Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Cell Mol Immunol ; 21(6): 575-588, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38632385

ABSTRACT

Neonates are susceptible to inflammatory disorders such as necrotizing enterocolitis (NEC) due to their immature immune system. The timely appearance of regulatory immune cells in early life contributes to the control of inflammation in neonates, yet the underlying mechanisms of which remain poorly understood. In this study, we identified a subset of neonatal monocytes characterized by high levels of neuropilin-1 (Nrp1), termed Nrp1high monocytes. Compared with their Nrp1low counterparts, Nrp1high monocytes displayed potent immunosuppressive activity. Nrp1 deficiency in myeloid cells aggravated the severity of NEC, whereas adoptive transfer of Nrp1high monocytes led to remission of NEC. Mechanistic studies showed that Nrp1, by binding to its ligand Sema4a, induced intracellular p38-MAPK/mTOR signaling and activated the transcription factor KLF4. KLF4 transactivated Nos2 and enhanced the production of nitric oxide (NO), a key mediator of immunosuppression in monocytes. These findings reveal an important immunosuppressive axis in neonatal monocytes and provide a potential therapeutic strategy for treating inflammatory disorders in neonates.


Subject(s)
Animals, Newborn , Enterocolitis, Necrotizing , Inflammation , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors , Monocytes , Neuropilin-1 , Monocytes/metabolism , Monocytes/immunology , Animals , Neuropilin-1/metabolism , Neuropilin-1/genetics , Inflammation/pathology , Inflammation/immunology , Humans , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/genetics , Enterocolitis, Necrotizing/immunology , Enterocolitis, Necrotizing/metabolism , Enterocolitis, Necrotizing/prevention & control , Mice , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Mice, Inbred C57BL , Infant, Newborn , p38 Mitogen-Activated Protein Kinases/metabolism , Mice, Knockout
2.
J Virol ; 97(3): e0176422, 2023 03 30.
Article in English | MEDLINE | ID: mdl-36779760

ABSTRACT

Respiratory syncytial virus (RSV) infects more than 60% of infants in their first year of life. Since an experimental formalin-inactivated (FI) RSV vaccine tested in the 1960s caused enhanced respiratory disease (ERD), few attempts have been made to vaccinate infants. ERD is characterized by Th2-biased responses, lung inflammation, and poor protective immune memory. Innate immune memory displays an increased nonspecific effector function upon restimulation, a process called trained immunity, or a repressed effector function upon restimulation, a process called tolerance, which participates in host defense and inflammatory disease. Mycobacterium bovis bacillus Calmette-Guérin (BCG) given at birth can induce trained immunity as well as heterologous Th1 responses. We speculate that BCG given at birth followed by FI-RSV may alleviate ERD and enhance protection through promoting trained immunity and balanced Th immune memory. Neonatal mice were given BCG at birth and then vaccinated with FI-RSV+Al(OH)3. BCG/FI-RSV+Al(OH)3 induced trained macrophages, tissue-resident memory T cells (TRM), and specific cytotoxic T lymphocytes (CTL) in lungs and inhibited Th2 and Th17 cell immune memory, all of which contributed to inhibition of ERD and increased protection. Notably, FI-RSV+Al(OH)3 induced tolerant macrophages, while BCG/FI-RSV+Al(OH)3 prevented the innate tolerance through promoting trained macrophages. Moreover, inhibition of ERD was attributed to trained macrophages or TRM in lungs but not memory T cells in spleens. Therefore, BCG given at birth to regulate trained immunity and TRM may be a new strategy for developing safe and effective RSV killed vaccines for young infants. IMPORTANCE RSV is the leading cause of severe lower respiratory tract infection of infants. ERD, characterized by Th2-biased responses, inflammation, and poor immune memory, has been an obstacle to the development of safe and effective killed RSV vaccines. Innate immune memory participates in host defense and inflammatory disease. BCG given at birth can induce trained immunity as well as heterologous Th1 responses. Our results showed that BCG/FI-RSV+Al(OH)3 induced trained macrophages, TRM, specific CTL, and balanced Th cell immune memory, which contributed to inhibition of ERD and increased protection. Notably, FI-RSV+Al(OH)3 induced tolerant macrophages, while BCG/FI-RSV+Al(OH)3 prevented tolerance through promoting trained macrophages. Moreover, inhibition of ERD was attributed to trained macrophages or TRM in lungs but not memory T cells in spleens. BCG at birth as an adjuvant to regulate trained immunity and TRM may be a new strategy for developing safe and effective RSV killed vaccines for young infants.


