87 results on '"David A. Bernlohr"'
Search Results
2. Slc43a3 is a regulator of free fatty acid flux
- Author
-
Salman Azhar, Wei Wang, Wen-Jun Shen, Qui Shuo, Yiqiang Zhang, Xiaoming Hou, Kathrin B. Hasbargen, Fredric B. Kraemer, and David A. Bernlohr
- Subjects
Adult ,Male ,0301 basic medicine ,Amino Acid Transport Systems ,Adipose tissue ,Fatty Acids, Nonesterified ,030204 cardiovascular system & hematology ,Biochemistry ,Cell Line ,Mice ,Young Adult ,03 medical and health sciences ,chemistry.chemical_compound ,Adenosine Triphosphate ,0302 clinical medicine ,Endocrinology ,Lipid droplet ,Adipocyte ,Cyclic AMP ,Animals ,Humans ,RNA, Messenger ,Research Articles ,chemistry.chemical_classification ,Membrane Transport Proteins ,Fatty acid ,Biological Transport ,Cell Biology ,Cell biology ,Solute carrier family ,030104 developmental biology ,Gene Expression Regulation ,chemistry ,Membrane protein ,Gene Knockdown Techniques ,Female ,Efflux ,Flux (metabolism) - Abstract
Adipocytes take up long chain FAs through diffusion and protein-mediated transport, whereas FA efflux is considered to occur by diffusion. To identify potential membrane proteins that are involved in regulating FA flux in adipocytes, the expression levels of 55 membrane transporters without known function were screened in subcutaneous adipose samples from obese patients before and after bariatric surgery using branched DNA methodology. Among the 33 solute carrier (SLC) transporter family members screened, the expression of 14 members showed significant changes before and after bariatric surgery. One of them, Slc43a3, increased about 2.5-fold after bariatric surgery. Further investigation demonstrated that Slc43a3 is highly expressed in murine adipose tissue and induced during adipocyte differentiation in primary preadipocytes and in OP9 cells. Knockdown of Slc43a3 with siRNA in differentiated OP9 adipocytes reduced both basal and forskolin-stimulated FA efflux, while also increasing FA uptake and lipid droplet accumulation. In contrast, overexpression of Slc43a3 decreased FA uptake in differentiated OP9 cells and resulted in decreased lipid droplet accumulation. Therefore, Slc43a3 seems to regulate FA flux in adipocytes, functioning as a positive regulator of FA efflux and as a negative regulator of FA uptake.
- Published
- 2020
- Full Text
- View/download PDF
3. Unconventional Secretion of Adipocyte Fatty Acid Binding Protein 4 Is Mediated By Autophagic Proteins in a Sirtuin-1–Dependent Manner
- Author
-
Ellie K. Bohm, David A. Bernlohr, Shin-ichiro Imai, Michael W. McBurney, Ajeetha Josephrajan, Douglas G. Mashek, Do Hyung Kim, and Ann V. Hertzel
- Subjects
0301 basic medicine ,Endocrinology, Diabetes and Metabolism ,ATG5 ,Autophagy-Related Proteins ,Adipose tissue ,030209 endocrinology & metabolism ,Stimulation ,Protein Serine-Threonine Kinases ,Fatty Acid-Binding Proteins ,Fatty acid-binding protein ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Sirtuin 1 ,Adipocytes ,Autophagy ,Internal Medicine ,Animals ,Autophagy-Related Protein-1 Homolog ,Secretion ,Gene Silencing ,Mice, Knockout ,Gene knockdown ,biology ,Chemistry ,Cell biology ,030104 developmental biology ,Adipose Tissue ,Knockout mouse ,biology.protein ,Beclin-1 ,Insulin Resistance ,Obesity Studies - Abstract
Fatty acid binding protein 4 (FABP4) is a leaderless lipid carrier protein primarily expressed by adipocytes and macrophages that not only functions intracellularly but is also secreted. The secretion is mediated via unconventional mechanism(s), and in a variety of species, metabolic dysfunction is correlated with elevated circulating FABP4 levels. In diabetic animals, neutralizing antibodies targeting serum FABP4 increase insulin sensitivity and attenuate hepatic glucose output, suggesting the functional importance of circulating FABP4. Using animal and cell-based models, we show that FABP4 is secreted from white, but not brown, adipose tissue in response to lipolytic stimulation in a sirtuin-1 (SIRT1)–dependent manner via a mechanism that requires some, but not all, autophagic components. Silencing of early autophagic genes such as Ulk1/2, Fip200, or Beclin-1 or chemical inhibition of ULK1/2 or VPS34 attenuated secretion, while Atg5 knockdown potentiated FABP4 release. Genetic knockout of Sirt1 diminished secretion, and serum FABP4 levels were undetectable in Sirt1 knockout mice. In addition, blocking SIRT1 by EX527 attenuated secretion while activating SIRT1 by resveratrol-potentiated secretion. These studies suggest that FABP4 secretion from adipocytes is regulated by SIRT1 and requires early autophagic components.
- Published
- 2019
- Full Text
- View/download PDF
4. Adipose Lipocalin 2 overexpression protects against age-related decline in thermogenic function of adipose tissue and metabolic deterioration
- Author
-
Yingjie Wu, David A. Bernlohr, Jessica A. Deis, Chengyu Liu, Xiaoli Chen, and Hong Guo
- Subjects
Male ,Lipocalin 2 ,0301 basic medicine ,Aging ,lcsh:Internal medicine ,medicine.medical_specialty ,Adipose Tissue, White ,Adipose tissue ,030209 endocrinology & metabolism ,White adipose tissue ,Adipose beiging ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Insulin resistance ,AMP-Activated Protein Kinase Kinases ,Lipocalin-2 ,Fibrosis ,Internal medicine ,Adipocyte ,medicine ,Animals ,lcsh:RC31-1245 ,Molecular Biology ,2. Zero hunger ,business.industry ,Thermogenesis ,Cell Biology ,Adipose Tissue, Beige ,Lipid Metabolism ,medicine.disease ,3. Good health ,Mice, Inbred C57BL ,Glucose ,030104 developmental biology ,Endocrinology ,Liver ,chemistry ,Adipogenesis ,Healthspan ,Original Article ,Steatosis ,business ,Protein Kinases ,Dyslipidemia - Abstract
Objectives Aging increases the risk for development of adipose tissue dysfunction, insulin resistance, dyslipidemia, and liver steatosis. Lipocalin 2 (Lcn2) deficient mice are more prone to diet-induced obesity and metabolic dysfunction, indicating a protective role for Lcn2 in younger mice. In this study, we determined whether overexpressing Lcn2 in adipose tissue can protect against age-related metabolic deterioration. Methods We developed ap2-promoter-driven Lcn2 transgenic (Tg) mice and aged Lcn2 Tg mice for the metabolic assessments. Results We found decreased adipocyte size in inguinal white adipose tissue (iWAT) from 10-month-old Lcn2 Tg mice relative to WT. This was accompanied by increased markers of adipogenesis in iWAT and attenuation of the age-related decline in AMP-activated protein kinase (AMPK) phosphorylation in adipose tissue depots. In addition to improvements in adipose tissue function, whole-body metabolic homeostasis was maintained in aged Lcn2 Tg mice. This included improved glucose tolerance and reduced serum triglycerides in older Lcn2 Tg mice relative to WT mice. Further, liver morphology and liver lipid levels were improved in older Lcn2 Tg mice, alongside a decrease in markers of liver inflammation and fibrosis. Conclusions We demonstrate that overexpression of Lcn2 in adipose tissue not only preserves adipose tissue function during aging but also promotes maintenance of glucose tolerance, decreases dyslipidemia, and prevents liver lipid accumulation and steatosis., Highlights • Lcn2 overexpression in adipose tissue promotes beiging of iWAT and BAT mitochondrial oxidation. • Lcn2 overexpression in adipose tissue improves thermogenic adaptation to cold in younger mice. • Adipose Lcn2 overexpression prevents age-related decline in thermogenic gene expression in brown and beige fat tissue. • Lcn2 overexpression in adipose tissue preserves adipose tissue function during aging. • Adipose Lcn2 overexpression promotes metabolic homeostasis and prevents age-related liver lipid accumulation.
- Published
- 2019
- Full Text
- View/download PDF
5. Defective internal allosteric network imparts dysfunctional ATP/substrate-binding cooperativity in oncogenic chimera of protein kinase A
- Author
-
Simon M. Sandford, Fernando Porcelli, Cristina Olivieri, V. S. Manu, David D. Thomas, Donald K. Blumenthal, Susan S. Taylor, Caitlin Walker, Yingjie Wang, Gianluigi Veglia, David A. Bernlohr, and Adak Karamafrooz
- Subjects
0301 basic medicine ,genetic structures ,QH301-705.5 ,Kinase ,Chemistry ,Allosteric regulation ,Medicine (miscellaneous) ,Cooperativity ,Fusion protein ,General Biochemistry, Genetics and Molecular Biology ,Article ,PRKACA ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Heat shock protein ,Phosphorylation ,Biology (General) ,General Agricultural and Biological Sciences ,Protein kinase A ,Solution-state NMR - Abstract
An aberrant fusion of the DNAJB1 and PRKACA genes generates a chimeric protein kinase (PKA-CDNAJB1) in which the J-domain of the heat shock protein 40 is fused to the catalytic α subunit of cAMP-dependent protein kinase A (PKA-C). Deceivingly, this chimeric construct appears to be fully functional, as it phosphorylates canonical substrates, forms holoenzymes, responds to cAMP activation, and recognizes the endogenous inhibitor PKI. Nonetheless, PKA-CDNAJB1 has been recognized as the primary driver of fibrolamellar hepatocellular carcinoma and is implicated in other neoplasms for which the molecular mechanisms remain elusive. Here we determined the chimera’s allosteric response to nucleotide and pseudo-substrate binding. We found that the fusion of the dynamic J-domain to PKA-C disrupts the internal allosteric network, causing dramatic attenuation of the nucleotide/PKI binding cooperativity. Our findings suggest that the reduced allosteric cooperativity exhibited by PKA-CDNAJB1 alters specific recognitions and interactions between substrates and regulatory partners contributing to dysregulation., Olivieri, Walker, Karamafrooz et al. show that the fusion of the dynamic J-domain to PKA-C (PKA-CDNAJB1) disrupts the internal allosteric network, attenuating the nucleotide/PKI binding cooperativity. This study suggests that the reduced allosteric cooperativity may contribute to the pathology that PKA-CDNAJB1 drives.
- Published
- 2021
6. Histone Carbonylation Is a Redox-Regulated Epigenomic Mark That Accumulates with Obesity and Aging
- Author
-
David A. Bernlohr, Yuxiang Sun, Tong Zhou, Yue Chen, Michael B. O'Connor, Amy K. Hauck, and Ambuj Upadhyay
- Subjects
0301 basic medicine ,Physiology ,Protein Carbonylation ,Clinical Biochemistry ,4-HNE (4-hydroxynonenal) ,Adipose tissue ,histone ,carbonylation ,medicine.disease_cause ,Biochemistry ,Article ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,medicine ,Molecular Biology ,Epigenomics ,adipose ,biology ,Chemistry ,lcsh:RM1-950 ,aging ,Cell Biology ,In vitro ,4-HHE (4-hydroxy hexenal) ,Cell biology ,030104 developmental biology ,Histone ,lcsh:Therapeutics. Pharmacology ,epigenomics ,biology.protein ,Carbonylation ,030217 neurology & neurosurgery ,Oxidative stress - Abstract
Oxidative stress is a hallmark of metabolic disease, though the mechanisms that define this link are not fully understood. Irreversible modification of proteins by reactive lipid aldehydes (protein carbonylation) is a major consequence of oxidative stress in adipose tissue and the substrates and specificity of this modification are largely unexplored. Here we show that histones are avidly modified by 4-hydroxynonenal (4-HNE) in vitro and in vivo. Carbonylation of histones by 4-HNE increased with age in male flies and visceral fat depots of mice and was potentiated in genetic (ob/ob) and high-fat feeding models of obesity. Proteomic evaluation of in vitro 4-HNE- modified histones led to the identification of both Michael and Schiff base adducts. In contrast, mapping of sites in vivo from obese mice exclusively revealed Michael adducts. In total, we identified 11 sites of 4-hydroxy hexenal (4-HHE) and 10 sites of 4-HNE histone modification in visceral adipose tissue. In summary, these results characterize adipose histone carbonylation as a redox-linked epigenomic mark associated with metabolic disease and aging.
- Published
- 2020
7. Stearic Acid Induces CD11c Expression in Proinflammatory Macrophages via Epidermal Fatty Acid Binding Protein
- Author
-
Shujun Liu, Jill Suttles, Jiaqing Hao, Margot P. Cleary, Edward R. Sauter, Yuwen Zhang, Bing Li, Yanwen Sun, David A. Bernlohr, and Jun Zeng
- Subjects
0301 basic medicine ,medicine.drug_class ,Immunology ,Mice, Obese ,CD11c ,Inflammation ,Fatty Acid-Binding Proteins ,Monocytes ,Article ,Fatty acid-binding protein ,Proinflammatory cytokine ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,medicine ,Animals ,Immunology and Allergy ,Obesity ,Retinoid ,Receptor ,integumentary system ,Chemistry ,Macrophages ,Binding protein ,Fatty Acids ,CD11c Antigen ,Up-Regulation ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,030220 oncology & carcinogenesis ,lipids (amino acids, peptides, and proteins) ,medicine.symptom ,Stearic Acids - Abstract
Obesity is associated with elevated levels of free fatty acids (FAs) and proinflammatory CD11c+ macrophages. However, whether and how free FAs contribute to CD11c+ macrophage differentiation and proinflammatory functions remain unclear. Here we report that dietary saturated FAs, but not unsaturated FAs, promoted the differentiation and function of CD11c+ macrophages. Specifically, we demonstrated that stearic acid (SA) significantly induced CD11c expression in monocytes through activation of the nuclear retinoid acid receptor. More importantly, cytosolic expression of epidermal FA binding protein (E-FABP) in monocytes/macrophages was shown to be critical to the mediation of the SA-induced effect. Depletion of E-FABP not only inhibited SA-induced CD11c upregulation in macrophages in vitro but also abrogated high-saturated-fat diet–induced skin lesions in obese mouse models in vivo. Altogether, our data demonstrate a novel mechanism by which saturated FAs promote obesity-associated inflammation through inducing E-FABP/retinoid acid receptor–mediated differentiation of CD11c+ macrophages.
