Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Santi Maneewatch is active.

Publication


Featured researches published by Santi Maneewatch.


Journal of Proteomics | 2009

Human monoclonal ScFv neutralize lethal Thai cobra, Naja kaouthia, neurotoxin.

Kasem Kulkeaw; Yuwaporn Sakolvaree; Potjanee Srimanote; Pongsri Tongtawe; Santi Maneewatch; Nitat Sookrung; Anchalee Tungtrongchitr; Pramuan Tapchaisri; Hisao Kurazono; Wanpen Chaicumpa

Animal derived anti-Naja. kaouthia (Thai cobra) venom is used for specific treatment of the snake bitten victims. Many recipients develop allergic reaction or anti-isotype response which causes serum sickness. A better therapeutic antibody is needed. In this study, long alpha-neurotoxin was purified from the N. kaouthia holovenom and verified by 2D-LC/MS-MS. The toxin was used as antigen in a phage bio-panning to select phage clones displaying human single chain variable antibody fragments (HuScFv) from a phage display antibody library constructed from immunoglobulin genes of non-immunized Thai blood donors. HuScFv that specifically bound to the neurotoxin were produced from huscfv-phagemid transformed E. coli clones and affinity purified. The HuScFv could neutralize toxicity of the N. kaouthia neurotoxin and rescued the envenomized mice from the neurotoxin mediated lethality. Peptide mimotope of the neutralizing HuScFv matched with an amino acid sequence (epitope) located in the loop-3 of the N. kaouthia long alpha-neurotoxin which functions in acetylcholine receptor binding. The mimotope is also similar to peptide sequences found on other snake venom neurotoxins implying a possibility of the HuScFv to exert pan-neutralizing activity against multiple snake neurotoxins.


Bioconjugate Chemistry | 2010

A Human Single Chain Transbody Specific to Matrix Protein (M1) Interferes with the Replication of Influenza A Virus

Ornnuthchar Poungpair; Anek Pootong; Santi Maneewatch; Potjanee Srimanote; Pongsri Tongtawe; Thaweesak Songserm; Pramuan Tapchaisri; Wanpen Chaicumpa

A cell penetrating format of human single chain antibody (HuScFv) specific to matrix protein (M1) of influenza A virus was produced by molecular linking of the gene sequence encoding the HuScFv (huscfv) to a protein transduction domain, i.e., penetratin (PEN) of the Drosophila homeodomain. DNA of a recombinant phagemid vector carrying the huscfv was used as a platform template in a three-step PCR for generating a nucleotide sequence encoding a 16 amino acid PEN peptide. The PEN-HuScFv had negligible cytotoxicity on living MDCK cells. They were readily translocated across the cell membrane and bound to native M1 in the A/H5N1-infected cells as revealed by immunofluorescent confocal microscopy. The PEN-HuScFv, when used to treat the influenza virus infected cells, reduced the number of viruses released from the cells. In conclusion, the cell penetrating M1-specific HuScFv, a transbody, produced in this study affected the influenza A virus life cycle in living mammalian cells. While the molecular mechanisms of the PEN-HuScFv need more investigation, the reagent warrants further testing in animals before developing it into a human immunotherapeutic anti-influenza formula.


PLOS ONE | 2012

Cell Penetrable Humanized-VH/VHH That Inhibit RNA Dependent RNA Polymerase (NS5B) of HCV

Kanyarat Thueng-in; Jeeraphong Thanongsaksrikul; Potjanee Srimanote; Kunan Bangphoomi; Ornnuthchar Poungpair; Santi Maneewatch; Kiattawee Choowongkomon; Wanpen Chaicumpa

NS5B is pivotal RNA dependent RNA polymerase (RdRp) of HCV and NS5B function interfering halts the virus infective cycle. This work aimed to produce cell penetrable humanized single domain antibodies (SdAb; VH/VHH) that interfere with the RdRp activity. Recombinant NS5BΔ55 of genotype 3a HCV with de novo RNA synthetic activity was produced and used in phage biopanning for selecting phage clones that displayed NS5BΔ55 bound VH/VHH from a humanized-camel VH/VHH display library. VH/VHH from E. coli transfected with four selected phage clones inhibited RdRp activity when tested by ELISA inhibition using 3′di-cytidylate 25 nucleotide directed in vitro RNA synthesis. Deduced amino acid sequences of two clones showed VHH hallmark and were designated VHH6 and VHH24; other clones were conventional VH, designated VH9 and VH13. All VH/VHH were linked molecularly to a cell penetrating peptide, penetratin. The cell penetrable VH9, VH13, VHH6 and VHH24 added to culture of Huh7 cells transfected with JHF-1 RNA of genotype 2a HCV reduced the amounts of RNA intracellularly and in culture medium implying that they inhibited the virus replication. VH/VHH mimotopes matched with residues scattered on the polymerase fingers, palm and thumb which were likely juxtaposed to form conformational epitopes. Molecular docking revealed that the antibodies covered the RdRp catalytic groove. The transbodies await further studies for in vivo role in inhibiting HCV replication.