Subject(s)
BCG Vaccine , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus Vaccines , Animals , Mice , BCG Vaccine/administration & dosage , BCG Vaccine/immunology , Lung/immunology , Macrophages/immunology , Mice, Inbred BALB C , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/administration & dosage , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Human/immunology , Spleen/immunology , Th1 Cells/immunology , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology
3.
Front Immunol ; 13: 977235, 2022.
Article in English | MEDLINE | ID: mdl-36211408

ABSTRACT

Background: Infants with respiratory syncytial virus (RSV)-associated bronchiolitis are at increased risk of childhood asthma. Recent studies demonstrated that certain infections induce innate immune memory (also termed trained immunity), especially in macrophages, to respond more strongly to future stimuli with broad specificity, involving in human inflammatory diseases. Metabolic reprogramming increases the capacity of the innate immune cells to respond to a secondary stimulation, is a crucial step for the induction of trained immunity. We hypothesize that specific metabolic reprogramming of lung trained macrophages induced by neonatal respiratory infection is crucial for childhood allergic asthma. Objective: To address the role of metabolic reprogramming in lung trained macrophages induced by respiratory virus infection in allergic asthma. Methods: Neonatal mice were infected and sensitized by the natural rodent pathogen Pneumonia virus of mice (PVM), a mouse equivalent strain of human RSV, combined with ovalbumin (OVA). Lung CD11b+ macrophages in the memory phase were re-stimulated to investigate trained immunity and metabonomics. Adoptive transfer, metabolic inhibitor and restore experiments were used to explore the role of specific metabolic reprogramming in childhood allergic asthma. Results: PVM infection combined with OVA sensitization in neonatal mice resulted in non-Th2 (Th1/Th17) type allergic asthma following OVA challenge in childhood of mice. Lung CD11b+ macrophages in the memory phage increased, and showed enhanced inflammatory responses following re-stimulation, suggesting trained macrophages. Adoptive transfer of the trained macrophages mediated the allergic asthma in childhood. The trained macrophages showed metabolic reprogramming after re-stimulation. Notably, proline biosynthesis remarkably increased. Inhibition of proline biosynthesis suppressed the development of the trained macrophages as well as the Th1/Th17 type allergic asthma, while supplement of proline recovered the trained macrophages as well as the allergic asthma. Conclusion: Proline metabolism reprogramming of trained macrophages induced by early respiratory infection combined with allergen sensitization contributes to development of allergic asthma in childhood. Proline metabolism could be a well target for prevention of allergic asthma in childhood.


Subject(s)
Asthma , Hypersensitivity , Respiratory Syncytial Virus Infections , Respiratory Tract Infections , Allergens , Animals , Humans , Macrophages , Mice , Mice, Inbred BALB C , Ovalbumin , Proline
4.
Plant J ; 112(5): 1159-1175, 2022 12.
Article in English | MEDLINE | ID: mdl-36214418

ABSTRACT

Chrysanthemum (Chrysanthemum morifolium) is well known as a photoperiod-sensitive flowering plant. However, it has also evolved into a temperature-sensitive ecotype. Low temperature can promote the floral transition of the temperature-sensitive ecotype, but little is known about the underlying molecular mechanisms. Here, we identified MADS AFFECTING FLOWERING 2 (CmMAF2), a putative MADS-box gene, which induces floral transition in response to low temperatures independent of day length conditions in this ecotype. CmMAF2 was shown to bind to the promoter of the GA biosynthesis gene CmGA20ox1 and to directly regulate the biosynthesis of bioactive GA1 and GA4 . The elevated bioactive GA levels activated LEAFY (CmLFY) expression, ultimately initiating floral transition. In addition, CmMAF2 expression in response to low temperatures was directly activated by CmC3H1, a CCCH-type zinc-finger protein upstream. In summary, our results reveal that the CmC3H1-CmMAF2 module regulates flowering time in response to low temperatures by regulating GA biosynthesis in the temperature-sensitive chrysanthemum ecotype.