- Published
- 2018
- Full Text
- View/download PDF
8. Perilipin 5 and liver fatty acid binding protein function to restore quiescence in mouse hepatic stellate cells
- Author
-
Shizhong Zheng, Elizabeth P. Newberry, Alan D. Attie, David A. Bernlohr, William S. Blaner, Jianguo Lin, Nicholas O. Davidson, Anping Chen, and Mark P. Keller
- Subjects
Male ,0301 basic medicine ,Endogeny ,QD415-436 ,Fatty Acid-Binding Proteins ,Perilipin-5 ,Biochemistry ,Fatty acid-binding protein ,Small Molecule Libraries ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,stellate cell activation ,Lipid droplet ,lipid metabolism ,Gene expression ,Hepatic Stellate Cells ,Animals ,Protein kinase A ,Research Articles ,Cells, Cultured ,Mice, Knockout ,Chemistry ,perilipins ,AMPK ,Lipid Droplets ,Cell Biology ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,030220 oncology & carcinogenesis ,Perilipin ,Hepatic stellate cell ,Female ,lipids (amino acids, peptides, and proteins) - Abstract
Hepatic stellate cell (HSC) activation occurs along with decreased Perilipin5 (Plin5) and liver fatty acid-binding protein (L-Fabp) expression and coincident lipid droplet (LD) depletion. Conversely, the activated phenotype is reversible in WT HSCs upon forced expression of Plin5. Here, we asked if L-Fabp expression is required for Plin5-mediated rescue of the quiescent phenotype. Lentiviral Plin5 transduction of passaged L-Fabp(−/−) HSCs failed to reverse activation markers or restore lipogenic gene expression and LD formation. However, adenoviral L-Fabp infection of lentiviral Plin5 transduced L-Fabp(−/−) HSCs restored both the quiescent phenotype and LD formation, an effect also mediated by adenoviral intestine-Fabp or adipocyte-Fabp. Expression of exogenous Plin5 in activated WT HSCs induced a transcriptional program of lipogenic gene expression including endogenous L-Fabp, but none of the other FABPs. We further demonstrated that selective, small molecule inhibition of endogenous L-Fabp also eliminated the ability of exogenous Plin5 to rescue LD formation and reverse activation of WT HSCs. This functional coordination of L-Fabp with Plin5 was 5′-AMP-activated protein kinase (AMPK)-dependent and was eliminated by AMPK inhibition. Taken together, our results indicate that L-Fabp is required for Plin5 to activate a transcriptional program that restores LD formation and reverses HSC activation.
- Published
- 2018
- Full Text
- View/download PDF
9. Is Disrupted Nucleotide-Substrate Cooperativity a Common Trait for Cushing's Syndrome Driving Mutations of Protein Kinase A?
- Author
-
Jiali Gao, Yingjie Wang, Caitlin Walker, David A. Bernlohr, Davide Calebiro, Gianluigi Veglia, Cristina Olivieri, Manu V S, and Susan S. Taylor
- Subjects
binding cooperativity ,Protein Conformation ,Protein subunit ,Allosteric regulation ,Mutant ,Cushing's syndrome ,Cooperativity ,medicine.disease_cause ,Article ,Substrate Specificity ,cAMP-dependent protein kinase A ,Allosteric Regulation ,Structural Biology ,Catalytic Domain ,Settore BIO/10 - Biochimica ,medicine ,Humans ,Phosphorylation ,Protein kinase A ,Cushing Syndrome ,Molecular Biology ,Settore CHIM/02 - Chimica Fisica ,Cyclic AMP-Dependent Protein Kinase Catalytic Subunits ,Mutation ,allostery ,Nucleotides ,Kinase ,Chemistry ,Cyclic AMP-Dependent Protein Kinases ,Cell biology ,Phenotype - Abstract
Somatic mutations in the PRKACA gene encoding the catalytic α subunit of protein kinase A (PKA-C) are responsible for cortisol-producing adrenocortical adenomas. These benign neoplasms contribute to the development of Cushing's syndrome. The majority of these mutations occur at the interface between the two lobes of PKA-C and interfere with the enzyme's ability to recognize substrates and regulatory (R) subunits, leading to aberrant phosphorylation patterns and activation. Rarely, patients with similar phenotypes carry an allosteric mutation, E31V, located at the C-terminal end of the αA-helix and adjacent to the αC-helix, but structurally distinct from the PKA-C/R subunit interface mutations. Using a combination of solution NMR, thermodynamics, kinetic assays, and molecular dynamics simulations, we show that the E31V allosteric mutation disrupts central communication nodes between the N- and C- lobes of the enzyme as well as nucleotide-substrate binding cooperativity, a hallmark for kinases' substrate fidelity and regulation. For both orthosteric (L205R and W196R) and allosteric (E31V) Cushing’s syndrome mutants, the loss of binding cooperativity is proportional to the density of the intramolecular allosteric network. This structure–activity relationship suggests a possible common mechanism for Cushing's syndrome driving mutations in which decreased nucleotide/substrate binding cooperativity is linked to loss in substrate fidelity and dysfunctional regulation.
- Published
- 2021
- Full Text
- View/download PDF
10. Fatty acid binding protein 4/aP2-dependent BLT1R expression and signaling
- Author
-
Ann V. Hertzel, Hongliang Xu, David A. Bernlohr, Nicholas Kvalheim, and Michael Downey
- Subjects
Lipopolysaccharides ,0301 basic medicine ,Receptors, Leukotriene B4 ,Inflammation ,lipid mediators ,QD415-436 ,Fatty Acid-Binding Proteins ,Biochemistry ,Fatty acid-binding protein ,Proinflammatory cytokine ,lipids ,Gene Knockout Techniques ,Mice ,03 medical and health sciences ,Endocrinology ,leukotrienes ,medicine ,Animals ,Gene silencing ,Uncoupling Protein 2 ,RNA, Messenger ,adipocyte protein 2 ,Research Articles ,biology ,Chemistry ,Leukotriene B4 Receptor 1 ,Cell Biology ,Lipid signaling ,macrophages ,Cell biology ,RAW 264.7 Cells ,030104 developmental biology ,Gene Expression Regulation ,inflammation ,biology.protein ,Signal transduction ,medicine.symptom ,Reactive Oxygen Species ,Signal Transduction - Abstract
Previous studies have shown that reduced levels of the adipocyte fatty acid binding protein (FABP)4 (AFABP/aP2), result in metabolic improvement including potentiated insulin sensitivity and attenuated atherosclerosis. Mechanistically, pharmacologic or genetic inhibition of FABP4 in macrophages upregulates UCP2, attenuates reactive oxygen species (ROS) production, polarizes cells toward the anti-inflammatory M2 state, and reduces leukotriene (LT) secretion. At the protein level, FABP4 stabilizes LTA4 toward chemical hydrolysis, thereby potentiating inflammatory LTC4 synthesis. Herein, we extend the FABP4-LT axis and demonstrate that genetic knockout of FABP4 reduces expression of the major macrophage LT receptor, LTB4 receptor 1 (BLT1R), via a ROS-dependent mechanism. Consistent with inflammation driving BLT1R expression, M1 polarized macrophages express increased levels of BLT1R relative to M2 polarized macrophages and treatment with proinflammatory lipopolysaccharide increased BLT1R mRNA and protein expression. In FABP4 knockout macrophages, silencing of UCP2, increased ROS levels and led to increased expression of BLT1R mRNA. Similarly, addition of exogenous H2O2 upregulated BLT1R expression, whereas the addition of a ROS scavenger, N-acetyl cysteine, decreased BLT1R levels. As compared with WT macrophages, LTB4-BLT1R-dependent JAK2-phosphorylation was reduced in FABP4 knockout macrophages. In summary, these results indicate that FABP4 regulates the expression of BLT1R and its downstream signaling via control of oxidative stress in macrophages.
- Published
- 2017
- Full Text
- View/download PDF
11. Stearoyl-CoA Desaturase Mediated Monounsaturated Fatty Acid Availability Supports Humoral Immunity
- Author
-
John A. Copland, Xingxing Zhu, Xian Zhou, Hu Zeng, Virginia Smith Shapiro, Taro Hitosugi, Jie Sun, Zhenqing Ye, Chaofan Li, David A. Bernlohr, and Yanfeng Li
- Subjects
Chemistry ,Endoplasmic reticulum ,Autophagy ,food and beverages ,Germinal center ,Metabolism ,Cell biology ,Stearoyl-CoA Desaturase ,medicine.anatomical_structure ,Immune system ,Humoral immunity ,medicine ,lipids (amino acids, peptides, and proteins) ,B cell - Abstract
Immune cells can metabolize glucose, amino acids, and fatty acids (FAs) to generate energy. The role of different FA species, and their impacts on humoral immunity remains poorly understood. Here we report that proliferating B cells require monounsaturated FAs (MUFA) to maintain mitochondrial metabolism and mTOR activity, and to prevent excessive autophagy and endoplasmic reticulum (ER) stress. Furthermore, B cell extrinsic Stearoyl-CoA desaturase (SCD) activity generates MUFA to support early B cell development and germinal center (GC) formationin vivoduring immunization and influenza infection. Thus, SCD-mediated MUFA production is critical for humoral immunity.
- Published
- 2020
- Full Text
- View/download PDF
12. Lipid receptors and signaling in adipose tissue
- Author
-
David A. Bernlohr, Timothy D. O'Connell, and Ann V. Hertzel
- Subjects
chemistry.chemical_classification ,Cell signaling ,biology ,CD36 ,Fatty acid ,Adipose tissue ,White adipose tissue ,Lipid signaling ,Cell biology ,chemistry ,biology.protein ,lipids (amino acids, peptides, and proteins) ,Receptor ,G protein-coupled receptor - Abstract
Beyond their role as macronutrients, nonesterified fatty acids (free fatty acids, FFAs) and their derivatives (prostanoids and eicosanoids) serve as signaling molecules regulating a variety of processes including inflammation, metabolism, and gene expression. While traditionally considered only to be esterified with coenzyme A staging the lipid for phospholipid and triacylglycerol biogenesis, unesterified fatty acids can diffuse laterally in the plasma membrane and interact with receptors thereby providing metabolic opportunities to elaborate a variety of second messenger systems. Receptors for fatty acids (CD36/SR-B2, GPCRs), prostaglandins (DP/EP receptors) and leukotrienes (LT receptors) are abundantly expressed by fat cells and are themselves controlled lipid signaling. Moreover, such receptors are variably expressed in different adipose depots implying specificity of signaling systems, particularly in white adipose tissue versus brown or beige fat. This chapter will summarize our current awareness of fatty acid receptors expressed by adipocytes and the signaling pathways that are affected downstream of lipid binding.
- Published
- 2020
- Full Text
- View/download PDF
13. Cushing’s syndrome driver mutation disrupts protein kinase A allosteric network, altering both regulation and substrate specificity
- Author
-
David A. Bernlohr, Adak Karamafrooz, Davide Calebiro, Jiali Gao, Cristina Olivieri, Jordan Casby, Caitlin Walker, Susan S. Taylor, Gianluigi Veglia, Kerstin Bathon, and Yingjie Wang
- Subjects
Hydrocortisone ,Mutant ,Allosteric regulation ,Biophysics ,Cooperativity ,medicine.disease_cause ,Substrate Specificity ,03 medical and health sciences ,0302 clinical medicine ,Allosteric Regulation ,Structural Biology ,Catalytic Domain ,medicine ,Humans ,skin and connective tissue diseases ,Protein kinase A ,Cushing Syndrome ,Research Articles ,030304 developmental biology ,chemistry.chemical_classification ,Cyclic AMP-Dependent Protein Kinase Catalytic Subunits ,0303 health sciences ,Mutation ,Multidisciplinary ,Kinase ,SciAdv r-articles ,Cyclic AMP-Dependent Protein Kinases ,Cell biology ,Enzyme ,chemistry ,Adrenocortical Adenoma ,Phosphorylation ,sense organs ,030217 neurology & neurosurgery ,Research Article - Abstract
A single mutation in protein kinase A remodels the intramolecular allostery, changing substrate specificity and regulation., Genetic alterations in the PRKACA gene coding for the catalytic α subunit of the cAMP-dependent protein kinase A (PKA-C) are linked to cortisol-secreting adrenocortical adenomas, resulting in Cushing’s syndrome. Among those, a single mutation (L205R) has been found in up to 67% of patients. Because the x-ray structures of the wild-type and mutant kinases are essentially identical, the mechanism explaining aberrant function of this mutant remains under active debate. Using NMR spectroscopy, thermodynamics, kinetic assays, and molecular dynamics simulations, we found that this single mutation causes global changes in the enzyme, disrupting the intramolecular allosteric network and eliciting losses in nucleotide/pseudo-substrate binding cooperativity. Remarkably, by rewiring its internal allosteric network, PKA-CL205R is able to bind and phosphorylate non-canonical substrates, explaining its changes in substrate specificity. Both the lack of regulation and change in substrate specificity reveal the complex role of this mutated kinase in the formation of cortisol-secreting adrenocortical adenomas.
