Fish Immunoglobulins

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Developmental and Comparative Immunology 115 (2021) 103873

Contents lists available at ScienceDirect

Developmental and Comparative Immunology


journal homepage: www.elsevier.com/locate/devcompimm

Review

The immunoglobulins of cartilaginous fishes


Hanover Matz a, b, Danish Munir c, James Logue a, Helen Dooley a, b, *
a
Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
b
Institute of Marine and Environmental Technology, Baltimore, MD, USA
c
School of Biological Sciences, University of Aberdeen, Aberdeen, UK

A R T I C L E I N F O A B S T R A C T

Keywords: Cartilaginous fishes, comprising the chimeras, sharks, skates, and rays, split from the common ancestor with
Cartilaginous fish other jawed vertebrates approx. 450 million years ago. Being the oldest extant taxonomic group to possess an
Shark immunoglobulin (Ig)-based adaptive immune system, examination of this group has taught us much about the
Antibody
evolution of adaptive immunity, as well as the conserved and taxon-specific characteristics of Igs. Significant
IgM
progress has been made analyzing sequences from numerous genomic and transcriptomic data sets. These
IgNAR
Memory findings have been supported by additional functional studies characterizing the Igs and humoral response of
B cells sharks and their relatives. This review will summarize what we have learned about the genomic organization,
protein structure, and in vivo function of these Ig isotypes in cartilaginous fishes and highlight the areas where
our knowledge is still lacking.

1. Introduction 1996; Greenberg et al., 1996; Kunihiko and Susumu, 1988), are
orthologous to IgM and IgD (respectively) in other vertebrate groups
Since adaptive immunity arose in the common ancestor of all (Ohta and Flajnik, 2006). In addition to these ‘conventional’ IgH-IgL
vertebrate lineages, it has diversified into a complex arsenal of cellular isotypes elasmobranchs possess a third isotype IgNAR, an IgH chain
and molecular defenses that each phylogenetic branch of both the ec­ homodimer that does not associate with L chain (Greenberg et al., 1995;
totherms (cold-blooded organisms) and endotherms (warm-blooded Roux et al., 1998). Further, cartilaginous fishes have four IgL chain
organisms) has preserved or altered to varying degrees. However, the isotypes: kappa (κ), lambda (λ), sigma (σ), and sigma-2 (σ-2; also called
fundamental molecular armament of humoral immunity in all gna­ σ-cart prior to its discovery in lobe-finned fishes) (Criscitiello and Flaj­
thostomes (jawed vertebrates) are immunoglobulins (Igs) or antibodies nik, 2007; Saha et al., 2014). This review will summarize what we have
(Abs). It is these molecules, expressed as heterodimers of heavy (H) and learned about the genomic organization, protein structure, and in vivo
light (L) chains on the surface of B cells and secreted by plasma cells, that function of these Ig isotypes in cartilaginous fishes in the context of
provide the host adaptive immune system with exquisite specificity to immune system evolution.
protect against pathogens via a myriad of functional roles.
Cartilaginous fish (Chondrichthyes) split from the common ancestor 2. Cartilaginous fish immune organs
with other jawed vertebrates approx. 450 million years ago (Inoue et al.,
2010) and have two extant subclasses, the holocephalans (chimeras, Like mammals, T cell maturation occurs in the cartilaginous fish
such as elephant sharks and rat fishes) and the better known elasmo­ thymus, which is paired and located dorsomedial to the gills (Fig. 1)
branchs (sharks, rays, and skates). Being the oldest extant taxonomic (Luer et al., 1995; Miracle et al., 2001). Lacking bone marrow, carti­
group to possess an Ig-based adaptive immune system (Flajnik and laginous fish B cell lymphopoiesis primarily occurs in special sites
Rumfelt, 2000) examination of this group has taught us much about the known as the epigonal organ, associated with the gonads, and the Leydig
evolution of adaptive immunity, as well as the conserved and organ, embedded within the wall of the esophagus (Fig. 1). Not all
taxon-specific characteristics of Igs. Sharks and their relatives have three elasmobranch species have a Leydig organ, for example, while found in
IgH chain isotypes: the first two, IgM (μ) (Clem et al., 1967; Marchalonis little skate (Leucoraja erinacea) it is not present in nurse sharks (Gin­
and Edelman, 1966) and IgW (ω) (Anderson et al., 1999; Berstein et al., glymostoma cirratum). Histologically both organs are similar in structure,

* Corresponding author. Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA.
E-mail address: hdooley@som.umaryland.edu (H. Dooley).

https://doi.org/10.1016/j.dci.2020.103873
Received 2 July 2020; Received in revised form 12 September 2020; Accepted 16 September 2020
Available online 23 September 2020
0145-305X/© 2020 Elsevier Ltd. All rights reserved.
H. Matz et al. Developmental and Comparative Immunology 115 (2021) 103873

with the spongy appearance of mammalian bone marrow (Luer et al., lymphopoietic organs (epigonal and Leydig organ) along with terminal
2004). Cartilaginous fishes, like other ectothermic vertebrates, lack deoxynucleotidyl transferase (TdT), which inserts N-nucleotides at the
lymph nodes (Zapata and Amemiya, 2000). Consequently, adaptive rearrangement junctions (Rumfelt et al., 2002). Rearrangement is pri­
immune responses occur primarily in the spleen, although there is some marily restricted within a cluster, and rarely occurs between different
evidence of lymphocyte activity in the gills, gut, and several other tis­ clusters (Malecek et al., 2008). Unlike mice and humans, where there is
sues (Hart et al., 1986; Rumfelt et al., 2002; Tomonaga et al., 1986). The a strict order of segmental rearrangement (always D to J first, then V to
shark spleen is highly vascularized and contains both red pulp and DJ for Ig heavy chains) the V, D, and J segments of shark Ig clusters can
defined white pulp regions. Organized zones of B cells are evident and recombine in any order (e.g. V to D1, D1 to D2, and D3 to J first round
may facilitate clonal selection and somatic hypermutation (SHM) (Cas­ rearrangements have all been observed; H. Dooley, unpublished data),
tro et al., 2013; Rumfelt et al., 2002), although true germinal centers and multiple Ig genes are accessible for recombination at one time
(GCs) have not been identified in any ectotherms to date (Zapata and (Malecek et al., 2008). Despite this, single cell PCRs amplified only one
Amemiya, 2000; Zapata et al., 1996). As the spleen houses both naïve B IgH V region transcript per B cell in the clearnose skate (Raja eglanteria)
cells and differentiated plasma cells, it is the major site of Ig expression (Eason and Litman, 2002) and no overlap is observed between IgM- and
and secretion (Castro et al., 2013; Rumfelt et al., 2002). While the epi­ IgNAR-secreting cells in nurse sharks (Rumfelt et al., 2002), indicating
gonal organ is hypothesized to be the home of long-lived plasma cells both isotypic and allelic exclusion occur. More recent studies have
(Castro et al., 2013) secretory Ig transcript has also been found in the shown that shark IgM clusters operate autonomously during VDJ
pancreas, gill, liver, kidney, and olfactory organ of nurse sharks (Rum­ recombination. This stochastic activation of IgH genes for rearrange­
felt et al., 2004a). Thus, as in mammals, Igs seemingly play a defensive ment initiation, combined with a restricted time window of accessibility,
role throughout all cartilaginous fish tissues. likely help maintain IgH chain exclusion (Zhu et al., 2011). It is hy­
pothesized that once a productive V region is produced from any of the
3. Genomic organization of cartilaginous fish Igs and repertoire accessible clusters it prevents rearrangement and expression of the
generation remaining Ig clusters, thereby maintaining B cell clonality (Zhu et al.,
2011).
Most vertebrate Ig genes are organized in a translocon configuration Based upon the limited number of V, (D) and J segments within a
(Fig. 2), in which multiple variable (V), diversity (D; only present in IgH single cartilaginous fish cluster it might be concluded that the shark
chains), and joining (J) segments are located upstream of constant (C) primary immune repertoire is restricted in diversity. However, several
domains for all Ig isotypes. In contrast, cartilaginous fish IgH and IgL mechanisms compensate for this. First, as noted above, every Ig isotype
chain genes are arranged in a cluster configuration (Fig. 2), in which a has multiple clusters with extensive germline-encoded heterogeneity
single V segment, one or more D segments (again, IgH chains only), and across complementarity determining regions (CDR)1 and 2 (Rast et al.,
a single J segment lie upstream of C region exons of a single isotype 1998; Rumfelt et al., 2004b). Second, each heavy chain cluster contains
(Hinds and Litman, 1986). The number of clusters varies between iso­ multiple D segments and thus requires multiple rearrangement events
types and between cartilaginous fish species: for example, nurse sharks (e.g. 4 for IgNAR), with associated trimming and N/P-addition, to
have ~15 IgM clusters (each having V, D, J segments and a set of Cμ generate a functional V region (Malecek et al., 2008). Finally, cartilag­
domains) and 3 IgNAR clusters (V, multiple Ds, J, and CNAR domains) inous fish have four IgL chain isotypes that are among the most heter­
while the spiny dogfish (Squalus acanthias) has >100 IgM and 10–20 ogenous of any vertebrate studied to date (Criscitiello and Flajnik, 2007;
IgNAR clusters (Malecek et al., 2005; Smith et al., 2012). While most Ig Fleurant et al., 2004). Thus, the weight of evidence suggests the primary
segments are unjoined in the cartilaginous fish germline genome, a small repertoire of cartilaginous fishes is just as diverse (or possibly even more
portion are found to be partially (VD-J) or fully (VDJ or VJ) diverse) than that of mammals. Study of random IgNAR cDNAs obtained
pre-rearranged or “germline-joined”; e.g. nurse sharks have both from peripheral blood lymphocyte samples showed a much lower mu­
germline-joined IgM and IgNAR clusters (Diaz et al., 2002; Kokubu et al., tation rate in transmembrane transcripts compared to secretory tran­
1988; Rumfelt et al., 2001). These germline-joined nurse shark IgH scripts, and only mutations in the latter were biased towards
genes are preferentially expressed early in ontogeny and are hypothe­ replacement mutations in the CDRs (Diaz et al., 1998). This indicates
sized to play unique protective and/or homeostatic roles in neonates that shark Igs undergo activation induced cytidine deaminase (AID)-­
(Diaz et al., 2002; Rumfelt et al., 2001). mediated somatic diversification following antigen stimulation akin to
Cartilaginous fishes use the same recombination mechanisms as mammals, rather than using mutation to generate the primary repertoire
mammals to generate a primary Ig repertoire, with recombination- like birds and rabbits. Interestingly, the mutation frequency of IgNAR is
activation gene (RAG1 and RAG2) enzymes active in the exceptionally high, surpassing even the upper limits reported for

Fig. 1. The major immunological organs of cartilaginous fishes and their relative positions.

2
H. Matz et al. Developmental and Comparative Immunology 115 (2021) 103873

Fig. 2. Schematic showing the different Ig gene arrangements in mammals and sharks. Mammalian Igs are found in a translocon arrangement, with multiple V
segments, D segments, and J segments arranged upstream of the C regions for all the different Ig isotypes. In contrast, cartilaginous fish Igs are found in a cluster
configuration, where a single V segment, two or more D segments, a J segment, and the C regions for a single isotype are present in each cluster, and multiple clusters
are present for each isotype. C regions are shown in grey, the segments that make up the variable region are colored by isotype.

mammalian V regions (Diaz et al., 1999). Study of random nurse shark 4. The Ig isotypes of cartilaginous fishes
IgNAR and IgL transcripts showed mutations accumulate in a pattern
similar to mammalian IgG, targeting AGC/T hotspots, but with a high 4.1. IgM
proportion of tandem mutations rather than singlets (Diaz et al., 1999;
Greenberg et al., 1995; Lee et al., 2002). The fact that high replacement A protein with characteristics similar to IgM was first discovered in
to synonymous (R/S) ratios were observed in the CDRs of some clones the plasma of cartilaginous fishes in the mid-1960s (Clem and Small Jr,
suggests positive selection can occur despite the apparent absence of 1967; Clem et al., 1967; Marchalonis and Edelman, 1966) and was
GCs (Diaz et al., 2002). Thus, the ability of cartilaginous fishes to originally thought to be the only isotype present. Indeed, in nurse shark
generate and diversify a primary Ig repertoire is by no means IgM makes up roughly half of the total serum protein and is present in
sub-optimal. Indeed, by all measures, sharks are as capable as mammals two forms, a monomeric form (mIgM or 7S) and a pentameric (pIgM or
of combating the potential universe of pathogens they may encounter. 19S) form (Fig. 3) in approximately equal amounts (Clem et al., 1967;
The cluster organization of cartilaginous fish Ig genes and absence of Small et al., 1970). The two forms do not interconvert (Small et al.,
recognizable classical (i.e. mammalian-like) switch (S) regions (Zhu 1970) and are hypothesized to be produced by different B cell lineages
et al., 2012) suggests this group lacks tetrapod-like class switch (Dooley and Flajnik, 2005).
recombination, which so far has only been identified as far back as Secretion of pIgM is coordinated with J chain expression in adult
amphibians (Muβmann et al., 1997). However, recent studies of nurse nurse sharks, and it has also been found that pIgM/J chain + nurse shark
shark Ig transcripts have shown that the V regions from one cluster may splenic B cells do not express the transcription factor Blimp-1, which
be expressed fused to the C domains of another, despite being separated controls plasma cell differentiation, and express lower levels of IgM
by large (>120 Kb) distances (Zhang et al., 2013; Zhu et al., 2012). transcript than mIgM B cells (Castro et al., 2013). In contrast, mIgM is
Unlike mammals this form of ‘switching’ is adirectional, i.e. IgW V re­ expressed in Blimp-1+/J chain- B cells (Castro et al., 2013) and is likely
gions can be expressed with IgM C regions and vice versa (Zhu et al., produced in a T cell-dependent manner (Clem et al., 1967; Dooley and
2012). As the number of switched transcripts is higher in immunized Flajnik, 2005; Marchalonis and Edelman, 1966), with antigen specific
animals it is hypothesized to occur via AID-initiated DNA lesions that titers of mIgM rising over the course of an immune response. Coupled
prompt recombination between the clusters (Zhu et al., 2012). We do with its non-specific nature, pIgM is thought to act as an innate-like,
not yet know how this process is directed or its biological consequence; ‘first line of defense’ against invading pathogens until a strong
however, it is possible this is the predecessor of conventional CSR as antigen-specific response can be developed. A lack of serum albumin in
found in other vertebrates. Thus, most of the fundamental components sharks suggests pIgM may also act as a carrier molecule or in osmo­
of Ig repertoire diversification, including V(D)J recombination, somatic regulation, in addition to acting as an innate defender (Dooley and
hypermutation (SHM), and a potential precursor of CSR were all present Flajnik, 2006; Fellows and Hird, 1981; Fellows et al., 1980). Like IgM in
in the common ancestor of jawed vertebrates. other vertebrates, the large size of pIgM restricts it to the vasculature,
while mIgM is capable of penetrating tissues similar to mammalian IgG
(Small et al., 1970).
Both the transmembrane and secreted forms of IgM contain 4 C do­
mains (Rumfelt et al., 2004a) but, interestingly, a third form with only 3
C domains (expressed from a separate cluster where Cμ2 is deleted,

3
H. Matz et al. Developmental and Comparative Immunology 115 (2021) 103873

Fig. 3. Illustration of the transmembrane and secretory immunoglobulin isoforms found in cartilaginous fishes to date. Constant domains are shown in grey while the
variable domains of each are colored by isotype. Secreted IgM pentamers are stabilized by joining (J) chain.

