106 results on '"Pichyangkul S"'
Search Results
2. Activation of γδ T Cells in Malaria: Interaction of Cytokines and a Schizont-Associated Plasmodium falciparum Antigen
- Author
-
Pichyangkul, S., Saengkrai, P., Yongvanitchit, K., Stewart, A., and Heppner, D. G.
- Published
- 1997
3. Cigarette smoke extract modulates human β-defensin-2 and interleukin-8 expression in human gingival epithelial cells
- Author
-
Mahanonda, R., Sa-Ard-Iam, N., Eksomtramate, M., Rerkyen, P., Phairat, B., Schaecher, K. E., Fukuda, M. M., and Pichyangkul, S.
- Published
- 2009
- Full Text
- View/download PDF
4. CpG ODN enhances uptake of bacteria by mouse macrophages
- Author
-
UTAISINCHAROEN, P., KESPICHAYAWATTANA, W., ANUNTAGOOL, N., CHAISURIYA, P., PICHYANGKUL, S., KRIEG, A. M., and SIRISINHA, S.
- Published
- 2003
5. CpG ODN activates NO and iNOS production in mouse macrophage cell line (RAW 264·7)
- Author
-
UTAISINCHAROEN, P, ANUNTAGOOL, N, CHAISURIYA, P, PICHYANGKUL, S, and SIRISINHA, S
- Published
- 2002
6. CpG oligodeoxynucleotide enhances immunity against blood-stage malaria infection in mice parenterally immunized with a yeast-expressed 19 kDa carboxyl-terminal fragment of Plasmodium yoelii merozoite surface protein-1 (MSP1 19) formulated in oil-based Montanides
- Author
-
Hirunpetcharat, C., Wipasa, J., Sakkhachornphop, S., Nitkumhan, T., Zheng, Y.Z., Pichyangkul, S., Krieg, A.M., Walsh, D.S., Heppner, D.G., and Good, M.F.
- Published
- 2003
- Full Text
- View/download PDF
7. PURIFICATION OF LYMPHOTOXIN FROM RPMI-1788 CELL LINE SUPERNATE
- Author
-
Pichyangkul, S., primary, Hill, N.O., additional, and Khan, A., additional
- Published
- 1982
- Full Text
- View/download PDF
8. Evaluation of the safety and immunogenicity of Plasmodium falciparum apical membrane antigen 1, merozoite surface protein 1 or RTS,S vaccines with adjuvant system AS02A administered alone or concurrently in rhesus monkeys
- Author
-
Pichyangkul, S., primary, Tongtawe, P., additional, Kum-Arb, U., additional, Yongvanitchit, K., additional, Gettayacamin, M., additional, Hollingdale, M.R., additional, Limsalakpetch, A., additional, Stewart, V.A., additional, Lanar, D.E., additional, Dutta, S., additional, Angov, E., additional, Ware, L.A., additional, Bergmann-Leitner, E.S., additional, House, B., additional, Voss, G., additional, Dubois, M.C., additional, Cohen, J.D., additional, Fukuda, M.M., additional, Heppner, D.G., additional, and Miller, R.S., additional
- Published
- 2009
- Full Text
- View/download PDF
9. Effects of IL-17 on Human Gingival Fibroblasts
- Author
-
Mahanonda, R., primary, Jitprasertwong, P., additional, Sa-Ard-Iam, N., additional, Rerkyen, P., additional, Charatkulangkun, O., additional, Jansisyanont, P., additional, Nisapakultorn, K., additional, Yongvanichit, K., additional, and Pichyangkul, S., additional
- Published
- 2008
- Full Text
- View/download PDF
10. Preclinical Evaluation of the Safety and Immunogenicity of a Vaccine Consisting of Plasmodium falciparum Liver-Stage Antigen 1 with Adjuvant AS01B Administered Alone or Concurrently with the RTS,S/AS01B Vaccine in Rhesus Primates
- Author
-
Pichyangkul, S., primary, Kum-Arb, U., additional, Yongvanitchit, K., additional, Limsalakpetch, A., additional, Gettayacamin, M., additional, Lanar, D. E., additional, Ware, L. A., additional, Stewart, V. A., additional, Heppner, D. G., additional, Mettens, P., additional, Cohen, J. D., additional, Ballou, W. R., additional, and Fukuda, M. M., additional
- Published
- 2008
- Full Text
- View/download PDF
11. Heterologous prime-boost immunization in rhesus macaques by two, optimally spaced particle-mediated epidermal deliveries of Plasmodium falciparum circumsporozoite protein-encoding DNA, followed by intramuscular RTS,S/AS02A☆☆☆
- Author
-
WALSH, D, primary, GETTAYACAMIN, M, additional, LEITNER, W, additional, LYON, J, additional, STEWART, V, additional, MARIT, G, additional, PICHYANGKUL, S, additional, GOSI, P, additional, TONGTAWE, P, additional, and KESTER, K, additional
- Published
- 2006
- Full Text
- View/download PDF
12. Pre-clinical evaluation of the malaria vaccine candidate . MSP1 formulated with novel adjuvants or with alum
- Author
-
PICHYANGKUL, S, primary, GETTAYACAMIN, M, additional, MILLER, R, additional, LYON, J, additional, ANGOV, E, additional, TONGTAWE, P, additional, RUBLE, D, additional, HEPPNERJR, D, additional, KESTER, K, additional, and BALLOU, W, additional
- Published
- 2004
- Full Text
- View/download PDF
13. Monocyte Activation by Porphyromonas gingivalis LPS in Aggressive Periodontitis with the Use of Whole-blood Cultures
- Author
-
Mahanonda, R., primary, Sa-Ard-Iam, N., additional, Charatkulangkun, O., additional, Promsudthi, A., additional, Schifferle, R.E., additional, Yongvanichit, K., additional, and Pichyangkul, S., additional
- Published
- 2004
- Full Text
- View/download PDF
14. CpG oligodeoxynucleotide enhances immunity against blood-stage malaria infection in mice parenterally immunized with a yeast-expressed 19 kDa carboxyl-terminal fragment of Plasmodium yoelii merozoite surface protein-1 (MSP119) formulated in oil-based Montanides
- Author
-
Hirunpetcharat, C., primary, Wipasa, J., additional, Sakkhachornphop, S., additional, Nitkumhan, T., additional, Zheng, Y.Z., additional, Pichyangkul, S., additional, Krieg, A.M., additional, Walsh, D.S., additional, Heppner, D.G., additional, and Good, M.F., additional
- Published
- 2003
- Full Text
- View/download PDF
15. Regulation of Leukocyte Adhesion Molecules CD11b/CD18 and Leukocyte Adhesion Molecule-1 on Phagocytic Cells Activated by Malaria Pigment
- Author
-
Pichyangkul, S., primary, Webster, H. K., additional, Yongvanitchit, K., additional, Saengkrai, P., additional, Kyle, D. E., additional, and Heppner, D. G., additional
- Published
- 1997
- Full Text
- View/download PDF
16. Activation of γδ T Cells in Malaria: Interaction of Cytokines and a Schizont‐AssociatedPlasmodium falciparumAntigen
- Author
-
Pichyangkul, S., primary, Saengkrai, P., additional, and Yongvanitchit, K., additional
- Published
- 1997
- Full Text
- View/download PDF
17. Evaluation of the safety and immunogenicity of Plasmodium falciparum apical membrane antigen 1, merozoite surface protein 1 or RTS,S vaccines with adjuvant system AS02A administered alone or concurrently in rhesus monkeys
- Author
-
Pichyangkul, S., Tongtawe, P., Kum-Arb, U., Yongvanitchit, K., Gettayacamin, M., Hollingdale, M.R., Limsalakpetch, A., Stewart, V.A., Lanar, D.E., Dutta, S., Angov, E., Ware, L.A., Bergmann-Leitner, E.S., House, B., Voss, G., Dubois, M.C., Cohen, J.D., Fukuda, M.M., Heppner, D.G., and Miller, R.S.
- Subjects
- *
IMMUNOGENETICS , *MEDICATION safety , *PROTOZOAN vaccines , *PLASMODIUM falciparum , *ANTIGENS , *MICROBIAL proteins , *LABORATORY monkeys , *DRUG administration , *IMMUNE response - Abstract
Abstract: In an effort to broaden the immune response induced by the RTS,S/AS02A,vaccine, we have evaluated the immunogenicity of the RTS,S antigen when combined with MSP142 and with AMA1, antigens derived from the asexual blood stage. The objectives of this study were (i) to determine whether MSP142 and AMA1 vaccines formulated with the AS02A Adjuvant System were safe and immunogenic in the rhesus monkey model; (ii) to investigate whether MSP142 or AMA1 induced immune interference to each other, or to RTS,S, when added singly or in combinations at a single injection site; (iii) in the event of immune interference, to determine if this could be reduced when antigens were administered at separate sites. We found that MSP142 and AMA1 were safe and immunogenic, eliciting antibodies, and Th1 and Th2 responses using IFN-γ and IL-5 as markers. When malaria antigens were delivered together in one formulation, MSP142 and RTS,S reduced AMA1-specific antibody responses as measured by ELISA however, only MSP142 lowered parasite growth inhibitory activity of anti-AMA1 antibodies as measured by in vitro growth inhibition assay. Unlike RTS,S, MSP142 significantly reduced AMA1 IFN-γ and IL-5 responses. MSP142 suppression of AMA1 IFN-γ responses was not seen in animals receiving RTS,S+AMA1+MSP142 suggesting that RTS,S restored IFN-γ responses. Conversely, AMA1 had no effect on MSP142 antibody and IFN-γ and IL-5 responses. Neither AMA1 alone or combined with MSP142 affected RTS,S antibody or IFN-γ and IL-5 responses. Immune interference by MSP142 on AMA1 antibody responses was also evident when AMA1, MSP142 and RTS,S were administered concurrently at separate sites. These results suggest that immune interference may be complex and should be considered for the design of multi-antigen, multi-stage vaccines against malaria. [Copyright &y& Elsevier]
- Published
- 2009
- Full Text
- View/download PDF
18. Cigarette smoke extract modulates human beta-defensin-2 and interleukin-8 expression in human gingival epithelial cells.