Subject(s)
Chrysanthemum , Chrysanthemum/physiology , Gibberellins/metabolism , Temperature , Flowers/metabolism , Gene Expression Regulation, Plant , Plant Proteins/genetics , Plant Proteins/metabolism , Photoperiod
5.
Curr Med Sci ; 39(3): 363-370, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31209804

ABSTRACT

Respiratory syncytial virus (RSV) infection is the primary cause of respiratory disease in infants. The formalin-inactivated RSV (FI-RSV) vaccine resulted in an enhanced respiratory disease (ERD) in infants upon natural RSV infection, which is a major obstacle for development of safe and efficacious vaccines. Excessive and uncontrolled Th immune responses could be involved in the ERD. Agonists of TLRs are used as adjuvants to guide the type of immune response induced by vaccines. We evaluated the impact of lipopolysaccharide (LPS), the agonist of TLR4, on ERD as the adjuvant of FI-RSV. The results showed that LPS remarkably inhibited FI-RSV-enhanced lung inflammation, mucus production, airway inflammatory cell infiltration, and inflammatory cytokines following RSV challenge. Interestingly, LPS inhibited both Th2 and Th17 type cytokines in lungs of FI-RSV-immunized mice following RSV challenge, without an increase in the Th1 type cytokines, suggesting a controlled immune response. In contrast, Pam3Cys and Poly(I:C), the agonist of TLR1/2 or TLR3, partly inhibited FI-RSV-enhanced lung inflammation. Pam3Cys inhibited Th17 type cytokine IL-17, but promoted both Th1 and Th2 type cytokines. Poly(I:C) inhibited Th2 and Th17 type cytokines, but promoted Th1 type cytokines. In addition, LPS promoted IgG and IgG2a antibody production, which might provide protection from RSV challenge. These results suggest that LPS inhibits ERD without impairment in antibody production and protection, and the mechanism appears to be related with regulation of Th responses induced by FI-RSV.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antibodies, Viral/biosynthesis , Lipopolysaccharides/pharmacology , Pneumonia/prevention & control , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/adverse effects , Respiratory Syncytial Viruses/drug effects , Animals , Female , Formaldehyde , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-2/genetics , Interleukin-2/immunology , Lipoproteins/pharmacology , Mice , Mice, Inbred C57BL , Pneumonia/etiology , Pneumonia/immunology , Pneumonia/pathology , Poly I-C/pharmacology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus Vaccines/administration & dosage , Respiratory Syncytial Viruses/immunology , Respiratory Syncytial Viruses/pathogenicity , Th1 Cells/drug effects , Th1 Cells/immunology , Th1 Cells/virology , Th1-Th2 Balance/drug effects , Th17 Cells/drug effects , Th17 Cells/immunology , Th17 Cells/virology , Th2 Cells/drug effects , Th2 Cells/immunology , Th2 Cells/virology , Toll-Like Receptor 2/agonists , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/immunology , Toll-Like Receptor 3/agonists , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/immunology , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Vaccination , Vaccines, Inactivated
6.
Hum Vaccin Immunother ; 15(11): 2684-2694, 2019.
Article in English | MEDLINE | ID: mdl-31021703

ABSTRACT

Respiratory syncytial virus (RSV) is a major respiratory pathogen in infants. The early formalin-inactivated RSV not only failed to protect infants against infection, but also was associated with enhanced pulmonary inflammatory disease upon natural infection. A safe and effective vaccine should prevent the inflammatory disease and provide protection. Immune memory is the cornerstone of vaccines. In this study, we evaluated three types of immune memory T cells, antibodies, and lung inflammation of a vaccine candidate G1F/M2, which includes a neutralizing epitope fragment of RSV G protein and a cytotoxic T lymphocyte epitope of M2 protein, with toll-like receptor 9 agonist CpG2006 as an adjuvant by intranasal (i.n.) and intraperitoneal (i.p.) immunization protocols. The results indicated that immunization of mice with G1F/M2 + CpG i.p. induced significantly higher level of CD4+ or CD8+ central memory (TCM), Th1-type effector memory (TEM), and balanced ratio of IgG1/IgG2a, but lower level of lung tissue-resident memory (TRM), compared with immunization with G1F/M2 + CpG i.n., G1F/M2 i.n., or G1F/M2 i.p. Following RSV challenge, the mice immunized with G1F/M2 + CpG i.p. showed higher level of Th1-type responses, remarkably suppressed inflammatory cytokines and histopathology in lungs, compared with mice immunized with G1F/M2 + CpG i.n., G1F/M2 i.n., or G1F/M2 i.p. These results suggested that high level of TCM and Th1 type of TEM in spleens may contribute to inhibition of lung inflammation, while high level of TRM in lungs and lack of or weak Th1-type immune memory in spleens may promote lung inflammation following RSV challenge.