making it structurally convergent with mammalian IgG) predominates preplasmablasts also lack Blimp-1 expression (Kallies et al., 2007), and
in the plasma of neonatal animals and pups (Rumfelt et al., 2001). This thus the absence of Blimp-1 in pIgM B cells would support model 2.
IgM form, IgM1gj, forms both monomers and dimers, has an entirely However, if pIgM are truly a transitional stage preceding mIgM cells
germline-joined (VDJ) V domain (Rumfelt et al., 2001) and preferen­ their high proportion and presence in the shark spleen regardless of
tially associates with a germline-joined L chain ((Lee et al., 2000) Flajnik immune state would suggest that this stage is more long-lived than seen
& Hsu, unpublished data), giving this Ig a completely predetermined in mammals (Castro et al., 2013). More work is required to prove which
binding site. As pups age the levels of pIgM and mIgM gradually increase of these two models, or a combination of both, explains the role of pIgM
to become the dominant Ig forms. Like pIgM secreting cells, IgM1gj B and mIgM shark B cell lineages.
cells are J chain+/Blimp-1-, and while they can be found in the spleen of
nurse shark pups, they are only found in the epigonal organ of adult 4.2. IgNAR
animals (Castro et al., 2013).
There are two proposed models for how the pIgM and mIgM B cells First reported in 1995, IgNAR is a novel Ig isotype that has so far only
arise in the shark: model 1 suggests that these cells types are determined been found in elasmobranchs. Like the novel heavy chain antibodies
at the stem cell stage, and that pIgM and mIgM-secreting cells are found in Camelidae (camels, llamas, and their relatives), IgNAR is also a
derived from distinct lineages akin to B1 and B2 cell populations in homodimer of IgH chains that does not associate with IgL chains
mammals. Model 2 suggests that pIgM cells are a distinct developmental (Greenberg et al., 1995; Roux et al., 1998), although the two isotypes
stage preceding mIgM cells, akin to plasmablasts (Castro et al., 2013; evolved convergently (Nguyen et al., 1998, 1999). In nurse shark there is
Dooley and Flajnik, 2005). The early appearance of pIgM B cells in a single secretory form of IgNAR where an N-terminal V domain (VNAR)
ontogeny coupled with their lack of Blimp-1 expression, suggesting a is joined to five C (C1–C5) domains (Fig. 3) (Greenberg et al., 1996;
potential for self-renewal, is similar to the phenotype of mammalian B1 Rumfelt et al., 2004a). A second form of secretory IgNAR, where C1 is
cells which do not require Blimp-1 upregulation for antibody secretion spliced directly to C4 (i.e. ΔC2-C3), has recently been found in the
(Tumang et al., 2005), supporting model 1. However, mouse whitespotted bamboo shark (Chiloscyllium plagiosum) (Zhang et al.,

4
H. Matz et al. Developmental and Comparative Immunology 115 (2021) 103873

2020). Membrane bound IgNAR is also expressed as long (five C do­ dramatically alter the structural topology of the variable loops. Based
mains) and short forms (three C domains), however here the short form upon the number and position of non-canonical cysteine residues,
is missing the C4 and C5 domains (Rumfelt et al., 2004a). Phylogenetic VNARs are categorized into several ‘types’ (Fig. 4). Type I VNARs, so far
and structural analyses revealed that VNARs share greater structural identified only in nurse sharks (Greenberg et al., 1995; Matz and Dooley,
homology with T cell receptor (TCR) and IgL V domains than conven­ 2019), carry an even number of cysteines (2, 4, or 6) in CDR3 and one
tional IgH V regions (Greenberg et al., 1995; Richards and Nelson, 2000; each in FR2 and FR4. Disulphide bond formation between these residues
Stanfield et al., 2004). VNAR-like domains are also found in NAR-TCR, a causes the CDR3 loop to be pinned tightly against the side of the
doubly rearranging T cell receptor unique to cartilaginous fishes, sug­ molecule (Stanfield et al., 2004). Type II VNAR domains, identified in
gesting a common ancestral origin for these domains (Criscitiello et al., several shark species (Liu et al., 2007; Nuttall et al., 2001; Zielonka
2006). As the constant domains (C2–C5) of IgNAR are homologous to et al., 2014), carry additional cysteines in CDR1 and CDR3 which results
those of IgW, it is hypothesized that IgNAR originated through the in­ in an intra-molecular disulphide bond that brings the loops closer
vasion of an IgW cluster by the V region of NAR-TCR (Criscitiello et al., together, forming a protrusive ‘finger-like’ structure at the top of the
2006; Greenberg et al., 1996). Lacking light chains IgNAR does not have domain (Streltsov et al., 2005). Type III VNARs, so far found only in
the combinatorial diversity generated through VH-VL pairing in con­ nurse sharks, also carry extra non-canonical cysteines in CDR1 and
ventional antibodies. However, as mentioned above, this deficiency is CDR3. However, because two of the three diversity regions of type III
more than compensated by the multiple rearrangement events required VNARs are germline joined, they show less diversity in CDR3 compared
to assemble their three D regions (Fig. 2). This generates CDR3s that are to type I or II VNARs (Diaz et al., 2002). Like IgM1gj, type III VNARs are
highly diverse in both length (7–34 aa) and sequence composition, mainly expressed during neonatal development, before maturation of
bringing considerable heterogeneity to the primary repertoire (Diaz the antigen-driven response; however, some expression is maintained in
et al., 1998; Flajnik and Dooley, 2009; Greenberg et al., 1995). Upon the epigonal organ into adulthood (Diaz et al., 2002). Type IV domains
exposure to antigen VNARs are further diversified by extremely high (alternately called type IIb) only have the canonical cysteines possessed
levels of SHM (Diaz et al., 1999). by all Ig domains, which may make their paratope topology more flex­
IgNAR levels in nurse shark serum are approximately 10x lower than ible (Kovalenko et al., 2013). The VNAR-antigen complexes crystalized
those observed for IgM, likely due to the presence of fewer IgNAR thus far indicate that VNARs have a propensity to target clefts and re­
clusters and the apparent susceptibility of IgNAR proteins to proteolysis cesses on their target antigens. Indeed, VNARs appear to exploit this
(Dooley and Flajnik, 2005; Roux et al., 1998). In nurse shark, IgNAR is binding strategy to increase their surface area of interaction with a
found only as a monomer, but in spiny dogfish and small spotted cat­ target, burying as much surface area in the interaction as a conventional
shark (Scyliorhinus canicula) it has been found as both a monomer and a H-L chain antibody, and facilitating high affinity interactions (Fig. 4).
multimer (Crouch et al., 2013; Smith et al., 2012). Finally, VNARs exist that do not conform to any of the classical types
The role of IgNAR in the adaptive response is undoubtedly enhanced described above; for example, in a recent analysis of 1.2 million VNAR
by the diversity of VNAR structures which enable this isotype to bind a transcripts isolated from naïve nurse sharks, a small percentage (~5%)
wide array of epitopes. VNAR domains act as independent soluble units, could not be classified into any of the traditional types based upon
that are attached to the C1 domain by an extended flexible tether rather cysteine number or location (Feng et al., 2019). Further structural and
than a conventional Ig hinge region (Roux et al., 1998). While VNARs functional characterization is needed to understand the biophysical
adopt the typical conformation of Ig superfamily domains, a large properties of these previously undescribed VNAR types.
portion of framework region 2 (FR2) and CDR2 is deleted (Greenberg The inherent attributes of VNARs, i.e., their small size, unique
et al., 1995) giving this domain its characteristically small size (approx. structural topologies, propensity to bind clefts and pockets on target
12 kDa). The loss of CDR2 is compensated by two loops of high amino antigens, and their high thermostability, make VNARs interesting
acid diversity, hypervariable region 2 (HV2) that forms a belt-like prospects as future diagnostic and therapeutic agents (reviewed in (Matz
structure around the VNAR domain and hypervariable region 4 (HV4) and Dooley, 2019).
that lies at the top of the domain next to CDR1 (Stanfield et al., 2004). In
addition to the cysteines that typify Ig domains, VNARs also contain
non-canonical cysteines that can form additional disulphide bonds and

