Skip to main content

This is a preprint.

It has not yet been peer reviewed by a journal.

The National Library of Medicine is running a pilot to include preprints that result from research funded by NIH in PMC and PubMed.

bioRxiv logoLink to bioRxiv
[Preprint]. 2023 Oct 20:2023.10.20.563165. [Version 1] doi: 10.1101/2023.10.20.563165

The widely used Ucp1-CreEvdr transgene elicits complex developmental and metabolic phenotypes

Manasi Suchit Halurkar 1, Oto Inoue 1, Rajib Mukherjee 1, Christian Louis Bonatto Paese 2,#, Molly Duszynski 3, Samantha A Brugmann 2,5,6, Hee-Woong Lim 4,5, Joan Sanchez-Gurmaches 1,2,5,*
PMCID: PMC10614962  PMID: 37904917

Summary:

Bacterial artificial chromosome transgenic models, including most Cre-recombinases, enable potent interrogation of gene function in vivo but require rigorous validation as limitations emerge. Due to its high relevance to metabolic studies, we performed comprehensive analysis of the Ucp1-CreEvdr line which is widely used for brown fat research. Hemizygotes exhibited major brown and white fat transcriptomic dysregulation, indicating potential altered tissue function. Ucp1-CreEvdr homozygotes also show high mortality, growth defects, and craniofacial abnormalities. Mapping the transgene insertion site revealed insertion in chromosome 1 accompanied by large genomic alterations disrupting several genes expressed in a range of tissues. Notably, Ucp1-CreEvdr transgene retains an extra Ucp1 gene copy that may be highly expressed under high thermogenic burden. Our multi-faceted analysis highlights a complex phenotype arising from the presence of the Ucp1-CreEvdr transgene independently of the intended genetic manipulations. Overall, comprehensive validation of transgenic mice is imperative to maximize discovery while mitigating unexpected, off-target effects.

Introduction:

Mouse transgenic models such as overexpressors, reporters, and Cre-recombinases empower spatial and temporal genetic manipulation enabling unparalleled interrogation of gene function in vivo. These models have driven discoveries across biological scales, from molecular processes to whole body physiology and have become indispensable for elucidating the foundations of health and disease.

Most transgenic mouse alleles in use today have been generated via bacterial artificial chromosome (BAC) technology1,2. This involves inserting sequences of interest (e.g. Cre-recombinase) into BAC plasmids (–150–350 kb) containing regulatory elements that confer spatiotemporal expression. For instance, insertion after a promoter sequence permits cell type- or stage-specific Cre-recombinase expression. The modified BAC, including contextual sequences, is then randomly integrated into the host genome in a nonspecific, stochastic manner, typically forming a multicopy concatemer3.

While revolutionary, Cre-driver lines generated by BAC transgenesis carry potential limitations that are rarely investigated. Validation of Cre-drivers is usually restricted to verification of specific expression in the targeted cell type. Initiatives like the CrePortal4,5 have been invaluable for collating Cre expression data and provide a valuable resource to guide appropriate use of Cre-drivers. Yet other limitations associated with BAC transgenesis are rarely examined: (1) The insertion site of transgenes are mostly unknown; only 5.03% and 3.40% of all transgenic and Cre alleles, respectively, have mapped integration sites collated in Mouse Genome Informatics [Figure S1AB]; (2) insertion can result in large genomic abnormalities that are not routinely inspected6,7,8 and additionally the insertion may directly influence the phenotypes observed by different mechanisms912; (3) passenger sequences are virtually never reported but may lead to unintended phenotypes13; (4) Cre transgenes are largely used in hemizygosity masking phenotypes that would otherwise be evident14,15; (5) the common absence of Cre-only control groups precludes assessment of perturbations directly attributable to the presence of the Cre transgene or protein itself.

BAC transgenics have been instrumental for generating adipose-specific Cre driver lines to dissect the biology of the highly thermogenic brown adipose tissue (BAT) and the energy storing white adipose tissue (WAT)1618. Cre-recombinase lines utilizing the adiponectin promoter enabled targeting of all adipocytes1921. Promoter elements from UnCoupling Protein 1 (Ucp1) have conferred selective Cre-recombinase expression in brown adipocytes. Although other BAT Cre-targeting tools existed22, and other tools to target brown adipocytes are used23, two Ucp1-Cre drivers dominate the literature currently: the constitutive Ucp1-CreEvdr line from the Rosen lab24 and the tamoxifen inducible Ucp1-CreERT2Biat line from the Wolfrum lab25. Both lines show remarkable specificity, full penetrance, and robust activity on brown adipocytes2632. Among the two, the Ucp1-CreEvdr line has been more widely adopted, featuring in 78.85% of manuscripts in the Mouse Genome Informatics records. This skewed utilization may be due to greater accessibility in open repositories and concerns about tamoxifen effects on adipocytes33,34.

Despite the extensive use of the constitutive Ucp1-CreEvdr allele, comprehensive validation of this driver line is lacking. Here, we perform an in-depth characterization of Ucp1-CreEvdr using genetic, genomic and physiologic approaches. Transcriptomic analysis showed substantial gene expression changes in both brown and white adipose tissues of hemizygous Ucp1-CreEvdr mice compared to wild-type littermate controls. Ucp1-CreEvdr homozygotes show high mortality, craniofacial abnormalities, and growth retardation. Molecular characterization of the Ucp1-CreEvdr transgene insertion site demonstrated substantial genomic alterations including disruptions of several genes at the integration locus in chromosome 1. Notably, the Ucp1-CreEvdr transgene retains an additional Ucp1 gene that may exhibit strong expression under high thermogenic burden. These effects suggest unintended consequence on brown adipose tissue function by Ucp1-CreEvdr. More broadly, our study highlights the critical need for extensive validation of BAC transgenic drivers.

Results:

UCP1-CreEvdr Homozygosity Induces Lethality, Growth Impairment, and Craniofacial Abnormalities.

While attempting to generate a Ucp1-CreEvdr mediated deletion of a Ucp1 floxed allele, we were unable to identify mice homozygous for the Ucp1 floxed allele and simultaneously harboring the Ucp1-CreEvdr transgene through standard genotyping (see below). Given that germline Ucp1 knockout mice are viable, embryonic lethality due to Ucp1 deficiency does not explain this bias in genotyping ratios. Thus, we ponder the question on whether the Ucp1-CreEvdr transgene itself may underlie the observed effects.

To rigorously evaluate the Ucp1-CreEvdr model for its use in discovery-based research, we generated control, hemizygous, and homozygous littermates by crossing Ucp1-CreEvdr hemizygous [Figure 1A]. We designated them as controls, 1xUcp1-CreEvdr and 2xUcp1-CreEvdr mice, respectively. We reasoned that this strategy would enable comprehensive assessment of potential developmental, physiological, and molecular perturbations arising from this widely utilized Cre driver line.

Figure 1:

Figure 1:

(A) Experimental strategy for the generation of control, Ucp1-CreEvdr hemizygous (1xUcp1-CreEvdr) and Ucp1-CreEvdr homozygous (2xUcp1-CreEvdr) mice.

(B) Expected and observed offspring genotypes obtained from 1xUcp1-CreEvdr to 1xUcp1-CreEvdr crosses separated by sex. N=251 pups from 46 litters. Statistical significance was calculated using Chi-square test.

(C) Kaplan-Meier survival plot of control, 1xUcp1-CreEvdr and 2xUcp1-CreEvdr mice from 3 to 6 weeks of age. n= 60 controls, 152 1xUcp1-CreEvdr, 23 2xUcp1-CreEvdr. Statistical significance was calculated using Log-rank (Mantel-Cox) test.

(D) Representative photographs of alizarin red S stained skulls of control, 1xUcp1-CreEvdr and 2xUcp1-CreEvdr mice. n=2 females and 2 males.

(E) Skull shape index (ration between condylobasal length and the interorbital constriction length) of control, 1xUcp1-CreEvdr and 2xUcp1-CreEvdr mice. n=2 females and 2 males.

(F) Body weights of control, 1xUcp1-CreEvdr and 2xUcp1-CreEvdr females. n= 7 controls, 9 1xUcp1-CreEvdr, 7 2xUcp1-CreEvdr.

(G) BAT weights of control, 1xUcp1-CreEvdr and 2xUcp1-CreEvdr females. n= 7 controls, 9 1xUcp1-CreEvdr, 5 2xUcp1-CreEvdr.

(H) WAT weights of control, 1xUcp1-CreEvdr and 2xUcp1-CreEvdr females. n= 7 controls, 9 1xUcp1-CreEvdr, 5 2xUcp1-CreEvdr.

(I) Liver weight of control, 1xUcp1-CreEvdr and 2xUcp1-CreEvdr females. n= 7 controls, 9 1xUcp1-CreEvdr, 5 2xUcp1-CreEvdr.

(J) Skeletal muscles weight of control, 1xUcp1-CreEvdr and 2xUcp1-CreEvdr females. n= 7 controls, 9 1xUcp1-CreEvdr, 5 2xUcp1-CreEvdr.

(K) Other organs weight of control, 1xUcp1-CreEvdr and 2xUcp1-CreEvdr females. n= 7 controls, 9 1xUcp1-CreEvdr, 5 2xUcp1-CreEvdr.

(L) Representative H&E images of fat depots and liver from control, 1xUcp1-CreEvdr and 2xUcp1-CreEvdr females. n= 4 per genotype.

Unless otherwise noted, data are mean + SEM and statistical significance was calculated using one-way ANOVA followed by Tukey’s multiple comparisons test. *P < 0.05, **P < 0.01, ***P < 0.001.