Subject(s)
Immunologic Memory , Lung/immunology , Oligodeoxyribonucleotides/immunology , Pneumonia/prevention & control , Respiratory Syncytial Virus Vaccines/immunology , Spleen/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Viral/blood , Female , Lung/pathology , Mice , Mice, Inbred BALB C , Oligodeoxyribonucleotides/administration & dosage , Pneumonia/etiology , Respiratory Syncytial Virus Vaccines/adverse effects , Respiratory Syncytial Virus Vaccines/genetics , Respiratory Syncytial Virus, Human , Spleen/pathology , Th1 Cells/immunology , Th2 Cells/immunology , Vaccines, Synthetic/adverse effects , Vaccines, Synthetic/immunology , Viral Proteins/immunology
7.
J Virol ; 92(8)2018 04 15.
Article in English | MEDLINE | ID: mdl-29386287

ABSTRACT

Respiratory syncytial virus (RSV) is the main cause of acute lower respiratory tract infection (ALRI) in children worldwide. Virus-host interactions affect the progression and prognosis of the infection. Autophagy plays important roles in virus-host interactions. Respiratory epithelial cells serve as the front line of host defense during RSV infection, However, it is still unclear how they interact with RSV. In this study, we found that RSV induced autophagy that favored RSV replication and exacerbated lung pathology in vivo Mechanistically, RSV induced complete autophagy flux through reactive oxygen species (ROS) generation and activation of the AMP-activated protein kinase/mammalian target of rapamycin (AMPK-MTOR) signaling pathway in HEp-2 cells. Furthermore, we evaluated the functions of autophagy in RSV replication and found that RSV replication was increased in HEp-2 cells treated with rapamycin but decreased remarkably in cells treated with 3-methylademine (3-MA) or wortmannin. Knockdown key molecules in the autophagy pathway with short hairpinp RNA (shRNA) against autophagy-related gene 5 (ATG5), autophagy-related gene 7 (ATG7), or BECN1/Beclin 1 or treatment with ROS scavenger N-acetyl-l-cysteine (NAC) and AMPK inhibitor (compound C) suppressed RSV replication. 3-MA or shATG5/BECN1 significantly decreased cell viability and increased cell apoptosis at 48 hours postinfection (hpi). Blocking apoptosis with Z-VAD-FMK partially restored virus replication at 48 hpi. Those results provide strong evidence that autophagy may function as a proviral mechanism in a cell-intrinsic manner during RSV infection.IMPORTANCE An understanding of the mechanisms that respiratory syncytial virus utilizes to interact with respiratory epithelial cells is critical to the development of novel antiviral strategies. In this study, we found that RSV induces autophagy through a ROS-AMPK signaling axis, which in turn promotes viral infection. Autophagy favors RSV replication through blocking cell apoptosis at 48 hpi. Mechanistically, RSV induces mitophagy, which maintains mitochondrial homeostasis and therefore decreases cytochrome c release and apoptosis induction. This study provides a novel insight into this virus-host interaction, which may help to exploit new antiviral treatments targeting autophagy processes.


Subject(s)
Apoptosis , Autophagy , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Viruses/physiology , Virus Replication , AMP-Activated Protein Kinases/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Autophagy-Related Protein 5/metabolism , Autophagy-Related Protein 7/metabolism , Beclin-1/metabolism , Cell Line , Humans , Respiratory Syncytial Virus Infections/pathology , TOR Serine-Threonine Kinases/metabolism
8.
J Virol ; 91(10)2017 05 15.
Article in English | MEDLINE | ID: mdl-28275186

