplantation and Infection, Department of Microbiology and Immunology, Stanford University School of Medicine, Palo Alto, CA 94304, USA.
| |
Collapse
|
8
|
Lichterfeld M, Gao C, Yu XG. An ordeal that does not heal: understanding barriers to a cure for HIV-1 infection. Trends Immunol 2022; 43:608-616. [PMID: 35905706 PMCID: PMC9346997 DOI: 10.1016/j.it.2022.06.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/10/2022] [Accepted: 06/12/2022] [Indexed: 12/23/2022]
Abstract
With more than 38 million people living with HIV-1 (PLWH) worldwide, developing a cure for HIV-1 remains a major global health priority. Lifelong persistence of HIV-1 is frequently attributed to a pool of stable, transcriptionally silent HIV-1 proviruses, which are unaffected by currently available antiretroviral therapy (ART) or host immune activity. In this opinion article, we propose a more dynamic interpretation of HIV-1 reservoir cell biology and argue that HIV-1 proviruses frequently display residual viral transcriptional activity, making them vulnerable to longitudinal immune-mediated selection processes. Such mechanisms may, over extended periods of ART, induce an attenuated viral reservoir profile characterized by intact proviruses preferentially integrated into heterochromatin locations. We suggest that intensifying and accelerating naturally occurring selection mechanisms might represent a promising strategy for finding a potential cure for HIV-1 infection.
Collapse
Affiliation(s)
- Mathias Lichterfeld
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA; Infectious Disease Division, Brigham and Women's Hospital, Boston, MA 02115, USA.
| | - Ce Gao
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA; Infectious Disease Division, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Xu G Yu
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA; Infectious Disease Division, Brigham and Women's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
9
|
Li W, Meng J, Ma X, Lin J, Lu X. Advanced materials for the delivery of vaccines for infectious diseases. BIOSAFETY AND HEALTH 2022. [DOI: 10.1016/j.bsheal.2022.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
10
|
Guo Y, Hu K, Li Y, Lu C, Ling K, Cai C, Wang W, Ye D. Targeting TNF-α for COVID-19: Recent Advanced and Controversies. Front Public Health 2022; 10:833967. [PMID: 35223745 PMCID: PMC8873570 DOI: 10.3389/fpubh.2022.833967] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 01/17/2022] [Indexed: 12/14/2022] Open
Abstract
Recent advances in the pathophysiologic understanding of coronavirus disease 2019 (COVID-19) suggests that cytokine release syndrome (CRS) has an association with the severity of disease, which is characterized by increased tumor necrosis factor α (TNF-α), interleukin (IL)-6, IL-2, IL-7, and IL-10. Hence, managing CRS has been recommended for rescuing severe COVID-19 patients. TNF-α, one of the pro-inflammatory cytokines commonly upregulated in acute lung injury, triggers CRS and facilitates SARS-CoV-2 interaction with angiotensin-converting enzyme 2 (ACE2). TNF-α inhibitors, therefore, may serve as an effective therapeutic strategy for attenuating disease progression in severe SARS-CoV-2 infection. Below, we review the possibilities and challenges of targeting the TNF-α pathway in COVID-19 treatment.
Collapse
Affiliation(s)
- Yi Guo
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ke Hu
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuxuan Li
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chanjun Lu
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ken Ling
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chuanqi Cai
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weici Wang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Weici Wang
| | - Dawei Ye
- Department of Pancreatic-Biliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Department of Cancer Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Dawei Ye
| |
Collapse
|
11
|
Shiozawa S, Tsumiyama K, Miyazaki Y, Uto K, Sakurai K, Nakashima T, Matsuyama H, Doi A, Tarui M, Izumikawa M, Kimura M, Fujita Y, Satonaka C, Horiuchi T, Matsubara T, Oribe M, Yamane T, Kagawa H, Li QZ, Mizuno K, Mukai Y, Murakami K, Enya T, Tsukimoto S, Hakata Y, Miyazawa M, Shiozawa K. DOCK8-expressing T follicular helper cells newly generated beyond self-organized criticality cause systemic lupus erythematosus. iScience 2022; 25:103537. [PMID: 34977502 PMCID: PMC8689056 DOI: 10.1016/j.isci.2021.103537] [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: 05/12/2021] [Revised: 10/01/2021] [Accepted: 11/24/2021] [Indexed: 11/24/2022] Open
Abstract
Pathogens including autoantigens all failed to induce systemic lupus erythematosus (SLE). We, instead, studied the integrity of host's immune response that recognized pathogen. By stimulating TCR with an antigen repeatedly to levels that surpass host's steady-state response, self-organized criticality, SLE was induced in mice normally not prone to autoimmunity, wherein T follicular helper (Tfh) cells expressing the guanine nucleotide exchange factor DOCK8 on the cell surface were newly generated. DOCK8+Tfh cells passed through TCR re-revision and induced varieties of autoantibody and lupus lesions. They existed in splenic red pulp and peripheral blood of active lupus patients, which subsequently declined after therapy. Autoantibodies and disease were healed by anti-DOCK8 antibody in the mice including SLE-model (NZBxNZW) F1 mice. Thus, DOCK8+Tfh cells generated after repeated TCR stimulation by immunogenic form of pathogen, either exogenous or endogenous, in combination with HLA to levels that surpass system's self-organized criticality, cause SLE. Autoimmunity seldom takes place under integrated steady-state immune response Repeated invasion by pathogen, such as measles virus, is not exceptional but routine in life DOCK8+Tfh is generated upon TCR overstimulation by pathogen beyond self-organized criticality Newly generated DOCK8+Tfh induces autoantibodies and SLE, i.e., autoimmunity
Collapse
Affiliation(s)
- Shunichi Shiozawa
- Institute for Rheumatic Diseases, 944-25 Fujita, Katoshi 673-1462, Japan.,Department of Medicine, Kyushu University Beppu Hospital, 4546 Tsurumihara, Beppu 874-0838, Japan.,Division of Bioregulation, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Sumaku, Kobe 654-0142, Japan.,Department of Medicine, Rheumatology and Orthopedic Surgery, Matsubara Mayflower Hospital, 944-25 Fujita, Katoshi 673-1462, Japan
| | - Ken Tsumiyama
- Institute for Rheumatic Diseases, 944-25 Fujita, Katoshi 673-1462, Japan.,Department of Medicine, Kyushu University Beppu Hospital, 4546 Tsurumihara, Beppu 874-0838, Japan.,Division of Bioregulation, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Sumaku, Kobe 654-0142, Japan.,Department of Medicine, Rheumatology and Orthopedic Surgery, Matsubara Mayflower Hospital, 944-25 Fujita, Katoshi 673-1462, Japan
| | - Yumi Miyazaki
- Department of Medicine, Kyushu University Beppu Hospital, 4546 Tsurumihara, Beppu 874-0838, Japan.,Division of Bioregulation, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Sumaku, Kobe 654-0142, Japan
| | - Kenichi Uto
- Division of Bioregulation, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Sumaku, Kobe 654-0142, Japan
| | - Keiichi Sakurai
- Institute for Rheumatic Diseases, 944-25 Fujita, Katoshi 673-1462, Japan.,Department of Medicine, Kyushu University Beppu Hospital, 4546 Tsurumihara, Beppu 874-0838, Japan
| | - Toshie Nakashima
- Division of Bioregulation, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Sumaku, Kobe 654-0142, Japan
| | - Hiroko Matsuyama
- Division of Bioregulation, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Sumaku, Kobe 654-0142, Japan
| | - Ai Doi
- Division of Bioregulation, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Sumaku, Kobe 654-0142, Japan
| | - Miho Tarui
- Division of Bioregulation, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Sumaku, Kobe 654-0142, Japan
| | - Manabu Izumikawa
- Division of Bioregulation, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Sumaku, Kobe 654-0142, Japan
| | - Mai Kimura
- Division of Bioregulation, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Sumaku, Kobe 654-0142, Japan
| | - Yuko Fujita
- Division of Bioregulation, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Sumaku, Kobe 654-0142, Japan
| | - Chisako Satonaka
- Division of Bioregulation, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Sumaku, Kobe 654-0142, Japan
| | - Takahiko Horiuchi
- Department of Medicine, Kyushu University Beppu Hospital, 4546 Tsurumihara, Beppu 874-0838, Japan
| | - Tsukasa Matsubara
- Department of Medicine, Rheumatology and Orthopedic Surgery, Matsubara Mayflower Hospital, 944-25 Fujita, Katoshi 673-1462, Japan
| | - Motohiro Oribe
- Oribe Clinic, 1-8-15 Higashi-Odori, Oita 870-0823, Japan
| | - Takashi Yamane
- Department of Rheumatology, Kakogawa City Hospital, 439 Honmachi, Kakogawa 675-8611, Japan
| | - Hidetoshi Kagawa
- Department of Medicine, Red Cross Society Himeji Hospital, 1-12-1 Shimoteno, Himeji 670-8540, Japan
| | - Quan-Zhen Li
- Department of Immunology, University of Texas Southwestern Medical Center, 6001 Forest Park Road/ND 6.504, Dallas, TX 75390-8814, USA
| | - Keiko Mizuno
- Drug Discovery Platform, KAN Research Institute, Inc., 6-8-2 Minatojimaminamicho, Kobe 650-0047, Japan
| | - Yohei Mukai
- Drug Discovery Platform, KAN Research Institute, Inc., 6-8-2 Minatojimaminamicho, Kobe 650-0047, Japan
| | - Kazuhiro Murakami
- Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsujima, Aobaku 981-8558, Japan
| | - Takuji Enya
- Department of Immunology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka 589-8511, Japan.,Department of Pediatrics, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka 589-8511, Japan
| | - Shota Tsukimoto
- Department of Immunology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka 589-8511, Japan.,Department of Anesthesiology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka 589-8511, Japan
| | - Yoshiyuki Hakata
- Department of Immunology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka 589-8511, Japan
| | - Masaaki Miyazawa
- Department of Immunology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka 589-8511, Japan.,Kindai University Anti-Aging Center, 3-4-1 Kowakae, Higashi-Osaka, Osaka 577-8502, Japan
| | - Kazuko Shiozawa
- Department of Medicine, Rheumatology and Orthopedic Surgery, Matsubara Mayflower Hospital, 944-25 Fujita, Katoshi 673-1462, Japan.,Rheumatology and Collagen Disease Center, Hyogo Prefectural Kakogawa Medical Center, 203 Kanno, Kakogawa 675-8555, Japan
| |
Collapse
|
12
|
Abstract
ABSTRACT The ongoing coronavirus disease 2019 (COVID-19) pandemic has swept over the world and causes thousands of deaths. Although the clinical features of COVID-19 become much clearer than before, there are still further problems with the pathophysiological process and treatments of severe patients. One primary problem is with the paradoxical immune states in severe patients with COVID-19. Studies indicate that Severe Acute Respiratory Syndrome Coronavirus 2 can attack the immune system, manifested as a state of immunosuppression with a decrease in lymphocytes, whereas a state of hyperinflammation, presenting as elevated cytokine levels, is also detected in COVID-19. Therefore, discussing the specific status of immunity in COVID-19 will contribute to the understanding of its pathophysiology and the search for appropriate treatments. Here, we review all the available literature concerning the different immune states in COVID-19 and the underlying pathophysiological mechanisms. In addition, the association between immune states and the development and severity of disease as well as the impact on the selection of immunotherapy strategies are discussed in our review.