To unambiguously discriminate transgene copy number, we developed a quantitative copy number assay to detect Cre in genomic DNA rather than relying on endpoint PCR genotyping [Figure S1C]. At three weeks of age, we find significantly fewer 2xUcp1-CreEvdr mice than the expected Mendelian ratio of 25% [Figure 1B]. Specifically, only 14.04% of females and 16.06% of males are homozygous across 251 pups from 46 litters [Figure 1B]. Analysis of both sexes together reveals that 2xUcp1-CreEvdr comprises just 15.14% of the offspring, reflecting approximately 60% survival [Figure S1D]. Sex distribution is unaffected, indicating no differential penetrance between sexes [Figure S1E]. Moreover, over 40% of 2xUcp1-CreEvdr die spontaneously from 3 to 6 weeks of age, while 1xUcp1-CreEvdr and controls show no mortality [Figure 1C]. This mortality phenotype occurs with no indication of malaise in 2xUcp1-CreEvdr mice. The dramatic reduction in viability and high postnatal lethality in 2xUcp1-CreEvdr mice suggests profound biological perturbations by the Ucp1-CreEvdr transgene.

To understand the potential effects of Ucp1-CreEvdr, we next examined body weights of controls, 1xUcp1-CreEvdr and 2xUcp1-CreEvdr mice from three to six weeks old. 1xUcp1-CreEvdr female and male mice are indistinguishable from controls [Figure S1FG]. However, female and male 2xUcp1-CreEvdr mice display 15% and 15–19%, respectively, lower body weights from 3–6 weeks of age compared to controls and hemizygotes [Figure S1FG]. Additionally, 2xUcp1-CreEvdr appeared to have calvarial defects. To more carefully characterize this dysmorphology, we dissected the heads of six-week-old controls, 1xUcp1-CreEvdr and 2xUcp1-CreEvdr mice and performed Alizarin Red staining. As suspected, 2xUcp1-CreEvdr mice have a more domed, less elongated skull [Figure 1D]. Specifically, the frontal bones appear reduced, while the parietal bones appear increased in size. This dysmorphology resulted in a significantly reduced condylobasal to interorbital constriction length in 2xUcp1-CreEvdr mice [Figure 1E]. Since frontal bones are neural crest derived and parietal bones are mesodermally-derived, this may indicate a differential effect of the homozygosity of the Ucp1-CreEvdr transgene in the development or cross-communication of these two populations35,36. Together, these data unveil craniofacial dysmorphologies and growth retardation in 2xUcp1-CreEvdr mice.

To better understand the effects of the Ucp1-CreEvdr transgene on mouse growth, we performed comprehensive tissue dissections at 6 weeks. Despite lower total body mass in 2xUcp1-CreEvdr females [Figure 1F], dissection of individual fat and lean tissues show that body weight reduction is surprisingly not due to a homogeneously global reduction of weight of each independent tissue. In particular, 2xUcp1-CreEvdr females show no change in BAT depots weights, including interscapular (iBAT), subscapular (sBAT) and cervical (cBAT) compared to control littermates [Figure 1G]. However, posterior subcutaneous or inguinal (psWAT), retroperitoneal (rWAT) and perigonadal (pgWAT) WAT depots are severely impacted, with 39%, 53% and 60% decrease in weight respectively [Figure 1H]. Beyond WAT, only quadriceps mass differs in 2xUcp1-CreEvdr females compared to controls [Figure 1IK]. Male homozygotes also exhibit similar decrease in body weight and dramatic WAT depletion along with reductions in liver, quadriceps and gastrocnemius mass [Figure S1HM]. Thus, tissue-specific effects underlie the global growth retardation in 2xUcp1-CreEvdr.

Histological analysis of iBAT and psWAT reveals no major changes between genotypes in either females or males [Figure 1L, S1P]. However, adipocytes in pgWAT of 2xUcp1-CreEvdr females and males appear to be smaller in size [Figure 1L, S1P]. This suggests that the changes in psWAT and pgWAT weights may be due to different mechanisms involving the generation of adipocytes or control of their size. We next investigated whether aberrant Cre expression could explain the dramatic WAT defects in 2xUcp1-CreEvdr. As expected, Cre mRNA is undetectable in all fat depots of control mice [Figure S1NO]. In female iBAT, Cre expression correlates with transgene copy number, with 3.16-fold higher levels in 2xUcp1-CreEvdr than 1xUcp1-CreEvdr. However, in psWAT and pgWAT of 2xUcp1-CreEvdr females, Cre levels remain hardly detectable and unchanged compared to 1xUcp1-CreEvdr [Figure S1N]. Analysis of male fat depots show similar results [Figure S1O]. This tissue distribution expression argues against Cre misexpression driving WAT perturbations in 2xUcp1-CreEvdr mice.

The UCP1-CreEvdr Transgene is Inserted in Chromosome 1, Disrupts Genomic Integrity, and Harbors an Extra Ucp1 Gene Copy.

To date, the genomic integration site and structure of the Ucp1-CreEvdr transgene are unknown. To elucidate the integration site of the Ucp1-CreEvdr transgene, we performed targeted locus amplification (TLA, Cergentis) in a hemizygous male. TLA is an unbiased, genome-wide method that utilizes sequence-specific inverse PCR of a circularized genomic DNA library following NlaIII fragmentation and crosslinking. Subsequent deep sequencing of PCR products enables mapping of the transgene insertion locus37.

TLA using Cre-specific primers reveals the Ucp1-CreEvdr transgene integrated into chromosome 1, cytoband A5 [Figure S2A, 2A]. As expected, Cre primers also detects homology near the endogenous Ucp1 locus in chromosome 8, indicating inclusion of surrounding Ucp1 genomic sequences in the transgene [Figure 2A]. Primer pairs surrounding Ucp1 produces high signal levels at the Ucp1 locus [Figure S2AB]. TLA maps the concatemer insertion site of Ucp1-CreEvdr to chr1:20,962,125–21,016,858 [Figure 2B]. Integration induces a –54 kb deletion flanking the insertion sites, along with a 3’ –280 kb inversion [Figure 2B]. This directly deletes or inverts the entirety or large portions of 4 genes (Paqr8, Efhc1, Tram2, Tmem14a) [Figure 2B]. Additionally, the concatemer localizes in close proximity to other 7 genes (Il17a, Il17f, Mcm3, Gsta3, Khdc1a, Khdc1c, Khdc1b). Several noncoding sequences within or in close proximity to the concatemer may also be affected [Figure 2B]. The majority of the genes directly affected or in close proximity to the Ucp1-CreEvdr concatemer exhibit low expression in adipose tissues, but they are selectively highly expressed in an array of other tissues [Figure 2C]. Knockout mouse models have not been generated for each coding gene affected by the Ucp1-CreEvdr transgene [Figure 2D]. However, out those generated, only the Mcm3 knockout mice show prenatal lethality with complete penetrance38 [Figure 2D]. Although this is quite distinct from what we observe in 2xUcp1-CreEvdr mice [Figure 1BC, S1DE], the genomic disruption induced by the Ucp1-CreEvdr concatemer may contribute to the above identified perturbations.

Figure 2:

Figure 2:

(A) Whole genome TLA mapping analysis of 1xUcp1-CreEvdr genome using primers specifics for the sequences of Cre.

(B) Schematic representation of the identified integration site of the Ucp1-CreEvdr transgene.

(C) Gene expression of coding genes surrounding the Ucp1-CreEvdr transgene from the Mouse ENCODE transcriptome data.

(D) Knockout mortality phenotype association to each coding gene surrounding the Ucp1-CreEvdr transgene in MGI.

(E) Coverage of BAC 148M1 inserted within the Ucp1-CreEvdr transgene as determined by TLA.

(F) de novo reconstruction of the CRE-proximal part of Ucp1-CreEvdr transgene mRNA from iBAT RNA-seq data. UTR: untranslated region; NLS: nuclear location signal; CDS: coding sequence; Unk: unknown.

(G) Copy number assay of Cre of control, 1xUcp1-CreEvdr and 2xUcp1-CreEvdr mice. n= 3 per genotype.

(H) Copy number assay of Ucp1 of control, 1xUcp1-CreEvdr and 2xUcp1-CreEvdr mice. n= 8 per genotype.

(I) Absolute copy number by ddPCR of Cre of control, 1xUcp1-CreEvdr and 2xUcp1-CreEvdr mice. n= 3 per genotype.

Next, we explored the Ucp1-CreEvdr concatemer structure. To do this, we first analyzed the TLA data. We find that –75% of the original BAC used to generate the Ucp1-CreEvdr mice (BAC 148M1), which covers –230Mb of chromosome 8 surrounding the Ucp1 gene, is incorporated with the transgene in chromosome 1 [Figure 2E]. Notably, this includes an entire extra copy of the Ucp1 gene. To further elucidate transgene structure, we performed de novo assembly of Cre-proximal transgene sequence of iBAT RNA-seq reads in Ucp1-CreEvdr mice using the Cre-recombinase coding sequence as bait. Upstream of the Cre coding sequence, we identified the proximal Ucp1 5’UTR sequence followed by the start codon and SV40 nuclear localization signal [Figure 2F]. The Cre coding sequence is followed at 3’ by a 3’UTR and a short sequence of unknown function [Figure 2F]. The length of sequencing fragments limits the extend of the transgenic Ucp1 gene we can detect as part of the Ucp1-CreEvdr transgene unambiguously against the endogenous copy; however, we find Cre transgene bound to Ucp1 exons 1 and part of exon 2 [Figure 2F]. The presence of Ucp1 mRNA within the Ucp1-CreEvdr transgene transcript suggests that the extra copy of Ucp1 gene may be expressed.