Vaccine | 2010

Heterosubtypic immunity to influenza mediated by liposome adjuvanted H5N1 recombinant protein vaccines.

Kanyarat Thueng-in; Santi Maneewatch; Potjanee Srimanote; Thaweesak Songserm; Pramuan Tapchaisri; Nitat Sookrung; Pongsri Tongtawe; Sunee Channarong; Wanpen Chaicumpa

A non-egg, non-culture based influenza vaccine that intervenes large influenza outbreaks and protects against heterosubtypic infections is needed. Candidates of such vaccine are likely to be conserved influenza virus proteins or their coding DNA. The vaccine must be conveniently produced at reasonable cost, safe, highly immunogenic and should be able to recall rapidly the immunological memory upon the antigenic re-exposure. In this study vaccines made of full length recombinant NP and M2 of the H5N1 influenza A virus were entrapped either alone or together into liposome (L) made of phosphatidylcholine and cholesterol. The vaccines (L-NP, L-M2 or L-NP+M2) and mocks (L or PBS) were safe without causing any adverse reaction in the intramuscularly injected mice. They were readily immunogenic at a single dose and a recalled response could be detected within one day post booster. Cytokine and antibody data indicated that the vaccines induced a Th1 bias immune response. NP containing vaccines stimulated a marked increase of cytotoxic lymphocytes, i.e., CD8(+), intracellular IFNγ(+) cells, while M2 containing vaccines elicited good antibody response which neutralized infectivity of heterologous influenza viruses. Although the three vaccines elicited different immunological defense factors; nevertheless, they similarly and readily abrogated lung histopathology mediated by viruses belonging to different H5N1 clade/subclade and heterosubtypes including swine H1N1 and human H1N1/2009 viruses. They protected the vaccinated mice against lethal challenges with mouse adapted avian H5N1 virus. The liposome adjuvanted vaccines which demonstrated high protective efficacy in mice warrant testing further in a non-rodent model as well as in humans.


Journal of Virological Methods | 2009

Human single chain monoclonal antibody that recognizes matrix protein of heterologous influenza A virus subtypes.

Ornnuthchar Poungpair; Wanpen Chaicumpa; Kasem Kulkeaw; Santi Maneewatch; Kanyarat Thueng-in; Potjanee Srimanote; Pongsri Tongtawe; Thaweesak Songserm; Porntippa Lekcharoensuk; Pramuan Tapchaisri

Matrix protein (M1) is predominant and has pivotal role in the influenza A virus replication and assembly. It is therefore an attractive target for antiviral drugs, siRNA studies, and therapeutic antibodies. Nevertheless, therapeutic antibody that interferes with the M1 multiplex function has never been developed. In this study, human single monoclonal antibody fragments (HuScFvs) to M1 were generated. Full length recombinant M1 (rM1) was produced from cDNA prepared from genome of highly pathogenic avian influenza virus, A/H5N1. The rM1 was used as an antigen in phage bio-panning to select phage clones displaying HuScFv from a human antibody phage display library. Several phage clones displaying HuScFv bound to the rM1 and harboring the respective huscfv gene inserts were isolated. RFLP experiments revealed multiple DNA banding patterns which indicated epitope/affinity diversity of the HuScFv. The HuScFv were tested for their binding to native M1 of homologous and heterologous influenza A viruses using ELISA as well as incorporating immunostaining and immunofluorescence studies with infected MDCK cells. One such protein produced from a selected phage clone blocked binding of M1 to viral RNA. The HuScFv in their in vivo functional format, e.g. cell-penetrating molecules, should be developed and tested as a broad spectrum anti-A/influenza.