- Author
-
Mahanonda R, Sa-Ard-Iam N, Eksomtramate M, Rerkyen P, Phairat B, Schaecher KE, Fukuda MM, and Pichyangkul S
- Abstract
BACKGROUND AND OBJECTIVE: Human gingival epithelial cells (HGECs) are continually exposed to oral bacteria and to other harmful agents. Their responses to stimuli are critical in maintaining periodontal homeostasis. The aim of this study was to investigate the modulating effect of cigarette smoke extract (CSE) on the innate immune responses of HGECs. MATERIAL AND METHODS: Toll-like receptor (TLR) expression of HGECs was determined by reverse transcriptase-polymerase chain reaction (RT-PCR). The effect of CSE or nicotine on the expression of the antimicrobial peptide human beta-defensin-2 (hBD-2) and the pro-inflammatory cytokine interleukin (IL)-8 in stimulated HGEC cultures was evaluated by RT-PCR and enzyme-linked immunosorbent assay. RESULTS: The HGECs expressed mRNA of TLRs 1, 2, 3, 5, 6, 9, 10, and minimally of TLR4, but not of TLRs 7 or 8. Stimulation of HGECs with highly purified TLR2, 3 or 5 ligands led to expression of hBD-2 and of IL-8. Enhancement of hBD-2 and IL-8 was observed in HGECs after combined stimulation with Porphyromonas gingivalis lipopolysaccharide (TLR2 ligand) and tumour necrosis factor-alpha, compared with stimulation using either agent alone. After CSE exposure, hBD-2 expression was markedly reduced in stimulated HGEC cultures, whereas IL-8 expression was markedly increased. These effects were also observed, but were markedly attenuated, upon nicotine treatment. CONCLUSION: Human gingival epithelial cells play a critical role in orchestrating the innate immune responses of periodontal tissue via TLR signalling. Our results represent the first demonstration that CSE can modulate HGEC function by suppressing hBD-2 and enhancing IL-8 production, and this may be, in part, a possible mechanism which promotes periodontal disease. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
19. Activation of gammadelta T cells in malaria: interaction of cytokines and a schizont-associated Plasmodium falciparum antigen.
- Author
-
Pichyangkul, S, Saengkrai, P, Yongvanitchit, K, Stewart, A, and Heppner, D G
- Abstract
A soluble Plasmodium falciparum antigen that specifically stimulates gammadelta T cells has been found associated predominantly with schizonts rather than ring forms, trophozoites, or gametocytes. This schizont-associated antigen (SAA) is resistant to protease digestion, is anionic at pH 8.5, is heat- and pH-resistant, and contains a phosphate group(s) that is crucial for biologic activity. Partially purified SAA induced proliferative responses and interferon-gamma production by gammadelta T cells. These stimulatory effects were greatly enhanced by monocyte-derived cytokines, interleukin (IL)-10, IL-12, and IL-1beta, but not by tumor necrosis factor-alpha. Taken together, these results suggest that concurrent stimulation of gammadelta T cells by SAA and by cytokines released from activated monocytes (IL-10, IL-12, IL-1beta) may represent the major mechanism underlying the selective activation of gammadelta T cells that is consistently observed in clinical cases of P. falciparum infection. [ABSTRACT FROM AUTHOR]
- Published
- 1997
20. Preclinical Evaluation of the Safety and Immunogenicity of a Vaccine Consisting of Plasmodium falciparumLiver-Stage Antigen 1 with Adjuvant AS01B Administered Alone or Concurrently with the RTS,S/AS01B Vaccine in Rhesus Primates
- Author
-
Pichyangkul, S., Kum-Arb, U., Yongvanitchit, K., Limsalakpetch, A., Gettayacamin, M., Lanar, D. E., Ware, L. A., Stewart, V. A., Heppner, D. G., Mettens, P., Cohen, J. D., Ballou, W. R., and Fukuda, M. M.
- Abstract
ABSTRACTSeveral lines of evidence suggest that targeting pre-erythrocytic-stage parasites for malaria vaccine development can provide sterile immunity. The objectives of this study were (i) to evaluate preclinically the safety and immunogenicity of a new recombinant pre-erythrocytic-stage antigen, liver-stage antigen 1 (LSA1), in nonhuman primates; and (ii) to investigate the potential for immune interference between LSA1 and the leading malaria vaccine candidate, RTS,S, by comparing the immune responses after single-antigen vaccination to responses after simultaneous administration of both antigens at separate sites. Using a rhesus monkey model, we found that LSA1 formulated with the GlaxoSmithKline proprietary adjuvant system AS01B (LSA1/AS01B) was safe and immunogenic, inducing high titers of antigen-specific antibody and CD4+T-cell responses, as monitored by the production of interleukin-2 and gamma interferon, using intracellular cytokine staining. RTS,S/AS01B vaccination was well tolerated and demonstrated robust antibody and moderate CD4+T-cell responses to circumsporozoite protein (CSP) and HBsAg. Positive CD8+T-cell responses to HBsAg were detected, whereas the responses to CSP and LSA1 were negligible. For both LSA1/AS01B and RTS,S/AS01B, no statistically significant differences were observed between individual and concurrent administration in the magnitude or duration of antibody and T-cell responses. Our results revealed that both pre-erythrocytic-stage antigens were safe and immunogenic, administered either separately or simultaneously to rhesus monkeys, and that no significant immune cross interference occurred with concurrent separate-site administration. The comparison of the profiles of immune responses induced by separate-site and single-site vaccinations with LSA1 and RTS,S warrants further investigation.
- Published
- 2008
- Full Text
- View/download PDF
21. Isolation and characterization of rhesus blood dendritic cells using flow cytometry
- Author
-
Pichyangkul, S., Saengkrai, P., Yongvanitchit, K., Limsomwong, C., Gettayacamin, M., Walsh, D. S., Stewart, V. A., Ballou, W. R., and Heppner, D. G.
- Published
- 2001
- Full Text
- View/download PDF
22. Whole blood cultures to assess the immunostimulatory activities of CpG oligodeoxynucleotides
- Author
-
Pichyangkul, S., Yongvanitchit, K., Kum-arb, U., Krieg, A. M., Heppner, D. G., and Walsh, D. S.
- Published
- 2001
- Full Text
- View/download PDF
23. Potentiation of Cytotoxic Effect of Lymphotoxin by Anti-Cancer Drugs and Elevated Temperatures
- Author
-
Pichyangkul, S., primary and Khan, A., additional
- Published
- 1986
- Full Text
- View/download PDF
24. Enhancement of Cell-Mediated Cytotoxicity by Recombinant Tumor Necrosis Factor (TNF )
- Author
-
Shimamoto, Y., primary, Pichyangkul, S., additional, and Khan, A., additional
- Published
- 1988
- Full Text
- View/download PDF
25. CpG oligodeoxynucleotide enhances immunity against blood-stage malaria infection in mice parenterally immunized with a yeast-expressed 19 kDa carboxyl-terminal fragment of Plasmodium yoelii merozoite surface protein-1 (MSP119) formulated in oil-based Montanides
- Author
-
Hirunpetcharat, C., Wipasa, J., Sakkhachornphop, S., Nitkumhan, T., Zheng, Y.Z., Pichyangkul, S., Krieg, A.M., Walsh, D.S., Heppner, D.G., and Good, M.F.
- Subjects
- *
MALARIA vaccines , *PROTEINS - Abstract
The 19 kDa carboxyl-terminal fragment of Plasmodium yoelii merozoite surface protein-1 (MSP119), an analog of the leading falciparum malaria vaccine candidate, induces protective immunity to challenge infection when formulated with complete/incomplete Freund’s adjuvant (CFA/IFA), an adjuvant unsuitable for use in humans. In this study, we investigate Montanide ISA51 and Montanide ISA720 as well as CpG oligodeoxynucleotide (ODN) as adjuvants for induction of immunity to MSP119. Mice immunized with MSP119 adjuvanted with Montanide ISA51 were protected even though some mice experienced low-grade parasitemia before resolving the infection. Mice immunized with MSP119 adjuvanted with Montanide ISA720 showed delayed patent parasitemia with all mice ultimately succumbing to infection. Interestingly, when the synthetic CpG ODN 1826 was included in either Montanide formulation, mice were completely protected with no parasites detected in the blood. MSP119-specific antibodies in MSP119-immunized mice adjuvanted with Montanide ISA51 or Montanide ISA720 showed predominantly IgG1 antibody and low levels of IgG2a. CpG ODN 1826 significantly enhanced both IgG1 and IgG2a antibody responses in Montanide ISA51-adjuvanted mice but significantly enhanced only the IgG2a antibody response in Montanide ISA720-adjuvanted mice. To investigate the relative roles of antibody and CD4+ T cells in protection, MSP119-immunized mice adjuvanted with Montanide ISA720 and CpG ODN 1826 were depleted of CD4+ T cells just prior to challenge. Results showed that three of nine immunized/T cell depleted mice died following infection. These results suggest that antibody and CD4+ T cells are critical for protection following immunization with MSP119 adjuvanted with Montanide and CpG ODN and that the formulation of a human malaria vaccine candidate in Montanide ISA720 or ISA51 together with human compatible CpG ODN would be useful for improving efficacy. [Copyright &y& Elsevier]
- Published
- 2003
- Full Text
- View/download PDF
26. Superior protection in a relapsing Plasmodium cynomolgi rhesus macaque model by a chemoprophylaxis with sporozoite immunization regimen with atovaquone-proguanil followed by primaquine.
