1
|
Effect pf Fermented Rapeseed Meal as a Feed Component on the Redox and Immune System of Pregnant Sows and their Offspring. ANNALS OF ANIMAL SCIENCE 2022. [DOI: 10.2478/aoas-2021-0034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Abstract
The aim of the study was to assess the effect of dried fermented rapeseed meal (FRSM) in diets for sows on blood redox and immunological parameters, taking into account the physiological period (pregnancy or lactation) and age (primiparous vs multiparous sows). The experiment also aimed to determine how FRSM administered to pregnant sows, affects the antioxidant and immune systems of piglets. The animals were divided into 4 groups of 15 animals each. Control groups of primiparous (CG) and multiparous sows (CS) received a standard diet for pregnant or lactating sows. Experimental groups of primiparous (EG) and multiparous sows (ES) received feed with a 4% share of FRSM in place of soybean meal up to day 100 of gestation, 9% share of FRSM from day 100 of gestation to day 7 of lactation, and then again 4% share of FRSM until the end of lactation. In the blood plasma of pregnant sows fed diet with FRSM addition, higher FRAP value and vitamin C, uric acid (UA), immunoglobulin IgG content, lymphocytes (LYM) count and a lower content of malondialdehyde (MDA), lipid hydroperoxides (LOH), immunoglobulin IgM was noted than in the blood plasma of control sows. Both primiparous and multiparous lactation sows whose feed included FRSM had higher catalase (CAT) activity, higher FRAP, vitamin C, immunoglobulin IgG and IL-6 content, and lower UA content than the control sows. Piglets born to sows fed diet with FRSM addition had significantly higher FRAP values, vitamin C, IgG, and IL-6 content and white blood cells (WBC) count and lower MDA and UA content in the blood plasma than piglets born to sows from control group. Multiparous sows compared to primiparous sows had higher CAT activity, and higher vitamin C, LOH, creatinine (CREAT), and IgM content. Elevated FRAP, and CREAT levels and reduced MDA content were also observed in the plasma of the multiparous sows compared to primiparous sows during lactation. Multiparous lactation sows compared to primiparous sows had lower WBC count, and IgG and IgM content. Piglets born to multiparous sows had higher FRAP values, LOH content and IgA content while lower MDA content compared to piglets born to primiparous sows. The inclusion of dried fermented rapeseed meal in feed for sows significantly stimulates antioxidant processes in primiparous and multiparous sows and in their piglets. The inclusion of dried fermented rapeseed meal in the diet of sows stimulates antioxidant processes in primiparous and multiparous sows and in their piglets. This is responsible for stimulation of the immune system (increased LYM counts and IgG titres in the blood plasma). The improved antioxidant status in the plasma suggests that dried fermented rapeseed meal stimulated the immune system of pregnant and lactating sows and their newborn offspring.
Collapse
|
2
|
Hajavi J, Esmaeili SA, Varasteh AR, Vazini H, Atabati H, Mardani F, Momtazi-Borojeni AA, Hashemi M, Sankian M, Sahebkar A. The immunomodulatory role of probiotics in allergy therapy. J Cell Physiol 2018; 234:2386-2398. [PMID: 30192002 DOI: 10.1002/jcp.27263] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 07/24/2018] [Indexed: 12/14/2022]
Abstract
The increased incidence of allergic disorders may be the result of a relative fall in microbial induction in the intestinal immune system during infancy and early childhood. Probiotics have recently been proposed as viable microorganisms for the prevention and treatment of specific allergic diseases. Different mechanisms have been considered for this probiotic property, such as generation of cytokines from activated pro-T-helper type 1 after bacterial contact. However, the effects of its immunomodulatory potential require validation for clinical applications. This review will focus on the currently available data on the benefits of probiotics in allergy disease.
Collapse
Affiliation(s)
- Jafar Hajavi
- Department of Basic Sciences, Faculty of Allied Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Seyed-Alireza Esmaeili
- Immunology Research Center, BuAli Research Institute, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Abdol-Reza Varasteh
- Allergy Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Vazini
- Nursing Department, Basic Sciences Faculty, Hamedan Branch, Islamic Azad University, Hamedan, Iran
| | - Hadi Atabati
- Leishmaniasis Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Fatemeh Mardani
- Immunology Research Center, BuAli Research Institute, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir A Momtazi-Borojeni
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Biotechnology, Nanotechnology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Hashemi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mojtaba Sankian
- Immunology Research Center, BuAli Research Institute, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
3
|
Tolerogenic Dendritic Cells on Transplantation: Immunotherapy Based on Second Signal Blockage. J Immunol Res 2015; 2015:856707. [PMID: 26543876 PMCID: PMC4620289 DOI: 10.1155/2015/856707] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 06/23/2015] [Accepted: 06/29/2015] [Indexed: 12/16/2022] Open
Abstract
Dendritic cells (DCs), the most important professional antigen-presenting cells (APC), play crucial role in both immunity and tolerance. It is well known that DCs are able to mount immune responses against foreign antigens and simultaneously tolerate self-antigens. Since DCs can be modulated depending on the surrounding microenvironment, they can act as a bridge between innate and adaptive immunity. However, the mechanisms that support this dual role are not entirely clear. Recent studies have shown that DCs can be manipulated ex vivo in order to trigger their tolerogenic profile, what can be a tool to be used in clinical trials aiming the treatment of various diseases and the prevention of transplant rejection. In this sense, the blockage of costimulatory molecules on DC, in the attempt of inhibiting the second signal in the immunological synapse, can be considered as one of the main strategies under development. This review brings an update on current therapies using tolerogenic dendritic cells modulated with costimulatory blockers with the aim of reducing transplant rejection. However, although there are current clinical trials using tolerogenic DC to treat allograft rejection, the actual challenge is to modulate these cells in order to maintain a permanent tolerogenic profile.