Fig. 4. Structural analyses show that VNARs (blue) can complex their target antigens (gold) using a range of different binding modalities. The VNAR CDR1 is shown
in yellow, CDR3 in red, HV2 in green, and HV4 in cyan in all panels. As can be seen in (a) the type I VNAR 5A7 contacts HEL via CDR1 and a folded CDR3 that is
constrained by disulfide bonding to the FR2 and FR4 (PDB 1T6V) while (b) the type II VNAR PBLA8 contacts hen egg lysozyme (HEL) via CDR1 and an extended
CDR3 loop projecting deep into HELs enzymatic cleft (PDB 2I25). In contrast, (c) the type IV/IIb VNAR E06 binds its target human serum albumin (HSA) through
CDR3 and HV2 (PDB 4HGK), and (d) the synthetic VNAR D01 contacts Aurora-A kinase via CDR1, CDR3, and HV2 (PDB 5L8L). From Matz and Dooley (2019). (For
interpretation of the references to color in this figure legend, the reader is referred to the Web version of this article.)

5
H. Matz et al. Developmental and Comparative Immunology 115 (2021) 103873

4.3. IgW antibodies for this isotype, will yield further insights regarding the
evolution of this isotype and its contribution to humoral immunity.
IgW was first identified in ocellate spot skate (Raja kenojei) and little
skate (Raja erinacea) (Harding et al., 1990; Kunihiko et al., 1984) where 4.4. IgL chains
it was named IgR and IgX, and later in nurse shark as IgNARC (Green­
berg et al., 1996) then sandbar shark (Carcharhinus plumbeus) as IgW Early immunization studies recognized that shark Igs contained L
(Berstein et al., 1996). With the discovery of an additional ortholog in chains akin to canonical mammalian Igs (Clem et al., 1967). It is now
lungfish (Ota et al., 2003) the cartilaginous fish Igs were reconciled into known that cartilaginous fishes have four L chain isotypes. The
one isotype, IgW, and recognized as orthologs of tetrapod IgD (Ohta and nomenclature of Ig L chains in chondrichthyans was confusing for many
Flajnik, 2006). Thus, both IgM-like and IgW/D-like molecules must have years, as it was difficult to determine the classification of L chains across
been present in the ancestor of all jawed vertebrates (Ohta and Flajnik, vertebrate taxa due to large phylogenetic distances (Dooley and Flajnik,
2006). Curiously, IgW appears to be lost from the elephant shark (Cal­ 2006; Smith et al., 2014). It is now established that sharks possess λ
lorhinchus milii) genome, a holocephalan and ancient chondrichthyan (formerly Type II/NS3), κ (formerly Type III/NS4), σ, and σ-cart/σ-2
(Venkatesh et al., 2014), but has been found in every elasmobranch (formerly Type I/NS5) L chains (Criscitiello and Flajnik, 2007). Like the
species yet examined. H chains, shark Ig L chain genes are organized in a cluster configuration
Several forms of IgW have been discovered. Based on transcript and (Greenberg et al., 1993; Shamblott and Litman, 1989). Of these loci in
protein S35 labeling, there exists both long (6 and 8Cω) and short (2 and nurse shark, κ and σ-cart contain both germline-joined and unjoined or
4Cω) secreted forms of IgW (Fig. 3) (Greenberg et al., 1996; Rumfelt split clusters, σ contains no germline-joined clusters, and all λ clusters
et al., 2004a; Zhang et al., 2013). Transcript data has also confirmed the are germline-joined (Fig. 5) (Criscitiello and Flajnik, 2007; Fleurant
existence of two transmembrane forms of IgW (2 and 4Cω) (Rumfelt et al., 2004; Lee et al., 2000; Rast et al., 1994). The number of clusters of
et al., 2004a; Zhang et al., 2013). The short secretory form (2Cω) has a each isotype, like Ig H chains, varies between shark species, as does the
long, cysteine-rich tail, unlike that of any other Ig, suggesting its effector proportion of split versus germline-joined clusters (Fleurant et al., 2004;
function is different from other isoforms (Rumfelt et al., 2004a). It is Hohman et al., 1992; Rast et al., 1994).
likely that the different forms are generated through alternative splicing It has been proposed that germline-joined Igs arose as a consequence
(Zhang et al., 2013). Additionally, transcript for a 6Cω form in which the of RAG activity in the germline (Lee et al., 2000), however, it should be
leader is spliced directly to the C1, resulting in a molecule lacking the V noted that other scenarios have not been entirely ruled out; for example,
domain (IgWΔV), was discovered in spiny dogfish and nurse shark retrotransposition of a rearranged VDJ transcript back into the germline
(Smith et al., 2012). The existence of IgWΔV in two species of shark is a possibility, but the presence of a split leader (Anderson et al., 1995)
separated by ~200 million years of evolution, at least at the transcript makes this explanation far less likely. While it might be thought that
level, suggests this form fulfils a unique functional role. It has been germline-joined L chains would restrict the antibody repertoire, λ L
speculated that the C domains of IgW/D can act as pattern recognition chain loci hypermutate at high rates in an antigen-driven fashion that
molecules (Edholm et al., 2010; Seifert et al., 2009), binding directly to results in tandem mutations, leading to diverse sequences despite a lack
pathogens then triggering antimicrobial and immunostimulatory re­ of junctional diversity (Lee et al., 2002). For isotypes with split clusters
sponses (Chen et al., 2009). The discovery of IgWΔV in sharks can help that undergo V-J rearrangement (e.g. κ and σ-cart) the majority of clones
us put this hypothesis to the test. examined carry N nucleotide additions, vastly increasing light chain
A lack of monoclonal Abs (mAbs) for IgW has made it difficult to CDR3 repertoire diversity (Fleurant et al., 2004).
study its function at the protein level, but the plethora of transcript The major outstanding question from L chain research in shark is, if
forms suggests IgW (along with IgNAR) has several diverse functions. these four clades are present in the oldest phylogenetic group with Ig-
Serum protein levels of IgW are low in adult nurse shark, however based immunity, what was the model for their evolution and the bio­
transcript levels are high in spleen, epigonal, pancreas, thymus, and gill, logical significance of each isotype? As might be predicted, neither of
suggesting IgW plays a role in mucosal protection (Rumfelt et al., 2004a; these questions has been simple to answer. Several models are proposed
Smith et al., 2012; Zhang et al., 2013). The short secreted form of IgW for the origins of these L chain isotypes and how Ig antigen receptors
varies greatly in expression levels, both between juvenile and adult evolved (Criscitiello and Flajnik, 2007), but what is apparent is that
nurse sharks, and between individuals in the same cohort (Rumfelt et al., σ-cart is a “dead-end” isotype found only in cartilaginous fishes, σ is
2004a). In situ hybridization shows transcription of the IgW long maintained only in ectothermic vertebrate lineages (Criscitiello and
secretory form overlaps with that of J chain in the spleen and epigonal Flajnik, 2007; Schwager et al., 1991), and only κ and λ are maintained in
organ of nurse sharks, suggesting it may form multimers (Castro et al., the endothermic vertebrate lineages. The functional difference between
2013). κ and λ is not even clear in mammals (Hershberg and Shlomchik, 2006;
We hypothesize that the diverse forms of secretory IgW generated Sverremark et al., 2000; Townsend et al., 2016), so the benefit of four
through splicing provide a means to control the effector functions trig­ different L chain isotypes in cartilaginous fish is equally unclear.
gered (as has been observed for the full-length and short (ΔFc) forms of
mallard duck IgY (Humphrey et al., 2004)), however this requires 5. Ig responses in sharks
formal testing. The presence of two IgW Tm forms also remains a mys­
tery, although the Tm forms have fewer flexible hinge regions, possibly Immunization studies performed in the mid-1960s suggested that the
helping reduce their proteolytic shedding from the B cell surface (Smith antibody responses of cartilaginous fishes differed substantially from
et al., 2012; Zhang et al., 2013). Thus, it is possible cells expressing those of mammals; following a long (2–3 month) lag period serum IgM
different versions of IgW-Tm have different activation requirements levels climbed slowly. While the proportion of mIgM to pIgM changed in
(Zhang et al., 2013). So far, no germline-joined versions of IgW have some studies, binding affinities did not appear to increase appreciably
been discovered, suggesting there are not innate roles early in ontogeny over the course of most responses (Clem and Small Jr, 1967; Clem et al.,
for this isotype (Rumfelt et al., 2004a). Unlike IgM and IgD in mammals, 1967; Marchalonis and Edelman, 1966). Thus, for a long time, it was
IgM and IgW do not appear to be expressed simultaneously on the sur­ thought that the antibody response in cartilaginous fishes was somewhat
face of cartilaginous fish B cells (Eason et al., 2004). While the role of ‘lacking’ when compared to mammals. However, pioneering work in
IgD is not fully understood even in mammals, comparative studies have nurse sharks determined how to generate true antigen-specific humoral
already increased our knowledge of this enigmatic isotype (Chen et al., responses and revealed that sharks are capable of affinity maturing their
2009; Edholm et al., 2010). We believe that further investigation of Ig repertoires (Dooley and Flajnik, 2005). While the affinity maturation
shark IgW, which would be greatly facilitated by the development of observed was 10 fold rather than the ~100 fold change seen in mice

6
H. Matz et al. Developmental and Comparative Immunology 115 (2021) 103873

Fig. 5. Like IgH chains, cartilaginous fish IgL chains can be found in both split and germline-joined configurations. Notably, the λ chains of all species examined to
date are all germline-joined and σ are split, while the other isotypes are present in varying ratios of split and joined in different species.

(Dooley et al., 2006), it is presumed to be no less effective at protecting immunity? The discovery that cartilaginous fishes employ V(D)J
the host against pathogens. recombination and somatic hypermutation to generate a highly diverse
Functional studies indicate distinct functional roles for pIgM and repertoire of binding specificities, can affinity mature their Ig pool, and
mIgM in the shark humoral response. Low affinity, high avidity pIgM maintain antigen-specific clones to provide long-term immunological
can be found prior to injection of antigen and affinities of antigen- memory (as detailed earlier) indicates these traits - long-associated with
specific pIgM do not increase during an antigen-driven immune Ig-mediated protection in mammals - were almost certainly present in
response, suggesting that affinity maturation does not occur in this Ig the common ancestor of all jawed vertebrates. The apparent division of
form (Dooley and Flajnik, 2005; Leslie and Clem, 1970; Voss and Sigel, function between cartilaginous fish innate-like Igs (pIgM and IgM1gj)
1972). It has been shown that pIgM serum levels can increase exclu­ and adaptive Igs (mIgM and IgNAR) suggests B1-like and B2-like cell
sively in response to bacterial carbohydrate, suggesting a T cell inde­ lineages arose early in vertebrate evolution. Segregation of these cells
pendent response (Clem and Leslie, 1971; Shankey and Clem, 1980a, b). followed by sequencing of their V region repertoires would allow us to
In contrast, mIgM affinity matures in an antigen-driven manner during test this hypothesis, not a simple task when species-specific monoclonal
the humoral response. This response includes a delay in mIgM produc­ antibodies are lacking.
tion following antigen exposure, an increase in antibody affinity over Further, the differential use of multiple Ig isotypes, combined with
the duration of the response, and maintenance of long term, the presence of multiple forms of some isotypes (Dooley and Flajnik,
antigen-specific mIgM memory (Dooley and Flajnik, 2005; Eve et al., 2005; Rumfelt et al., 2004a), indicates that cartilaginous fishes can
2020; Rumfelt et al., 2002). That mIgM undergoes affinity maturation in tailor their response to achieve different immunological outcomes like
an antigen-driven manner suggests there is a mechanism in sharks for B other vertebrates. However, significant work is required to understand
cell selection reminiscent of mammalian GCs; however, as noted above, the effector functions mediated by the different shark Ig isotypes. The
true GCs that would mediate this process have not yet been observed in development of additional isotype-specific mAbs would greatly advance
cartilaginous fishes (Zapata and Amemiya, 2000). these studies; currently, the research tools available only allow for
Akin to mIgM, antigen specific IgNAR titers also rise slowly after limited study of IgM and IgNAR. Once the in vivo roles of each isotype
immunization and exhibit affinity maturation over the course of a have been established it should also become easier to understand the
response. The lag-period necessary to generate an antigen-driven IgNAR functional consequences of the atypical form of CSR found in sharks.
response, coupled with a lack of IgNAR in neonatal serum, strongly Such mAbs would also facilitate flow cytometry experiments, permitting
suggests that IgNAR responses require T cell help (Dooley and Flajnik, the isolation of isotype-specific B cells from their native sites for further
2005; Rumfelt et al., 2002). characterization.
The Ig titers achieved through immunization in nurse sharks persist The ability of cartilaginous fishes to affinity mature their Ig reper­
up to 1–3 years before returning to initial pre-immunization levels, toire (Dooley and Flajnik, 2005; Dooley et al., 2006) in the apparent
suggesting long-term circulating protection against antigens. Further, absence of GCs is also a point that requires further exploration. We
antigen-specific Ig expression can be re-stimulated long (>8 years) after hypothesize the presence of alternative selective environments (perhaps
primary antigen exposure, clearly demonstrating B cell memory (Dooley evolutionary precursors to mammalian-like GCs?) that facilitate the
and Flajnik, 2005; Eve et al., 2020). Collectively, this data demonstrates selection and expansion of antigen-specific B cell clones. The cloning of
that sharks are capable of Ig responses of the same caliber as endo­ cartilaginous fish AID (Conticello et al., 2005; Quinlan et al., 2017) and
therms, albeit on longer timescales. orthologs of other molecules key to the germinal center reaction in
mammals, will allow this hypothesis to be put to the test.
6. Perspective and future directions Now that long term memory has unequivocally been proven in
cartilaginous fishes (Eve et al., 2020), we next need to confirm where
From what has been learned about Igs in sharks and their relatives, long-lived memory B cells reside while awaiting recall and the signaling
what conclusions can we draw about the evolution of adaptive molecules required to recruit and sustain them. It is hypothesized that