- Published
- 2019
- Full Text
- View/download PDF
14. Mitochondrial Oxidative Stress And Adipocyte Protein Carbonylation
- Author
-
Amy K. Hauck, Yue Chen, David A. Bernlohr, and Yimao Huang
- Subjects
chemistry.chemical_compound ,chemistry ,Adipocyte ,Protein Carbonylation ,Genetics ,medicine ,medicine.disease_cause ,Molecular Biology ,Biochemistry ,Oxidative stress ,Biotechnology ,Cell biology - Published
- 2019
- Full Text
- View/download PDF
15. Author Correction: Defective internal allosteric network imparts dysfunctional ATP/substrate-binding cooperativity in oncogenic chimera of protein kinase A
- Author
-
Donald K. Blumenthal, Yingjie Wang, Sanford M. Simon, Caitlin Walker, V. S. Manu, Fernando Porcelli, David D. Thomas, Cristina Olivieri, Gianluigi Veglia, Susan S. Taylor, David A. Bernlohr, and Adak Karamafrooz
- Subjects
QH301-705.5 ,Recombinant Fusion Proteins ,Allosteric regulation ,Medicine (miscellaneous) ,Cooperativity ,Molecular Dynamics Simulation ,Ligands ,General Biochemistry, Genetics and Molecular Biology ,Chimera (genetics) ,Adenosine Triphosphate ,Allosteric Regulation ,Catalytic Domain ,Cyclic AMP ,Humans ,Phosphorylation ,Biology (General) ,Author Correction ,Protein kinase A ,Cyclic AMP-Dependent Protein Kinase Catalytic Subunits ,Binding Sites ,Chemistry ,Substrate (chemistry) ,HSP40 Heat-Shock Proteins ,Peptide Fragments ,Cell biology ,General Agricultural and Biological Sciences ,Solution-state NMR ,Protein Binding - Abstract
A Correction to this paper has been published: https://doi.org/10.1038/s42003-021-02006-3
- Published
- 2021
16. Lipocalin 2, a Regulator of Retinoid Homeostasis and Retinoid-mediated Thermogenic Activation in Adipose Tissue
- Author
-
Hong Guo, William S. Blaner, Hongfeng Jiang, Jessica A. Deis, Xiaoli Chen, Rocio Foncea, Sheila M. O'Byrne, David A. Bernlohr, and Yuanyuan Zhang
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,medicine.drug_class ,Adipose tissue macrophages ,Adipocytes, White ,Retinol transport ,Retinoic acid ,Adipose tissue ,Tretinoin ,White adipose tissue ,Lipocalin ,Biology ,Biochemistry ,Mice ,Retinoids ,03 medical and health sciences ,chemistry.chemical_compound ,Lipocalin-2 ,Internal medicine ,medicine ,Animals ,Homeostasis ,Obesity ,Retinoid ,Molecular Biology ,Uncoupling Protein 1 ,Mice, Knockout ,Membrane Proteins ,Thermogenesis ,Cell Biology ,Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha ,Metabolism ,Adipocytes, Brown ,030104 developmental biology ,Endocrinology ,Adipose Tissue ,chemistry ,Retinol-Binding Proteins, Plasma ,Protein Binding - Abstract
We have recently characterized the role of lipocalin 2 (Lcn2) as a new adipose-derived cytokine in the regulation of adaptive thermogenesis via a non-adrenergic pathway. Herein, we explored a potential non-adrenergic mechanism by which Lcn2 regulates thermogenesis and lipid metabolism. We found that Lcn2 is a retinoic acid target gene, and retinoic acid concurrently stimulated UCP1 and Lcn2 expression in adipocytes. Lcn2 KO mice exhibited a blunted effect of all-trans-retinoic acid (ATRA) on body weight and fat mass, lipid metabolism, and retinoic acid signaling pathway activation in adipose tissue under the high fat diet-induced obese condition. We further demonstrated that Lcn2 is required for the full action of ATRA on the induction of UCP1 and PGC-1α expression in brown adipocytes and the restoration of cold intolerance in Lcn2 KO mice. Interestingly, we discovered that Lcn2 KO mice have decreased levels of retinoic acid and retinol in adipose tissue. The protein levels of STRA6 responsible for retinol uptake were significantly decreased in adipose tissue. The retinol transporter RBP4 was increased in adipose tissue but decreased in the circulation, suggesting the impairment of RBP4 secretion in Lcn2 KO adipose tissue. Moreover, Lcn2 deficiency abolished the ATRA effect on RBP4 expression in adipocytes. All the data suggest that the decreased retinoid level and action are associated with impaired retinol transport and storage in adipose tissue in Lcn2 KO mice. We conclude that Lcn2 plays a critical role in regulating metabolic homeostasis of retinoids and retinoid-mediated thermogenesis in adipose tissue.
- Published
- 2016
- Full Text
- View/download PDF
17. Stearoyl-CoA Desaturase-Mediated Monounsaturated Fatty Acid Availability Supports Humoral Immunity
- Author
-
Taro Hitosugi, David A. Bernlohr, Xian Zhou, Yanfeng Li, Hu Zeng, Xingxing Zhu, John A. Copland, Zhenqing Ye, Jie Sun, Chaofan Li, and Virginia Smith Shapiro
- Subjects
0301 basic medicine ,Mice, Transgenic ,General Biochemistry, Genetics and Molecular Biology ,Article ,Fatty Acids, Monounsaturated ,03 medical and health sciences ,Gene Knockout Techniques ,Mice ,0302 clinical medicine ,Immune system ,Orthomyxoviridae Infections ,medicine ,Autophagy ,Animals ,B cell ,B-Lymphocytes ,Chemistry ,Endoplasmic reticulum ,TOR Serine-Threonine Kinases ,Germinal center ,food and beverages ,Metabolism ,Endoplasmic Reticulum Stress ,Orthomyxoviridae ,Cell biology ,Immunity, Humoral ,Mitochondria ,Mice, Inbred C57BL ,Stearoyl-CoA Desaturase ,030104 developmental biology ,medicine.anatomical_structure ,Humoral immunity ,lipids (amino acids, peptides, and proteins) ,030217 neurology & neurosurgery - Abstract
SUMMARY Immune cells can metabolize glucose, amino acids, and fatty acids (FAs) to generate energy. The roles of different FA species and their impacts on humoral immunity remain poorly understood. Here, we report that proliferating B cells require monounsaturated FAs (MUFAs) to maintain mitochondrial metabolism and mTOR activity and to prevent excessive autophagy and endoplasmic reticulum (ER) stress. Furthermore, B cell-extrinsic stearoyl-CoA desaturase (SCD) activity generates MUFA to support early B cell development and germinal center (GC) formation in vivo during immunization and influenza infection. Thus, SCD-mediated MUFA production is critical for humoral immunity., Graphical Abstract, In Brief Zhou et al. show that monounsaturated fatty acids (MUFAs), generated by stearoyl-CoA desaturase (SCD), support B cell mitochondrial metabolism and mTOR activity and promote B cell development and humoral immune responses. These data establish MUFA availability as a key regulator for humoral immunity and a potential therapeutic target.
- Published
- 2021
18. Lipid Droplet-Derived Monounsaturated Fatty Acids Traffic via PLIN5 to Allosterically Activate SIRT1
- Author
-
Mallory P. Franklin, Mark J. Graham, Linnea E. Mead, Timothy D. Heden, Kenneth K. Karanja, Bruce A. Witthuhn, Laurie L. Parker, Charles P. Najt, Douglas G. Mashek, Lisa S. Chow, Minervo Perez, Mara T. Mashek, Jason L. Heier, Salmaan A. Khan, and David A. Bernlohr
- Subjects
Male ,Transcription, Genetic ,Allosteric regulation ,Endogeny ,Biology ,Perilipin-5 ,Article ,Cell Line ,Fatty Acids, Monounsaturated ,03 medical and health sciences ,0302 clinical medicine ,Allosteric Regulation ,Sirtuin 1 ,Lipid droplet ,Animals ,Lipolysis ,Olive Oil ,Molecular Biology ,Cells, Cultured ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,Fatty Acids ,food and beverages ,Fatty acid ,Biological Transport ,Lipase ,Lipid Droplets ,Cell Biology ,Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha ,Diet ,Cell biology ,Mice, Inbred C57BL ,chemistry ,Mitochondrial biogenesis ,Perilipin ,lipids (amino acids, peptides, and proteins) ,030217 neurology & neurosurgery ,Function (biology) - Abstract
Lipid droplets (LDs) provide a reservoir for triacylglycerol storage and are a central hub for fatty acid trafficking and signaling in cells. Lipolysis promotes mitochondrial biogenesis and oxidative metabolism via a SIRT1/PGC-1α/PPARα-dependent pathway through an unknown mechanism. Herein, we identify that monounsaturated fatty acids (MUFAs) allosterically activate SIRT1 towards select peptide-substrates such as PGC-1α. MUFAs enhance PGC-1α/PPARα signaling and promote oxidative metabolism in cells and animal models in a SIRT1 dependent manner. Moreover, we characterize the LD protein perilipin 5 (PLIN5), which is known to enhance mitochondrial biogenesis and function, to be a fatty acid binding protein that preferentially binds LD-derived monounsaturated fatty acids (MUFAs) and traffics them to the nucleus following cAMP/PKA-mediated lipolytic stimulation. Thus, these studies identify the first-known endogenous allosteric modulators of SIRT1 and characterize a LD-nuclear signaling axis that underlies the known metabolic benefits of MUFAs and PLIN5.
- Published
- 2020
- Full Text
- View/download PDF
19. Adipose oxidative stress and protein carbonylation
- Author
-
David A. Bernlohr, Ann V. Hertzel, Yimao Huang, and Amy K. Hauck
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Protein Carbonylation ,Adipose tissue ,medicine.disease_cause ,Biochemistry ,03 medical and health sciences ,chemistry.chemical_compound ,Insulin resistance ,Lipid oxidation ,Adipocyte ,Internal medicine ,medicine ,Animals ,Humans ,Molecular Biology ,chemistry.chemical_classification ,Reactive oxygen species ,030102 biochemistry & molecular biology ,Endoplasmic reticulum ,JBC Reviews ,Proteins ,Cell Biology ,medicine.disease ,Oxidative Stress ,030104 developmental biology ,Endocrinology ,chemistry ,Adipose Tissue ,Reactive Oxygen Species ,Oxidative stress - Abstract
Increased oxidative stress and abundance of reactive oxygen species (ROS) are positively correlated with a variety of pathophysiologies, including cardiovascular disease, type 2 diabetes, Alzheimer's disease, and neuroinflammation. In adipose biology, diabetic obesity is correlated with increased ROS in an age- and depot-specific manner and is mechanistically linked to mitochondrial dysfunction, endoplasmic reticulum (ER) stress, potentiated lipolysis, and insulin resistance. The cellular quality control systems that homeostatically regulate oxidative stress in the lean state are down-regulated in obesity as a consequence of inflammatory cytokine pressure leading to the accumulation of oxidized biomolecules. New findings have linked protein, DNA, and lipid oxidation at the biochemical level, and the structures and potential functions of protein adducts such as carbonylation that accumulate in stressed cells have been characterized. The sum total of such regulation and biochemical changes results in alteration of cellular metabolism and function in the obese state relative to the lean state and underlies metabolic disease progression. In this review, we discuss the molecular mechanisms and events underlying these processes and their implications for human health and disease.
- Published
- 2018
20. Obesity-induced protein carbonylation in murine adipose tissue regulates the DNA-binding domain of nuclear zinc finger proteins
- Author
-
David A. Bernlohr, Wendy S. Hahn, Amy K. Hauck, Yue Chen, Raphael Petegrosso, Rui Kuang, and Tong Zhou
- Subjects
0301 basic medicine ,Protein Carbonylation ,Biochemistry ,DNA-binding protein ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Animals ,Amino Acid Sequence ,Obesity ,Nuclear protein ,Molecular Biology ,Transcription factor ,Zinc finger ,Cell Nucleus ,Aldehydes ,Chemistry ,Nuclear Proteins ,Zinc Fingers ,Cell Biology ,DNA-binding domain ,Cell biology ,DNA-Binding Proteins ,Oxidative Stress ,030104 developmental biology ,Metabolism ,Adipose Tissue ,Lipid modification ,Carbonylation ,030217 neurology & neurosurgery - Abstract
In obesity-linked insulin resistance, oxidative stress in adipocytes leads to lipid peroxidation and subsequent carbonylation of proteins by diffusible lipid electrophiles. Reduction in oxidative stress attenuates protein carbonylation and insulin resistance, suggesting that lipid modification of proteins may play a role in metabolic disease, but the mechanisms remain incompletely understood. Herein, we show that in vivo, diet-induced obesity in mice surprisingly results in preferential carbonylation of nuclear proteins by 4-hydroxy-trans-2,3-nonenal (4-HNE) or 4-hydroxy-trans-2,3-hexenal (4-HHE). Proteomic and structural analyses revealed that residues in or around the sites of zinc coordination of zinc finger proteins, such as those containing the C2H2 or MATRIN, RING, C3H1, or N4-type DNA-binding domains, are particularly susceptible to carbonylation by lipid aldehydes. These observations strongly suggest that carbonylation functionally disrupts protein secondary structure supported by metal coordination. Analysis of one such target, the nuclear protein estrogen-related receptor γ (ERR-γ), showed that ERR-γ is modified by 4-HHE in the obese state. In vitro carbonylation decreased the DNA-binding capacity of ERR-γ and correlated with the obesity-linked down-regulation of many key genes promoting mitochondrial bioenergetics. Taken together, these findings reveal a novel mechanistic connection between oxidative stress and metabolic dysfunction arising from carbonylation of nuclear zinc finger proteins, such as the transcriptional regulator ERR-γ.
