Skip to main content

Update on the human and mouse lipocalin (LCN) gene family, including evidence the mouse Mup cluster is result of an “evolutionary bloom”

Abstract

Lipocalins (LCNs) are members of a family of evolutionarily conserved genes present in all kingdoms of life. There are 19 LCN-like genes in the human genome, and 45 Lcn-like genes in the mouse genome, which include 22 major urinary protein (Mup) genes. The Mup genes, plus 29 of 30 Mup-ps pseudogenes, are all located together on chromosome (Chr) 4; evidence points to an “evolutionary bloom” that resulted in this Mup cluster in mouse, syntenic to the human Chr 9q32 locus at which a single MUPP pseudogene is located. LCNs play important roles in physiological processes by binding and transporting small hydrophobic molecules —such as steroid hormones, odorants, retinoids, and lipids—in plasma and other body fluids. LCNs are extensively used in clinical practice as biochemical markers. LCN-like proteins (18–40 kDa) have the characteristic eight β-strands creating a barrel structure that houses the binding-site; LCNs are synthesized in the liver as well as various secretory tissues. In rodents, MUPs are involved in communication of information in urine-derived scent marks, serving as signatures of individual identity, or as kairomones (to elicit fear behavior). MUPs also participate in regulation of glucose and lipid metabolism via a mechanism not well understood. Although much has been learned about LCNs and MUPs in recent years, more research is necessary to allow better understanding of their physiological functions, as well as their involvement in clinical disorders.

Introduction

Lipocalins (LCNs) are members of a family that includes a diverse group of low-molecular-weight (18–40 kDa) proteins. The larger members of this family undergo cleavage to form the ultimate LCN protein. Comprising usually 150–180 amino-acid residues, these proteins belong to the calycin superfamily and are widely dispersed throughout all kingdoms of life [1]. LCNs are evolutionarily conserved and share an eight-stranded antiparallel β-sheet structure; this forms a “barrel” which is the internal ligand-binding site that interacts with and transports small hydrophobic molecules—such as steroid hormones, odorants (e.g., pheromones), retinoids, and lipids [2, 3].

There are three main structurally conserved regions (SCR1, SCR2, SCR3) that are shared in the lipocalin fold; these represent a moiety composed of three loops that are close to each other in the three-dimensional structure of the β-strands that make up the barrel [4,5,6,7]. Based on the SRCs, two separate groups have been proposed: the kernel LCNs and the outlier LCNs [3]. The kernel LCNs represent a core set of proteins sharing the three characteristic motifs, while the outlier LCNs, which are more divergent family members, typically share only one or two motifs [4]. Based on this categorization—retinoic acid-binding protein-4 (RBP4), α1-microglobulin (A1M), apolipoprotein D (APOD), complement C8 gamma chain (C8G), prostaglandin D2 synthase (PTGDS), and the major urinary proteins (MUPs)—have all been classified as kernel lipocalins, while odorant-binding proteins (OBP2A, OBP2B) and von Ebner’s gland protein (LCN1) are included in the outlier category [4, 7].

Depending on the structure of the individual LCN, the binding-site pocket can accommodate molecules of various sizes and shapes—thus contributing to the diversity of functions within this protein family [5]. Lipocalin crystal structures confirm the highly conserved eight continuously-hydrogen-bonded antiparallel β-strand domains creating the barrel.

The fatty acid-binding protein (FABP) gene family is considered a related, but distinct, subfamily of the calycin superfamily [8] and will not be discussed further here. Another subset of the lipocalins worthy of mention is the immunocalin subfamily. These include α1-acid glycoprotein, α1-microglobulin/bikunin precursor, and glycodelin, each of which exert significant immunomodulatory effects in cell culture [9, 10]; interestingly, all three are encoded by genes in the human Chr 9q32-34 region—together with at least four other lipocalins (neutrophil gelatinase-associated lipocalin, complement factor γ-subunit, tear prealbumin, and prostaglandin D synthase), which also might exert anti-inflammatory and/or antimicrobial activity [11].

Lipocalin family in humans

Among bacteria, plants, fungi, and animals, more than 1000 LCN genes have been identified to date. Nineteen LCN genes, encoding functional LCN proteins, exist in the human genome (Table 1). Figure 1 dendrogram shows the evolutionary relatedness of these human LCN proteins.

Table 1 List of all human LCN and mouse Lcn genes—with official gene symbols, full protein name, aliases, chromosomal locations, isoforms, National Center for Biotechnology Information (NCBI) RefSeq mRNA accession numbers, NCBI RefSeq protein accession numbers, and total number of amino acids (# of AAs) [information retrieved and confirmed from https://www.genenames.org/needs a close bracket]
Fig. 1
figure 1

Dendrogram of lipocalins (LCNs) in the human genome. Although the names listed are the official human gene symbols [https://www.genenames.org/], this dendrogram is based on the alignment of proteins (listed in Table 1), using multiple sequence alignment by CLUSTALW (http://www.genome.jp/tools/clustalw/)

In humans, LCNs are located in blood plasma and other body fluids such as tears and genital secretions, in which they serve as carriers for a variety of small molecules [5]. LCNs also can play important roles in disease such as diabetic retinopathy [12], and, as a result, they are extensively used clinically as biochemical markers. For example, A1M (α1-microglobulin/bikunin precursor, encoded by the AMBP gene) is a biomarker of proteinuria and indicator of declining renal function [13].

Lipocalin-1 (LCN1; human tear pre-albumin, or von Ebner’s gland protein) is one of four major proteins in human tears, acting as a lipid sponge on the ocular surface [14, 15]. LCN1 is produced by lacrimal glands and secreted into tear fluid. Decreased LCN1 levels are associated with Sjögren’s disease, LASIK-induced dry-eye disease [16], and diabetic retinopathy [12].

Lipocalin-2 (LCN2; also known as neutrophil gelatinase-associated lipocalin) mediates various inflammatory processes by suppressing macrophage interleukin-10 (IL10) production [17, 18]. Several studies have shown that LCN2 gene expression in adipose tissue is elevated in insulin-resistant states [19, 20]. LCN2 is also involved in kidney development and used as a biomarker for acute and chronic renal injury [21].

Odorant-binding proteins 2A and 2B (encoded by the OBP2A and OBP2B genes) are members of the LCN family. OBP2A is highly expressed in the oral sphere (e.g., nasal mucus, salivary, and lacrimal glands), whereas OBP2B is expressed in endocrine organs (e.g., mammary gland and prostate) [22]. Functioning as soluble-carrier proteins, OBP2A and OBP2B can bind reversibly to odorants [23].

The AMBP gene encodes α1-microglobulin/bikunin precursor protein; α1-MG (A1M) is the lipocalin—derived from proteolytic cleavage of AMBP [24]. A1M is secreted into plasma, where it can exist free, or bound, to immunoglobulin-A or albumin. Although the molecular weight of A1M is 27.0 kDa, it is freely filtered through the glomerulus and reabsorbed by proximal tubular cells [25]; for this reason, A1M is a biomarker of proteinuria, i.e., increased levels in urine indicate a defect in proximal tubules. A1M is considered to be a major factor for progressive impairment of renal function, as well as for early diagnosis of acute allograft rejection [13, 24, 26]. Recent studies have shown A1M to be expressed in rat retinal explants and to have oxygen radical-scavenging and reductase properties; these findings suggest that A1M might protect against oxidative stress and possibly be involved in the response to retinal detachment [27, 28].