Collapse
Affiliation(s)
- Ye Liu
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
- Clinical Research Center of Hubei Critical Care Medicine, Hubei, China
| | - Yiming Li
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
- Clinical Research Center of Hubei Critical Care Medicine, Hubei, China
| | - Dongxue Xu
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
- Clinical Research Center of Hubei Critical Care Medicine, Hubei, China
| | - Jing Zhang
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
- Clinical Research Center of Hubei Critical Care Medicine, Hubei, China
| | - Zhiyong Peng
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
- Clinical Research Center of Hubei Critical Care Medicine, Hubei, China
| |
Collapse
|
13
|
Krolik M, Csepregi L, Hartmann F, Engetschwiler C, Flatz L. Recombinant lymphocytic choriomeningitis virus-based vaccine vector protects type I interferon receptor deficient mice from viral challenge. Vaccine 2021; 39:1257-1264. [PMID: 33518468 DOI: 10.1016/j.vaccine.2021.01.047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 12/07/2020] [Accepted: 01/16/2021] [Indexed: 01/20/2023]
Abstract
Reverse genetically engineered recombinant lymphocytic choriomeningitis virus (rLCMV) is a novel vaccine vector platform. Here, we investigate the safety and efficacy of rLCMV in mice lacking a functional type I interferon system with high susceptibility to viral infections. Propagation-deficient rLCMV vector expressing ovalbumin as a model antigen is cleared from type I interferon receptor-deficient mice (Ifnar-/-) within seven days post vaccination. In Ifnar-/-, induction of vaccine antigen specific T cells is delayed compared to wild type animals. However, immunization of Ifnar-/- results in potent memory formation and generates multifunctional cytotoxic CD8+ T cells. Most importantly, Ifnar-/- vaccinated with rLCMV are protected from a challenge with the aggressive LCMV Clone 13. Our data provide evidence for an excellent safety profile with maintained efficacy in immunocompromised animals.
Collapse
Affiliation(s)
- Michal Krolik
- Institute of Immunobiology, Kantonsspital St.Gallen, Rorschacher Strasse 95, 9007 St.Gallen, Switzerland
| | - Lucia Csepregi
- Institute of Immunobiology, Kantonsspital St.Gallen, Rorschacher Strasse 95, 9007 St.Gallen, Switzerland
| | - Fabienne Hartmann
- Institute of Immunobiology, Kantonsspital St.Gallen, Rorschacher Strasse 95, 9007 St.Gallen, Switzerland
| | - Céline Engetschwiler
- Institute of Immunobiology, Kantonsspital St.Gallen, Rorschacher Strasse 95, 9007 St.Gallen, Switzerland
| | - Lukas Flatz
- Institute of Immunobiology, Kantonsspital St.Gallen, Rorschacher Strasse 95, 9007 St.Gallen, Switzerland; Department of Dermatology, University Hospital Zurich, Gloriastrasse 31, 8091 Zurich, Switzerland; Department of Oncology and Hematology, Kantonsspital St.Gallen, Rorschacher Strasse 95, 9007 St.Gallen, Switzerland; Department of Dermatology, Kantonsspital St.Gallen, Rorschacher Strasse 95, 9007 St.Gallen, Switzerland.
| |
Collapse
|
14
|
Chunder R, Schropp V, Kuerten S. B Cells in Multiple Sclerosis and Virus-Induced Neuroinflammation. Front Neurol 2020; 11:591894. [PMID: 33224101 PMCID: PMC7670072 DOI: 10.3389/fneur.2020.591894] [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: 08/05/2020] [Accepted: 10/05/2020] [Indexed: 01/02/2023] Open
Abstract
Neuroinflammation can be defined as an inflammatory response within the central nervous system (CNS) mediated by a complex crosstalk between CNS-resident and infiltrating immune cells from the periphery. Triggers for neuroinflammation not only include pathogens, trauma and toxic metabolites, but also autoimmune diseases such as neuromyelitis optica spectrum disorders and multiple sclerosis (MS) where the inflammatory response is recognized as a disease-escalating factor. B cells are not considered as the first responders of neuroinflammation, yet they have recently gained focus as a key component involved in the disease pathogenesis of several neuroinflammatory disorders like MS. Traditionally, the prime focus of the role of B cells in any disease, including neuroinflammatory diseases, was their ability to produce antibodies. While that may indeed be an important contribution of B cells in mediating disease pathogenesis, several lines of recent evidence indicate that B cells are multifunctional players during an inflammatory response, including their ability to present antigens and produce an array of cytokines. Moreover, interaction between B cells and other cellular components of the immune system or nervous system can either promote or dampen neuroinflammation depending on the disease. Given that the interest in B cells in neuroinflammation is relatively new, the precise roles that they play in the pathophysiology and progression of different neuroinflammatory disorders have not yet been well-elucidated. Furthermore, the possibility that they might change their function during the course of neuroinflammation adds another level of complexity and the puzzle remains incomplete. Indeed, advancing our knowledge on the role of B cells in neuroinflammation would also allow us to tackle these disorders better. Here, we review the available literature to explore the relationship between autoimmune and infectious neuroinflammation with a focus on the involvement of B cells in MS and viral infections of the CNS.
Collapse
Affiliation(s)
- Rittika Chunder
- Institute of Anatomy and Cell Biology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Verena Schropp
- Institute of Anatomy and Cell Biology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Stefanie Kuerten
- Institute of Anatomy and Cell Biology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
15
|
Perez-Shibayama C, Islander U, Lütge M, Cheng HW, Onder L, Ring SS, De Martin A, Novkovic M, Colston J, Gil-Cruz C, Ludewig B. Type I interferon signaling in fibroblastic reticular cells prevents exhaustive activation of antiviral CD8 + T cells. Sci Immunol 2020; 5:5/51/eabb7066. [PMID: 32917792 DOI: 10.1126/sciimmunol.abb7066] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 08/20/2020] [Indexed: 12/16/2022]
Abstract
Fibroblastic reticular cells (FRCs) are stromal cells that actively promote the induction of immune responses by coordinating the interaction of innate and adaptive immune cells. However, whether and to which extent immune cell activation is determined by lymph node FRC reprogramming during acute viral infection has remained unexplored. Here, we genetically ablated expression of the type I interferon-α receptor (Ifnar) in Ccl19-Cre+ cells and found that sensing of type I interferon imprints an antiviral state in FRCs and thereby preserves myeloid cell composition in lymph nodes of naive mice. During localized lymphocytic choriomeningitis virus infection, IFNAR signaling precipitated profound phenotypic adaptation of all FRC subsets enhancing antigen presentation, chemokine-driven immune cell recruitment, and immune regulation. The IFNAR-dependent shift of all FRC subsets toward an immunostimulatory state reduced exhaustive CD8+ T cell activation. In sum, these results unveil intricate circuits underlying type I IFN sensing in lymph node FRCs that enable protective antiviral immunity.