- Author
-
Yongvanitchit K, Kum-Arb U, Limsalakpetch A, Im-Erbsin R, Ubalee R, Spring MD, Vesely BA, Waters N, and Pichyangkul S
- Subjects
- Animals, Primaquine therapeutic use, Sporozoites, Macaca mulatta, Immunization, Chemoprevention, CD8-Positive T-Lymphocytes, Drug Combinations, Plasmodium cynomolgi, Malaria Vaccines, Proguanil, Atovaquone
- Abstract
Background: To gain a deeper understanding of protective immunity against relapsing malaria, this study examined sporozoite-specific T cell responses induced by a chemoprophylaxis with sporozoite (CPS) immunization in a relapsing Plasmodium cynomolgi rhesus macaque model., Methods: The animals received three CPS immunizations with P. cynomolgi sporozoites, administered by mosquito bite, while under two anti-malarial drug regimens. Group 1 (n = 6) received artesunate/chloroquine (AS/CQ) followed by a radical cure with CQ plus primaquine (PQ). Group 2 (n = 6) received atovaquone-proguanil (AP) followed by PQ. After the final immunization, the animals were challenged with intravenous injection of 10
4 P. cynomolgi sporozoites, the dose that induced reliable infection and relapse rate. These animals, along with control animals (n = 6), were monitored for primary infection and subsequent relapses. Immunogenicity blood draws were done after each of the three CPS session, before and after the challenge, with liver, spleen and bone marrow sampling and analysis done after the challenge., Results: Group 2 animals demonstrated superior protection, with two achieving protection and two experiencing partial protection, while only one animal in group 1 had partial protection. These animals displayed high sporozoite-specific IFN-γ T cell responses in the liver, spleen, and bone marrow after the challenge with one protected animal having the highest frequency of IFN-γ+ CD8+ , IFN-γ+ CD4+ , and IFN-γ+ γδ T cells in the liver. Partially protected animals also demonstrated a relatively high frequency of IFN-γ+ CD8+ , IFN-γ+ CD4+ , and IFN-γ+ γδ T cells in the liver. It is important to highlight that the second animal in group 2, which experienced protection, exhibited deficient sporozoite-specific T cell responses in the liver while displaying average to high T cell responses in the spleen and bone marrow., Conclusions: This research supports the notion that local liver T cell immunity plays a crucial role in defending against liver-stage infection. Nevertheless, there is an instance where protection occurs independently of T cell responses in the liver, suggesting the involvement of the liver's innate immunity. The relapsing P. cynomolgi rhesus macaque model holds promise for informing the development of vaccines against relapsing P. vivax., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
27. Author Correction: A Pvs25 mRNA vaccine induces complete and durable transmission-blocking immunity to Plasmodium vivax.
- Author
-
Kunkeaw N, Nguitragool W, Takashima E, Kangwanrangsan N, Muramatsu H, Tachibana M, Ishino T, Lin PJC, Tam YK, Pichyangkul S, Tsuboi T, Pardi N, and Sattabongkot J
- Published
- 2024
- Full Text
- View/download PDF
28. A Pvs25 mRNA vaccine induces complete and durable transmission-blocking immunity to Plasmodium vivax.
- Author
-
Kunkeaw N, Nguitragool W, Takashima E, Kangwanrangsan N, Muramatsu H, Tachibana M, Ishino T, Lin PJC, Tam YK, Pichyangkul S, Tsuboi T, Pardi N, and Sattabongkot J
- Abstract
Plasmodium vivax (P. vivax) is the major malaria parasite outside of Africa and no vaccine is available against it. A vaccine that interrupts parasite transmission (transmission-blocking vaccine, TBV) is considered highly desirable to reduce the spread of P. vivax and to accelerate its elimination. However, the development of a TBV against this pathogen has been hampered by the inability to culture the parasite as well as the low immunogenicity of the vaccines developed to date. Pvs25 is the most advanced TBV antigen candidate for P. vivax. However, in previous phase I clinical trials, TBV vaccines based on Pvs25 yielded low antibody responses or had unacceptable safety profiles. As the nucleoside-modified mRNA-lipid nanoparticle (mRNA-LNP) vaccine platform proved to be safe and effective in humans, we generated and tested mRNA-LNP vaccines encoding several versions of Pvs25 in mice. We found that in a prime-boost vaccination schedule, all Pvs25 mRNA-LNP vaccines elicited robust antigen-specific antibody responses. Furthermore, when compared with a Pvs25 recombinant protein vaccine formulated with Montanide ISA-51 adjuvant, the full-length Pvs25 mRNA-LNP vaccine induced a stronger and longer-lasting functional immunity. Seven months after the second vaccination, vaccine-induced antibodies retained the ability to fully block P. vivax transmission in direct membrane feeding assays, whereas the blocking activity induced by the protein/ISA-51 vaccine dropped significantly. Taken together, we report on mRNA vaccines targeting P. vivax and demonstrate that Pvs25 mRNA-LNP outperformed an adjuvanted Pvs25 protein vaccine suggesting that it is a promising candidate for further testing in non-human primates., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
29. Preclinical evaluation of immunogenicity, efficacy and safety of a recombinant plant-based SARS-CoV-2 RBD vaccine formulated with 3M-052-Alum adjuvant.
- Author
-
Phoolcharoen W, Shanmugaraj B, Khorattanakulchai N, Sunyakumthorn P, Pichyangkul S, Taepavarapruk P, Praserthsee W, Malaivijitnond S, Manopwisedjaroen S, Thitithanyanont A, Srisutthisamphan K, Jongkaewwattana A, Tomai M, Fox CB, and Taychakhoonavudh S
- Subjects
- Animals, Mice, Rats, Rats, Sprague-Dawley, Aluminum Hydroxide, Adjuvants, Immunologic, Antibodies, Neutralizing, Macaca fascicularis, Antibodies, Viral, Spike Glycoprotein, Coronavirus genetics, Immunogenicity, Vaccine, SARS-CoV-2, COVID-19 prevention & control
- Abstract
Cost-effective, and accessible vaccines are needed for mass immunization to control the ongoing coronavirus disease 2019 (COVID-19), especially in low- and middle-income countries (LMIC).A plant-based vaccine is an attractive technology platform since the recombinant proteins can be easily produced at large scale and low cost. For the recombinant subunit-based vaccines, effective adjuvants are crucial to enhance the magnitude and breadth of immune responses elicited by the vaccine. In this study, we report a preclinical evaluation of the immunogenicity, efficacy and safety of a recombinant plant-based SARS-CoV-2 RBD vaccine formulated with 3M-052 (TLR7/8 agonist)-Alum adjuvant. This vaccine formulation, named Baiya SARS-CoV-2 Vax 2, induced significant levels of RBD-specific IgG and neutralizing antibody responses in mice. A viral challenge study using humanized K18-hACE2 mice has shown that animals vaccinated with two doses of Baiya SARS-CoV-2 Vax 2 established immune protection against SARS-CoV-2. A study in nonhuman primates (cynomolgus monkeys) indicated that immunization with two doses of Baiya SARS-CoV-2 Vax 2 was safe, well tolerated, and induced neutralizing antibodies against the prototype virus and other viral variants (Alpha, Beta, Gamma, Delta, and Omicron subvariants). The toxicity of Baiya SARS-CoV-2 Vax 2 was further investigated in Jcl:SD rats, which demonstrated that a single dose and repeated doses of Baiya SARS-CoV-2 Vax 2 were well tolerated and no mortality or unanticipated findings were observed. Overall, these preclinical findings support further clinical development of Baiya SARS-CoV-2 Vax 2., Competing Interests: Declaration of Competing Interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: ST and WP from Chulalongkorn University are founders/shareholders of Baiya Phytopharm Co., Ltd. Thailand. CBF from the Access to Advanced Health Institute is an inventor on patents and applications claiming rights in formulations of TLR 7/8 agonists such as 3M-052 and methods for using said formulations (e.g. WO/2017/200852A1)., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
30. Lymphostatin, a virulence factor of attaching and effacing Escherichia coli , inhibits proliferation and cytokine responses of human T cells in a manner associated with cell cycle arrest but not apoptosis or necrosis.
- Author
-
Ruamsap N, Riyapa D, Janesomboon S, Stevens JM, Pichyangkul S, Pattanapanyasat K, Demons ST, Stevens MP, and Korbsrisate S
- Subjects
- Apoptosis, CD8-Positive T-Lymphocytes immunology, Cell Cycle Checkpoints immunology, Cell Division, Cell Proliferation physiology, Cytokines biosynthesis, Cytokines immunology, Enteropathogenic Escherichia coli immunology, Enteropathogenic Escherichia coli pathogenicity, Humans, Interleukin-2, Interleukin-4, Leukocytes, Mononuclear immunology, Necrosis, Virulence Factors immunology, Bacterial Toxins immunology, Escherichia coli immunology, Escherichia coli pathogenicity, Escherichia coli Infections immunology, Escherichia coli Proteins immunology, T-Lymphocytes immunology
- Abstract
Lymphostatin is a virulence factor of enteropathogenic E. coli (EPEC) and non-O157 serogroup enterohaemorrhagic E. coli . Previous studies using whole-cell lysates of EPEC showed that lymphostatin inhibits the mitogen-activated proliferation of bulk human peripheral blood mononuclear cells (PBMCs) and the production of cytokines IL-2, IL-4, IL-5, and IFN-γ. Here, we used highly purified lymphostatin and PBMC-derived T cells to show that lymphostatin inhibits anti-CD3/anti-CD28-activated proliferation of human CD4
+ and CD8+ T cells and blocks the synthesis of IL-2, IL-4, IL-10 and IFN-γ without affecting cell viability and in a manner dependent on an N-terminal DTD glycosyltransferase motif. Such inhibition was not observed with T cells activated by phorbol 12-myristate 13-acetate and ionomycin, implying that lymphostatin targets T cell receptor signaling. Analysis of the expression of CD69 indicated that lymphostatin suppresses T cell activation at an early stage and no impacts on apoptosis or necrosis were observed. Flow cytometric analysis of the DNA content of lymphostatin-treated CD4+ and CD8+ T cells showed a concentration- and DTD-dependent accumulation of the cells in the G0/G1 phase of the cell cycle, and corresponding reduction of the percentage of cells in S phase. Consistent with this, we found a marked reduction in the abundance of cyclins D3, E and A and loss of phosphorylated Rb over time in activated T cells from 8 donors treated with lymphostatin. Moreover, the cyclin-dependent kinase (cdk) inhibitor p27kip1 , which inhibits progression of the cell cycle at G1 by acting on cyclin E-cdk2 or cyclin D-cdk4 complexes, was found to be accumulated in lymphostatin-treated T cells. Analysis of the abundance of phosphorylated kinases involved in signal transduction found that 30 of 39 were reduced in abundance following lymphostatin treatment of T cells from 5 donors, albeit not significantly so. Our data provide novel insights into the mode of action of lymphostatin on human T lymphocytes., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Ruamsap, Riyapa, Janesomboon, Stevens, Pichyangkul, Pattanapanyasat, Demons, Stevens and Korbsrisate.)- Published
- 2022
- Full Text
- View/download PDF
31. Prevalence of CYP2D6 Genotypes and Predicted Phenotypes in a Cohort of Cambodians at High Risk for Infections with Plasmodium vivax .