TLA cannot discern the number of repetitions occurring within the concatemer. To clearly determine the copy number of Ucp1 and Cre genes within the Ucp1-CreEvdr concatemer, we employed two quantitative PCR-based techniques. First, we developed copy number assays against the Ucp1 intron 3 to assess the number of copies of Ucp1 gene in genomic DNA of controls, 1xUcp1-CreEvdr and 2xUcp1-CreEvdr mice. Control littermates were used as reference for two copies of Ucp1 gene [Figure 2GH]. In contrast, 1xUcp1-CreEvdr mice have three copies and 2xUcp1-CreEvdr mice have four copies of the Ucp1 gene [Figure 2GH]. However, this assay requires calibration with reference samples, limiting its ability to discern Cre copy number within the transgene concatemer. To solve this, we used digital droplet PCR (ddPCR)39 to quantify the absolute copy number of Cre in HaeIII-digested genomic DNA. As anticipated, control mice contained no copies of Cre. In contrast, 1xUcp1-CreEvdr mice harbored a single Cre copy, while 2xUcp1-CreEvdr mice contained two copies per genome [Figure 2I]. Taken together, these complementary assays indicate that the Ucp1-CreEvdr transgene structure comprises one additional copy of the Ucp1 gene and a single copy of Cre.

UCP1-CreEvdr Transgene Induces Profound Effects in BAT and WAT Biology.

Because Ucp1-CreEvdr directly affects fat size, we next examined if the Ucp1-CreEvdr transgene directly impacts fat biology. First, we performed unbiased whole genome expression profiling of iBAT and psWAT from control, 1xUcp1-CreEvdr and 2xUcp1-CreEvdr female mice. Strikingly, the presence of the transgene profoundly alters the transcriptomic landscape of both fat depots. In 1xUcp1-CreEvdr iBAT, 1012 genes are upregulated and 905 downregulated compared to controls [Figure 3A]. Even more dramatic effects are evident in 1xUcp1-CreEvdr psWAT, with 3742 genes upregulated and 3130 downregulated despite barely detectable transgene expression [Figure 3B, S1N]. Comparisons between 2xUcp1-CreEvdr and control females reveal similar transcriptomic perturbations, with over 10-fold more altered genes in psWAT (8313) than in iBAT (760) [Figure 3CD]. A lower number of genes are significantly different in iBAT and psWAT when comparing 1xUcp1-CreEvdr and 2xUcp1-CreEvdr [Figure S3AB]. Remarkably, this suggest that the major effect appears from having just one copy of the Ucp1-CreEvdr transgene. The dramatic transcriptomic effects in Ucp1-CreEvdr fat depots, especially the psWAT, suggest either potent inter-tissue consequences or major effects from transgene insertion. In summary, unbiased transcriptional profiling indicates that the Ucp1-CreEvdr transgene may profoundly impact the molecular state of both brown and white adipose tissues.

Figure 3:

Figure 3:

(A) RNA-seq comparing female control and 1xUcp1-CreEvdr iBAT gene expression (left). Each dot represents one gene. Corresponding GO analysis (right). Genes and pathways significantly enriched in controls are labeled in orange and those enriched in 1xUcp1-CreEvdr are labeled in red.

(B) RNA-seq comparing female control and 1xUcp1-CreEvdr psWAT gene expression (left). Each dot represents one gene. Corresponding GO analysis (right). Genes and pathways significantly enriched in controls are labeled in orange and those enriched in 1xUcp1-CreEvdr are labeled in red.

(C) RNA-seq comparing female control and 2xUcp1-CreEvdr iBAT gene expression (left). Each dot represents one gene. Corresponding GO analysis (right). Genes and pathways significantly enriched in controls are labeled in orange and those enriched in 2xUcp1-CreEvdr are labeled in brown.

(D) RNA-seq comparing female control and 2xUcp1-CreEvdr psWAT gene expression (left). Each dot represents one gene. Corresponding GO analysis (right). Genes and pathways significantly enriched in controls are labeled in orange and those enriched in 2xUcp1-CreEvdr are labeled in brown.

(E) qPCR analysis of iBAT of control, 1xUcp1-CreEvdr and 2xUcp1-CreEvdr females at 6 weeks of age. n= 6.

(F) qPCR analysis of psWAT of control, 1xUcp1-CreEvdr and 2xUcp1-CreEvdr females at 6 weeks of age. n= 6.

(G) qPCR analysis of pgWAT of control, 1xUcp1-CreEvdr and 2xUcp1-CreEvdr females at 6 weeks of age. n= 6.

(H) Rectal temperature of control and 1xUcp1-CreEvdr females undergoing acute cold challenge. n= 3 controls, 4 1xUcp1-CreEvdr. Statistical significance was calculated using unpaired t-test within timepoint.

(I) Tail temperature of control and 1xUcp1-CreEvdr females undergoing acute cold challenge. n= 3 controls, 4 1xUcp1-CreEvdr. Statistical significance was calculated using unpaired t-test within timepoint.

(J) BAT temperature of control and 1xUcp1-CreEvdr females undergoing acute cold challenge. n= 3 controls, 4 1xUcp1-CreEvdr. Statistical significance was calculated using unpaired t-test within timepoint.

(K) qPCR analysis of iBAT of control and 1xUcp1-CreEvdr females after cold challenge or maintained at room temperature. n= 3. Statistical significance was calculated using unpaired t-test between room temperature (RT) and cold samples.

(L) qPCR analysis of psWAT of control and 1xUcp1-CreEvdr females after cold challenge or maintained at room temperature. n= 3. Statistical significance was calculated using unpaired t-test between room temperature (RT) and cold samples.

(M) qPCR analysis of pgWAT of control and 1xUcp1-CreEvdr females after cold challenge or maintained at room temperature. n= 3. Statistical significance was calculated using unpaired t-test between room temperature (RT) and cold samples.

Unless otherwise noted, data are mean + SEM and statistical significance was calculated using one-way ANOVA followed by Tukey’s multiple comparisons test. *P < 0.05, **P < 0.01, ***P < 0.001. For RNA-seq, differential genes were selected by false discovery rate (FDR) < 0.05 with no fold-change cut-off.

Pathway analysis of the significantly altered genes reveals downregulation of mitochondrial activity pathways (e.g., electron transport chain, respiratory chain, energy generation) in 1xUcp1-CreEvdr psWAT [Figure 3B]. Conversely, mRNA biology pathways are upregulated in 1xUcp1-CreEvdr psWAT [Figure 3B]. 1xUcp1-CreEvdr iBAT display irregularities in diverse pathways not directly related to energy metabolism [Figure 3A]. Similar patterns are observed in 2xUcp1-CreEvdr mice, with psWAT exhibiting suppressed energy generation pathways and elevated mRNA biology [Figure 3D]. In contrast to 1xUcp1-CreEvdr iBAT, 2xUcp1-CreEvdr iBAT uniquely show upregulation of lipid metabolism including very long chain fatty acid metabolism [Figure 3C]. iBAT of 2xUcp1-CreEvdr appears to be enriched in genes related to multiple terms of fatty acid metabolism while psWAT of 2xUcp1-CreEvdr is depleted of them [Figure S3AB]. Collectively, these results poised a scenario in which a single copy of the Ucp1-CreEvdr transgene may affect energy metabolism and thermogenesis pathways in iBAT and psWAT and these effects are heightened in 2xUcp1-CreEvdr mice.

We next used qPCR to verify whether thermogenic gene expression is affected by the Ucp1-CreEvdr transgene. In iBAT, key markers of the classic thermogenesis pathway are largely unchanged across control, 1xUcp1-CreEvdr and 2xUcp1-CreEvdr females, apart from increased Elovl3 in 2xUcp1-CreEvdr [Figure 3E]. In contrast, psWAT of 1xUcp1-CreEvdr females display an approximately 50% reduction in thermogenic genes (i.e., Ucp1, Prdm16, Ppgc1a, Cidea, Cox7a), but not Elovl3 [Figure 3F]. These suppressions are amplified in 2xUcp1-CreEvdr psWAT with reductions of 98% in Ucp1, 69% in Prdm16 and 94% in Cidea [Figure 3F]. Similar thermogenic depletion is evident in 2xUcp1-CreEvdr pgWAT of females, including 96% lower Ucp1 expression on average [Figure 3G]. Male iBAT and psWAT, but not pgWAT, show similar results to females suggesting a gender specific effect on pgWAT [Figure S3CE]. Collectively, these results indicate that the presence of the Ucp1-CreEvdr transgene impairs expression of thermogenic genes in psWAT.

Given the profound effects of Ucp1-CreEvdr on thermogenic gene expression, we hypothesized that the transgene alone, without additional genetic manipulation, would impact cold response. To test this, room temperature-acclimated control and 1xUcp1-CreEvdr female mice were exposed to 4°C for 6 hours. Intriguingly, 1xUcp1-CreEvdr mice exhibit elevated core body temperature, measured by a rectal thermometer, before cold exposure which normalized to control levels during cold [Figure 3H]. Under room temperature conditions, 1xUcp1-CreEvdr mice display lower tail but higher iBAT surface temperatures compared to controls [Figure 3IJ]. This suggests a scenario in which 1xUcp1-CreEvdr shows tail vasoconstriction and elevated iBAT thermogenesis as a physiological mechanism to increase body temperature. Upon cold exposure, 1xUcp1-CreEvdr tail temperature normalized while iBAT remain hyperactive [Figure 3IJ]. However, this is insufficient to maintain core temperature. Together, these data reveal dysfunctional thermogenic regulation and body temperature control in mice harboring just one copy of the Ucp1-CreEvdr transgene.

We next investigated the effects of acute cold exposure on control and 1xUcp1-CreEvdr mice. After 6 hours of cold exposure, 1xUcp1-CreEvdr mice exhibit slightly greater body weight loss compared to controls [Figure S3F]. iBAT weight is unchanged between genotypes; however, psWAT and pgWAT are smaller in 1xUcp1-CreEvdr mice [Figure S3F], indicating increased lipid utilization to maintain body temperature. Cold-induced thermogenic gene expression is largely similar between control and 1xUcp1-CreEvdr iBAT, with comparable upregulation of Ucp1 (–2.8-fold), and Elovl3 (–4.2-fold) [Figure 3K, S3H]. However, psWAT displayed blunted activation, with Ucp1 increasing –52-fold in controls but only –16-fold in 1xUcp1-CreEvdr by cold together with reduced upregulation of other markers such as Elovl3 [Figure 3L, S3I]. pgWAT shows no differences after cold treatment between controls and 1xUcp1-CreEvdr mice [Figure 3M, S3J]. Histological analysis aligns with the gene expression data, revealing fewer multilocular adipocytes in 1xUcp1-CreEvdr psWAT after cold exposure, while iBAT and pgWAT appeared unaffected [Figure S3G]. Together, these data suggest an scenario of impaired psWAT thermogenic activation in response to acute cold stress in mice harboring the Ucp1-CreEvdr transgene.