Collapse
|
4
|
Baker MP, Reynolds HM, Lumicisi B, Bryson CJ. Immunogenicity of protein therapeutics: The key causes, consequences and challenges. SELF NONSELF 2014; 1:314-322. [PMID: 21487506 DOI: 10.4161/self.1.4.13904] [Citation(s) in RCA: 269] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/10/2010] [Revised: 09/20/2010] [Accepted: 10/07/2010] [Indexed: 12/15/2022]
Abstract
The immunogenicity of protein therapeutics has so far proven to be difficult to predict in patients, with many biologics inducing undesirable immune responses directed towards the therapeutic resulting in reduced efficacy, anaphylaxis and occasionally life threatening autoimmunity. The most common effect of administrating an immunogenic protein therapeutic is the development of a high affinity anti-therapeutic antibody response. Furthermore, it is clear from clinical studies that protein therapeutics derived from endogenous human proteins are capable of stimulating undesirable immune responses in patients, and as a consequence, the prediction and reduction of immunogenicity has been the focus of intense research. This review will outline the principle causes of the immunogenicity in protein therapeutics, and describe the development of pre-clinical models that can be used to aid in the prediction of the immunogenic potential of novel protein therapeutics prior to administration in man.
Collapse
Affiliation(s)
- Matthew P Baker
- Antitope Ltd.; Babraham Research Campus; Babraham, Cambridge UK
| | | | | | | |
Collapse
|
5
|
Sathish JG, Sethu S, Bielsky MC, de Haan L, French NS, Govindappa K, Green J, Griffiths CEM, Holgate S, Jones D, Kimber I, Moggs J, Naisbitt DJ, Pirmohamed M, Reichmann G, Sims J, Subramanyam M, Todd MD, Van Der Laan JW, Weaver RJ, Park BK. Challenges and approaches for the development of safer immunomodulatory biologics. Nat Rev Drug Discov 2013; 12:306-24. [PMID: 23535934 PMCID: PMC7097261 DOI: 10.1038/nrd3974] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Immunomodulatory biologics are a class of biotechnology-derived therapeutic products that are designed to engage immune-relevant targets and are indicated in the treatment and management of a range of diseases, including immune-mediated inflammatory diseases and malignancies. Despite their high specificity and therapeutic advantages, immmunomodulatory biologics have been associated with adverse reactions such as serious infections, malignancies and cytokine release syndrome, which arise owing to the on-target or exaggerated pharmacological effects of these drugs. Immunogenicity resulting in the generation of antidrug antibodies is another unwanted effect that leads to loss of efficacy and — rarely — hypersensitivity reactions. For some adverse reactions, mitigating and preventive strategies are in place, such as stratifying patients on the basis of responsiveness to therapy and the risk of developing adverse reactions. These strategies depend on the availability of robust biomarkers for therapeutic efficacy and the risk of adverse reactions: for example, seropositivity for John Cunningham virus is a risk factor for progressive multifocal leukoencephalopathy. The development of effective biomarkers will greatly aid these strategies. The development and design of safer immunomodulatory biologics is reliant on a detailed understanding of the nature of the disease, target biology, the interaction of the target with the immunomodulatory biologic and the inherent properties of the biologic that elicit unwanted effects. The availability of in vitro and in vivo models that can be used to predict adverse reactions associated with immunomodulatory biologics is central to the development of safer immunomodulatory biologics. Some progress has been made in developing in vitro and in silico tests for predicting cytokine release syndrome and immunogenicity, but there is still a lack of models for effectively predicting infections and malignancies. Two pathways can be followed in designing and developing safer immunomodulatory biologics. The first pathway involves generating a biologic that engages an alternative target or mechanism to produce the desired pharmacodynamic effect without the associated adverse reaction, and is followed when the adverse reaction cannot be dissociated from the target biology. The second pathway involves redesigning the biologic to 'engineer out' components within the biologic structure that trigger adverse effects or to alter the nature of the target–biologic interactions.
Owing to their specificity, immunomodulatory biologics generally have better safety profiles than small-molecule drugs. However, adverse effects such as an increased risk of infections or cytokine release syndrome are of concern. Here, Park and colleagues discuss the current strategies used to predict and mitigate these adverse effects and consider how they can be used to inform the development of safer immunomodulatory biologics. Immunomodulatory biologics, which render their therapeutic effects by modulating or harnessing immune responses, have proven their therapeutic utility in several complex conditions including cancer and autoimmune diseases. However, unwanted adverse reactions — including serious infections, malignancy, cytokine release syndrome, anaphylaxis and hypersensitivity as well as immunogenicity — pose a challenge to the development of new (and safer) immunomodulatory biologics. In this article, we assess the safety issues associated with immunomodulatory biologics and discuss the current approaches for predicting and mitigating adverse reactions associated with their use. We also outline how these approaches can inform the development of safer immunomodulatory biologics.