Collapse
Affiliation(s)
| | - Ulrika Islander
- Krefting Research Centre, Department of Internal Medicine and Clinical Nutrition, University of Gothenburg, Gothenburg, Sweden
| | - Mechthild Lütge
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Hung-Wei Cheng
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Lucas Onder
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Sandra S Ring
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Angelina De Martin
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Mario Novkovic
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Julia Colston
- Department of Medicine, Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, UK
| | - Cristina Gil-Cruz
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland. .,Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
16
|
Huang M, Wang Y, Ye J, Da H, Fang S, Chen L. Dynamic changes of T-lymphocyte subsets and the correlations with 89 patients with coronavirus disease 2019 (COVID-19). ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1145. [PMID: 33240994 PMCID: PMC7576080 DOI: 10.21037/atm-20-5479] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background In December 2019, an outbreak of coronavirus disease 2019 (COVID-19), caused by a novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), occurred in Wuhan City, Hubei Province, China. The coronavirus has spread throughout the world, posing a severe threat to human health. By using flow cytometry, here we observed the dynamic changes of peripheral blood T lymphocyte subsets in COVID-19 patients, with an attempt to explore their roles in the pathogenesis of COVID-19 and their impacts on prognosis. Methods Eighty-nine COVID-19 patients were divided into a moderate group (n=70) and the severe/critical group (n=19) according to the disease severity. Furthermore, the severe/critical patients were divided into the improved group (n=14) and unimproved group (n=5) according to the outcomes. The absolute peripheral blood lymphocytes counts and subsets, including CD45+, CD3+, CD4+, and CD8+, in the acute phase, and flow cytometry measured the recovery phase for all patients. Then, the results were compared with those in the normal control group. Results The absolute counts of lymphocytes, T lymphocytes, and their subsets decreased during the acute phase in COVID-19 patients, especially in the severe/critical group. The T-lymphocyte count reached the lowest point on the 14th day in the severe/critical group. It rose with fluctuations to the normal level in the improved group as the immune function recovered; in the unimproved group, however, the T-lymphocyte count remained at a low level or even continued to decrease. The percentages of CD4+ and CD8+ T lymphocytes showed no visible change in the improved group; however, the percentage of CD8+ T cells dropped in the unimproved group, resulting in higher CD4+/CD8+ ratio. Conclusions T lymphocytes count, and their subsets can be used for monitoring the immune functions and predicting the prognosis of COVID-19 patients.
Collapse
Affiliation(s)
- Mingxiang Huang
- Fuzhou Pulmonary Hospital & Fujian Medical University Clinical Teaching Hospital, Fuzhou, China
| | - Yao Wang
- Fuzhou Pulmonary Hospital & Fujian Medical University Clinical Teaching Hospital, Fuzhou, China
| | - Jing Ye
- Fuzhou Pulmonary Hospital & Fujian Medical University Clinical Teaching Hospital, Fuzhou, China
| | - Hongqiang Da
- Fuzhou Pulmonary Hospital & Fujian Medical University Clinical Teaching Hospital, Fuzhou, China
| | - Sufang Fang
- Fuzhou Pulmonary Hospital & Fujian Medical University Clinical Teaching Hospital, Fuzhou, China
| | - Lizhou Chen
- Fuzhou Pulmonary Hospital & Fujian Medical University Clinical Teaching Hospital, Fuzhou, China
| |
Collapse
|
17
|
De D, Pandhi D. Use of Immunosuppressants/Immunomodulators in Autoimmune/Inflammatory Dermatologic Diseases during COVID-19 Pandemic-General Recommendation Based on Available Evidence. Indian Dermatol Online J 2020; 11:526-533. [PMID: 32832437 PMCID: PMC7413441 DOI: 10.4103/idoj.idoj_414_20] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/05/2020] [Accepted: 06/19/2020] [Indexed: 12/24/2022] Open
Affiliation(s)
- Dipankar De
- Department of Dermatology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Deepika Pandhi
- Department of Dermatology and STD, University College of Medical Sciences and GTB Hospital, Delhi, India E-mail:
| |
Collapse
|
18
|
Bashir S, Paeshuyse J. Construction of Antibody Phage Libraries and Their Application in Veterinary Immunovirology. Antibodies (Basel) 2020; 9:E21. [PMID: 32503103 PMCID: PMC7345743 DOI: 10.3390/antib9020021] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/26/2020] [Accepted: 04/28/2020] [Indexed: 12/14/2022] Open
Abstract
Antibody phage display (APD) technology has revolutionized the field of immunovirology with its application in viral disease diagnostics and antiviral therapy. This robust and versatile technology allows the expression of an antibody fused to a phage coat protein on the surface of a filamentous phage. The DNA sequence coding for the antibody is packaged within the phage, linking the phenotype to genotype. Antibody phage display inherits the ability to rapidly generate and modify or improve high-affinity monoclonal antibodies, rendering it indispensable in immunology. In the last two decades, phage-display-derived antibodies have been extensively used in human medicine as diagnostic and therapeutic modalities. Recently, they are also gaining significant ground in veterinary medicine. Even though these advancements are mainly biased towards economically important animals such as chicken, cattle, and pigs, they are laying the foundation of fulfilling the unmet needs of veterinary medicine as antibody-based biologics in viral diagnostics, therapeutics, and immunoprophylaxis. This review provides a brief overview of the construction of antibody phage libraries and their application in diagnosis, prevention, and control of infectious viral diseases in veterinary medicine in detail.
Collapse
Affiliation(s)
| | - Jan Paeshuyse
- Department of Biosystems, Division of Animal and Human Health Engineering, Laboratory of Host Pathogen Interaction in Livestock, KU Leuven University, 3000 Leuven, Belgium;
| |
Collapse
|
19
|
Panova V, Attig J, Young GR, Stoye JP, Kassiotis G. Antibody-induced internalisation of retroviral envelope glycoproteins is a signal initiation event. PLoS Pathog 2020; 16:e1008605. [PMID: 32453763 PMCID: PMC7274472 DOI: 10.1371/journal.ppat.1008605] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 06/05/2020] [Accepted: 05/05/2020] [Indexed: 12/31/2022] Open
Abstract
As obligate parasites, viruses highjack, modify and repurpose the cellular machinery for their own replication. Viral proteins have, therefore, evolved biological functions, such as signalling potential, that alter host cell physiology in ways that are still incompletely understood. Retroviral envelope glycoproteins interact with several host proteins, extracellularly with their cellular receptor and anti-envelope antibodies, and intracellularly with proteins of the cytoskeleton or sorting, endocytosis and recirculation pathways. Here, we examined the impact of endogenous retroviral envelope glycoprotein expression and interaction with host proteins, particularly antibodies, on the cell, independently of retroviral infection. We found that in the commonly used C57BL/6 substrains of mice, where murine leukaemia virus (MLV) envelope glycoproteins are expressed by several endogenous MLV proviruses, the highest expressed MLV envelope glycoprotein is under the control of an immune-responsive cellular promoter, thus linking MLV envelope glycoprotein expression with immune activation. We further showed that antibody ligation induces extensive internalisation from the plasma membrane into endocytic compartments of MLV envelope glycoproteins, which are not normally subject to constitutive endocytosis. Importantly, antibody binding and internalisation of MLV envelope glycoproteins initiates signalling cascades in envelope-expressing murine lymphocytic cell lines, leading to cellular activation. Similar effects were observed by MLV envelope glycoprotein ligation by its cellular receptor mCAT-1, and by overexpression in human lymphocytic cells, where it required an intact tyrosine-based YXXΦ motif in the envelope glycoprotein cytoplasmic tail. Together, these results suggest that signalling potential is a general property of retroviral envelope glycoproteins and, therefore, a target for intervention. The outcome of viral infection depends on the balance between host immunity and the ability of the virus to avoid, evade or subvert it. The envelope glycoproteins of diverse viruses, including retroviruses, are displayed on the surface of virions and of infected cells and thus constitute the major target of the host antibody response. Antibody responses are elicited not only against infectious viruses we acquire during our life-history, but also against the numerous retroviral envelopes encoded by our genome and acquired during our species’ life-history. In turn, viruses have evolved ways to reduce exposure of their envelope glycoproteins to the host immune system, including constitutive endocytosis or antibody-induced internalisation. Using murine leukaemia viruses as models of infectious and endogenous retroviruses, we show that antibody binding to retroviral envelopes induces extensive internalisation of the envelope-antibody complex and initiates signalling cascades, ultimately leading to transcriptional activation of envelope glycoprotein-expressing lymphocytes. We further show that expression of endogenous retroviral envelopes is coupled to physiological lymphocyte activation, integrating them with the immune response. These findings reveal an unexpected layer of interaction between the host antibody response and retroviral envelope glycoproteins, which could be considered immune receptors.
Collapse
Affiliation(s)
- Veera Panova
- Retroviral Immunology, The Francis Crick Institute, United Kingdom
| | - Jan Attig
- Retroviral Immunology, The Francis Crick Institute, United Kingdom
| | - George R. Young
- Retrovirus-Host Interactions, The Francis Crick Institute, London, United Kingdom
| | - Jonathan P. Stoye
- Retrovirus-Host Interactions, The Francis Crick Institute, London, United Kingdom
- Department of Medicine, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - George Kassiotis
- Retroviral Immunology, The Francis Crick Institute, United Kingdom
- Department of Medicine, Faculty of Medicine, Imperial College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
20
|
Niklasson B, Lindquist L, Klitz W, Netherlands Brain Bank, Englund E. Picornavirus Identified in Alzheimer's Disease Brains: A Pathogenic Path? J Alzheimers Dis Rep 2020; 4:141-146. [PMID: 32587947 PMCID: PMC7306919 DOI: 10.3233/adr-200174] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2020] [Indexed: 12/25/2022] Open
Abstract
We investigated formalin-fixed postmortem brain tissue from the hippocampus region of 18 AD cases and 11 age-matched controls using a polyclonal antibody against Ljungan virus (LV) capsid protein 1. Evidence of a LV antigen was found in all AD cases but in none of the control specimens (p < 0.0001). The antibodies reacted with neurons and astrocytes and also showed distinct positive reaction in the amyloid/neuritic plaques. The possible role of an incompletely characterized picornavirus as the etiologic agent in AD open up the possibility of treatment with antiviral therapy directed against picornaviruses. The positive result of such treatment in a small number of patients is presented separately back to back to this report.
Collapse
Affiliation(s)
- Bo Niklasson
- Jordbro Primary Health Care Center, Stockholm, Sweden
| | - Lars Lindquist
- Department of Medicine Huddinge, Division of Infectious Diseases, Karolinska Institutet, Stockholm, Sweden
| | - William Klitz
- Department of Integrative Biology, University of California, Berkeley, CA, USA
| | - Netherlands Brain Bank
- Netherlands Brain Bank, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Elisabet Englund
- Department of Clinical Sciences, Division of Pathology, University of Lund, Lund, Sweden
| |
Collapse
|
21
|
Abstract
Mucosal-associated invariant T (MAIT) cells have been attracting increasing attention over the last few years as a potent unconventional T cell subset. Three factors largely account for this emerging interest. Firstly, these cells are abundant in humans, both in circulation and especially in some tissues such as the liver. Secondly is the discovery of a ligand that has uncovered their microbial targets, and also allowed for the development of tools to accurately track the cells in both humans and mice. Finally, it appears that the cells not only have a diverse range of functions but also are sensitive to a range of inflammatory triggers that can enhance or even bypass T cell receptor–mediated signals—substantially broadening their likely impact in health and disease. In this review we discuss how MAIT cells display antimicrobial, homeostatic, and amplifier roles in vivo, and how this may lead to protection and potentially pathology.
Collapse
Affiliation(s)
- Nicholas M. Provine
- Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Headington, Oxford OX3 9DU, United Kingdom
| | - Paul Klenerman
- Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Headington, Oxford OX3 9DU, United Kingdom
- NIHR Biomedical Research Centre, John Radcliffe Hospital, Headington, Oxford OX3 9DU, United Kingdom
| |
Collapse
|
22
|
TUFAN A, AVANOĞLU GÜLER A, MATUCCI-CERINIC M. COVID-19, immune system response, hyperinflammation and repurposing antirheumatic drugs. Turk J Med Sci 2020; 50:620-632. [PMID: 32299202 PMCID: PMC7195984 DOI: 10.3906/sag-2004-168] [Citation(s) in RCA: 289] [Impact Index Per Article: 57.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 04/16/2020] [Indexed: 12/21/2022] Open
Abstract
In the Wuhan Province of China, in December 2019, the novel coronavirus 2019 (COVID-19) has caused a severe involvement of the lower respiratory tract leading to an acute respiratory syndrome. Subsequently, coronavirus 2 (SARS-CoV-2) provoked a pandemic which is considered a life-threatening disease. The SARS-CoV-2, a family member of betacoronaviruses, possesses single-stranded positive-sense RNA with typical structural proteins, involving the envelope, membrane, nucleocapsid and spike proteins that are responsible for the viral infectivity, and nonstructural proteins. The effectual host immune response including innate and adaptive immunity against SARS-Cov-2 seems crucial to control and resolve the viral infection. However, the severity and outcome of the COVID-19 might be associated with the excessive production of proinflammatory cytokines “cytokine storm” leading to an acute respiratory distress syndrome. Regretfully, the exact pathophysiology and treatment, especially for the severe COVID-19, is still uncertain. The results of preliminary studies have shown that immune-modulatory or immune-suppressive treatments such as hydroxychloroquine, interleukin (IL)-6 and IL-1 antagonists, commonly used in rheumatology, might be considered as treatment choices for COVID-19, particularly in severe disease. In this review, to gain better information about appropriate anti-inflammatory treatments, mostly used in rheumatology for COVID-19, we have focused the attention on the structural features of SARS-CoV-2, the host immune response against SARS-CoV-2 and its association with the cytokine storm.
Collapse
Affiliation(s)
- Abdurrahman TUFAN
- Department of Internal Medicine & Rheumatology, Faculty of Medicine, Gazi University, AnkaraTurkey
| | - Aslıhan AVANOĞLU GÜLER
- Department of Internal Medicine & Rheumatology, Faculty of Medicine, Gazi University, AnkaraTurkey
| | - Marco MATUCCI-CERINIC
- Department of Experimental and Clinical Medicine, University of Florence, FlorenceItaly
| |
Collapse
|
23
|
Ludewig B. Legends of allergy/immunology: Rolf Zinkernagel and the co-discovery of MHC restriction together with Peter Doherty. Allergy 2019; 74:1409-1411. [PMID: 30916402 DOI: 10.1111/all.13796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 03/13/2019] [Accepted: 03/17/2019] [Indexed: 11/30/2022]
Affiliation(s)
- Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| |
Collapse
|
24
|
Fribourg M, Anderson L, Fischman C, Cantarelli C, Perin L, La Manna G, Rahman A, Burrell BE, Heeger PS, Cravedi P. T-cell exhaustion correlates with improved outcomes in kidney transplant recipients. Kidney Int 2019; 96:436-449. [PMID: 31040060 DOI: 10.1016/j.kint.2019.01.040] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 12/08/2018] [Accepted: 01/10/2019] [Indexed: 02/07/2023]
Abstract
Continuous antigen stimulation during chronic infection or malignancy can promote functional T cell silencing, a phenomenon called T cell exhaustion. The prevalence and impact of T cell exhaustion following organ transplantation, another immune stimulus with persistently high antigen load, are unknown. Here, we characterized serially collected peripheral blood mononuclear cells from 26 kidney transplant recipients using time-of-flight mass cytometry (CyTOF) to define distinct subsets of circulating exhausted T cells and their relationship to induction therapy and allograft function. We observed an increase in specific subsets of CD4+ and CD8+ exhausted T cells from pre-transplant to 6-months post-transplant, with greater increases in participants given anti-thymocyte globulin induction than in participants who received no induction or non-depleting induction. The percentages of exhausted T cells at 6 months correlated inversely with adenosine triphosphate (ATP) production (a surrogate of T cell function) and with allograft interstitial fibrosis. Guided by the CyTOF data, we delineated a PD-1+CD57- phenotype for CD4+ and CD8+ exhausted T cells, and confirmed that these cells have limited capacity for cytokine secretion and ATP production. In an independent cohort of 50 kidney transplant recipients, we confirmed the predicted increase of PD-1+CD57- exhausted T cells after lymphocyte-depleting induction therapy and its direct correlation with better allograft function. Our findings suggest that monitoring T cell exhaustion can be useful for post-transplant risk assessment and support the need to develop and test strategies aimed at augmenting T cell exhaustion following kidney transplantation.
Collapse
Affiliation(s)
- Miguel Fribourg
- Department of Medicine, Division of Nephrology and Translational Transplant Research Center, Recanati Miller Transplant Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Lisa Anderson
- Department of Medicine, Division of Nephrology and Translational Transplant Research Center, Recanati Miller Transplant Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Clara Fischman
- Department of Medicine, Division of Nephrology and Translational Transplant Research Center, Recanati Miller Transplant Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Chiara Cantarelli
- Department of Medicine, Division of Nephrology and Translational Transplant Research Center, Recanati Miller Transplant Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Laura Perin
- GOFARR Laboratory for Organ Regenerative Research and Cell Therapeutics in Urology, Children's Hospital Los Angeles, Division of Urology, Saban Research Institute, University of Southern California, Los Angeles, California, USA
| | - Gaetano La Manna
- Department of Experimental Diagnostic and Specialty Medicine (DIMES), Nephrology, Dialysis and Renal Transplant Unit, St. Orsola Hospital, University of Bologna, Bologna, Italy
| | - Adeeb Rahman
- Human Immune Monitoring Core, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | - Peter S Heeger
- Department of Medicine, Division of Nephrology and Translational Transplant Research Center, Recanati Miller Transplant Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Paolo Cravedi
- Department of Medicine, Division of Nephrology and Translational Transplant Research Center, Recanati Miller Transplant Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| |
Collapse
|
25
|
Kaur M, Kumar D, Butty V, Singh S, Esteban A, Fink GR, Ploegh HL, Sehrawat S. Galectin-3 Regulates γ-Herpesvirus Specific CD8 T Cell Immunity. iScience 2018; 9:101-119. [PMID: 30388704 PMCID: PMC6214866 DOI: 10.1016/j.isci.2018.10.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 09/17/2018] [Accepted: 10/10/2018] [Indexed: 02/02/2023] Open
Abstract
To gain insights into the molecular mechanisms and pathways involved in the activation of γ-herpesvirus (MHV68)-specific T cell receptor transnuclear (TN) CD8+ T cells, we performed a comprehensive transcriptomic analysis. Upon viral infection, we observed differential expression of several thousand transcripts encompassing various networks and pathways in activated TN cells compared with their naive counterparts. Activated cells highly upregulated galectin-3. We therefore explored the role of galectin-3 in influencing anti-MHV68 immunity. Galectin-3 was recruited at the immunological synapse during activation of CD8+ T cells and helped constrain their activation. The localization of galectin-3 to immune synapse was evident during the activation of both naive and memory CD8+ T cells. Galectin-3 knockout mice mounted a stronger MHV68-specific CD8+ T cell response to the majority of viral epitopes and led to better viral control. Targeting intracellular galectin-3 in CD8+ T cells may therefore serve to enhance response to efficiently control infections.
