Novel Camelid Antibody Fragments Targeting Recombinant Nucleoprotein of Araucaria hantavirus: A Prototype for an Early Diagnosis of Hantavirus Pulmonary Syndrome

In addition to conventional antibodies, camelids produce immunoglobulins G composed exclusively of heavy chains in which the antigen binding site is formed only by single domains called VHH. Their particular characteristics make VHHs interesting tools for drug-delivery, passive immunotherapy and high-throughput diagnosis. Hantaviruses are rodent-borne viruses of the Bunyaviridae family. Two clinical forms of the infection are known. Hemorrhagic Fever with Renal Syndrome (HFRS) is present in the Old World, while Hantavirus Pulmonary Syndrome (HPS) is found on the American continent. There is no specific treatment for HPS and its diagnosis is carried out by molecular or serological techniques, using mainly monoclonal antibodies or hantavirus nucleoprotein (N) to detect IgM and IgG in patient serum. This study proposes the use of camelid VHHs to develop alternative methods for diagnosing and confirming HPS. Phage display technology was employed to obtain VHHs. After immunizing one Lama glama against the recombinant N protein (prNΔ85) of a Brazilian hantavirus strain, VHH regions were isolated to construct an immune library. VHHs were displayed fused to the M13KO7 phage coat protein III and the selection steps were performed on immobilized prNΔ85. After selection, eighty clones recognized specifically the N protein. These were sequenced, grouped based mainly on the CDRs, and five clones were analyzed by western blot (WB), surface plasmon resonance (SPR) device, and ELISA. Besides the ability to recognize prNΔ85 by WB, all selected clones showed affinity constants in the nanomolar range. Additionaly, the clone KC329705 is able to detect prNΔ85 in solution, as well as the native viral antigen. Findings support the hypothesis that selected VHHs could be a powerful tool in the development of rapid and accurate HPS diagnostic assays, which are essential to provide supportive care to patients and reduce the high mortality rate associated with hantavirus infections.

[1]  Michael D. McLean,et al.  In Vivo Neutralization of α-Cobratoxin with High-Affinity Llama Single-Domain Antibodies (VHHs) and a VHH-Fc Antibody , 2013, PloS one.

[2]  Serge Muyldermans,et al.  Nanobodies: natural single-domain antibodies. , 2013, Annual review of biochemistry.

[3]  P. De Baetselier,et al.  Novel applications of nanobodies for in vivo bio-imaging of inflamed tissues in inflammatory diseases and cancer. , 2012, Immunobiology.

[4]  M. R. de Araújo,et al.  Hantavirus infection prevalence in wild rodents and human anti-hantavirus serological profiles from different geographic areas of South Brazil. , 2012, The American journal of tropical medicine and hygiene.

[5]  F. Ducancel,et al.  Molecular engineering of antibodies for therapeutic and diagnostic purposes , 2012, mAbs.

[6]  R. Weiss,et al.  Llama Antibody Fragments Recognizing Various Epitopes of the CD4bs Neutralize a Broad Range of HIV-1 Subtypes A, B and C , 2012, PloS one.

[7]  S. Muyldermans,et al.  Introduction to heavy chain antibodies and derived Nanobodies. , 2012, Methods in molecular biology.

[8]  R. Weiss,et al.  Llama-Derived Single Domain Antibodies to Build Multivalent, Superpotent and Broadened Neutralizing Anti-Viral Molecules , 2011, PloS one.

[9]  De-xin Li,et al.  Tracking hantavirus nucleocapsid protein using intracellular antibodies , 2010, Virology Journal.

[10]  S. Muyldermans,et al.  In vitro antiviral activity of single domain antibody fragments against poliovirus. , 2010, Antiviral research.

[11]  J. Kolkman,et al.  Nanobodies - from llamas to therapeutic proteins. , 2010, Drug discovery today. Technologies.

[12]  R. Weiss,et al.  Generation of a Family-specific Phage Library of Llama Single Chain Antibody Fragments That Neutralize HIV-1* , 2010, The Journal of Biological Chemistry.