Collapse
Affiliation(s)
- Jean G Sathish
- MRC Centre for Drug Safety Science and Institute of Translational Medicine, Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool L69 3GE, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Immunogenicity to biologics: mechanisms, prediction and reduction. Arch Immunol Ther Exp (Warsz) 2012; 60:331-44. [PMID: 22930363 DOI: 10.1007/s00005-012-0189-7] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 05/11/2012] [Indexed: 01/06/2023]
Abstract
Currently, there is a significant rise in the development and clinical use of a unique class of pharmaceuticals termed as Biopharmaceuticals or Biologics, in the management of a range of disease conditions with, remarkable therapeutic benefits. However, there is an equally growing concern regarding development of adverse effects like immunogenicity in the form of anti-drug antibodies (ADA) production and hypersensitivity. Immunogenicity to biologics represents a significant hurdle in the continuing therapy of patients in a number of disease settings. Efforts focussed on the identification of factors that contribute towards the onset of immunogenic response to biologics have led to reductions in the incidence of immunogenicity. An in-depth understanding of the cellular and molecular mechanism underpinning immunogenic responses will likely improve the safety profile of biologics. This review addresses the mechanistic basis of ADA generation to biologics, with emphasis on the role of antigen processing and presentation in this process. The article also addresses the potential contribution of complement system in augmenting or modulating this response. Identifying specific factors that influences processing and presentation of biologic-derived antigens in different genotype and disease background may offer additional options for intervention in the immunogenic process and consequently, the management of immunogenicity to biologics.
Collapse
|
7
|
Geem D, Medina-Contreras O, Kim W, Huang CS, Denning TL. Isolation and characterization of dendritic cells and macrophages from the mouse intestine. J Vis Exp 2012:e4040. [PMID: 22644046 DOI: 10.3791/4040] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Within the intestine reside unique populations of innate and adaptive immune cells that are involved in promoting tolerance towards commensal flora and food antigens while concomitantly remaining poised to mount inflammatory responses toward invasive pathogens. Antigen presenting cells, particularly DCs and macrophages, play critical roles in maintaining intestinal immune homeostasis via their ability to sense and appropriately respond to the microbiota. Efficient isolation of intestinal DCs and macrophages is a critical step in characterizing the phenotype and function of these cells. While many effective methods of isolating intestinal immune cells, including DCs and macrophages, have been described, many rely upon long digestions times that may negatively influence cell surface antigen expression, cell viability, and/or cell yield. Here, we detail a methodology for the rapid isolation of large numbers of viable, intestinal DCs and macrophages. Phenotypic characterization of intestinal DCs and macrophages is carried out by directly staining isolated intestinal cells with specific fluorescence-labeled monoclonal antibodies for multi-color flow cytometric analysis. Furthermore, highly pure DC and macrophage populations are isolated for functional studies utilizing CD11c and CD11b magnetic-activated cell sorting beads followed by cell sorting.
Collapse
Affiliation(s)
- Duke Geem
- Department of Pediatrics, Emory University
| | | | | | | | | |
Collapse
|
8
|
Li H, Liu X, Li Y, Hua Y, Zhi D, Pang G. Effects of the polysaccharide from Pholiota nameko on human cytokine network in serum. Int J Biol Macromol 2012; 50:164-70. [DOI: 10.1016/j.ijbiomac.2011.10.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Revised: 10/01/2011] [Accepted: 10/17/2011] [Indexed: 11/24/2022]
|
9
|
CD4 T cell depletion at the cervix during HIV infection is associated with accumulation of terminally differentiated T cells. J Virol 2011; 85:13333-41. [PMID: 21994461 DOI: 10.1128/jvi.05671-11] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
In blood, the accumulation of terminally differentiated (TD) T cells during HIV infection is associated with CD4 T cell loss and HIV disease progression. Here, we investigated the maintenance and functional characteristics of memory T cells at the cervix. We found that CD4 T cell depletion at the cervix mirrors CD4 depletion in blood. In all women, depletion of CD4 T cells at the cervix was associated with significant reductions in CD45RA- CCR7+ (central memory [CM]) T cells and the accumulation of CD45RA+ CCR7- (TD T cells). We determined whether inflammation in the genital tract was associated with the local differentiation of T cells at the cervix. In uninfected women, genital tract inflammation was associated with the accumulation of CD45RA- CCR7+ CM CD4 T cells and reduced frequencies of CD45RA+ CCR7- TD cells at the cervix. This finding may reflect the fact that, in the absence of HIV infection, TD T cells may be slowly lost in the presence of genital inflammation, while CD45RA- CCR7+ CM T cells are recruited to replenish the diminishing CD4 T cell pool. Following global stimulation with phorbol myristate acetate (PMA)-ionomycin, we noted a significant interleukin 2 (IL-2) deficit in both cervical and blood CD4 T cells from HIV-infected women compared to uninfected women, while gamma interferon (IFN-γ) production was similar, irrespective of HIV status. Few HIV-infected women had detectable IFN-γ and IL-2 HIV-specific T cell responses at the cervix, and these responses were significantly lower in magnitude than the corresponding responses in blood. These data suggest that CD4 depletion was associated with the accumulation of terminally differentiated T cell phenotypes at the cervical mucosa defective in their ability to produce IL-2. CD4 depletion and compromised immunity at the cervix may be accompanied by progressive decline of central memory-like T cells and development of T cells toward terminally differentiated phenotypes.