- Published
- 2018
21. Multi-omic profiling of tyrosine kinase inhibitor-resistant K562 cells suggests metabolic reprogramming to promote cell survival
- Author
-
David A. Bernlohr, Tzu Yi Yang, Laura J. Marholz, Brett M Noel, Laurie L. Parker, Vinh Nguyen, Connor Navis, Zohar Sachs, Steven B. Ouellette, Deborah M. Dickey, and Sarah J. Parker
- Subjects
Cell Survival ,medicine.drug_class ,Dasatinib ,Biology ,Biochemistry ,Article ,Tyrosine-kinase inhibitor ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,Gene expression ,medicine ,Humans ,Glycolysis ,Gene ,Protein Kinase Inhibitors ,030304 developmental biology ,0303 health sciences ,Kinase ,General Chemistry ,Protein-Tyrosine Kinases ,Cell biology ,respiratory tract diseases ,3. Good health ,Metabolic pathway ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Imatinib Mesylate ,Metabolome ,Cancer research ,K562 Cells ,Tyrosine kinase ,K562 cells - Abstract
Resistance to chemotherapy can occur through a wide variety of mechanisms. Resistance to tyrosine kinase inhibitors (TKIs) often arises from kinase mutations-however, “off-target” resistance occurs but is poorly understood. Previously, we established cell line resistance models for three TKIs used in chronic myeloid leukemia treatment, and found that resistance was not attributed entirely to failure of kinase inhibition. Here, we performed global, integrated proteomic and transcriptomic profiling of these cell lines to describe mechanisms of resistance at the protein and gene expression level. We used whole transcriptome sequencing and SWATH-based data-independent acquisition mass spectrometry (DIA-MS), which does not require isotopic labels and provides quantitative measurements of proteins in a comprehensive, unbiased fashion. The proteomic and transcriptional data were correlated to generate an integrated understanding of the gene expression and protein alterations associated with TKI resistance. We defined mechanisms of resistance and two novel markers, CA1 and alpha-synuclein, that were common to all TKIs tested. Resistance to all of the TKIs was associated with oxidative stress responses, hypoxia signatures, and apparent metabolic reprogramming of the cells. Metabolite profiling and glucose-dependence experiments showed that resistant cells had routed their metabolism through glycolysis (particularly through the pentose phosphate pathway) and exhibited disruptions in mitochondrial metabolism. These experiments are the first to report a global, integrated proteomic, transcriptomic and metabolic analysis of TKI resistance. These data suggest that although the mechanisms are complex, targeting metabolic pathways along with TKI treatment may overcome pan-TKI resistance.Key Points:Alterations to metabolism are a common feature of target-mutation-independent resistance in CML cells across multiple clinically relevant TKIs.Carbonic anhydrase 1 (CA1) and a-synuclein (SNCA) are novel markers of metabolic reprogramming in TKI resistant CML cells.
- Published
- 2018
- Full Text
- View/download PDF
22. FABP4 regulates eosinophil recruitment and activation in allergic airway inflammation
- Author
-
Mythili Dileepan, David A. Bernlohr, Sung Gil Ha, P. Sriramarao, Idil Bastan, Savita P. Rao, Xiao Na Ge, Kaylee Steen, and Yana G. Greenberg
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,Male ,Allergic airway inflammation ,Physiology ,MAP Kinase Signaling System ,Inflammation ,Fatty Acid-Binding Proteins ,Fatty acid-binding protein ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cell Movement ,Physiology (medical) ,Cell Adhesion ,Hypersensitivity ,Medicine ,Animals ,Mice, Knockout ,Mitogen-Activated Protein Kinase 1 ,Mitogen-Activated Protein Kinase 3 ,business.industry ,Allergic asthma ,Cell Biology ,Eosinophil ,respiratory system ,Eosinophils ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,Immunology ,Cytokines ,medicine.symptom ,business ,Intracellular ,030215 immunology ,Research Article - Abstract
Fatty acid binding protein 4 (FABP4), a member of a family of lipid-binding proteins, is known to play a role in inflammation by virtue of its ability to regulate intracellular events such as lipid fluxes and signaling. Studies have indicated a proinflammatory role for FABP4 in allergic asthma although its expression and function in eosinophils, the predominant inflammatory cells recruited to allergic airways, were not investigated. We examined expression of FABP4 in murine eosinophils and its role in regulating cell recruitment in vitro as well as in cockroach antigen (CRA)-induced allergic airway inflammation. CRA exposure led to airway recruitment of FABP4-expressing inflammatory cells, specifically eosinophils, in wild-type (WT) mice. FABP4 expression in eosinophils was induced by TNF-α as well as IL-4 and IL-13. FABP4-deficient eosinophils exhibited markedly decreased cell spreading/formation of leading edges on vascular cell adhesion molecule-1 and significantly decreased adhesion to intercellular adhesion molecule-1 associated with reduced β2-integrin expression relative to WT cells. Furthermore, FABP4-deficient eosinophils exhibited decreased migration, F-actin polymerization, calcium flux, and ERK(1/2) phosphorylation in response to eotaxin-1. In vivo, CRA-challenged FABP4-deficient mice exhibited attenuated eosinophilia and significantly reduced airway inflammation (improved airway reactivity, lower IL-5, IL-13, TNF-α, and cysteinyl leukotriene C4 levels, decreased airway structural changes) compared with WT mice. In conclusion, expression of FABP4 in eosinophils is induced during conditions of inflammation and plays a proinflammatory role in the development of allergic asthma by promoting eosinophil adhesion and migration and contributing to the development of various aspects of airway inflammation.
- Published
- 2018
23. Metabolic functions of FABPs—mechanisms and therapeutic implications
- Author
-
David A. Bernlohr and Gökhan S. Hotamisligil
- Subjects
Inflammation ,Protein family ,Drug discovery ,Endocrinology, Diabetes and Metabolism ,Gene Expression ,Lipid metabolism ,Biology ,Fatty Acid-Binding Proteins ,Lipid Metabolism ,Article ,Fatty acid-binding protein ,Cell biology ,Mice ,Endocrinology ,Biochemistry ,Extracellular ,Animals ,Humans ,Glucose homeostasis ,Secretion ,Intracellular - Abstract
Intracellular and extracellular interactions with proteins enables the functional and mechanistic diversity of lipids. Fatty acid-binding proteins (FABPs) were originally described as intracellular proteins that can affect lipid fluxes, metabolism and signalling within cells. As the functions of this protein family have been further elucidated, it has become evident that they are critical mediators of metabolism and inflammatory processes, both locally and systemically, and therefore are potential therapeutic targets for immunometabolic diseases. In particular, genetic deficiency and small molecule-mediated inhibition of FABP4 (also known as aP2) and FABP5 can potently improve glucose homeostasis and reduce atherosclerosis in mouse models. Further research has shown that in addition to their intracellular roles, some FABPs are found outside the cells, and FABP4 undergoes regulated, vesicular secretion. The circulating form of FABP4 has crucial hormonal functions in systemic metabolism. In this Review we discuss the roles and regulation of both intracellular and extracellular FABP actions, highlighting new insights that might direct drug discovery efforts and opportunities for management of chronic metabolic diseases.
- Published
- 2015
- Full Text
- View/download PDF
24. Uncoupling Lipid Metabolism from Inflammation through Fatty Acid Binding Protein-Dependent Expression of UCP2
- Author
-
Kaylee Steen, Ann V. Hertzel, David A. Bernlohr, Qigui Wang, Jill Suttles, and Hongliang Xu
- Subjects
Lipopolysaccharides ,Adipose tissue ,Inflammation ,Fatty Acids, Nonesterified ,Mitochondrion ,Biology ,Endoplasmic Reticulum ,Fatty Acid-Binding Proteins ,Ion Channels ,Fatty acid-binding protein ,Cell Line ,Mitochondrial Proteins ,Mice ,medicine ,Animals ,Uncoupling Protein 2 ,Obesity ,RNA, Messenger ,Molecular Biology ,UCP3 ,Mice, Knockout ,Macrophages ,Endoplasmic reticulum ,food and beverages ,Articles ,Cell Biology ,Lipid Metabolism ,Molecular biology ,Mitochondria ,Cell biology ,Mice, Inbred C57BL ,Adipose Tissue ,Unfolded protein response ,lipids (amino acids, peptides, and proteins) ,medicine.symptom ,Reactive Oxygen Species ,Intracellular - Abstract
Chronic inflammation in obese adipose tissue is linked to endoplasmic reticulum (ER) stress and systemic insulin resistance. Targeted deletion of the murine fatty acid binding protein (FABP4/aP2) uncouples obesity from inflammation although the mechanism underlying this finding has remained enigmatic. Here, we show that inhibition or deletion of FABP4/aP2 in macrophages results in increased intracellular free fatty acids (FFAs) and elevated expression of uncoupling protein 2 (UCP2) without concomitant increases in UCP1 or UCP3. Silencing of UCP2 mRNA in FABP4/aP2-deficient macrophages negated the protective effect of FABP loss and increased ER stress in response to palmitate or lipopolysaccharide (LPS). Pharmacologic inhibition of FABP4/aP2 with the FABP inhibitor HTS01037 also upregulated UCP2 and reduced expression of BiP, CHOP, and XBP-1s. Expression of native FABP4/aP2 (but not the non-fatty acid binding mutant R126Q) into FABP4/aP2 null cells reduced UCP2 expression, suggesting that the FABP-FFA equilibrium controls UCP2 expression. FABP4/aP2-deficient macrophages are resistant to LPS-induced mitochondrial dysfunction and exhibit decreased mitochondrial protein carbonylation and UCP2-dependent reduction in intracellular reactive oxygen species. These data demonstrate that FABP4/aP2 directly regulates intracellular FFA levels and indirectly controls macrophage inflammation and ER stress by regulating the expression of UCP2.
- Published
- 2015
- Full Text
- View/download PDF
25. Down regulation of Peroxiredoxin-3 in 3T3-L1 adipocytes leads to oxidation of Rictor in the mammalian-target of rapamycin complex 2 (mTORC2)
- Author
-
Dalay H. Olson, Do Hyung Kim, Jovan Kuzmicic, David A. Bernlohr, Ji Man Park, Joel S. Burrill, and Wendy S. Hahn
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Peroxiredoxin III ,medicine.medical_treatment ,Biophysics ,Down-Regulation ,030209 endocrinology & metabolism ,Mechanistic Target of Rapamycin Complex 2 ,Biology ,Biochemistry ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Insulin resistance ,Downregulation and upregulation ,Internal medicine ,Mitochondrial unfolded protein response ,3T3-L1 Cells ,medicine ,Adipocytes ,Animals ,Insulin ,Phosphorylation ,Molecular Biology ,Protein kinase B ,TOR Serine-Threonine Kinases ,Biological Transport ,Cell Biology ,medicine.disease ,PRDX3 ,Acetylcysteine ,Mitochondria ,Mice, Inbred C57BL ,Insulin receptor ,Oxidative Stress ,030104 developmental biology ,Endocrinology ,Glucose ,Rapamycin-Insensitive Companion of mTOR Protein ,Multiprotein Complexes ,Unfolded protein response ,biology.protein ,Insulin Resistance ,Carrier Proteins ,Oxidation-Reduction ,Proto-Oncogene Proteins c-akt - Abstract
Mitochondrially-derived oxidative stress has been implicated in the development of obesity-induced insulin resistance and is correlated with down regulation of Peroxiredoxin-3 (Prdx3). Prdx3 knockout mice exhibit whole-body insulin resistance, while Prdx3 transgenic animals remain insulin sensitive when placed on a high fat diet. To define the molecular events linking mitochondrial oxidative stress to insulin action, Prdx3 was silenced in 3T3-L1 adipocytes (Prdx3 KD) and the resultant cells evaluated for mitochondrial function, endoplasmic reticulum stress (ER stress), mitochondrial unfolded protein response (mtUPR) and insulin signaling. Prdx3 KD cells exhibit a two-fold increase in H2O2, reduced insulin-stimulated glucose transport and attenuated S473 phosphorylation of the mTORC2 substrate, Akt. Importantly, the decrease in glucose uptake can be rescued by pre-treatment with the antioxidant N-acetyl-cysteine (NAC). The changes in insulin sensitivity occur independently from activation of the ER stress or mtUPR pathways. Analysis of mTORC2, the complex responsible for phosphorylating Akt at S473, reveals increased cysteine oxidation of Rictor in Prdx3 KD cells that can be rescued with NAC. Taken together, these data suggest mitochondrial dysfunction in adipocytes may attenuate insulin signaling via oxidation of the mammalian-target of rapamycin complex 2 (mTORC2).
- Published
- 2017
26. Lipocalin 2 Regulates Brown Fat Activation via a Nonadrenergic Activation Mechanism
- Author
-
Jessica A. Deis, David A. Bernlohr, Aníbal G. Armién, Douglas G. Mashek, Mara G. Mashek, DonSanjiv Ariyakumar, Xiaoli Chen, Hong Guo, Yuanyuan Zhang, and Ming Zhao
- Subjects
medicine.medical_specialty ,p38 mitogen-activated protein kinases ,Gene Expression ,Adipose tissue ,Peroxisome proliferator-activated receptor ,Mitochondrion ,Biology ,p38 Mitogen-Activated Protein Kinases ,Biochemistry ,Ion Channels ,Mitochondrial Proteins ,Mice ,Catecholamines ,Adipose Tissue, Brown ,Lipocalin-2 ,Internal medicine ,Brown adipose tissue ,Peroxisomes ,medicine ,Animals ,Phosphorylation ,Receptor ,Molecular Biology ,Uncoupling Protein 1 ,Mice, Knockout ,Oncogene Proteins ,chemistry.chemical_classification ,integumentary system ,Thermogenesis ,Cell Biology ,Lipid Metabolism ,Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha ,Lipocalins ,Mitochondria ,Mice, Inbred C57BL ,PPAR gamma ,Metabolism ,Adipocytes, Brown ,Endocrinology ,medicine.anatomical_structure ,chemistry ,Oxidation-Reduction ,Acute-Phase Proteins ,Transcription Factors - Abstract
In this study, we report that lipocalin 2 (Lcn2), a recently characterized adipokine/cytokine, is a novel regulator of brown adipose tissue (BAT) activation by modulating the adrenergic independent p38 MAPK-PGC-1α-UCP1 pathway. Global Lcn2 knock-out (Lcn2(-/-)) mice have defective BAT thermogenic activation caused by cold stimulation and decreased BAT activity under high fat diet-induced obesity. Nevertheless, Lcn2(-/-) mice maintain normal sympathetic nervous system activation as evidenced by normal catecholamine release and lipolytic activity in response to cold stimulation. Further studies showed that Lcn2 deficiency impairs peroxisomal and mitochondrial oxidation of lipids and attenuates cold-induced Pgc1a and Ucp1 expression and p38 MAPK phosphorylation in BAT. Moreover, in vitro studies showed that Lcn2 deficiency reduces the thermogenic activity of brown adipocytes. Lcn2(-/-) differentiated brown adipocytes have significantly decreased expression levels of brown fat markers, decreased p38 MAPK phosphorylation, and decreased mitochondrial oxidation capacity. However, Lcn2(-/-) brown adipocytes have normal norepinephrine-stimulated p38 MAPK and hormone-sensitive lipase phosphorylation and Pgc1a and Ucp1 expression, suggesting an intact β-adrenergic signaling activation. More intriguingly, recombinant Lcn2 was able to significantly stimulate p38 MAPK phosphorylation in brown adipocytes. Activating peroxisome proliferator-activated receptor γ, a downstream effector of PGC-1α, by thiazolidinedione administration fully reverses the BAT function of Lcn2(-/-) mice. Our findings provide evidence for the novel role Lcn2 plays in oxidative metabolism and BAT activation via an adrenergic independent mechanism.
- Published
- 2014
- Full Text
- View/download PDF
27. Glutathionylated products of lipid peroxidation
- Author
-
Brigitte I. Frohnert and David A. Bernlohr
- Subjects
medicine.medical_specialty ,Histology ,glutathionylated lipids ,Adipose tissue macrophages ,Adipose tissue ,Inflammation ,Biology ,medicine.disease_cause ,Impaired glucose tolerance ,Lipid peroxidation ,chemistry.chemical_compound ,Insulin resistance ,Internal medicine ,Adipocyte ,medicine ,oxidative stress ,glutathione S-transferase ,lipid peroxidation ,Cell Biology ,medicine.disease ,Endocrinology ,chemistry ,inflammation ,Commentary ,medicine.symptom ,Oxidative stress - Abstract
Obesity-associated insulin resistance has long been linked to both increased adipocyte oxidative stress as well as the presence of inflammatory changes in adipose tissue, including the infiltration and activation of tissue-resident macrophages. In order to investigate the connections between obesity-associated oxidative stress in adipocytes and increased inflammation in adipose tissue associated with the development of insulin resistance, our laboratory recently demonstrated that adipocytes form glutathionylated products of oxidative stress including glutathionyl-4-hydroxy-2-nonenal (GS-HNE) and glutathionyl-1,4-dihydroxynonene (GS-DHN). The abundance of both GS-HNE and GS-DHN were increased in the visceral adipose tissue of ob/ob mice and diet-induced obese, insulin-resistant mice. Further, these products of lipid peroxidation were shown to induce inflammatory changes in macrophages. Finally, in a mouse model, overproduction of GS-HNE was associated with increased fasting glucose levels and moderately impaired glucose tolerance. Together, these findings suggest a novel mechanism by which obesity-induced oxidative stress in adipocytes may lead to activation of tissue-resident macrophages. As adipose tissue inflammation has been shown to play an important role in the development of insulin resistance, further study of this pathway may lead to potential interventions to attenuate the metabolic consequences of obesity.
- Published
- 2014
- Full Text
- View/download PDF
28. FABP4/aP2 Regulates Macrophage Redox Signaling and Inflammasome Activation via Control of UCP2
- Author
-
Hongliang Xu, David A. Bernlohr, and Kaylee Steen
- Subjects
Male ,0301 basic medicine ,Proteasome Endopeptidase Complex ,Inflammasomes ,Interleukin-1beta ,Caspase 1 ,SOD2 ,Bone Marrow Cells ,Mitochondrion ,Biology ,Diet, High-Fat ,Fatty Acid-Binding Proteins ,Antioxidants ,Proinflammatory cytokine ,03 medical and health sciences ,ATP-Dependent Proteases ,Downregulation and upregulation ,Heat shock protein ,NLR Family, Pyrin Domain-Containing 3 Protein ,medicine ,Animals ,Homeostasis ,Uncoupling Protein 2 ,Cysteine ,RNA, Messenger ,Molecular Biology ,Macrophages ,food and beverages ,Inflammasome ,Chaperonin 60 ,Hydrogen Peroxide ,Cell Biology ,Mitochondria ,Up-Regulation ,Cell biology ,PSMB5 ,Mice, Inbred C57BL ,Oxidative Stress ,Protein Subunits ,030104 developmental biology ,Stromal Cells ,Oxidation-Reduction ,Gene Deletion ,Research Article ,Signal Transduction ,medicine.drug - Abstract
Obesity-linked metabolic disease is mechanistically associated with the accumulation of proinflammatory macrophages in adipose tissue, leading to increased reactive oxygen species (ROS) production and chronic low-grade inflammation. Previous work has demonstrated that deletion of the adipocyte fatty acid-binding protein (FABP4/aP2) uncouples obesity from inflammation via upregulation of the uncoupling protein 2 (UCP2). Here, we demonstrate that ablation of FABP4/aP2 regulates systemic redox capacity and reduces cellular protein sulfhydryl oxidation and, in particular, oxidation of mitochondrial protein cysteine residues. Coincident with the loss of FABP4/aP2 is the upregulation of the antioxidants superoxide dismutase (SOD2), catalase, methionine sulfoxide reductase A, and the 20S proteasome subunits PSMB5 and αβ. Reduced mitochondrial protein oxidation in FABP4/aP2−/− macrophages attenuates the mitochondrial unfolded-protein response (mtUPR) as measured by expression of heat shock protein 60, Clp protease, and Lon peptidase 1. Consistent with a diminished mtUPR, FABP4/aP2−/− macrophages exhibit reduced expression of cleaved caspase-1 and NLRP3. Secretion of interleukin 1β (IL-1β), in response to inflammasome activation, is ablated in FABP4/aP2−/− macrophages, as well as in FABP4/aP2 inhibitor-treated cells, but partially rescued in FABP4/aP2-null macrophages when UCP2 is silenced. Collectively, these data offer a novel pathway whereby FABP4/aP2 regulates macrophage redox signaling and inflammasome activation via control of UCP2 expression.
- Published
- 2017
- Full Text
- View/download PDF
29. Oxidative stress and lipotoxicity
- Author
-
David A. Bernlohr and Amy K. Hauck
- Subjects
0301 basic medicine ,Protein Carbonylation ,QD415-436 ,medicine.disease_cause ,Biochemistry ,Antioxidants ,Lipid peroxidation ,03 medical and health sciences ,chemistry.chemical_compound ,Endocrinology ,oxidized lipids ,Cardiolipin ,medicine ,Animals ,Humans ,Aldehydes ,Endoplasmic reticulum ,Proteins ,Lipid metabolism ,Thematic Review Series ,Cell Biology ,Endoplasmic Reticulum Stress ,Lipid Metabolism ,Cell biology ,Oxidative Stress ,030104 developmental biology ,chemistry ,Lipotoxicity ,lipids/peroxidation ,Lipid Peroxidation ,Signal transduction ,cardiolipin ,Reactive Oxygen Species ,Oxidative stress ,Signal Transduction - Abstract
The α,β polyunsaturated lipid aldehydes are potent lipid electrophiles that covalently modify lipids, proteins, and nucleic acids. Recent work highlights the critical role these lipids play under both physiological and pathological conditions. Protein carbonylation resulting from nucleophilic attack of lysine, histidine, and cysteine residues is a major outcome of oxidative stress and functions as a redox-sensitive signaling mechanism with roles in autophagy, cell proliferation, transcriptional control, and apoptosis. In addition, protein carbonylation is implicated as an initiating factor in mitochondrial dysfunction and endoplasmic reticulum stress, providing a mechanistic connection between oxidative stress and metabolic disease. In this review, we discuss the generation and metabolism of reactive lipid aldehydes, as well as their signaling roles.
- Published
- 2016
30. Identification of a fatty acid binding protein4-UCP2 axis regulating microglial mediated neuroinflammation
- Author
-
Tammy A. Butterick, Joshua P. Nixon, Cayla M. Duffy, Hongliang Xu, and David A. Bernlohr
- Subjects
0301 basic medicine ,Anti-Inflammatory Agents ,Hypothalamus ,Palmitic Acid ,Nitric Oxide Synthase Type II ,Inflammation ,Mice, Transgenic ,Biology ,Fatty Acid-Binding Proteins ,Article ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Mice ,Fatty acid binding ,medicine ,Animals ,Uncoupling Protein 2 ,RNA, Small Interfering ,Molecular Biology ,Neuroinflammation ,Cell Line, Transformed ,Regulation of gene expression ,Microglia ,Arginase ,Tumor Necrosis Factor-alpha ,Calcium-Binding Proteins ,Microfilament Proteins ,Brain ,Cell Biology ,Cell biology ,Nitric oxide synthase ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,Immunology ,biology.protein ,Tumor necrosis factor alpha ,medicine.symptom ,Signal transduction ,Reactive Oxygen Species ,Signal Transduction - Abstract
Hypothalamic inflammation contributes to metabolic dysregulation and the onset of obesity. Dietary saturated fats activate microglia via a nuclear factor-kappa B (NFκB) mediated pathway to release pro-inflammatory cytokines resulting in dysfunction or death of surrounding neurons. Fatty acid binding proteins (FABPs) are lipid chaperones regulating metabolic and inflammatory pathways in response to fatty acids. Loss of FABP4 in peripheral macrophages via either molecular or pharmacologic mechanisms results in reduced obesity-induced inflammation via a UCP2-redox based mechanism. Despite the widespread appreciation for the role of FABP4 in mediating peripheral inflammation, the expression of FABP4 and a potential FABP4-UCP2 axis regulating microglial inflammatory capacity is largely uncharacterized. To that end, we hypothesized that microglial cells express FABP4 and that inhibition would upregulate UCP2 and attenuate palmitic acid (PA)-induced pro-inflammatory response. Gene expression confirmed expression of FABP4 in brain tissue lysate from C57Bl/6J mice and BV2 microglia. Treatment of microglial cells with an FABP inhibitor (HTS01037) increased expression of Ucp2 and arginase in the presence or absence of PA. Moreover, cells exposed to HTS01037 exhibited attenuated expression of inducible nitric oxide synthase (iNOS) compared to PA alone indicating reduced NFκB signaling. Hypothalamic tissue from mice lacking FABP4 exhibit increased UCP2 expression and reduced iNOS, tumor necrosis factor-alpha (TNF-α), and ionized calcium-binding adapter molecule 1 (Iba1; microglial activation marker) expression compared to wild type mice. Further, this effect is negated in microglia lacking UCP2, indicating the FABP4-UCP2 axis is pivotal in obesity induced neuroinflammation. To our knowledge, this is the first report demonstrating a FABP4-UCP2 axis with the potential to modulate the microglial inflammatory response.
- Published
- 2016
31. Adipose Fatty Acid Binding Protein Promotes Saturated Fatty Acid-Induced Macrophage Cell Death through Enhancing Ceramide Production
- Author
-
Bing Li, Jun Zeng, Edward R. Sauter, Shujun Liu, Jill Suttles, Enyu Rao, Yuwen Zhang, Jiaqing Hao, Margot P. Cleary, Yanwen Sun, and David A. Bernlohr
- Subjects
0301 basic medicine ,Programmed cell death ,Ceramide ,Immunology ,Blotting, Western ,Adipose tissue ,Inflammation ,Biology ,Ceramides ,Diet, High-Fat ,Fatty Acid-Binding Proteins ,Real-Time Polymerase Chain Reaction ,Fatty acid-binding protein ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Microscopy, Electron, Transmission ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,Animals ,Obesity ,Mice, Knockout ,Microscopy, Confocal ,Cell Death ,Binding protein ,Macrophages ,Fatty Acids ,Flow Cytometry ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,chemistry ,Biochemistry ,030220 oncology & carcinogenesis ,Gene Knockdown Techniques ,Saturated fatty acid ,lipids (amino acids, peptides, and proteins) ,medicine.symptom - Abstract
Macrophages play a critical role in obesity-associated chronic inflammation and disorders. However, the molecular mechanisms underlying the response of macrophages to elevated fatty acids (FAs) and their contribution to metabolic inflammation in obesity remain to be fully elucidated. In this article, we report a new mechanism by which dietary FAs, in particular, saturated FAs (sFAs), are able to directly trigger macrophage cell death. We demonstrated that excess sFAs, but not unsaturated FAs, induced the production of cytotoxic ceramides (Cers) in macrophage cell lines. Most importantly, expression of adipose FA binding protein (A-FABP) in macrophages facilitated metabolism of excess sFAs for Cer synthesis. Inhibition or deficiency of A-FABP in macrophage cell lines decreased sFA-induced Cer production, thereby resulting in reduced cell death. Furthermore, we validated the role of A-FABP in promoting sFA-induced macrophage cell death with primary bone marrow–derived macrophages and high-fat diet–induced obese mice. Altogether, our data reveal that excess dietary sFAs may serve as direct triggers in induction of Cer production and macrophage cell death through elevated expression of A-FABP, thus establishing A-FABP as a new molecular sensor in triggering macrophage-associated sterile inflammation in obesity.
- Published
- 2016
32. Protein Carbonylation and Adipocyte Mitochondrial Function
- Author
-
Margaret A. Donoghue, David A. Bernlohr, Eric K. Long, Sergio Lavandero, Timothy J. Griffin, Edgar A. Arriaga, Jessica Curtis, Wendy S. Hahn, Aníbal G. Armién, Jacob J. Inda, Matthew D. Stone, David J. Droullard, and Jovan Kuzmicic
- Subjects
Protein Carbonylation ,Mitochondrion ,Biochemistry ,Mitochondrial Proteins ,Mice ,chemistry.chemical_compound ,Oxygen Consumption ,3T3-L1 Cells ,Adipocyte ,Adipocytes ,Animals ,Translocase ,Gene Silencing ,Inner mitochondrial membrane ,Molecular Biology ,biology ,Superoxide ,NADH dehydrogenase ,Cell Biology ,Mitochondria ,Metabolism ,chemistry ,biology.protein ,ATP–ADP translocase ,Insulin Resistance - Abstract
Carbonylation is the covalent, non-reversible modification of the side chains of cysteine, histidine, and lysine residues by lipid peroxidation end products such as 4-hydroxy- and 4-oxononenal. In adipose tissue the effects of such modifications are associated with increased oxidative stress and metabolic dysregulation centered on mitochondrial energy metabolism. To address the role of protein carbonylation in the pathogenesis of mitochondrial dysfunction, quantitative proteomics was employed to identify specific targets of carbonylation in GSTA4-silenced or overexpressing 3T3-L1 adipocytes. GSTA4-silenced adipocytes displayed elevated carbonylation of several key mitochondrial proteins including the phosphate carrier protein, NADH dehydrogenase 1α subcomplexes 2 and 3, translocase of inner mitochondrial membrane 50, and valyl-tRNA synthetase. Elevated protein carbonylation is accompanied by diminished complex I activity, impaired respiration, increased superoxide production, and a reduction in membrane potential without changes in mitochondrial number, area, or density. Silencing of the phosphate carrier or NADH dehydrogenase 1α subcomplexes 2 or 3 in 3T3-L1 cells results in decreased basal and maximal respiration. These results suggest that protein carbonylation plays a major instigating role in cytokine-dependent mitochondrial dysfunction and may be linked to the development of insulin resistance in the adipocyte.
- Published
- 2012
- Full Text
- View/download PDF
33. Stimulation of GLUT4 (glucose transporter isoform 4) storage vesicle formation by sphingolipid depletion
- Author
-
David A. Bernlohr, David L. Marks, Christine L. Wheatley, Zhi Jie Cheng, Richard E. Pagano, Eileen L. Holicky, Raman Deep Singh, and Teng Ke Wang
- Subjects
VAMP2 ,biology ,Glucose transporter ,nutritional and metabolic diseases ,Fluorescence recovery after photobleaching ,Cell Biology ,Glycosphingolipid ,Biochemistry ,Sphingolipid ,Transport protein ,Cell biology ,carbohydrates (lipids) ,chemistry.chemical_compound ,chemistry ,Insulin receptor substrate ,biology.protein ,lipids (amino acids, peptides, and proteins) ,Molecular Biology ,GLUT4 - Abstract
Insulin stimulates glucose transport in fat and skeletal muscle cells primarily by inducing the translocation of GLUT4 (glucose transporter isoform 4) to the PM (plasma membrane) from specialized GSVs (GLUT4 storage vesicles). Glycosphingolipids are components of membrane microdomains and are involved in insulin-regulated glucose transport. Cellular glycosphingolipids decrease during adipocyte differentiation and have been suggested to be involved in adipocyte function. In the present study, we investigated the role of glycosphingolipids in regulating GLUT4 translocation. We decreased glycosphingolipids in 3T3-L1 adipocytes using glycosphingolipid synthesis inhibitors and investigated the effects on GLUT4 translocation using immunocytochemistry, preparation of PM sheets, isolation of GSVs and FRAP (fluorescence recovery after photobleaching) of GLUT4–GFP (green fluorescent protein) in intracellular structures. Glycosphingolipids were located in endosomal vesicles in pre-adipocytes and redistributed to the PM with decreased expression at day 2 after initiation of differentiation. In fully differentiated adipocytes, depletion of glycosphingolipids dramatically accelerated insulin-stimulated GLUT4 translocation. Although insulin-induced phosphorylation of IRS (insulin receptor substrate) and Akt remained intact in glycosphingolipid-depleted cells, both in vitro budding of GLUT4 vesicles and FRAP of GLUT4–GFP on GSVs were stimulated. Glycosphingolipid depletion also enhanced the insulin-induced translocation of VAMP2 (vesicle-associated membrane protein 2), but not the transferrin receptor or cellubrevin, indicating that the effect of glycosphingolipids was specific to VAMP2-positive GSVs. Our results strongly suggest that decreasing glycosphingolipid levels promotes the formation of GSVs and, thus, GLUT4 translocation. These studies provide a mechanistic basis for recent studies showing that inhibition of glycosphingolipid synthesis improves glycaemic control and enhances insulin sensitivity in animal models of Type 2 diabetes.
- Published
- 2010
- Full Text
- View/download PDF
34. A novel role for fatty acid transport protein 1 in the regulation of tricarboxylic acid cycle and mitochondrial function in 3T3-L1 adipocytes
- Author
-
Brian M. Wiczer and David A. Bernlohr
- Subjects
oxidation ,Citric Acid Cycle ,QD415-436 ,Mitochondrion ,Biology ,Biochemistry ,Mice ,Endocrinology ,proteomics ,3T3-L1 Cells ,alpha-ketoglutarate dehydrogenase ,Adipocytes ,Animals ,fatty acid transport proteins ,Beta oxidation ,Cells, Cultured ,chemistry.chemical_classification ,Fatty Acid Transport Proteins ,Fatty acid ,Cell Biology ,Transport protein ,Cell biology ,Mice, Inbred C57BL ,Citric acid cycle ,mitochondria ,chemistry ,reconstitution ,Female ,Branched-chain alpha-keto acid dehydrogenase complex ,Oxoglutarate dehydrogenase complex ,Research Article - Abstract
Fatty acid transport proteins (FATPs) are integral membrane acyl-CoA synthetases implicated in adipocyte fatty acid influx and esterification. Whereas some FATP1 translocates to the plasma membrane in response to insulin, the majority of FATP1 remains within intracellular structures and bioinformatic and immunofluorescence analysis of FATP1 suggests the protein primarily resides in the mitochondrion. To evaluate potential roles for FATP1 in mitochondrial metabolism, we used a proteomic approach following immunoprecipitation of endogenous FATP1 from 3T3-L1 adipocytes and identified mitochondrial 2-oxoglutarate dehydrogenase. To assess the functional consequence of the interaction, purified FATP1 was reconstituted into phospholipid-containing vesicles and its effect on 2-oxoglutarate dehydrogenase activity evaluated. FATP1 enhanced the activity of 2-oxoglutarate dehydrogenase independently of its acyl-CoA synthetase activity whereas silencing of FATP1 in 3T3-L1 adipocytes resulted in decreased activity of 2-oxoglutarate dehydrogenase. FATP1 silenced 3T3-L1 adipocytes exhibited decreased tricarboxylic acid cycle activity, increased cellular NAD(+)/NADH, increased fatty acid oxidation, and increased lactate production indicative of altered mitochondrial energy metabolism. These results reveal a novel role for FATP1 as a regulator of tricarboxylic acid cycle activity and mitochondrial function.
- Published
- 2009
35. FATP1 mediates fatty acid-induced activation of AMPK in 3T3-L1 adipocytes
- Author
-
Brian M. Wiczer, G. Luke Machen, David A. Bernlohr, Sandra Lobo, and Lee M. Graves
- Subjects
Biophysics ,Carbohydrate metabolism ,Biochemistry ,Mice ,chemistry.chemical_compound ,AMP-Activated Protein Kinase Kinases ,3T3-L1 Cells ,Adipocyte ,Coenzyme A Ligases ,Adipocytes ,Animals ,Insulin ,Phosphorylation ,adipocyte protein 2 ,Protein kinase A ,Molecular Biology ,chemistry.chemical_classification ,biology ,Chemistry ,Fatty Acid Transport Proteins ,Fatty Acids ,AMPK ,Fatty acid ,Cell Biology ,Enzyme Activation ,biology.protein ,Protein Kinases - Abstract
Fatty acid transport proteins are integral membrane acyl-CoA synthetases implicated in adipocyte fatty acid influx and esterification. FATP-dependent production of AMP was evaluated using FATP4 proteoliposomes, and fatty acid-dependent activation of AMP-activated protein kinase (AMPK) was assessed in 3T3-L1 adipocytes. Insulin-stimulated fatty acid influx (palmitate or arachidonate) into cultured adipocytes resulted in an increase in the phosphorylation of AMPK and its downstream target acetyl-CoA carboxylase. Consistent with the activation of AMPK, palmitate uptake into 3T3-L1 adipocytes resulted in an increase in intracellular [AMP]/[ATP]. The fatty acid-induced increase in AMPK activation was attenuated in a cell line expressing shRNA targeting FATP1. Taken together, these results demonstrate that, in adipocytes, insulin-stimulated fatty acid influx mediated by FATP1 regulates AMPK and provides a potential regulatory mechanism for balancing de novo production of fatty acids from glucose metabolism with influx of preformed fatty acids via phosphorylation of acetyl-CoA carboxylase.
- Published
- 2009
- Full Text
- View/download PDF
36. Interaction of Adipocyte Fatty Acid-binding Protein (AFABP) and JAK2
- Author
-
David A. Bernlohr, Jill Suttles, Brian R. Thompson, Anna M. Mazurkiewicz-Muñoz, and Christin Carter-Su
- Subjects
chemistry.chemical_classification ,HEK 293 cells ,food and beverages ,Fatty acid ,Cell Biology ,Plasma protein binding ,Biology ,Biochemistry ,Fatty acid-binding protein ,chemistry.chemical_compound ,chemistry ,Adipocyte ,Fatty acid binding ,Phosphorylation ,Signal transduction ,Molecular Biology - Abstract
Adipocyte fatty acid-binding protein (AFABP/aP2) facilitates the intracellular solubilization and trafficking of lipids within the aqueous environment of the cell. Studies in the AFABP/aP2 knock-out mouse suggest that the protein may have roles in cellular processes broader than lipid transport. We present herein the finding that AFABP/aP2 interacts with JAK2 in a fatty acid-dependent manner. This interaction was established using yeast two-hybrid analysis, co-immunoprecipitation from adipose tissue, and 3T3-L1 adipocytes as well as in 293 cells overexpressing JAK2 and AFABP/aP2. Mutational analysis of AFABP/aP2 (R126L/Y128F) revealed that fatty acid binding activity is necessary for the interaction and that Asp18 of the helix-turn-helix motif forms a component of the interaction domain. Mutational analysis of JAK2 (Y1007F/Y1008F) revealed that AFABP/aP2 associates with the basal unphosphorylated form of the protein. Interleukin-6, but not interleukin-10, stimulated phosphorylation of STAT3, and induction of SOCS3 mRNA expression were potentiated in a time- and dose-dependent manner in macrophage cell lines derived from AFABP/aP2-EFABP/mal1 double knock-out mice relative to cells from wild type animals. These results suggest that ligand-bound AFABP/aP2 binds to and attenuates JAK2 signaling and establishes a new role for AFABP/aP2 as a fatty acid sensor affecting cellular metabolism via protein-protein interactions.
- Published
- 2009
- Full Text
- View/download PDF
37. Mapping of the Hormone-sensitive Lipase Binding Site on the Adipocyte Fatty Acid-binding Protein (AFABP)
- Author
-
David A. Bernlohr, Ann V. Hertzel, Brittany E. Juhlmann, Anne J. Smith, and Mark A. Sanders
- Subjects
chemistry.chemical_classification ,Alanine ,Mutagenesis ,food and beverages ,Fatty acid ,Helix-turn-helix ,Hormone-sensitive lipase ,Cell Biology ,Biology ,Biochemistry ,Amino acid ,chemistry ,Fatty acid binding ,Binding site ,Molecular Biology - Abstract
The hormone-sensitive lipase (HSL) and adipocyte fatty acid-binding protein (AFABP/aP2) form a physical complex that affects basal and hormone-stimulated adipocyte fatty acid efflux. Previous work has established that AFABP/aP2-HSL complex formation requires that HSL be in its activated, phosphorylated form and AFABP/aP2 have a bound fatty acid. To identify the HSL binding site of AFABP/aP2 a combination of alanine-scanning mutagenesis and fluorescence resonance energy transfer was used. Mutation of Asp17, Asp18, Lys21, or Arg30 (but not other amino acids in the helix-turn-helix region) to alanine inhibited interaction with HSL without affecting fatty acid binding. The cluster of residues on the helical domain of AFABP/aP2 form two ion pairs (Asp17-Arg30 and Asp18-Lys21) and identifies the region we have termed the charge quartet as the HSL interaction site. To demonstrate direct association, the non-interacting AFABP/aP2-D18K mutant was rescued by complementary mutation of HSL (K196E). The charge quartet is conserved on other FABPs that interact with HSL such as the heart and epithelial FABPs but not on non-interacting proteins from the liver or intestine and may be a general protein interaction domain utilized by fatty acid-binding proteins in regulatory control of lipid metabolism.
- Published
- 2008
- Full Text
- View/download PDF
38. Loss of Fatty Acid Binding Protein 4/aP2 Reduces Macrophage Inflammation Through Activation of SIRT3
- Author
-
Kaylee Steen, Hongliang Xu, Ann V. Hertzel, and David A. Bernlohr
- Subjects
0301 basic medicine ,Lipopolysaccharides ,Male ,SIRT3 ,Inflammation ,Fatty Acid-Binding Proteins ,Protective Agents ,Fatty acid-binding protein ,Proinflammatory cytokine ,03 medical and health sciences ,Mice ,Endocrinology ,Sirtuin 3 ,medicine ,Animals ,adipocyte protein 2 ,Molecular Biology ,Beta oxidation ,Original Research ,biology ,Lysine ,Macrophages ,Fatty Acids ,food and beverages ,Acetylation ,General Medicine ,Cell biology ,Mitochondria ,Up-Regulation ,Nitric oxide synthase ,Mice, Inbred C57BL ,Oxidative Stress ,030104 developmental biology ,RAW 264.7 Cells ,Biochemistry ,biology.protein ,Signal transduction ,medicine.symptom ,Reactive Oxygen Species ,Oxidation-Reduction ,Signal Transduction - Abstract
Activation of proinflammatory macrophages plays an important role in the pathogenesis of insulin resistance, type 2 diabetes, and atherosclerosis. Previous work using high fat-fed mice has shown that ablation of the adipocyte fatty acid binding protein (FABP4/aP2) in macrophages leads to an antiinflammatory state both in situ and in vivo, and the mechanism is linked, in part, to increased intracellular monounsaturated fatty acids and the up-regulation of uncoupling protein 2. Here, we show that loss of FABP4/aP2 in macrophages additionally induces sirtuin 3 (SIRT3) expression and that monounsaturated fatty acids (C16:1, C18:1) lead to increased SIRT3 protein expression. Increased expression of SirT3 in FABP4/aP2 null macrophages occurs at the protein level with no change in SirT3 mRNA. When compared with controls, silencing of SIRT3 in Raw246.7 macrophages leads to increased expression of inflammatory cytokines, inducible nitric oxide synthase and cyclooxygenase 2. In contrast, loss of SIRT3 in FABP4/aP2-deficient macrophages attenuates the suppressed inflammatory signaling, reduced reactive oxygen species production, lipopolysaccharide-induced mitochondrial dysfunction, and increased fatty acid oxidation. These results suggest that the antiinflammatory phenotype of FABP4/aP2 null mice is mediated by increased intracellular monounsaturated fatty acids leading to the increased expression of both uncoupling protein 2 and SirT3.
- Published
- 2016
39. Interaction of the Adipocyte Fatty Acid-binding Protein with the Hormone-sensitive Lipase
- Author
-
Brian R. Thompson, Anne J. Smith, David A. Bernlohr, and Mark A. Sanders
- Subjects
Alanine ,food and beverages ,Hormone-sensitive lipase ,Cell Biology ,Biology ,Biochemistry ,Fatty acid-binding protein ,Lipid droplet ,Fatty acid binding ,Phosphorylation ,Binding site ,Isoleucine ,Molecular Biology - Abstract
Adipocyte fatty acid-binding protein (AFABP/aP2) forms a physical complex with the hormone-sensitive lipase (HSL) and AFABP/aP2-null mice exhibit reduced basal and hormone-stimulated lipolysis. To identify the determinants affecting the interaction fluorescence resonance energy transfer (FRET) imaging was used in conjunction with a mutagenesis strategy to evaluate the roles AFABP/aP2 fatty acid binding and HSL phosphorylation have in complex formation as well as determine the HSL binding site on AFABP/aP2. The nonfatty acid binding mutant of AFABP/aP2 (R126Q) failed to form a FRET-competent complex with HSL either under basal or forskolin-stimulated conditions, indicating that lipid binding is required for association. Once bound to HSL and on the surface of the lipid droplet, YFP-AFABP/aP2 (but not YFP-HSL) exhibited energy transfer between the fusion protein and BODIPY-C12-labeled triacylglycerol. Serine to alanine mutations at the two PKA phosphorylation sites of HSL (659 and 660), or at the AMPK phosphorylation sites (565), blocked FRET between HSL and AFABP/aP2. Substitution of isoleucine for lysine at position 21 of AFABP/aP2 (K21I), but not 31 (K31I), resulted in a non-HSL-binding protein indicating that residues on helix αI of AFABP/aP2 define a component of the HSL binding site. These results indicate that the ligand-bound form of AFABP/aP2.interacts with the activated, phosphorylated HSL and that the association is likely to be regulatory; either delivering FA to inhibit HSL (facilitating feedback inhibition) or affecting multicomponent complex formation on the droplet surface.
- Published
- 2007
- Full Text
- View/download PDF
40. Gene Expression Profiling of the Human Maternal-Fetal Interface Reveals Dramatic Changes between Midgestation and Term
- Author
-
Lenore Pereira, Andrej Sali, Y. Jean Yang, Matthew Gormley, Agnes C. Paquet, Susan J. Fisher, Virginia D. Winn, Mallur S. Madhusudhan, Kui Tzu V. Feng, David A. Bernlohr, Susan McDonagh, and Ronit Haimov-Kochman
- Subjects
medicine.medical_specialty ,Term Birth ,Placenta ,Basal plate (neural tube) ,Gestational Age ,Biology ,Models, Biological ,Endocrinology ,Pregnancy ,Internal medicine ,Gene expression ,medicine ,Humans ,Gene Regulatory Networks ,Maternal-Fetal Exchange ,Regulation of gene expression ,Genetics ,Fetus ,Gene Expression Profiling ,Gene Expression Regulation, Developmental ,Placentation ,Cell biology ,Transplantation ,Gene expression profiling ,medicine.anatomical_structure ,Female - Abstract
Human placentation entails the remarkable integration of fetal and maternal cells into a single functional unit. In the basal plate region (the maternal-fetal interface) of the placenta, fetal cytotrophoblasts from the placenta invade the uterus and remodel the resident vasculature and avoid maternal immune rejection. Knowing the molecular bases for these unique cell-cell interactions is important for understanding how this specialized region functions during normal pregnancy with implications for tumor biology and transplantation immunology. Therefore, we undertook a global analysis of the gene expression profiles at the maternal-fetal interface. Basal plate biopsy specimens were obtained from 36 placentas (14–40 wk) at the conclusion of normal pregnancies. RNA was isolated, processed, and hybridized to HG-U133A&B Affymetrix GeneChips. Surprisingly, there was little change in gene expression during the 14- to 24-wk interval. In contrast, 418 genes were differentially expressed at term (37–40 wk) as compared with midgestation (14–24 wk). Subsequent analyses using quantitative PCR and immunolocalization approaches validated a portion of these results. Many of the differentially expressed genes are known in other contexts to be involved in differentiation, motility, transcription, immunity, angiogenesis, extracellular matrix dissolution, or lipid metabolism. One sixth were nonannotated or encoded hypothetical proteins. Modeling based on structural homology revealed potential functions for 31 of these proteins. These data provide a reference set for understanding the molecular components of the dialogue taking place between maternal and fetal cells in the basal plate as well as for future comparisons of alterations in this region that occur in obstetric complications.
- Published
- 2007
- Full Text
- View/download PDF
41. Perilipin Targets a Novel Pool of Lipid Droplets for Lipolytic Attack by Hormone-sensitive Lipase
- Author
-
Robert B. Silver, Hsiao Ping H Moore, Emilio P. Mottillo, James G. Granneman, and David A. Bernlohr
- Subjects
Perilipin-1 ,Lipolysis ,Adipose tissue ,Hormone-sensitive lipase ,White adipose tissue ,Biology ,Transfection ,Biochemistry ,Cell Line ,Mice ,Structure-Activity Relationship ,3T3-L1 Cells ,Lipid droplet ,Adipocytes ,Image Processing, Computer-Assisted ,Animals ,Humans ,Phosphorylation ,Molecular Biology ,Triglycerides ,Microscopy, Confocal ,Lipid metabolism ,Cell Biology ,Sterol Esterase ,Lipid Metabolism ,Phosphoproteins ,Lipids ,Protein Structure, Tertiary ,Drug Combinations ,Adipose Tissue ,Microscopy, Fluorescence ,Perilipin ,Proteoglycans ,Collagen ,Laminin ,Carrier Proteins ,Protein Binding - Abstract
Adipocytes serve as the principal energy reservoir of the body; however, the subcellular organization of the machinery regulating lipid trafficking and metabolism is poorly understood. Mobilization of stored triglyceride is thought be controlled by interactions among intracellular lipases and proteins that coat lipid storage droplets. A major limitation of previous studies of hormone-mediated lipolysis, however, is the use of cultured model adipocytes whose three-dimensional architectures do not resemble those in real adipose tissue. To address this limitation, we investigated the intracellular targeting of perilipin, a major lipid coat protein, and hormone-sensitive lipase in three preparations that exhibit more appropriate morphologies: 3T3-L1 adipocytes grown in three-dimensional matrix, dissociated mature adipocytes from mouse adipose tissue, and adipocytes within intact fat pads. High resolution imaging of native and fluorescently tagged proteins indicate that: 1) perilipin preferentially targets a special class of peripheral lipid storage droplets, but not the major or central lipid storage droplets, 2) the peripheral droplets are the sites of attack by hormone-sensitive lipase, and 3) perilipin and hormone-sensitive lipase are continuously colocalized following lipolytic activation. These results indicate that in white adipose tissue, lipolysis takes place in a specialized subcellular domain that is distinct from the major lipid storage site and is defined by perilipin.
- Published
- 2005
- Full Text
- View/download PDF
42. Enzymatic Properties of Purified Murine Fatty Acid Transport Protein 4 and Analysis of Acyl-CoA Synthetase Activities in Tissues from FATP4 Null Mice
- Author
-
Thomas Herrmann, Angela M. Hall, Brian M. Wiczer, Wolfgang Stremmel, and David A. Bernlohr
- Subjects
Very long chain fatty acid ,Lignoceric acid ,Biochemistry ,Catalysis ,Substrate Specificity ,Mice ,chemistry.chemical_compound ,Coenzyme A Ligases ,Animals ,adipocyte protein 2 ,Molecular Biology ,Beta oxidation ,chemistry.chemical_classification ,biology ,Fatty Acid Transport Proteins ,Fatty Acids ,Membrane Transport Proteins ,Fatty acid ,Cell Biology ,Molecular biology ,Triacsin C ,Kinetics ,chemistry ,COS Cells ,Free fatty acid receptor ,biology.protein ,lipids (amino acids, peptides, and proteins) - Abstract
Fatty acid transport protein 4 (FATP4) is an integral membrane protein expressed in the plasma and internal membranes of the small intestine and adipocyte as well as in the brain, kidney, liver, skin, and heart. FATP4 has been hypothesized to be bifunctional, exhibiting both fatty acid transport and acyl-CoA synthetase activities that work in concert to mediate fatty acid influx across biological membranes. To determine whether FATP4 is an acyl-CoA synthetase, the murine protein was engineered to contain a C-terminal FLAG epitope tag, expressed in COS1 cells via adenovirus-mediated infection and purified to near homogeneity using alpha-FLAG affinity chromatography. Kinetic analysis of the enzyme was carried out for long chain (palmitic acid, C16:0) and very long chain (lignoceric acid, C24:0) fatty acids as well as for ATP and CoA. FATP4 exhibited substrate specificity for C16:0 and C24:0 fatty acids with a V(max)/K(m) (C16:0)/V(max)/K(m) (C24:0) of 1.5. Like purified FATP1, FATP4 was insensitive to inhibition by triacsin C but was sensitive to feedback inhibition by acyl-CoA. Although purified FATP4 exhibited high levels of palmitoyl-CoA and lignoceroyl-CoA synthetase activity, extracts from the skin and intestine of FATP4 null mice exhibited reduced esterification for C24:0, but not C16:0 or C18:1, suggesting that in vivo, defects in very long chain fatty acid uptake may underlie the skin disorder phenotype of null mice.
- Published
- 2005
- Full Text
- View/download PDF
43. Physical Association between the Adipocyte Fatty Acid-binding Protein and Hormone-sensitive Lipase
- Author
-
David A. Bernlohr, Brian R. Thompson, Fredric B. Kraemer, Mark A. Sanders, Anne J. Smith, and Constantine Londos
- Subjects
chemistry.chemical_classification ,Yellow fluorescent protein ,food and beverages ,Fatty acid ,Context (language use) ,Hormone-sensitive lipase ,Cell Biology ,Transfection ,Biology ,Biochemistry ,chemistry.chemical_compound ,Förster resonance energy transfer ,chemistry ,Adipocyte ,Lipid droplet ,biology.protein ,lipids (amino acids, peptides, and proteins) ,Molecular Biology - Abstract
Previous in vitro studies have established that hormone sensitive lipase (HSL) and adipocyte fatty acid-binding protein (AFABP) form a physical complex that presumably positions the FABP to accept a product fatty acid generated during catalysis. To assess AFABP-HSL interaction within a cellular context, we have used lipocytes derived from 293 cells (C8PA cells) and examined physical association using fluorescence resonance energy transfer. Transfection of C8PA cells with cyan fluorescent protein (CFP)-HSL, yellow fluorescent protein (YFP)-adipocyte FABP, or YFP-liver FABP revealed that under basal conditions each protein was cytoplasmic. In the presence of 20 μm forskolin, CFP-HSL translocated to the triacylglycerol droplet, coincident with BODIPY-FA labeled depots. Fluorescence resonance energy transfer analysis demonstrated that CFP-HSL associated with YFP-adipocyte FABP in both basal and forskolin-treated cells. In contrast, little if any fluorescence resonance energy transfer could be detected between CFP-HSL and YFP-liver FABP. These results suggest that a pre-lipolysis complex containing at least AFABP and HSL exists and that the complex translocates to the surface of the lipid droplet.
- Published
- 2004
- Full Text
- View/download PDF
44. Fatty acid binding proteins stabilize leukotriene A4
- Author
-
Robert C. Murphy, Douglas F. Dyckes, Jennifer S. Dickinson Zimmer, and David A. Bernlohr
- Subjects
biology ,half-life ,Chemistry ,Leukotriene A4 ,Biological activity ,Cell Biology ,QD415-436 ,Biochemistry ,Fatty acid-binding protein ,stomatognathic diseases ,Lipoxygenase ,chemistry.chemical_compound ,Endocrinology ,Biosynthesis ,transcellular biosynthesis ,biology.protein ,Arachidonic acid ,lipids (amino acids, peptides, and proteins) ,leukotriene biosynthesis ,Binding site ,Transcellular - Abstract
Leukotriene A(4) (LTA(4)) is a chemically reactive conjugated triene epoxide product derived from 5-lipoxygenase oxygenation of arachidonic acid. At physiological pH, this reactive compound has a half-life of less than 3 s at 37 degrees C and approximately 40 s at 4 degrees C. Regardless of this aqueous instability, LTA(4) is an intermediate in the formation of biologically active leukotrienes, which can be formed through either intracellular or transcellular biosynthesis. Previously, epithelial fatty acid binding protein (E-FABP) present in RBL-1 cells was shown to increase the half-life of LTA(4) to approximately 20 min at 4 degrees C. Five FABPs (adipocyte FABP, intestinal FABP, E-FABP, heart/muscle FABP, and liver FABP) have now been examined and also found to increase the half-life of LTA(4) at 4 degrees C to approximately 20 min with protein present. Stabilization of LTA(4) was examined when arachidonic acid was present to compete with LTA(4) for the binding site on E-FABP. Arachidonate has an apparent higher affinity for E-FABP than LTA(4) and was able to completely block stabilization of the latter. When E-FABP is not saturated with arachidonate, FABP can still stabilize LTA(4). Several lipoxygenase products, including 5-hydroxyeicosatetraenoic acid, 5,6-dihydroxyeicosatetraenoic acid, and leukotriene B(4), were found to have no effect on the stability of LTA(4) induced by E-FABP even when present at concentrations 3-fold higher than LTA(4).
- Published
- 2004
45. Stabilization of Leukotriene A4 by Epithelial Fatty Acid-binding Protein in the Rat Basophilic Leukemia Cell
- Author
-
Dennis R. Voelker, Robert C. Murphy, Jennifer S. Dickinson Zimmer, and David A. Bernlohr
- Subjects
Immunoprecipitation ,Blotting, Western ,Nerve Tissue Proteins ,Peptide ,Fatty Acid-Binding Proteins ,Peptide Mapping ,Biochemistry ,Mass Spectrometry ,Fatty acid-binding protein ,chemistry.chemical_compound ,Cytosol ,Sequence Analysis, Protein ,Protein purification ,Tumor Cells, Cultured ,medicine ,Animals ,Trypsin ,Eye Proteins ,Molecular Biology ,Chromatography, High Pressure Liquid ,Immunosorbent Techniques ,chemistry.chemical_classification ,Leukotriene A4 ,Cell Biology ,Peptide Fragments ,Rats ,Leukemia, Basophilic, Acute ,chemistry ,Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization ,Electrophoresis, Polyacrylamide Gel ,lipids (amino acids, peptides, and proteins) ,Arachidonic acid ,Carrier Proteins ,Half-Life ,medicine.drug - Abstract
Leukotriene A(4) (LTA(4)) is a chemically unstable triene epoxide product of 5-lipoxygenase metabolism of arachidonic acid. Despite this chemical reactivity and its synthesis at the perinuclear membrane, LTA(4) is enzymatically converted into the cysteinyl leukotrienes and leukotriene B(4). Furthermore, LTA(4) participates in transcellular biosynthesis and is thus transferred between cells as an intact molecule. A cytosolic fatty acid-binding protein present in the rat basophilic leukemia cells was identified using mass spectrometry. This protein was determined to be the stabilizing factor present in the cell cytosol responsible for increasing the effective chemical half-life of LTA(4). Rat epithelial fatty acid-binding protein (E-FABP) was isolated using partial protein purification and immunoprecipitation. In-gel digestion with trypsin followed by peptide fingerprint analysis using matrix-assisted laser desorption ionization mass spectrometry and sequencing the major tryptic peptide obtained from liquid chromatography/mass spectrometry/mass spectrometry analysis identified E-FABP in the active fraction. Semi-quantitative Western blot analysis indicated that E-FABP in the cytosolic fraction of RBL-1 cells was present at approximately 1-3 pmol/10(6) cells. E-FABP (9 microm) was tested for its ability to stabilize LTA(4), and at 37 degrees C E-FABP was able to increase the half-life of LTA(4) from the previously reported half-life less than 3 s to a half-life of approximately 7 min. These results present a novel function for the well studied fatty acid-binding protein as a participant in leukotriene biosynthesis that permits LTA(4) to be available for further enzymatic processing in various cellular regions.
- Published
- 2004
- Full Text
- View/download PDF
46. Fatty Acid-binding Protein-Hormone-sensitive Lipase Interaction
- Author
-
Fredric B. Kraemer, David A. Bernlohr, Wen-Jun Shen, Assumpta Bennaars-Eiden, James R. Ross, and Anne E. Jenkins-Kruchten
- Subjects
chemistry.chemical_classification ,food and beverages ,Fatty acid ,Hormone-sensitive lipase ,Cell Biology ,Butyrate ,Biochemistry ,Fatty acid-binding protein ,chemistry.chemical_compound ,Enzyme ,chemistry ,Adipocyte ,Fatty acid binding ,Lipolysis ,lipids (amino acids, peptides, and proteins) ,Molecular Biology - Abstract
Adipose lipolysis is mediated, in part, via interaction of fatty acid-binding protein (FABP) with hormone-sensitive lipase (HSL). Mice with reduced FABP content in fat (adipocyte FABP null) exhibit diminished fat cell lipolysis, whereas transgenic mice with increased FABP content in fat (epithelial FABP transgenic) exhibit enhanced lipolysis. To examine the relationship between the binding of FABP to HSL and activation of catalytic activity, isothermal titration microcalorimetry as well as kinetic analysis using a variety of FABP isoforms have been employed. In the absence of fatty acids, no FABP-HSL association could be demonstrated for any FABP form. However, in the presence of 10 μm oleate, A-FABP and E-FABP each bound to HSL with high affinity (Kd of 0.5 and 3 nm, respectively) in a ∼1:1 molar stoichiometry, whereas liver FABP and intestinal FABP did not exhibit any association. To compare binding to catalysis, each FABP isoform was incubated with HSL in vitro, and enzymatic activity was assessed. Importantly, each FABP form stimulated HSL activity ∼2-fold using cholesteryl oleate as substrate but exhibited no activation using p-nitrophenyl butyrate. The activation by A-FABP was dependent upon its fatty acid binding properties because a non-fatty acid binding mutant, R126Q, failed to activate HSL. These results suggest that binding and activation of HSL by FABPs are separate and distinct functions and that HSL contains a site for fatty acid binding that allows for FABP association.
- Published
- 2003
- Full Text
- View/download PDF
47. Covalent Modification of Epithelial Fatty Acid-binding Protein by 4-Hydroxynonenal in Vitro and in Vivo
- Author
-
David A. Bernlohr, Rebecca J. Kapphahn, LeeAnn Higgins, Ann V. Hertzel, Deborah A. Ferrington, and Assumpta Bennaars-Eiden
- Subjects
Gel electrophoresis ,Chemistry ,Electrospray ionization ,Cell Biology ,Biochemistry ,Fatty acid-binding protein ,In vitro ,4-Hydroxynonenal ,Lipid peroxidation ,chemistry.chemical_compound ,In vivo ,lipids (amino acids, peptides, and proteins) ,Guanidine ,Molecular Biology - Abstract
4-Hydroxynonenal (4-HNE) is a cytotoxic α,β-unsaturated acyl aldehyde that is naturally produced from lipid peroxidation and cleavage in response to oxidative stress and aging. Such reactive lipids covalently modify cellular target proteins, thereby affecting biological structure and function. Herein we report the identification of the epithelial fatty acid-binding protein (E-FABP) as a molecular target for 4-HNE modification both in vitro and in vivo. 4-HNE covalently modified (t < 60 s) E-FABPin vitro, as revealed by a combination of matrix-assisted laser desorption ionization-time of flight mass spectrometry and immunochemical reactivity using antibodies directed to 4-HNE-protein conjugates. Identification of Cys-120 as the major site of modification was determined through tandem mass spectral sequencing of tryptic peptides, as well as analysis of E-FABP mutants C120A, C127A, and C120A/C127A. The in vitro modification of Cys-120 by 4-HNE was relatively insensitive to pH (6.4–8.4), and temperature (4–37 °C) but was markedly potentiated by noncovalently bound fatty acids. 4-HNE-modified E-FABP was more stable than unmodified E-FABP to chemical denaturation by guanidine hydrochloride, as assessed by changes in intrinsic tryptophan fluorescence. Analysis of soluble protein extracts from rat retina with antibodies directed to 4-HNE-protein conjugates revealed immunoreactivity with a 15-kDa protein that was identified by electrospray ionization and matrix-assisted laser desorption ionization-time of flight mass spectrometry as E-FABP. Evaluation of retinal pigment epithelial cell extracts derived from E-FABP null mice by two-dimensional gel electrophoresis using anti-4-HNE antibodies revealed increased modification in the null cells relative to those from wild type cells. These results indicate that E-FABP is a molecular target for 4-HNE modification and the hypothesis that E-FABP functions as an antioxidant protein by scavenging reactive lipids through covalent modification of Cys-120.
- Published
- 2002
- Full Text
- View/download PDF
48. Adenovirus-mediated gene transfer in primary murine adipocytes
- Author
-
David A. Bernlohr, Ann V. Hertzel, and Mark A. Sanders
- Subjects
green fluorescent protein ,Regulation of gene expression ,Reporter gene ,adipocytes ,Promoter ,adenovirus ,QD415-436 ,Cell Biology ,Biology ,medicine.disease_cause ,Biochemistry ,Molecular biology ,Green fluorescent protein ,Adenoviridae ,chemistry.chemical_compound ,Endocrinology ,chemistry ,Adipocyte ,medicine ,Luciferase ,reporter genes ,Gene - Abstract
The transfer of genes into primary murine adipocytes using an adenovirus system has been developed. A recombinant adenovirus was constructed (expressing green fluorescent protein [GFP] under the control of the strong cytomegalovirus [CMV] promoter and a luciferase reporter gene under the control of the weak adipocyte promoter keratinocyte lipid-binding protein [KLBP/FABP5]) and incubated with primary adipocytes from C57BL/6J mice. Analysis of infected cells by confocal microscopy detected GFP expression in both the cytoplasm and nucleus of adipocytes with a 64% efficiency of infection. To demonstrate the applicability of this method in the study of gene regulation, adenovirus-infected adipocytes exhibited significant levels of luciferase activity even from a weak promoter. TPA treatment of infected adipocytes increased luciferase activity, consistent with previous studies indicating that the KLBP/FABP5 gene is up-regulated by phorbol esters. These results provide an efficient, convenient, and sensitive method to transiently infect primary murine adipocytes, facilitating protein expression or permitting analysis of reporter gene activity from both viral and endogenous promoters.
- Published
- 2000
- Full Text
- View/download PDF
49. Regulation of fatty acid transporters in mammalian cells
- Author
-
Brigitte I. Frohnert and David A. Bernlohr
- Subjects
Mammals ,chemistry.chemical_classification ,Chemistry ,Fatty Acids ,Fatty acid ,Biological Transport ,Transporter ,Fasting ,Cell Biology ,Biochemistry ,Absorption ,Diffusion ,Adipocytes ,Animals ,Humans ,Intestinal Mucosa ,Carrier Proteins ,Triglycerides - Published
- 2000
- Full Text
- View/download PDF
50. Targeted disruption of the adipocyte lipid-binding protein (aP2 protein) gene impairs fat cell lipolysis and increases cellular fatty acid levels
- Author
-
Natalie Ribarik Coe, Melanie A. Simpson, and David A. Bernlohr
- Subjects
medicine.medical_specialty ,Adipose tissue ,QD415-436 ,adipocyte ,Biochemistry ,Fatty acid-binding protein ,chemistry.chemical_compound ,Endocrinology ,Internal medicine ,Adipocyte ,medicine ,Lipolysis ,adipocyte protein 2 ,chemistry.chemical_classification ,biology ,food and beverages ,Fatty acid ,Cell Biology ,medicine.anatomical_structure ,chemistry ,lipolysis ,biology.protein ,fatty acid ,Keratinocyte ,lipid binding proteins ,Intracellular - Abstract
The availability of mice containing an adipocyte lipid-binding protein (ALBP/aP2) gene disruption allowed for a direct examination of the presumed role of lipid-binding proteins in the mobilization and trafficking of intracellular fatty acids. Total body and epididymal fat pad weights, as well as adipose cell morphology, were unaltered in male ALBP/aP2 disrupted mice when compared to their wild-type littermates. Analysis of adipocytes isolated from wild-type and ALBP/aP2 null mice revealed that a selective 40- and 13-fold increase in the level of the keratinocyte lipid-binding protein (KLBP) mRNA and protein, respectively, accompanied the ALBP/aP2 gene disruption. Although KLBP protein was significantly up-regulated, the total lipid-binding protein level decreased 8-fold as a consequence of the disruption. There was no appreciable difference in the rate of fatty acid influx or esterification in adipocytes of wild-type and ALBP/aP2 null animals. To the contrary, basal lipolysis decreased approximately 40% in ALBP/aP2 nulls as compared to wild-type littermates. The glycerol release from isproterenol-stimulated ALBP/aP2 null fat cells was similarly reduced by ∼35%. Consistent with a decrease in basal efflux, the non-esterified fatty acid (NEFA) level was nearly 3-fold greater in adipocytes from ALBP/aP2 nulls as compared to wild-type animals. The significant decrease in both basal and isoproterenol-stimulated lipolysis in adipose tissue of ALBP/aP2 null mice supports the model whereby intracellular lipid-binding proteins function as lipid chaperones, facilitating the movement of fatty acids out of the fat cell.—Ribarik Coe, N., M. A. Simpson, and D. A. Bernlohr. 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.
- Published
- 1999
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.