[13]  L. de Noronha,et al.  Production and characterization of monoclonal antibodies against the recombinant nucleoprotein of Araucaria hantavirus. , 2009, Journal of virological methods.

[14]  C. Duyckaerts,et al.  Single-domain antibodies recognize selectively small oligomeric forms of amyloid beta, prevent Abeta-induced neurotoxicity and inhibit fibril formation. , 2009, Molecular immunology.

[15]  S. Muyldermans,et al.  General Strategy to Humanize a Camelid Single-domain Antibody and Identification of a Universal Humanized Nanobody Scaffold* , 2009, Journal of Biological Chemistry.

[16]  S. Muyldermans,et al.  Single-domain antibodies as building blocks for novel therapeutics. , 2008, Current opinion in pharmacology.

[17]  Michael D. McLean,et al.  Cloning, expression, and characterization of a single-domain antibody fragment with affinity for 15-acetyl-deoxynivalenol. , 2008, Molecular immunology.

[18]  S. Muyldermans,et al.  Antibody Fragments as Probe in Biosensor Development , 2008, Sensors.

[19]  L. Saif,et al.  Llama-Derived Single-Chain Antibody Fragments Directed to Rotavirus VP6 Protein Possess Broad Neutralizing Activity In Vitro and Confer Protection against Diarrhea in Mice , 2008, Journal of Virology.

[20]  L. Figueiredo,et al.  Expression of a hantavirus N protein and its efficacy as antigen in immune assays. , 2008, Brazilian journal of medical and biological research = Revista brasileira de pesquisas medicas e biologicas.

[21]  T. Surrey,et al.  Selection of Genetically Encoded Fluorescent Single Domain Antibodies Engineered for Efficient Expression in Escherichia coli* , 2007, Journal of Biological Chemistry.

[22]  Andrew Hayhurst,et al.  Rapid assembly of sensitive antigen-capture assays for Marburg virus, using in vitro selection of llama single-domain antibodies, at biosafety level 4. , 2007, The Journal of infectious diseases.

[23]  H. de Haard,et al.  Properties, production, and applications of camelid single-domain antibody fragments , 2007, Applied Microbiology and Biotechnology.

[24]  C. N. Duarte Dos Santos,et al.  Hantavirus infection in Brazil: development and evaluation of an enzyme immunoassay and immunoblotting based on N recombinant protein. , 2007, Diagnostic microbiology and infectious disease.

[25]  Chang-yuan Wang,et al.  Detection of circulating antigen with a MAb-based sandwich-ELISA and its comparison with specific IgM detection in sera of patients with hemorrhagic fever with renal syndrome. , 2007, Hybridoma.

[26]  James B Delehanty,et al.  Facile generation of heat-stable antiviral and antitoxin single domain antibodies from a semisynthetic llama library. , 2006, Analytical chemistry.

[27]  Lennart Hammarstrom,et al.  Reduction in morbidity of rotavirus induced diarrhoea in mice by yeast produced monovalent llama-derived antibody fragments. , 2006, Vaccine.

[28]  T. Niewold,et al.  Selection and optimization of proteolytically stable llama single-domain antibody fragments for oral immunotherapy , 2006, Applied Microbiology and Biotechnology.

[29]  C. Probst,et al.  Hantaviruses in Central South America: Phylogenetic analysis of the S segment from HPS cases in Paraná, Brazil , 2005, Journal of medical virology.

[30]  S. Goldenberg,et al.  Clinical survey of hantavirus in southern Brazil and the development of specific molecular diagnosis tools. , 2005, The American journal of tropical medicine and hygiene.

[31]  Å. Lundkvist,et al.  Anti-idiotypic antibodies against Puumala virus glycoprotein-specific monoclonal antibodies inhibit virus infection in cell cultures , 2005, Archives of Virology.

[32]  J. McCormick,et al.  Monoclonal antibodies to three strains of hantaviruses: Hantaan, R22, and Puumala , 2005, Archives of Virology.

[33]  D. Krüger,et al.  Nucleocapsid protein of cell culture-adapted Seoul virus strain 80–39: Analysis of its encoding sequence, expression in yeast and immuno-reactivity , 2004, Virus Genes.