Collapse
|
10
|
Fousteri G, Dave A, Morin B, Omid S, Croft M, von Herrath MG. Nasal cardiac myosin peptide treatment and OX40 blockade protect mice from acute and chronic virally-induced myocarditis. J Autoimmun 2011; 36:210-20. [PMID: 21333491 PMCID: PMC3079777 DOI: 10.1016/j.jaut.2011.01.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2010] [Revised: 01/17/2011] [Accepted: 01/18/2011] [Indexed: 02/05/2023]
Abstract
Myocarditis poses a severe health problem, can lead to dilated cardiomyopathy (DCM) and death, and is thought to be triggered by infections. Enteroviruses such as Coxsackie virus B3 (CVB3) have been implicated as a culprit, since they can cause acute and chronic heart disease in susceptible mice. CVB was detected in human cardiac myocytes in some cases, whereas acute CVB infection was thought to have caused death. Here we studied, whether nasal administration of cardiac myosin (CM) major histocompatibility class (MHC) II peptides CM₉₄₇-₉₆₀ and CM₇₃₅-₇₄₇ and OX40 blockade would be able to ameliorate immunopathology and heart disease in BALB/C mice infected with CVB3. We found that nasal CM-peptide prophylactic treatment significantly reduced myocarditis and mortality by enhancing Treg and IL-10 induction and that blockade of OX40 signaling could reduce heart inflammation when administered late during pathogenesis. Altogether, these results chart the way for novel prevention and intervention strategies for viral myocarditis.
Collapse
Affiliation(s)
| | - Amy Dave
- Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, California, United States, 92037
| | - Bret Morin
- Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, California, United States, 92037
| | - Shaida Omid
- Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, California, United States, 92037
| | - Michael Croft
- Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, California, United States, 92037
| | - Matthias G von Herrath
- Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, California, United States, 92037
| |
Collapse
|
11
|
van de Pol MA, Lutter R, Smids BS, Weersink EJM, van der Zee JS. Synbiotics reduce allergen-induced T-helper 2 response and improve peak expiratory flow in allergic asthmatics. Allergy 2011; 66:39-47. [PMID: 20716319 DOI: 10.1111/j.1398-9995.2010.02454.x] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Previous studies suggest that pre/probiotics can be used in the prevention and treatment of early allergic disease in newborns and young children. OBJECTIVE To determine the effect of treatment with synbiotics (90% short-chain galacto-oligosaccharides, 10% long-chain fructo-oligosaccharides: Immunofortis(®) and Bifidobacterium breve M-16V) on allergic responses in adults with established allergic asthma. Primary outcome was allergen-induced bronchial inflammation as represented by eosinophil counts. METHODS Twenty-nine patients with asthma and house dust mite (HDM) allergy were randomized in a double-blind, parallel design to receive placebo or synbiotics for 4 weeks. At study entry and after treatment, a bronchial allergen challenge with HDM was performed, followed by lung function tests, collection of blood (in/ex vivo IL-5) and induced sputum (inflammatory parameters). During treatment, a diary was kept with peak expiratory flow (PEF) and asthma scores. RESULTS Treatment did not affect the allergen-induced increase in sputum eosinophils at 6 and 24 h after challenge. Likewise, other parameters for bronchial inflammation and early and late changes in lung function did not differ upon treatment. Both the morning and evening PEF, however, significantly increased during synbiotics treatment (morning P = 0.003, evening P = 0.011). Also, the increase in serum IL-5 after allergen challenge was significantly inhibited by synbiotics (P = 0.034), as was ex vivo allergen-induced Th2-cytokine (IL-5 and IL-4+ IL-13) production by PBMCs (P = 0.046). In vivo (24 h) and ex vivo IL-5 production were associated. CONCLUSION Four-week treatment with synbiotics had no effect on bronchial inflammation and LAR, but did significantly reduce systemic production of Th2-cytokines after allergen challenge and improved PEF.
Collapse
Affiliation(s)
- M A van de Pol
- Department of Pulmonology, Academic Medical Center (University of Amsterdam), Amsterdam, the Netherlands
| | | | | | | | | |
Collapse
|
12
|
Westendorf AM, Fleissner D, Hansen W, Buer J. T cells, dendritic cells and epithelial cells in intestinal homeostasis. Int J Med Microbiol 2009; 300:11-8. [PMID: 19781991 DOI: 10.1016/j.ijmm.2009.08.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The mucosal immune system of the intestinal tract is continuously exposed to both potential pathogens and beneficial commensal microorganism. A variety of mechanisms contribute to the ability of the gut to either react or remain tolerant to antigen present in the intestinal lumen. Antigens of the gut commensals are not simply ignored, but rather trigger an active immunosuppressive process, which prevents the outcome of immunopathology. The aim of this review is to provide an update on the mechanism of intestinal homeostasis, with particular focus on the complex crosstalk between T cells, dendritic cells and intestinal epithelial cells.