- Author
-
Spring MD, Lon C, Sok S, Sea D, Wojnarski M, Chann S, Kuntawunginn W, Kheang Heng T, Nou S, Arsanok M, Sriwichai S, Vanachayangkul P, Lin JT, Manning JE, Jongsakul K, Pichyangkul S, Satharath P, Smith PL, Dysoley L, Saunders DL, and Waters NC
- Subjects
- Artemisinins therapeutic use, Asian People genetics, Cambodia, Drug Therapy, Combination, Endemic Diseases, Gene Frequency, Genotype, Humans, Pharmacogenomic Variants, Phenotype, Plasmodium vivax, Polymorphism, Genetic, Quinolines therapeutic use, Recurrence, Treatment Failure, Antimalarials therapeutic use, Cytochrome P-450 CYP2D6 genetics, Malaria, Vivax drug therapy, Primaquine therapeutic use
- Abstract
Clinical failure of primaquine (PQ) has been demonstrated in people with CYP450 2D6 genetic polymorphisms that result in reduced or no enzyme activity. The distribution of CYP2D6 genotypes and predicted phenotypes in the Cambodian population is not well described. Surveys in other Asian countries have shown an approximate 50% prevalence of the reduced activity CYP2D6 allele *10, which could translate into increased risk of PQ radical cure failure and repeated relapses, making interruption of transmission and malaria elimination difficult to achieve. We determined CYP2D6 genotypes from 96 volunteers from Oddor Meanchey Province, Cambodia, an area endemic for Plasmodium vivax . We found a 54.2% frequency of the *10 allele, but in approximately half of our subjects, it was paired with a normal activity allele, either *1 or *2. The prevalence of *5, a null allele, was 9.4%. Overall predicted phenotype percentages were normal metabolizers, 46%; intermediate metabolizers, 52%; and poor metabolizers, 1%.
- Published
- 2020
- Full Text
- View/download PDF
32. Memory T cell subsets in healthy gingiva and periodontitis tissues.
- Author
-
Mahanonda R, Champaiboon C, Subbalekha K, Sa-Ard-Iam N, Yongyuth A, Isaraphithakkul B, Rerkyen P, Charatkulangkun O, and Pichyangkul S
- Subjects
- CD4-Positive T-Lymphocytes, CD8-Positive T-Lymphocytes, Humans, Immunologic Memory, T-Lymphocyte Subsets, Gingiva, Periodontitis
- Abstract
Background: In the gingival sulcus, effective and balanced innate and adaptive immune responses against subgingival plaque microbiome are crucial to maintain immune homeostasis. In this study, we investigated the memory T cell subsets in healthy gingiva and periodontitis tissues., Methods: Anatomical localization of T cells (CD3
+ , CD4+ , and CD8+ ) in healthy gingiva and periodontitis tissues were examined immunohistochemically. Subsets of memory T cells from isolated gingival cells were analyzed by flow cytometry using a cocktail of monoclonal antibodies (anti-CD69, anti-CD103, anti-CD45RA, anti-CCR7, anti-CD28, and anti-CD95). Intracellular cytokine staining of interleukin (IL)-17 and interferon (IFN)-γ expression on memory T cells in periodontitis tissues was also investigated., Results: We found that healthy gingiva contains two memory T cell populations; a CD69- recirculating population and a CD69+ gingiva-resident memory T cell population. CD4+ T cells with transitional memory (TTM ) phenotype (CD45RA- CCR7- CD28+ CD95+ ) constitute the major subset within these two populations. A significant increase in the proportion of CD4+ CD69+ CD103- memory T cells was observed in periodontitis tissues compared with healthy gingiva. CD4+ memory T cells from periodontitis tissues produced either IL-17 or IFN-γ whereas CD8+ memory T cells produced only IFN-γ., Conclusions: Our findings suggest that recirculating and gingiva-resident memory T cells could represent an important part of the immune surveillance network in the connective tissue, maintaining periodontal homeostasis. Imbalance of subgingival bacterial communities could damage gingival barrier allowing bacterial antigens to get access to the deeper connective tissue where they activate memory T cells leading to deleterious inflammation; a hallmark of periodontitis., (©2018 The Authors. Journal of Periodontology Published by Wiley Periodicals, Inc. on behalf of American Academy of Periodontology.)- Published
- 2018
- Full Text
- View/download PDF
33. Chemoprophylaxis with sporozoite immunization in P. knowlesi rhesus monkeys confers protection and elicits sporozoite-specific memory T cells in the liver.
- Author
-
Pichyangkul S, Spring MD, Yongvanitchit K, Kum-Arb U, Limsalakpetch A, Im-Erbsin R, Ubalee R, Vanachayangkul P, Remarque EJ, Angov E, Smith PL, and Saunders DL
- Subjects
- Animals, Anopheles parasitology, CD4-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes immunology, Macaca mulatta, Malaria immunology, Plasmodium growth & development, Plasmodium pathogenicity, Chemoprevention methods, Immunization methods, Immunologic Memory, Liver immunology, Malaria prevention & control, Plasmodium immunology, Sporozoites immunology
- Abstract
Whole malaria sporozoite vaccine regimens are promising new strategies, and some candidates have demonstrated high rates of durable clinical protection associated with memory T cell responses. Little is known about the anatomical distribution of memory T cells following whole sporozoite vaccines, and immunization of nonhuman primates can be used as a relevant model for humans. We conducted a chemoprophylaxis with sporozoite (CPS) immunization in P. knowlesi rhesus monkeys and challenged via mosquito bites. Half of CPS immunized animals developed complete protection, with a marked delay in parasitemia demonstrated in the other half. Antibody responses to whole sporozoites, CSP, and AMA1, but not CelTOS were detected. Peripheral blood T cell responses to whole sporozoites, but not CSP and AMA1 peptides were observed. Unlike peripheral blood, there was a high frequency of sporozoite-specific memory T cells observed in the liver and bone marrow. Interestingly, sporozoite-specific CD4+ and CD8+ memory T cells in the liver highly expressed chemokine receptors CCR5 and CXCR6, both of which are known for liver sinusoid homing. The majority of liver sporozoite-specific memory T cells expressed CD69, a phenotypic marker of tissue-resident memory (TRM) cells, which are well positioned to rapidly control liver-stage infection. Vaccine strategies that aim to elicit large number of liver TRM cells may efficiently increase the efficacy and durability of response against pre-erythrocytic parasites., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2017
- Full Text
- View/download PDF
34. Human Memory B Cells in Healthy Gingiva, Gingivitis, and Periodontitis.
- Author
-
Mahanonda R, Champaiboon C, Subbalekha K, Sa-Ard-Iam N, Rattanathammatada W, Thawanaphong S, Rerkyen P, Yoshimura F, Nagano K, Lang NP, and Pichyangkul S
- Subjects
- Enzyme-Linked Immunospot Assay, Flow Cytometry, Humans, Immunohistochemistry, Immunologic Memory immunology, Real-Time Polymerase Chain Reaction, B-Lymphocyte Subsets immunology, Gingiva immunology, Gingivitis immunology, Periodontitis immunology, Plasma Cells immunology
- Abstract
The presence of inflammatory infiltrates with B cells, specifically plasma cells, is the hallmark of periodontitis lesions. The composition of these infiltrates in various stages of homeostasis and disease development is not well documented. Human tissue biopsies from sites with gingival health (n = 29), gingivitis (n = 8), and periodontitis (n = 21) as well as gingival tissue after treated periodontitis (n = 6) were obtained and analyzed for their composition of B cell subsets. Ag specificity, Ig secretion, and expression of receptor activator of NF-κB ligand and granzyme B were performed. Although most of the B cell subsets in healthy gingiva and gingivitis tissues were CD19(+)CD27(+)CD38(-) memory B cells, the major B cell component in periodontitis was CD19(+)CD27(+)CD38(+)CD138(+)HLA-DR(low) plasma cells, not plasmablasts. Plasma cell aggregates were observed at the base of the periodontal pocket and scattered throughout the gingiva, especially apically toward the advancing front of the lesion. High expression of CXCL12, a proliferation-inducing ligand, B cell-activating factor, IL-10, IL-6, and IL-21 molecules involved in local B cell responses was detected in both gingivitis and periodontitis tissues. Periodontitis tissue plasma cells mainly secreted IgG specific to periodontal pathogens and also expressed receptor activator of NF-κB ligand, a bone resorption cytokine. Memory B cells resided in the connective tissue subjacent to the junctional epithelium in healthy gingiva. This suggested a role of memory B cells in maintaining periodontal homeostasis., (Copyright © 2016 by The American Association of Immunologists, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
35. Antibody profiles to plasmodium merozoite surface protein-1 in Cambodian adults during an active surveillance cohort with nested treatment study.
