Phenotypic Mutation 'potbelly' (pdf version)
Allelepotbelly
Mutation Type nonsense
Chromosome6
Coordinate29,068,971 bp (GRCm39)
Base Change T ⇒ A (forward strand)
Gene Lep
Gene Name leptin
Chromosomal Location 29,060,220-29,073,875 bp (+) (GRCm39)
MGI Phenotype FUNCTION: [Summary is not available for the mouse gene. This summary is for the human ortholog.] This gene encodes a protein that is secreted by white adipocytes, and which plays a major role in the regulation of body weight. This protein, which acts through the leptin receptor, functions as part of a signaling pathway that can inhibit food intake and/or regulate energy expenditure to maintain constancy of the adipose mass. This protein also has several endocrine functions, and is involved in the regulation of immune and inflammatory responses, hematopoiesis, angiogenesis and wound healing. Mutations in this gene and/or its regulatory regions cause severe obesity, and morbid obesity with hypogonadism. This gene has also been linked to type 2 diabetes mellitus development. [provided by RefSeq, Jul 2008]
PHENOTYPE: Homozygotes are obese, hyperphagic, have low activity, high metabolic efficiency, impaired thermogenesis, infertility and short lifespan in addition to varying other abnormalities. Strain background affects severity and course of diabetes. Heterozygotes survive fasting longer than control mice. [provided by MGI curators]
Accession Number

Ncbi Refseq: NM_008493.3; MGI:104663

MappedYes 
Amino Acid Change Cysteine changed to Stop codon
Institutional SourceBeutler Lab
Gene Model predicted gene model for protein(s): [ENSMUSP00000067046] [ENSMUSP00000130087]
AlphaFold P41160
SMART Domains Protein: ENSMUSP00000067046
Gene: ENSMUSG00000059201
AA Change: C7*

DomainStartEndE-ValueType
signal peptide 1 21 N/A INTRINSIC
Pfam:Leptin 23 167 5.8e-71 PFAM
Predicted Effect probably null
SMART Domains Protein: ENSMUSP00000130087
Gene: ENSMUSG00000059201
AA Change: C7*

DomainStartEndE-ValueType
signal peptide 1 21 N/A INTRINSIC
Pfam:Leptin 22 98 5.9e-50 PFAM
Predicted Effect probably null
Meta Mutation Damage Score 0.9755 question?
Is this an essential gene? Non Essential (E-score: 0.000) question?
Phenotypic Category Autosomal Recessive
Candidate Explorer Status loading ...
Single pedigree
Linkage Analysis Data
Penetrance  
Alleles Listed at MGI

All alleles(6) : Targeted(3) Spontaneous(2) Chemically induced(1)

Lab Alleles
AlleleSourceChrCoordTypePredicted EffectPPH Score
potbelly2 UTSW 6 29069089 missense possibly damaging 0.95
R0009:Lep UTSW 6 29068971 nonsense probably null
R1190:Lep UTSW 6 29071173 nonsense probably null
R1545:Lep UTSW 6 29070831 missense probably damaging 1.00
R1585:Lep UTSW 6 29069089 missense possibly damaging 0.95
R5253:Lep UTSW 6 29070862 missense probably damaging 1.00
R9113:Lep UTSW 6 29071093 missense probably damaging 0.98
R9757:Lep UTSW 6 29069083 missense probably benign 0.07
Z1176:Lep UTSW 6 29070969 missense possibly damaging 0.50
Z1177:Lep UTSW 6 29071096 missense probably damaging 1.00
Z1177:Lep UTSW 6 29071095 missense probably damaging 1.00
Mode of Inheritance Autosomal Recessive
Local Stock Live Mice
MMRRC Submission 036787-MU
Last Updated 2016-12-08 11:53 AM by Anne Murray
Record Created 2012-11-01 2:14 PM by Hexin Shi
Record Posted 2013-01-03
Phenotypic Description
Figure 1.  Ventral (left) and dorsal (right) views of potbelly mice.  In each picture, potbelly mice (right mouse) is shown next to a wild-type animal (left mouse).
Figure 2.  Fat ratio (as determined from MRI) of potbelly and wild-type littermates.