Collapse
Affiliation(s)
- Astrid M Westendorf
- Institute of Medical Microbiology, University Hospital Essen, Hufelandstrasse 55, D-45122 Essen, Germany.
| | | | | | | |
Collapse
|
13
|
Mason KL, Huffnagle GB. Control of mucosal polymicrobial populations by innate immunity. Cell Microbiol 2009; 11:1297-305. [DOI: 10.1111/j.1462-5822.2009.01347.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
14
|
Shang L, Thirunarayanan N, Viejo-Borbolla A, Martin AP, Bogunovic M, Marchesi F, Unkeless JC, Ho Y, Furtado GC, Alcami A, Merad M, Mayer L, Lira SA. Expression of the chemokine binding protein M3 promotes marked changes in the accumulation of specific leukocytes subsets within the intestine. Gastroenterology 2009; 137:1006-18, 1018.e1-3. [PMID: 19501588 PMCID: PMC2736321 DOI: 10.1053/j.gastro.2009.05.055] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2009] [Revised: 05/12/2009] [Accepted: 05/28/2009] [Indexed: 01/09/2023]
Abstract
BACKGROUND & AIMS Chemokines are small proteins that direct leukocyte trafficking under homeostatic and inflammatory conditions. We analyzed the differential expression of chemokines in distinct segments of the intestine and investigated the importance of chemokines for the distribution of leukocytes in the intestine during homeostatic and inflammatory conditions. METHODS We analyzed messenger RNA for all known chemokines in different segments of the gut by quantitative polymerase chain reaction. To study the effect of multiple-chemokine blockade in the gut, we generated transgenic mice that expressed the chemokine binding protein M3 in the intestine (V-M3 mice). We used flow cytometry to evaluate the changes in the numbers of leukocytes. RESULTS We observed distinct chemokine expression profiles in the 6 segments of the gut. Some chemokines were expressed throughout the intestine (CCL28, CCL6, CXCL16, and CX3CL1), whereas others were expressed preferentially in the small (CCL25 and CCL5) or large intestine (CCL19, CCL21, and CXCL5). Expression of the chemokine blocker M3 in intestinal epithelial cells resulted in reduced numbers of B and T cells in Peyer's patches, reduced numbers of intraepithelial CD8alphabeta(+)/TCRalphabeta(+) and CD8alphaalpha(+)/TCRalphabeta(+) T cells, and reduced numbers of lamina propria CD8(+) T cells. Strikingly, M3 expression markedly reduced the number of eosinophils and macrophages in the small and large intestines. Dextran sulfate sodium treatment of control mice led to marked changes in the expression of chemokines and in the number of myeloid cells in the colon. These cellular changes were significantly attenuated in the presence of M3. CONCLUSIONS Our study reveals a complex pattern of chemokine expression in the intestine and indicates that chemokines are critical for leukocyte accumulation in the intestine during homeostasis and inflammation.
Collapse
Affiliation(s)
- Limin Shang
- Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029
| | | | - Abel Viejo-Borbolla
- Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029
- Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid), Cantoblanco, Madrid, 28049, Spain
| | - Andrea P. Martin
- Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029
| | | | - Federica Marchesi
- Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029
| | - Jay C. Unkeless
- Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029
| | - Yin Ho
- Department of Medicine, University of Cambridge, Addenbrooke’s Hospital, Cambridge, CB2 2QQ, United Kingdom
| | - Glaucia C. Furtado
- Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029
| | - Antonio Alcami
- Department of Medicine, University of Cambridge, Addenbrooke’s Hospital, Cambridge, CB2 2QQ, United Kingdom
- Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid), Cantoblanco, Madrid, 28049, Spain
| | - Miriam Merad
- Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029
| | - Lloyd Mayer
- Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029
| | - Sergio A. Lira
- Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029
| |
Collapse
|
15
|
B-1 cells modulate oral tolerance in mice. Immunol Lett 2009; 124:63-9. [PMID: 19389426 DOI: 10.1016/j.imlet.2009.04.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2009] [Revised: 04/07/2009] [Accepted: 04/12/2009] [Indexed: 12/12/2022]
Abstract
Although the origin and functions of B-1 cells are controversial, they are considered as a cellular element of innate immunity due to their ability to produce natural autoantibodies of the IgM type. These antibodies are encoded by a relatively limited repertoire of V genes, and their resulting diversity is smaller than that produced by conventional B cells. B-1 cells constitute the larger fraction of B cells in the peritoneal cavity and migrate to non-specific inflammation sites. In addition, they contribute to the production of IgA antibodies in the intestinal lamina propria. It has been demonstrated that they participate in the induction and maintenance of peripheral tolerance. Herein, the participation of B-1 cells in inducing oral tolerance is evaluated. Unexpectedly, BALB/Xid mice, the animals deficient in B-1 cells, are not tolerized to OVA but instead are responsive to oral immunization. Conversely, BALB/c mice respond to oral tolerance to this antigen. We used these biological characteristics of these animals to investigate whether B-1 cells are involved in the induction of oral tolerance to OVA. Results show that B-1 cells from BALB/c mice, treated orally with OVA and adoptively transferred to BALB/Xid mice were able to suppress local hypersensitivity reaction and lymphoproliferative cellular response observed in BALB/Xid mice. These data demonstrate that B-1 cells have regulatory properties and are involved in the induction of oral tolerance.
