Gene transfer using viral or nonviral vectors enables the ability to manipulate specific cells and tissues for gene silencing, protein overexpression, or genome modification. Despite the widespread application of viral- and non-viral-mediated gene transfer to liver, heart, skeletal muscle, and the central nervous system, its use in adipose tissue has been limited. This is largely because adipose tissue is distributed throughout the body in distinct depots and adipocytes make up a minority of the cells within the tissue, making transduction difficult. Currently, there is no consensus methodology for efficient gene transfer to adipose tissue and many studies report conflicting information with regard to transduction efficiency and vector biodistribution. In this review, we summarize the challenges associated with gene transfer to adipose tissue and report on innovations that improve efficacy. We describe how vector and route of administration are the two key factors that influence transduction efficiency and outline a “gold standard” approach and experimental workflow for validating gene transfer to adipose tissue. Lastly, we speculate on how CRISPR/Cas9 can be integrated to improve adipose tissue research.

Viral and Nonviral Gene Transfer to Adipose Tissue

Introduction to Gene Transfer

Our understanding of physiology is heightened by our ability to manipulate cells and tissues. Through gene transfer, we can deliver genetic agents for gene silencing, protein overexpression, or genome modification to further our understanding of biological phenomena. To accomplish gene transfer in a tissue-specific manner, one must consider two critical components: the vector and the route of administration. An ideal vector shields its cargo from degradation, targets a specific tissue, facilitates unloading of its cargo into cells, and evades the host immune response. Both viral and nonviral vectors meet these criteria (13). Secondly, to ensure the vector reaches its desired destination, one must determine an appropriate route of administration. In general, vectors are administered systemically or locally, depending on the tissue of interest. Gene transfer has largely been aimed at the liver, heart, skeletal muscles, and central nervous system; however, its application to adipose tissue has been met with limited success. Collectively, adipose tissues comprise the largest endocrine organ in the body with important regulatory functions for energy balance and metabolism, thus making it an attractive focal point for understanding obesity and the metabolic syndrome. In this section, we will outline how both vector and route of administration influence transduction efficiency. By careful optimization of these two factors, gene transfer can be accomplished in an adipose-specific fashion.

Targeting Adipose Tissue: Challenges and Limitations

Adipocytes are located throughout the body mainly in subcutaneous, visceral, and marrow depots as white (WAT), brown (BAT), or more specialized adipose tissues (46). At the cellular level, mature adipocytes comprise only 11–40% of the total cell population in adipose tissue, which also contains preadipocytes, stem cells, macrophages, endothelial cells, and vasculature, collectively referred to as stromal vascular cells (7). WAT constitutes the bulk of adipose tissue and is found in subcutaneous and visceral depots. In mice, the subcutaneous WAT includes the interscapular, subscapular, axillary, and cervical depots in the anterior and the dorso-lumbar, inguinal, and gluteal depots in the posterior. Subcutaneous WAT are also closely associated with lymphatic tissues and in females comprise part of the mammary glands. Visceral adipocytes are contained within the peritoneal cavity and include the mesenteric, perirenal, retroperitoneal, and gonadal (epididymal/ovarian) depots. An inability to expand these depots to store excess energy is typically associated with insulin resistance and metabolic dysfunction (5,6). Additionally, there are several smaller depots including the intramuscular, periarticular, paracardial, epicardial, retro-orbital, and dermal WAT. By contrast, BAT is predominantly found in the interscapular depot but also in the periaortic, perirenal, and intercostal depots (8). Brown adipocytes are characterized by a multilocular lipid droplet morphology and are rich in mitochondria (46,8). In response to temperatures below thermoneutrality (lower than ∼30°C in mice), brown adipocytes express uncoupling protein 1 (UCP1), a key mechanism for adaptive nonshivering thermogenesis (8). Although much smaller in terms of adipose tissue mass, BAT metabolism is the predominant energy consumer in mice at cold temperatures, making it an attractive therapeutic target for treating obesity (8,9).

Given the diversity and distribution of adipose tissue, gene transfer has remained challenging. In vivo manipulation has been limited to the generation of Cre-Lox and/or drug-inducible transgenic models. Cre-Lox systems enable the expression of Cre recombinase under the control of adipocyte-specific promoters. Several adipocyte-specific promoters including fatty acid binding protein-4 (FABP4) and adiponectin (AdipoQ) have been developed (10). Although both have been used extensively, FABP4-Cre activity has been detected in the brain, endothelial cells, macrophages, adipocyte progenitors, and embryonic tissues (1113). Thus, the field has shifted to conditional (AdipoQ-Cre) or inducible (AdipoQ-CreERT, AdipoChaser) lines (1315). While useful, Cre mice must be bred to transgenic mice harboring a floxed gene. In some cases, a new floxed line must also be generated, which is time-consuming and expensive and involves large cohorts of animals. Therefore, more efficient strategies, such as gene transfer, would be beneficial to the field.

Step 1: Selection of an Appropriate Vector

Adipose tissue is very heterogenous, and in many cases, vectors transduce stromal vascular cells more efficiently than the adipocytes (7). Thus, a vector must successfully navigate the adipose tissue microenvironment to home to the adipocytes. Important considerations in selection of a vector should include cargo, tropism, expression profile, and immune response (Table 1). In this section, we will discuss advancements using viral and nonviral vectors for transducing adipocytes.

Table 1

Viral and nonviral vector properties and affinity for adipose tissue

VectorCargoGenome size (kb)Affinity for adipose tissueExpression profileImmune responseGenome integrationReferences
Lentivirus ssRNA 9.7 Adipocytes, stromal vascular cells, cultured preadipocytes and adipocytes Stable (permanent) Low Yes 16,17,49  
Adenovirus dsDNA 26–46 Transient (weeks) High No 1826  
AAV ssDNA 4.7 Transient (months to years) Very low Rare 2730,41,42,4448  
Nonviral DNA, mRNA, shRNA, protein Not restricted Stromal vascular cells, cultured preadipocytes Transient (hours to days) Low to moderate No 3237,50,6062  
VectorCargoGenome size (kb)Affinity for adipose tissueExpression profileImmune responseGenome integrationReferences
Lentivirus ssRNA 9.7 Adipocytes, stromal vascular cells, cultured preadipocytes and adipocytes Stable (permanent) Low Yes 16,17,49  
Adenovirus dsDNA 26–46 Transient (weeks) High No 1826  
AAV ssDNA 4.7 Transient (months to years) Very low Rare 2730,41,42,4448  
Nonviral DNA, mRNA, shRNA, protein Not restricted Stromal vascular cells, cultured preadipocytes Transient (hours to days) Low to moderate No 3237,50,6062  

Viral vectors including lentivirus, adenovirus, and AAV can transduce adipose tissues as well as cultured preadipocytes and adipocytes. Of the classes of viruses, AAV has been the most widely used owing to its tropism, expression profile, and low immune response. Nonviral vectors including organic and inorganic nanoparticles have been shown to transduce adipose tissue vasculature and macrophages in addition to cultured preadipocytes. Further research is required to confirm nonviral transduction of adipocytes in vivo. dsDNA, double-stranded DNA; ssRNA, single-stranded RNA.

Viral Vectors.

Different classes of viruses, including lentivirus, adenovirus, and adeno-associated virus (AAV), have all been used for gene transfer to adipose tissue. While lentiviruses and adenoviruses efficiently transduce adipocytes in vitro, efforts to mediate gene transfer in vivo have been less successful. For example, lentiviruses target CD4+ cells and bind to the human adenovirus C receptor, which is not expressed by adipocytes (16,17). Moreover, lentiviruses integrate into the genome leading to insertional mutagenesis (16). While several studies using adenovirus have been performed, concerns regarding its strong immunologic response and generation of antibodies against its capsid proteins have caused the field to shift to AAV (1826). AAV is characterized by low immunogenicity, epichromosomal expression, and a multitude of serotypes and capsid variants (27).

Seminal work from the Bosch laboratory demonstrated that AAV8 and AAV9 preferentially transduce white and brown adipocytes with variable transduction efficiencies across depots (28). Following systemic delivery, AAV8 and AAV9 GFP were highly expressed in the epididymal WAT and interscapular BAT but marginally expressed in the inguinal, retroperitoneal, and mesenteric depots. Interestingly, variability in GFP expression was observed across different strains of mice including C57/BL/6J, ICR, ob/ob, and db/db, suggesting that genetic background may influence transduction efficiency (28,29). For improvement of tropism, recombinant AAV (rAAVs) have been engineered by capsid shuffling to increase affinity for adipose tissue. For example, Liu et al. (30) generated rAAVs harboring GFP (Rec2-GFP) and demonstrated increased adipocyte transduction in comparison with the natural AAV serotypes 1, 8, and 9. Rec2 vectors were outfitted with Cre recombinase and injected into the inguinal WAT depots of floxed–insulin receptor mice, which led to ∼53% knockout of the insulin receptor and 50% decrease in inguinal WAT mass. Interestingly, these findings are in contrast to AdipoQ-Cre–mediated knockout mice, which develop lipodystrophy and metabolic dysfunction characterized by insulin resistance and hyperglycemia (31). This might be explained by the fact that Rec2-Cre knockout occurs in the adult mouse, whereas AdipoQ-Cre causes recombination early in adipocyte development.

Nonviral Vectors.

As an alternative approach, nonviral vectors, including organic and inorganic nanoparticles, have also been explored for gene transfer to adipose tissue. Nonviral nanoscale vectors (<200 nm in diameter) have unique characteristics that make them attractive targets for gene transfer and pharmacologic applications. For example, nonviral vectors can be functionalized with macromolecules to impart tissue specificity (2,3). Efforts to impart adipose specificity have largely focused on conjugating nonviral vectors with the peptide sequence CKGGRAKDC (adipocyte-targeting sequence [ATS]), which binds to the prohibitin receptor on the subcutaneous WAT vasculature (32). Hossen et al. (33) formulated ATS-conjugated nanoparticles that accumulated in the adipose tissue vasculature of mice with diet-induced obesity. In another study, cadmium-based quantum dots functionalized with ATS targeted the WAT of obese Wistar rats (34). While the mechanism by which ATS-based vectors escape the vasculature to transduce adipocytes remains unknown, research has shown that targeting adipose tissue vasculature directly can influence the surrounding tissue. For example, Xue et al. (35) developed polymer-based nanoparticles conjugated to ATS and loaded with rosiglitazone or prostaglandin E2 analog. In treated obese mice, WAT vascular density was increased coupled with the upregulation of the BAT markers Ucp1, Cidea, and Dio2. Thus, modulation of adipose tissues can be achieved by targeting the vasculature.

Modulation of adipose tissue can also be achieved by targeting the resident macrophages to suppress obesity-induced inflammation. In the obese state, endogenous low-molecular-weight hyaluronic acid binds to CD44-expressing cells to trigger inflammation and insulin resistance (36). Rho et al. (36) developed hyaluronic acid nanoparticles (HA-NPs) to block this pathway and reduce inflammation. HA-NP–treated obese mice had significant reductions in body weight, glucose tolerance, and insulin resistance. Additionally, HA-NPs reduced macrophage infiltration and production of inflammatory cytokines in epididymal WAT, likely by outcompeting low-molecular-weight hyaluronic acid for CD44 binding sites. It should be emphasized, however, that HA-NPs accumulated predominantly in liver, lungs, and kidneys, with only marginal transduction observed in the epididymal, subcutaneous, and brown depots. In another study, Ma et al. (37) synthesized dextran conjugates loaded with dexamethasone to reduce inflammation in obese mice. Impressively, a single dose reduced the expression of TNFα, IL-6, and MCP-1 in the gonadal, perirenal, mesenteric, and subcutaneous depots. Although these studies do not involve gene transfer per se, it can be reasoned that genetic cargo can be incorporated into these nonviral vectors. In fact, several nonviral nanoscale vectors have entered clinical trials for the delivery of siRNA to the vasculature (38). Thus, future strategies may involve non-viral-mediated gene transfer to adipocytes, vasculature, or macrophages.

Insights Into Targeted Vector Design.

Although both viral and nonviral vectors have been investigated (Fig. 1), there is no consensus method for transducing adipose tissue largely because adipocyte specificity remains a major limitation. Therefore, future success will depend on creating targeted vectors that enhance tissue tropism and impart tissue-specific transcriptional control. For improvement of tissue tropism, viral envelope glycoproteins have been modified with ligands, peptides, and antibodies. Yang et al. (39) created an efficient method that incorporates antibodies onto the viral surface and enables the virus to bind to a specific cell type and deliver its transgene. While this method has not been used to transduce adipocytes directly, one can envision the use of adipocyte-specific antibodies against adipocyte-selective targets such as the amino acid transporter ASC-1 or the brown cell surface markers PAT2 or P2RK5 to improve tropism (40). Adipose-specific promoters such as AdipoQ or UCP1 can limit off-target transgene expression to impart transcriptional control (10,13). It should be noted that the large size of the AdipoQ locus required for adipocyte specificity restricts its use with AAV, although “mini” promoters, such as mini/UCP1, have been implemented (28). Another strategy uses tissue-specific miRNAs targeting posttranscriptional regulatory elements of the transgene. For example, Jimenez et al. (28) incorporated multiple miRNAs to prevent transgene expression in liver and heart but retained transgene expression in WAT and BAT, respectively. Thus, targeted vector design that enhances tropism and imparts adipose-selective transcriptional control can significantly improve transduction efficiency and limit off-target expression. A combination of these strategies will likely emerge as the consensus method for gene transfer to adipose tissue. It should be emphasized that careful consideration should be paid to validating transduction efficiency and selectivity, either by incorporating a fluorescent probe or by separating the adipocytes from stromal vascular cells. We will now turn our attention to the second critical component of delivery: route of administration.

Figure 1

Viral and nonviral vectors have been used for transducing adipose tissue. Adipose tissue is very heterogenous, and adipocytes represent a minority of cells in adipose tissue. Thus, a vector must successfully navigate through a variety of cell types to home to the target cell. Viral vectors have been used to transduce adipocytes. Nonviral vectors have been used to transduce the adipose tissue vasculature and resident macrophages to elicit physiological changes to the tissue.

Figure 1

Viral and nonviral vectors have been used for transducing adipose tissue. Adipose tissue is very heterogenous, and adipocytes represent a minority of cells in adipose tissue. Thus, a vector must successfully navigate through a variety of cell types to home to the target cell. Viral vectors have been used to transduce adipocytes. Nonviral vectors have been used to transduce the adipose tissue vasculature and resident macrophages to elicit physiological changes to the tissue.

Close modal

Step 2: Optimizing the Route of Administration

Generally speaking, vector administration can proceed via systemic or local delivery (Fig. 2). Ideally, a vector should transduce every adipocyte across all adipose depots, although this goal has never been fully realized. Therefore, in selection of a route one should carefully consider the experimental aims, anatomical location, degree of vascularization, and invasiveness in order to optimize transduction efficiency.

Figure 2

Adipocyte transduction efficiency is highly dependent on route of administration. Gene transfer to adipose tissue has proceeded through both systemic and local delivery of viral or nonviral vectors. Systemic delivery via intravenous injection, intraperitoneal injection, or oral gavage accumulates largely in the liver and reticuloendothelial organs even when vectors are outfitted with transcriptional control features. By contrast, local delivery via intra-WAT or intra-BAT injection retains the vector at the site of the injection with minimal leak to the liver. Arrow size indicates degree of vector distribution.

Figure 2

Adipocyte transduction efficiency is highly dependent on route of administration. Gene transfer to adipose tissue has proceeded through both systemic and local delivery of viral or nonviral vectors. Systemic delivery via intravenous injection, intraperitoneal injection, or oral gavage accumulates largely in the liver and reticuloendothelial organs even when vectors are outfitted with transcriptional control features. By contrast, local delivery via intra-WAT or intra-BAT injection retains the vector at the site of the injection with minimal leak to the liver. Arrow size indicates degree of vector distribution.

Close modal
Systemic Delivery via Intravenous Injection.

Intravenous injection capitalizes on the circulatory system’s direct access to all tissues in the body. Despite this, the varying degree of vascularization in each adipose depot coupled with vector accumulation in reticuloendothelial organs (liver, lung, heart, and kidney) limits adipocyte transduction efficiency (1,2,5,6). Successful gene transfer using intravenous injection via the tail vein has only been accomplished with adenovirus or AAV; however, the field has since shifted exclusively to AAV because of its favorable properties. For example, O’Neill et al. (41) intravenously injected AAV2/8 encoding leptin to leptin-deficient mice. A single injection increased plasma leptin levels to 7% of that observed in wild-type mice, which led to decreased body weight, food intake, and fasting insulin levels and improved glucose tolerance. In another study, rAAV8 vectors expressing perilipin A were intravenously injected into mice (42). Treated mice showed elevated expression of perilipin A in visceral and subcutaneous WAT and the liver with a decrease in serum free fatty acids. Interestingly, however, both of these studies reported transgene expression in reticuloendothelial organs despite having transcriptional control features, thus highlighting the off-target issues associated with systemic delivery.

Systemic Delivery via Intraperitoneal Injection.

Intraperitoneal injection can be rapidly accomplished in conscious animals with minimal restraint. Vectors injected intraperitoneally are primarily absorbed into the mesenteric vessels that drain into the portal vein and pass through the liver (43). Huang et al. (44) injected mice with Rec2-GFP vectors encoding a liver-specific miRNA to prevent off-target expression. Substantial GFP expression was observed in the epididymal WAT but not in other adipose tissues examined or in reticuloendothelial organs. GFP was exchanged for leptin and injected into leptin-deficient mice, which increased lean mass and circulating leptin levels, improved glucose tolerance, and reversed hepatic steatosis. An important caveat to this study is that vectors transduced only one adipose depot despite being injected via a systemic route.

Systemic Delivery via Oral Gavage.

Enteral delivery through oral gavage is economical, convenient, and relatively safe and requires moderate technical skill (43). In a proof of concept study, Huang et al. (45) examined the effect of oral gavage on the biodistribution of Rec2 vectors. Interestingly, Rec2-GFP expression was only detected in the BAT, inguinal WAT, and liver and not in the stomach, intestine, heart, kidney, or brain. Rec2-Cre vectors were orally administered to floxed-VEGF mice for targeted deletion in BAT. Cold exposure (4°C for 4 h) led to an inability to defend core body temperature, decreased BAT mass, and VEGF protein depletion by ∼80%. VEGF was then overexpressed with use of orally administered Rec2, which led to an increase in BAT mass and VEGF, UCP1, and CD31 protein levels. The potential mechanism by which Rec2 bypasses the gastrointestinal system to preferentially transduce BAT has not been described; however, the authors postulate that the lymphatic system may play a role, as Rec2 was detected in the mesenteric adipose depot, which is rich in lymph nodes.

Local Delivery via Intradepot Injection.

In contrast to systemic delivery, local delivery is a more targeted approach in which vectors are administered directly into a tissue. Although slightly more invasive, time-consuming, and resource heavy, local delivery via intradepot injection retains vectors at the site of injection, enabling the study of depot-specific function (27,43,46). For adipose tissue, intradepot injection has proceeded through intra-WAT or intra-BAT injection of viral and nonviral vectors. Before attempting intradepot injection, we recommend reviewing references (46,27,46) for guidance on how to access these tissues.

Local Delivery via Intra-WAT Injection.

Mizukami et al. (47) were the first group to optimize intra-WAT injection by administering AAV1-erythropoietin into the subcutaneous WAT. Treated mice showed elevated plasma erythropoietin concentration compared with controls. The transduced subcutaneous WAT was then surgically removed, causing plasma erythropoietin concentrations to return to baseline, indicating that increased circulating erythropoietin was the result of gene transfer to WAT specifically. Work from the Bosch laboratory explored AAV-mediated gene transfer to overexpress fibroblast growth factor 21 as a therapeutic strategy for treating obesity (48). AAV8 that expressed fibroblast growth factor 21 was administered directly into liver, epididymal WAT, or skeletal muscle of obese mice. Interestingly, a single injection into the epididymal WAT led to a dose-dependent reduction in adipocyte size, improvement in insulin sensitivity, and prevention of hepatic steatosis, results that were comparable with those of both liver- and skeletal muscle–specific injections. Collectively, these studies demonstrate that a single dose of AAV to one adipose depot is sufficient to mediate systemic changes to metabolism.

Lentiviruses have also been used for intra-WAT gene transfer applications. Gnad et al. (49) injected a lentivirus expressing the A2A receptor into the inguinal WAT of obese mice as an antiobesity therapy. Stimulation of sympathetic drive causes BAT to release adenosine, which binds to the A2A receptor to increase energy expenditure. It was hypothesized that overexpression of the A2A receptor in inguinal WAT, which has the capacity for browning, might have therapeutic potential. Interestingly, lentiviral expression of the A2A receptor significantly increased thermogenic and lipogenic markers and led to a brown adipocyte–like morphology in the inguinal WAT with increased expression of Ucp1, Pgc1α, and Prdm16.

Only one study has attempted intra-WAT injection of a nonviral vector for gene transfer. The Kim laboratory synthesized an ATS-oligopeptide complex to administer shRNA to silence fatty acid binding protein-4 (FABP4) and combat obesity (50). Obese mice were administered the complex via intra-WAT injection on one side of the “abdominal” adipose tissue. Strikingly, FABP4 expression was decreased by 70% compared with the contralateral depot. Treated mice also showed significant improvements in insulin sensitivity and reduced blood glucose levels. Perhaps the most surprising data are that treated mice lost nearly 20% of their body weight following a single administration to one WAT depot. While these data are interesting, they also raise several red flags. Firstly, the mechanism by which the ATS-oligopeptide-shRNA complex binds to prohibitin on adipose tissue vasculature and escapes to transduce the adipocytes has not been explained. Secondly, the authors do not report changes in energy balance, lipolysis, inflammation, apoptosis, or cell death or changes in tissue mass in the injected WAT depot, leading us to question how silencing FABP4 in one adipose tissue equates to a 20% reduction in body weight. This study is also in direct contradiction with previous work that shows FABP4 knockdown or knockout in obese mice increases body weight and fat mass without changes in glucose homeostasis (51,52). Moreover, it does not report on FABP5 expression, which is compensatory to loss of FABP4 (53).

Local Delivery via Intra-BAT Injection.

Similar to intra-WAT injection, AAV has remained the most commonly used vector for intra-BAT injection. Two aforementioned studies attempted intra-BAT injection using AAVs. Jimenez et al. (28) showed that intra-BAT injection of AAV9 encoding VEGF164 under the control of the mini/UCP1 promoter increased vessel density and VEGF164 expression specifically in BAT. In the second study, Liu et al. (30) administered Rec2-Cre bilaterally into the BAT of floxed–insulin receptor mice, which decreased BAT size by 50% and expression of insulin receptor protein by ∼60%. Surprisingly, disruption of insulin receptor signaling in BAT induced gene expression changes in the hypothalamus, highlighting that depot-specific gene transfer can be used to study the interaction between adipose tissue and the central nervous system.

The “Gold Standard” Approach for Adipose Tissue Gene Transfer

Although significant progress has been made using viral and nonviral vectors, the field lacks a gold standard approach. Experimental aims will always dictate the selection of a vector and route of administration; however, to guide the reader we propose the following set of guidelines:

  • 1) Transcriptionally controlled AAVs currently show the most promise for transducing adipocytes in adipose tissues. Researchers should prioritize rAAV, AAV8, or AAV9 that incorporates an adipose-specific promoter–driven transgene (mini/AdipoQ or mini/UCP1), an miRNA to limit transgene expression in off-target tissues, and a reporter molecule to validate transduction efficiency.

  • 2) To maximize adipocyte transduction efficiency and limit accumulation in off-target tissues, researchers should administer vectors locally via intra-WAT or intra-BAT injection.

  • 3) For validation of your experiment, follow the gold standard approach outlined in Fig. 3. For validation of adipocyte transduction, vectors should be outfitted with fluorescent probes or administered to reporter lines such as the mT/mG mouse (54). Successful transduction is characterized by a fluorescent “halo” surrounding the adipocyte. As adipocytes represent only a small fraction of the total cells in adipose tissue, they should be separated from stromal vascular cells by buoyant-density centrifugation, and both fractions should be analyzed for transduction.

Figure 3

Our proposed “gold standard” approach for validation of adipose tissue gene transfer. For maximal transduction efficiency and minimal accumulation in off-target tissues, vectors (preferably AAV) should be administered locally through intradepot injection. The most commonly targeted adipose depots are the interscapular BAT, the posterior subcutaneous WAT, and the gonadal WAT. For validation of successful gene transfer, AAV vectors should be outfitted with a reporter such as a fluorescent protein and/or Cre recombinase. Dissected tissues should be imaged or fixed and stained to confirm adipocyte transduction. In this example, mT/mG mice were administered AAV8-Cre via local injection into the posterior subcutaneous WAT. GFP expression is indicative of successful gene transfer and expression of Cre within the adipocytes. Adipose tissue should then be digested and fractionated to separate adipocytes from stromal vascular cells. The isolated adipocytes and stromal vascular cells can then be analyzed for specificity of transgene expression or cultured for further analyses.

Figure 3

Our proposed “gold standard” approach for validation of adipose tissue gene transfer. For maximal transduction efficiency and minimal accumulation in off-target tissues, vectors (preferably AAV) should be administered locally through intradepot injection. The most commonly targeted adipose depots are the interscapular BAT, the posterior subcutaneous WAT, and the gonadal WAT. For validation of successful gene transfer, AAV vectors should be outfitted with a reporter such as a fluorescent protein and/or Cre recombinase. Dissected tissues should be imaged or fixed and stained to confirm adipocyte transduction. In this example, mT/mG mice were administered AAV8-Cre via local injection into the posterior subcutaneous WAT. GFP expression is indicative of successful gene transfer and expression of Cre within the adipocytes. Adipose tissue should then be digested and fractionated to separate adipocytes from stromal vascular cells. The isolated adipocytes and stromal vascular cells can then be analyzed for specificity of transgene expression or cultured for further analyses.

Close modal

While we have focused on transducing adipocytes, it should be noted that targeting the vasculature or resident macrophages is an alternative strategy for manipulating adipose tissues. This is particularly true for nonviral vectors, although continued research is required.

CRISPR/Cas9: The Future of Adipose Tissue Gene Transfer

CRISPR/Cas9 Gene Transfer in Adipose Tissue

With the emergence of powerful gene-editing technologies like CRISPR/Cas9, researchers can modify virtually any region of the genome. CRISPR/Cas9 is composed of two components: the Cas9 endonuclease and a programmable single-guide RNA (sgRNA) that directs Cas9 where to create double-strand breaks in genomic DNA (55). CRISPR/Cas9 is thus easily amendable to viral or nonviral gene transfer, as its constitutive parts can be genetically encoded. Despite the adoption of CRISPR/Cas9 gene transfer to a variety of tissues (5659), only one study reports its use in adipose tissue (60). We recommend caution, however, as this study uses the ATS-oligopeptide complex described above for CRISPR-interference silencing of FABP4 (50,60).

Two studies have recently reported on in vitro delivery of CRISPR/Cas9 to cultured adipocyte precursors. The Czech laboratory successfully transduced isolated primary preadipocytes with nonviral vectors loaded with Cas9 protein and an sgRNA targeted to Nrip1 (61). Treated cells showed a mutation frequency of 43.8% in the Nrip1 locus coupled with increased expression of the brown-like genes Ucp1, Cidea, Pgc1α, Prdm16, and Cpt1b. Importantly, no off-target mutations were detected and CRISPR/Cas9 did not affect the capacity to differentiate. In a second study, Kamble et al. (62) electroporated human preadipocytes with a ribonucleoprotein complex of Cas9 and an sgRNA targeted to Pparg or Fkbp5. Impressively, treated cells achieved 90% efficiency and did not require selection or clonal isolation of edited cells. While these studies demonstrate in vitro proof of principle, the field has yet to report successful in vivo gene transfer of CRISPR/Cas9 to adipose tissue.

The Future of Adipose Tissue Genome Editing

CRISPR/Cas9 gene transfer to adipose tissue is in its infancy; however, we can gain several insights from its use in other tissues and apply these to the gold standard approach. For example, CRISPR/Cas9 gene transfer can proceed via exogenous expression or a combination of exogenous/endogenous expression of its components. In the exogenous method, both Cas9 and sgRNA are delivered via a vector. In the exogenous/endogenous method, the sgRNA is delivered to a Cas9-expressing mouse line (63,64). Thus, rAAVs can be encoded with one or multiple sgRNAs and administered locally to AdipoQ-Cre–dependent Cas9 mice to enable spatial control (63). Cas9 expression can also be temporally regulated using a doxycycline-inducible line developed by Dow et al. (64). Ultimately, CRISPR/Cas9 has immense potential for elucidating the underlying mechanisms to a myriad of metabolic diseases. For instance, future application of CRISPR/Cas9 might entail homology-directed repair to correct single nucleotide polymorphisms associated with aberrant metabolism (65). Particular focus should be paid to integrating CRISPR/Cas9 with well-established gene transfer techniques such as AAV and Cre-Lox mouse models (63,64). Moreover, we also recommend a strong adherence to the gold standard approach to validate transduction efficiency. With these powerful technologies in hand, the field of adipose tissue biology is poised to enter a new era of research.

Acknowledgments. The authors thank members of the MacDougald laboratory for their helpful suggestions and review of the manuscript.

Funding. This review was supported by grants from the National Institutes of Health to O.A.M. (R24 DK092759, R01 DK121759, and R01 DK125513) and S.M.R. (T32 GM835326 and F31 DK12272301).

Duality of Interest. No potential conflicts of interest relevant to this article were reported.

Author Contributions. S.M.R. researched and wrote the manuscript. O.A.M. provided scientific guidance and editorial assistance.

1.
Mingozzi
F
,
High
KA
.
Therapeutic in vivo gene transfer for genetic disease using AAV: progress and challenges
.
Nat Rev Genet
2011
;
12
:
341
355
2.
Nayerossadat
N
,
Maedeh
T
,
Ali
PA
.
Viral and nonviral delivery systems for gene delivery
.
Adv Biomed Res
2012
;
1
:
27
3.
Yang
N
.
An overview of viral and nonviral delivery systems for microRNA
.
Int J Pharm Investig
2015
;
5
:
179
181
4.
Bagchi
DP
,
Forss
I
,
Mandrup
S
,
MacDougald
OA
.
SnapShot: niche determines adipocyte character I
.
Cell Metab
2018
;
27
:
264
264.e1
5.
Bagchi
DP
,
Forss
I
,
Mandrup
S
,
MacDougald
OA
.
SnapShot: niche determines adipocyte character II
.
Cell Metab
2018
;
27
:
266
266.e1
6.
Bagchi
DP
,
MacDougald
OA
. Identification and
dissection of diverse mouse adipose depots
.
J Vis Exp
2019
;
149
:
10.3791/59499
7.
Parlee
SD
,
Lentz
SI
,
Mori
H
,
MacDougald
OA
.
Quantifying size and number of adipocytes in adipose tissue
.
Methods Enzymol
2014
;
537
:
93
122
8.
Cannon
B
,
Nedergaard
J
.
Brown adipose tissue: function and physiological significance
.
Physiol Rev
2004
;
84
:
277
359
9.
Chechi
K
,
Carpentier
AC
,
Richard
D
.
Understanding the brown adipocyte as a contributor to energy homeostasis
.
Trends Endocrinol Metab
2013
;
24
:
408
420
10.
Jeffery
E
,
Berry
R
,
Church
CD
, et al
.
Characterization of Cre recombinase models for the study of adipose tissue
.
Adipocyte
2014
;
3
:
206
211
11.
Mullican
SE
,
Tomaru
T
,
Gaddis
CA
,
Peed
LC
,
Sundaram
A
,
Lazar
MA
.
A novel adipose-specific gene deletion model demonstrates potential pitfalls of existing methods
.
Mol Endocrinol
2013
;
27
:
127
134
12.
Urs
S
,
Harrington
A
,
Liaw
L
,
Small
D
.
Selective expression of an aP2/fatty acid binding protein 4-Cre transgene in non-adipogenic tissues during embryonic development
.
Transgenic Res
2006
;
15
:
647
653
13.
Lee
KY
,
Russell
SJ
,
Ussar
S
, et al
.
Lessons on conditional gene targeting in mouse adipose tissue
.
Diabetes
2013
;
62
:
864
874
14.
Eguchi
J
,
Wang
X
,
Yu
S
, et al
.
Transcriptional control of adipose lipid handling by IRF4
.
Cell Metab
2011
;
13
:
249
259
15.
Wang
QA
,
Scherer
PE
.
The AdipoChaser mouse: a model tracking adipogenesis in vivo
.
Adipocyte
2014
;
3
:
146
150
16.
Quinonez
R
,
Sutton
RE
.
Lentiviral vectors for gene delivery into cells
.
DNA Cell Biol
2002
;
21
:
937
951
17.
Kobinger
GP
,
Weiner
DJ
,
Yu
QC
,
Wilson
JM
.
Filovirus-pseudotyped lentiviral vector can efficiently and stably transduce airway epithelia in vivo
.
Nat Biotechnol
2001
;
19
:
225
230
18.
Wold
WS
,
Toth
K
.
Adenovirus vectors for gene therapy, vaccination and cancer gene therapy
.
Curr Gene Ther
2013
;
13
:
421
433
19.
Yang
Y
,
Nunes
FA
,
Berencsi
K
,
Furth
EE
,
Gönczöl
E
,
Wilson
JM
.
Cellular immunity to viral antigens limits E1-deleted adenoviruses for gene therapy
.
Proc Natl Acad Sci U S A
1994
;
91
:
4407
4411
20.
Yang
Y
,
Ertl
HC
,
Wilson
JM
.
MHC class I-restricted cytotoxic T lymphocytes to viral antigens destroy hepatocytes in mice infected with E1-deleted recombinant adenoviruses
.
Immunity
1994
;
1
:
433
442
21.
Muzzin
P
,
Eisensmith
RC
,
Copeland
KC
,
Woo
SL
.
Correction of obesity and diabetes in genetically obese mice by leptin gene therapy
.
Proc Natl Acad Sci U S A
1996
;
93
:
14804
14808
22.
Lee
S
,
Zhang
H
,
Chen
J
,
Dellsperger
KC
,
Hill
MA
,
Zhang
C
.
Adiponectin abates diabetes-induced endothelial dysfunction by suppressing oxidative stress, adhesion molecules, and inflammation in type 2 diabetic mice
.
Am J Physiol Heart Circ Physiol
2012
;
303
:
H106
H115
23.
Lo
JC
,
Ljubicic
S
,
Leibiger
B
, et al
.
Adipsin is an adipokine that improves β cell function in diabetes
.
Cell
2014
;
158
:
41
53
24.
Boström
P
,
Wu
J
,
Jedrychowski
MP
, et al
.
A PGC1-α-dependent myokine that drives brown-fat-like development of white fat and thermogenesis
.
Nature
2012
;
481
:
463
468
25.
Rao
RR
,
Long
JZ
,
White
JP
, et al
.
Meteorin-like is a hormone that regulates immune-adipose interactions to increase beige fat thermogenesis
.
Cell
2014
;
157
:
1279
1291
26.
Svensson
KJ
,
Long
JZ
,
Jedrychowski
MP
, et al
.
A secreted Slit2 fragment regulates adipose tissue thermogenesis and metabolic function
.
Cell Metab
2016
;
23
:
454
466
27.
Huang
W
,
Queen
NJ
,
Cao
L
.
rAAV-mediated gene delivery to adipose tissue
.
Methods Mol Biol
2019
;
1950
:
389
405
28.
Jimenez
V
,
Muñoz
S
,
Casana
E
, et al
.
In vivo adeno-associated viral vector-mediated genetic engineering of white and brown adipose tissue in adult mice
.
Diabetes
2013
;
62
:
4012
4022
29.
Zincarelli
C
,
Soltys
S
,
Rengo
G
,
Rabinowitz
JE
.
Analysis of AAV serotypes 1-9 mediated gene expression and tropism in mice after systemic injection
.
Mol Ther
2008
;
16
:
1073
1080
30.
Liu
X
,
Magee
D
,
Wang
C
, et al
.
Adipose tissue insulin receptor knockdown via a new primate-derived hybrid recombinant AAV serotype
.
Mol Ther Methods Clin Dev
2014
;
1
:
8
31.
Qiang
G
,
Whang Kong
H
,
Xu
S
, et al
.
Lipodystrophy and severe metabolic dysfunction in mice with adipose tissue-specific insulin receptor ablation
.
Mol Metab
2016
;
5
:
480
490
32.
Kolonin
MG
,
Saha
PK
,
Chan
L
,
Pasqualini
R
,
Arap
W
.
Reversal of obesity by targeted ablation of adipose tissue
.
Nat Med
2004
;
10
:
625
632
33.
Hossen
MN
,
Kajimoto
K
,
Akita
H
,
Hyodo
M
,
Harashima
H
.
Vascular-targeted nanotherapy for obesity: unexpected passive targeting mechanism to obese fat for the enhancement of active drug delivery
.
J Control Release
2012
;
163
:
101
110
34.
Thovhogi
N
,
Sibuyi
NRS
,
Onani
MO
,
Meyer
M
,
Madiehe
AM
.
Peptide-functionalized quantum dots for potential applications in the imaging and treatment of obesity
.
Int J Nanomedicine
2018
;
13
:
2551
2559
35.
Xue
Y
,
Xu
X
,
Zhang
X-Q
,