Other members of the LCN family include apolipoproteins D (APOD) and M (APOM)—which interestingly exhibit structural similarities to LCNs rather than to other apolipoproteins. APOD is an atypical apolipoprotein, because it is highly expressed in mammalian tissues such as liver, kidney, and central nervous system. APOD is a component of HDL cholesterol. Recent studies have shown that abnormal APOD expression is associated with altered lipid metabolism; three distinct missense mutations (Phe36Val, Tyr108Cys, and Thr158Lys) in African populations link APOD with metabolic syndrome [29]. A recent study showed that APOM, which resides in the plasma HDL fraction, acts as a chaperone for sphingosine-1-phosphate (S1P) and facilitates interaction between S1P and plasma HDL, thereby exhibiting a vasculoprotective effect [30].

The protein encoded by the complement C8 gamma chain gene (C8G) is one of the three subunits present in complement component 8 (C8). It is an oligomeric protein composed of three non-identical sub-units (α, 64-kDa; β, 64-kDa; γ, 22-kDa); the gamma chain is the only one that belongs to the lipocalin family [31]. C8 is part of the membrane-attack complex (MAC) that participates in irreversible association of the complement proteins C5b, C6, C7, and C9 to form a cytolytic complex that inserts into, and directly lyses, microbes [32]. Activation of complement triggers the assembly of MAC, which is then deployed to kill a wide range of Gram-negative bacteria [33]. Two functionally distinct C8-deficiency states have been identified: the first reflects a lack of the alpha and gamma chains and has been reported in Afro-Caribbean, Hispanic, and Japanese populations; the second results from lack of the beta chain and is found mainly in Caucasians [34, 35]. Deficiency of C8 complement is a very rare primary immunodeficiency associated with invasive and recurrent infections by Neisseria meningitidis [32, 36, 37].

Orosomucoids (ORM1 and ORM2), α1-acid glycoproteins (trivial name AGPs), belong to the subfamily of immunocalins. ORM1 is an acute phase protein secreted by hepatocytes in response to inflammation, with its expression being regulated by pro-inflammatory cytokines such as IL1 and IL6, the chemokine IL8, and glucocorticoids [38]. ORM1 and ORM2 are polymorphic proteins—commonly referred to as ORM/AGP with four variants in humans: AGP F1; AGP F2; AGP S, encoded by the ORM1 gene; and AGP A, encoded by the ORM2 gene [39]. AGPs are important members of the lipocalin family, because their capacity to bind to basic drugs can affect plasma free drug concentrations, playing a key role in a drug’s volume of distribution, metabolism, and therapeutic effect [40]. The ORM1 and ORM2 proteins have been recently identified as predictive urinary biomarkers for rheumatoid arthritis [41]. In addition, they are predictive markers for systemic lupus [42] and chronic inflammation [43].

The progestagen-associated endometrial protein (PAEP) is a secreted immunosuppressive glycoprotein (28 kDa), also termed glycodelin, i.e., one of the immunocalins. Studies have shown that PAEP downregulation can lead to abortion during the first trimester—due to increased activation of the immune system [44, 45]. In addition, PAEP has been found expressed in many tumors (e.g., gynecological malignancies, lung cancer, and melanoma) [46,47,48].

The protein encoded by the prostaglandin D2 synthase (PTGDS) gene is a glutathione-independent prostaglandin synthase (PTGDS). PTGDS is involved in the arachidonic acid cascade, converting prostaglandin H2 to prostaglandin D2 (PGD2), and is preferentially expressed in brain [49]. Increased PTGDS expression has been shown in patients having attention deficit hyperactivity disorder, compared with patients having bipolar disorder [50]. Another study suggests that dysregulated PTGDS mRNA expression is associated with rapid-cycling bipolar depression [49]. Enhanced PTGDS expression has also been associated with various malignancies [51,52,53,54,55].

Plasma retinol-binding protein 4 (RBP4) is a 21-kDa transporter of all-trans-retinol and belongs to the lipocalin family [56, 57]. RBP4 circulates in plasma as a moderately tight 1:1 M complex with vitamin A. RBP4 is secreted mainly by hepatocytes and also by adipose tissue [58]. In humans, increased circulating RBP4 levels have been correlated with obesity [59], insulin resistance, and type-2 diabetes [60, 61]. Insulin resistance has been long considered to play a key role in the development of non-alcoholic fatty liver disease (NAFLD) [62]—which is associated with altered RBP4 levels. Information in the literature on this association, however, is controversial. Several studies have reported significantly increased RBP4 levels in patients with NAFLD [63,64,65,66], whereas other studies have shown no difference on RBP4 levels between control and NAFLD groups [67, 68].

There is limited information in the literature regarding human LCN6, LCN8, LCN9, LCN10, LCN12, or LCN15.

Lipocalin family in mice

Lipocalins have been extensively studied in the mouse. Forty-five proteins belong to this family in mice (Table 1), which also includes major urinary proteins (MUPs) as members of this family (Fig. 2). All of the LCN genes are expressed in both humans and mice (Table 1), with the only exception of LCN1, which is found only in human—whereas Lcn3, Lcn4, Lcn5, Lcn11, Lcn16, Lcn17, and all the functional Mup genes are found in mouse but not human.

Fig. 2
figure 2

Dendrogram of mouse Lcn and Mup proteins. Although the names listed are the official mouse gene symbols [http://www.informatics.jax.org/], this dendrogram is based on the alignment of proteins (listed in Tables 1 and 2), using multiple sequence alignment by CLUSTALW (http://www.genome.jp/tools/clustalw/)

MUPs are intriguing small proteins (19–21 kDa) found in mouse urine; in rats, MUPs are known as α2u-globulins [69,70,71,72]. (For the purpose of this review, MUPs will refer to major urinary proteins in both mouse and rat.) While the presence of proteinuria is considered in humans to be a pathological renal condition, this is not the case for mice or rats [70]. Under physiological conditions, rodents excrete substantial levels of protein in urine, with MUPs accounting for > 90% of total protein content [70, 73, 74], playing a key role in chemo-signaling between animals to coordinate social behavior [75]. MUPs represent highly homologous proteoforms that control the release of volatile pheromones for urinary scent marks by transporting them into the vomeronasal organ (VNO) [76, 77]. MUPs bind pheromones within the hydrophobic calyx of the protein structure where hydrophobic binding sites exist for small lipophilic ligands. The affinity of each MUP for specific ligands varies, according to its subtype [70, 78], and depends on the amino-acid sequence in the binding domain [79, 80]. MUP affinity is most affected by polymorphisms that influence amino acids on the luminal surface of the ligand-binding domain (pocket)—rather than on the protein surface where most sequence differences are observed [73]. In addition, MUPs may act as direct stimulants of pheromone receptors [81].

MUPs are primarily synthesized in post-pubescent mouse liver in response to various hormones such as testosterone, growth hormone, thyroxine, insulin, and glucocorticoids [82, 83]. MUP synthesis is sex-dependent—resulting in (three- to fourfold) higher protein concentrations in post-pubescent males than female mice [84].