Collapse
|
16
|
Abstract
Human noroviruses cause more than 90% of epidemic nonbacterial gastroenteritis. However, the role of B cells and antibody in the immune response to noroviruses is unclear. Previous studies have demonstrated that human norovirus specific antibody levels increase upon infection, but they may not be protective against infection. In this report, we used murine norovirus (MNV), an enteric norovirus, as a model to determine the importance of norovirus specific B cells and immune antibody in clearance of norovirus infection. We show here that mice genetically deficient in B cells failed to clear primary MNV infection as effectively as wild-type mice. In addition, adoptively transferred immune splenocytes derived from B-cell-deficient mice or antibody production-deficient mice were unable to efficiently clear persistent MNV infection in RAG1(-/-) mice. Further, adoptive transfer of either polyclonal anti-MNV serum or neutralizing anti-MNV monoclonal antibodies was sufficient to reduce the level of MNV infection both systemically and in the intestine. Together, these data demonstrate that antibody plays an important role in the clearance of MNV and that immunoglobulin G anti-norovirus antibody can play an important role in clearing mucosal infection.
Collapse
|
17
|
Bracklein T, Theise S, Metzler A, Spiess BM, Richter M. Activity of feline interferon-omega after ocular or oral administration in cats as indicated by Mx protein expression in conjunctival and white blood cells. Am J Vet Res 2007; 67:1025-32. [PMID: 16740097 DOI: 10.2460/ajvr.67.6.1025] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To assess the biological response to recombinant feline interferon-omega (rFeIFN-omega) following ocular or oral administration in cats via estimation of Mx protein expression in conjunctival cells (CCs) and WBCs. ANIMALS 10 specific pathogen-free cats. PROCEDURES In multiple single-dose drug experiments, each cat received various concentrations of rFeIFN-omega administered topically into both eyes (50 to 10,000 U/eye) and orally (200 to 20,000 units). The same cats received saline (0.9% NaCl) solution topically and orally as control treatments. The CCs and WBCs were collected prior to treatment (day 0), on day 1, and every third or seventh day thereafter until samples yielded negative results for Mx protein. Samples were examined for Mx protein expression via immunohistochemistry and immunoblotting procedures involving murine anti-Mx protein monoclonal antibody M143. RESULTS After topical application of 10,000 U of rFeIFN-omega/eye, CCs stained for Mx protein for a minimum of 7 days, whereas WBCs were positive for Mx protein for a minimum of 31 days. After topical application of lower concentrations, CCs did not express Mx protein, in contrast to WBCs, which stained for Mx protein at 1,000 units for at least 1 day. Following oral administration, Mx protein was expressed in WBCs at rFeIFN-omega concentrations as low as 200 units, whereas CCs did not stain for Mx protein at any concentration. CONCLUSIONS AND CLINICAL RELEVANCE Results indicate that Mx protein expression (a marker of the biological response to rFeIFN-omega) in CCs and WBCs of rFeIFN-omega-treated cats depends on the dose of rFeIFN-omega, site of administration, and cell type.
Collapse
Affiliation(s)
- Thorsten Bracklein
- Ophthalmology Unit, Department of Small Animals, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 260, 8057 Zurich, Switzerland
| | | | | | | | | |
Collapse
|
18
|
Castellaneta A, Abe M, Morelli AE, Thomson AW. Identification and characterization of intestinal Peyer's patch interferon-alpha producing (plasmacytoid) dendritic cells. Hum Immunol 2005; 65:104-13. [PMID: 14969765 DOI: 10.1016/j.humimm.2003.10.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2003] [Revised: 10/10/2003] [Accepted: 10/13/2003] [Indexed: 11/18/2022]
Abstract
Recently, a subset of murine dendritic cells (DC) has been identified that resembles human plasmacytoid (pDC) the principal interferon-alpha (IFN-alpha) producing cells in blood. In this study, C57BL/10 (B10;H2b) mice were treated with fms-like tyrosine 3 kinase Ligand (Flt3L; 10 microg/d; i.p.; 10 days) that expands DC selectively in vivo. Putative pDC (CD11c+B220+) were identified in the subepithelial dome and in interfollicular regions of intestinal Peyer's patches (PP) from both normal and Flt3L-treated animals. Freshly-isolated, immunobead-purified CD11c+ DC from PP were flow-sorted to obtain lineage- (CD11b-CD19-) CD11c+ B220+ DC (purity>96%). Flow cytometric analysis revealed that these sorted PPpDC were negative for surface markers associated with myeloid DC (CD11b) and expressed only low levels of the "lymphoid-related" DC marker CD8alphaalpha+. They expressed low levels of costimulatory molecules and moderate MHC class II. They proved weak stimulators of naïve allogeneic (C3H; H2k) T-cell proliferation. Cytospin preparations of sorted CD11c+B220+ cells revealed plasmacytoid morphology similar to that of human pDC. Immunocytochemistry and enzyme immunoassay revealed that, within 24-hour culture with Herpes simplex virus (10 p.f.u./cell), a subpopulation of stimulated (but not unstimulated) CD11c+B220+ DC produced and secreted IFN-alpha. This novel DC subset may play important roles in innate and adaptive immune responses of the gut and in the regulation of mucosal immune reactions.