7
H. Matz et al. Developmental and Comparative Immunology 115 (2021) 103873

long-lived memory B cells reside in the epigonal organ of nurse shark Dooley, H., Flajnik, M., 2006. Antibody repertoire development in cartilaginous fish.
Dev. Comp. Immunol. 30, 43–56.
(Castro et al., 2013) however the work performed so far has been unable
Dooley, H., Flajnik, M.F., 2005. Shark immunity bites back: affinity maturation and
to prove this. It may be that the frequency and diversity of the memory response in the nurse shark, Ginglymostoma cirratum. Eur. J. Immunol. 35,
antigen-specific B cells retained for memory are few in number, as has 936–945.
previously been observed in studies of mammalian immunological 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.
memory (Mesin et al., 2020) making these cells difficult to find. It would Proc. Natl. Acad. Sci. U. S. A 103, 1846–1851.
also be interesting to establish if there is a division in B cell fates in Eason, D.D., Litman, G.W., 2002. Haplotype Exclusion: the Unique Case Presented by
sharks, for example, between long-lived plasma cells and memory B cells Multiple Immunoglobulin Gene Loci in Cartilaginous Fish, Seminars in Immunology.
Elsevier, pp. 145–152.
as observed in mammals. Eason, D.D., Litman, R.T., Luer, C.A., Kerr, W., Litman, G.W., 2004. Expression of
Moving forward we anticipate the growing number of sequence individual immunoglobulin genes occurs in an unusual system consisting of multiple
datasets for cartilaginous fishes and application of new technologies, independent loci. Eur. J. Immunol. 34, 2551–2558.
Edholm, E.-S., Bengtén, E., Stafford, J.L., Sahoo, M., Taylor, E.B., Miller, N.W.,
such as single cell sequencing and high-throughput proteomics, will Wilson, M., 2010. Identification of two IgD+ B cell populations in channel catfish,
greatly facilitate our work. Such approaches should help overcome the Ictalurus punctatus. J. Immunol. 185, 4082–4094.
paucity of molecular tools available for cartilaginous fishes and better Eve, O., Matz, H., Dooley, H., 2020. Proof of long-term immunological memory in
cartilaginous fishes. Dev. Comp. Immunol. 108, 103674.
our understanding of adaptive immunity in this key lineage. As our Fellows, F., Hird, F., 1981. Fatty acid binding proteins in the serum of various animals.
knowledge increases, it will become easier to distinguish lineage- Comp. Biochem. Physiol. Part B: Comparative Biochemistry 68, 83–87.
specific traits from those shared across jawed vertebrates, and un­ Fellows, F.C., Hird, F., McLean, R.M., Walker, T., 1980. A survey of the non-esterified
fatty acids and binding proteins in the plasmas of selected animals. Comp. Biochem.
doubtedly result in a better understanding of Ig-mediated adaptive im­
Physiol. Part B: Comparative Biochemistry 67, 593–597.
munity in both human and non-human animals. Feng, M., Bian, H., Wu, X., Fu, T., Fu, Y., Hong, J., Fleming, B.D., Flajnik, M.F., Ho, M.,
2019. Construction and next-generation sequencing analysis of a large phage-
Acknowledgements displayed VNAR single-domain antibody library from six naive nurse sharks.
Antibody therapeutics 2, 1–11.
Flajnik, M., Rumfelt, L., 2000. The Immune System of Cartilaginous Fish, Origin and
HM is supported by NIH-NIAID predoctoral fellowship Evolution of the Vertebrate Immune System. Springer, pp. 249–270.
F31AI147532. DM was supported by an Elphinstone PhD Scholarship Flajnik, M.F., Dooley, H., 2009. The Generation and Selection of Single-Domain, V
Region Libraries from Nurse Sharks. In: Antibody Phage Display. Springer,
awarded by the University of Aberdeen. pp. 71–82.
Fleurant, M., Changchien, L., Chen, C.-T., Flajnik, M.F., Hsu, E., 2004. Shark Ig light
References chain junctions are as diverse as in heavy chains. J. Immunol. 173, 5574–5582.
Greenberg, A.S., Avila, D., Hughes, M., Hughes, A., 1995. A new antigen receptor gene
family that undergoes rearrangement and extensive somatic diversification in sharks.
Anderson, M.K., Shamblott, M.J., Litman, R.T., Litman, G.W., 1995. Generation of
Nature 374, 168.
immunoglobulin light chain gene diversity in Raja erinacea is not associated with
Greenberg, A.S., Hughes, A.L., Guo, J., Avila, D., McKinney, E.C., Flajnik, M.F., 1996.
somatic rearrangement, an exception to a central paradigm of B cell immunity.
A novel “chimeric” antibody class in cartilaginous fish: IgM may not be the
J. Exp. Med. 182, 109–119.
primordial immunoglobulin. Eur. J. Immunol. 26, 1123–1129.
Anderson, M.K., Strong, S.J., Litman, R.T., Luer, C.A., Amemiya, C.T., Rast, J.P.,
Greenberg, A.S., Steiner, L., Kasahara, M., Flajnik, M.F., 1993. Isolation of a shark
Litman, G.W., 1999. A long form of the skate IgX gene exhibits a striking
immunoglobulin light chain cDNA clone encoding a protein resembling mammalian
resemblance to the new shark IgW and IgNARC genes. Immunogenetics 49, 56–67.
kappa light chains: implications for the evolution of light chains. Proc. Natl. Acad.
Berstein, R., Schluter, S.F., Shen, S., Marchalonis, J.J., 1996. A new high molecular
Sci. Unit. States Am. 90, 10603–10607.
weight immunoglobulin class from the carcharhine shark: implications for the
Harding, F.A., Amemiya, C.T., Litman, R.T., Cohen, N., Litman, G.W., 1990. Two distinct
properties of the primordial immunoglobulin. Proc. Natl. Acad. Sci. Unit. States Am.
immunoglobulin heavy chain isotypes in a primitive, cartilaginous fish, Raja
93, 3289–3293.
erinacea. Nucleic Acids Res. 18, 6369–6376.
Castro, C.D., Ohta, Y., Dooley, H., Flajnik, M.F., 2013. Noncoordinate expression of J-
Hart, S., Wrathmell, A., Harris, J., 1986. Ontogeny of gut-associated lymphoid tissue
chain and B limp-1 define nurse shark plasma cell populations during ontogeny. Eur.
(GALT) in the dogfishScyliorhinus canicula L. Vet. Immunol. Immunopathol. 12,
J. Immunol. 43, 3061–3075.
107–116.
Chen, K., Xu, W., Wilson, M., He, B., Miller, N.W., Bengten, E., Edholm, E.-S., Santini, P.
Hershberg, U., Shlomchik, M.J., 2006. Differences in potential for amino acid change
A., Rath, P., Chiu, A., 2009. Immunoglobulin D enhances immune surveillance by
after mutation reveals distinct strategies for κ and λ light-chain variation. Proc. Natl.
activating antimicrobial, proinflammatory and B cell–stimulating programs in
Acad. Sci. Unit. States Am. 103, 15963–15968.
basophils. Nat. Immunol. 10, 889.
Hinds, K., Litman, G., 1986. Major reorganization of immunoglobulin VH segmental
Clem, L., Small Jr., P., 1967. Phylogeny of immunoglobulin structure and function: I.
elements during vertebrate evolution. Nature 320, 546.
Immunoglobulins of the lemon shark. J. Exp. Med. 125, 893–920.
Hohman, V.S., Schluter, S.F., Marchalonis, J.J., 1992. Complete sequence of a cDNA
Clem, L.W., De Boutaud, F., Sigel, M.M., 1967. Phylogeny of immunoglobulin structure
clone specifying sandbar shark immunoglobulin light chain: gene organization and
and function: II. Immunoglobulins of the nurse shark. J. Immunol. 99, 1226–1235.
implications for the evolution of light chains. Proc. Natl. Acad. Sci. Unit. States Am.
Clem, L.W., Leslie, G.A., 1971. Production of 19S IgM antibodies with restricted
89, 276–280.
heterogeneity from sharks. Proc. Natl. Acad. Sci. Unit. States Am. 68, 139–141.
Humphrey, B.D., Calvert, C.C., Klasing, K.C., 2004. The ratio of full length IgY to
Conticello, S.G., Thomas, C.F.J., Petersen-Mahrt, S.K., Neuberger, M.S., 2005. Evolution
truncated IgY in immune complexes affects macrophage phagocytosis and the acute
of the AID/APOBEC family of polynucleotide (Deoxy)cytidine deaminases. Mol. Biol.
phase response of mallard ducks (Anas platyrhynchos). Dev. Comp. Immunol. 28,
Evol. 22, 367–377.
665–672.
Criscitiello, M.F., Flajnik, M.F., 2007. Four primordial immunoglobulin light chain
Inoue, J.G., Miya, M., Lam, K., Tay, B.-H., Danks, J.A., Bell, J., Walker, T.I.,
isotypes, including λ and κ, identified in the most primitive living jawed vertebrates.
Venkatesh, B., 2010. Evolutionary origin and phylogeny of the modern
Eur. J. Immunol. 37, 2683–2694.
holocephalans (Chondrichthyes: chimaeriformes): a mitogenomic perspective. Mol.
Criscitiello, M.F., Saltis, M., Flajnik, M.F., 2006. An evolutionarily mobile antigen
Biol. Evol. 27, 2576–2586.
receptor variable region gene: doubly rearranging NAR-TcR genes in sharks. Proc.
Kallies, A., Hasbold, J., Fairfax, K., Pridans, C., Emslie, D., McKenzie, B.S., Lew, A.M.,
Natl. Acad. Sci. U.S.A. 103, 5036–5041.
Corcoran, L.M., Hodgkin, P.D., Tarlinton, D.M., Nutt, S.L., 2007. Initiation of
Crouch, K., Smith, L.E., Williams, R., Cao, W., Lee, M., Jensen, A., Dooley, H., 2013.
plasma-cell differentiation is independent of the transcription factor Blimp-1.
Humoral immune response of the small-spotted catshark, Scyliorhinus canicula. Fish
Immunity 26, 555–566.
Shellfish Immunol. 34, 1158–1169.
Kokubu, F., Hinds, K., Litman, R., Shamblott, M., Litman, G., 1988. Complete structure
Diaz, M., Greenberg, A.S., Flajnik, M.F., 1998. Somatic hypermutation of the new antigen
and organization of immunoglobulin heavy chain constant region genes in a
receptor gene (NAR) in the nurse shark does not generate the repertoire: possible
phylogenetically primitive vertebrate. EMBO J. 7, 1979–1988.
role in antigen-driven reactions in the absence of germinal centers. Proc. Natl. Acad.
Kovalenko, O.V., Olland, A., Piché-Nicholas, N., Godbole, A., King, D., Svenson, K.,
Sci. Unit. States Am. 95, 14343–14348.
Calabro, V., Müller, M.R., Barelle, C.J., Somers, W., 2013. Atypical antigen
Diaz, M., Stanfield, R.L., Greenberg, A.S., Flajnik, M.F., 2002. Structural analysis,
recognition mode of a shark immunoglobulin new antigen receptor (IgNAR) variable
selection, and ontogeny of the shark new antigen receptor (IgNAR): identification of
domain characterized by humanization and structural analysis. J. Biol. Chem. 288,
a new locus preferentially expressed in early development. Immunogenetics 54,
17408–17419.
501–512.
Kunihiko, K., Susumu, T., 1988. The second immunoglobulin class is commonly present
Diaz, M., Velez, J., Singh, M., Cerny, J., Flajnik, M.F., 1999. Mutational pattern of the
in cartilaginous fish belonging to the order Rajiformes. Mol. Immunol. 25, 115–120.
nurse shark antigen receptor gene (NAR) is similar to that of mammalian Ig genes
Kunihiko, K., Susumu, T., Tadashi, K., 1984. A second class of immunoglobulin other
and to spontaneous mutations in evolution: the translesion synthesis model of
than IgM present in the serum of a cartilaginous fish, the skate, Raja kenojei:
somatic hypermutation. Int. Immunol. 11, 825–833.
isolation and characterization. Mol. Immunol. 21, 397–404.