ABSTRACT

Respiratory syncytial virus (RSV) is the leading cause of childhood hospitalizations. The formalin-inactivated RSV (FI-RSV) vaccine-enhanced respiratory disease (ERD) has been an obstacle to the development of a safe and effective killed RSV vaccine. Agonists of Toll-like receptor (TLR) have been shown to regulate immune responses induced by FI-RSV. Notch signaling plays critical roles during the differentiation and effector function phases of innate and adaptive immune responses. Cross talk between TLR and Notch signaling pathways results in fine-tuning of TLR-triggered innate inflammatory responses. We evaluated the impact of TLR and Notch signaling on ERD in a murine model by administering CpG, an agonist of TLR9, in combination with L685,458, an inhibitor of Notch signaling during FI-RSV immunization. Activation with CpG or deficiency of MyD88-dependent TLR signaling did not alleviate airway inflammation in FI-RSV-immunized mice. Activation or inhibition of Notch signaling with Dll4, one of the Notch ligands, or L685,458 did not suppress FI-RSV-enhanced airway inflammation either. However, the CpG together with L685,458 markedly inhibited FI-RSV-enhanced airway hyperresponsiveness, weight loss, and lung inflammation. Interestingly, CpG plus L685,458 completely inhibited FI-RSV-associated Th17 and Th17-associated proinflammatory chemokine responses in lungs following RSV challenge but not Th1 or Th2, memory responses. In addition, FI-RSV plus CpG plus L685,458 promoted protective CD8+ lung tissue-resident memory (TRM) cells. These results indicate that activation of TLR signaling combined with inhibition of Notch signaling prevent FI-RSV ERD, and the mechanism appears to involve suppressing proinflammatory Th17 memory responses and promoting protective TRM in lungs.IMPORTANCE RSV is the most important cause of lower respiratory tract infections in infants. The FI-RSV-enhanced respiratory disease (ERD) is a major impediment to the development of a safe and effective killed RSV vaccine. Using adjuvants to regulate innate and adaptive immune responses could be an effective method to prevent ERD. We evaluated the impact of TLR and Notch signaling on ERD by administering CpG, an agonist of TLR9, in combination with L685,458, an inhibitor of Notch signaling, during FI-RSV immunization. The data showed that treatment of TLR or Notch signaling alone did not suppress FI-RSV-enhanced airway inflammation, while CpG plus L685,458 markedly inhibited ERD. The mechanism appears to involve suppressing Th17 memory responses and promoting tissue-resident memory cells. Moreover, these results suggest that regulation of lung immune memory with adjuvant compounds containing more than one immune-stimulatory molecule may be a good strategy to prevent FI-RSV ERD.


Subject(s)
Adjuvants, Immunologic/chemistry , Immunologic Memory , Lung/immunology , Oligodeoxyribonucleotides/pharmacology , Receptors, Notch/metabolism , Th17 Cells/immunology , Animals , Bronchial Hyperreactivity/chemically induced , CD8-Positive T-Lymphocytes/immunology , Carbamates/pharmacology , Dipeptides/pharmacology , Formaldehyde , Humans , Inflammation , Lung/virology , Mice , Mice, Inbred BALB C , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Human/immunology , Signal Transduction , Toll-Like Receptor 1/immunology , Toll-Like Receptor 1/metabolism , Vaccines, Inactivated/adverse effects , Vaccines, Inactivated/immunology
9.
Virus Res ; 167(2): 138-45, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22698878

ABSTRACT

Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract illness in infants and young children. Host immune response has been implicated in both the protection and immunopathological mechanisms. Pattern recognition receptors (PRRs) expressed on innate immune cells during RSV infection recognize the RSV-associated molecular patterns and activate innate immune cells as well as mediate airway inflammation, protective immune response, and pulmonary immunopathology. The resident and recruited innate immune cells play important roles in the protection and pathogenesis of an RSV disease by expressing these PRRs. Agonist-binding PRRs are the basis of many adjuvants that are essential for most vaccines. In the present review, we highlight recent advances in the innate immune recognition of and responses to RSV through PRRs, including toll-like receptors (TLRs), retinoic acid-inducible gene (RIG)-I-like receptors (RLRs), and nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs). We also describe the role of PRRs in the design of RSV vaccines.


Subject(s)
Receptors, Pattern Recognition/immunology , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Human/immunology , Respiratory Tract Infections/immunology , Respiratory Tract Infections/pathology , Humans , Immunity, Innate , Respiratory Tract Infections/virology
10.
J Virol ; 86(8): 4505-17, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22301139

ABSTRACT

Respiratory syncytial virus (RSV) is the most important cause of lower respiratory tract disease in young children. In the 1960s, infants vaccinated with formalin-inactivated RSV developed a more severe disease characterized by excessive inflammatory immunopathology in lungs upon natural RSV infection. The fear of causing the vaccine-enhanced disease (VED) is an important obstacle for development of safe and effective RSV vaccines. The recombinant vaccine candidate G1F/M2 immunization also led to VED. It has been proved that cellular memory induced by RSV vaccines contributed to VED. Interleukin-27 (IL-27) and IL-23 regulate Th1, Th17, and/or Th2 cellular immune responses. In this study, mice coimmunized with pcDNA3-IL-27 and G1F/M2 were fully protected and, importantly, did not develop vaccine-enhanced inflammatory responses and immunopathology in lungs after RSV challenge, which was correlated with moderate Th1-, suppressed Th2-, and Th17-like memory responses activated by RSV. In contrast, G1F/M2- or pcDNA3-IL-23+G1F/M2-immunized mice, in which robust Th2- and Th17-like memory responses were induced, developed enhanced pulmonary inflammation and severe immunopathology. Mice coimmunized with G1F/M2 and the two cytokine plasmids exhibited mild inflammatory responses as well as remarkable Th1-, suppressed Th2-, and Th17-like memory responses. These results suggested that Th1-, Th2-, and Th17-like memory responses and, in particular, excessive Th2- and Th17-like memory responses were closely associated with VED; IL-27 may inhibit VED following respiratory syncytial virus infection by regulating cellular memory responses.