MUP expression is stimulated by androgens and leads to higher expression levels in adult males than in females, as well as immature males [70, 85]. Because their expression is stimulated by androgens, MUP synthesis is gender-dependent with higher (three- to fourfold) protein levels occurring in adult males than in females or immature males [70, 85]. For example, in C57/BL6 mice, MUPs represent 3.5–4% of total protein synthesized in male liver, but only 0.6–0.9% in female liver [86]. Mup mRNA is also expressed in a number of secretory tissues—such as nasal tissue, mammary, salivary, submaxillary, and lacrimal glands [87, 88]—as well as skeletal muscle, kidney, brain, spleen, heart, epididymal adipocytes, and brown adipose tissue [89,90,91]. MUP synthesis is initiated in response to different hormonal signals during various developmental stages; for example, liver synthesis of MUPs begins at onset of puberty and on through adulthood [92], whereas MUP synthesis in lacrimal gland starts 1 to 2 weeks before onset of puberty and continues into adulthood [83]. In addition, the specific Mup mRNA subtype produced varies from tissue to tissue [93] (Table 2).

Table 2 Mouse tissues known to express Mup mRNA [93]

The MUP gene cluster in mouse and human genomes

Interestingly, the mouse Mup gene cluster (22 protein-coding genes; Table 3) can be divided into two subgroups. The first group (Mup3, Mup4, Mup5, Mup6, Mup20, and Mup21) is slightly older (Fig. 2) and contains a more divergent class of genes. The second group comprises the remaining 16 Mup genes, which share almost 99% sequence identity [75, 94]. The predicted gene, previously designated Gm21320 (“gene model 21320”), has now been renamed Mup22, cf. [http://www.informatics.jax.org/].

Table 3 List of all mouse Mup genes [http://www.informatics.jax.org/], with official gene symbols, aliases, chromosomal locations, isoforms, National Center for Biotechnology Information (NCBI) RefSeq mRNA accession numbers, NCBI RefSeq protein accession numbers, and total number of amino acids (# of AAs) [information retrieved and confirmed from https://www.ncbi.nlm.nih.gov/genome.]

As members of the LCN family, MUPs exhibit conservation in the common three-dimensional structure of the protein family, i.e., a central area pocket formed by eight hydrophobic β-strand domains that form a barrel (Fig. 3) [81, 95]. This structure enables the MUPs to serve as carrier proteins for small lipophilic molecules such as pheromones and other chemical signals [78, 81]. All 22 mouse Mup protein-coding genes are located in a cluster (the Mup locus) on Chr 4 (Fig. 4 a, b) [96]. There are also 29 Mup-ps pseudogenes in the Chr 4 Mup cluster (intriguingly, the one remaining pseudogene, Mup-ps22, is located on Chr 11).

Fig. 3
figure 3

Structure of prototypical mouse urinary protein. The crystal structure consists of eight β-strands, forming a calyx-shaped barrel (red); this encloses an internal ligand-binding site. There are also an α-helix (green) and four 310-helices (blue); the hydrophobic pocket is located inside the barrel. AB, BC, CD, DE, EF, FG, GH, HI, AND βI denote the amino-acid segments between the β-strands (This diagram taken from Ref. [95])

Fig. 4
figure 4

Chromosomal location of mouse Mup genes and pseudogenes. a The Mup cluster region, located at 60,498,012 Mb to 60,501,960 Mb (red vertical rectangle). Taken from the Ensembl genome browser. b The Chr 4 region (in greater detail)—showing ten of the 22 Mup genes (Gm21320 is Mup22) in the Mup cluster and 12 of the 29 Mup-ps pseudogenes

An “evolutionary bloom” is defined when one sees a recent, phylogenetically independent proliferation of close paralogs or lineage-specific gene family expansion [97]. Examples of this phenomenon have been extensively studied in the large and diverse cytochrome P450 superfamily [97]. For example, the koala’s ability to detoxify eucalyptus leaves appears to be due to an evolutionary bloom within a cytochrome P450 gene group; the koala’s CYP2C subfamily was found to comprise 31 putative protein-coding functional enzymes, compared to 15 Cyp2c genes in mouse and just four CYP2C genes in human [98]. Another example is the mouse Scgb gene superfamily—which includes a number of encoded androgen-binding proteins involved in mate selection [99]; this is fascinating, because the Mup cluster (described herein) also encodes proteins involved in mate selection. It has been suggested that these evolutionary blooms might represent simply a stochastic process [97]. However, it is more likely these blooms are the result of environmental pressures needed for the organism to survive (i.e., find food, avoid predators, and reproduce) at a particular moment in evolutionary time.

Mup gene polymorphisms in rat and mouse have shown significant differences. Yet, such differences have not been seen in genomes of other mammalian species for which whole-genome sequences have been explored [75]. Although the amino acid sequence of MUP homologs between rat and mouse is ~ 65%, there is a characteristic six amino-acid consensus sequence (Glu-Glu-Ala-Ser-Ser-Thr) that remains highly conserved between these two species [100]. In general, species differences in MUP proteins appear to be mainly due to glycosylated MUP amino-acid residues that occur in rats, but not mice [100]. Most other mammalian species (e.g., dog, baboon, gorilla, and chimpanzee) have only one functional protein-coding MUP gene, except for horse that has three functional MUP genes [75].

The human MUP-related gene is a pseudogene (MUPP, located at Chr 9q32). Using the UCSC genome browser [https://genome.ucsc.edu/], one can visualize that human Chr 9q32 is syntenic to mouse Chr 4 at 60,498,012 Mb to 60,501,960 Mb, where the Mup cluster of 22 Mup genes is located; in fact, the human ZFP37 and mouse Zfp37 gene flank the “MUP region” in both human and mouse, respectively. The human MUPP locus exhibits a high degree of sequence similarity to mouse Mup functional genes but contains coding-sequence disruptions that prevent the gene product from being formed [101]. The human MUPP shows a G > A transition (relative to the chimpanzee MUP sequence) that disrupts a splice-donor site [75]; this is interesting because this G > A mutation has not been observed in mammals other than humans [101]. The human MUPP pseudogene sequence is most similar to the mouse Mup-ps4 pseudogene [75].

One of the main functions of MUP proteins is to promote aggressive behavior through binding to vomeronasal pheromone receptors (V2Rs) in the accessory olfactory neural pathway. Even though there is a co-expansion of MUPs and V2Rs in mouse, rat, and opossum—all human V2R receptors have become inactive, possibly leading to the pseudogenization of the single human MUP gene [102, 103]. In other words, the absence of the specific V2R removed the selection pressure for a functional MUP ligand.

Parallel expansions of Mup clusters

The last common ancestor of rat and mouse had either a single, or a small number of, Mup genes [75]. By determining the extent of Mup gene expansions across non-rodent lineages, Logan and colleagues were able to identify orthologs of the Slc46a2 and Zfp37 genes (and the contiguous genomic sequence spanning the interval between these two genes) in nine additional placental mammals [75]. Whereas C57BL/6J mice have a cluster of 22 distinct Mup genes on Chr 4 and rats have nine distinct Mup genes, mammalian species such as dog, pig, baboon, chimpanzee, bush baby, and orangutan—each has a single Mup gene (with no evidence of additional pseudogenes). By contrast, the human genome has only the one pseudogene.

A neighbor-joining dendrogram of human LCN and mouse MUP proteins is illustrated in Fig. 5; subfamilies can be distinguished based on evolutionary divergence. Note that all mouse MUPs are clustered into a subgroup near the top of the dendrogram, whereas the human LCNs are split into several different branches—due to the high degree of divergence of LCN proteins. The mouse Mup cluster divergence is most closely associated with human LCN9 and PAEP (Fig. 5). Note that the evolutionarily oldest human LCN genes include ORM1, ORM2, APOM, APOD, RBP4, and LCN8.