8
H. Matz et al. Developmental and Comparative Immunology 115 (2021) 103873

Lee, S.S., Fitch, D., Flajnik, M.F., Hsu, E., 2000. Rearrangement of immunoglobulin genes Rumfelt, L.L., Diaz, M., Lohr, R.L., Mochon, E., Flajnik, M.F., 2004a. Unprecedented
in shark germ cells. J. Exp. Med. 191, 1637–1648. multiplicity of Ig transmembrane and secretory mRNA forms in the cartilaginous
Lee, S.S., Tranchina, D., Ohta, Y., Flajnik, M.F., Hsu, E., 2002. Hypermutation in shark fish. J. Immunol. 173, 1129–1139.
immunoglobulin light chain genes results in contiguous substitutions. Immunity 16, Rumfelt, L.L., Lohr, R.L., Dooley, H., Flajnik, M.F., 2004b. Diversity and repertoire of
571–582. IgW and IgM VH families in the newborn nurse shark. BMC Immunol. 5, 8.
Leslie, G.A., Clem, L.W., 1970. Reactivity of normal shark immunoglobulins with Saha, N.R., Ota, T., Litman, G.W., Hansen, J., Parra, Z., Hsu, E., Buonocore, F.,
nitrophenyl ligands. J. Immunol. 105, 1547–1552. Canapa, A., Cheng, J.F., Amemiya, C.T., 2014. Genome complexity in the coelacanth
Liu, J.L., Anderson, G.P., Delehanty, J.B., Baumann, R., Hayhurst, A., Goldman, E.R., is reflected in its adaptive immune system. J. Exp. Zool. B Mol. Dev. Evol. 322,
2007. Selection of cholera toxin specific IgNAR single-domain antibodies from a 438–463.
naive shark library. Mol. Immunol. 44, 1775–1783. Schwager, J., Bürckert, N., Schwager, M., Wilson, M., 1991. Evolution of
Luer, C.A., Walsh, C.J., Bodine, A., Wyffels, J.T., Scott, T.R., 1995. The elasmobranch immunoglobulin light chain genes: analysis of Xenopus IgL isotypes and their
thymus: anatomical, histological, and preliminary functional characterization. contribution to antibody diversity. EMBO J. 10, 505–511.
J. Exp. Zool. 273, 342–354. Seifert, M., Steimle-Grauer, S.A., Goossens, T., Hansmann, M.L., Brauninger, A.,
Luer, C.A., Walsh, C.J., Bodine, A.B., 2004. The Immune System of Sharks, Skates, and Kuppers, R., 2009. A model for the development of human IgD-only B cells:
Rays, Biology of Sharks and Their Relatives. CRC Press, pp. 377–403. genotypic analyses suggest their generation in superantigen driven immune
Malecek, K., Brandman, J., Brodsky, J.E., Ohta, Y., Flajnik, M.F., Hsu, E., 2005. Somatic responses. Mol. Immunol. 46, 630–639.
hypermutation and junctional diversification at Ig heavy chain loci in the nurse Shamblott, M., Litman, G., 1989. Genomic organization and sequences of
shark. J. Immunol. 175, 8105–8115. immunoglobulin light chain genes in a primitive vertebrate suggest coevolution of
Malecek, K., Lee, V., Feng, W., Huang, J.L., Flajnik, M.F., Ohta, Y., Hsu, E., 2008. immunoglobulin gene organization. EMBO J. 8, 3733–3739.
Immunoglobulin heavy chain exclusion in the shark. PLoS Biol. 6. Shankey, T.V., Clem, L.W., 1980a. Phylogeny of immunoglobulin structure and
Marchalonis, J., Edelman, G., 1966. Polypeptide chaini of immunoglobulins from the function—VIII: intermolecular heterogeneity of shark 19S IgM antibodies to
smooth dogfish (Mustelus canis). Science 154, 1567–1568. pneumococcal polysaccharide. Mol. Immunol. 17, 365–375.
Matz, H., Dooley, H., 2019. Shark IgNAR-derived binding domains as potential Shankey, T.V., Clem, L.W., 1980b. Phylogeny of immunoglobulin structure and function.
diagnostic and therapeutic agents. Dev. Comp. Immunol. 90, 100–107. IX. Intramolecular heterogeneity of shark 19S IgM antibodies to the dinitrophenyl
Mesin, L., Schiepers, A., Ersching, J., Barbulescu, A., Cavazzoni, C.B., Angelini, A., hapten. J. Immunol. 125, 2690–2698.
Okada, T., Kurosaki, T., Victora, G.D., 2020. Restricted clonality and limited Small, P.A., Klapper, D.G., Clem, L.W., 1970. Half-lives, body distribution and lack of
germinal center reentry characterize memory B cell reactivation by boosting. Cell interconversion of serum 19S and 7S IgM of sharks. J. Immunol. 105, 29–37.
180, 92–106 e111. Smith, L.E., Crouch, K., Cao, W., Müller, M.R., Wu, L., Steven, J., Lee, M., Liang, M.,
Miracle, A.L., Anderson, M.K., Litman, R.T., Walsh, C.J., Luer, C.A., Rothenberg, E.V., Flajnik, M.F., Shih, H.H., 2012. Characterization of the immunoglobulin repertoire
Litman, G.W., 2001. Complex expression patterns of lymphocyte-specific genes of the spiny dogfish (Squalus acanthias). Dev. Comp. Immunol. 36, 665–679.
during the development of cartilaginous fish implicate unique lymphoid tissues in Smith, S.L., Sim, R.B., Flajnik, M.F., 2014. Immunobiology of the Shark. CRC Press.
generating an immune repertoire. Int. Immunol. 13, 567–580. Stanfield, R.L., Dooley, H., Flajnik, M.F., Wilson, I.A., 2004. Crystal structure of a shark
Mußmann, R., Courtet, M., Schwager, J., Du Pasquier, L., 1997. Microsites for single-domain antibody V region in complex with lysozyme. Science 305,
immunoglobulin switch recombination breakpoints from Xenopus to mammals. Eur. 1770–1773.
J. Immunol. 27, 2610–2619. Streltsov, V.A., Carmichael, J.A., Nuttall, S.D., 2005. Structure of a shark IgNAR antibody
Nguyen, V.K., Hamers, R., Wyns, L., Muyldermans, S., 1999. Loss of splice consensus variable domain and modeling of an early-developmental isotype. Protein Sci. 14,
signal is responsible for the removal of the entire C(H)1 domain of the functional 2901–2909.