Collapse
Affiliation(s)
- Antonino Castellaneta
- Department of Surgery, the Thomas E Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | | | | | | |
Collapse
|
19
|
Simioni PU, Fernandes LGR, Gabriel DL, Tamashiro WMSC. Induction of Systemic Tolerance in Normal but not in Transgenic Mice Through Continuous Feeding of Ovalbumin. Scand J Immunol 2004; 60:257-66. [PMID: 15320882 DOI: 10.1111/j.0300-9475.2004.01454.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The ingestion of most dietary protein can cause systemic tolerance, and such tolerance is easier to induce in younger than in older mice. In this study, we examined whether oral tolerance to ovalbumin (OVA) could be induced in OVA-T-cell receptor (OVA-TCR)-specific transgenic mice. Continuous feeding or gavage with OVA induced tolerance, measured as reduced antibody production, in young and aged BALB/c mice, in a dose-dependent manner, but this effect was not observed in transgenic mice. Once BALB/c mice became tolerant, this state was maintained for over 44 weeks, although the tolerant state could be reversed by adoptive cell transfer. DO11.10 mice did not become tolerant upon continuous feeding with OVA, and the adoptive transfer of naïve cells increased the levels of specific antibodies in their sera after antigenic challenge. The immunization schedule used here leads to a Th2-dependent antibody response in normal BALB/c mice. However, the same schedule induced both Th1- and Th2-antibody responses in transgenic mice. Dendritic cells (DC) from tolerant BALB/c mice were less efficient in the induction of the proliferation of cocultured T cells from both BALB/c and DO11.10 mice, as well as Th1 [interleukin (IL)-2 and interferon (IFN)-gamma] and Th2 (IL-4 and IL-10) cytokine production. The DC from DO11.10 transgenic mice were equally efficient in the induction of T-cell proliferation in both normal and transgenic mice, as well as in the induction of Th1 and Th2 cytokines, whether or not the mice consumed OVA. Transforming growth factor (TGF)-beta secretion was significantly lower in the supernatants of T cells from both normal and transgenic mice cocultured with DC from DO11.10 mice that had consumed OVA, while it was significantly higher in the presence of DC from normal tolerant mice, thus implicating TGF-beta as a regulatory cytokine in oral tolerance in the murine model.
Collapse
Affiliation(s)
- P U Simioni
- Department of Microbiology and Immunology, Institute of Biology, State University of Campinas, Campinas, SP, Brazil
| | | | | | | |
Collapse
|
20
|
Epardaud M, Bonneau M, Payot F, Cordier C, Mégret J, Howard C, Schwartz-Cornil I. Enrichment for a CD26hi SIRP- subset in lymph dendritic cells from the upper aero-digestive tract. J Leukoc Biol 2004; 76:553-61. [PMID: 15197234 DOI: 10.1189/jlb.0404223] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Dendritic cells (DC) have been reported to migrate in afferent lymph in the steady state. However, it is unknown whether DC traffic is modulated by the nature of the drained tissue. To analyze the influence of mucosal versus cutaneous microenvironments on the constitutive DC release, we exploited a novel technique of lymph cannulation in sheep, which allowed a comparison of afferent lymph DC migrating from the head mucosae [cervical DC (CerDC)] with DC migrating from skin [prescapular DC (PresDC)]. The migration rate was lower for CerDC than for PresDC. Compared with PresDC, CerDC contained a higher proportion of the CD26hi signal regulatory protein (SIRP)- DC subset. It is interesting that cytoplasmic apoptotic DNA as well as cytokeratin-positive inclusions were primarily detected among CD26hi SIRP- DC, an observation similar to that made in rats, which leads to the suggestion that this subset was involved in self-antigen presentation and tolerance induction. After the inoculation of cholera toxin (CT) onto the oro-nasal mucosae, migration of CD26hi SIRP- and CD26lo SIRP+ DC was accelerated in lymph, indicating that the effect of CT on DC mobilization is not subset-specific. Our results show that a mucosal environment influences DC output and the relative DC subset representation in lymph. This modulation of DC traffic to lymph nodes by mucosal surfaces is likely to affect the bias of the mucosal immune responses.
Collapse
|
21
|
Björkstén B. Effects of intestinal microflora and the environment on the development of asthma and allergy. SPRINGER SEMINARS IN IMMUNOPATHOLOGY 2004; 25:257-70. [PMID: 15007630 DOI: 10.1007/s00281-003-0142-2] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2003] [Accepted: 08/26/2003] [Indexed: 01/11/2023]
Abstract
The aim of previous research into the causes of allergic diseases, including asthma was mostly to identify potential risk factors in the environment. No major risk factors have been identified, however. Over the past 10 years, focus has, therefore, more been directed towards protective factors that could enhance the development of tolerance to allergens which were previously encountered early in life, but are now lost in modern affluent societies. In particular, the role of childhood infections has been discussed, but so far these studies have not been conclusive. Recent epidemiological studies and experimental research suggest that the microbial environment and exposure to microbial products in infancy modifies immune responses and enhances the development of tolerance to ubiquitous allergens. The intestinal microflora may play a particular role in this respect, as it is the major external driving force in the maturation of the immune system after birth, and animal experiments have shown it to be a prerequisite for normal development of oral tolerance. Recent studies have shown differences in the composition of the microflora between healthy and allergic infants in countries with a high and low prevalence of allergies and between healthy and allergic infants within such countries. These differences are apparent within the first week of life and thus precede clinical symptoms. The use of live microorganisms that might be beneficial to health has a long tradition and the safety is well documented. Very recently, several prospective intervention studies, modifying the gut flora from birth have yielded encouraging results and may suggest a new mode of primary prevention of allergy in the future.