UCP1-CreEvdr transgene has the potential to express high levels of UCP1.

The discovery of an additional transgenic Ucp1 gene within the Ucp1-CreEvdr transgene raised the question of its potential functionality. We hypothesized that this transgene derived Ucp1 could contribute to overall UCP1 levels. However, the transgenic Ucp1 sequence is identical to endogenous C57Bl6/J Ucp1, precluding its discrimination from the native genes. To overcome this limitation, we adopted a tissue-specific approach utilizing Ucp1-floxed mice (Ucp1tm1a(EUCOMM)Hmgu, EUCOMM)40 harboring LoxP sites flanking exon 2 [Figure S4AB]. By crossing Ucp1-floxed mice with Ucp1-CreEvdr animals, we would selectively ablate endogenous Ucp1 while preserving the transgenic variant. This would allow us to assess the functional impact of the transgenic Ucp1 gene within the Ucp1-CreEvdr concatemer.

However, standard endpoint PCR genotyping failed to identify any mice homozygous for Ucp1-floxed (Ucp1-fl/fl) and positive for Ucp1-CreEvdr (aka. Ucp1-fl/flUcp1-CreEvdr mice)[Fig 4A]. Concurrently, an excess proportion of heterozygous Ucp1-floxed (Ucp1-fl/+) carriers of Ucp1-CreEvdr (aka. Ucp1-fl/+Ucp1-CreEvdr mice) was observed [Fig 4A]. As explained above, embryonic lethality due to Ucp1 deficiency is not expected.

Figure 4:

Figure 4:

(A) Expected and observed offspring genotypes obtained from end-point PCR genotyping and FRT copy number assay. n=131 pups. Statistical significance was calculated using Chi-square test.

(B) Copy number assay of FRT. n= 4.

(C) BAT weights of control, Ucp1-fl/+Ucp1-CreEvdr and Ucp1-fl/flUcp1-CreEvdr males. n= 14 control, 13 Ucp1-fl/+Ucp1-CreEvdr, 9 Ucp1-fl/flUcp1-CreEvdr.

(D) WAT weights of control, Ucp1-fl/+Ucp1-CreEvdr and Ucp1-fl/flUcp1-CreEvdr males. n= 14 control, 13 Ucp1-fl/+Ucp1-CreEvdr, 9 Ucp1-fl/flUcp1-CreEvdr.

(E) Representative H&E images of fat depots from control, Ucp1-fl/+Ucp1-CreEvdr and Ucp1-fl/flUcp1-CreEvdr males. n= 4. Scale bar, 50μm.

(F) qPCR analysis of Ucp1 in adipose tissue depots of control, Ucp1-fl/+Ucp1-CreEvdr and Ucp1-fl/flUcp1-CreEvdr males. Values are relative to those of iBAT control. n= 8.

(G) Western blot of iBAT protein lysates of control, Ucp1-fl/+Ucp1-CreEvdr and Ucp1-fl/flUcp1-CreEvdr males.

(H) Body of control and Ucp1-fl/flUcp1-CreERT2Biat males. n= 7 control, 6 Ucp1-fl/flUcp1-CreERT2Biat.

(I) BAT weights of control and Ucp1-fl/flUcp1-CreERT2Biat males. n= 7 control, 6 Ucp1-fl/flUcp1-CreERT2Biat.

(J) WAT weights of control and Ucp1-fl/flUcp1-CreERT2Biat males. n= 7 control, 6 Ucp1-fl/flUcp1-CreERT2Biat.

(K) Western blot of iBAT protein lysates of control and Ucp1-fl/flUcp1-CreERT2Biat males.

(L) BAT temperature of control and Ucp1-fl/flUcp1-CreEvdr males undergoing acute cold challenge. n= 6 controls, 3 Ucp1-fl/flUcp1-CreEvdr. Statistical significance was calculated using unpaired t-test within timepoint.

(M) Tail temperature of control and Ucp1-fl/flUcp1-CreEvdr males undergoing acute cold challenge. n= 6 controls, 3 Ucp1-fl/flUcp1-CreEvdr. Statistical significance was calculated using unpaired t-test within timepoint.

(N) Rectal temperature of control and Ucp1-fl/flUcp1-CreEvdr males undergoing acute cold challenge. n= 6 controls, 3 Ucp1-fl/flUcp1-CreEvdr. Statistical significance was calculated using unpaired t-test within timepoint.

(O) Rectal temperature of control and Ucp1-fl/flUcp1-CreERT2Biat males undergoing acute cold challenge. n= 3 controls, 3 Ucp1-fl/flUcp1-CreEvdr. Statistical significance was calculated using unpaired t-test within timepoint.

Unless otherwise noted, data are mean + SEM. Statistical significance was calculated using unpaired t-test or one-way ANOVA followed by Tukey’s multiple comparisons test. *P < 0.05, **P < 0.01, ***P < 0.001.

Thus, we ponder the hypothesis that the transgenic Ucp1 gene within the Ucp1-CreEvdr concatemer [Fig 2E] would mask the floxed status of the endogenous Ucp1 alleles by yielding a wildtype band in endpoint PCR genotyping. To overcome this, we obtained the sequences surrounding the FRT site present in floxed allele (tm1c) of the Ucp1tm1a(EUCOMM)Hmgu mice [Figure S4AB]. Next, we designed a copy number assay specific to detect this FRT sequence [Figure S4AB]. We used wildtype, Ucp1-fl/+ and Ucp1-fl/fl mice to calibrate our assay to zero, one and two copies of FRT [Figure 4B]. Using this approach, we readily distinguished Ucp1-fl/+Ucp1-CreEvdr mice harboring one FRT copy from Ucp1-fl/flUcp1-CreEvdr mice with two FRT copies [Figure 4B]. Genotyping with the FRT copy number assay revealed expected Mendelian ratios of control, Ucp1-fl/+Ucp1-CreEvdr, and Ucp1-fl/flUcp1-CreEvdr progeny [Figure 4A], proving that this strategy overcomes the confounding effects from the transgenic Ucp1 sequence.

At 6 weeks of age, total body weight is equivalent between control, Ucp1-fl/+Ucp1-CreEvdr, and Ucp1-fl/flUcp1-CreEvdr mice [Figure S4C]. BAT depots weights are similar between controls and Ucp1-fl/+Ucp1-CreEvdr [Figure 4C]. However, Ucp1-fl/flUcp1-CreEvdr mice display marked –2-fold increase in weight in all BAT [Figure 4C]. WAT, liver or muscle tissue weights are unchanged across genotypes [Figure 4D, S4DF]. In summary, targeted BAT-specific Ucp1 ablation elicit pronounced BAT growth without impacting body and WAT weight.

Histological examination reveals the iBAT hypertrophy in Ucp1-fl/flUcp1-CreEvdr mice is attributable to uniformly enlarged brown adipocytes engorged with excessive lipid [Figure 4E]. In contrast, WAT depots morphology is largely unaffected by genotype [Figure 4E]. This shows a lack of morphological change compensation in WAT by the targeted deletion of Ucp1 in BAT.

qPCR analysis reveals a –50% and –70% reduction in iBAT Ucp1 mRNA in Ucp1-fl/+Ucp1-CreEvdr and Ucp1-fl/flUcp1-CreEvdr mice, respectively, compared to controls [Figure 4F]. Cre mRNA is equal in iBAT of Ucp1-fl/+Ucp1-CreEvdr and Ucp1-fl/flUcp1-CreEvdr mice [Figure S4G]. iBAT also shows a compensatory increased expression of classic thermogenic key markers Ppargc1a, Cox7a, and Elovl3 [Figure S4J]. In psWAT, Ucp1 expression is unaltered; however Ucp1-fl/flUcp1-CreEvdr mice display elevated expression of Cidea, Cox7a, and Elovl3 [Figure 4F, S4K]. pgWAT gene expression is largely unchanged [Figure 4F, S4L]. Cre mRNA expression is slightly upregulated in psWAT and pgWAT of Ucp1-fl/flUcp1-CreEvdr mice partially recapitulating a possible compensation in psWAT, but with levels still much lower than those found in iBAT [Figure S4HI]. Thus, BAT targeted Ucp1 ablation induces depots-specific effects, including a rather unique selective compensatory thermogenic activation in iBAT and psWAT.

The high residual Ucp1 mRNA expression in iBAT of Ucp1-fl/flUcp1-CreEvdr mice was unexpected. This is because, first, Ucp1 is restricted to mature brown adipocytes and second, because Ucp1-CreEvdr drives recombination in essentially all brown adipocytes, as we and others have previously shown28,41. To further investigate this, we examined UCP1 protein levels by western blot. Remarkably, iBAT lysates of Ucp1-fl/flUcp1-CreEvdr mice retained variable but high UCP1 protein levels, averaging approximately 70% of control [Figure 4G]. Given the broad UCP1-CreEvdr-mediated excision in brown adipocytes, the substantial UCP1 retained seems unlikely to be derived from endogenous Ucp1 genes. This paradoxical preservation of UCP1 protein suggests functionally significant expression from the transgenic Ucp1 within the UCP1-CreEvdr concatemer.

As a control, we crossed Ucp1-floxed mice with the tamoxifen-inducible Ucp1-CreERT2Biat allele. Tamoxifen treatment of Ucp1-fl/flUcp1-CreERT2Biat mice does not change body weight but increases BAT weights with a slight enlargement of pgWAT and liver, while other fats and tissues are unchanged [Figure 4HJ, S4MO]. Critically, tamoxifen treatment leads to highly efficient ablation of UCP1 protein in iBAT [Figure 4K]. This confirms that the Ucp1-floxed allele can be efficiently deleted and reinforce the hypothesis that the lack of UCP1 deletion in Ucp1-fl/flUcp1-CreEvdr may arise from ectopic transgene expression.