Subject(s)
Immunity, Cellular , Immunologic Memory , Interleukin-17/metabolism , Lung Diseases/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Viruses/immunology , Adjuvants, Immunologic , Animals , Cell Line , Female , Humans , Immunity, Humoral , Inflammation Mediators/metabolism , Lung Diseases/pathology , Mice , Mice, Inbred BALB C , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/metabolism , Th1 Cells/immunology , Th17 Cells/immunology , Th2 Cells/immunology , Vaccines, DNA/immunology , Vaccines, DNA/metabolism
11.
Cytokine ; 53(1): 1-7, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21035355

ABSTRACT

Respiratory syncytial virus (RSV) is the primary cause of serious lower respiratory tract illness in infants and young children worldwide. The mechanism is largely unknown. RSV stimulates airway epithelial cells and resident leukocytes to release cytokines. Cytokines and chemokines involved in host response to RSV infection are thought to play a central role in the pathogenesis. In addition, RSV infection early in life has been associated with the development of asthma in later childhood. It is likely that the persistence of cytokines and chemokines in fully recovered patients with RSV in the long term can provide a substratum for the development of subsequent asthma. This review describes the genetic factors in cytokines and chemokines associated with severity of RSV disease, cytokines and chemokines synthesis in RSV infection, and the role of these innate immune components in RSV-associated asthma.


Subject(s)
Asthma/etiology , Asthma/immunology , Chemokines/immunology , Respiratory Syncytial Virus Infections/complications , Respiratory Syncytial Virus Infections/immunology , Asthma/genetics , Asthma/physiopathology , Chemokines/genetics , Humans , Respiratory Syncytial Virus Infections/genetics , Respiratory Syncytial Virus Infections/physiopathology
12.
Antiviral Res ; 84(1): 23-30, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19646486

ABSTRACT

Hepatitis C virus (HCV) has emerged as the major pathogen of liver disease worldwide. The mechanisms of HCV infection and interaction with a host are poorly understood. What exactly is required for efficient control of HCV infection is largely unknown. Standard treatment combining interferon-alpha (IFN-alpha) and ribavirine is effective in about 50% of the treated patients, however associated with significant toxicity and cost. Therefore, the development of new drugs or vaccines is urgently needed. An efficient vaccine against HCV infection requires induction of broad cellular and humoral immune responses against several viral proteins. We have engineered the combined vaccine candidate mT+mE1, an inclusion of multiple epitopes from HCV NS3, core (C) and E1 proteins. mT contains multiple conserved CD4(+) and CD8(+) T cell epitopes from HCV NS3 and C proteins. mE1 is based on eight dominant neutralizing epitopes of E1 protein from six HCV genotypes. In current study, we showed that immunization with mT+mE1 induced high titers of IgG, IgG1 and IgG2a antibodies to mE1, and high level of NS3- or C-specific CTLs. Furthermore, mT+mE1 elicited a Th1-biased immune response with secretion of high amounts of IFN-gamma, compared with mT alone. Prophylactic as well as therapeutic administration of mT+mE1 in BALB/c mice led to protecting mice against SP2/0 tumor cells expressing HCV NS3 protein. These results suggested that mT+mE1 elicited strong humoral immune responses and multiple specific cellular immune responses. The vaccine candidate is now being tested in pre-clinical trials.