Collapse
Affiliation(s)
- Manpreet Kaur
- Indian Institute of Science Education and Research Mohali, Sector 81 SAS Nagar, PO Manauli, Mohali, Knowledge City 140306, Punjab, India
| | - Dhaneshwar Kumar
- Indian Institute of Science Education and Research Mohali, Sector 81 SAS Nagar, PO Manauli, Mohali, Knowledge City 140306, Punjab, India
| | - Vincent Butty
- Whitehead Institute for Biomedical Research, 9 Cambridge Center, Cambridge 02142 MA, USA
| | - Sudhakar Singh
- Indian Institute of Science Education and Research Mohali, Sector 81 SAS Nagar, PO Manauli, Mohali, Knowledge City 140306, Punjab, India
| | - Alexandre Esteban
- Whitehead Institute for Biomedical Research, 9 Cambridge Center, Cambridge 02142 MA, USA
| | - Gerald R Fink
- Whitehead Institute for Biomedical Research, 9 Cambridge Center, Cambridge 02142 MA, USA
| | - Hidde L Ploegh
- Whitehead Institute for Biomedical Research, 9 Cambridge Center, Cambridge 02142 MA, USA.
| | - Sharvan Sehrawat
- Indian Institute of Science Education and Research Mohali, Sector 81 SAS Nagar, PO Manauli, Mohali, Knowledge City 140306, Punjab, India.
| |
Collapse
|
26
|
Wu X, Wu P, Shen Y, Jiang X, Xu F. CD8 + Resident Memory T Cells and Viral Infection. Front Immunol 2018; 9:2093. [PMID: 30283442 PMCID: PMC6156262 DOI: 10.3389/fimmu.2018.02093] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 08/24/2018] [Indexed: 12/24/2022] Open
Abstract
Tissue-resident memory T (Trm) cells are a subset of recently identified memory T cells that mainly reside and serve as sentinels in non-lymphoid peripheral tissues. Unlike the well-characterized circulating central memory T (Tcm) cells and effector memory T (Tem) cells, Trm cells persist in the tissues, do not recirculate into blood, and offer immediate protection against pathogens upon reinfection. In this review, we focus on CD8+ Trm cells and briefly introduce their characteristics, development, maintenance, and function during viral infection. We also discuss some unresolved problems, such as how CD8+ Trm cells adapt to the local tissue microenvironment, how Trm cells interact with other immune cells during their development and maintenance, and the mechanisms by which CD8+ Trm cells confer immune protection. We believe that a better understanding of these problems is of great clinical and therapeutic value and may contribute to more effective vaccination and treatments against viral infection.
Collapse
Affiliation(s)
- Xuejie Wu
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Pin Wu
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yifei Shen
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, United States
| | - Xiaodong Jiang
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, United States
| | - Feng Xu
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
27
|
Abstract
Antibodies play a crucial role in virus control. The production of antibodies requires virus-specific B cells to encounter viral antigens in lymph nodes, become activated, interact with different immune cells, proliferate and enter specific differentiation programmes. Each step occurs in distinct lymph node niches, requiring a coordinated migration of B cells between different subcompartments. The development of multiphoton intravital microscopy has enabled researchers to begin to elucidate the precise cellular and molecular events by which lymph nodes coordinate humoral responses. This Review discusses recent studies that clarify how viruses interfere with antibody responses, highlighting how these mechanisms relate to our topological and temporal understanding of B cell activation within secondary lymphoid organs.
Collapse
Affiliation(s)
- Mirela Kuka
- Division of Immunology, Transplantation and Infectious Diseases and Experimental Imaging Center, IRCCS San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Via Olgettina 58, Milan 20132, Italy
| | - Matteo Iannacone
- Division of Immunology, Transplantation and Infectious Diseases and Experimental Imaging Center, IRCCS San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Via Olgettina 58, Milan 20132, Italy
| |
Collapse
|
28
|
Affiliation(s)
- Paul Klenerman
- Translational Gastroenterology Unit and Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford OX1 3SY, UK.
| | - Eleanor J Barnes
- Translational Gastroenterology Unit and Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford OX1 3SY, UK
| |
Collapse
|
29
|
Abstract
Viral hepatitis in poultry is a complex disease syndrome caused by several viruses belonging to different families including avian hepatitis E virus (HEV), duck hepatitis B virus (DHBV), duck hepatitis A virus (DHAV-1, -2, -3), duck hepatitis virus Types 2 and 3, fowl adenoviruses (FAdV), and turkey hepatitis virus (THV). While these hepatitis viruses share the same target organ, the liver, they each possess unique clinical and biological features. In this article, we aim to review the common and unique features of major poultry hepatitis viruses in an effort to identify the knowledge gaps and aid the prevention and control of poultry viral hepatitis. Avian HEV is an Orthohepevirus B in the family Hepeviridae that naturally infects chickens and consists of three distinct genotypes worldwide. Avian HEV is associated with hepatitis-splenomegaly syndrome or big liver and spleen disease in chickens, although the majority of the infected birds are subclinical. Avihepadnaviruses in the family of Hepadnaviridae have been isolated from ducks, snow geese, white storks, grey herons, cranes, and parrots. DHBV evolved with the host as a noncytopathic form without clinical signs and rarely progressed to chronicity. The outcome for DHBV infection varies by the host's ability to elicit an immune response and is dose and age dependent in ducks, thus mimicking the pathogenesis of human hepatitis B virus (HBV) infections and providing an excellent animal model for human HBV. DHAV is a picornavirus that causes a highly contagious virus infection in ducks with up to 100% flock mortality in ducklings under 6 wk of age, while older birds remain unaffected. The high morbidity and mortality has an economic impact on intensive duck production farming. Duck hepatitis virus Types 2 and 3 are astroviruses in the family of Astroviridae with similarity phylogenetically to turkey astroviruses, implicating the potential for cross-species infections between strains. Duck astrovirus (DAstV) causes acute, fatal infections in ducklings with a rapid decline within 1-2 hr and clinical and pathologic signs virtually indistinguishable from DHAV. DAstV-1 has only been recognized in the United Kingdom and recently in China, while DAstV-2 has been reported in ducks in the United States. FAdV, the causative agent of inclusion body hepatitis, is a Group I avian adenovirus in the genus Aviadenovirus. The affected birds have a swollen, friable, and discolored liver, sometimes with necrotic or hemorrhagic foci. Histologic lesions include multifocal necrosis of hepatocytes and acute hepatitis with intranuclear inclusion bodies in the nuclei of the hepatocytes. THV is a picornavirus that is likely the causative agent of turkey viral hepatitis. Currently there are more questions than answers about THV, and the pathogenesis and clinical impacts remain largely unknown. Future research in viral hepatic diseases of poultry is warranted to develop specific diagnostic assays, identify suitable cell culture systems for virus propagation, and develop effective vaccines.
Collapse
Affiliation(s)
- Danielle M Yugo
- A Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, 1981 Kraft Drive, Blacksburg, VA 24061-0913
| | - Ruediger Hauck
- B Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, CA 95616
| | - H L Shivaprasad
- C California Animal Health and Food Safety Laboratory System, University of California-Davis, Tulare, CA 93274
| | - Xiang-Jin Meng
- A Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, 1981 Kraft Drive, Blacksburg, VA 24061-0913
| |
Collapse
|
30
|
Morsica G, Bagaglio S, Spagnuolo V, Castagna A, Di Serio C, Galli A, Della Torre L, Andolina A, Pramov A, Uberti-Foppa C. Immune response to hepatitis B vaccination in HIV-positive individuals with isolated antibodies against hepatitis B core antigen: Results of a prospective Italian study. PLoS One 2017; 12:e0184128. [PMID: 28863182 PMCID: PMC5581175 DOI: 10.1371/journal.pone.0184128] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 08/18/2017] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND AND AIM Antibodies against hepatitis B core antigen (anti-HBc) are found in 14-44% of patients with HIV infection, but it is still unclear whether hepatitis B virus (HBV) vaccination should be recommended for HIV-positive subjects with isolated anti-HBc (IAHBc). We examined the rate of anamnestic and primary responses (ARs and PRs) and associated factors in a group of HIV-infected patients with an IAHBc profile. METHODS This prospective study recruited 25 HIV-positive patients with anti-HBc alone who were vaccinated against HBV infection. Those without an AR (anti-hepatitis B envelope antigen [anti-HBs] levels of <10 U/L) or who were hypo-responsiveness (anti-HBs levels of >10 but <100 U/L) four weeks after the first dose of vaccine underwent a full course of vaccinations. Their clinical and virological data, including the presence of occult hepatitis B infection (OBI), were evaluated in accordance with the vaccination schedule. RESULTS Six of the 25 patients (24%) showed an AR, four of whom had anti-HBs levels of <100 U/L. Ten of 19 (52.6%) remaining patients became seroprotected after the third dose. OBI was detected in four of the six patients with an AR, two of the 10 patients with a PR, and none of the nine patients who did not respond. Multivariate analysis showed that an AR was associated with the presence of OBI (P = 0.0162), and a PR was associated with HCV antibody status. (P = 0.0191). CONCLUSIONS Our data suggest that testing for anti-HBc alone may not be a reliable means of assessing protection from HBV infection in HIV-positive patients. OBI-positive patients may benefit from a single vaccine dose. Anti-HCV serostatus may affect PRs.
Collapse
Affiliation(s)
- Giulia Morsica
- Division of Infectious Diseases, IRCCS, Ospedale San Raffaele, Milan, Italy
- * E-mail:
| | - Sabrina Bagaglio
- Division of Infectious Diseases, IRCCS, Ospedale San Raffaele, Milan, Italy
| | | | | | - Clelia Di Serio
- Vita-Salute San Raffaele University, CUSSB (University Centre for Statistics in the Biomedical Sciences), Milan, Italy
| | - Andrea Galli
- Division of Infectious Diseases, IRCCS, Ospedale San Raffaele, Milan, Italy
| | | | - Andrea Andolina
- Division of Infectious Diseases, IRCCS, Ospedale San Raffaele, Milan, Italy
| | - Alexander Pramov
- Vita-Salute San Raffaele University, CUSSB (University Centre for Statistics in the Biomedical Sciences), Milan, Italy
| | | |
Collapse
|
31
|
Gandhi GR, Santos VS, Denadai M, da Silva Calisto VK, de Souza Siqueira Quintans J, de Oliveira e Silva AM, de Souza Araújo AA, Narain N, Cuevas LE, Júnior LJQ, Gurgel RQ. Cytokines in the management of rotavirus infection: A systematic review of in vivo studies. Cytokine 2017; 96:152-160. [DOI: 10.1016/j.cyto.2017.04.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 04/07/2017] [Accepted: 04/10/2017] [Indexed: 01/31/2023]
|
32
|
Cabrera-Perez J, Badovinac VP, Griffith TS. Enteric immunity, the gut microbiome, and sepsis: Rethinking the germ theory of disease. Exp Biol Med (Maywood) 2016; 242:127-139. [PMID: 27633573 DOI: 10.1177/1535370216669610] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Sepsis is a poorly understood syndrome of systemic inflammation responsible for hundreds of thousands of deaths every year. The integrity of the gut epithelium and competence of adaptive immune responses are notoriously compromised during sepsis, and the prevalent assumption in the scientific and medical community is that intestinal commensals have a detrimental role in the systemic inflammation and susceptibility to nosocomial infections seen in critically ill, septic patients. However, breakthroughs in the last decade provide strong credence to the idea that our mucosal microbiome plays an essential role in adaptive immunity, where a human host and its prokaryotic colonists seem to exist in a carefully negotiated armistice with compromises and benefits that go both ways. In this review, we re-examine the notion that intestinal contents are the driving force of critical illness. An overview of the interaction between the microbiome and the immune system is provided, with a special focus on the impact of commensals in priming and the careful balance between normal intestinal flora and pathogenic organisms residing in the gut microbiome. Based on the data in hand, we hypothesize that sepsis induces imbalances in microbial populations residing in the gut, along with compromises in epithelial integrity. As a result, normal antigen sampling becomes impaired, and proliferative cues are intermixed with inhibitory signals. This situates the microbiome, the gut, and its complex immune network of cells and bacteria, at the center of aberrant immune responses during and after sepsis.
Collapse
Affiliation(s)
- Javier Cabrera-Perez
- 1 Microbiology, Immunology, and Cancer Biology Graduate Program, University of Minnesota Medical School, Minneapolis, MN 55455, USA.,2 Medical Scientist Training Program, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Vladimir P Badovinac
- 3 Department of Pathology, University of Minnesota Medical School, Minneapolis, MN 55455, USA.,4 Interdisciplinary Program in Immunology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Thomas S Griffith
- 1 Microbiology, Immunology, and Cancer Biology Graduate Program, University of Minnesota Medical School, Minneapolis, MN 55455, USA.,5 Department of Urology, University of Minnesota Medical School, Minneapolis, MN 55455, USA.,6 Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA.,7 Masonic Cancer Center, University of Minnesota Medical School, Minneapolis, MN 55455, USA.,8 Minneapolis VA Health Care System, Minneapolis, MN 55417, USA
| |
Collapse
|
33
|
Affiliation(s)
- Michael A. Steller
- Program in Women's Oncology, Women and Infants' Hospital, Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Brown University School of Medicine, Providence, Rhode Island; St. Elizabeth's Medical Center, Division of Gynecologic Oncology, 736 Cambridge Street, Boston, MA 02135-2997
| |
Collapse
|
34
|
Sanchez-Fueyo A, Markmann JF. Immune Exhaustion and Transplantation. Am J Transplant 2016; 16:1953-7. [PMID: 26729653 DOI: 10.1111/ajt.13702] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 11/09/2015] [Accepted: 12/13/2015] [Indexed: 01/25/2023]
Abstract
Exhaustion of lymphocyte function through chronic exposure to a high load of foreign antigen is well established for chronic viral infection and antitumor immunity and has been found to be associated with a distinct molecular program and characteristic cell surface phenotype. Although exhaustion has most commonly been studied in the context of CD8 viral responses, recent studies indicate that chronic antigen exposure may affect B cells, NK cells and CD4 T cells in a parallel manner. Limited information is available regarding the extent of lymphocyte exhaustion development in the transplant setting and its impact on anti-graft alloreactivity. By analogy to the persistence of a foreign virus, the large mass of alloantigen presented by an allograft in chronic residence could provide an ideal setting for exhausting donor-reactive T cells. The extent of T cell exhaustion occurring with various allografts, the kinetics of its development, whether exhaustion is influenced positively or negatively by different immunosuppressants, and the impact of exhaustion on graft survival and tolerance development remains a fertile area for investigation. Harnessing or encouraging the natural processes of exhaustion may provide a novel means to promote graft survival and transplantation tolerance.
Collapse
Affiliation(s)
- A Sanchez-Fueyo
- Institute of Liver Studies, Division of Transplantation Immunology and Mucosal Biology, Medical Research Council Centre for Transplantation, Faculty of Life Sciences and Medicine, King's College London University, King's College Hospital, London, UK
| | - J F Markmann
- Division of Transplant Surgery, Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
35
|
Affiliation(s)
- Akiko Iwasaki
- Howard Hughes Medical Institute, Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520;
| |
Collapse
|
36
|
Stipp SR, Iniguez A, Wan F, Wodarz D. Timing of CD8 T cell effector responses in viral infections. ROYAL SOCIETY OPEN SCIENCE 2016; 3:150661. [PMID: 26998338 PMCID: PMC4785989 DOI: 10.1098/rsos.150661] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 01/22/2016] [Indexed: 06/05/2023]
Abstract
CD8 T cell or cytotoxic T lymphocyte (CTL) responses are an important branch of the immune system in the fight against viral infections. The dynamics of anti-viral CTL responses have been characterized in some detail, both experimentally and with mathematical models. An interesting experimental observation concerns the timing of CTL responses. A recent study reported that in pneumonia virus of mice the effector CTL tended to arrive in the lung only after maximal virus loads had been achieved, an observation that seems at first counterintuitive because prevention of pathology would require earlier CTL-mediated activity. A delay in CTL-mediated effector activity has also been quoted as a possible explanation for the difficulties associated with CTL-based vaccines. This paper uses mathematical models to show that in specific parameter regimes, delayed CTL effector activity can be advantageous for the host in the sense that it can increase the chances of virus clearance. The increased ability of the CTL to clear the infection, however, is predicted to come at the cost of acute pathology, giving rise to a trade-off, which is discussed in the light of evolutionary processes. This work provides a theoretical basis for understanding the described experimental observations.
Collapse
Affiliation(s)
- Shaun R. Stipp
- Institute for Mathematical Behavioral Sciences, University of California, Irvine, CA, USA
| | - Abdon Iniguez
- Mathematical and Computational Systems Biology, University of California, Irvine, CA, USA
| | - Frederic Wan
- Department of Mathematics, University of California, Irvine, CA, USA
| | - Dominik Wodarz
- Department of Mathematics, University of California, Irvine, CA, USA
- Department of Ecology and Evolutionary Biology, University of California, Irvine, CA, USA
| |
Collapse
|
37
|
Attar BM, Van Thiel D. A New Twist to a Chronic HCV Infection: Occult Hepatitis C. Gastroenterol Res Pract 2015; 2015:579147. [PMID: 26221136 PMCID: PMC4495183 DOI: 10.1155/2015/579147] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Revised: 04/15/2015] [Accepted: 05/24/2015] [Indexed: 02/07/2023] Open
Abstract
Background. The prevalence of occult hepatitis C infection (OCI) in the population of HCV-RNA negative but anti-HCV positive individuals is presently unknown. OCI may be responsible for clinically overt recurrent disease following an apparent sustained viral response (SVR) weeks to years later. Purpose. To review the available current literature regarding OCI, prevalence, pathogenic mechanisms, clinical characteristics, and future directions. Data Sources. Searching MEDLINE, article references, and national and international meeting abstracts for the diagnosis of OCI (1990-2014). Data Synthesis. The long-term followup of individuals with an OCI suggests that the infection can be transient with the loss of detectable HCV-RNA in PPBMCs after 12-18 months or alternatively exist intermittently and potentially long term. The ultimate outcome of HCV infection is decided by interplay between host immune responses, antiviral therapies, and the various well-identified viral evasion mechanisms as well as the presence of HCV infection within extrahepatic tissues. Conclusion. The currently widely held assumption of a HCV-cure in individuals having had "SVR" after 8-12 weeks of a course of DAA therapy as recently defined may not be entirely valid. Careful longitudinal followup utilizing highly sensitive assays and unique approaches to viral isolation are needed.
Collapse
Affiliation(s)
- Bashar M. Attar
- Division of Gastroenterology and Hepatology, Cook County Health and Hospitals System, 1901 West Harrison Street, Chicago, IL 60612, USA
- Rush University Medical Center, Chicago, IL 60612, USA
| | - David Van Thiel
- Advanced Liver and Gastrointestinal Disease Center, Berwyn, IL 60402, USA
| |
Collapse
|
38
|
Kløverpris HN, McGregor R, McLaren JE, Ladell K, Harndahl M, Stryhn A, Carlson JM, Koofhethile C, Gerritsen B, Keşmir C, Chen F, Riddell L, Luzzi G, Leslie A, Walker BD, Ndung'u T, Buus S, Price DA, Goulder PJ. CD8+ TCR Bias and Immunodominance in HIV-1 Infection. THE JOURNAL OF IMMUNOLOGY 2015; 194:5329-45. [PMID: 25911754 DOI: 10.4049/jimmunol.1400854] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 02/25/2015] [Indexed: 12/25/2022]
Abstract
Immunodominance describes a phenomenon whereby the immune system consistently targets only a fraction of the available Ag pool derived from a given pathogen. In the case of CD8(+) T cells, these constrained epitope-targeting patterns are linked to HLA class I expression and determine disease progression. Despite the biological importance of these predetermined response hierarchies, little is known about the factors that control immunodominance in vivo. In this study, we conducted an extensive analysis of CD8(+) T cell responses restricted by a single HLA class I molecule to evaluate the mechanisms that contribute to epitope-targeting frequency and antiviral efficacy in HIV-1 infection. A clear immunodominance hierarchy was observed across 20 epitopes restricted by HLA-B*42:01, which is highly prevalent in populations of African origin. Moreover, in line with previous studies, Gag-specific responses and targeting breadth were associated with lower viral load set-points. However, peptide-HLA-B*42:01 binding affinity and stability were not significantly linked with targeting frequencies. Instead, immunodominance correlated with epitope-specific usage of public TCRs, defined as amino acid residue-identical TRB sequences that occur in multiple individuals. Collectively, these results provide important insights into a potential link between shared TCR recruitment, immunodominance, and antiviral efficacy in a major human infection.
Collapse
Affiliation(s)
- Henrik N Kløverpris
- Department of Paediatrics, University of Oxford, Oxford OX1 3SY, United Kingdom; Department of International Health, Immunology, and Microbiology, University of Copenhagen, 2200-Copenhagen N, Denmark; KwaZulu-Natal Research Institute for Tuberculosis and HIV, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban 4001, South Africa;
| | - Reuben McGregor
- Department of Paediatrics, University of Oxford, Oxford OX1 3SY, United Kingdom
| | - James E McLaren
- Institute of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom
| | - Kristin Ladell
- Institute of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom
| | - Mikkel Harndahl
- Department of International Health, Immunology, and Microbiology, University of Copenhagen, 2200-Copenhagen N, Denmark
| | - Anette Stryhn
- Department of International Health, Immunology, and Microbiology, University of Copenhagen, 2200-Copenhagen N, Denmark
| | | | - Catherine Koofhethile
- HIV Pathogenesis Program, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban 4013, South Africa
| | - Bram Gerritsen
- Theoretical Biology Group, Utrecht University, 3584 CH Utrecht, the Netherlands
| | - Can Keşmir
- Theoretical Biology Group, Utrecht University, 3584 CH Utrecht, the Netherlands
| | - Fabian Chen
- Department of Sexual Health, Royal Berkshire Hospital, Reading RG1 5AN, United Kingdom
| | - Lynn Riddell
- Department of Genitourinary Medicine, Northamptonshire Healthcare National Health Service Trust, Northampton General Hospital, Cliftonville, Northampton NN1 5BD, United Kingdom
| | - Graz Luzzi
- Department of Sexual Health, Wycombe Hospital, High Wycombe HP11 2TT, United Kingdom
| | - Alasdair Leslie
- KwaZulu-Natal Research Institute for Tuberculosis and HIV, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban 4001, South Africa
| | - Bruce D Walker
- Ragon Institute of MGH, MIT, and Harvard, Boston, MA 02129; Howard Hughes Medical Institute, Chevy Chase, MD 20815; and
| | - Thumbi Ndung'u
- KwaZulu-Natal Research Institute for Tuberculosis and HIV, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban 4001, South Africa; HIV Pathogenesis Program, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban 4013, South Africa; Max Planck Institute for Infection Biology, D-10117 Berlin, Germany
| | - Søren Buus
- Department of International Health, Immunology, and Microbiology, University of Copenhagen, 2200-Copenhagen N, Denmark
| | - David A Price
- Institute of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom
| | - Philip J Goulder
- Department of Paediatrics, University of Oxford, Oxford OX1 3SY, United Kingdom
| |
Collapse
|
39
|
Srivastava RK, Utley A, Shrikant PA. Rapamycin: A rheostat for CD8(+) T-cell-mediated tumor therapy. Oncoimmunology 2014; 1:1189-1190. [PMID: 23170275 PMCID: PMC3494641 DOI: 10.4161/onci.20663] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Vaccines that generate Ag-specific CD8(+) T-cell responses of appropriate quality, magnitude and duration are highly desirable. The ability of mTOR to regulate CD8(+) T-cell functional differentiation must be exploited for clinical benefit. In a recent paper, we report that varying the regimen of rapamycin administration regulates viral vaccine-induced CD8(+) T-cell responses for tumor immunity. These observations validate the use of rapamycin in vaccination strategies and demonstrate the efficacy of memory CD8(+) T-cell responses for tumor immunity.
Collapse
|
40
|
Ferreira M, Veiga-Fernandes H. Pre-birth world and the development of the immune system: mum's diet affects our adult health: new insight on how the diet during pregnancy permanently influences offspring health and immune fitness. Bioessays 2014; 36:1213-20. [PMID: 25382781 DOI: 10.1002/bies.201400115] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Secondary lymphoid organs form in utero through an inherited and well-established developmental program. However, maternal non-heritable features can have a major impact on the gene expression of the embryo, hence influencing the future health of the offspring. Recently, maternal retinoids were shown to regulate the formation of immune structures, shedding light on the role of maternal nutrition in the genetic signature of emergent immune cells. Here we highlight evidence showing how the maternal diet influences the establishment of the immune system, and we also discuss how unbalanced maternal diets may set the response to infection and vaccination in the progeny.
Collapse
Affiliation(s)
- Manuela Ferreira
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Lisboa, Portugal
| | | |
Collapse
|
41
|
Junttila N, Lévêque N, Magnius L, Kabue J, Muyembe-Tamfum JJ, Maslin J, Lina B, Norder H. Complete coding regions of the prototypes enterovirus B93 and C95: Phylogenetic analyses of the P1 and P3 regions of EV-B and EV-C strains. J Med Virol 2014; 87:485-97. [DOI: 10.1002/jmv.24062] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/01/2014] [Indexed: 01/30/2023]
Affiliation(s)
- N. Junttila
- MTC; Karolinska Institutet; Stockholm Sweden
| | - N. Lévêque
- Clinical and Molecular Virology Unit; University Hospital Faculty of Medicine; Reims France
- Laboratory of Virology, National Enterovirus Laboratory; Hospices Civils de Lyon; France
| | | | - J.P. Kabue
- National Institute of Biomedical Research; Kinshasa, Democratic Republic of the Congo
| | - J. J. Muyembe-Tamfum
- National Institute of Biomedical Research; Kinshasa, Democratic Republic of the Congo
| | - J. Maslin
- Department of Biology; Saint-Anne Military Hospital; Toulon France
| | - B. Lina
- Laboratory of Virology, National Enterovirus Laboratory; Hospices Civils de Lyon; France
| | - H. Norder
- MTC; Karolinska Institutet; Stockholm Sweden
- Department of Infectious Diseases/Section of Clinical Virology; Institute of Biomedicine; University of Gothenburg; Gothenburg Sweden
| |
Collapse
|
42
|
Zhang X, Jia J, Dong J, Yu F, Ma N, Li M, Liu X, Liu W, Li T, Liu D. HLA-DQ polymorphisms with HBV infection: different outcomes upon infection and prognosis to lamivudine therapy. J Viral Hepat 2014; 21:491-8. [PMID: 24750255 DOI: 10.1111/jvh.12159] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Two recent genome-wide studies showed that the single-nucleotide polymorphisms in the HLA-DQ region (rs2856718 and rs9275572) were associated with chronic hepatitis B virus infection and chronic hepatitis C virus-associated hepatocellular carcinoma in Japanese patients. We tested the effects of the two single-nucleotide polymorphisms for all major HBV outcomes and lamivudine treatment in Han Chinese. A total of 1649 samples were enrolled, and peripheral blood samples were collected in this study. The single-nucleotide polymorphisms in the HLA-DQ region were genotyped using matrix-assisted laser desorption/ionization time of flight mass spectrometry. Our study demonstrated the clear relevance of HLA-DQ rs2856718 and rs9275572 with HBV susceptibility, natural clearance and HBV-associated HCC. HLA-DQ rs2856718G and rs9275572A were strongly associated with decreased risk of chronic HBV infection (odds ratio = 0.641; P = 2.64 × 10(-4) ; odds ratio = 0.627, P = 7.22 × 10(-5) ) and HBV natural clearance (odds ratio = 0.610; P = 4.80 × 10(-4) ; odds ratio = 0.714, P = 0.013). Moreover, rs9275572A was also associated with development of cirrhosis and hepatocellular carcinoma (odds ratio = 0.632, P = 0.008). In addition, we showed for the first time to our knowledge that rs9275572 was a predictor for lamivudine therapy (viral response: odds ratio = 2.599, P = 4.43 × 10(-4) ; biochemical response: odds ratio = 2.279, P = 4.23 × 10(-4) ). Our study suggested that HLA-DQ loci were associated with both HBV clearance and HBV-related diseases and outcomes of lamivudine treatment in Han Chinese.
Collapse
Affiliation(s)
- X Zhang
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, Shi Jiazhuang, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Perez-Shibayama C, Gil-Cruz C, Pastelin-Palacios R, Cervantes-Barragan L, Hisaki E, Chai Q, Onder L, Scandella E, Regen T, Waisman A, Isibasi A, Lopez-Macias C, Ludewig B. IFN-γ-producing CD4+ T cells promote generation of protective germinal center-derived IgM+ B cell memory against Salmonella Typhi. THE JOURNAL OF IMMUNOLOGY 2014; 192:5192-200. [PMID: 24778443 DOI: 10.4049/jimmunol.1302526] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Abs play a significant role in protection against the intracellular bacterium Salmonella Typhi. In this article, we investigated how long-term protective IgM responses can be elicited by a S. Typhi outer-membrane protein C- and F-based subunit vaccine (porins). We found that repeated Ag exposure promoted a CD4(+) T cell-dependent germinal center reaction that generated mutated IgM-producing B cells and was accompanied by a strong expansion of IFN-γ-secreting T follicular helper cells. Genetic ablation of individual cytokine receptors revealed that both IFN-γ and IL-17 are required for optimal germinal center reactions and production of porin-specific memory IgM(+) B cells. However, more profound reduction of porin-specific IgM B cell responses in the absence of IFN-γR signaling indicated that this cytokine plays a dominant role. Importantly, mutated IgM mAbs against porins exhibited bactericidal capacity and efficiently augmented S. Typhi clearance. In conclusion, repeated vaccination with S. Typhi porins programs type I T follicular helper cell responses that contribute to the diversification of B cell memory and promote the generation of protective IgM Abs.