Hybridoma | 2008

Monoclonal Antibodies to LipL32 Protect Against Heterologous Leptospira spp. Challenge

Santi Maneewatch; Yuwaporn Sakolvaree; Patcharin Saengjaruk; Potjanee Srimanote; Pramuan Tapchaisri; Pongsri Tongtawe; Buppa Klaysing; Surasakdi Wongratanacheewin; Manas Chongsa-nguan; Wanpen Chaicumpa

A non-culture-based leptospirosis vaccine that cross-protects against infection caused by heterologous Leptospira spp. should replace the currently available products, which are qualitatively and quantitatively inadequate. With that in mind, two murine hybridomas secreting monoclonal antibodies (MAb) binding only to homogenates of pathogenic Leptospira spp., and not of the saprophytic L. biflexa, serogroup Patoc, serovar Patoc, were produced. The MAbs of both clones neutralized Leptospira-mediated human red blood cell lysis in vitro and rescued hamsters from lethal infection with heterologous Leptospira spp. The orthologous Leptospira spp. protein carrying the MAb epitope(s) was identified by two-dimensional gel electrophoresis (2DE)-based proteomics and database search. The epitopes of the MAbs were located on the major outer membrane protein LipL32 of the pathogenic Leptospira spp. The MAbs in their humanized version are potential leptospirosis immunotherapeutics. They are also suitable as detection reagents in antigen-based assays for the rapid diagnosis of leptospirosis. Recombinant LipL32 is a good candidate for a broad spectrum, non-culture-based leptospirosis vaccine.


Virology Journal | 2013

Human monoclonal ScFv that bind to different functional domains of M2 and inhibit H5N1 influenza virus replication

Tippawan Pissawong; Santi Maneewatch; Kanyarat Thueng-in; Potjanee Srimanote; Fonthip Dong-din-on; Jeeraphong Thanongsaksrikul; Thaweesak Songserm; Pongsri Tongtawe; Kunan Bangphoomi; Wanpen Chaicumpa

BackgroundNovel effective anti-influenza agent that tolerates influenza virus antigenic variation is needed. Highly conserved influenza virus M2 protein has multiple pivotal functions including ion channel activity for vRNP uncoating, anti-autophagy and virus assembly, morphogenesis and release. Thus, M2 is an attractive target of anti-influenza agents including small molecular drugs and specific antibodies.MethodsFully human monoclonal single chain antibodies (HuScFv) specific to recombinant and native M2 proteins of A/H5N1 virus were produced from huscfv-phagemid transformed E. coli clones selected from a HuScFv phage display library using recombinant M2 of clade 1 A/H5N1 as panning antigen. The HuScFv were tested for their ability to inhibit replication of A/H5N1 of both homologous and heterologous clades. M2 domains bound by HuScFv of individual E. coli clones were identified by phage mimotope searching and computerized molecular docking.ResultsHuScFv derived from four huscfv-phagemid transformed E. coli clones (no. 2, 19, 23 and 27) showed different amino acid sequences particularly at the CDRs. Cells infected with A/H5N1 influenza viruses (both adamantane sensitive and resistant) that had been exposed to the HuScFv had reduced virus release and intracellular virus. Phage peptide mimotope search and multiple alignments revealed that conformational epitopes of HuScFv2 located at the residues important for ion channel activity, anti-autophagy and M1 binding; epitopic residues of HuScFv19 located at the M2 amphipathic helix and cytoplasmic tail important for anti-autophagy, virus assembly, morphogenesis and release; epitope of HuScFv23 involved residues important for the M2 activities similar to HuScFv2 and also amphipathic helix residues for viral budding and release while HuScFv27 epitope spanned ectodomain, ion channel and anti-autophagy residues. Results of computerized homology modelling and molecular docking conformed to the epitope identification by phages.ConclusionsHuScFv that bound to highly conserved epitopes across influenza A subtypes and human pathogenic H5N1clades located on different functional domains of M2 were produced. The HuScFv reduced viral release and intracellular virus of infected cells. While the molecular mechanisms of the HuScFv await experimental validation, the small human antibody fragments have high potential for developing further as a safe, novel and mutation tolerable anti-influenza agent especially against drug resistant variants.


Protein Engineering Design & Selection | 2009

Humanized-monoclonal antibody against heterologous Leptospira infection

Santi Maneewatch; Yuwaporn Sakolvaree; Pramuan Tapchaisri; Patcharin Saengjaruk; Thaweesak Songserm; Surasakdi Wongratanachewin; Pongsri Tongtawe; Potjanee Srimanote; Urai Chaisri; Wanpen Chaicumpa