The potbelly phenotype was identified among ENU-induced G3 mutant mice (Figure 1).  Preliminary data indicate potbelly mice develop severe obesity (Figure 1 & 2), but not diabetes.

Nature of Mutation

The potbelly mutation corresponds to a T to A transversion at position 21 of the Lep transcript (ENSMUST00000069789; version NCBIM37) in exon 2 of 3 total exons.

1 ATGTGCTGGAGACCCCTGTGTCGGTTCCTGTGGCTTTGGTCC

1 -M--C--W--R--P--L--C--R--F--L--W--L--W--S-

The mutated nucleotide is indicated in red lettering and results in a conversion of cysteine 7 to a stop codon.

Illustration of Mutations in
Gene & Protein
Protein Prediction

Figure 3. Mouse leptin domains. Mouse leptin contains a signal peptide (SP) at aa 1-21 that is cleaved during protein maturation.  The residues that consitute the four-helix bundle (helices A-D) are labeled.  The cysteines at aa 117 and 167 form a intramolecular disulfide bond between the C-terminus and the beginning of the CD loop. The residues at 140-142 (i.e., Tyr (Y)-140, Ser (S)-141, and Thr (T)-142) are required for leptin-mediated activation of the LepR. The potbelly mutation results in coding of a premature stop codon at amino acid 7 within the N-terminal signal peptide region.

The Lep (alternatively, ob) gene encodes leptin, a highly conserved [e.g., mouse and human leptin share 84% sequence identity (1-3)] adipocyte-secreted hormone and member of the class I helical cytokine family [Figure 3; (4-9)]. Leptin regulates several physiological processes including appetite, energy homeostasis, body weight, neuroendocrine systems [i.e., the growth hormone axis, thyroid axis, hypothalamic-pituitary-gonadal axis, and adrenal axis (10-13)], immune functions (i.e., thymic homeostasis, secretion of IL-1 and TNF-α, and promotion of Th1-cell differentiation [reviewed in (14)]), and glycaemia [reviewed in (15;16)].

Figure 4. The crystal structure of human leptin. The model is based on PDB: 1AX8. Leptin has a four-helix bundle (helices A-D) with an up-up-down-down topology. Parallel to the helical bundle is a hydrophobic cylindrical core formed from conserved residues of the four alpha helices. Leptin has a small helical segment (i.e., helix E) within the loop between helices C and D.

The crystal structure of human leptin has been solved [PDB: 1AX8; (17); Figure 4].  In order to prevent leptin aggregation and to generate a biologically active and soluble leptin, amino acid Trp (W) 100 was mutated to a Glu (E100) (17). The leptin (E100) tertiary structure resembles that of other class I cytokines [e.g., granulocyte colony-stimulating factor (G-CSF), IL-6, gp130 family members, leukemia inhibitory factor (LIF), ciliary neurotrophic factor (CNTF), and human growth hormone (hGH)] in that it has a four-helix bundle (helices A-D) with an up-up-down-down topology (17-19). A nuclear magnetic resonance (NMR) study determined that in mouse leptin, Helix A is amino acids 3-24, Helix B is aa 51-67, Helix C is aa 72-94, and Helix D is aa 122-141 (18). Parallel to the helical bundle is a hydrophobic cylindrical core formed from conserved residues of the four alpha helices (17). Leptin differs structurally from other class I cytokines in that it has a small helical segment (i.e., helix E) within the loop between helices C and D [aa 95-121 (17)]; the other cytokines have kinks in the middle of helix A, D, or B (4;17).  The length and position of the helices in leptin are most similar to cytokines of the short-helix family (20) such as interleukin-2 (21), interleukin-4 (22), and macrophage-colony stimulating factor (23).