camel IGG2A heavy-chain antibodies. Mol. Immunol. 36, 515–524. Sverremark, E., Rietz, C., Fernández, C., 2000. κ-deficient mice are non-responders to
Nguyen, V.K., Muyldermans, S., Hamers, R., 1998. The specific variable domain of camel dextran B512: is this unresponsiveness due to specialization of the κ and λ Ig
heavy-chain antibodies is encoded in the germline. J. Mol. Biol. 275, 413–418. repertoires? Int. Immunol. 12, 431–438.
Nuttall, S.D., Krishnan, U.V., Hattarki, M., De Gori, R., Irving, R.A., Hudson, P.J., 2001. Tomonaga, S., Kobayashi, K., Hagiwara, K., Yamaguchi, K., Awaya, K., 1986. Gut-
Isolation of the new antigen receptor from wobbegong sharks, and use as a scaffold associated lymphoid tissue in elasmobranchs. Zool. Sci. 3, 453–458.
for the display of protein loop libraries. Mol. Immunol. 38, 313–326. Townsend, C.L., Laffy, J.M., Wu, Y.-C.B., Silva O’Hare, J., Martin, V., Kipling, D.,
Ohta, Y., Flajnik, M., 2006. IgD, like IgM, is a primordial immunoglobulin class Fraternali, F., Dunn-Walters, D.K., 2016. Significant differences in physicochemical
perpetuated in most jawed vertebrates. Proc. Natl. Acad. Sci. Unit. States Am. 103, properties of human immunoglobulin kappa and lambda CDR3 regions. Front.
10723–10728. Immunol. 7, 388.
Ota, T., Rast, J.P., Litman, G.W., Amemiya, C.T., 2003. Lineage-restricted retention of a Tumang, J.R., Frances, R., Yeo, S.G., Rothstein, T.L., 2005. Spontaneously Ig-secreting B-
primitive immunoglobulin heavy chain isotype within the Dipnoi reveals an 1 cells violate the accepted paradigm for expression of differentiation-associated
evolutionary paradox. Proc. Natl. Acad. Sci. Unit. States Am. 100, 2501–2506. transcription factors. J. Immunol. 174, 3173–3177.
Quinlan, E.M., King, J.J., Amemiya, C.T., Hsu, E., Larijani, M., 2017. Biochemical Venkatesh, B., Lee, A.P., Ravi, V., Maurya, A.K., Lian, M.M., Swann, J.B., Ohta, Y.,
regulatory features of activation-induced cytidine deaminase remain conserved from Flajnik, M.F., Sutoh, Y., Kasahara, M., 2014. Elephant shark genome provides unique
lampreys to humans. Mol. Cell Biol. 37. insights into gnathostome evolution. Nature 505, 174.
Rast, J.P., Amemiya, C.T., Litman, R.T., Strong, S.J., Litman, G.W., 1998. Distinct Voss, E.W., Sigel, M.M., 1972. Valence and temporal change in affinity of purified 7S and
patterns of IgH structure and organization in a divergent lineage of chrondrichthyan 18S nurse shark anti-2, 4 dinitrophenyl antibodies. J. Immunol. 109, 665–673.
fishes. Immunogenetics 47, 234–245. Zapata, A., Amemiya, C., 2000. Phylogeny of Lower Vertebrates and Their
Rast, J.P., Anderson, M.K., Litman, R.T., Margittai, M., Litman, G.W., Ota, T., Immunological Structures, Origin and Evolution of the Vertebrate Immune System.
Shamblott, M.J., 1994. Immunoglobulin light chain class multiplicity and alternative Springer, pp. 67–107.
organizational forms in early vertebrate phylogeny. Immunogenetics 40, 83–99. Zapata, A., Torroba, M., Sacedon, R., Varas, A., Vicente, A., 1996. Structure of the
Richards, M., Nelson, J., 2000. The evolution of vertebrate antigen receptors: a lymphoid organs of elasmobranchs. J. Exp. Zool. Part A: Ecological Genetics and
phylogenetic approach. Mol. Biol. Evol. 17, 146–155. Physiology 275, 125–143.
Roux, K.H., Greenberg, A.S., Greene, L., Strelets, L., Avila, D., McKinney, E.C., Flajnik, M. Zhang, C., Du Pasquier, L., Hsu, E., 2013. Shark IgW C region diversification through
F., 1998. Structural analysis of the nurse shark (new) antigen receptor (NAR): RNA processing and isotype switching. J. Immunol. 191, 3410–3418.
molecular convergence of NAR and unusual mammalian immunoglobulins. Proc. Zhang, W., Qin, L., Cai, X., Juma, S.N., Xu, R., Wei, L., Wu, Y., Cui, X., Chen, G., Liu, L.,
Natl. Acad. Sci. Unit. States Am. 95, 11804–11809. 2020. Sequence structure character of IgNAR Sec in whitespotted bamboo shark
Rumfelt, L., McKinney, E., Taylor, E., Flajnik, M., 2002. The development of primary and (Chiloscyllium plagiosum). Fish Shellfish Immunol. 102, 140–144.
secondary lymphoid tissues in the nurse shark Ginglymostoma cirratum: B-cell zones Zhu, C., Feng, W., Weedon, J., Hua, P., Stefanov, D., Ohta, Y., Flajnik, M.F., Hsu, E.,
precede dendritic cell immigration and T-cell zone formation during ontogeny of the 2011. The multiple shark Ig H chain genes rearrange and hypermutate
spleen. Scand. J. Immunol. 56, 130–148. autonomously. J. Immunol. 187, 2492–2501.
Rumfelt, L.L., Avila, D., Diaz, M., Bartl, S., McKinney, E.C., Flajnik, M.F., 2001. A shark Zhu, C., Lee, V., Finn, A., Senger, K., Zarrin, A.A., Du Pasquier, L., Hsu, E., 2012. Origin
antibody heavy chain encoded by a nonsomatically rearranged VDJ is preferentially of immunoglobulin isotype switching. Curr. Biol. 22, 872–880.
expressed in early development and is convergent with mammalian IgG. Proc. Natl. Zielonka, S., Weber, N., Becker, S., Doerner, A., Christmann, A., Christmann, C., Uth, C.,
Acad. Sci. Unit. States Am. 98, 1775–1780. Fritz, J., Schäfer, E., Steinmann, B., 2014. Shark Attack: high affinity binding
proteins derived from shark vNAR domains by stepwise in vitro affinity maturation.
J. Biotechnol. 191, 236–245.

You might also like