Subject(s)
Hepacivirus/immunology , Hepatitis C/immunology , Viral Core Proteins/immunology , Viral Envelope Proteins/immunology , Viral Hepatitis Vaccines/immunology , Viral Nonstructural Proteins/immunology , Animals , Antibodies, Viral/blood , Epitopes/administration & dosage , Epitopes/genetics , Epitopes/immunology , Female , Hepacivirus/genetics , Hepatitis C/virology , Humans , Mice , Mice, Inbred BALB C , Protein Engineering , Vaccines, Combined/administration & dosage , Viral Core Proteins/administration & dosage , Viral Core Proteins/genetics , Viral Envelope Proteins/administration & dosage , Viral Envelope Proteins/genetics , Viral Hepatitis Vaccines/administration & dosage , Viral Hepatitis Vaccines/genetics , Viral Nonstructural Proteins/administration & dosage , Viral Nonstructural Proteins/genetics
13.
Biochem Biophys Res Commun ; 377(2): 495-499, 2008 Dec 12.
Article in English | MEDLINE | ID: mdl-18851947

ABSTRACT

Respiratory syncytial virus (RSV) is a major respiratory pathogen in newborns. Neonate vaccine should induce strong protective immunity. We have engineered a subunit vaccine candidate G1F/M2. A major problem in developing subunit vaccines is their limited immunogenicity. Aluminium adjuvants with a long history of use with routine childhood vaccines have some limitations, especially inability to elicit CTL response. There is a need for alternative adjuvants. Heat shock proteins (HSPs) are characterized as potent immunoadjuvants. In this study, HSP70-like protein 1 (HSP70L1) gene was cloned. The recombinant protein HSP70L1 was expressed in E. coli, purified and renaturated. We evaluated the potential of HSP70L1 used as the adjuvant of G1F/M2. G1F/M2 was chemically cross-linked with HSP70L1 (HSP-G1F/M2). HSP70L1 enhanced significantly the immunogenicity and protective effect of G1F/M2. HSP-G1F/M2 induced significant higher levels of antibodies, neutralizing antibodies and CTL activity than unadjuvanted G1F/M2. The antibody titers induced by HSP-G1F/M2 were similar to that by G1F/M2+Alum. RSV-specific CTL activity induced by HSP-G1F/M2 was stronger than that by G1F/M2+Alum. Interestingly, the protective effect of HSP-G1F/M2 against RSV was significantly stronger than that of G1F/M2+Alum. The results suggest that HSP70L1 is a potent adjuvant of G1F/M2.


Subject(s)
Adjuvants, Immunologic , HSP70 Heat-Shock Proteins/immunology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antibody Formation , Cell Line, Tumor , Cloning, Molecular , Female , HSP70 Heat-Shock Proteins/administration & dosage , HSP70 Heat-Shock Proteins/genetics , Humans , Mice , Mice, Inbred BALB C , Respiratory Syncytial Virus Vaccines/administration & dosage , Respiratory Syncytial Virus Vaccines/genetics , T-Lymphocytes, Cytotoxic/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology
14.
Vaccine ; 25(42): 7422-8, 2007 Oct 16.
Article in English | MEDLINE | ID: mdl-17850930

ABSTRACT

Respiratory syncytial virus (RSV) is the primary cause of serious lower respiratory tract illness in young children. We have engineered a recombinant candidate vaccine G1F/M2, consisting of a cytotoxic T lymphocyte (CTL) epitope of RSV-M2 protein and a domain of RSV-G protein. In this study, the long-term immunogenicity and protective effect were evaluated. In G1F/M2-immunized mice, special antibodies lasted for more than 19 weeks, and the IgG1/IgG2a ratio remained a balanced level till the end of the study, suggesting mixed Th1/Th2 type of responses. Concomitantly, G1F/M2 elicited long-lived RSV-specific CTL activity that was detectable at 12 weeks after the final immunization. Stronger CTL responses were induced with immunization once more at 13 weeks after the last immunization in G1F/M2-primed mice than those in F/M2-primed mice. These results suggest that G1F/M2-induced long-lasting balanced humoral and cellular immunity responses, and immunological memory in mice. Furthermore, following RSV challenge, long-term protective efficacy was observed. RSV replication in lungs of G1F/M2-primed mice elicited also mixed Th1/Th2 responses, a property that is considered advantageous for the safety of an RSV vaccine. Therefore, G1F/M2 is a promising RSV subunit vaccine.