The 167 amino acid leptin has a 21 amino acid N-terminal signal sequence that is cleaved during leptin maturation (1-3;16;24).  Vertebrate leptins have two conserved cysteine residues (Cys117 and Cys167 in mouse leptin) that form an intramolecular disulfide bond between the C-terminus and the beginning of the CD loop (17;25).  The function of the disulfide bond has been debated.  Initial studies reported that the disulfide bond was essential for folding and receptor binding (17).  However, an in vitro study on a mutant with mutations at the conserved cysteines did not exhibit changes in leptin function (25). In contrast, another in vitro study showed that mutations at either (or both) cysteines resulted in impaired leptin secretion and the formation of aggregates (26).  It was proposed that the discrepancies in the in vitro studies was due to the use of different expression systems (26): Imagawa et al. (25) studied mutant leptin purified from E. coli inclusion bodies; Boute et al. (26) studied leptin secretion in mammalian cells Three conserved leptin receptor (LepR; see the records for Business class, Cherub, and Well-upholstered) binding sites on leptin have been identified: site I is on the face of helix D and is proposed to bind the cytokine receptor homology 1 (CRH1) or CRH2 domain of the LepR, site II is composed of residues on the surface of helices A and C and binds the CRH2 domain of LepR, and site III is at the N-terminus of helix D at the interface of the N-terminus of helix D and the AB loop and binds the immunoglobulin-like domain of LepR (1;4;27;28). Binding site II is proposed to be the main high affinity binding site for receptor-ligand interaction (29-31), while site III is proposed to function in forming the active multimeric complex and activating the receptor (27;32).  Mutagenesis of mouse and human leptins in the proximity of binding site III have identified Tyr-140, Ser-141, and Thr-142 as essential for activation, but not binding, of leptin to LepR (27;32). In addition to the three binding sites in leptin, the highly conserved GLDFIP sequence at aa 38-43 in human leptin is required for LepR activation (4;33;34).

The potbelly mutation results in coding of a premature stop codon at amino acid 7 within the N-terminal signal peptide region (aa 1-21).

Expression/Localization

Lep mRNA is highly expressed in white adipose tissue (35), stomach, and liver (2;36-39). Lep is expressed at lower levels in brown adipose tissue, heart, placenta and fetal tissues (40;41), the pituitary gland (42;43), primary and secondary lymphoid organs (19), bone marrow, mammary epithelium, ovaries, skeletal muscle, and the brain in humans and other mammalian species [(44); reviewed in (4;7;16;45;46)].  Although expression of Lep has been verified in the rat brain, expression is not detected in the brain of the mouse (7;47).

Lep expression is regulated in fasting/feeding and diabetes several hormones (e.g., insulin (48-50), glucocorticoid (51;52), and adrenergic agents (35)) regulate Lep. Lep expression is also proportional to the size of adipose cells: smaller cells express less Lep mRNA (53).  It is proposed that changes in the cell wall and plasma membrane tension can initiate signaling to enhance Lep expression (53).

Leptin secretion into the bloodstream is mediated by changes in several factors [reviewed in (16)]. (i) The amount of leptin produced by the body is proportional to the amount of white adipose tissue in the body [(54); reviewed in (7)]. (ii) Fat distribution leads to differential leptin levels in that subcutaneous fat produces more leptin than omental (i.e., abdominal) fat (55). (iii) Sex hormones differentially regulate the levels of leptin, resulting in higher leptin levels in men, independent of adipose tissue levels [(56-58); reviewed in (16)]. Furthermore, leptin levels are the highest in women during the luteal (i.e., secretory) phase of the menstrual cycle when progesterone levels are the highest (59).  (iv) Leptin secretion exhibits a pulsatile release pattern (60) and there is an inverse relation between the rapidity of the leptin fluctuations in the plasma with those of adrenocorticotropic hormone and cortisol.  The amplitude of leptin pulses is greater in obese versus lean individuals [(60); reviewed in (16)]. (v) Leptin expression is regulated in a circadian-based manner; leptin is lowest between early afternoon and mid-afternoon and it is at its highest between midnight and early morning [(61;62); reviewed in (16)]. (vi) Leptin concentrations are low in fetuses and high in neonates, possibly to regulate different developmental processes (i.e., maintaining neural stem cells or progenitor cells in fetuses and regulating the differentiation of neurons in neonates) (63).

Leptin is transported across the blood-brain barrier via the short receptor form of the LepR (LepRa; see the record for Business class) and an unidentified receptor isoform localized to non-neuronal cells in the meninges, choroid plexus, and blood vessels [(64;65); reviewed in (7)].

Background
Leptin, a systemic hormone, regulates multiple functions of the body including energy utilization and storage, various endocrine axes, bone metabolism, thermoregulation, angiogenesis, immunity and inflammation. For a comprehensive review on leptin-associated signaling and functions please see the record for Business class. Table 1 summarizes the current knowledge of leptin-mediated functions.

Table 1.  Summary of leptin-associated functions [reviewed in (3;4;7;27)]

Function

Details

References

Regulation of energy expenditure, food intake, weight loss, and diabetes

  • Leptin acts on the hypothalamus to signal when the body has enough energy, subsequently inhibiting appetite 
  • Leptin influences energy balance through the hypothalamic -pituitary-adrenal (HPA) and hypothalamic -pituitary-thyroid (HPT) axes
  • Leptin stimulates corticotropin releasing hormone, which suppresses appetite
  • Morbid obesity can lead to the development of type 2 diabetes
  • Recombinant leptin produces weight loss in the Lepob/Lepob mouse

(1;66-74)

Promotion of linear growth

  • Leptin influences energy balance
  • Leptin induces mitosis (e.g., chondrocytes of the epiphyseal growth plate)
  • Leptin stimulates pituitary growth hormone

(75;76)

Promotes onset of puberty and the ovulatory cycle in mammals

  • Puberty is promoted via leptin-mediated action on mammalian target of rapamycin (mTOR)
  • Rise in leptin levels precedes onset of puberty
  • Human mutations can cause hypogonadotropic hypogonadism

(63;76-81)

Embryonic and fetal development

  • Lepob/Lepob mice are sterile; leptin supplementation rescues infertility phenotype
  • Leptin regulates the levels of β3-integrin, IL-1, LIF, IL-1 receptor, and VEGF receptor 2 during implantation
  • Leptin promotes oligodendroglial and cortical neuron development in the embryonic cerebral cortex
  • Fetal plasma leptin concentration is correlated with weight, length, and head circumference at birth; maternal leptin concentration shows a negative correlation with fetal growth in humans

(63;82-86)

Neural development

  • Lepob/Lepob mice have reduced brain weight and DNA content; leptin treatment reverses the phenotype

(63;82;87)

Bone development, growth, and homeostasis

  • Low levels of leptin are linked to osteoporosis/stress fractures in women with hypothalamic amenorrhea
  • Leptin deficiency leads to decreased bone formation

(16;88-93)

Lung development, function, and diseases

  • Leptin is expressed in fetal lung tissue; expression is enhanced during alveolar differentiation in rat lung fibroblasts
  • Leptin is a stimulant of ventilation
  • Leptin levels are decreased in patients with obstructive sleep apnea
  • Leptin may regulate the infiltration and survival of inflammatory cells in the submucosa of chronic obstructive pulmonary disease (COPD)

(94-97)

Cardiac function

  • Leptin receptor-mediated activation of signal transducer and activator of transcription-3 (STAT3) mediates cardiac remodeling after injury and in heart failure
  • Leptin acts as a growth factor in cardiac myocytes
  • Increased plasma leptin levels is a potential independent risk factor for coronary heart disease

(98-105)

Immune function

  • Leptin enhances T-cell-mediated immune responses by signaling through the long form of the LepR on CD4+ T lymphocytes
  • Leptin can shift T-cell responses toward a Th1 type, with increased secretion of pro-inflammatory cytokines interleukin-2 and interferon-γ and decreased interleukin-4 production
  • Humans with congenital leptin deficiency have a much higher incidence of infection-related death during childhood
  • Leptin induces the proliferation, differentiation, and function of hemopoietic cells

(14;106-110)

Regulates the development of autoimmune diseases

  • Lepob/Lepob mice show reduced experimentally induced colitis, arthritis, and experimental auto-immune encephalomyelitis

(111-116)

Promotes atherosclerosis

  • Lepob/Lepob mice appear to be resistant to diet-induced atherosclerosis

(83;116;117)

Thyroid function

  • Thyroid-releasing hormone is positively regulated by leptin

(68)

Stress response

  • Leptin blunts the rise of ACTH and corticosterone and inhibits glucocorticoid synthesis and secretion in the adrenal cortex

(118;119)

Induction of angiogenesis

  • In vivo, leptin induces neovascularization in corneas from normal rats, but not in corneas from fa/fa Zucker rats, which lack functional leptin receptors

(120)

Enhancement of wound healing

  • Excisional wounds treated with a neutralizing anti-leptin antibodies had reduced healing progression and wound closure and contraction were prevented

(121)

Cancer

  • Leptin is often overexpressed, along with the LepR, in tumor tissues
  • Increased levels of leptin can be indicative of poor prognosis

(122-125)

In humans, mutations in LEP are linked to morbid obesity, with or without hypogonadism [OMIM: +164160; (1;79;126;127)].  Homozygosity in reported mutations [i.e., frameshift mutation at ΔG133 that leads to a premature stop codon (126), missense mutation R105W (79), and missense mutation at N103K (128)] result in an inability to produce/secrete leptin [reviewed in (1)].  Heterozygosity of a mutant leptin (frameshift ΔG133) can cause increased body weight (129).  In addition to obesity, patients with leptin deficiency can also exhibit many of the phenotypes listed in Table 1.  Administration of leptin to patients has been shown to treat and/or reverse several of the phenotypes [(130-132); reviewed in (16)].

Lep mouse models

The Lepob (ob) strain (MGI:1856424) has a nonsense mutation in Lep that results in coding of a premature stop codon at aa 105 (2); the mutant Lep mRNA is expressed 20-fold higher than wild-type animals although serum leptin levels are undetectable (3).  The Lepob/Lepob mouse is morbidly obese and is characterized by hyperinsulinemia, hyperglucocorticoidemia, hypothalamic hypothyroidism, defects in cell-mediated and humoral immunity [i.e., Thy 1.2-positive lymphocytes are reduced, lower plaque forming cell response, increase in natural killer cell activity, and an increase in antibody-dependent cell-mediated cytotoxicity (133)], impaired thermogenesis, hyperglycemia, insulin resistance, altered central nervous system activity, reduced metabolic rate of brown adipose tissue, infertility, and lethargy [(19;134-137); reviewed in (4;24)]. There are slight strain differences in the severity and course of the diabetes phenotype exhibited by the Lepob/Lepob mice. Upon treatment with leptin, female Lepob/Lepob mice can ovulate and give birth [(78); reviewed in (16)] due to the stimulation of secretion of luteinizing hormone (138). Studies have shown that Lepob/Lepob mice are resistant to actively and passively induced experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis (19;139).  Following leptin administration, the Lepob/Lepob mice become susceptible to the disease (139).  Researchers determined that the resistance to EAE was due to a reduced proliferative response to myelin antigens and an increased IL-4 response; treatment with leptin converted the Th2 to a Th1-type cytokine response and the subsequent secretion of IFN-α and to an IgG1-to-IgG2a isotype shift switch (19). In addition, administration of leptin to wild-type animals promoted an increase in proinflammatory cytokines and IgG2a production, causing a more severe disease phenotype (19).

A second spontaneous mutation, Lepob-2J (MGI:1858048), has a retroviral-like transposon inserted into the first intron of Lep (140).  The insertion of the transposon leads to the production of chimeric RNAs in which the first exon of Lep is spliced to sequences in the insertion (140).  In this mutant, the mature Lep RNA is not synthesized (140). The Lepob-2J/ Lepob-2J mouse is phenotypically indistinguishable from the ob/ob strain.

A third mutant mouse model has an ENU-induced T-to-A mutation in exon 3 of Lep, resulting in a Val (V) to Glu (E) exchange at amino acid 145 (V145E) [MGI: 4880027; (1)]. This residue is in the N-terminal region of helix D, a domain that contains LepR binding site III.   The V145E mutation does not alter the binding of leptin to the LepR (1).  In contrast to the spontaneous models, the level of circulating leptin was increased in the V145E mice (1). Homozygous V145E mice share similar phenotypes to the spontaneous mutants mentioned above and they also exhibit adipocyte hypertrophy and hyperplasia as well as liver steatosis (i.e., fatty infiltration) (1).

Putative Mechanism

The phenotype of the potbelly mice mimic other Lep mouse models [see "Background", above; (1;2;140)] in that they exhibit morbid obesity.  Expression and secretion of Lep in the potbelly mice has not been examined. Other leptin-related phenotypes and functions (as seen in Table 1) have not been tested in potbelly.

Primers PCR Primer
potbelly_pcr_F: CTCTTCCCAGGGGTACACATTTCAC
potbelly_pcr_R: CGATACTGGCAGTACAACTGAGCAG

Sequencing Primer
potbelly_seq_F: CAGGGGTACACATTTCACTAATC
potbelly_seq_R: CTGTATTTCAGCAGGCAGC
Genotyping

Potbelly genotyping is performed by PCR amplifying regions on Lep that contain the mutation. The PCR product is subsequently sequenced to detect the nucleotide change.

The following primers were used for PCR amplification and sequencing of Lep:

Primers for PCR amplification

LEP_F: 5’- CTCTTCCCAGGGGTACACATTTCAC -3’

LEP_R: 5’- CGATACTGGCAGTACAACTGAGCAG -3’

Primers for Sequencing

LEP_Seq_F: 5’- CAGGGGTACACATTTCACTAATC -3’

LEP_Seq_R: 5’- CTGTATTTCAGCAGGCAGC -3’

The following sequence of 574 nucleotides is amplified (Genbank genomic region NC_000072 for linear DNA sequence of Lep):  

ctcttcccag gggtacacat ttcactaatc taggttccat aatgaattgt ctttgacttt          

ggcaagatag tagcaagtta gggaagaaag cacattttat ccgtccacat cctatagcag      

gatggcagca ggaccattgg atggattcat attgggctct tgaaaagtgt cattcattct      

gtctgtaggt gcaagaagaa gaagatccca gggaggaaaa tgtgctggag acccctgtgt      

cggttcctgt ggctttggtc ctatctgtct tatgttcaag cagtgcctat ccagaaagtc      

caggatgaca ccaaaaccct catcaagacc attgtcacca ggatcaatga catttcacac      

acggtaggag tctcatgggg ggacaaagat gtaggactag aaccagagtc tgagaaacat      

gtcatgcacc tcctagaagc tgagagttta taagcctcga gtgtacatta tttctggtca      

tggctcttgt cactgctgcc tgctgaaata cagggctgag tggttccatt tctaaaccca      

gcatctagac tgctcagttg tactgccagt atcg

PCR primer binding sites are underlined; sequencing primer binding sites are highlighted in gray; the mutated nucleotide is shown in red text.

References
Science Writers Anne Murray
Illustrators Diantha La Vine
AuthorsHexin Shi, Ying Wang, Bruce Beutler