Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Med Vet Entomol ; 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38478926

ABSTRACT

Aedes albopictus is highly prevalent in the northern part of West Bengal and is considered to be responsible for the recent dengue outbreaks in this region. Control of this vector is largely relied on the use of synthetic pyrethroids, which can lead to the development of resistance. In the present study, larvae of three wild Ae. albopictus populations from the dengue-endemic regions were screened for deltamethrin resistance, and the role of cytochrome P450 monooxygenases (CYPs) was investigated in deltamethrin exposed and unexposed larvae. Two populations were incipient resistant, and one population was completely resistant against deltamethrin. Monooxygenase titration assay revealed the involvement of CYPs in deltamethrin resistance along with an induction effect of deltamethrin exposure. Gene expression studies revealed differential expression of five CYP6 family genes, CYP6A8, CYP6P12, CYP6A14, CYP6N3 and CYP6N6, with high constitutive expression of CYP6A8 and CYP6P12 in all the populations before and after deltamethrin exposure. From these findings, it was evident that CYPs play an important role in the development of deltamethrin resistance in the Ae. albopictus populations in this region.

2.
Front Cell Dev Biol ; 10: 925971, 2022.
Article in English | MEDLINE | ID: mdl-35800892

ABSTRACT

Adipose tissue macrophages are a major immune cell type contributing to homeostatic maintenance and pathological adipose tissue remodeling. However, the mechanisms underlying macrophage recruitment and polarization in adipose tissue during obesity remain poorly understood. Previous studies have suggested that the gap junctional protein, connexin 43 (Cx43), plays a critical role in macrophage activation and phagocytosis. Herein, we investigated the macrophage-specific roles of Cx43 in high fat diet (HFD)-induced pathological remodeling of adipose tissue. Expression levels of Cx43 were upregulated in macrophages co-cultured with dying adipocytes in vitro, as well as in macrophages associated with dying adipocytes in the adipose tissue of HFD-fed mice. Cx43 knockdown reduced lipopolysaccharide (LPS)-induced ATP release from macrophages and decreased inflammatory responses of macrophages co-cultured with dying adipocytes. Based on global gene expression profiling, macrophage-specific Cx43-knockout (Cx43-MKO) mice were resistant to HFD-induced inflammatory responses in adipose tissue, potentially via P2X7-mediated signaling pathways. Cx43-MKO mice exhibited reduced HFD-induced macrophage recruitment in adipose tissue. Moreover, Cx43-MKO mice showed reduced inflammasome activation in adipose tissues and improved glucose tolerance. Collectively, these findings demonstrate that Cx43 expression in macrophages facilitates inflammasome activation, which, in turn, contributes to HFD-induced metabolic dysfunction.

3.
Nat Commun ; 13(1): 4084, 2022 07 14.
Article in English | MEDLINE | ID: mdl-35835749

ABSTRACT

Dysregulation of adipose tissue plasmalogen metabolism is associated with obesity-related metabolic diseases. We report that feeding mice a high-fat diet reduces adipose tissue lysoplasmalogen levels and increases transmembrane protein 86 A (TMEM86A), a putative lysoplasmalogenase. Untargeted lipidomic analysis demonstrates that adipocyte-specific TMEM86A-knockout (AKO) increases lysoplasmalogen content in adipose tissue, including plasmenyl lysophosphatidylethanolamine 18:0 (LPE P-18:0). Surprisingly, TMEM86A AKO increases protein kinase A signalling pathways owing to inhibition of phosphodiesterase 3B and elevation of cyclic adenosine monophosphate. TMEM86A AKO upregulates mitochondrial oxidative metabolism, elevates energy expenditure, and protects mice from metabolic dysfunction induced by high-fat feeding. Importantly, the effects of TMEM86A AKO are largely reproduced in vitro and in vivo by LPE P-18:0 supplementation. LPE P-18:0 levels are significantly lower in adipose tissue of human patients with obesity, suggesting that TMEM86A inhibition or lysoplasmalogen supplementation might be therapeutic approaches for preventing or treating obesity-related metabolic diseases.


Subject(s)
Plasmalogens , Thermogenesis , Adipocytes/metabolism , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Diet, High-Fat/adverse effects , Energy Metabolism/physiology , Homeostasis , Humans , Hydrolases , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/metabolism , Plasmalogens/metabolism , Thermogenesis/physiology
4.
Metabolism ; 130: 155159, 2022 05.
Article in English | MEDLINE | ID: mdl-35150731

ABSTRACT

INTRODUCTION: The mobilization and catabolism of lipid energy is a central function of adipocytes that is under the control of the ß-adrenergic signaling pathway, and defects in ß-adrenergic signaling in adipocytes have been linked to obesity and obesity-related metabolic diseases. Receptor expression-enhancing proteins (REEPs) are endoplasmic reticulum (ER) proteins that play critical roles in subcellular targeting of receptor signaling complexes. Examination of gene expression profiles indicates that, among REEPs expressed in adipocytes, REEP6 expression is uniquely upregulated by sympathetic nervous system activation, suggesting involvement in regulating adrenergic signal transduction. OBJECTIVE: The aim of this study was to assess the contribution of REEP6 to the thermogenic activation of adipocytes and characterize the metabolic consequences of REEP6 deficiency in vivo. METHODS: Expression levels of Reep6 in adipose tissue were examined by using public transcriptomic data and validated by Western blot and qPCR analyses. Adipocyte-specific regulatory roles of REEP6 were investigated in vitro in C3H10T1/2 adipocytes and in primary adipocytes obtained from REEP6 KO mice. Effects of in vivo REEP6 deficiency on energy expenditure were measured by indirect calorimetry. Mitochondrial content in adipose tissue was accessed by immunoblot, mitochondrial DNA analysis, and confocal and electron microscopy. Effects of REEP6 KO on obesity-induced metabolic dysfunction were tested in a high-fat diet-induced obesity mouse model by glucose tolerance test, Western blot, and histological analyses. RESULTS: REEP6 expression is highly enriched in murine adipocytes and is sharply upregulated upon adipocyte differentiation and by cold exposure. Inactivation of REEP6 in mice increased adiposity, and reduced energy expenditure and cold tolerance. REEP6 KO severely reduced protein kinase A-mediated signaling in BAT and greatly reduced mitochondrial mass. The effect of REEP6 inactivation on diminished ß-adrenergic signaling was reproduced in cultured adipocytes, indicating that this effect is cell-autonomous. REEP6 KO also suppressed expression of adenylate cyclase 3 (Adcy3) in brown adipose tissue and knockdown of REEP6 in adipocytes reduced targeting of ADCY3 to the plasma membrane. Lastly, REEP6 KO exacerbated high-fat diet-induced insulin resistance and inflammation in adipose tissue. CONCLUSIONS: This study indicates that REEP6 plays an important role in ß-adrenergic signal transduction in adipocytes involving the expression and trafficking of Adcy3. Genetic inactivation of REEP6 reduces energy expenditure, increases adiposity, and the susceptibility to obesity-related metabolic dysfunction.


Subject(s)
Adipocytes , Adrenergic Agents , Adipocytes/metabolism , Adipose Tissue, Brown/metabolism , Adrenergic Agents/metabolism , Animals , Diet, High-Fat , Energy Metabolism/genetics , Eye Proteins/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/metabolism , Signal Transduction , Thermogenesis/genetics
5.
J Contemp Dent Pract ; 23(12): 1199-1202, 2022 Dec 01.
Article in English | MEDLINE | ID: mdl-37125516

ABSTRACT

AIM: To evaluate the effectiveness of gelatin sponge [Abgel] with injectable platelet-rich fibrin (i-PRF) in the surgical treatment of mandibular Grade II furcation defects in endodontically involved teeth. MATERIALS AND METHODS: The present study was a single-center clinical trial wherein 20 mandibular grade II furcation defects were treated with gelatin sponge combined with i-PRF results were compared both clinically and radiographically at baseline, 3, and 6 months postoperatively. Statistical analysis was done using Statistical package for social sciences (SPSS) we software. For pre and post comparison, paired t-test, analysis of variance (ANOVA) and Wilcoxon test were used. RESULTS: There was a statistically highly significant improvement seen in all the clinical parameters vertical clinical attachment level (V-CAL), horizontal clinical attachment level (H-CAL) and probing pocket depth (PPD) and radiographic parameters at baseline and 6 months postoperatively p < 0.01. CONCLUSION: Open flap debridement along with Abgel combined with i-PRF is an effective treatment modality in reducing the horizontal and vertical component of grade II furcation defects. CLINICAL SIGNIFICANCE: Gelatin sponge with i-PRF is a cost-effective treatment modality in achieving periodontal regeneration.


Subject(s)
Furcation Defects , Platelet-Rich Fibrin , Humans , Gelatin/therapeutic use , Furcation Defects/diagnostic imaging , Furcation Defects/surgery , Treatment Outcome , Molar/surgery , Guided Tissue Regeneration, Periodontal/methods , Periodontal Attachment Loss
6.
Biomol Ther (Seoul) ; 29(5): 455-464, 2021 Sep 01.
Article in English | MEDLINE | ID: mdl-34162770

ABSTRACT

Uncontrolled inflammation is considered the pathophysiological basis of many prevalent metabolic disorders, such as nonalcoholic fatty liver disease, diabetes, obesity, and neurodegenerative diseases. The inflammatory response is a self-limiting process that produces a superfamily of chemical mediators, called specialized proresolving mediators (SPMs). SPMs include the ω-3-derived family of molecules, such as resolvins, protectins, and maresins, as well as arachidonic acid-derived (ω-6) lipoxins that stimulate and promote resolution of inflammation, clearance of microbes, and alleviation of pain and promote tissue regeneration via novel mechanisms. SPMs function by binding and activating G protein-coupled receptors, such as FPR2/ALX, GPR32, and ERV1, and nuclear orphan receptors, such as RORα. Recently, several studies reported that SPMs have the potential to attenuate lipid metabolism disorders. However, the understanding of pharmacological aspects of SPMs, including tissue-specific biosynthesis, and specific SPM receptors and signaling pathways, is currently limited. Here, we summarize recent advances in the role of SPMs in resolution of inflammatory diseases with metabolic disorders, such as nonalcoholic fatty liver disease and obesity, obtained from preclinical animal studies. In addition, the known SPM receptors and their intracellular signaling are reviewed as targets of resolution of inflammation, and the currently available information on the therapeutic effects of major SPMs for metabolic disorders is summarized.

7.
Diabetes ; 70(9): 2000-2013, 2021 09.
Article in English | MEDLINE | ID: mdl-34187836

ABSTRACT

Transmembrane 4 L six family member 5 (TM4SF5) functions as a sensor for lysosomal arginine levels and activates the mammalian target of rapamycin complex 1 (mTORC1). While the mTORC1 signaling pathway plays a key role in adipose tissue metabolism, the regulatory function of TM4SF5 in adipocytes remains unclear. In this study we aimed to establish a TM4SF5 knockout (KO) mouse model and investigated the effects of TM4SF5 KO on mTORC1 signaling-mediated autophagy and mitochondrial metabolism in adipose tissue. TM4SF5 expression was higher in inguinal white adipose tissue (iWAT) than in brown adipose tissue and significantly upregulated by a high-fat diet (HFD). TM4SF5 KO reduced mTORC1 activation and enhanced autophagy and lipolysis in adipocytes. RNA sequencing analysis of TM4SF5 KO mouse iWAT showed that the expression of genes involved in peroxisome proliferator-activated receptor α signaling pathways and mitochondrial oxidative metabolism was upregulated. Consequently, TM4SF5 KO reduced adiposity and increased energy expenditure and mitochondrial oxidative metabolism. TM4SF5 KO prevented HFD-induced glucose intolerance and inflammation in adipose tissue. Collectively, the results of our study demonstrate that TM4SF5 regulates autophagy and lipid catabolism in adipose tissue and suggest that TM4SF5 could be therapeutically targeted for the treatment of obesity-related metabolic diseases.


Subject(s)
Adipose Tissue/metabolism , Autophagy/genetics , Membrane Proteins/genetics , Obesity/genetics , Animals , Diet, High-Fat , Energy Metabolism/genetics , Female , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Membrane Proteins/metabolism , Mice , Mice, Knockout , Obesity/metabolism , Signal Transduction/genetics
8.
Nat Metab ; 3(3): 428-441, 2021 03.
Article in English | MEDLINE | ID: mdl-33758424

ABSTRACT

Obesity reduces adipocyte mitochondrial function, and expanding adipocyte oxidative capacity is an emerging strategy to improve systemic metabolism. Here, we report that serine/threonine-protein kinase 3 (STK3) and STK4 are key physiological suppressors of mitochondrial capacity in brown, beige and white adipose tissues. Levels of STK3 and STK4, kinases in the Hippo signalling pathway, are greater in white than brown adipose tissues, and levels in brown adipose tissue are suppressed by cold exposure and greatly elevated by surgical denervation. Genetic inactivation of Stk3 and Stk4 increases mitochondrial mass and function, stabilizes uncoupling protein 1 in beige adipose tissue and confers resistance to metabolic dysfunction induced by high-fat diet feeding. Mechanistically, STK3 and STK4 increase adipocyte mitophagy in part by regulating the phosphorylation and dimerization status of the mitophagy receptor BNIP3. STK3 and STK4 expression levels are elevated in human obesity, and pharmacological inhibition improves metabolic profiles in a mouse model of obesity, suggesting STK3 and STK4 as potential targets for treating obesity-related diseases.


Subject(s)
Adipocytes/metabolism , Energy Metabolism , Mitophagy , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Animals , Cell Line , Humans , Intracellular Signaling Peptides and Proteins , Mice , Mice, Knockout , Obesity/prevention & control , Obesity/therapy , Protein Serine-Threonine Kinases/genetics , Serine-Threonine Kinase 3
9.
Int J Obes (Lond) ; 45(1): 122-129, 2021 01.
Article in English | MEDLINE | ID: mdl-32467614

ABSTRACT

BACKGROUND/OBJECTIVES: Polymethoxyselenoflavone (PMSF) is a compound that substitutes the oxygen atom in a flavonoid with selenium. This study aimed to investigate the effects of PMSFs on lipid metabolism in adipocytes and their anti-obesity potential. SUBJECTS/METHODS: To test lipolytic and thermogenic effects of the compounds in vitro, adipocytes differentiated from immortalized pre-brown adipocyte progenitors and pre-white adipocyte cell lines were treated with 19 PMSFs. The expression levels of brown adipocyte markers and genes related to mitochondrial metabolism were analyzed by qPCR and western blot. In vivo anti-obesity effect was investigated using diet-induced obesity mouse models and adipocyte-specific ATGL knockout mice. RESULTS: The qPCR analysis identified 2-(3,4-dimethoxyphenyl)-4H-selenochromen-4-one (DMPSC) as the most potent brown adipogenic candidate among the 19 compounds tested in this study. DMPSC treatment significantly increased the mitochondrial content and oxidative metabolism in adipocytes in vitro. Mechanistically, DMPSC treatment increased lipolysis through activation of PKA downstream signaling. Consistently, the in vivo treatment of DMPSC increased energy consumption, reduced body weight, and improved glucose tolerance in mice fed with high-fat diets. Moreover, DMPSC treatment increased brown adipocyte marker expression and mitochondrial content in adipose tissue of mice. The anti-obesity effects were absent in adipocyte-specific ATGL knockout mice, indicating that the DMPSC effect is mediated by cytosolic lipase-dependent mechanisms. CONCLUSIONS: Collectively, our results indicated that DMPSC exerted anti-obesity effects partially through the PKA signaling-mediated activation of lipolysis and brown adipose tissue metabolism.


Subject(s)
Adipocytes, Brown/drug effects , Anti-Obesity Agents/pharmacology , Flavonoids/pharmacology , Lipolysis/drug effects , Selenium Compounds/pharmacology , 3T3-L1 Cells , Adipocytes, Brown/metabolism , Animals , Disease Models, Animal , Male , Mice , Mice, Knockout , Obesity/metabolism
10.
Aging Dis ; 11(3): 575-587, 2020 May.
Article in English | MEDLINE | ID: mdl-32489703

ABSTRACT

Aging-related adipose tissue dysfunction contributes to the progression of chronic metabolic diseases. We investigated the role of age-dependent expression of a neurotrophin, brain-derived neurotrophic factor (BDNF) in adipose tissue. Pro-BDNF expression was elevated in epididymal white adipose tissue (eWAT) with advanced age, which was associated with the reduction in sympathetic innervation. Interestingly, BDNF expression was enriched in PDGFRα+ adipocyte progenitors isolated from eWAT, with age-dependent increase in expression. In vitro pro-BDNF treatment caused apoptosis in adipocytes differentiated from C3H10T1/2 cells, and siRNA knockdown of sortilin mitigated these effects. Tamoxifen-inducible PDGFRα+ cell-specific deletion of BDNF (BDNFPdgfra KO) reduced pro-BDNF expression in eWAT, prevented age-associated declines in sympathetic innervation and mitochondrial content in eWAT, and improved insulin sensitivity. Moreover, BDNFPdgfra KO mice showed reduced expression of aging-induced inflammation and senescence markers in eWAT. Collectively, these results identified the upregulation of pro-BDNF expression in adipocyte progenitors as a feature of visceral white adipose tissue aging and suggested that inhibition of BDNF expression in adipocyte progenitors is potentially beneficial to prevent aging-related adipose tissue dysfunction.

11.
Mol Metab ; 39: 101005, 2020 09.
Article in English | MEDLINE | ID: mdl-32344065

ABSTRACT

OBJECTIVE: Beclin1 is a core molecule of the macroautophagy machinery. Although dysregulation of macroautophagy is known to be involved in metabolic disorders, the function of Beclin1 in adipocyte metabolism has not been investigated. In the present study, we aimed to study the role of Beclin1 in lipolysis and mitochondrial homeostasis of adipocytes. METHODS: Autophagic flux during lipolysis was examined in adipocytes cultured in vitro and in the adipose tissue of mice. Adipocyte-specific Beclin1 knockout (KO) mice were used to investigate the activities of Beclin1 in adipose tissues. RESULTS: cAMP/PKA signaling increased the autophagic flux in adipocytes differentiated from C3H10T1/2 cells. In vivo autophagic flux was higher in the brown adipose tissue (BAT) than that in the white adipose tissue and was further increased by the ß3 adrenergic receptor agonist CL316243. In addition, surgical denervation of BAT greatly reduced autophagic flux, indicating that sympathetic nerve activity is a major regulator of tissue autophagy. Adipocyte-specific KO of Beclin1 led to a hypertrophic enlargement of lipid droplets in BAT and impaired CL316243-induced lipolysis/lipid mobilization and energy expenditure. While short-term effects of Beclin1 deletion were characterized by an increase in mitochondrial proteins, long-term Beclin1 deletion led to severe disruption of autophagy, resulting in mitochondrial loss, and dramatically reduced the expression of genes involved in lipid metabolism. Consequently, adipose tissue underwent increased activation of cell death signaling pathways, macrophage recruitment, and inflammation, particularly in BAT. CONCLUSIONS: The present study demonstrates the critical roles of Beclin1 in the maintenance of lipid metabolism and mitochondrial homeostasis in adipose tissues.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Beclin-1/genetics , Gene Deletion , Lipolysis/genetics , Mitochondria/genetics , Mitochondria/metabolism , Adipocytes/ultrastructure , Adipose Tissue, Brown/metabolism , Animals , Autophagy/genetics , Beclin-1/metabolism , Cell Line , Cyclic AMP , Cyclic AMP-Dependent Protein Kinases/metabolism , DNA Copy Number Variations , Immunity , Lipid Metabolism , Mice , Mice, Knockout , Mitochondria/ultrastructure , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Receptors, Adrenergic, beta-3/metabolism , Signal Transduction , Thermogenesis/genetics
12.
J Nutr Biochem ; 76: 108300, 2020 02.
Article in English | MEDLINE | ID: mdl-31812908

ABSTRACT

Coumestrol is a dietary phytoestrogen with estrogen-mimicking characteristics. This study investigated the molecular mechanisms of antiobesity effects of coumestrol. Two weeks of coumestrol treatment reduced body weight and improved glucose tolerance of high-fat diet (HFD)-fed mice. Notably, coumestrol treatment reduced adiposity but expanded brown adipose tissue mass. In addition, coumestrol treatment induced up-regulation of brown adipocyte markers and lipolytic gene expression in adipose tissue. Mechanistically, coumestrol induced an increase in mitochondrial contents of brown adipose tissue, which was associated with up-regulation of adenosine monophosphate-activated protein kinase and sirtuin 1. In vitro knockdown of estrogen receptor 1 inhibited the effect of coumestrol on brown adipogenic marker expression, increase in mitochondrial contents and oxygen consumption rate in brown adipocytes. Furthermore, lineage tracing of platelet-derived growth factor receptor A-positive (PDGFRA+) adipocyte progenitors confirmed increased levels of de novo brown adipogenesis from PDGFRA+ cells by coumestrol treatment. In conclusion, our results indicate that coumestrol has antiobesity effects through the expansion and activation of brown adipose tissue metabolism.


Subject(s)
Adipose Tissue, Brown/metabolism , Coumestrol/pharmacology , Obesity/drug therapy , Obesity/metabolism , Adipocytes, Beige/drug effects , Adipogenesis , Adipose Tissue, Brown/drug effects , Adipose Tissue, White/metabolism , Adiposity , Animals , Body Weight , Diet, High-Fat , Glucose Tolerance Test , Lipolysis , Male , Mice , Mice, Inbred C57BL , Phytoestrogens/pharmacology
13.
Mol Metab ; 29: 86-98, 2019 11.
Article in English | MEDLINE | ID: mdl-31668395