To test for functionality, we performed acute cold challenges by exposing mice at 6°C for 6 hours. Intriguingly, Ucp1-fl/flUcp1-CreEvdr mice exhibit rather elevated core and BAT temperatures before starting cold exposure, compared to littermate controls [Figure 4LN]. Unexpectedly, Ucp1-fl/flUcp1-CreEvdr mice are proficient in maintaining core body temperature during cold exposure at the same level than littermate controls [Figure 4N]. In stark contrast, Ucp1-fl/flUcp1-CreERT2Biat mice rapidly become hypothermic at 6°C, reflecting the efficient ablation of UCP1 in BAT [Figure 4O]. Together, these results indicate that the UCP1 protein present in Ucp1-fl/flUcp1-CreEvdr BAT, which may be bestowed by the Ucp1-CreEvdr allele, may confer sufficient thermogenic capacity to preserve body temperature.

Discussion

The Cre-Lox system is invaluable for spatial and temporal dissection gene function, tracing lineages, and labeling cells. However, the validation of Cre-recombinase transgenes in the literature is usually incomplete. In particular, the UCP1-CreEvdr transgene transformed BAT and metabolism research. However, our findings reveal several key unexpected caveats of this widely used line including (1) increased mortality, growth defects, and craniofacial alterations in homozygosity; (2) substantial genomic disruptions; (3) profound impacts on BAT and psWAT function in both hemizygosity and homozygosity; and (4) potential misexpression of Ucp1 itself under high thermogenic burden. These findings highlight the importance of carefully considering the validation of transgenic lines before embarking on experimental studies.

BAC Cre-drivers are usually validated only by examining spatiotemporal recombination, fueling comprehensive databases that guide selection among a large and growing number of Cre-recombinases available4,5. Causes for unexpected transgene expression include insertion effects on local regulation or integration of sequences leading to ectopic expression. Alternatively, unanticipated activity may reflect previously unknown endogenous gene expression. Adipocyte-targeting Cre lines exemplify these issues. The promoter of the fatty acid binding protein 4 gene, also known as adipocyte protein 2 (aP2), was used to generate two independent aP2-Cre mouse models with the intention to specifically target mature adipocytes42,43. However, these aP2-Cre models were found to inefficiently target mature adipocytes while exhibiting broad recombination in the brain, endothelial cell in adipose tissues, macrophages, adipocyte progenitors, and elsewhere5,16,20,4447. This prompted development of Cre lines driven by the promoter of adiponectin19,20. However, Ucp1-CreEvdr also exhibits widespread brain activity41,48, including regions controlling feeding and non-shivering thermogenesis4951. Importantly, very low endogenous Ucp1 expression partially overlaps with these brain areas41,48, suggesting Ucp1-CreEvdr partially recaptures native Ucp1 regulation. However, if this reflects endogenous Ucp1 expression or expression of the Ucp1 gene found within the UCP1-CreEvdr transgene, that we find here, is not known at this point.

The recent close examination of some BAC transgenic lines, beyond cellular expression, has shown that the transgenes themselves can result in phenotypes leading to potential misinterpretations of the intended genetic modifications5254. Random BAC transgene insertion is frequently associated with substantial genomic alterations, often disrupting gene coding sequences and creating small or large rearrangements6,7,8. As expected, Ucp1-CreEvdr mice exhibit major structural variations at the insertion site including relatively large deletions and inversions. Historically, the position in the genome and the genomic alterations induced by the random insertion of a transgene have not been systematically examined. This is partially due to previous low-resolution techniques like FISH or linkage mapping which poorly defined insertion sites and structures. However, new sequencing approaches such as whole genome sequencing and TLA enable fine mapping of insertion locus, disruption effects, and integrated sequences37. For instance, whole genome sequencing and TLA both revealed Adipoq-CreEvdr transgene inserted into the Tbx18 gene on chromosome 919,55,56, perturbing Tbx18 expression and adding passenger gene copies with possible widespread effects56. Additionally, BAC transgenes normally integrate as concatemers leading to multiple full or partial copies of the transgene3,57,58. Using ddPCR, we find that Cre coding sequence is present in a single copy in Ucp1-CreEvdr mice. This was also the case for the Adipoq-CreEvdr transgene56. However, ddPCR analysis is limited to a small specific sequence via specific primers; thus, if partial coding or non-coding sequences are present, they may not be detected by ddPCR39. TLA is also not capable of defining the order or number of copies within the inserted concatemer in a transgenic line. Defining insertion sites and the full structure including inserted sequences may only be possible by new long-range genome sequencing7,59,60. Additionally, transgenic strains are bred for generations, allowing accrual of modifications in transgenic sequences and the genetically linked endogenous sequences over time. Furthermore, Cre-drivers with initially robust expression can become leaky or lose activity. Monitoring integrated sequences over mouse line generations could enhance integrity of lines maintained and avoid misinterpretations.

The genetic alterations induced by random transgene insertion and the passenger sequences inserted can lead to confounding genetic effects that are dependent on the transgene rather than the intended genetic alteration. In Ucp1-CreEvdr mice, we observe drastic transcriptional dysregulation in iBAT and psWAT, suggesting major changes in tissue function. Moreover, interactions between transgene effects and specific genetic manipulations (e.g., deletions) are unpredictable. While linking fat transcriptomics to whole body physiology is challenging, these data indicate that Ucp1-CreEvdr the transgene has the potential to profoundly perturb adipose function. In this sense, 1xUcp1-CreEvdr mice show altered body temperature dynamics and distinct cold reactions when compared to controls. Compared to Adipoq-CreEvdr mice, which show minimal adipose tissues gene expression changes56, the Ucp1-CreEvdr effects are considerable. Beyond targeted tissues, transgenes can reprogram untargeted tissues due to genomic disruption and passenger genes. For Ucp1-CreEvdr, the extra Ucp1 gene seems highly expressed under high thermogenic demand, exemplifying how passenger genes can have unexpected impact. Overall, transgene insertion effects are diverse and context dependent. Thus, thorough characterization of each line is essential to parse transgene-specific artifacts from intended genetic effects.

Homozygosity often reveals phenotypes undetectable in heterozygotes, as most loss-of-function mutations display recessive inheritance. Thus, generating homozygous BAC transgenic models can uncover cryptic transgene-dependent effects. For example, crosses of hemizygous Adipoq-CreEvdr mice have not produced homozygous mice suggesting lethality, although this experiment may have been underpowered55. The unexpected high mortality and other major effects caused by homozygosity of Ucp1-CreEvdr imply impacts beyond adipose tissue. The multiple genes that are directly affected by the insertion site and that are expressed in a range of tissues may be responsible for these phenotypes. Moving forward, evaluating homozygous transgenic models, despite logistic challenges, may be a strong paradigm to ensure detection of subtle artifacts.

A limitation across published studies finding unexpected transgenic effects is that they usually lack mechanistic resolution. For example, the underlying causes of 2xUcp1-CreEvdr mortality and 1xUcp1-CreEvdr fat transcriptional changes remain unresolved. However, this knowledge gap in the literature is reasonable given the manifold possibilities. Effects could arise from chromosomal rearrangements, long-distance interactions, 3D conformation changes, or passenger sequences within the BAC, among other potential mechanisms. Moreover, unraveling any single mechanism may provide limited core insights into normal and pathogenic biology, as insertion effects likely stem from complex interactions only arising in the context of the transgenic line analyzed. While mechanistic details are invaluable, delineating the precise causes underlying insertion artifacts would require substantial efforts unlikely to significantly produce advancements. As such, the mechanistic ambiguity in these studies is expected.

New techniques for generating transgenic mice can mitigate issues with random BAC insertion, bolster rigor and drive discovery. CRISPR-directed insertion at known safe harbor loci provides control over transgene placement. Additionally, rational design of regulatory sequences enables precise spatiotemporal expression, avoiding complications from passenger DNA in large BAC constructs. As these and other innovations become widespread, they will complement and enhance previous data obtained with BAC transgenics.

The effects we report here may justify the reevaluation of some prior work using Ucp1-CreEvdr mice to clarify confounding outcomes. However, such assessments will be complicated by the unpredictable interactions between the Ucp1-CreEvdr effects and intended genetic changes. The strength and specificity of Ucp1-CreEvdr transgene in brown adipocytes ensures that this line remains useful in verified contexts. In future studies, control groups with only the Ucp1-CreEvdr transgene could be incorporated to parse its specific effects. Orthogonal tools like additional Cre lines (e.g., inducible Ucp1-CreERT2Biat allele) can also confirm results. Finally, new Cre-recombinase drivers may be generated, using methodologies described above, to confirm previous results. Though challenging, careful experimental design and layered validation can distinguish between effects dependent on the transgene, gene manipulation, or the interaction between the two.

In conclusion, validation of research tools is a requirement of several funding agencies, yet standards for doing so remain opaque. While BAC transgenics have revolutionized basic and biomedical research, limitations have become increasingly apparent. Overall, transparent validation, cautious interpretation, and technological innovations will maximize scientific rigor of BAC transgenics as future tools to catalyze discovery.

STAR METHODS

Lead contact and material availability

Further information and request for resources and reagents should be directed to and will be fulfilled by the Lead contact, Joan Sanchez-Gurmaches (juan.sanchezgurmaches@cchmc.org).

Experimental model and subject details

Mice and mice housing

All mice used in this study were in C57Bl6/J background. UCP1-Cre (JAX stock 024670), R26R-mTmG mice (JAX stock 007676), Ucp1-CreER mice were described before25. Ucp1 flox mice were obtained from the EUCOMM program (C57BL/6N-Ucp1tm1a(EUCOMM)Hmgu/Ieg) after removal of the LacZ and neomycin cassette by Flippase.