Fig. 5
figure 5

Dendrogram of human LCNs and mouse MUPs, combined. Although the names listed are the official human gene symbols and mouse Mup gene symbols, this dendrogram was based on the alignment of proteins (listed in Tables 1 and 3) using multiple sequence alignment by CLUSTALW (http://www.genome.jp/tools/clustalw/). Note that the human LCN9 gene is evolutionarily closest to the mouse Mup cluster in this dendrogram

Functions of MUPs in mice

MUPs and chemical communication

Due to their influence on pheromones, MUPs appear to be involved in regulating transmission of social signals—such as identity, territorial marking, and mate choice [104,105,106]. Most pheromones are small volatile molecules that influence aggression, mating, feeding, and territorial behavior within the same species [103, 107]. Mice use pheromones as cues to regulate social behaviors. Neurons that detect pheromones reside in at least two separate organs within the nasal cavity: the vomeronasal organ (VNO) and the main olfactory epithelium (MOE). Each pheromone molecule is thought to activate a dedicated subset of these sensory neurons—similar to the manner in which odorants are received by dedicated subsets of mammalian olfactory receptors. However, the identity of the responding neurons that regulate specific social behaviors remains largely unknown.

Pheromones have a short half-life, which can be prolonged by binding to the characteristic barrel pocket, in the MUP protein. In addition, gradual release of a pheromone from a MUP protein allows the half-life of these airborne odor signals to be extended, e.g., to be used as mammalian scent marks [108, 109].

MUPs are also linked to reproductive success in males [110] and to social behavior, by adjusting the animal’s odor profile in response to different stimuli. This function underscores how social environment plays an important role in MUP production in both male and female mice. For example, MUP synthesis is upregulated in a male mouse housed with a female, but downregulated when a male mouse is housed with other males only [111]. Perhaps related to this, MUPs can be predictive of the onset of aggressive and dispersal behavior among male mice [103].

In addition to serving as pheromone carriers, MUPs can function as pheromones themselves. They facilitate chemical information exchange to convey specific information (e.g., gender, social and reproductive status) between animals [70]. Recent research has revealed that MUPs also act as kairomones, causing a fear reaction in response to predators [112]. For example, the rat kairomone that triggers defensive behavior in mice is encoded by the Mup13 gene [112].

Members of the MUP family are known to be involved in intraspecies interplay, especially male-male aggression in mice [103]. Female mice are attracted to urine-borne male pheromones. MUP20, for example, has been shown to be rewarding and attractive to female mice. MUPs excreted by male mice can also influence reproductive behavior and promote female attraction [103, 113,114,115]. The molecular mechanism promoting spontaneous ovulation involves direct stimulation of VNO nerves by four residues on the NH2-terminus of MUP proteins [116]. MUPs may also be involved in mediating individual recognition and inbreeding avoidance [117, 118].

MUPs and metabolism

MUPs also appear to be involved in energy metabolism—actions reminiscent of the lipocalins that have been implicated clinically in lipid disorders and metabolic syndrome caused by obesity and type-2 diabetes [119, 120]. For example, mouse MUP1 regulates systemic glucose metabolism by modulating the hepatic gluconeogenic and/or lipogenic programs [121,122,123]. Caloric restriction dramatically reduces MUP1 expression in mouse liver [124, 125] and appears to decrease MUP4 and MUP5 expression, as well [124, 126].

Decreased hepatic MUP1 levels have been linked to obesity and type-2 diabetes in mice with either genetic (leptin receptor-deficient db/db) or dietary fat-induced obesity [121, 127]. Similar decreases in MUP1 are found in extrahepatic organs—such as adipose tissue and the hypothalamus—after caloric restriction [128, 129]. Furthermore, MUP1 was also found to lower blood glucose levels by inhibiting expression of phosphoenolpyruvate carboxykinase and glucose-6-phosphatase, two rate-limiting enzymes for gluconeogenesis [127]. These studies suggest that mouse MUP1, and possibly other MUP family members, are playing key roles in energy metabolism and potentially contributing to the development of metabolic diseases such as type-2 diabetes.

Human LCN-like genes and their mouse orthologs

We have listed the human LCN-related genes in Table 1 and mouse Lcn-like genes plus the Mup cluster genes in Table 3. What are the percent similarities of proteins—if one compares human LCN-like genes with their mouse orthologs? Among the 19 LCN-like genes in the human genome (Table 4), 17 have mouse orthologs, whereas human LCN1 and PAEP do not. Within the LCN cluster, LCN6 and LCN8 exhibit the highest percent similarity: 74 and 70%, respectively. Among all 19 LCN-like genes, RBP4 and APOM reveal the highest percent similarity (86% and 81%, respectively); LCN15 and OBP2A display the lowest percent similarity (39%) to their mouse orthologs.

Table 4 List of the 19 LCN-like genes in the human genome and their similarity to their mouse orthologs, expressed as percent identity (%)

Human and mouse ancestors are estimated to have diverged from one another ~ 80 million years ago. Table 4 confirms the relatively rapid rate of evolutionary divergence by the LCN-like genes, which is consistent with their function of requiring evolutionarily quick adaptation to changing environments; this is similar to, e.g., beta-defensin (DEFB) genes, which number almost four dozen in the human genome and encode broad-spectrum antimicrobial cationic peptides [130]. Out of 19 LCN-like genes, the appearance of two novel human genes (LCN1 and PAEP) during the past ~ 80 million years is further evidence of an enhanced evolutionary rate for this gene superfamily.

Rapid rates of evolutionary divergence stand in sharp contrast to, e.g., highly conserved transcription factors, whose human-mouse orthologs are generally > 95% similar in protein sequence. In fact, assaying for complementation of lethal growth defects in yeast, almost half (47%) of the yeast genes could be successfully humanized [131], and the yeast-human divergence occurred well over one billion years ago.

Conclusions

Lipocalins (LCNs) are members of a family of evolutionarily conserved small proteins that possess a binding pocket. The LCN proteins (18–40 kDa) are encoded by 19 human LCN-related genes and 45 mouse Lcn-related genes. LCN proteins are expressed in numerous tissues and play important roles in physiological processes by transporting molecules in plasma and other body fluids. In humans, LCNs are extensively used clinically as biochemical markers in various diseases—such as diabetic renal disease, systemic lupus erythematosus, and chronic inflammation.

In mice, major urinary proteins (MUPs) are also members of the lipocalin family. The Mup cluster of 22 functional protein-coding Mup genes (plus 29 of 30 Mup-ps pseudogenes) is confined to mouse Chr 4 and represents an “evolutionary bloom,” because only one or a few MUP genes are functional in other mammals. In fact, no functional MUP gene exists in the human genome—although a human MUPP pseudogene located at Chr 9q32 is syntenic to the Mup cluster on Chr 4.

The MUP protein structure contains a conserved “barrel” formed by the eight β-chains having the characteristic central hydrophobic pocket binding-site. Mouse MUP proteins are expressed mainly in the liver, secreted into the bloodstream, and excreted by the kidney. MUPs are involved in the communication of information in urine-derived scent marks and can also serve as pheromones themselves. Circulating MUPs may also contribute to regulation of nutrient metabolism—possibly by suppressing hepatic gluconeogenic and lipid metabolism. However, it still remains unclear how MUPs, especially mouse MUP1, regulate energy metabolism and the gluconeogenic pathway. Further studies will be needed to shed light on these mechanisms.