[34]  G. Darai,et al.  A Major Antigenic Domain of Hantaviruses is Located on the Aminoproximal Site of the Viral Nucleocapsid Protein , 2004, Virus Genes.

[35]  S. Muyldermans,et al.  Emergence and evolution of functional heavy-chain antibodies in Camelidae. , 2003, Developmental and comparative immunology.

[36]  R. Prichard,et al.  Immunization with a tubulin-rich preparation from Trypanosoma brucei confers broad protection against African trypanosomosis. , 2002, Experimental parasitology.

[37]  Jamshid Tanha,et al.  Selection by phage display of llama conventional V(H) fragments with heavy chain antibody V(H)H properties. , 2002, Journal of immunological methods.

[38]  L. Wyns,et al.  Single‐domain antibody fragments with high conformational stability , 2002, Protein science : a publication of the Protein Society.

[39]  S. Muyldermans,et al.  Heavy-chain antibodies in Camelidae; a case of evolutionary innovation , 2002, Immunogenetics.

[40]  K. Ratanabanangkoon,et al.  Production of potent polyvalent antivenom against three elapid venoms using a low dose, low volume, multi-site immunization protocol. , 2001, Toxicon : official journal of the International Society on Toxinology.

[41]  S. Muyldermans,et al.  Single domain camel antibodies: current status. , 2001, Journal of biotechnology.

[42]  L. Wyns,et al.  LOSS OF SPLICE CONSENSUS SIGNAL IS RESPONSIBLE FOR THE REMOVAL OF THE ENTIRE CH1 DOMAIN OF THE FUNCTIONAL CAMEL IGG2A HEAVY-CHAIN ANTIBODIES' , 1999 .

[43]  M. Hämäläinen,et al.  Identification and optimization of regeneration conditions for affinity-based biosensor assays. A multivariate cocktail approach. , 1999, Analytical chemistry.

[44]  L. Wyns,et al.  Comparison of llama VH sequences from conventional and heavy chain antibodies. , 1997, Molecular immunology.

[45]  L. Wyns,et al.  Selection and identification of single domain antibody fragments from camel heavy‐chain antibodies , 1997, FEBS letters.

[46]  B. Hjelle,et al.  Hantaviruses: a global disease problem. , 1997, Emerging infectious diseases.

[47]  A. Vaheri,et al.  Hantaviruses: genome structure, expression and evolution. , 1996, The Journal of general virology.

[48]  Å. Lundkvist,et al.  A major antigenic domain for the human humoral response to Puumala virus nucleocapsid protein is located at the amino-terminus. , 1996, Journal of virological methods.

[49]  S. Muyldermans,et al.  Sequence and structure of VH domain from naturally occurring camel heavy chain immunoglobulins lacking light chains. , 1994, Protein engineering.

[50]  P. R. Sibbald,et al.  CDR3 length in antigen-specific immune receptors , 1994, The Journal of experimental medicine.

[51]  U. Kent Purification of antibodies using ammonium sulfate fractionation or gel filtration. , 1994, Methods in molecular biology.

[52]  J. Childs,et al.  Genetic identification of a hantavirus associated with an outbreak of acute respiratory illness. , 1993, Science.

[53]  S. Muyldermans,et al.  Naturally occurring antibodies devoid of light chains , 1993, Nature.

[54]  C. Schmaljohn,et al.  Expression of non-conserved regions of the S genome segments of three hantaviruses: evaluation of the expressed polypeptides for diagnosis of haemorrhagic fever with renal syndrome. , 1993, The Journal of general virology.

[55]  G. Darai,et al.  A novel mu-capture enzyme-linked immunosorbent assay based on recombinant proteins for sensitive and specific diagnosis of hemorrhagic fever with renal syndrome , 1993, Journal of clinical microbiology.

[56]  R. Karlsson,et al.  Kinetic analysis of monoclonal antibody-antigen interactions with a new biosensor based analytical system. , 1991, Journal of immunological methods.

[57]  S. Loefas,et al.  Immobilization of proteins to a carboxymethyldextran-modified gold surface for biospecific interaction analysis in surface plasmon resonance sensors. , 1991, Analytical biochemistry.