Patients with leptospirosis are commonly treated with antibiotics. Jarisch-Herxheimer reaction caused by toxic bacterial substances massively released as a result of the antibiotic mediated-bacterial lysis occurs in some patients which may aggravate the existing severe clinical manifestations. In this study, a humanized-murine single-chain monoclonal antibody (HuScFv) was produced and tested as an alternative of antibiotics for treatment of leptospirosis. Complementary DNA was prepared from total RNA of a murine hybridoma clone secreting monoclonal antibody (MAb) specific to LipL32 of pathogenic Leptospira spp. The MAb had therapeutic efficacy in Leptospira challenged hamsters. The VH and VL coding sequences were amplified using the cDNA as a template. The sequences were linked to form a single-chain variable murine DNA fragment (muscFv). CDR sequences of the muscFv were grafted onto the best matching human VH and VL immunoglobulin frameworks. After cloning of the humanized murine DNA sequences (huscFv) into a phagemid vector and the vector was introduced into competent Escherichia coli, the HuScFv was produced. On the same weight basis, the HuScFv possessed equal neutralizing activities to the murine ScFv counterpart against heterologous Leptospira-mediated hemolysis in vitro and rescued hamsters from a heterologous Leptospira lethal challenge. The HuScFv antibody has high therapeutic potential as an alternative to antibiotics for human leptospirosis, especially for drug hypersensitive patients.


Antiviral Research | 2013

Human monoclonal ScFv specific to NS1 protein inhibits replication of influenza viruses across types and subtypes

Rungrueang Yodsheewan; Santi Maneewatch; Potjanee Srimanote; Kanyarat Thueng-in; Thaweesak Songserm; Fonthip Dong-din-on; Kunan Bangphoomi; Nitat Sookrung; Kiattawee Choowongkomon; Wanpen Chaicumpa

Currently, there is a need of new anti-influenza agents that target influenza virus proteins other than ion channel M2 and neuraminidase. Non-structural protein-1 (NS1) is a highly conserved multifunctional protein which is indispensable for the virus replication cycle. In this study, fully human single chain antibody fragments (HuScFv) that bound specifically to recombinant and native NS1 were produced from three huscfv-phagemid transformed Escherichia coli clones (nos. 3, 10 and 11) selected from a human ScFv phage display library. Western blot analysis, mimotope searching/epitope identification, homology modeling/molecular docking and phage mimotope ELISA inhibition indicated that HuScFv of clone no. 3 reacted with NS1 R domain important for host innate immunity suppression; HuScFv of clone nos. 10 and 11 bound to E domain sites necessary for NS1 binding to the host eIF4GI and CPSF30, respectively. The HuScFv of all clones could enter the influenza virus infected cells and interfered with the NS1 activities leading to replication inhibition of viruses belonging to various heterologous A subtypes and type B by 2-64-fold as semi-quantified by hemagglutination assay. Influenza virus infected cells treated with representative HuScFv (clone 10) had up-expression of IRF3 and IFN-β genes by 14.75 and 4.95-fold, respectively, in comparison with the controls, indicating that the antibodies could restore the host innate immune response. The fully human single chain antibodies have high potential for developing further as a safe (adjunctive) therapeutic agent for mitigating, if not abrogating, severe symptoms of influenza.


Biochemical and Biophysical Research Communications | 2016

Human transbodies to VP40 inhibit cellular egress of Ebola virus-like particles.

Salma Teimoori; Watee Seesuay; Surasak Jittavisutthikul; Urai Chaisri; Nitat Sookrung; Jaslan Densumite; Nawannaporn Saelim; Monrat Chulanetra; Santi Maneewatch; Wanpen Chaicumpa

A direct acting anti-Ebola agent is needed. VP40, a conserved protein across Ebolavirus (EBOV) species has several pivotal roles in the virus life cycle. Inhibition of VP40 functions would lessen the virion integrity and interfere with the viral assembly, budding, and spread. In this study, cell penetrable human scFvs (HuscFvs) that bound to EBOV VP40 were produced by phage display technology. Gene sequences coding for VP40-bound-HuscFvs were subcloned from phagemids into protein expression plasmids downstream to a gene of cell penetrating peptide, i.e., nonaarginine (R9). By electron microscopy, transbodies from three clones effectively inhibited egress of the Ebola virus-like particles from human hepatic cells transduced with pseudo-typed-Lentivirus particles carrying EBOV VP40 and GP genes. Computerized simulation indicated that the effective HuscFvs bound to multiple basic residues in the cationic patch of VP40 C-terminal domain which are important in membrane-binding for viral matrix assembly and virus budding. The transbodies bound also to VP40 N-terminal domain and L domain peptide encompassed the PTAPPEY (WW binding) motif, suggesting that they might confer VP40 function inhibition through additional mechanism(s). The generated transbodies are worthwhile tested with authentic EBOV before developing to direct acting anti-Ebola agent for preclinical and clinical trials.

Collaboration


Dive into the Santi Maneewatch's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge