リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

大学・研究所にある論文を検索できる 「Analysis of brownbanded bamboo shark (Chiloscyllium punctatum) immunoglobulin novel antigen receptor (IgNAR) as a potential immunotherapeutic」の論文概要。リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

コピーが完了しました

URLをコピーしました

論文の公開元へ論文の公開元へ
書き出し

Analysis of brownbanded bamboo shark (Chiloscyllium punctatum) immunoglobulin novel antigen receptor (IgNAR) as a potential immunotherapeutic

Dehinga, Prasadi Nayanashani De Silva 東京大学 DOI:10.15083/0002004933

2022.06.22

概要

Immune system is important for animals to fight against invading pathogens and antigenic agents. It contains two parts mainly innate immunity and acquired immunity. During evolution, cartilaginous fish are the first jawed vertebrates having immunoglobulins (Igs) which is also called as antibodies. There are three types of Igs found in sharks namely, IgM, IgW and immunoglobulin novel antigen receptor (IgNAR). IgNAR was reported first by Greenburg et al., in 1995 as heavy chain homodimer. Since then many scientists focused on this antibody due to its advantageous characteristics in biomedical research.

There are only two naturally occurring heavy chain only antibodies (lacking light chain) were reported, one from camelids called VHH and the other one is IgNAR from cartilaginous fish. Both of these molecules have smaller variable regions of which variable region of IgNAR (vNAR) considered as the smallest antibody like molecule in nature (~ 12 – 15 kDa). The reasons for the smaller size are due to lack of light chain and devoid of second complementary determining region (CDR). IgNAR was found to be stable in harsh environments such as high temperature and pH variations. This might be an adaptation of the Ig which circulates in shark blood with high ammonia concentration. The higher stability was believed to be due to the presence of non-canonical cysteine molecules present in the variable domains of vNAR.

Use of monoclonal antibodies on diagnostics and treatments is becoming more popular recently. Due to larger molecular size of the conventional antibodies (eg: IgG 150 kDa), it is difficult to reach the epitopes particularly in cellular spaces and cancer cells. Hence scientists focused on engineering the antibodies to develop smaller, antigen specific and high affinity monoclonal antibodies. Variable region of shark IgNAR which comply with those features is being investigated to develop immunotherapeutics (anti-Ebola, anti-TNFα) and diagnostics (anti malaria AMA1, anti-Cholera). Various techniques have been used to develop these nanobodies such as immunization of shark and development of semi-synthetic antibodies.

There are four types of IgNARs classified to date based on the presence and location of cysteine residue in the variable region. Currently several sharks and rays have been investigated for their IgNAR gene sequence. Although they all have non-canonical cysteine residues, type I IgNAR was reported only from nurse shark (Ginglymostoma cirratum). Most of the investigators also performed immunizations and it revealed that different shark species have been responding differently against antigens. Hence, the aim of this study was to develop a novel platform to isolate antigen specific vNAR from brownbanded bamboo shark (Chiloscyllium punctatum) by in vivo affinity maturation.

1. Characterization of brownbanded bamboo shark IgNAR constant region
IgNAR has two regions namely, the variable region and constant region. Constant region consists of five domains C1 to C5. These domains help the integrity of the IgNAR antibody and also maintains flexibility at the hinge region during antigen binding. Study on brownbanded bamboo shark focused on the evolutionary compliance with the conventional antibodies particularly human Igs. In this study, brownbanded bamboo shark (Chiloscyllium punctatum) was selected as the model species to characterize the IgNAR constant region sequence by next generation sequencing (NGS) method. Peripheral blood leukocytes (PBL) were used to isolate mRNA and synthesized cDNA for the library having 20 million raw reads (Illumina, MiSeq). Average sequence length after paired end assembly was 218.4 bp (35 - 301 bp) with total of 4.5 billion residues. Transcriptome analysis of complete constant region with five domains and conserved cysteine residues revealed the presence of two distinct types of IgNAR in brownbanded bamboo shark. The C1 domain alone displayed 13 unique sequences that might resemble the number of IgNAR CH gene clusters. Furthermore, phylogenetic analysis showed that the relationship with order Orectolobiformes, of which nurse shark (Ginglymostoma cirratum) and wobbegong shark (Orectolobus maculatus) as the highest similarity. The alignment of human IgG constant regions indicated the conserved nature of cysteine residues throughout the evolution of Igs. Through this study, understanding of the characteristics of IgNAR constant domains used to maintain its structural stability will become useful information for engineering monoclonal antibodies in the future.

2. Diverse response by vNAR of brownbanded bamboo shark against antigen
Shark vNAR is widely used in therapeutic and diagnostic research due to its smaller size which enables tissue penetration, simple architecture and higher stability of the molecule that helps antibody engineering easier and cost effective. Developing highly specific and higher affinity antibody is important to minimize adverse effects. Presence of cysteine in variable region is useful to maintain structural integrity and stabilize the antigen binding regions. Number of cysteine residues also play an important role in the formation of diverse antigen-binding surfaces. Therefore, studying the responses against antigen and screening cysteine expression in CDR domains is important prior to synthesis of the therapeutic or diagnostic nanobodies. This study aimed at understanding the diverse nature of vNAR from brownbanded bamboo shark and the individual response against antigen at various time of exposures. Adult sharks were immunized to determine antigenic responses of vNAR by injecting hen egg lysozyme (HEL) and collected the peripheral blood leukocytes (PBL) for cDNA library synthesis. Three-step tailed PCR was designed and vNAR library was obtained using Illumina amplicon sequencing. The vNAR library with 72% productive sequences was consisted of 8.41 GB data (19.3 million sequences). More mutations were found in CDR-1 than CDR-3 which is typical for type II IgNAR when affinity maturation occurs due to somatic hypermutation. However, CDR1 and CDR3 variations were reduced as number of antigen exposures increased. Type II IgNAR sequences were abundant in only one shark and found different patterns expressed in different individuals. Interestingly, type I sequence found in one shark after first exposure to antigen at a very low expression level (1/100,000). In addition to nurse shark this is the first report of type I IgNAR from another species. There was an increase of IgNAR titer along with vNAR sequences obtained after HEL exposure. This study reveals the diversity of vNAR among individual sharks and also variability during the number of exposures. Hence it is suggestive of determining the highly antigen specific vNAR gene sequence prior to development of monoclonal antibodies from shark origin. This study shows the platform of synthesizing large cDNA library by amplicon sequencing and reduces the time, labour and cost of conventional cloning methods.

3. Transcriptome analysis revealed overall positive immune response against antigen
The transcriptome analysis and RNA-Seq data provides insights to gene expressions and reveals lot of information. Currently few shark species including nurse shark tissues such as heart, spleen, kidney were studied. Genomic study on Elephant shark (Callorhynchus milii) showed that adaptive immune system of cartilaginous fish has T helper cells that were highly restricted and unconditional antigen-binding ability. Furthermore, it lacks CD4 and related cytokine receptors as well. However very few studies have done on transcriptomic analysis of shark species and almost none of the studies investigated the immune response before and after antigen exposure in brownbanded bamboo shark. The aim of this study was to perform transcriptome analysis on the response of PBL from brownbanded bamboo shark against HEL. The RNA-seq library was prepared from PBL of pre-immunized (control) and immunized brownbanded bamboo sharks. Nearly 40 million clean sequence reads were obtained including 56 % immunized, 46 % control sequences. After antigen exposure, genes on negative regulation of Ig production was down regulated while regulation of complement activation, lectin pathway and regulation of cellular defense response were upregulated. Based on the transcriptomic analysis brownbanded bamboo shark shows great adaptive immune response against antigen which makes it a suitable candidate for immune research.

4. Strategy to synthesize brownbanded bamboo shark vNAR based monoclonal antibody
Development of monoclonal antibody is trending these days having around 20 – 30 products in the market and over 100 in clinical trials. It is predicted to be increasing and advancing with the use of smaller, recombinant antibody fragments and engineered antibody variants such as nanobodies. Shark vNAR having interesting features fit for immunotherapy is a potential candidate and this study performed to develop a platform to synthesize monoclonal antibody from brownbanded bamboo shark vNAR origin. Adult sharks were immunized with hen egg lysosome (HEL) and cDNA amplicon library prepared using mRNA from peripheral blood leukocytes. After paired end assembly unique sequences were listed based on the abundance and expression during prior and after immunization were compared. The sequences expressed after third immunization with higher abundance were selected and obtained anti-HEL vNAR model (nurse shark anti-HEL vNAR was used as template). Using Chimera (UCSF) software, strength of antigen binding by the number of contacts between antigen-antibody complex was determined. In order to synthesize ant-HEL nanobody, the highest affinity vNAR gene sequence was inserted into mammalian expression vector pEHX1.1-N.S.-IgNAR-Fc (from Omasa lab, Osaka University). The protocol has been developed to express the vNAR antibody using chicken hamster ovary (CHO) cell line. This novel platform for isolation and expression of antigen specific vNAR may be useful for the development of monoclonal nanobodies from shark origin at less cost and time compared to the conventional methods such as phage display.

5. Future research potential
Shark IgNAR was first reported in 1995 by Flajnik and co-workers. Since then many studies have been carried out on characterization of IgNAR in different shark species and synthesis of antigen specific vNAR by immunization or molecular engineering. However, there are few drawbacks of using shark vNAR; firstly, due to the smaller size (~12 – 15 kDa) it is easily filter through kidney glomeruli causing less retention time (or half-life), secondly, adverse effects due to rejection of the antibody from host. Hence this study focused on developing IgNAR against albumin, which is a main plasma protein in mammals with higher molecular weight (66.5 kDa). A combination of HEL and bovine serum albumin (BSA) were injected to brownbanded bamboo sharks and synthesized a cDNA library with amplicons covering full-length vNAR gene sequences. Similar to the previous study with HEL, potential anti-BSA vNAR gene sequences were determined by using anti-HSA vNAR template from spiny dogfish. The target binding residues such as, Trp, Asp, Tyr and Pro were prominent in anti-BSA vNAR CDR3 sequence in brownbanded bamboo shark. According to ELISA, plasma IgNAR response against HEL became less compared to BSA. The sequences derived from this study will be useful for the expression of nanobodies using CHO cells in further research.
Overall, this research investigated brownbanded bamboo shark naiive IgNAR and immunized variations. The transcriptome analysis will provide insights to the antigenic response by PBLs. In addition, development of novel platform to synthesize full length vNAR amplicon library and screening highly specific gene sequences will be useful in developing nanobodies with minimal adverse effects in the future.

この論文で使われている画像

参考文献

Benichou, J., Ben-Hamo, R., Louzoun., Y. & Efroni, S. 2012. Rep-Seq: Uncovering the immunological repertoire through next-generation sequencing. Immunology, 135(3), 183–191.

Bolger, A.M., Lohse, M. & Usadel, B. 2014. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics, 30(15), 2114–2120.

Bray N.L., Pimentel H., Melsted P. and Pachter L., 2016 Near-optimal probabilistic RNA seq quantification. Nat. Biotechnol., 34 (5), 525-527.

Crouch, K., Smith, L.E., Williams, R., Cao, W., Lee, M., Jensen, A. & Dooley, H. 2013. Humoral immune response of the small-spotted catshark, Scyliorhinus canicula. Fish and Shellfish Immunology, 34(5), 1158–1169.

De los Rios, M., Criscitiello, M. & Smider, V. 2015. Structural and genetic diversity in antibody repertoires from diverse species. Current Opinion in Structural Biology, 33, 27-41.

Diaz, M., Greenberg, A.S. & Flajnik, M.F., 1998. Somatic hypermutation of the new antigen receptor gene (NAR) in the nurse shark does not generate the repertoire: possible role in antigen-driven reactions in the absence of germinal centers. Proceedings of the National Academy of Sciences of the United States of America, 95(24), 14343–14348.

Diaz, M., Stanfield, R.L., Greenberg, A.S. & Flajnik, M.F., 2002. Structural analysis, selection, and ontogeny of the shark new antigen receptor (IgNAR): Identification of a new locus preferentially expressed in early development. Immunogenetics, 54(7), 501–512.

Dooley, H., Flajnik, M.F. & Porter, A.J., 2003. Selection and characterization of naturally occurring single-domain (IgNAR) antibody fragments from immunized sharks by phage display. Molecular Immunology, 40(1), 25–33.

Dooley, H. & Flajnik, M.F., 2005. Shark immunity bites back: Affinity maturation and memory response in the nurse shark, Ginglymostoma cirratum. European Journal of Immunology, 35(3), 936–945.

Dooley, H. & Flajnik, M.F., 2006. Antibody repertoire development in cartilaginous fish. Developmental and Comparative Immunology, 30(1–2), 43–56.

Dooley, H., Stanfield, R.L., Brady R.A. & Flajnik M.F., 2006. First molecular and biochemical analysis of in vivo affinity maturation in an ectothermic vertebrate. Proceedings of the National Academy of Sciences of the United States of America, 103(6), 1846–51.

Feige, M.J., Grawert, M.A., Marcinowski, M., Henning, J., Behnke, J., Auslander, D.Herold, E.M., Peschek, J., Castro, C.D., Flajnik, M., Hendershot, L.M., Sattler, M., Groll, M. & Buchner, J., 2014. The structural analysis of shark IgNAR antibodies reveals evolutionary principles of immunoglobulins. Proceedings of the National Academy of Sciences of the United States of America, 111(22), 8155–60.

Felsenstein, J., 1985. Confidence limits on phylogenies: An approach using the bootstrap. Evolution, 39, 783-791

Feng, M., Bian, H., Wu, X., Fu, T., Fu, Y., Hong, J., Flemming, B.D., Flajnik, M.F. & Ho, M., 2019. Construction and next-generation sequencing analysis of a large phage-displayed VNAR single-domain antibody library from six naïve nurse sharks. Antibody Therapeutics. 2 (1), 1-11.

Flajnik,M.F. Rumfelt, L.L., 2000. The immune system of cartilagenous fish. Curr. Top. Microbiol. Immunol, 248, 249-270

Flajnik, M.F., 2002. Comparative analyses of immunoglobulin genes: surprises and portents. Nature reviews. Immunology, 2(9), 688–98.

Flajnik, M.F. & Dooley, H., 2009. The generation and selection of single-domain, v region libraries from nurse sharks. Methods in molecular biology (Clifton, N.J.), 562, 71–82.

Flajnik, M.F. & Kasahara, M., 2010. Origin and evolution of the adaptive immune system: genetic events and selective pressures. Nature reviews. Genetics, 11(1), 47–59.

Flajnik, M.F., Deschacht, N. & Muyldermans, S., 2011. A case of convergence: Why did a simple alternative to canonical antibodies arise in Sharks and Camels? PLoS Biology, 9(8), 1-5.

Gilbert, P.W. & Wood, F.G., 1957. Method of anesthetizing large sharks and rays safely and rapidly. Science, 126(3266), 212–213.

Grabherr MG, Haas BJ, Yassour M, Levin JZ, Thompson DA, Amit I, Adiconis X, Fan L, Raychowdhury R, Zeng Q, Chen Z, Mauceli E, Hacohen N, Gnirke A, Rhind N, di Palma F, Birren BW, Nusbaum C, Lindblad-Toh K, Friedman N, Regev A., 2011. Full-length transcriptome assembly from RNA-seq data without a reference genome. Nat Biotechnol, 29(7), 644-652.

Greenberg, A.S., Avila, D., Hughes, M., Hughes, A., McKinney, E.C. & Flajnik, M.F., 1995. A new antigen receptor gene family that undergoes rearrangement and extensive somatic diversification in sharks. Nature, 374(6518), 168–73.

Griffiths K, Dolezal O, Parisi K, Angerosa J, Dogovski C, Barraclough M, Sanalla A, Casey J, González I, Perugini M, Nuttall S, Foley M., 2013. Shark variable new antigen receptor (VNAR) single domain antibody fragments: stability and diagnostic applications. Antibodies, 2, 66–81.

Hall, T.A., 1999. BioEdit: a user-friendly biological sequence alignment editor and analysis program for Windows 95/98/NT. Nucl. Acids Symp, 41, .95–98.

Hamers-Casterman, C., Atarhouch, T., Muyldemans, S., Robinson, G., Hamers, C., Songa, E.B., Bendahman N. and Hamers, R., 1993. Naturally occurring antibodies devoid of light chains. Nature, 363(6428), 446–448.

Hara, Y., Yamaguchi, K., Onimaru, K. et al. 2018. Shark genomes provide insights into elasmobranch evolution and the origin of vertebrates. Nat Ecol Evol 2, 1761–1771

Hinds, K.R. & Litman, G.W., 1986. Major reorganization of immunoglobulin VH segmental elements during vertebrate evolution. Nature, 320(6062), 546–9.

Honda, Y., Kondo, H., Caipang, C.M.A., Hirono, I. & Aoki, T., 2010. cDNA cloning of the immunoglobulin heavy chain genes in banded houndshark Triakis scyllium. Fish and Shellfish Immunology, 29(5), 854–861.

Honma H., Okabe K. and Chiba A., 1984. Comparative histology of the Leydig and Epigonal orggans in some elasmobranchs. Japanese Journal of Ichthhyology, 31 (1), 47-54

Huang, Jinghe; Doria-Rose, Nicole A.; Longo, Nancy S.; Laub, Leo; Lin, Chien-Li; Turk, Ellen; Kang, Byong H.; Migueles, Stephen A.; Bailer, Robert T., 2013. Isolation of human monoclonal antibodies from peripheral blood B cells. Nature Protocols. 8 (10), 1907–1915.

Hinds KR, Litman GW., 1986. Major reorganization of immunoglobulin VH segmental elements during vertebrate evolution. Nature. 320, 546–549.

Khan K.H., 2013. Gene expression in mammalian cells and its applications. Advanced Pharmaceutical Bulletin. 3(2), 257-263.

Kokubu F, Litman R, Shamblott MJ, Hinds K, Litman GW., 1988. Diverse organization of immunoglobulin VH gene loci in a primitive vertebrate. EMBO J., 7, 3413–3422. Kopsidas, G., Roberts, A.S., Coia, G., Streltsov, V.A. & Nuttall, S.D., 2006. In vitro improvement of a shark IgNAR antibody by Qß replicase mutation and ribosome display mimics in vivo affinity maturation. Immunology Letters, 107(2), 163–168.

Kovaleva, M., Ferguson, L., Steven, J., Porter, A. & Barelle, C., 2014. Shark variable new antigen receptor biologics - a novel technology platform for therapeutic drug development. Expert opinion on biological therapy, 14(10), 1527–1539.

Kroeker, A.L.; Berard, A.R.; Coombs, K.M., 2012. Fishing beyond the peer: Future omic analyses of virus-host Interactions. J. Virol. Antivir. Res. 1(1),.

Kubota, T., Niwa, R., Satoh M., Akinaga S., Shitara, K. & Hanai, N., 2009. Engineered therapeutic antibodies with improved effector functions. Cancer Science, 100(9), 1566–1572.

Kumar S., Stecher, G., Tamura, K., 2016. MEGA7: Molecular Evolutionary Genetics Analysis version 7.0 for bigger datasets. Molecular Biology and Evolution, 33, 1870-1874

Lee J.S., Park H.J. Kim Y.H. and Lee G.M., 2010. Protein reference mapping of dihydrofolate reductase deficient CHO DG44 cell lines using 2-dimensional electrophoresis. Proteomics. 10 (12), 2292-2302.

Leow C.H., Fischer K., Leow C.Y., Cheng Q., Chuah C. and McCarthy J., 2017. Single domain antibodies as new biomarker detectors. Diagnostics. 7, 52.

Li, S., Lefrank, M.P., Miles, J.J., Alamyar, E., Giudicelli, V., Durox, P., Freeman, J.D., Corbin, V.D., Scheerlink, J.P., Frohman, M.A., Cameron, P.U., Plebanski, M., Loveland, B., Burrows, S.R., Papenfuss A.T. & Gowans, E.J., 2013. IMGT/HighV QUEST paradigm for T cell receptor IMGT clonotype diversity and next generation repertoire immunoprofiling. Nat. Commun. 4:2333

Litman, G.W., Anderson, M.K. & Rast, J.P., 1999. Evolution of antigen binding receptors. Annual Review of Immunology, 17(1), 109–147.

Love M.I., Huber W. and Anders S., 2014. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550.

Magor, B.G., 2015. Antibody Affinity Maturation in Fishes-Our Current Understanding. Biology, 4(3), 512–24.

Marra N.J., Richards V.P., Early A., Bogdanowicz S.M., Bitar P.D.P., Stanhope M.J. and Shivji M.S., 2017. Comparative transcriptomics of elasmobranch and teleosts highlight important processes in adaptive immunity and regional endothermy. BMC Genomics, 18, 87

Marchalonis, J.J., Schluter, S.F., Bernstein, R.M., Shen S., &. Edmundson, A.B., 1998. Phylogenetic emergence and molecular evolution of the immunoglobulin family, Adv. Immunol, 70, 417-506

Matz, H. & Dooley H., 2019. Shark IgNAR-derived binding domains aas potential diagnostic and therapeutic agents. Developmental and Comparative Immunology, 90, 100-107.

McCullough, K.C. & Summerfield, A., 2005. Basic Concepts of Immune Response and Defense Development. ILAR Journal, 46(3), 230–240.

Miracle, A.L., Anderson, M.K., Litman, R.T., Walsh, C.J., Luer, C.A., Rothenberg, E.V. & Litman, G.W., 2001. Complex expression patterns of lymphocyte-specific genes during the development of cartilaginous fish implicate unique lymphoid tissues in generating an immune repertoire. International immunology, 13(4), 567–580.

Morozova, O.; Hirst, M.; Marra, M.A., 2009. Applications of new sequencing technologies for transcriptome analysis. Annu. Rev. Genom. Hum. Genet. 10, 135– 151.

Müller, M.R., Saunders, K., Grace, C. et. Al., 2012. Improving the pharmacokinetic properties of biologics by fusion to an anti-HSA shark VNAR domain. mAbs, 4(6), 673–685.

Mulley J.F., Hargreaves A.D., Hegarty M.J., Heller R.S., Swain M.T., 2014 Transcriptomic analysis of the lesser spotted catshark (Scyliorhinus canicula) pancreas, liver and brain reveals molecular level conservation of vertebrate pancreas function. BMC Genomics, 15, 1074.

Mutz, K.O.; Heilkenbrinker, A.; Lönne, M.; Walter, J.G.; Stahl, F. 2013. Transcriptome analysis using next-generation sequencing. Curr. Opin. Biotechnol., 24, 22–30.

Nikolova, V., Koo, C., Abrahim, S.A. et al., 2009. Differential roles for membrane-bound and soluble syndecan-1 (CD138) in breast cancer progression. Carcinogenesis, 30(3), 397–407.

Nuttall, S.D., Krishnan, U.V., Hattarki, M., De Gori, R., Irving, R.A. & Hudson, P.J., 2001. Isolation of the new antigen receptor from wobbegong sharks, and use as a scaffold for the display of protein loop libraries. Molecular Immunology, 38(4), 313–326.

Ogino S., Gulley M.L., Dunnen J.T., Wilson R.B. and the Association for Molecular Pathology Training and Education Committee. 2007. Standard mutation nomenclature in molecular diagnostics. Practical and educational challenges. Journal of Molecular Diagnostics, 9(1), 1-7.

Ohtani, M., Hikima, J., Jung, T.S., Kondo, H., Hirono, I. & Aoki, T., 2013. Construction of an Artificially Randomized IgNAR Phage Display Library: Screening of Variable Regions that Bind to Hen Egg White Lysozyme. Marine Biotechnology, 15(1), 56–62.

Omasa T, Onitsuka M, Kim W.D., 2010. Cell engineering and cultivation of chinese hamster ovary (CHO) cells. Curr Pharm Biotechnol, 11, 233–240.

Oulion S, Debiais-Thibaud M, D’Aubenton-Carafa Y, Thermes C, Da Silva C, Bernard- Samain S, Gavory F, Wincker P, Mazan S, Casane D., 2010. Evolution of Hox gene clusters in gnathostomes: insights from a survey of a shark (Scyliorhinus canicula) transcriptome. Mol Biol Evol. 27, 2829–2838.

Pettersen E.F., Goddard T.D., Huang C.C., Couch G.S., Greenblatt D.M., Meng E.C., Ferrin T.E., 2004. UCSF Chimera – a visualization system for exploratory research and analysis. J Comput Chem, 13, 1605-1612.

Pimentel H., Bray N.L., Puente S., Melsted P. and Pachter L., 2017. Differential analysis of RNA-seq incorporating quantification uncertainty. Nat. Methods. 14 (7), 687- 690.

Presta, L.G., 2008. Molecular engineering and design of therapeutic antibodies. Curr Opin Immunol, 20(4), 460–470.

Puck T.T., Cieciura S.J. & Robinson A., 1958. Genetics of somatic mammalian cells. III. Long-term cultivation of euploid cells from human and animal subjects. J Exp Med. 108(6), 945-956.

Reichert J.M., 2012. Marketed therapeutic antibodies compendium. MAbs. 4, 413-415.

Richards V.P., Suzuki H, Stanhope M.J, Shivji M.S. 2013. Characterization of the heart transcriptome of the white shark (Carcharodon carcharias). BMC Genomics, 14, 697.

Rumfelt, L.L., Avila D., Marilyn D., Barti S.,McKinney E.C. & Flajnik M.A. 2001. A shark antibody heavy chain encoded by a nonsomatically rearranged VDJ is preferentially expressed in early development and is convergent with mammalian IgG. Proceedings of the National Academy of Sciences of the United States of America, 98(4), 1775–1780.

Saitou N. and M. Nei, 1987. The neighbor-joining method: A new method for reconstructing phylogenetic trees. Molecular Biology and Evolution. 4, 406-425.

Schroeder, H.W. & Cavacini, L., 2010. Structure and Function of Immunoglobulins. Journal of Allergy and Clinical Immunology, 125(202) S41-S52.

Seeber, S., Ros, F., Thorey, I., Tiefenthaler, G., Kaluza, K., Lifke, V., Fischer, J., André A., Klostermann, S. & Endl, J. 2014. A Robust High Throughput Platform to Generate Functional Recombinant Monoclonal Antibodies Using Rabbit B Cells from Peripheral Blood. PLoS ONE. 9 (2): e86184.

Simmons D.P., Abregu F.A., Krishnan U.V., Proll D.F., Streltsov V.A., Doughty L, Hattarki M.K. & Nuttall S.D. 2006. Dimerisation strategies for shark IgNAR single domain antibody fragments. J. of Immunol Methods. 315(1-2), 171-84.

Skerra A. 2007. Alternative non-antibody scaffolds for molecular recognition. Curr Opin Biotechnol, 18, 295-304.

Smith, M., Warmolts, D., Thoney, D. & Hueter, R. 2004. Elasmobranch husbandry manual: Captive care of sharks, rays, and their relatives. The Elasmobranch Husbandry Manual : Captive Care of Sharks , Rays and their Relatives, 307-323.

Smith, L.E., Crouch, K., Cao, W. et al., 2012. Characterization of the immunoglobulin repertoire of the spiny dogfish (Squalus acanthias). Developmental & Comparative Immunology, 36(4), 665–679.

Stanfield, R.L., Dooley,H., Verdino, P., Flajnik, M.F. & Wilson, I.A., 2007. Maturation of Shark Single-domain (IgNAR) Antibodies: Evidence for Induced-fit Binding. Journal of Molecular Biology, 367(2), 358–372.

Streltsov, V.A., Carmichael, J.A. & Nuttall, S.D., 2005. Structure of a shark IgNAR antibody variable domain and modeling of an early-developmental isotype. Protein science : a publication of the Protein Society, 14(11), 2901–9.

Sudhakar A., Kumar G & El-Matbouli M. 2018. Transcriptome analysis based on RNA- Seq in understanding pathogenic mechanisms of diseases and immune system of fish: A comprehensive review. International Journal of Molecular Sciences. 19, 245

Takagi Y., Kikuchi T., Wada R. and Omasa T. 2017. The enhancement of antibody concentration and achievement of high cell density CHO cell cultivation by adding nucleoside. Cytotechnology. 69, 511-521.

Takechi M, Takeuchi M, Ota K.G, Nishimura O, Mochii M, Itomi K, Adachi N, Takahashi M, Fujimoto S, Tarui H, Okabe M, Aizawa S, Kuratani S. 2011. Overview of the transcriptome profiles identified in hagfish, shark, and bichir: current issues arising from some nonmodel vertebrate taxa. J Exp Zool Part B Mol Dev Evol, 316B, 526–546.

Tonegawa, S., 1983. Somatic generation of antibody diversity. Nature, 302(5909), 575– 581.