Farokhzad
OC
,
Langer
R
.
Preventing diet-induced obesity in mice by adipose tissue transformation and angiogenesis using targeted nanoparticles
.
Proc Natl Acad Sci U S A
2016
;
113
:
5552
5557
36.
Rho
JG
,
Han
HS
,
Han
JH
, et al
.
Self-assembled hyaluronic acid nanoparticles: implications as a nanomedicine for treatment of type 2 diabetes
.
J Control Release
2018
;
279
:
89
98
37.
Ma
L
,
Liu
T-W
,
Wallig
MA
, et al
.
Efficient targeting of adipose tissue macrophages in obesity with polysaccharide nanocarriers
.
ACS Nano
2016
;
10
:
6952
6962
38.
Cullis
PR
,
Hope
MJ
.
Lipid nanoparticle systems for enabling gene therapies
.
Mol Ther
2017
;
25
:
1467
1475
39.
Yang
L
,
Bailey
L
,
Baltimore
D
,
Wang
P
.
Targeting lentiviral vectors to specific cell types in vivo
.
Proc Natl Acad Sci U S A
2006
;
103
:
11479
11484
40.
Ussar
S
,
Lee
KY
,
Dankel
SN
, et al
.
ASC-1, PAT2, and P2RX5 are cell surface markers for white, beige, and brown adipocytes
.
Sci Transl Med
2014
;
6
:
247ra103
41.
O’Neill
SM
,
Hinkle
C
,
Chen
S-J
, et al
.
Targeting adipose tissue via systemic gene therapy
.
Gene Ther
2014
;
21
:
653
661
42.
Uhrig-Schmidt
S
,
Geiger
M
,
Luippold
G
, et al
.
Gene delivery to adipose tissue using transcriptionally targeted rAAV8 vectors
.
PLoS One
2014
;
9
:
e116288
43.
Turner
PV
,
Brabb
T
,
Pekow
C
,
Vasbinder
MA
.
Administration of substances to laboratory animals: routes of administration and factors to consider
.
J Am Assoc Lab Anim Sci
2011
;
50
:
600
613
44.
Huang
W
,
Liu
X
,
Queen
NJ
,
Cao
L
.
Targeting visceral fat by intraperitoneal delivery of novel AAV serotype vector restricting off-target transduction in liver
.
Mol Ther Methods Clin Dev
2017
;
6
:
68
78
45.
Huang
W
,
McMurphy
T
,
Liu
X
,
Wang
C
,
Cao
L
.
Genetic manipulation of brown fat via oral administration of an engineered recombinant adeno-associated viral serotype vector
.
Mol Ther
2016
;
24
:
1062
1069
46.
Gómez-Banoy
N
,
Lo
JC
.
Genetic manipulation with viral vectors to assess metabolism and adipose tissue function
.
Methods Mol Biol
2017
;
1566
:
109
124
47.
Mizukami
H
,
Mimuro
J
,
Ogura
T
, et al
.
Adipose tissue as a novel target for in vivo gene transfer by adeno-associated viral vectors
.
Hum Gene Ther
2006
;
17
:
921
928
48.
Jimenez
V
,
Jambrina
C
,
Casana
E
, et al
.
FGF21 gene therapy as treatment for obesity and insulin resistance
.
EMBO Mol Med
2018
;
10
:
e8791
49.
Gnad
T
,
Scheibler
S
,
von Kügelgen
I
, et al
.
Adenosine activates brown adipose tissue and recruits beige adipocytes via A2A receptors
.
Nature
2014
;
516
:
395
399
50.
Won
Y-W
,
Adhikary
PP
,
Lim
KS
,
Kim
HJ
,
Kim
JK
,
Kim
YH
.
Oligopeptide complex for targeted non-viral gene delivery to adipocytes
.
Nat Mater
2014
;
13
:
1157
1164
51.
Yang
R
,
Castriota
G
,
Chen
Y
, et al
.
RNAi-mediated germline knockdown of FABP4 increases body weight but does not improve the deranged nutrient metabolism of diet-induced obese mice
.
Int J Obes
2011
;
35
:
217
225
52.
Coe
NR
,
Simpson
MA
,
Bernlohr
DA
.
Targeted disruption of the adipocyte lipid-binding protein (aP2 protein) gene impairs fat cell lipolysis and increases cellular fatty acid levels
.
J Lipid Res
1999
;
40
:
967
972
53.
Hotamisligil
GS
,
Johnson
RS
,
Distel
RJ
,
Ellis
R
,
Papaioannou
VE
,
Spiegelman
BM
.
Uncoupling of obesity from insulin resistance through a targeted mutation in aP2, the adipocyte fatty acid binding protein
.
Science
1996
;
274
:
1377
1379
54.
Muzumdar
MD
,
Tasic
B
,
Miyamichi
K
,
Li
L
,
Luo
L
.
A global double-fluorescent Cre reporter mouse
.
Genesis
2007
;
45
:
593
605
55.
Doudna
JA
,
Charpentier
E
.
Genome editing. The new frontier of genome engineering with CRISPR-Cas9
.
Science
2014
;
346
:
1258096
56.
Swiech
L
,
Heidenreich
M
,
Banerjee
A
, et al
.
In vivo interrogation of gene function in the mammalian brain using CRISPR-Cas9
.
Nat Biotechnol
2015
;
33
:
102
106
57.
Yin
H
,
Song
C-Q
,
Dorkin
JR
, et al
.
Therapeutic genome editing by combined viral and non-viral delivery of CRISPR system components in vivo
.
Nat Biotechnol
2016
;
34
:
328
333
58.
Guo
Y
,
VanDusen
NJ
,
Zhang
L
, et al
.
Analysis of cardiac myocyte maturation using CASAAV, a platform for rapid dissection of cardiac myocyte gene function in vivo
.
Circ Res
2017
;
120
:
1874
1888
59.
Finn
JD
,
Smith
AR
,
Patel
MC
, et al
.
A single administration of CRISPR/Cas9 lipid nanoparticles achieves robust and persistent in vivo genome editing
.
Cell Rep
2018
;
22
:
2227
2235
60.
Chung
JY
,
Ain
QU
,
Song
Y
,
Yong
S-B
,
Kim
Y-H
.
Targeted delivery of CRISPR interference system against Fabp4 to white adipocytes ameliorates obesity, inflammation, hepatic steatosis, and insulin resistance
.
Genome Res
2019
;
29
:
1442
1452
61.
Shen
Y
,
Cohen
JL
,
Nicoloro
SM
, et al
.
CRISPR-delivery particles targeting nuclear receptor-interacting protein 1 (Nrip1) in adipose cells to enhance energy expenditure
.
J Biol Chem
2018
;
293
:
17291
17305
62.
Kamble
PG
,
Hetty
S
,
Vranic
M
, et al
.
Proof-of-concept for CRISPR/Cas9 gene editing in human preadipocytes: deletion of FKBP5 and PPARG and effects on adipocyte differentiation and metabolism
.
Sci Rep
2020
;
10
:
10565
63.
Platt
RJ
,
Chen
S
,
Zhou
Y
, et al
.
CRISPR-Cas9 knockin mice for genome editing and cancer modeling
.
Cell
2014
;
159
:
440
455
64.
Dow
LE
,
Fisher
J
,
O’Rourke
KP
, et al
.
Inducible in vivo genome editing with CRISPR-Cas9
.
Nat Biotechnol
2015
;
33
:
390
394
65.
Cheng
M
,
Mei
B
,
Zhou
Q
, et al
.
Computational analyses of obesity associated loci generated by genome-wide association studies
.
PLoS One
2018
;
13
:
e0199987
Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. More information is available at https://www.diabetesjournals.org/content/license.