- Author
-
Spring MD, Pichyangkul S, Lon C, Gosi P, Yongvanichit K, Srichairatanakul U, Limsalakpeth A, Chaisatit C, Chann S, Sriwichai S, Auayapon M, Chaorattanakawee S, Dutta S, Prom S, Meng Chour C, Walsh DS, Angov E, and Saunders DL
- Subjects
- Adult, Antigens, Protozoan immunology, Artemisinins therapeutic use, Enzyme-Linked Immunosorbent Assay, Female, Humans, Malaria drug therapy, Malaria immunology, Malaria, Falciparum drug therapy, Male, Plasmodium falciparum immunology, Plasmodium falciparum pathogenicity, Plasmodium vivax immunology, Plasmodium vivax pathogenicity, Young Adult, Antibodies, Protozoan immunology, Malaria, Falciparum immunology, Merozoite Surface Protein 1 immunology
- Abstract
Background: In addition to evidence for a protective role of antibodies to the malaria blood stage antigen merozoite surface protein 1 (MSP1), MSP1 antibodies are also considered as a marker of past malaria exposure in sero-epidemiological studies., Methods: In order to better assess the potential use of MSP1 serology in malaria chemoprophylaxis trials in endemic areas, an analysis for the prevalence of antibodies to both Plasmodium falciparum and Plasmodium vivax MSP142 in healthy Cambodian adults was conducted at two sites as part of an active, observational cohort evaluating the efficacy of dihydroartemisinin-piperaquine (DP) for uncomplicated malaria (ClinicalTrials.gov identifier NCT01280162)., Results: Rates of baseline sero-positivity were high (59 and 73% for PfMSP142 and PvMSP142, respectively), and titers higher in those who lived in a higher transmission area, although there was little correlation in titers between the two species. Those volunteers who subsequently went on to develop malaria had higher baseline MSP142 titers than those who did not for both species. Titers to both antigens remained largely stable over the course of the 4-6 month study, except in those infected with P. falciparum who had multiple recurrences., Conclusion: These findings illuminate the difficulties in using MSP142 serology as either a screening criterion and/or biomarker of exposure in chemoprophylaxis studies. Further work remains to identify useful markers of malarial infection and/or immunity.
- Published
- 2016
- Full Text
- View/download PDF
36. Tissue Distribution of Memory T and B Cells in Rhesus Monkeys following Influenza A Infection.
- Author
-
Pichyangkul S, Yongvanitchit K, Limsalakpetch A, Kum-Arb U, Im-Erbsin R, Boonnak K, Thitithayanont A, Jongkaewwattana A, Wiboon-ut S, Mongkolsirichaikul D, Mahanonda R, Spring M, Chuang I, Mason CJ, and Saunders DL
- Subjects
- Age Factors, Animals, Antigens, CD immunology, Antigens, CD metabolism, Antigens, Differentiation, T-Lymphocyte immunology, Antigens, Differentiation, T-Lymphocyte metabolism, B-Lymphocytes metabolism, B-Lymphocytes virology, Bone Marrow immunology, Bone Marrow metabolism, Bone Marrow virology, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, CD8-Positive T-Lymphocytes virology, Cells, Cultured, Host-Pathogen Interactions immunology, Humans, Influenza A Virus, H1N1 Subtype physiology, Integrin alpha Chains immunology, Integrin alpha Chains metabolism, Interferon-gamma immunology, Interferon-gamma metabolism, Interleukin-2 immunology, Interleukin-2 metabolism, Lectins, C-Type immunology, Lectins, C-Type metabolism, Lung immunology, Lung metabolism, Lung virology, Lymph Nodes immunology, Lymph Nodes metabolism, Lymph Nodes virology, Macaca mulatta metabolism, Macaca mulatta virology, Mediastinum virology, Orthomyxoviridae Infections metabolism, Orthomyxoviridae Infections virology, Spleen immunology, Spleen metabolism, Spleen virology, T-Lymphocytes metabolism, T-Lymphocytes virology, Time Factors, B-Lymphocytes immunology, Immunologic Memory immunology, Influenza A Virus, H1N1 Subtype immunology, Macaca mulatta immunology, Orthomyxoviridae Infections immunology, T-Lymphocytes immunology
- Abstract
Studies of influenza-specific immune responses in humans have largely assessed systemic responses involving serum Ab and peripheral blood T cell responses. However, recent evidence indicates that tissue-resident memory T (TRM) cells play an important role in local murine intrapulmonary immunity. Rhesus monkeys were pulmonary exposed to 2009 pandemic H1N1 virus at days 0 and 28 and immune responses in different tissue compartments were measured. All animals were asymptomatic postinfection. Although only minimal memory immune responses were detected in peripheral blood, a high frequency of influenza nucleoprotein-specific memory T cells was detected in the lung at the "contraction phase," 49-58 d after second virus inoculation. A substantial proportion of lung nucleoprotein-specific memory CD8(+) T cells expressed CD103 and CD69, phenotypic markers of TRM cells. Lung CD103(+) and CD103(-) memory CD8(+) T cells expressed similar levels of IFN-γ and IL-2. Unlike memory T cells, spontaneous Ab secreting cells and memory B cells specific to influenza hemagglutinin were primarily observed in the mediastinal lymph nodes. Little difference in systemic and local immune responses against influenza was observed between young adult (6-8 y) and old animals (18-28 y). Using a nonhuman primate model, we revealed substantial induction of local T and B cell responses following 2009 pandemic H1N1 infection. Our study identified a subset of influenza-specific lung memory T cells characterized as TRM cells in rhesus monkeys. The rhesus monkey model may be useful to explore the role of TRM cells in local tissue protective immunity after rechallenge and vaccination., (Copyright © 2015 by The American Association of Immunologists, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
37. Dihydroartemisinin-piperaquine failure associated with a triple mutant including kelch13 C580Y in Cambodia: an observational cohort study.
- Author
-
Spring MD, Lin JT, Manning JE, Vanachayangkul P, Somethy S, Bun R, Se Y, Chann S, Ittiverakul M, Sia-ngam P, Kuntawunginn W, Arsanok M, Buathong N, Chaorattanakawee S, Gosi P, Ta-aksorn W, Chanarat N, Sundrakes S, Kong N, Heng TK, Nou S, Teja-isavadharm P, Pichyangkul S, Phann ST, Balasubramanian S, Juliano JJ, Meshnick SR, Chour CM, Prom S, Lanteri CA, Lon C, and Saunders DL
- Subjects
- Adolescent, Adult, Aged, Antimalarials pharmacology, Artemisinins pharmacology, Cambodia, Cohort Studies, Female, Humans, Male, Middle Aged, Mutation, Missense, Plasmodium falciparum genetics, Plasmodium falciparum isolation & purification, Point Mutation, Protozoan Proteins genetics, Quinolines pharmacology, Randomized Controlled Trials as Topic, Treatment Failure, Young Adult, Antimalarials therapeutic use, Artemisinins therapeutic use, Drug Resistance, Malaria, Falciparum drug therapy, Malaria, Falciparum parasitology, Plasmodium falciparum drug effects, Quinolines therapeutic use
- Abstract
Background: Dihydroartemisinin-piperaquine has been adopted as first-line artemisinin combination therapy (ACT) for multidrug-resistant Plasmodium falciparum malaria in Cambodia because of few remaining alternatives. We aimed to assess the efficacy of standard 3 day dihydroartemisinin-piperaquine treatment of uncomplicated P falciparum malaria, with and without the addition of primaquine, focusing on the factors involved in drug resistance., Methods: In this observational cohort study, we assessed 107 adults aged 18-65 years presenting to Anlong Veng District Hospital, Oddar Meanchey Province, Cambodia, with uncomplicated P falciparum or mixed P falciparum/Plasmodium vivax infection of between 1000 and 200,000 parasites per μL of blood, and participating in a randomised clinical trial in which all had received dihydroartemisinin-piperaquine for 3 days, after which they had been randomly allocated to receive either primaquine or no primaquine. The trial was halted early due to poor dihydroartemisinin-piperaquine efficacy, and we assessed day 42 PCR-corrected therapeutic efficacy (proportion of patients with recurrence at 42 days) and evidence of drug resistance from the initial cohort. We did analyses on both the intention to treat (ITT), modified ITT (withdrawals, losses to follow-up, and those with secondary outcomes [eg, new non-recrudescent malaria infection] were censored on the last day of follow-up), and per-protocol populations of the original trial. The original trial was registered with ClinicalTrials.gov, number NCT01280162., Findings: Between Dec 10, 2012, and Feb 18, 2014, we had enrolled 107 patients in the original trial. Enrolment was voluntarily halted on Feb 16, 2014, before reaching planned enrolment (n=150) because of poor efficacy. We had randomly allocated 50 patients to primaquine and 51 patients to no primaquine groups. PCR-adjusted Kaplan-Meier risk of P falciparum 42 day recrudescence was 54% (95% CI 45-63) in the modified ITT analysis population. We found two kelch13 propeller gene mutations associated with artemisinin resistance--a non-synonymous Cys580Tyr substitution in 70 (65%) of 107 participants, an Arg539Thr substitution in 33 (31%), and a wild-type parasite in four (4%). Unlike Arg539Thr, Cys580Tyr was accompanied by two other mutations associated with extended parasite clearance (MAL10:688956 and MAL13:1718319). This combination triple mutation was associated with a 5·4 times greater risk of treatment failure (hazard ratio 5·4 [95% CI 2·4-12]; p<0·0001) and higher piperaquine 50% inhibitory concentration (triple mutant 34 nM [28-41]; non-triple mutant 24 nM [1-27]; p=0·003) than other infections had. The drug was well tolerated, with gastrointestinal symptoms being the most common complaints., Interpretation: The dramatic decline in efficacy of dihydroartemisinin-piperaquine compared with what was observed in a study at the same location in 2010 was strongly associated with a new triple mutation including the kelch13 Cys580Tyr substitution. 3 days of artemisinin as part of an artemisinin combination therapy regimen might be insufficient. Strict regulation and monitoring of antimalarial use, along with non-pharmacological approaches to malaria resistance containment, must be integral parts of the public health response to rapidly accelerating drug resistance in the region., Funding: Armed Forces Health Surveillance Center/Global Emerging Infections Surveillance and Response System, Military Infectious Disease Research Program, National Institute of Allergy and Infectious Diseases, and American Society of Tropical Medicine and Hygiene/Burroughs Wellcome Fund., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
38. Pre-existing cross-reactive antibodies to avian influenza H5N1 and 2009 pandemic H1N1 in US military personnel.
- Author
-
Pichyangkul S, Krasaesub S, Jongkaewwattana A, Thitithanyanont A, Wiboon-Ut S, Yongvanitchit K, Limsalakpetch A, Kum-Arb U, Mongkolsirichaikul D, Khemnu N, Mahanonda R, Garcia JM, Mason CJ, Walsh DS, and Saunders DL
- Subjects
- Adult, Aged, Antibodies, Neutralizing, Female, Humans, Influenza, Human blood, Influenza, Human immunology, Male, Middle Aged, Neutralization Tests, Odds Ratio, United States, Antibodies, Viral blood, Influenza A Virus, H1N1 Subtype immunology, Influenza A Virus, H5N1 Subtype immunology, Influenza, Human virology, Military Personnel
- Abstract
We studied cross-reactive antibodies against avian influenza H5N1 and 2009 pandemic (p) H1N1 in 200 serum samples from US military personnel collected before the H1N1 pandemic. Assays used to measure antibodies against viral proteins involved in protection included a hemagglutination inhibition (HI) assay and a neuraminidase inhibition (NI) assay. Viral neutralization by antibodies against avian influenza H5N1 and 2009 pH1N1 was assessed by influenza (H5) pseudotyped lentiviral particle-based and H1N1 microneutralization assays. Some US military personnel had cross-neutralizing antibodies against H5N1 (14%) and 2009 pH1N1 (16.5%). The odds of having cross-neutralizing antibodies against 2009 pH1N1 were 4.4 times higher in subjects receiving more than five inactivated whole influenza virus vaccinations than those subjects with no record of vaccination. Although unclear if the result of prior vaccination or disease exposure, these pre-existing antibodies may prevent or reduce disease severity.