Venkatesh B, Lee AP, Ravi V, Maurya AK, Lian MM, Swann JB, Ohta Y, Flajnik MF, Sutoh Y, Kasahara M, Hoon S, Gangu V, Roy SW, Irimia M, Korzh V, Kondrychyn I, Lim ZW, Tay B-H, Tohari S, Kong KW, Ho S, Lorente-Galdos B, Quilez J, Marques-Bonet T, Raney BJ, Ingham PW, Tay A, Hillier LW, Minx P, Boehm T, et al. 2014. Elephant shark genome provides unique insights into gnathostome evolution. Nature, 505, 174–179.

Walsh G. 2014. Biopharmaceutical benchmarks. Nat Biotechnol, 32, 992–1000.

Wang, F., Ekiert, D.C., Ahmad, I., Yu, W., Zhang, Y., Bazirgan, O., Torkamani, A., Raudsepp, T., Mwangi, W., Criscitiello, M.F., Wilson, I.A., Schultz, P.G. & Smider, V.V. 2013. Reshaping antibody diversity. Cell, 153(6), 1379-1393.

Wang, Z.; Gerstein, M.; Snyder, M. 2009. RNA-Seq: A revolutionary tool for transcriptomics. Nat. Rev. Genet, 10, 57–63.

Wang Q, Arighi C.N., King B.L., Polson S.W., Vincent J., Chen C, Huang H., Kingham B.F., Page S.T., Rendino M.F., Thomas W.K., Udwary D.W., Wu C.H. 2012. Community annotation and bioinformatics workforce development in concert-Little Skate Genome Annotation Workshops and Jamborees. Database (Oxford). 2012; 2012:bar064.

Wardemann, H., Yurasov, S., Schaefer, A., Young, James W.; Meffre, E. & Nussenzweig, M.C. 2003. Predominant autoantibody production by early human B cell precursors. Science. 301 (5638), 1374–1377.

Warrington, R., Watson, W., Kim H.L. & Antonetti R. 2011. An introduction to immunology and immunopathology. Allergy, Asthma & Clinical Immunology, 7(Suppl 1):S1, 1-8.

Waterhouse, A., Bertoni, M., Bienert, S., Studer, G., Tauriello, G., Gumienny, R., Heer, F.T., de Beer, T.A.P., Rempfer, C., Bordoli, L., Lepore, R., Schwede, T. 2018. SWISS-MODEL: homology modelling of protein structures and complexes. Nucleic Acids Res. 46(W1), W296-W303.

Williams, F., A. & Barclay, A.N., 1988. The immunoglobulin superfamily domains for cell surface recognition. Annual Review of Immunology, 6, 381–405.

Wu, J., Mao, X., Cai, T., Luo, J., Wei, L. 2006. KOBAS server: a web-based platform for automated annotation and pathway identification. Nucleic Acids Res. 34, W720– W724.

Wurm F.M. & Hacker D., 2011. First CHO genome. Nat Biotechnol. 29 (8), 718-720.

Xie, C., Mao, X., Huang, J., Ding, Y., Wu, J., Dong, S., Kong, L., Gao, G., Li, C. & Wei, L. 2011. KOBAS 2.0: a web server for annotation and identification of enriched pathways and diseases. Nucleic Acids Research, 39, W316–W322.

Zhang, J., Kobert K., Fluori T. & Stamatakis, A. 2014. PEAR: A fast and accurate Illumina Paired-End reAd mergeR. Bioinformatics, 30(5), 614–620.

Zielonka, S., Weber, N., Becker, S. et al., 2014. Shark Attack: High affinity binding proteins derived from shark vNAR domains by stepwise in vitro affinity maturation. Journal of Biotechnology, 191, 236–245.

Zielonka, S., Empting, M., Grzeschik, J., et al., 2015. Structural insights and biomedical potential of IgNAR scaffolds from sharks. mAbs, 7(1), 15–25.

Zuckerkandl E. & Pauling, L. 1965. Evolutionary divergence and convergence in proteins. Edited in Evolving Genes and Proteins by V. Bryson and H.J. Vogel. Academic Press, New York. 97-166

参考文献をもっと見る

全国の大学の
卒論・修論・学位論文

一発検索!

この論文の関連論文を見る