Unless noted otherwise, mice were housed in the CCHMC Animal Medicine Facility in a clean room set at 22°C and 45% humidity on a daily 12h light/dark cycle, and kept in ventilated racks fed ad libitum with a standard chow diet, with bedding changed every two weeks. See figure legend for specific age and number of mice used. All animal experiments were approved by the CCHMC IACUC.

For long term temperature acclimation experiments, mice were housed in rodent incubators (7001–33 series, Caron) in pairs within the facilities of animal medicine of CCHMC. Room temperature group mice were co-housed in the same facility as the mice in rodent incubators. Mouse cages were changed weekly using components pre-adjusted to temperature. No cage enrichment was used in this set of experiments.

Method Details

MGI alleles and publications search

MGI database was searched with search word “transgenic” on 12/16/2022. Out of the 10,166 results, mice with Transgenic allele type were selected. Out of these, mouse models with location indicated as “unknown” were assigned as the location of the transgene unidentified group. The remainder mouse models were automatically assigned to transgene with known location group. To analyze Cre-driver mouse models the symbol of each mouse model was search for containing “Cre”. Out of the 1,968 mouse models found, location was assigned as above. Number of publications assigned to specific transgenic mice were found in MGI on July 2023.

GWAS associated traits and mouse mortality phenotype search.

Mouse mortality phenotype associated to gene knockouts was searched in the MGI database. GWAS associated traits to specific genes were search in the Phenotype-Genotype Integrator of the NCBI.

Growth and Survival

Body weight and survival of mice was followed starting at week 3 of age as tail snips were taken for genotyping. Body weights and survival was recorded weekly after that until 6 weeks of age.

Tamoxifen treatment

Tamoxifen was dissolved in corn oil/ethanol (9:1 vol/vol) at 2mg/mL by shaking at 4°C overnight. 6 week old mice were injected with 2 mg/day/mouse for 5 times in a period of seven days. A subgroup of mice was additionally injected for four days during the third week after first injection. Mice were sacrificed three weeks after first injection.

Tissue dissection

Tissues were carefully dissected to avoid surrounding tissue contamination. Adipose tissue notation used here was described previously61. Mice were dissected at early morning without fasting or any other alteration, unless noted in the figure legend.

Acute cold exposure

Mice were placed at 4°C early in the morning of the experiment in overnight pre-chilled caging with free access to pre-chilled water with or without food.

Temperature measurements

Internal temperature was recorded by using a rectal thermometer probe (RET-3, Braintree Scientific Inc.). BAT and tail temperatures were obtained using an infrared thermal camera (FLIR T530 24°) in lightly anesthetized mice and analyzed with FLIR tools.

Tissue histology

Tissue pieces were fixed in 10% formalin. Embedding, sectioning and Hematoxylin and Eosin (H&E) staining was done by the CCHMC Pathology Core facility.

Craniofacial morphometric analysis

Samples were incubated in 0.005% Alizarin Red S (Sigma-Aldrich, A5533) in 1% KOH for 24 hours at room temperature and cleared in 1% KOH for 72 hours. Once cleared, samples were incubated in Glycerol:KOH 1% (50:50) solution. For imaging and long-term storage, samples were kept in 100% glycerol. Stained skulls were imaged using a Leica M165 FC stereo microscope system for measurements. The condylobasal length and the interorbital constriction length were measured in ImageJ. The ratio between them was used as skull shape defining factor.

qPCR analysis

Tissues were homogenized in a FASTPREP-24 (MP Biomedicals) using Qiazol (Qiagen). Total RNA was isolated using RNeasy kit (Qiagen), retrotranscription was done using High Capacity cDNA reverse transcription kit (#4368813, Applied Biosystems) and analyzed in a QuanStudio 3 real-time PCR machine (Thermofisher). Primer sequences are shown in Table S1.

Western blot analysis

Tissues were homogenized in a FASTPREP-24 (MP Biomedicals) and lysed in RIPA buffer (150 mM NaCl, 50 mM Hepes at pH 7.4, 0.1% SDS, 1% Triton X-100, 2 mM EDTA, 0.5% Na-deoxycholate) containing a protease and phosphatase inhibitor cocktail. Protein lysates (typically 10mg per lane) were mixed with 5X SDS sample buffer and run in SDS acrylamide/bis-acrylamide gels (typically 10 or 12%), transferred to PVDF membranes and detected with specific antibodies as specified in Table S2.

Copy number assays

Genomic DNA was isolated from tails or liver using the DNeasy Blood and Tissue kit spin columns (Qiagen) and diluted to 1 ng/μL. Copy number assays were done using Taqman ® copy number assays (ThermoFisher) using predesigned oligonucleotides assays using Tfrc as reference gene (ThermoFisher)(Table S4). qPCR was performed on a QuantStudio 6Flex real-time PCR system using the following protocol: 95°C for 10min followed by 40 cycles of 95°C for 15 s and 60°C for 60 s with manual Ct threshold at 0.2 and Autobaseline on. Results were analyzed on Copy Caller 2.0 software (ThermoFisher).

Digital droplet PCR

Reaction mixture was composed of 10 uL 2x ddPCR Supermix (without dUTPs; Bio-Rad, Hercules CA), 1 uL each of the proves against housekeeping and target gene (Bio-Rad)(Table S4), 1uL of HaeIII (NEB, R0108), 50 ng of DNA template, and adjusted to a final volume of 20 uL. Droplets were generated in a 96-well polypropylene plate (Bio-Rad) using the QX200 droplet generator (Bio-Rad). The plate containing the water-in-oil emulsions was sealed with foil using a PX1 PCR Plate Sealer (Bio-Rad) and placed in a C1000 Touch Thermal Cycler (Bio-Rad). The following conditions were used for amplification: 95°C for 10 minutes, 94°C for 30 seconds and 60°C for 1 minute (40 cycles 2°C/sec ramp rate), a 10-minute hold at 98°C, and a final hold at 4°C. The plate was processed using the QX200 droplet reader (Bio-Rad). Results were analyzed using QuantaSoft Analysis Pro software version 1.0.596.

Targeted Locus Amplification

UCP1-Cre transgene location and genetic rearrangements associated were approximated by Targeted Locus Amplification (TLA)(Cergentis B.V.) in splenocytes. Splenocytes were isolated from 8 weeks old UCP1-Cre hemizygous mice by pushing the spleen through a 40 mm mesh and collecting in 10% fetal bovine serum in PBS. After red blood cell lysis and washes with 10% fetal bovine serum in PBS, around 10 million spleen cells were aliquoted in cryovials in freezing media (10% fetal bovine serum, 10% DMSO, in PBS). TLA analysis was performed by Cergentis B.V. as previously reported37 with six independent pairs of primers (Table S3) using mouse mm10 genome as host reference.

Whole genome gene expression profile.

RNA-seq reads were aligned to UCSC mouse genome 10 mm using STAR aligner62. Only uniquely aligned reads were used for downstream analysis. Raw read counts for each gene were measured using FeatureCounts in the subread package63 with an option, “-s 2 -O --fracOverlap 0.8”. Differential gene expression analysis was performed using EdgeR64. Genes with Fold-change > 1.5 and FDR < 0.05 were selected as differentially expressed genes. Gene ontology analysis was performed using Enrichr65.

De novo assembly.

To build a Cre transgene sequence, we performed incremental alignment and de novo assembly. Initially, we built a STAR reference combining mm10 and the known Cre CDS sequence. Read 1 and 2 from each RNA-seq sample were aligned to the combined reference separately, where we selected read pairs for de novo assembly if at least one out of the pair is aligned to the Cre reference. And then, we pooled the selected read pairs and performed de novo assembly using Trinity66. Given the design of Cre transgene, we anticipated that the assembled sequence should fully cover Cre CDS and span from Cre CDS sequence to the 5’ half and 3’half of the Ucp1 exon #1. We observed that the very first assembly results fully cover the Cre CDS and connect between Ucp1 5’ half and Cre CDS with a small gap of unknown sequence but not the 3’half. Therefore, we updated the Cre reference with the assembled sequence and repeated these steps of alignment, selection, and de novo assembly until the assembly result reaches the 3’ half of the Ucp1 exon #1, which happened after 4th round. The final assembly results were assessed and annotated using known references and Blast67.

Figure design.

Figures were made in Adobe Illustrator. Several figures were created with BioRender.com.

QUANTIFICATION AND STATISTICAL ANALYSIS

Data are presented as mean+s.e.m., unless stated otherwise. Unpaired t-test, analysis of variance (one or two ways) followed by Tukey’s multiple comparisons, Chi-square and Log-rank (Mantel-Cox) as appropriate, were used to determine statistical significance. No pre-test was used to choose sample size. Statistical analysis was done using GraphPad Prism except for global RNA expression (see methods). The number of mice used per experiment is stated in each figure legend. In all panels, *P < 0.05, **P < 0.01, ***P < 0.001.

Supplementary Material

Supplement 1

Highlights:

  • Hemizygous Ucp1-CreEvdr mice exhibit substantial brown and white fat tissue dysregulation.

  • Homozygous Ucp1-CreEvdr mice display high mortality, growth defects, and craniofacial abnormalities.

  • The Ucp1-CreEvdr transgene integration resulted in major genomic disruptions affecting multiple genes.

  • The Ucp1-CreEvdr transgene retains a possibly functional extra Ucp1 copy.