Collapse
Affiliation(s)
- Bengt Björkstén
- Centre for Allergy Research and Department of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden.
| |
Collapse
|
22
|
Bozić F, Bilić V, Valpotić I. Levamisole mucosal adjuvant activity for a live attenuated Escherichia coli oral vaccine in weaned pigs. J Vet Pharmacol Ther 2003; 26:225-31. [PMID: 12755907 DOI: 10.1046/j.1365-2885.2003.00458.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The present study tested the hypothesis that levamisole exerts its immunopotentiating activity in weaned pigs vaccinated against colibacillosis by priming the lymphocytes and macrophages in the mesenteric lymph node (MLN). Ten weaned piglets were used and allocated into two equal groups. The experimental group was intramuscularly primed with levamisole at an immunostimulatory dose of 2.5 mg/kg given daily, in three consecutive days, and controls received saline according to the same schedule. Both groups were orally vaccinated with the vaccinal Escherichia coli strain on day 0 and challenged with the virulent E. coli strain 7 days later. All pigs were killed on postchallenge day 6. Upon virulent challenge the health status of the two groups was evaluated by clinical observations, and expression of CD25, SWC7 and SWC9 activation antigens by MLN and spleen T and B cells and macrophages, respectively, was tested using flow cytometry. Priming by levamisole significantly contributed to the effectiveness of a live attenuated oral vaccine against porcine postweaning colibacillosis, as evidenced by a good health status of primed vaccinated vs. un-primed vaccinated pigs. The CD3+, CD25+ and SWC9+ MLN but not spleen T cells and macrophages increased in experimental vs. control pigs, implying that levamisole exerts its potentiating activity in the MLN by augmenting both recruitment and activation of cells that participate in cell-mediated immunity.
Collapse
Affiliation(s)
- F Bozić
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Zagreb, Zagreb, Croatia.
| | | | | |
Collapse
|
23
|
Meritet JF, Maury C, Tovey MG. Induction of tolerance to recombinant therapeutic proteins. J Interferon Cytokine Res 2001; 21:1031-8. [PMID: 11798460 DOI: 10.1089/107999001317205150] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The specific IgM and IgG antibody responses to subcutaneous (s.c.) treatment of mice with recombinant human IFN-alpha2a (rHuIFN-alpha2a) or IFN-beta were inhibited in a dose-dependent manner by prior oromucosal (o.m.) administration of rHuIFN-alpha2a or IFN-beta, respectively. Pretreatment of animals once a day for 7 days by the o.m. route with the highest dose of IFN-alpha2a tested (10(7) IU) resulted in complete inhibition of the peak IFN-alpha2a-specific IgG antibody response detected 28 days after subsequent s.c. injection of IFN-alpha2a (p < 0.001). Similarly, prior o.m. administration of 1-10 microg rHuGM-CSF per day for 7 days resulted in a statistically significant (p < 0.001) inhibition of the peak GM-CSF-specific IgG antibody response detected 28 days after s.c. administration of GM-CSF. In contrast, prior o.m. treatment with a quantity of bovine serum albumin (BSA) (100 microg) or human serum albumin (HSA) (10 microg) equivalent, respectively, to the protein content of the highest dose of IFN-alpha2a or GM-CSF administered by the o.m. route, did not affect significantly the IFN-alpha2a-specific or GM-CSF-specific IgG antibody responses detected on subsequent s.c. administration of IFN-alpha2a or GM-CSF. Oromucosal administration of IFN-alpha2a, IFN-beta, or GM-CSF alone did not induce detectable IFN-alpha2a-specific, IFN-beta-specific, or GM-CSF-specific IgM or IgG antibody responses at any of the time points tested. These results suggest that short-term o.m. administration of a recombinant protein is an effective means of inducing peripheral tolerance to subsequent parenteral administration of a therapeutic protein.
Collapse
Affiliation(s)
- J F Meritet
- Laboratory of Viral Oncology, UPR 9045 CNRS, Institut André Lwoff/IFR 2249, 94801 Villejuif, France
| | | | | |
Collapse
|
24
|
Abstract
Immunologists typically study the immune responses induced in the spleen or peripheral lymph nodes after parenteral immunization with antigen and poorly defined experimental adjuvants. However, most antigens enter the body through mucosal surfaces. It is now clear that the microenvironment in these mucosal barriers has a marked influence on the immune response that ultimately ensues. Nowhere is the microenvironment more influential than in the gut-associated lymphoid tissue (GALT). The GALT must constantly distinguish harmless antigens that are present in food or on commensal bacteria from pathogenic assault by microbes. It is perhaps not surprising, then, that the GALT contains more lymphocytes than all of the secondary lymphoid organs combined.
Collapse
Affiliation(s)
- C Nagler-Anderson
- Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Charlestown 02129, USA.
| |
Collapse
|
25
|
Gao ZH, Williams GM. Vascular endothelial-cell turnover: a new factor in the vascular microenvironment of the liver. Trends Immunol 2001; 22:421-2. [PMID: 11491091 DOI: 10.1016/s1471-4906(01)01988-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|