- Published
- 2014
- Full Text
- View/download PDF
39. Comparison of the immune responses induced by soluble and particulate Plasmodium vivax circumsporozoite vaccine candidates formulated in AS01 in rhesus macaques.
- Author
-
Vanloubbeeck Y, Pichyangkul S, Bayat B, Yongvanitchit K, Bennett JW, Sattabongkot J, Schaecher K, Ockenhouse CF, Cohen J, and Yadava A
- Subjects
- Animals, Antibodies, Protozoan blood, CD4-Positive T-Lymphocytes immunology, Cytokines metabolism, Escherichia coli genetics, Gene Expression, Macaca mulatta, Malaria Vaccines administration & dosage, Malaria Vaccines genetics, Plasmodium vivax genetics, Protozoan Proteins genetics, Saccharomyces cerevisiae genetics, Vaccines, Subunit administration & dosage, Vaccines, Subunit genetics, Vaccines, Subunit immunology, Vaccines, Synthetic administration & dosage, Vaccines, Synthetic genetics, Vaccines, Synthetic immunology, Vaccines, Virus-Like Particle administration & dosage, Vaccines, Virus-Like Particle genetics, Vaccines, Virus-Like Particle immunology, Adjuvants, Immunologic administration & dosage, Malaria Vaccines immunology, Plasmodium vivax immunology, Protozoan Proteins immunology
- Abstract
We have designed a pre-erythrocytic vaccine candidate based on the Plasmodium vivax circumsporozoite (CSV) protein, which includes its N- and C-terminal parts and a truncated region containing repeat sequences from both the VK210 and the VK247 P. vivax subtypes. Two versions of this vaccine candidate were made: a soluble recombinant protein expressed in Escherichia coli, designated VMP001 and a particulate antigen expressed in Saccharomyces cerevisiae, designated CSV-S,S. The latter is composed of CSV-S, a fusion protein between VMP001 and hepatitis B surface antigen (HBsAg), and free HBsAg co-expressed in yeast and self-assembling into mixed particles. Both antigen versions, adjuvanted with AS01, were shown to be immunogenic in rhesus monkeys. CSV-S,S/AS01 induced higher levels of VMP001-specific antibodies than did VMP001/AS01. Antibody responses against the N- and C-terminal regions of CSV and the VK210 repeat motif were of a similar magnitude following immunization with either the soluble or the particulate antigen. However, antibodies against the AGDR region, a potentially protective B cell epitope, were only detected after immunization with CSV-S,S. Analysis of the induced CD4(+) T cells highlighted different cytokine profiles depending on the antigen form. These results warrant further clinical evaluation of these two vaccine candidates to assess the added value of a particulate versus soluble form of CSV, in terms of both immunogenicity and protective efficacy., (Copyright © 2013 Elsevier Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
40. Generation and preclinical evaluation of a DENV-1/2 prM+E chimeric live attenuated vaccine candidate with enhanced prM cleavage.
- Author
-
Keelapang P, Nitatpattana N, Suphatrakul A, Punyahathaikul S, Sriburi R, Pulmanausahakul R, Pichyangkul S, Malasit P, Yoksan S, and Sittisombut N
- Subjects
- Aedes, Animals, Antibodies, Neutralizing blood, Antibodies, Viral blood, Antibody Formation, Dengue Virus immunology, Drug Evaluation, Preclinical, Macaca mulatta, Mice, Mice, Inbred BALB C, Reassortant Viruses genetics, Reassortant Viruses immunology, Vaccines, Attenuated immunology, Viremia prevention & control, Dengue prevention & control, Dengue Vaccines immunology, Dengue Virus genetics, Viral Envelope Proteins immunology
- Abstract
In the absence of a vaccine or sustainable vector control measures, illnesses caused by dengue virus infection remain an important public health problem in many tropical countries. During the export of dengue virus particles, furin-mediated cleavage of the prM envelope protein is usually incomplete, thus generating a mixture of immature, partially mature and mature extracellular particles. Variations in the arrangement and conformation of the envelope proteins among these particles may be associated with their different roles in shaping the antibody response. In an attempt to improve upon live, attenuated dengue vaccine approaches, a mutant chimeric virus, with enhanced prM cleavage, was generated by introducing a cleavage-enhancing substitution into a chimeric DENV-1/2 virus genome, encoding the prM+E sequence of a recent DENV-1 isolate under an attenuated DENV-2 genetic background. A modest increase in virus specific infectivity observed in the mutant chimeric virus affected neither the attenuation phenotype, when assessed in the suckling mouse neurovirulence model, nor multiplication in mosquitoes. The two chimeric viruses induced similar levels of anti-DENV-1 neutralizing antibody response in mice and rhesus macaques, but more efficient control of viremia during viral challenge was observed in macaques immunized with the mutant chimeric virus. These results indicate that the DENV-1/2 chimeric virus, with enhanced prM cleavage, could be useful as an alternative live, attenuated vaccine candidate for further tests in humans., (Copyright © 2013 Elsevier Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
41. Protective efficacy of baculovirus dual expression system vaccine expressing Plasmodium falciparum circumsporozoite protein.
- Author
-
Iyori M, Nakaya H, Inagaki K, Pichyangkul S, Yamamoto DS, Kawasaki M, Kwak K, Mizukoshi M, Goto Y, Matsuoka H, Matsumoto M, and Yoshida S
- Subjects
- Animals, Antibodies, Neutralizing blood, Antibodies, Neutralizing immunology, Antibodies, Protozoan blood, Antibodies, Protozoan immunology, Antibody Formation immunology, Antigens, Protozoan genetics, Baculoviridae immunology, Cell Line, Disease Models, Animal, Gene Expression, Gene Order, Genetic Vectors genetics, Genetic Vectors immunology, Humans, Macaca mulatta, Malaria Vaccines genetics, Mice, Plasmodium falciparum genetics, Protozoan Proteins genetics, T-Lymphocytes immunology, Vaccines, DNA genetics, Vaccines, DNA immunology, Antigens, Protozoan immunology, Baculoviridae genetics, Malaria Vaccines immunology, Malaria, Falciparum prevention & control, Plasmodium falciparum immunology, Protozoan Proteins immunology
- Abstract
We have previously developed a new malaria vaccine delivery system based on the baculovirus dual expression system (BDES). In this system, expression of malaria antigens is driven by a dual promoter consisting of the baculovirus-derived polyhedrin and mammal-derived cytomegalovirus promoters. To test this system for its potential as a vaccine against human malaria parasites, we investigated immune responses against the newly developed BDES-based Plasmodium falciparum circumsporozoite protein vaccines (BDES-PfCSP) in mice and Rhesus monkeys. Immunization of mice with BDES-PfCSP induced Th1/Th2-mixed type immune responses with high PfCSP-specific antibody (Ab) titers, and provided significant protection against challenge from the bites of mosquitoes infected with a transgenic P. berghei line expressing PfCSP. Next, we evaluated the immunogenicity of the BDES-PfCSP vaccine in a rhesus monkey model. Immunization of BDES-PfCSP elicited high levels of anti-PfCSP Ab responses in individual monkeys. Moreover, the sera from the immunized monkeys remarkably blocked sporozoite invasion of HepG2 cells. Taken together with two animal models, our results indicate that this novel vaccine platform (BDES) has potential clinical application as a vaccine against malaria.
- Published
- 2013
- Full Text
- View/download PDF
42. Evaluation of in vitro cross-reactivity to avian H5N1 and pandemic H1N1 2009 influenza following prime boost regimens of seasonal influenza vaccination in healthy human subjects: a randomised trial.
- Author
-
Bethell D, Saunders D, Jongkaewwattana A, Kramyu J, Thitithayanont A, Wiboon-ut S, Yongvanitchit K, Limsalakpetch A, Kum-Arb U, Uthaimongkol N, Garcia JM, Timmermans AE, Peiris M, Thomas S, Engering A, Jarman RG, Mongkolsirichaikul D, Mason C, Khemnu N, Tyner SD, Fukuda MM, Walsh DS, and Pichyangkul S
- Subjects
- Adolescent, Adult, Animals, Antibodies, Viral blood, Antibodies, Viral immunology, Birds, Feasibility Studies, Female, Health, Humans, Immunization, Secondary adverse effects, Influenza A Virus, H1N1 Subtype immunology, Influenza A Virus, H1N1 Subtype physiology, Influenza A Virus, H5N1 Subtype immunology, Influenza A Virus, H5N1 Subtype physiology, Influenza in Birds prevention & control, Influenza, Human epidemiology, Influenza, Human immunology, Male, Middle Aged, Orthomyxoviridae physiology, Pilot Projects, Safety, Seasons, T-Lymphocytes immunology, T-Lymphocytes virology, Vaccination adverse effects, Viral Vaccines adverse effects, Viral Vaccines immunology, Young Adult, Cross Reactions, Immunization, Secondary methods, Influenza in Birds immunology, Influenza, Human prevention & control, Orthomyxoviridae immunology, Pandemics prevention & control, Vaccination methods
- Abstract
Introduction: Recent studies have demonstrated that inactivated seasonal influenza vaccines (IIV) may elicit production of heterosubtypic antibodies, which can neutralize avian H5N1 virus in a small proportion of subjects. We hypothesized that prime boost regimens of live and inactivated trivalent seasonal influenza vaccines (LAIV and IIV) would enhance production of heterosubtypic immunity and provide evidence of cross-protection against other influenza viruses., Methods: In an open-label study, 26 adult volunteers were randomized to receive one of four vaccine regimens containing two doses of 2009-10 seasonal influenza vaccines administered 8 (±1) weeks apart: 2 doses of LAIV; 2 doses of IIV; LAIV then IIV; IIV then LAIV. Humoral immunity assays for avian H5N1, 2009 pandemic H1N1 (pH1N1), and seasonal vaccine strains were performed on blood collected pre-vaccine and 2 and 4 weeks later. The percentage of cytokine-producing T-cells was compared with baseline 14 days after each dose., Results: Subjects receiving IIV had prompt serological responses to vaccine strains. Two subjects receiving heterologous prime boost regimens had enhanced haemagglutination inhibition (HI) and neutralization (NT) titres against pH1N1, and one subject against avian H5N1; all three had pre-existing cross-reactive antibodies detected at baseline. Significantly elevated titres to H5N1 and pH1N1 by neuraminidase inhibition (NI) assay were observed following LAIV-IIV administration. Both vaccines elicited cross-reactive CD4+ T-cell responses to nucleoprotein of avian H5N1 and pH1N1. All regimens were safe and well tolerated., Conclusion: Neither homologous nor heterologous prime boost immunization enhanced serum HI and NT titres to 2009 pH1N1 or avian H5N1 compared to single dose vaccine. However heterologous prime-boost vaccination did lead to in vitro evidence of cross-reactivity by NI; the significance of this finding is unclear. These data support the strategy of administering single dose trivalent seasonal influenza vaccine at the outset of an influenza pandemic while a specific vaccine is being developed., Trial Registration: ClinicalTrials.gov NCT01044095.
- Published
- 2013
- Full Text
- View/download PDF
43. MxA expression induced by α-defensin in healthy human periodontal tissue.
- Author
-
Mahanonda R, Sa-Ard-Iam N, Rerkyen P, Thitithanyanont A, Subbalekha K, and Pichyangkul S
- Subjects
- 2',5'-Oligoadenylate Synthetase, Antimicrobial Cationic Peptides immunology, Antimicrobial Cationic Peptides metabolism, Epithelial Cells cytology, Epithelial Cells metabolism, GTP-Binding Proteins biosynthesis, Gingiva cytology, Gingiva metabolism, Humans, Influenza A Virus, H5N1 Subtype immunology, Influenza A Virus, H5N1 Subtype metabolism, Influenza, Human immunology, Influenza, Human metabolism, Interferon Type I immunology, Interferon Type I metabolism, Myxovirus Resistance Proteins, Protein Serine-Threonine Kinases immunology, Protein Serine-Threonine Kinases metabolism, Secretory Leukocyte Peptidase Inhibitor immunology, Secretory Leukocyte Peptidase Inhibitor metabolism, alpha-Defensins metabolism, beta-Defensins immunology, beta-Defensins metabolism, Cathelicidins, Epithelial Cells immunology, GTP-Binding Proteins immunology, Gene Expression Regulation immunology, Gingiva immunology, alpha-Defensins immunology
- Abstract
Although periodontal tissue is continually challenged by microbial plaque, it is generally maintained in a healthy state. To understand the basis for this, we investigated innate antiviral immunity in human periodontal tissue. The expression of mRNA encoding different antiviral proteins, myxovirus resistance A (MxA), protein kinase R (PKR), oligoadenylate synthetase (OAS), and secretory leukocyte protease inhibitor (SLPI) were detected in both healthy tissue and that with periodontitis. Immunostaining data consistently showed higher MxA protein expression in the epithelial layer of healthy gingiva as compared with tissue with periodontitis. Human MxA is thought to be induced by type I and III interferons (IFNs) but neither cytokine type was detected in healthy periodontal tissues. Treatment in vitro of primary human gingival epithelial cells (HGECs) with α-defensins, but not with the antimicrobial peptides β-defensins or LL-37, led to MxA protein expression. α-defensin was also detected in healthy periodontal tissue. In addition, MxA in α-defensin-treated HGECs was associated with protection against avian influenza H5N1 infection and silencing of the MxA gene using MxA-targeted-siRNA abolished this antiviral activity. To our knowledge, this is the first study to uncover a novel pathway of human MxA induction, which is initiated by an endogenous antimicrobial peptide, namely α-defensin. This pathway may play an important role in the first line of antiviral defense in periodontal tissue., (© 2012 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2012
- Full Text
- View/download PDF
44. Evaluation of the safety and immunogenicity in rhesus monkeys of a recombinant malaria vaccine for Plasmodium vivax with a synthetic Toll-like receptor 4 agonist formulated in an emulsion.
- Author
-
Lumsden JM, Pichyangkul S, Srichairatanakul U, Yongvanitchit K, Limsalakpetch A, Nurmukhambetova S, Klein J, Bertholet S, Vedvick TS, Reed SG, Sattabongkot J, Bennett JW, Polhemus ME, Ockenhouse CF, Howard RF, and Yadava A
- Subjects
- Adjuvants, Immunologic, Animals, Antigens, Protozoan immunology, CD4-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes, Emulsions, Enzyme-Linked Immunosorbent Assay, Fluorescent Antibody Technique, Immunoglobulin G blood, Interferon-gamma biosynthesis, Interleukin-2 biosynthesis, Interleukin-2 metabolism, Lipid A immunology, Macaca mulatta, Malaria, Vivax immunology, Protozoan Proteins immunology, Toll-Like Receptor 4 immunology, Tumor Necrosis Factor-alpha biosynthesis, Vaccines, Synthetic immunology, Malaria Vaccines immunology, Malaria, Vivax prevention & control, Plasmodium vivax immunology, Toll-Like Receptor 4 agonists
- Abstract
Plasmodium vivax is the major cause of malaria outside sub-Saharan Africa and inflicts debilitating morbidity and consequent economic impacts in developing countries. In order to produce a P. vivax vaccine for global use, we have previously reported the development of a novel chimeric recombinant protein, VMP001, based on the circumsporozoite protein (CSP) of P. vivax. Very few adjuvant formulations are currently available for human use. Our interest is to evaluate second-generation vaccine formulations to identify novel combinations of adjuvants capable of inducing strong, long-lasting immune responses. In this study rhesus monkeys were immunized intramuscularly three times with VMP001 in combination with a stable emulsion (SE) or a synthetic Toll-like receptor 4 (TLR4) agonist (glucopyranosyl lipid A [GLA]) in SE (GLA-SE). Sera and peripheral blood mononuclear cells (PBMCs) were tested for the presence of antigen-specific humoral and cellular responses, respectively. All groups of monkeys generated high titers of anti-P. vivax IgG antibodies, as detected by enzyme-linked immunosorbent assays (ELISAs) and immunofluorescence assays. In addition, all groups generated a cellular immune response characterized by antigen-specific CD4(+) T cells secreting predominantly interleukin-2 (IL-2) and lesser amounts of tumor necrosis factor (TNF). We conclude that the combination of VMP001 and GLA-SE is safe and immunogenic in monkeys and may serve as a potential second-generation vaccine candidate against P. vivax malaria.
- Published
- 2011
- Full Text
- View/download PDF
45. A single injection of 19 kda carboxy-terminal fragment of Plasmodium yoelii merozoite surface protein 1 (PyMSP1(19)) formulated with Montanide ISA and CpG ODN induces protective immune response in mice.
- Author
-
Hirunpetcharat C, Mahakunkijcharoen Y, Jeamwattanalert P, Kittigul L, Mahannop P, and Pichyangkul S
- Subjects
- Adjuvants, Immunologic metabolism, Animals, Female, Malaria Vaccines administration & dosage, Malaria Vaccines chemistry, Mannitol administration & dosage, Mannitol analogs & derivatives, Mannitol chemistry, Mannitol immunology, Merozoite Surface Protein 1 administration & dosage, Merozoite Surface Protein 1 chemistry, Mice, Mice, Inbred BALB C, Oleic Acids administration & dosage, Oleic Acids chemistry, Oleic Acids immunology, Oligodeoxyribonucleotides administration & dosage, Oligodeoxyribonucleotides chemistry, Immunoglobulin G blood, Immunoglobulin G immunology, Malaria Vaccines immunology, Merozoite Surface Protein 1 immunology, Oligodeoxyribonucleotides immunology, Plasmodium yoelii immunology
- Abstract
Objective: To investigate the efficacy of a vaccine formulation of the 19 kDa conserved carboxyl-terminal fragment of Plasmodium yoelii merozoite surface protein-1 (PyMSP1(19)) formulated with CpG ODN 1826 and Montanide ISA51 or ISA720 when used to immunize mice by a single injection., Methods: Groups of BALB/c mice were immunized parenterally with one, two or four injections with PBS or PyMSP1(19) formulated with CpG ODN in ISA51 or ISA720. Sera were collected weekly and assessed for total IgG and IgG subclass titers. Protection was tested by challenge infection with P. yoelii YM., Results: Interestingly, single injection immunization showed the same kinetics of antibody responses as two- or four-injection immunization. However, the peak antibody response induced by PyMSP1(19) in CpG ODN and ISA51 appeared earlier than that induced by PyMSP1(19) in CpG ODN and ISA720 (28 days vs 41 days). At day 63 after the first injection, the PyMSP1(19)-specific IgG antibody levels by single injection and four-injection immunizations were not different. However, the levels of the IgG2a antibody subclass were significantly lower by single injection immunization with PyMSP1(19) in CpG ODN and ISA720. The antibodies were sustained at high levels for at least 20 weeks. After challenge infection, all mice immunized by a single injection of PyMSP1(19) in CpG ODN and ISA51 survived with low-grade parasitemia, while 50% of mice immunized with PyMSP1(19) in CpG ODN and ISA720 died with high levels of parasitemia., Conclusion: These findings suggest that MSP1(19) immunization by a single injection can induce protective immunity, particularly when formulated with an appropriate strong adjuvant.
- Published
- 2011
46. Innate antiviral immunity of periodontal tissue.
- Author
-
Mahanonda R, Sa-Ard-Iam N, Rerkyen P, Champaiboon C, Vanavit N, and Pichyangkul S
- Subjects
- Humans, Mouth virology, Mouth Diseases etiology, Mouth Diseases virology, Saliva immunology, Salivary Proteins and Peptides immunology, Signal Transduction immunology, Signal Transduction physiology, Toll-Like Receptors immunology, Toll-Like Receptors physiology, Virus Diseases complications, Immunity, Innate immunology, Mouth immunology, Mouth Diseases immunology, Virus Diseases immunology
- Published
- 2011
- Full Text
- View/download PDF
47. Department of Defense influenza and other respiratory disease surveillance during the 2009 pandemic.
- Author
-
Burke RL, Vest KG, Eick AA, Sanchez JL, Johns MC, Pavlin JA, Jarman RG, Mothershead JL, Quintana M, Palys T, Cooper MJ, Guan J, Schnabel D, Waitumbi J, Wilma A, Daniels C, Brown ML, Tobias S, Kasper MR, Williams M, Tjaden JA, Oyofo B, Styles T, Blair PJ, Hawksworth A, Montgomery JM, Razuri H, Laguna-Torres A, Schoepp RJ, Norwood DA, Macintosh VH, Gibbons T, Gray GC, Blazes DL, Russell KL, Rubenstein J, Hathaway K, Gibbons R, Yoon IK, Saunders D, Gaywee J, Stoner M, Timmermans A, Shrestha SK, Velasco JM, Alera MT, Tannitisupawong D, Myint KS, Pichyangkul S, Woods B, Jerke KH, Koenig MG, Byarugaba DK, Mangen FW, Assefa B, Williams M, Brice G, Mansour M, Pimentel G, Sebeny P, Talaat M, Saeed T, Espinosa B, Faix D, Maves R, Kochel T, Smith J, Guerrero A, Maupin G, Sjoberg P, Duffy M, Garner J, Canas L, Macias E, Kuschner RA, Shanks D, Lewis S, Nowak G, Ndip LM, Wolfe N, and Saylors K
- Subjects
- Humans, Influenza, Human prevention & control, Military Medicine, Pandemics, Respiratory Tract Diseases prevention & control, United States epidemiology, United States Department of Defense, Global Health, Influenza A Virus, H1N1 Subtype, Influenza, Human epidemiology, Respiratory Tract Diseases epidemiology, Sentinel Surveillance
- Abstract
The Armed Forces Health Surveillance Center's Division of Global Emerging Infections Surveillance and Response System (AFHSC-GEIS) supports and oversees surveillance for emerging infectious diseases, including respiratory diseases, of importance to the U.S. Department of Defense (DoD). AFHSC-GEIS accomplishes this mission by providing funding and oversight to a global network of partners for respiratory disease surveillance. This report details the system's surveillance activities during 2009, with a focus on efforts in responding to the novel H1N1 Influenza A (A/H1N1) pandemic and contributions to global public health. Active surveillance networks established by AFHSC-GEIS partners resulted in the initial detection of novel A/H1N1 influenza in the U.S. and several other countries, and viruses isolated from these activities were used as seed strains for the 2009 pandemic influenza vaccine. Partners also provided diagnostic laboratory training and capacity building to host nations to assist with the novel A/H1N1 pandemic global response, adapted a Food and Drug Administration-approved assay for use on a ruggedized polymerase chain reaction platform for diagnosing novel A/H1N1 in remote settings, and provided estimates of seasonal vaccine effectiveness against novel A/H1N1 illness. Regular reporting of the system's worldwide surveillance findings to the global public health community enabled leaders to make informed decisions on disease mitigation measures and controls for the 2009 A/H1N1 influenza pandemic. AFHSC-GEIS's support of a global network contributes to DoD's force health protection, while supporting global public health.
- Published
- 2011
- Full Text
- View/download PDF
48. Antiviral immune responses in H5N1-infected human lung tissue and possible mechanisms underlying the hyperproduction of interferon-inducible protein IP-10.
- Author
-
Thitithanyanont A, Engering A, Uiprasertkul M, Ekchariyawat P, Wiboon-Ut S, Kraivong R, Limsalakpetch A, Kum-Arb U, Yongvanitchit K, Sa-Ard-Iam N, Rukyen P, Mahanonda R, Kawkitinarong K, Auewarakul P, Utaisincharoen P, Sirisinha S, Mason CJ, Fukuda MM, and Pichyangkul S
- Subjects
- Cells, Cultured, Chemokine CXCL10 antagonists & inhibitors, DEAD Box Protein 58, DEAD-box RNA Helicases metabolism, GTP-Binding Proteins biosynthesis, Humans, Interferon-alpha biosynthesis, Interferon-alpha pharmacology, Methylprednisolone pharmacology, Myxovirus Resistance Proteins, Receptors, Immunologic, Tumor Necrosis Factor-alpha metabolism, Tumor Necrosis Factor-alpha pharmacology, Chemokine CXCL10 biosynthesis, Influenza A Virus, H5N1 Subtype, Influenza, Human immunology, Lung immunology, Lung virology, Pneumonia, Viral immunology
- Abstract
Information on the immune response against H5N1 within the lung is lacking. Here we describe the sustained antiviral immune responses, as indicated by the expression of MxA protein and IFN-alpha mRNA, in autopsy lung tissue from an H5N1-infected patient. H5N1 infection of primary bronchial/tracheal epithelial cells and lung microvascular endothelial cells induced IP-10, and also up-regulated the retinoic acid-inducible gene-I (RIG-I). Down-regulation of RIG-I gene expression decreased IP-10 response. Co-culturing of H5N1-infected pulmonary cells with TNF-alpha led to synergistically enhanced production of IP-10. In the absence of viral infection, TNF-alpha and IFN-alpha also synergistically enhanced IP-10 response. Methylprednisolone showed only a partial inhibitory effect on this chemokine response. Our findings strongly suggest that both the H5N1 virus and the locally produced antiviral cytokines; IFN-alpha and TNF-alpha may have an important role in inducing IP-10 hyperresponse, leading to inflammatory damage in infected lung., (Published by Elsevier Inc.)
- Published
- 2010
- Full Text
- View/download PDF
49. Cross-reactive Antibodies against avian influenza virus A (H5N1).
- Author
-
Pichyangkul S, Jongkaewwattana A, Thitithanyanont A, Ekchariyawat P, Wiboon-ut S, Limsalakpetch A, Yongvanitchit K, Kum-Arb U, Mahanonda R, Utaisincharoen P, Sirisinha S, Mason CJ, and Fukuda MM
- Subjects
- Animals, Birds virology, Cross Reactions immunology, Humans, Immunoglobulins, Intravenous administration & dosage, Influenza A Virus, H1N1 Subtype immunology, Influenza A Virus, H3N2 Subtype immunology, Influenza A Virus, H5N1 Subtype physiology, Influenza in Birds virology, Influenza, Human virology, Neuraminidase antagonists & inhibitors, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, Immunoglobulins, Intravenous immunology, Influenza A Virus, H5N1 Subtype immunology
- Published
- 2009
- Full Text
- View/download PDF
50. Expression and function of Toll-like receptors on dendritic cells and other antigen presenting cells from non-human primates.
- Author
-
Ketloy C, Engering A, Srichairatanakul U, Limsalakpetch A, Yongvanitchit K, Pichyangkul S, and Ruxrungtham K
- Subjects
- Adjuvants, Immunologic pharmacology, Animals, Antigen-Presenting Cells immunology, Antigens, CD immunology, B-Lymphocytes immunology, Cytokines immunology, Female, Humans, Male, Mice, Mice, Inbred BALB C, RNA, Messenger biosynthesis, RNA, Messenger genetics, Reverse Transcriptase Polymerase Chain Reaction veterinary, Toll-Like Receptors biosynthesis, Toll-Like Receptors genetics, Dendritic Cells immunology, Macaca mulatta immunology, Toll-Like Receptors immunology
- Abstract
Antigen presenting cells (APCs), especially dendritic cells (DCs), play a crucial role in immune responses against infections by sensing microbial invasion through Toll-like receptors (TLRs). In this regard, TLR ligands are attractive candidates for use in humans and animal models as vaccine adjuvants. So far, no studies have been performed on TLR expression in non-human primates such as rhesus macaques. Therefore, we studied the TLR expression patterns in different subsets of APC in rhesus macaques and compared them to similar APC subsets in human. Also, expression was compared with corresponding DC subsets from different organs from mice. Here we show by semi-quantitative RT-PCR, that blood DC subsets of rhesus macaque expressed the same sets of TLRs as those of human but substantially differed from mouse DC subsets. Macaque myeloid DCs (MDCs) expressed TLR3, 4, 7 and 8 whereas macaque plasmacytoid DCs (PDCs) expressed only TLR7 and 9. Additionally, TLR expression patterns in macaque monocyte-derived dendritic cells (mo-DCs) (i.e., TLR3, 4, 8 and 9), monocytes (i.e., TLR4, 7, and 8) and B cells (i.e., TLR4, 7, 8, and 9) were also similar to their human counterparts. However, the responsiveness of macaque APCs to certain TLR ligands partially differed from that of human in terms of phenotype differentiation and cytokine production. Strikingly, in contrast to human mo-DCs, no IL-12p70 production was observed when macaque mo-DCs were stimulated with TLR ligands. In addition, CD40 and CD86 phenotypic responses to TLR8 ligand (poly U) in mo-DCs of macaque were higher than that of human. Despite these functional differences, our results provide important information for a rational design of animal models in evaluating TLR ligands as adjuvant in vivo.
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.