Acknowledgements:

This work was supported by grants from the American Heart Association (18CDA34080527 to J.S.-G; and 19POST34380545 to RM), the NIH (R21OD031907 to J.S.-G, R35 DE027557 to SAB), a CCHMC Trustee Award to J.S.-G, a CCHMC Trustee Award to HL, a Center for Pediatric Genomics grant (CCHMC) to J.S.-G and HL and a Center for Mendelian Genetics grant (CCHMC) to J.S.-G. OI is supported by a Japanese Heart Foundation Research Abroad Award 2021. This project was supported in part by NIH P30 DK078392 of the Digestive Diseases Research Core Center in Cincinnati. This project was supported in part by the National Center for Advancing Translational Sciences of the National Institutes of Health, under Award Number 2UL1TR001425-05A1. The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH. We thank Helmholtz Zentrum München - Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH) for providing the mutant allele:C57BL/6N-Ucp1tm1a(EUCOMM)Hmgu, EMMA/INFRAFRONTIER (www.infrafrontier.eu) for distributing mouse line (EM:05767). We thank Christian Wolfrum for UCP1-CreERT2 mice. We thank Matt Kofron (Nikon Confocal Imaging Core at CCHMC) for support on image acquisition and analysis. We thank Barbara Cannon and Jan Nedergaard (The Wenner-Gren Institute, Stockholm University) for UCP1-floxed mice and for critically reading the manuscript. We thank Evan D Rosen (BIDMC, Harvard) for critically reading the manuscript. We thank all members of the Sanchez-Gurmaches lab for valuable discussions.

Footnotes

Conflict of interest: The authors declare no conflict of interest.

References:

  • 1.Montoliu L. (2023). Transgenesis and Genome Engineering: A Historical Review. Methods in molecular biology (Clifton, N.J.) 2631, 1–32. 10.1007/978-1-0716-2990-1_1. [DOI] [PubMed] [Google Scholar]
  • 2.Yang X.W., Model P., and Heintz N. (1997). Homologous recombination based modification in Escherichia coli and germline transmission in transgenic mice of a bacterial artificial chromosome. Nature biotechnology 15, 859–865. 10.1038/nbt0997-859. [DOI] [PubMed] [Google Scholar]
  • 3.Smirnov A., and Battulin N. (2021). Concatenation of Transgenic DNA: Random or Orchestrated? Genes (Basel) 12. 10.3390/genes12121969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Perry M.N., Smith C.M., Onda H., Ringwald M., Murray S.A., and Smith C.L. (2022). Annotated expression and activity data for murine recombinase alleles and transgenes: the CrePortal resource. Mamm Genome 33, 55–65. 10.1007/s00335-021-09909-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Heffner C.S., Herbert Pratt C., Babiuk R.P., Sharma Y., Rockwood S.F., Donahue L.R., Eppig J.T., and Murray S.A. (2012). Supporting conditional mouse mutagenesis with a comprehensive cre characterization resource. Nature communications 3, 1218. 10.1038/ncomms2186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Goodwin L.O., Splinter E., Davis T.L., Urban R., He H., Braun R.E., Chesler E.J., Kumar V., van Min M., Ndukum J., et al. (2019). Large-scale discovery of mouse transgenic integration sites reveals frequent structural variation and insertional mutagenesis. Genome research 29, 494–505. 10.1101/gr.233866.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Nicholls P.K., Bellott D.W., Cho T.J., Pyntikova T., and Page D.C. (2019). Locating and Characterizing a Transgene Integration Site by Nanopore Sequencing. G3 (Bethesda) 9, 1481–1486. 10.1534/g3.119.300582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Cain-Hom C., Splinter E., van Min M., Simonis M., van de Heijning M., Martinez M., Asghari V., Cox J.C., and Warming S. (2017). Efficient mapping of transgene integration sites and local structural changes in Cre transgenic mice using targeted locus amplification. Nucleic acids research 45, e62. 10.1093/nar/gkw1329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Dinsmore C.J., Ke C.Y., and Soriano P. (2022). The Wnt1-Cre2 transgene is active in the male germline. Genesis (New York, N.Y. : 2000) 60, e23468. 10.1002/dvg.23468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Liu S., Lockhart J.R., Fontenard S., Berlett M., and Ryan T.M. (2020). Mapping the Chromosomal Insertion Site of the GFP Transgene of UBC-GFP Mice to the MHC Locus. J Immunol 204, 1982–1987. 10.4049/jimmunol.1901338. [DOI] [PubMed] [Google Scholar]
  • 11.Lee S., Kuramochi-Miyagawa S., Nagamori I., and Nakano T. (2022). Effects of transgene insertion loci and copy number on Dnmt3L gene silencing through antisense transgene-derived PIWI-interacting RNAs. Rna 28, 683–696. 10.1261/rna.078905.121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Laboulaye M.A., Duan X., Qiao M., Whitney I.E., and Sanes J.R. (2018). Mapping Transgene Insertion Sites Reveals Complex Interactions Between Mouse Transgenes and Neighboring Endogenous Genes. Front Mol Neurosci 11, 385. 10.3389/fnmol.2018.00385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Lewis A.E., Vasudevan H.N., O’Neill A.K., Soriano P., and Bush J.O. (2013). The widely used Wnt1-Cre transgene causes developmental phenotypes by ectopic activation of Wnt signaling. Developmental biology 379, 229–234. 10.1016/j.ydbio.2013.04.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Zheng H., López-Ferreras L., Krieger J.P., Fasul S., Cea Salazar V., Valderrama Pena N., Skibicka K.P., and Rinaman L. (2022). A Cre-driver rat model for anatomical and functional analysis of glucagon (Gcg)-expressing cells in the brain and periphery. Molecular metabolism 66, 101631. 10.1016/j.molmet.2022.101631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Yong C.S., Sharkey J., Duscio B., Venville B., Wei W.Z., Jones R.F., Slaney C.Y., Mir Arnau G., Papenfuss A.T., Schröder J., (2015). Embryonic Lethality in Homozygous Human Her-2 Transgenic Mice Due to Disruption of the Pds5b Gene. PloS one 10, e0136817. 10.1371/journal.pone.0136817. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Wolfrum C., and Straub L.G. (2019). Lessons from Cre-Mice and Indicator Mice. Handbook of experimental pharmacology 251, 37–54. 10.1007/164_2018_146. [DOI] [PubMed] [Google Scholar]
  • 17.Wang Q.A., Scherer P.E., and Gupta R.K. (2014). Improved methodologies for the study of adipose biology: insights gained and opportunities ahead. Journal of lipid research 55, 605–624. 10.1194/jlr.R046441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Kang S., Kong X., and Rosen E.D. (2014). Adipocyte-specific transgenic and knockout models. Methods in enzymology 537, 1–16. 10.1016/b978-0-12-411619-1.00001-x. [DOI] [PubMed] [Google Scholar]
  • 19.Eguchi J., Wang X., Yu S., Kershaw E.E., Chiu P.C., Dushay J., Estall J.L., Klein U., Maratos-Flier E., and Rosen E.D. (2011). Transcriptional control of adipose lipid handling by IRF4. Cell metabolism 13, 249–259. 10.1016/j.cmet.2011.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Jeffery E., Berry R., Church C.D., Yu S., Shook B.A., Horsley V., Rosen E.D., and Rodeheffer M.S. (2014). Characterization of Cre recombinase models for the study of adipose tissue. Adipocyte 3, 206–211. 10.4161/adip.29674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Wang Z.V., Deng Y., Wang Q.A., Sun K., and Scherer P.E. (2010). Identification and characterization of a promoter cassette conferring adipocyte-specific gene expression. Endocrinology 151, 2933–2939. 10.1210/en.2010-0136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Guerra C., Navarro P., Valverde A.M., Arribas M., Brüning J., Kozak L.P., Kahn C.R., and Benito M. (2001). Brown adipose tissue-specific insulin receptor knockout shows diabetic phenotype without insulin resistance. J Clin Invest 108, 1205–1213. 10.1172/jci13103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Sun K., Kusminski C.M., Luby-Phelps K., Spurgin S.B., An Y.A., Wang Q.A., Holland W.L., and Scherer P.E. (2014). Brown adipose tissue derived VEGF-A modulates cold tolerance and energy expenditure. Molecular metabolism 3, 474–483. 10.1016/j.molmet.2014.03.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Kong X., Banks A., Liu T., Kazak L., Rao R.R., Cohen P., Wang X., Yu S., Lo J.C., Tseng Y.H., et al. (2014). IRF4 is a key thermogenic transcriptional partner of PGC-1alpha. Cell 158, 69–83. 10.1016/j.cell.2014.04.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Rosenwald M., Perdikari A., Rulicke T., and Wolfrum C. (2013). Bi-directional interconversion of brite and white adipocytes. Nature cell biology 15, 659–667. 10.1038/ncb2740. [DOI] [PubMed] [Google Scholar]
  • 26.Jung S.M., Hung C.M., Hildebrand S.R., Sanchez-Gurmaches J., Martinez-Pastor B., Gengatharan J.M., Wallace M., Mukhopadhyay D., Martinez Calejman C., Luciano A.K., et al. (2019). Non-canonical mTORC2 Signaling Regulates Brown Adipocyte Lipid Catabolism through SIRT6-FoxO1. Molecular cell 75, 807–822.e808. 10.1016/j.molcel.2019.07.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Sanchez-Gurmaches J., Martinez Calejman C., Jung S.M., Li H., and Guertin D.A. (2019). Brown fat organogenesis and maintenance requires AKT1 and AKT2. Molecular metabolism 23, 60–74. 10.1016/j.molmet.2019.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Sanchez-Gurmaches J., Tang Y., Jespersen N.Z., Wallace M., Martinez Calejman C., Gujja S., Li H., Edwards Y.J.K., Wolfrum C., Metallo C.M., et al. (2018). Brown Fat AKT2 Is a Cold-Induced Kinase that Stimulates ChREBP-Mediated De Novo Lipogenesis to Optimize Fuel Storage and Thermogenesis. Cell metabolism 27, 195–209 e196. 10.1016/j.cmet.2017.10.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Lasar D., Rosenwald M., Kiehlmann E., Balaz M., Tall B., Opitz L., Lidell M.E., Zamboni N., Krznar P., Sun W., et al. (2018). Peroxisome Proliferator Activated Receptor Gamma Controls Mature Brown Adipocyte Inducibility through Glycerol Kinase. Cell reports 22, 760–773. 10.1016/j.celrep.2017.12.067. [DOI] [PubMed] [Google Scholar]
  • 30.Keinan O., Valentine J.M., Xiao H., Mahata S.K., Reilly S.M., Abu-Odeh M., Deluca J.H., Dadpey B., Cho L., Pan A., et al. (2021). Glycogen metabolism links glucose homeostasis to thermogenesis in adipocytes. Nature 599, 296–301. 10.1038/s41586-021-04019-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Chitraju C., Fischer A.W., Farese R.V. Jr., and Walther T.C. (2020). Lipid Droplets in Brown Adipose Tissue Are Dispensable for Cold-Induced Thermogenesis. Cell reports 33, 108348. 10.1016/j.celrep.2020.108348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Nayak G., Zhang K.X., Vemaraju S., Odaka Y., Buhr E.D., Holt-Jones A., Kernodle S., Smith A.N., Upton B.A., D’Souza S., et al. (2020). Adaptive Thermogenesis in Mice Is Enhanced by Opsin 3-Dependent Adipocyte Light Sensing. Cell reports 30, 672–686.e678. 10.1016/j.celrep.2019.12.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Liu L., Zou P., Zheng L., Linarelli L.E., Amarell S., Passaro A., Liu D., and Cheng Z. (2015). Tamoxifen reduces fat mass by boosting reactive oxygen species. Cell death & disease 6, e1586. 10.1038/cddis.2014.553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Ye R., Wang Q.A., Tao C., Vishvanath L., Shao M., McDonald J.G., Gupta R.K., and Scherer P.E. (2015). Impact of tamoxifen on adipocyte lineage tracing: Inducer of adipogenesis and prolonged nuclear translocation of Cre recombinase. Molecular metabolism 4, 771–778. 10.1016/j.molmet.2015.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Noden D.M., and Trainor P.A. (2005). Relations and interactions between cranial mesoderm and neural crest populations. J Anat 207, 575–601. 10.1111/j.1469-7580.2005.00473.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Jiang X., Iseki S., Maxson R.E., Sucov H.M., and Morriss-Kay G.M. (2002). Tissue origins and interactions in the mammalian skull vault. Developmental biology 241, 106–116. 10.1006/dbio.2001.0487. [DOI] [PubMed] [Google Scholar]
  • 37.de Vree P.J., de Wit E., Yilmaz M., van de Heijning M., Klous P., Verstegen M.J., Wan Y., Teunissen H., Krijger P.H., Geeven G., et al. (2014). Targeted sequencing by proximity ligation for comprehensive variant detection and local haplotyping. Nature biotechnology 32, 1019–1025. 10.1038/nbt.2959. [DOI] [PubMed] [Google Scholar]
  • 38.Chuang C.H., Wallace M.D., Abratte C., Southard T., and Schimenti J.C. (2010). Incremental genetic perturbations to MCM2–7 expression and subcellular distribution reveal exquisite sensitivity of mice to DNA replication stress. PLoS genetics 6, e1001110. 10.1371/journal.pgen.1001110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Hindson B.J., Ness K.D., Masquelier D.A., Belgrader P., Heredia N.J., Makarewicz A.J., Bright I.J., Lucero M.Y., Hiddessen A.L., Legler T.C., et al. (2011). High-throughput droplet digital PCR system for absolute quantitation of DNA copy number. Analytical chemistry 83, 8604–8610. 10.1021/ac202028g. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Skarnes W.C., Rosen B., West A.P., Koutsourakis M., Bushell W., Iyer V., Mujica A.O., Thomas M., Harrow J., Cox T., et al. (2011). A conditional knockout resource for the genome-wide study of mouse gene function. Nature 474, 337–342. 10.1038/nature10163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Claflin K.E., Flippo K.H., Sullivan A.I., Naber M.C., Zhou B., Neff T.J., Jensen-Cody S.O., and Potthoff M.J. (2022). Conditional gene targeting using UCP1-Cre mice directly targets the central nervous system beyond thermogenic adipose tissues. Molecular metabolism 55, 101405. 10.1016/j.molmet.2021.101405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Abel E.D., Peroni O., Kim J.K., Kim Y.B., Boss O., Hadro E., Minnemann T., Shulman G.I., and Kahn B.B. (2001). Adipose-selective targeting of the GLUT4 gene impairs insulin action in muscle and liver. Nature 409, 729–733. 10.1038/35055575. [DOI] [PubMed] [Google Scholar]
  • 43.He W., Barak Y., Hevener A., Olson P., Liao D., Le J., Nelson M., Ong E., Olefsky J.M., and Evans R.M. (2003). Adipose-specific peroxisome proliferator-activated receptor gamma knockout causes insulin resistance in fat and liver but not in muscle. Proc Natl Acad Sci U S A 100, 15712–15717. 10.1073/pnas.2536828100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Mullican S.E., Tomaru T., Gaddis C.A., Peed L.C., Sundaram A., and Lazar M.A. (2013). A novel adipose-specific gene deletion model demonstrates potential pitfalls of existing methods. Molecular endocrinology (Baltimore, Md.) 27, 127–134. 10.1210/me.2012-1267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Urs S., Harrington A., Liaw L., and Small D. (2006). Selective expression of an aP2/Fatty Acid Binding Protein 4-Cre transgene in non-adipogenic tissues during embryonic development. Transgenic research 15, 647–653. 10.1007/s11248-006-9000-z. [DOI] [PubMed] [Google Scholar]
  • 46.Lee K.Y., Russell S.J., Ussar S., Boucher J., Vernochet C., Mori M.A., Smyth G., Rourk M., Cederquist C., Rosen E.D., (2013). Lessons on conditional gene targeting in mouse adipose tissue. Diabetes 62, 864–874. 10.2337/db12-1089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Shan T., Liu W., and Kuang S. (2013). Fatty acid binding protein 4 expression marks a population of adipocyte progenitors in white and brown adipose tissues. FASEB journal : official publication of the Federation of American Societies for Experimental Biology 27, 277–287. 10.1096/fj.12-211516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Wang H., Willershauser M., Karlas A., Gorpas D., Reber J., Ntziachristos V., Maurer S., Fromme T., Li Y., and Klingenspor M. (2019). A dual Ucp1 reporter mouse model for imaging and quantitation of brown and brite fat recruitment. Molecular metabolism 20, 14–27. 10.1016/j.molmet.2018.11.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Perkins M.N., Rothwell N.J., Stock M.J., and Stone T.W. (1981). Activation of brown adipose tissue thermogenesis by the ventromedial hypothalamus. Nature 289, 401–402. 10.1038/289401a0. [DOI] [PubMed] [Google Scholar]
  • 50.Zhang K.X., D’Souza S., Upton B.A., Kernodle S., Vemaraju S., Nayak G., Gaitonde K.D., Holt A.L., Linne C.D., Smith A.N., et al. (2020). Violet-light suppression of thermogenesis by opsin 5 hypothalamic neurons. Nature 585, 420–425. 10.1038/s41586-020-2683-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Tran L.T., Park S., Kim S.K., Lee J.S., Kim K.W., and Kwon O. (2022). Hypothalamic control of energy expenditure and thermogenesis. Exp Mol Med 54, 358–369. 10.1038/s12276-022-00741-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Pomplun D., Florian S., Schulz T., Pfeiffer A.F., and Ristow M. (2007). Alterations of pancreatic beta-cell mass and islet number due to Ins2-controlled expression of Cre recombinase: RIP-Cre revisited; part 2. Horm Metab Res 39, 336–340. 10.1055/s-2007-976538. [DOI] [PubMed] [Google Scholar]
  • 53.Volovelsky O., Nguyen T., Jarmas A.E., Combes A.N., Wilson S.B., Little M.H., Witte D.P., Brunskill E.W., and Kopan R. (2018). Hamartin regulates cessation of mouse nephrogenesis independently of Mtor. Proc Natl Acad Sci U S A 115, 5998–6003. 10.1073/pnas.1712955115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Morimoto M., and Kopan R. (2009). rtTA toxicity limits the usefulness of the SP-C-rtTA transgenic mouse. Developmental biology 325, 171–178. 10.1016/j.ydbio.2008.10.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Wong A.M., Patel T.P., Altman E.K., Tugarinov N., Trivellin G., and Yanovski J.A. (2021). Characterization of the adiponectin promoter + Cre recombinase insertion in the Tg(Adipoq-cre)1Evdr mouse by targeted locus amplification and droplet digital PCR. Adipocyte 10, 21–27. 10.1080/21623945.2020.1861728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Farrar J.S., Lownik J.C., Way G.W., Rodriguez M.C., Celi F.S., and Martin R.K. (2021). Identification of the transgene insertion site for an adipocyte-specific adiponectin-cre model and characterization of the functional consequences. Adipocyte 10, 91–100. 10.1080/21623945.2021.1880083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Fan Y., Chen W., Wei R., Qiang W., Pearson J.D., Yu T., Bremner R., and Chen D. (2022). Mapping transgene insertion sites reveals the α-Cre transgene expression in both developing retina and olfactory neurons. Commun Biol 5, 411. 10.1038/s42003-022-03379-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Hinteregger B., Loeffler T., Flunkert S., Neddens J., Birner-Gruenberger R., Bayer T.A., Madl T., and Hutter-Paier B. (2020). Transgene integration causes RARB downregulation in homozygous Tg4–42 mice. Scientific reports 10, 6377. 10.1038/s41598-020-63512-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Amarasinghe S.L., Su S., Dong X., Zappia L., Ritchie M.E., and Gouil Q. (2020). Opportunities and challenges in long-read sequencing data analysis. Genome Biol 21, 30. 10.1186/s13059-020-1935-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Suzuki O., Koura M., Uchio-Yamada K., and Sasaki M. (2020). Analysis of the transgene insertion pattern in a transgenic mouse strain using long-read sequencing. Exp Anim 69, 279–286. 10.1538/expanim.19-0118. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplement 1

Articles from bioRxiv are provided here courtesy of Cold Spring Harbor Laboratory Preprints

RESOURCES