Collapse
Affiliation(s)
- Christian Perez-Shibayama
- Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland; Medical Research Unit on Immunochemistry, Specialties Hospital, National Medical Centre "Siglo XXI," Mexican Social Security Institute, Mexico City, C.P. 06020 Mexico
| | - Cristina Gil-Cruz
- Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland; Medical Research Unit on Immunochemistry, Specialties Hospital, National Medical Centre "Siglo XXI," Mexican Social Security Institute, Mexico City, C.P. 06020 Mexico
| | | | - Luisa Cervantes-Barragan
- Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland; Department of Pathology, Washington University School of Medicine, St. Louis, MO 63130; Department of Immunology, Washington University School of Medicine, St. Louis, MO 63130; and
| | - Emiliano Hisaki
- Medical Research Unit on Immunochemistry, Specialties Hospital, National Medical Centre "Siglo XXI," Mexican Social Security Institute, Mexico City, C.P. 06020 Mexico
| | - Qian Chai
- Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland
| | - Lucas Onder
- Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland
| | - Elke Scandella
- Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland
| | - Tommy Regen
- Institute for Molecular Medicine, University of Mainz, D-55131 Mainz, Germany
| | - Ari Waisman
- Institute for Molecular Medicine, University of Mainz, D-55131 Mainz, Germany
| | - Armando Isibasi
- Medical Research Unit on Immunochemistry, Specialties Hospital, National Medical Centre "Siglo XXI," Mexican Social Security Institute, Mexico City, C.P. 06020 Mexico
| | - Constantino Lopez-Macias
- Medical Research Unit on Immunochemistry, Specialties Hospital, National Medical Centre "Siglo XXI," Mexican Social Security Institute, Mexico City, C.P. 06020 Mexico
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland;
| |
Collapse
|
44
|
Recognition of Foreign Antigen and Foreign Major Histocompatibility Complex. Xenotransplantation 2014. [DOI: 10.1128/9781555818043.ch9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
45
|
Abstract
The peripheral T cell repertoire is sculpted from prototypic T cells in the thymus bearing randomly generated T cell receptors (TCR) and by a series of developmental and selection steps that remove cells that are unresponsive or overly reactive to self-peptide–MHC complexes. The challenge of understanding how the kinetics of T cell development and the statistics of the selection processes combine to provide a diverse but self-tolerant T cell repertoire has invited quantitative modeling approaches, which are reviewed here.
Collapse
Affiliation(s)
- Andrew J Yates
- Departments of Systems and Computational Biology, Microbiology and Immunology, Albert Einstein College of Medicine , New York, NY , USA
| |
Collapse
|
46
|
McMichael AJ, Haynes BF. Influenza vaccines: mTOR inhibition surprisingly leads to protection. Nat Immunol 2014; 14:1205-7. [PMID: 24240151 DOI: 10.1038/ni.2764] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
47
|
Abstract
γδ T cells, αβ T cells, and B cells are present together in all but the most primitive vertebrates, suggesting that each population contributes to host immune competence uniquely and that all three are necessary for maintaining immune competence. Functional and molecular analyses indicate that in infections, γδ T cells respond earlier than αβ T cells do and that they emerge late after pathogen numbers start to decline. Thus, these cells may be involved in both establishing and regulating the inflammatory response. Moreover, γδ T cells and αβ T cells are clearly distinct in their antigen recognition and activation requirements as well as in the development of their antigen-specific repertoire and effector function. These aspects allow γδ T cells to occupy unique temporal and functional niches in host immune defense. We review these and other advances in γδ T cell biology in the context of their being the major initial IL-17 producers in acute infection.
Collapse
|
48
|
Salisbury EM, Game DS, Lechler RI. Transplantation tolerance. Pediatr Nephrol 2014; 29:2263-72. [PMID: 24213880 PMCID: PMC4212135 DOI: 10.1007/s00467-013-2659-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 10/01/2013] [Accepted: 10/04/2013] [Indexed: 01/26/2023]
Abstract
Although transplantation has been a standard medical practice for decades, marked morbidity from the use of immunosuppressive drugs and poor long-term graft survival remain important limitations in the field. Since the first solid organ transplant between the Herrick twins in 1954, transplantation immunology has sought to move away from harmful, broad-spectrum immunosuppressive regimens that carry with them the long-term risk of potentially life-threatening opportunistic infections, cardiovascular disease, and malignancy, as well as graft toxicity and loss, towards tolerogenic strategies that promote long-term graft survival. Reports of "transplant tolerance" in kidney and liver allograft recipients whose immunosuppressive drugs were discontinued for medical or non-compliant reasons, together with results from experimental models of transplantation, provide the proof-of-principle that achieving tolerance in organ transplantation is fundamentally possible. However, translating the reconstitution of immune tolerance into the clinical setting is a daunting challenge fraught with the complexities of multiple interacting mechanisms overlaid on a background of variation in disease. In this article, we explore the basic science underlying mechanisms of tolerance and review the latest clinical advances in the quest for transplantation tolerance.
Collapse
Affiliation(s)
- Emma M. Salisbury
- Section of Immunobiology, Division of Immunology and Inflammation, Department of Medicine, Faculty of Medicine, Imperial College London, Sir Alexander Fleming Building, South Kensington Campus, Exhibition Road, London, SW7 2AZ UK
| | - David S. Game
- Department of Renal Medicine, Guy’s and St. Thomas’ NHS Foundation Trust, Guy’s Hospital, Great Maze Pond, London, SE1 9RT UK
| | - Robert I. Lechler
- King’s Health Partners Academic Health Sciences Centre, King’s College London, London, WC2R 2LS UK
| |
Collapse
|
49
|
Lee K, Takenaka H, Yoneda Y, Goto T, Sano K, Nakanishi M, Eguchi A, Okada M, Tashiro J, Sakurai K, Kubota T, Yoshida R. Differential Susceptibility of Cells Expressing Allogeneic MHC or Viral Antigen to Killing by Antigen-Specific CTL. Microbiol Immunol 2013; 48:15-25. [PMID: 14734854 DOI: 10.1111/j.1348-0421.2004.tb03483.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
CD8(+) cytotoxic T lymphocytes (CTLs) generated by immunization with allogeneic cells or viral infection are able to lyse allogeneic or virally infected in vitro cells (e.g., lymphoma and mastocytoma). In contrast, it is reported that CD8(+) T cells are not essential for allograft rejection (e.g., heart and skin), and that clearance of influenza or the Sendai virus from virus-infected respiratory epithelium is normal or only slightly delayed after a primary viral challenge of CD8-knockout mice. To address this controversy, we generated H-2(d)-specific CD8(+) CTLs by a mixed lymphocyte culture and examined the susceptibility of a panel of H-2(d) cells to CTL lysis. KLN205 squamous cell carcinoma, Meth A fibrosarcoma, and BALB/c skin components were found to be resistant to CTL-mediated lysis. This resistance did not appear to be related to a reduced expression of MHC class I molecules, and all these cells could block the recognition of H-2(d) targets by CTLs in cold target inhibition assays. We extended our observation by persistently infecting the same panel of cell lines with defective-interfering Sendai virus particles. The Meth A and KLN205 lines infected with a variant Sendai virus were resistant to lysis by Sendai virus-specific CTLs. The Sendai virus-infected Meth A and KLN205 lines were able to block the lysis of Sendai virus-infected targets by CTLs in cold target inhibition assays. Taken together, these results suggest that not all in vivo tissues may be sensitive to CTL lysis.
Collapse
Affiliation(s)
- Koutetsu Lee
- Department of Physiology, Osaka Medical College, Takatsuki, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Bocharov G, Luzyanina T, Cupovic J, Ludewig B. Asymmetry of Cell Division in CFSE-Based Lymphocyte Proliferation Analysis. Front Immunol 2013; 4:264. [PMID: 24032033 PMCID: PMC3759284 DOI: 10.3389/fimmu.2013.00264] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 08/19/2013] [Indexed: 12/31/2022] Open
Abstract
Flow cytometry-based analysis of lymphocyte division using carboxyfluorescein succinimidyl ester (CFSE) dye dilution permits acquisition of data describing cellular proliferation and differentiation. For example, CFSE histogram data enable quantitative insight into cellular turnover rates by applying mathematical models and parameter estimation techniques. Several mathematical models have been developed using different types of deterministic or stochastic approaches. However, analysis of CFSE proliferation assays is based on the premise that the label is halved in the two daughter cells. Importantly, asymmetry of protein distribution in lymphocyte division is a basic biological feature of cell division with the degree of the asymmetry depending on various factors. Here, we review the recent literature on asymmetric lymphocyte division and CFSE-based lymphocyte proliferation analysis. We suggest that division- and label-structured mathematical models describing CFSE-based cell proliferation should take into account asymmetry and time-lag in cell proliferation. Utilization of improved modeling algorithms will permit straightforward quantification of essential parameters describing the performance of activated lymphocytes.
Collapse
Affiliation(s)
- Gennady Bocharov
- Institute of Numerical Mathematics, Russian Academy of Sciences , Moscow , Russia
| | | | | | | |
Collapse
|