Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Ther ; 32(5): 1407-1424, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38429927

ABSTRACT

Maintaining functional adipose innervation is critical for metabolic health. We found that subcutaneous white adipose tissue (scWAT) undergoes peripheral neuropathy (PN) with obesity, diabetes, and aging (reduced small-fiber innervation and nerve/synaptic/growth-cone/vesicle markers, altered nerve activity). Unlike with nerve injuries, peripheral nerves do not regenerate with PN, and therefore new therapies are needed for treatment of this condition affecting 20-30 million Americans. Here, we validated a gene therapy approach using an adipocyte-tropic adeno-associated virus (AAV; serotype Rec2) to deliver neurotrophic factors (brain-derived neurotrophic factor [BDNF] and nerve growth factor [NGF]) directly to scWAT to improve tissue-specific PN as a proof-of-concept approach. AAVRec2-BDNF intra-adipose delivery improved tissue innervation in obese/diabetic mice with PN, but after longer periods of dietary obesity there was reduced efficacy, revealing a key time window for therapies. AAVRec2-NGF also increased scWAT innervation in obese mice and was more effective than BDNF, likely because Rec2 targeted adipocytes, the tissue's endogenous NGF source. AAVRec2-NGF also worked well even after 25 weeks of dietary obesity, unlike BDNF, which likely needs a vector that targets its physiological cellular source (stromal vascular fraction cells). Given the differing effects of AAVs carrying NGF versus BDNF, a combined therapy may be ideal for PN.


Subject(s)
Adipocytes , Brain-Derived Neurotrophic Factor , Dependovirus , Genetic Therapy , Genetic Vectors , Obesity , Subcutaneous Fat , Animals , Dependovirus/genetics , Obesity/therapy , Obesity/metabolism , Mice , Genetic Therapy/methods , Adipocytes/metabolism , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Subcutaneous Fat/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/genetics , Disease Models, Animal , Nerve Growth Factor/metabolism , Nerve Growth Factor/genetics , Nerve Growth Factors/metabolism , Nerve Growth Factors/genetics , Gene Transfer Techniques , Humans , Male , Peripheral Nervous System Diseases/therapy , Peripheral Nervous System Diseases/etiology , Peripheral Nervous System Diseases/metabolism , Peripheral Nervous System Diseases/genetics , Transduction, Genetic
2.
Adv Biol (Weinh) ; 8(1): e2300148, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37518850

ABSTRACT

Aging results in the progressive decline of muscle strength. Interventions to maintain muscle strength may mitigate the age-related loss of physical function, thus maximizing health span. The work on environmental enrichment (EE), an experimental paradigm recapitulating aspects of an active lifestyle, has revealed EE-induced metabolic benefits mediated by a brain-fat axis across the lifespan of mice. EE initiated at 18-month of age shows a trend toward an increased mean lifespan. While previous work described EE's influences on the aging dynamics of several central-peripheral processes, its influence on muscle remained understudied. Here, the impact of EE is investigated on motor function, neuromuscular physiology, and the skeletal muscle transcriptome. EE is initiated in 20-month-old mice for a five-month period. EE mice exhibit greater relative lean mass that is associated with improved mobility and hindlimb grip strength. Transcriptomic profiling of muscle tissue reveals an EE-associated enrichment of gene expression within several metabolic pathways related to oxidative phosphorylation and the TCA cycle. Many mitochondrial-related genes-several of which participate in the electron transport chain-are upregulated. Stress-responsive signaling pathways are downregulated because of EE. The results suggest that EE improves motor function-possibly through preservation of mitochondrial function-even late in life.


Subject(s)
Environment , Transcriptome , Mice , Animals , Brain , Gene Expression Profiling , Muscle, Skeletal
3.
Methods Mol Biol ; 2662: 167-181, 2023.
Article in English | MEDLINE | ID: mdl-37076680

ABSTRACT

Recombinant adeno-associated virus (AAV) vectors are attractive vehicles for gene therapy. Yet targeting adipose tissue is still a challenging task. We recently showed that a novel engineered hybrid serotype Rec2 displays high efficacy of gene transfer to both brown and white fat. Furthermore, the administration route influences the tropism and efficacy of Rec2 vector with oral administration transducing interscapular brown fat, while intraperitoneal injection preferentially targets visceral fat and liver. To restrict off-target transgene expression in the liver, we further develop a single rAAV vector harboring two expression cassettes: one using CBA promoter driving a transgene and another using a liver-specific albumin promoter driving a microRNA targeting the woodchuck posttranscriptional regulatory element (WPRE) sequence in this rAAV vector. In vivo studies by our lab and others have shown that the Rec2/dual-cassette vector system provides a powerful tool for gain-of-function and loss-of-function studies. Here we offer an updated protocol for AAV packaging and delivery to brown fat.


Subject(s)
Adipose Tissue, Brown , Genetic Vectors , Mice , Animals , Adipose Tissue, Brown/metabolism , Genetic Vectors/genetics , Mice, Inbred CBA , Gene Transfer Techniques , Genetic Therapy/methods , Dependovirus/genetics , Dependovirus/metabolism
4.
PLoS One ; 18(3): e0282566, 2023.
Article in English | MEDLINE | ID: mdl-36893171

ABSTRACT

BTBR T+ Itpr3tf/J (BTBR) mice are used as a model of autism spectrum disorder (ASD), displaying similar behavioral and physiological deficits observed in patients with ASD. Our recent study found that implementation of an enriched environment (EE) in BTBR mice improved metabolic and behavioral outcomes. Brain-derived neurotrophic factor (Bdnf) and its receptor tropomyosin kinase receptor B (Ntrk2) were upregulated in the hypothalamus, hippocampus, and amygdala by implementing EE in BTBR mice, suggesting that BDNF-TrkB signaling plays a role in the EE-BTBR phenotype. Here, we used an adeno-associated virus (AAV) vector to overexpress the TrkB full-length (TrkB.FL) BDNF receptor in the BTBR mouse hypothalamus in order to assess whether hypothalamic BDNF-TrkB signaling is responsible for the improved metabolic and behavioral phenotypes associated with EE. Normal chow diet (NCD)-fed and high fat diet (HFD)-fed BTBR mice were randomized to receive either bilateral injections of AAV-TrkB.FL or AAV-YFP as control, and were subjected to metabolic and behavioral assessments up to 24 weeks post-injection. Both NCD and HFD TrkB.FL overexpressing mice displayed improved metabolic outcomes, characterized as reduced percent weight gain and increased energy expenditure. NCD TrkB.FL mice showed improved glycemic control, reduced adiposity, and increased lean mass. In NCD mice, TrkB.FL overexpression altered the ratio of TrkB.FL/TrkB.T1 protein expression and increased phosphorylation of PLCγ in the hypothalamus. TrkB.FL overexpression also upregulated expression of hypothalamic genes involved in energy regulation and altered expression of genes involved in thermogenesis, lipolysis, and energy expenditure in white adipose tissue and brown adipose tissue. In HFD mice, TrkB.FL overexpression increased phosphorylation of PLCγ. TrkB.FL overexpression in the hypothalamus did not improve behavioral deficits in either NCD or HFD mice. Together, these results suggest that enhancing hypothalamic TrkB.FL signaling improves metabolic health in BTBR mice.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Noncommunicable Diseases , Animals , Mice , Autism Spectrum Disorder/metabolism , Autistic Disorder/genetics , Autistic Disorder/metabolism , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Disease Models, Animal , Hypothalamus/metabolism , Mice, Inbred C57BL , Mice, Inbred Strains , Receptor, trkB/genetics , Receptor, trkB/metabolism
5.
J Immunol ; 208(9): 2109-2121, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35418470

ABSTRACT

CD1d, a lipid Ag-presenting molecule for invariant NKT (iNKT) cells, is abundantly expressed on adipocytes and regulates adipose homeostasis through iNKT cells. CD1d gene expression was restored in visceral adipose tissue adipocytes of CD1d knockout (KO) mice to investigate the interactions between adipocytes and immune cells within adipose tissue. We developed an adipocyte-specific targeting recombinant adeno-associated viral vector, with minimal off-target transgene expression in the liver, to rescue CD1d gene expression in visceral adipose tissue adipocytes of CD1d KO mice, followed by assessment of immune cell alternations in adipose tissue and elucidation of the underlying mechanisms of alteration. We report that adeno-associated virus-mediated gene transfer of CD1d to adipocytes in CD1d KO mice fails to rescue iNKT cells but leads to massive and selective expansion of T cells within adipose tissue, particularly CD8+ T effector cells, that is associated with adipocyte NLRP3 inflammasome activation, dysregulation of adipocyte functional genes, and upregulation of apoptotic pathway proteins. An NLRP3 inhibitor has no effect on T cell phenotypes whereas depletion of CD8+ T cells significantly attenuates inflammasome activation and abolishes the dysregulation of adipocyte functional genes induced by adipocyte CD1d. In contrast, adipocyte overexpression of CD1d fails to induce T cell activation in wild-type mice or in invariant TCR α-chain Jα18 KO mice that have a normal lymphocyte repertoire except for iNKT cells. Our studies uncover an adipocyte CD1d → CD8+ T cell → adipocyte inflammasome cascade, in which CD8+ T cells function as a key mediator of adipocyte inflammation likely induced by an allogeneic response against the CD1d molecule.


Subject(s)
CD8-Positive T-Lymphocytes , Inflammasomes , Adipocytes , Animals , Antigens, CD1d , CD8-Positive T-Lymphocytes/metabolism , Inflammasomes/metabolism , Inflammation/metabolism , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
6.
Mol Ther Methods Clin Dev ; 20: 409-422, 2021 Mar 12.
Article in English | MEDLINE | ID: mdl-33575433

ABSTRACT

Fibroblast growth factor 21 (FGF21) is a peptide hormone that serves as a potent effector of energy homeostasis. Increasingly, FGF21 is viewed as a promising therapeutic agent for type 2 diabetes, fatty liver disease, and other metabolic complications. Exogenous administration of native FGF21 peptide has proved difficult due to unfavorable pharmacokinetic properties. Here, we utilized an engineered serotype adeno-associated viral (AAV) vector coupled with a dual-cassette design to selectively overexpress FGF21 in visceral adipose tissue of insulin-resistant BTBR T+Itpr3tf/J (BTBR) mice. Under high-fat diet conditions, a single, low-dose intraperitoneal injection of AAV-FGF21 resulted in sustained benefits, including improved insulin sensitivity, glycemic processing, and systemic metabolic function and reduced whole-body adiposity, hepatic steatosis, inflammatory cytokines, and adipose tissue macrophage inflammation. Our study highlights the potential of adipose tissue as a FGF21 gene-therapy target and the promise of minimally invasive AAV vectors as therapeutic agents for metabolic diseases.

7.
Mol Ther Methods Clin Dev ; 19: 236-249, 2020 Dec 11.
Article in English | MEDLINE | ID: mdl-33102616

ABSTRACT

Adipose tissue is one of the largest organs, playing important roles in physiology and pathologies of multiple diseases. However, research related to adeno-associated virus (AAV) targeting adipose tissue has been left far behind studies carried out in the liver, brain, heart, and muscle. Despite initial reports indicating poor performance, AAV-mediated gene delivery to adipose tissue has continued to rise during the past two decades. AAV8 and a novel engineered hybrid serotype, Rec2, have been shown to transduce adipose tissue more efficiently than other serotypes so far tested and have been applied in most of the in vivo studies. The Rec2 serotype displays high efficacy of gene transfer to both brown and white fat via local and systemic administration. This review summarizes the advances in developing AAV vectors with enhanced adipose tropism and restricting off-target transgene expression. We discuss the challenges and strategies to search for and generate novel serotypes with tropism tailoring for adipose tissue and develop AAV vector systems to improve adipose transgene expression for basic research and translational studies.

SELECTION OF CITATIONS
SEARCH DETAIL
...