ABSTRACT

OBJECTIVE: This study investigated the role of microRNAs generated from adipose tissue macrophages (ATMs) during adipose tissue remodeling induced by pharmacological and nutritional stimuli. METHODS: Macrophage-specific Dicer knockout (KO) mice were used to determine the roles of microRNA generated in macrophages in adipose tissue remodeling induced by the ß3-adrenergic receptor agonist CL316,243 (CL). RNA-seq was performed to characterize microRNA and mRNA expression profiles in isolated macrophages and PDGFRα+ adipocyte stem cells (ASCs). The role of miR-10a-5p was further investigated in cell culture, and in adipose tissue remodeling induced by CL treatment and high fat feeding. RESULTS: Macrophage-specific deletion of Dicer elevated pro-inflammatory gene expression and prevented CL-induced de novo beige adipogenesis in gonadal white adipose tissue (gWAT). Co-culture of ASCs with ATMs of wild type mice promoted brown adipocyte gene expression upon differentiation, but co-culture with ATMs of Dicer KO mice did not. Bioinformatic analysis of RNA expression profiles identified miR-10a-5p as a potential regulator of inflammation and differentiation in ATMs and ASCs, respectively. CL treatment increased levels of miR-10a-5p in ATMs and ASCs in gWAT. Interestingly, CL treatment elevated levels of pre-mir-10a in ATMs but not in ASCs, suggesting possible transfer from ATMs to ASCs. Elevating miR-10a-5p levels inhibited proinflammatory gene expression in cultured RAW 264.7 macrophages and promoted the differentiation of C3H10T1/2 cells into brown adipocytes. Furthermore, treatment with a miR-10a-5p mimic in vivo rescued CL-induced beige adipogenesis in Dicer KO mice. High fat feeding reduced miR-10a-5p levels in ATMs of gWAT, and treatment of mice with a miR-10a-5p mimic suppressed pro-inflammatory responses, promoted the appearance of new white adipocytes in gWAT, and improved systemic glucose tolerance. CONCLUSIONS: These results demonstrate an important role of macrophage-generated microRNAs in adipogenic niches and identify miR-10a-5p as a key regulator that reduces adipose tissue inflammation and promotes therapeutic adipogenesis.


Subject(s)
Adipose Tissue, White/metabolism , MicroRNAs/metabolism , 3' Untranslated Regions , Adipocytes, Brown/metabolism , Adipogenesis , Animals , Antagomirs/metabolism , Cell Differentiation , DEAD-box RNA Helicases/deficiency , DEAD-box RNA Helicases/genetics , Dioxoles/pharmacology , Down-Regulation/drug effects , Macrophage Activation/drug effects , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Nuclear Receptor Subfamily 1, Group F, Member 1/chemistry , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , RAW 264.7 Cells , Receptors, Adrenergic, beta-3/chemistry , Receptors, Adrenergic, beta-3/metabolism , Ribonuclease III/deficiency , Ribonuclease III/genetics , Stem Cells/cytology , Stem Cells/metabolism
14.
Sci Rep ; 8(1): 8856, 2018 06 11.
Article in English | MEDLINE | ID: mdl-29891910

ABSTRACT

15-lipoxygenase is involved in the generation of specialized pro-resolving lipid mediators that play essential roles in resolution and inflammatory responses. Here, we investigated anti-inflammatory role of Alox15 in skin homeostasis. We demonstrated that knockout (KO) of Alox15 led to hair loss and disrupted the structural integrity of the dorsal skin. Alox15 KO resulted in loss of hair follicle stem cells and abnormal transition of dermal adipocytes into fibroblasts. Alox15 deficiency increased infiltration of proinflammatory macrophages and upregulated proinflammatory and necroptotic signaling in dermal adipose tissue in the dorsal skin. Lipidomic analysis revealed severe loss of resolvin D2 in the dorsal skin of Alox15 KO mice compared to wild type controls. Treatment with resolvin D2 reduced skin inflammation in Alox15 KO mice. Collectively, these results indicate that Alox15-mediated production of resolvin D2 is required to maintain skin integrity by suppressing dermal inflammation.


Subject(s)
Arachidonate 12-Lipoxygenase/physiology , Arachidonate 15-Lipoxygenase/physiology , Docosahexaenoic Acids/metabolism , Inflammation , Skin/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Alopecia , Animals , Arachidonate 12-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/genetics , Cell Death , Fibroblasts/cytology , Gene Knockout Techniques , Hair Follicle/pathology , Homeostasis , Inflammation/metabolism , Inflammation Mediators/metabolism , Macrophages/cytology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL
...