Subject(s)
Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/genetics , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Human/genetics , Respiratory Syncytial Virus, Human/immunology , Animals , Antibodies, Viral/blood , Female , Humans , Immunoglobulin G/blood , Mice , Mice, Inbred BALB C , Recombinant Proteins/genetics , Recombinant Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Time Factors , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Proteins/genetics , Viral Proteins/immunology
15.
Vaccine ; 24(7): 941-7, 2006 Feb 13.
Article in English | MEDLINE | ID: mdl-16159685

ABSTRACT

Respiratory syncytial virus (RSV), an important pathogen of the lower respiratory tract, is responsible for severe illness both in new born and young children and in elderly people. However, development of a RSV vaccine has been hampered by the outcome of the infant trials in the 1960s with a formalin-inactivated RSV (FI-RSV) preparation. Previous studies in mice indicated that G protein immunization resulted in antibody and Th2-type response and failed to induce MHC I-restricted CD8(+) T-cell response. Vaccines designed to induce CD8(+) T-cell along with antibody response might be ideal. In the present report, a fusion protein G1F/M2 containing a RSV-G protein fragment (G: 125-225 amino acid) and a CD8(+) T-cell epitope from RSV-M2 protein was investigated. G1F/M2 was cloned, expressed in E. coli, purified and renaturated. In BALB/c mice, G1F/M2 induced not only humoral immunity but also cellular immunity. In addition, interestedly, G1F/M2 elicited balanced IgG1/IgG2a response. These results suggest that the fusion protein G1F/M2 is potential as a RSV subunit vaccine.


Subject(s)
Epitopes, T-Lymphocyte , Recombinant Fusion Proteins/immunology , Respiratory Syncytial Virus Vaccines/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccines, Synthetic/immunology , Viral Fusion Proteins/immunology , Animals , Antibodies, Viral/blood , Female , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Respiratory Syncytial Viruses/immunology , Vaccination
16.
Sheng Wu Gong Cheng Xue Bao ; 21(4): 534-9, 2005 Jul.
Article in Chinese | MEDLINE | ID: mdl-16176088

ABSTRACT

To construct plasmid of recombinant protein candidate vaccine of respiratory syncytial virus, express it in E. coli, and to investigate its immunogenicity and protective efficacy. A CD8+ T cell epitope from respiratory syncytial virus (RSV) M2 protein F/M2:81 - 95 and the G:125-225 (G1) gene fragments from RSV-G protein containing B cell epitopes were amplified by PCR method and then inserted into the prokaryotic expression vector pET-DsbA after bonding to a linker. The fusion protein DsbA-G1-Linker-F/M2:81-95 (D-G1LF/M2) was expressed successfully in E. coli BL21 (DE3). The product was proved to be RSV-specific by Western-blot. After purified by affinity chromatography on Ni+ Sepharose and renatured by gradient dialysis. D-G1LF/M2 was used to immune BALB/c mice. D-G1LF/M2 induced high anti-D-G1LF/M2 IgG, anti-RSV IgG and neutralizing antibody titers in serum and lung of BALB/c mice, and elicied RSV-specific CTL responses. The IgG subclass distribution revealed that IgG1/IgG2a ratio was 2.66. Viral titration indicated that D-G1LF/M2 could protect BALB/c mice against RSV challenge in lung.


Subject(s)
Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Human/immunology , Viral Envelope Proteins/genetics , Viral Proteins/genetics , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Escherichia coli/genetics , Escherichia coli/metabolism , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Mice , Mice, Inbred BALB C , Plasmids/genetics , Plasmids/immunology , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/biosynthesis , Respiratory Syncytial Virus Vaccines/genetics , Respiratory Syncytial Virus, Human/genetics , Viral Fusion Proteins/genetics
17.
Vaccine ; 23(22): 2869-75, 2005 Apr 22.
Article in English | MEDLINE | ID: mdl-15780735

ABSTRACT

In an effort to seek a means of inducing long lasting respiratory syncytial virus-specific CTL responses in mice, we constructed a new recombinant protein, DsbA-F/M2:81-95, by fusing carrier protein DsbA (disulfide bond isomerase) to the N-terminus of CTL chimeric epitope F/M2:81-95 of this virus. DsbA-F/M2:81-95 can induce effectively virus-specific CTL responses as well as protective immunity without association with enhanced disease. Furthermore, compared with F/M2:81-95 alone, it increases the longevity of CTL responses in vivo up to 2.93 folds. Our study emphasizes that appropriate stimulation of non-antigen-specific T helper cells is essential to induce long lasting CD8+ CTL, and also implies DsbA-F/M2:81-95 may be a promising candidate for RSV vaccine development since it is an efficacious and safe immunogen.


Subject(s)
Epitopes/genetics , Protein Disulfide-Isomerases/genetics , Respiratory Syncytial Viruses/immunology , T-Lymphocytes, Cytotoxic/immunology , Viral Vaccines/immunology , Animals , Base Sequence , DNA Primers , Electrophoresis, Polyacrylamide Gel , Female , Mice , Mice, Inbred BALB C , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/prevention & control , Viral Vaccines/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...