References

  1. Akerstrom B, Flower DR, Salier JP. Lipocalins: unity in diversity. Biochim Biophys Acta. 2000;1482(1–2):1–8.

    Article  CAS  PubMed  Google Scholar 

  2. di Masi A, et al. Human plasma lipocalins and serum albumin: plasma alternative carriers? J Control Release. 2016;228:191–205.

    Article  PubMed  CAS  Google Scholar 

  3. Flower DR, North AC, Attwood TK. Structure and sequence relationships in the lipocalins and related proteins. Protein Sci. 1993;2(5):753–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Flower DR. The lipocalin protein family: structure and function. Biochem J. 1996;318(Pt 1):1–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Schiefner A, Skerra A. The menagerie of human lipocalins: a natural protein scaffold for molecular recognition of physiological compounds. Acc Chem Res. 2015;48(4):976–85.

    Article  CAS  PubMed  Google Scholar 

  6. Bocskei Z, et al. Pheromone binding to two rodent urinary proteins revealed by X-ray crystallography. Nature. 1992;360(6400):186–8.

    Article  CAS  PubMed  Google Scholar 

  7. Ganfornina MD, et al. A phylogenetic analysis of the lipocalin protein family. Mol Biol Evol. 2000;17(1):114–26.

    Article  CAS  PubMed  Google Scholar 

  8. Sanchez D, Ganfornina Álvarez MD, Gutierrez G, Gauthier-Jauneau AC, Risler JL, Salier JP. In: Akerstrom B, editor. Lipocalin genes and their evolutionary history, in (Molecular Biology Intelligence Unit: Lipocalins): Landes Bioscience, Inc; 2006. p. 1–12.

  9. Akerstrom B, et al. alpha(1)-Microglobulin: a yellow-brown lipocalin. Biochim Biophys Acta. 2000;1482(1–2):172–84.

    Article  CAS  PubMed  Google Scholar 

  10. Alok A, Mukhopadhyay D, Karande AA. Glycodelin A, an immunomodulatory protein in the endometrium, inhibits proliferation and induces apoptosis in monocytic cells. Int J Biochem Cell Biol. 2009;41(5):1138–47.

    Article  CAS  PubMed  Google Scholar 

  11. Logdberg L, Wester L. Immunocalins: a lipocalin subfamily that modulates immune and inflammatory responses. Biochim Biophys Acta. 2000;1482(1–2):284–97.

    Article  CAS  PubMed  Google Scholar 

  12. Wang JC, et al. Detection of low-abundance biomarker lipocalin 1 for diabetic retinopathy using optoelectrokinetic bead-based immunosensing. Biosens Bioelectron. 2017;89(Pt 2):701–9.

    Article  CAS  PubMed  Google Scholar 

  13. Bazzi C, et al. Urinary excretion of IgG and alpha(1)-microglobulin predicts clinical course better than extent of proteinuria in membranous nephropathy. Am J Kidney Dis. 2001;38(2):240–8.

    Article  CAS  PubMed  Google Scholar 

  14. Karnati R, Laurie DE, Laurie GW. Lacritin and the tear proteome as natural replacement therapy for dry eye. Exp Eye Res. 2013;117:39–52.

    Article  CAS  PubMed  Google Scholar 

  15. Srinivasan S, et al. iTRAQ quantitative proteomics in the analysis of tears in dry eye patients. Invest Ophthalmol Vis Sci. 2012;53(8):5052–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Glasgow BJ, Gasymov OK. Focus on molecules: tear lipocalin. Exp Eye Res. 2011;92(4):242–3.

    Article  CAS  PubMed  Google Scholar 

  17. Moschen AR, et al. Lipocalin-2: a master mediator of intestinal and metabolic inflammation. Trends Endocrinol Metab. 2017;28(5):388–97.

    Article  CAS  PubMed  Google Scholar 

  18. Nairz M, et al. Lipocalin-2 ensures host defense against Salmonella typhimurium by controlling macrophage iron homeostasis and immune response. Eur J Immunol. 2015;45(11):3073–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Li L, et al. Serum retinol-binding protein 4 is associated with insulin secretion in Chinese people with normal glucose tolerance. J Diabetes. 2009;1(2):125–30.

    Article  CAS  PubMed  Google Scholar 

  20. Singh RG, et al. Role of human lipocalin proteins in abdominal obesity after acute pancreatitis. Peptides. 2017;91:1–7.

    Article  CAS  PubMed  Google Scholar 

  21. Wang E, et al. Overexpression of exogenous kidney-specific Ngal attenuates progressive cyst development and prolongs lifespan in a murine model of polycystic kidney disease. Kidney Int. 2017;91(2):412–22.

    Article  CAS  PubMed  Google Scholar 

  22. Lacazette E, Gachon AM. Pitiot G. A novel human odorant-binding protein gene family resulting from genomic duplicons at 9q34: differential expression in the oral and genital spheres. Hum Mol Genet. 2000;9(2):289–301.

    Article  CAS  PubMed  Google Scholar 

  23. Tegoni M, et al. Mammalian odorant binding proteins. Biochim Biophys Acta. 2000;1482(1–2):229–40.

    Article  CAS  PubMed  Google Scholar 

  24. Stubendorff B, et al. Urine protein profiling identified alpha-1-microglobulin and haptoglobin as biomarkers for early diagnosis of acute allograft rejection following kidney transplantation. World J Urol. 2014;32(6):1619–24.

    Article  CAS  PubMed  Google Scholar 

  25. Hong CY, et al. Urinary alpha1-microglobulin as a marker of nephropathy in type-2 diabetic Asian subjects in Singapore. Diabetes Care. 2003;26(2):338–42.

    Article  CAS  PubMed  Google Scholar 

  26. Amer H, et al. Urine high and low molecular weight proteins one-year post-kidney transplant: relationship to histology and graft survival. Am J Transplant. 2013;13(3):676–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Cederlund M, et al. Vitreous levels of oxidative stress biomarkers and the radical-scavenger alpha1-microglobulin/A1M in human rhegmatogenous retinal detachment. Graefes Arch Clin Exp Ophthalmol. 2013;251(3):725–32.

    Article  CAS  PubMed  Google Scholar 

  28. Akerstrom B, et al. The role of mitochondria, oxidative stress, and the radical-binding protein A1M in cultured porcine retina. Curr Eye Res. 2017;42(6):948–61.

    Article  PubMed  CAS  Google Scholar 

  29. Desai PP, et al. Genetic variation in the apolipoprotein D gene among African blacks and its significance in lipid metabolism. Atherosclerosis. 2002;163(2):329–38.

    Article  CAS  PubMed  Google Scholar 

  30. Christoffersen C, et al. Endothelium-protective sphingosine-1-phosphate provided by HDL-associated apolipoprotein M. Proc Natl Acad Sci U S A. 2011;108(23):9613–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Chiswell B, et al. Structural features of the ligand binding site on human complement protein C8gamma: a member of the lipocalin family. Biochim Biophys Acta. 2007;1774(5):637–44.

    Article  CAS  PubMed  Google Scholar 

  32. Serna M, et al. Structural basis of complement membrane attack complex formation. Nat Commun. 2016;7:10587.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Figueroa JE, Densen P. Infectious diseases associated with complement deficiencies. Clin Microbiol Rev. 1991;4(3):359–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Kotnik V, et al. Molecular, genetic, and functional analysis of homozygous C8 beta-chain deficiency in two siblings. Immunopharmacology. 1997;38(1–2):215–21.

    Article  CAS  PubMed  Google Scholar 

  35. Ross SC, Densen P. Complement deficiency states and infection: epidemiology, pathogenesis and consequences of neisserial and other infections in an immune deficiency. Medicine (Baltimore). 1984;63(5):243–73.

    Article  CAS  PubMed  Google Scholar 

  36. Arnold DF, et al. A novel mutation in a patient with a deficiency of the eighth component of complement associated with recurrent meningococcal meningitis. J Clin Immunol. 2009;29(5):691–5.

    Article  CAS  PubMed  Google Scholar 

  37. Dellepiane RM, et al. Invasive meningococcal disease in three siblings with hereditary deficiency of the 8(th) component of complement: evidence for the importance of an early diagnosis. Orphanet J Rare Dis. 2016;11(1):64.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Ceciliani F, Pocacqua V. The acute phase protein alpha1-acid glycoprotein: a model for altered glycosylation during diseases. Curr Protein Pept Sci. 2007;8(1):91–108.

    Article  CAS  PubMed  Google Scholar 

  39. Nishi K, et al. Structural insights into differences in drug-binding selectivity between two forms of human alpha1-acid glycoprotein genetic variants, the A and F1*S forms. J Biol Chem. 2011;286(16):14427–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Ohbatake Y, et al. Elevated alpha1-acid glycoprotein in gastric cancer patients inhibits the anticancer effects of paclitaxel, effects restored by co-administration of erythromycin. Clin Exp Med. 2016;16(4):585–92.

    Article  CAS  PubMed  Google Scholar 

  41. Gomes MB, Nogueira VG. Acute-phase proteins and microalbuminuria among patients with type-2 diabetes. Diabetes Res Clin Pract. 2004;66(1):31–9.

    Article  CAS  PubMed  Google Scholar 

  42. Watson L, et al. Urinary monocyte chemoattractant protein 1 and alpha 1 acid glycoprotein as biomarkers of renal disease activity in juvenile-onset systemic lupus erythematosus. Lupus. 2012;21(5):496–501.

    Article  CAS  PubMed  Google Scholar 

  43. Singh R, et al. Urinary biomarkers as indicator of chronic inflammation and endothelial dysfunction in obese adolescents. BMC Obes. 2017;4:11.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Toth B, et al. Glycodelin protein and mRNA is downregulated in human first trimester abortion and partially upregulated in mole pregnancy. J Histochem Cytochem. 2008;56(5):477–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Xu S, Venge P. Lipocalins as biochemical markers of disease. Biochim Biophys Acta. 2000;1482(1–2):298–307.

    Article  CAS  PubMed  Google Scholar 

  46. Ren S, et al. Functional characterization of the progestagen-associated endometrial protein gene in human melanoma. J Cell Mol Med. 2010;14(6b):1432–42.

    Article  CAS  PubMed  Google Scholar 

  47. Scholz C, et al. Glycodelin A is a prognostic marker to predict poor outcome in advanced stage ovarian cancer patients. BMC Res Notes. 2012;5:551.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Schneider MA, et al. Glycodelin: a new biomarker with immunomodulatory functions in non-mall cell lung cancer. Clin Cancer Res. 2015;21(15):3529–40.

    Article  CAS  PubMed  Google Scholar 

  49. Munkholm K, et al. Reduced mRNA expression of PTGDS in peripheral blood mononuclear cells of rapid-cycling bipolar disorder patients compared with healthy control subjects. Int J Neuropsychopharmacol. 2014;18(5):1–9.

    Google Scholar 

  50. Marin-Mendez JJ, et al. Differential expression of prostaglandin D2 synthase (PTGDS) in patients with attention deficit-hyperactivity disorder and bipolar disorder. J Affect Disord. 2012;138(3):479–84.

    Article  CAS  PubMed  Google Scholar 

  51. Kim GE, et al. Differentially expressed genes in matched normal, cancer, and lymph node metastases predict clinical outcomes in patients with breast cancer. Appl Immunohistochem Mol Morphol. 2018.

  52. Zhang B, et al. PGD2/PTGDR2 signaling restricts the self-renewal and tumorigenesis of gastric cancer. Stem Cells. 2018.

  53. Nault JC, et al. Argininosuccinate synthase 1 and periportal gene expression in sonic hedgehog hepatocellular adenomas. Hepatology. 2018;68(3):964–76.

    Article  CAS  PubMed  Google Scholar 

  54. Davalieva K, et al. Comparative proteomics analysis of urine reveals down-regulation of acute phase response signaling and LXR/RXR activation pathways in prostate cancer. Proteomes. 2017;6(1):1–25.

    Article  PubMed Central  Google Scholar 

  55. Omori K, et al. Lipocalin-type prostaglandin D synthase-derived PGD2 attenuates malignant properties of tumor endothelial cells. J Pathol. 2018;244(1):84–96.

    Article  CAS  PubMed  Google Scholar 

  56. Zhou Z, et al. Circulating retinol binding protein 4 levels in nonalcoholic fatty liver disease: a systematic review and meta-analysis. Lipids Health Dis. 2017;16(1):180.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Christou GA, Tselepis AD, Kiortsis DN. The metabolic role of retinol binding protein 4: an update. Horm Metab Res. 2012;44(1):6–14.

    Article  CAS  PubMed  Google Scholar 

  58. Newcomer ME, Ong DE. Plasma retinol binding protein: structure and function of the prototypic lipocalin. Biochim Biophys Acta. 2000;1482(1–2):57–64.

    Article  CAS  PubMed  Google Scholar 

  59. Codoner-Franch P, et al. Association of RBP4 genetic variants with childhood obesity and cardiovascular risk factors. Pediatr Diabetes. 2016;17(8):576–83.

    Article  CAS  PubMed  Google Scholar 

  60. Yang Q, et al. Serum retinol binding protein 4 contributes to insulin resistance in obesity and type-2 diabetes. Nature. 2005;436(7049):356–62.

    Article  CAS  PubMed  Google Scholar 

  61. Graham TE, et al. Retinol-binding protein 4 and insulin resistance in lean, obese, and diabetic subjects. N Engl J Med. 2006;354(24):2552–63.

    Article  CAS  PubMed  Google Scholar 

  62. Birkenfeld AL, Shulman GI. Nonalcoholic fatty liver disease, hepatic insulin resistance, and type-2 diabetes. Hepatology. 2014;59(2):713–23.

    Article  PubMed  Google Scholar 

  63. Seo JA, et al. Serum retinol-binding protein 4 levels are elevated in non-alcoholic fatty liver disease. Clin Endocrinol. 2008;68(4):555–60.

    Article  CAS  Google Scholar 

  64. Chen X, et al. Retinol binding protein-4 levels and non-alcoholic fatty liver disease: a community-based cross-sectional study. Sci Rep. 2017;7:45100.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Terra X, et al. Retinol binding protein-4 circulating levels were higher in nonalcoholic fatty liver disease vs. histologically normal liver from morbidly obese women. Obesity (Silver Spring). 2013;21(1):170–7.

    Article  CAS  Google Scholar 

  66. Wu H, et al. Serum retinol binding protein 4 and nonalcoholic fatty liver disease in patients with type-2 diabetes mellitus. Diabetes Res Clin Pract. 2008;79(2):185–90.

    Article  CAS  PubMed  Google Scholar 

  67. Cengiz C, et al. Serum retinol-binding protein 4 in patients with nonalcoholic fatty liver disease: does it have a significant impact on pathogenesis? Eur J Gastroenterol Hepatol. 2010;22(7):813–9.

    Article  CAS  PubMed  Google Scholar 

  68. Milner KL, et al. Adipocyte fatty acid binding protein levels relate to inflammation and fibrosis in nonalcoholic fatty liver disease. Hepatology. 2009;49(6):1926–34.

    Article  CAS  PubMed  Google Scholar 

  69. Beynon RJ, Hurst JL. Multiple roles of major urinary proteins in the house mouse, Mus domesticus. Biochem Soc Trans. 2003;31(Pt 1):142–6.

    Article  CAS  PubMed  Google Scholar 

  70. Gomez-Baena G, et al. The major urinary protein system in the rat. Biochem Soc Trans. 2014;42(4):886–92.

    Article  PubMed  CAS  Google Scholar 

  71. Mudge JM, et al. Dynamic instability of the major urinary protein gene family revealed by genomic and phenotypic comparisons between C57 and 129 strain mice. Genome Biol. 2008;9(5):R91.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  72. Thom MD, Stockley P, Jury F, Ollier WE, Beynon RJ, Hurst JL. The direct assessment of genetic heterozygosity through scent in the mouse. Curr Biol. 2008;(18)8:619–623.

  73. Beynon RJ, et al. Polymorphism in major urinary proteins: molecular heterogeneity in a wild mouse population. J Chem Ecol. 2002;28(7):1429–46.

    Article  CAS  PubMed  Google Scholar 

  74. Krop EJ, et al. Recombinant major urinary proteins of the mouse in specific IgE and IgG testing. Int Arch Allergy Immunol. 2007;144(4):296–304.

    Article  CAS  PubMed  Google Scholar 

  75. Logan DW, Marton TF, Stowers L. Species specificity in major urinary proteins by parallel evolution. PLoS One. 2008;3(9):e3280.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  76. Yang H, et al. Mup-knockout mice generated through CRISPR/Cas9-mediated deletion for use in urinary protein analysis. Acta Biochim Biophys Sin (Shanghai). 2016;48(5):468–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Enk VM, et al. Regulation of highly homologous major urinary proteins in house mice quantified with label-free proteomic methods. Mol Biosyst. 2016;12(10):3005–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Rajkumar R, et al. Primary structural documentation of the major urinary protein of the Indian commensal rat (Rattus rattus) using a proteomic platform. Protein Pept Lett. 2010;17(4):449–57.

    Article  CAS  PubMed  Google Scholar 

  79. Sharrow SD, et al. Pheromone binding by polymorphic mouse major urinary proteins. Protein Sci. 2002;11(9):2247–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Armstrong SD, et al. Structural and functional differences in isoforms of mouse major urinary proteins: a male-specific protein that preferentially binds a male pheromone. Biochem J. 2005;391(Pt 2):343–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Timm DE, et al. Structural basis of pheromone binding to mouse major urinary protein (MUP-I). Protein Sci. 2001;10(5):997–1004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Hastie ND, Held WA, Toole JJ. Multiple genes coding for the androgen-regulated major urinary proteins of the mouse. Cell. 1979;17(2):449–57.

    Article  CAS  PubMed  Google Scholar 

  83. Shaw PH, Held WA, Hastie ND. The gene family for major urinary proteins: expression in several secretory tissues of the mouse. Cell. 1983;32(3):755–61.

    Article  CAS  PubMed  Google Scholar 

  84. Beynon RJ, Hurst JL. Urinary proteins and the modulation of chemical scents in mice and rats. Peptides. 2004;25(9):1553–63.

    Article  CAS  PubMed  Google Scholar 

  85. Kaur AW, et al. Murine pheromone proteins constitute a context-dependent combinatorial code governing multiple social behaviors. Cell. 2014;157(3):676–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Berger FG, Szoka P. Biosynthesis of the major urinary proteins in mouse liver: a biochemical genetic study. Biochem Genet. 1981;19(11–12):1261–73.

    Article  CAS  PubMed  Google Scholar 

  87. Utsumi M, Ohno K, Kawasaki Y, Tamura M, Kubo T, Tohyama M. Expression of major urinary protein genes in the nasal glands associated with general olfaction. J Neurobiol. 1999;39(2):227–36.

    Article  CAS  PubMed  Google Scholar 

  88. Guo J, Zhou A, Moss RL. Urine and urine-derived compounds induce c-fos mRNA expression in accessory olfactory bulb. Neuroreport. 1997;8(7):1679–83.

    Article  CAS  PubMed  Google Scholar 

  89. Stopkova R, et al. Species-specific expression of major urinary proteins in the house mice (Mus musculus musculus and Mus musculus domesticus). J Chem Ecol. 2007;33(4):861–9.

    Article  CAS  PubMed  Google Scholar 

  90. Hui X, et al. Major urinary protein-1 increases energy expenditure and improves glucose intolerance through enhancing mitochondrial function in skeletal muscle of diabetic mice. J Biol Chem. 2009;284(21):14050–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Stopková R, et al. Mouse lipocalins (MUP, OBP, LCN) are co-expressed in tissues involved in chemical communication. Front Ecol Evol. 2016;4(47):1–11.

    Google Scholar 

  92. Derman E. Isolation of a cDNA clone for mouse urinary proteins: age- and sex-related expression of mouse urinary protein genes is transcriptionally controlled. Proc Natl Acad Sci U S A. 1981;78(9):5425–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Shahan K, Denaro M, et al. Expression of six mouse major urinary protein genes in the mammary, parotid, sublingual, submaxillary, and lachrymal glands and in the liver. Mol Cell Biol. 1987;7(5):1947–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Stopka P, et al. On the saliva proteome of the Eastern European house mouse (Mus musculus musculus) focusing on sexual signalling and immunity. Sci Rep. 2016;6:32481.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Kuser PR, et al. The X-ray structure of a recombinant major urinary protein at 1.75 A resolution. A comparative study of X-ray and NMR-derived structures. Acta Crystallogr D Biol Crystallogr. 2001;57(Pt 12):1863–9.

    Article  CAS  PubMed  Google Scholar 

  96. 2018. Available from: http://useast.ensembl.org/Mus_musculus/Location/View?db=core;g=ENSMUSG00000078683;r=4:60498012-60501960 . Accessed 13 Oct 2018.

  97. Feyereisen R. Arthropod CYPomes illustrate the tempo and mode in P450 evolution. Biochim Biophys Acta. 2011;1814(1):19–28.

    Article  CAS  PubMed  Google Scholar 

  98. Johnson RN, et al. Adaptation and conservation insights from the koala genome. Nat Genet. 2018;50(8):1102–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Jackson BC, et al. Update of the human secretoglobin (SCGB) gene superfamily and an example of ‘evolutionary bloom’ of androgen-binding protein genes within the mouse Scgb gene superfamily. Hum Genomics. 2011;5(6):691–702.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Cavaggioni A, Mucignat-Caretta C. Major urinary proteins, alpha(2U)-globulins and aphrodisin. Biochim Biophys Acta. 2000;1482(1–2):218–28.

    Article  CAS  PubMed  Google Scholar 

  101. Zhang ZD, et al. Identification and analysis of unitary pseudogenes: historic and contemporary gene losses in humans and other primates. Genome Biol. 2010;11(3):R26.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  102. Young JM, Trask BJ. V2R gene families degenerated in primates, dog and cow, but expanded in opossum. Trends Genet. 2007;23(5):212–5.

    Article  CAS  PubMed  Google Scholar 

  103. Chamero P, et al. Identification of protein pheromones that promote aggressive behaviour. Nature. 2007;450(7171):899–902.

    Article  CAS  PubMed  Google Scholar 

  104. Nelson AC, et al. Protein pheromone expression levels predict and respond to the formation of social dominance networks. J Evol Biol. 2015;28(6):1213–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Hurst JL, Beynon RJ. Scent wars: the chemobiology of competitive signalling in mice. Bioessays. 2004;26(12):1288–98.

    Article  CAS  PubMed  Google Scholar 

  106. Hurst JL, et al. Individual recognition in mice mediated by major urinary proteins. Nature. 2001;414(6864):631–4.

    Article  CAS  PubMed  Google Scholar 

  107. Deisig N, et al. Responses to pheromones in a complex odor world: sensory processing and behavior. Insects. 2014;5(2):399–422.

    Article  PubMed  PubMed Central  Google Scholar 

  108. Sengupta S, Smith DP. How drosophila detect volatile pheromones: signaling, circuits, and behavior. In: Mucignat-Caretta C, editor. Neurobiology of Chemical Communication. Boca Raton (FL): CRC Press/Taylor & Francis; 2014. Chapter 7.

    Google Scholar 

  109. Hurst JL, et al. Proteins in urine scent marks of male house mice extend the longevity of olfactory signals. Anim Behav. 1998;55(5):1289–97.

    Article  CAS  PubMed  Google Scholar 

  110. Thonhauser KE, et al. Scent marking increases male reproductive success in wild house mice. Anim Behav. 2013;86(5):1013–21.

    Article  PubMed  PubMed Central  Google Scholar 

  111. Janotova K, Stopka P. The level of major urinary proteins is socially regulated in wild Mus musculus musculus. J Chem Ecol. 2011;37(6):647–56.

    Article  CAS  PubMed  Google Scholar 

  112. Papes F, Logan DW, Stowers L. The vomeronasal organ mediates interspecies defensive behaviors through detection of protein pheromone homologs. Cell. 2010;141(4):692–703.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Roberts SA, et al. Darcin: a male pheromone that stimulates female memory and sexual attraction to an individual male's odour. BMC Biol. 2010;8:75.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  114. Roberts SA, et al. Pheromonal induction of spatial learning in mice. Science. 2012;338(6113):1462–5.

    Article  CAS  PubMed  Google Scholar 

  115. Kimoto H, Haga S, Sato K, Touhara K. Sex-specific peptides from exocrine glands stimulate mouse vomeronasal sensory neurons. Nature. 2005;437(7060):898-901.

  116. More L. Mouse major urinary proteins trigger ovulation via the vomeronasal organ. Chem Senses. 2006;31(5):393–401.

    Article  CAS  PubMed  Google Scholar 

  117. Cheetham SA, et al. Limited variation in the major urinary proteins of laboratory mice. Physiol Behav. 2009;96(2):253–61.

    Article  CAS  PubMed  Google Scholar 

  118. Sherborne AL, et al. The genetic basis of inbreeding avoidance in house mice. Curr Biol. 2007;17(23):2061–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Xiao Y, et al. Circulating lipocalin-2 and retinol-binding protein 4 are associated with intima-media thickness and subclinical atherosclerosis in patients with type-2 diabetes. PLoS One. 2013;8(6):e66607.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. De la Chesnaye E, et al. Lipocalin-2 plasmatic levels are reduced in patients with long-term type-2 diabetes mellitus. Int J Clin Exp Med. 2015;8(2):2853–9.

    PubMed  PubMed Central  Google Scholar 

  121. Xu A, Tso AW, Cheung BM, Wang Y, Wat NM, Fong CH, Yeung DC, Janus ED, Sham PC, Lam KS. Circulating adipocyte-fatty acid binding protein levels predict the development of the metabolic syndrome: a 5-year prospective study. Circulation. 2007;115(12):1537-1543.

  122. Baur JA, Pearson KJ, Price NL, Jamieson HA, Lerin C, Kalra A, Prabhu VV, Allard JS, Lopez-Lluch G, Lewis K, Pistell PJ, Poosala S, Becker KG, Boss O, Gwinn D, Wang M, Ramaswamy S, Fishbein KW, Spencer RG, Lakatta EG, Le Couteur D, Shaw RJ, Navas P, Puigserver P, Ingram DK, de Cabo R, Sinclair DA. Resveratrol improves health and survival of mice on a high-calorie diet. Nature. 2006;444(7117):337–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Zhou Y, Rui L. Major urinary protein regulation of chemical communication and nutrient metabolism. Vitam Horm. 2010;83:151–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Dhahbi JM, et al. Temporal linkage between the phenotypic and genomic responses to caloric restriction. Proc Natl Acad Sci U S A. 2004;101(15):5524–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Miller RA, et al. Gene expression patterns in calorically restricted mice: partial overlap with long-lived mutant mice. Mol Endocrinol. 2002;16(11):2657–66.

    Article  CAS  PubMed  Google Scholar 

  126. Giller K, et al. Major urinary protein 5, a scent communication protein, is regulated by dietary restriction and subsequent re-feeding in mice. Proc Biol Sci. 2013;280(1757):20130101.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Zhou Y, Jiang L, Rui L. Identification of MUP1 as a regulator for glucose and lipid metabolism in mice. J Biol Chem. 2009;284(17):11152–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. De Giorgio MR, Yoshioka M, St-Amand J. Feeding induced changes in the hypothalamic transcriptome. Clin Chim Acta. 2009;406(1–2):103–7.

    Article  PubMed  CAS  Google Scholar 

  129. van Schothorst EM, et al. Adipose gene expression response of lean and obese mice to short-term dietary restriction. Obesity (Silver Spring). 2006;14(6):974–9.

    Article  Google Scholar 

  130. Maxwell AI, Morrison GM, Dorin JR. Rapid sequence divergence in mammalian beta-defensins by adaptive evolution. Mol Immunol. 2003;40(7):413–21.

    Article  CAS  PubMed  Google Scholar 

  131. Kachroo AH, et al. Evolution. Systematic humanization of yeast genes reveals conserved functions and genetic modularity. Science. 2015;348(6237):921–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We appreciate our colleagues for careful reading of this manuscript and offering valuable advice. We thank David R. Nelson (University of Tennessee, Memphis) for his help in guiding us through the UCSC Genome Browser.

Author information

Authors and Affiliations

Authors

Contributions

GC participated in writing and editing manuscript, and generating Table 2 and Figs. 3 and 4. YW carried out the alignment of the proteins and generated Tables 1 and 2. MM participated in editing the manuscript. EAB participated in editing the manuscript. DCT participated in writing and editing the manuscript and provided insightful feedback. VV participated in conceiving the idea of this manuscript and in drafting and editing the manuscript. DWN participated in writing the manuscript, overlooking all the data provided and contributed in finalizing the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Vasilis Vasiliou.

Ethics declarations

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Charkoftaki, G., Wang, Y., McAndrews, M. et al. Update on the human and mouse lipocalin (LCN) gene family, including evidence the mouse Mup cluster is result of an “evolutionary bloom”. Hum Genomics 13, 11 (2019). https://doi.org/10.1186/s40246-019-0191-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40246-019-0191-9