Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
J Am Heart Assoc ; : e030084, 2023 Nov 10.
Article in English | MEDLINE | ID: mdl-37947148

ABSTRACT

Background Sterile inflammation caused by metabolic disorders impairs endothelial function; however, the underlying mechanism by which hyperglycemia induces inflammation remains obscure. Recent studies have suggested that stimulator of interferon genes (STING), a key cytosolic DNA sensor in the innate immune system, contributes to the pathogenesis of inflammatory diseases. This study examines the role of the STING in endothelial dysfunction in streptozotocin-induced diabetic mice. Methods and Results Injection of streptozotocin promoted the expression of STING and DNA damage markers in the aorta of wild-type mice. Streptozotocin elevated blood glucose and lipid levels in both wild-type and STING-deficient mice, which showed no statistical differences. Genetic deletion of STING ameliorated endothelial dysfunction as determined by the vascular relaxation in response to acetylcholine (P<0.001) and increased endothelial nitric oxide synthase phosphorylation in the aorta (P<0.05) in STZ-injected mice. Endothelium-independent vascular response to sodium nitroprusside did not differ. Treatment with a direct STING agonist, cyclic GMP-AMP, or mitochondrial DNA increased inflammatory molecule expression (eg, VCAM1 and IFNB) and decreased endothelial nitric oxide synthase phosphorylation in human umbilical vein endothelial cells, partially through the STING pathway. Cyclic GMP-AMP significantly impaired endothelial function of aortic segments obtained from wild-type mice, which was ameliorated in the presence of C-176, a STING inhibitor, or a neutralizing interferon-ß antibody. Furthermore, the administration of C-176 ameliorated endothelial dysfunction in STZ-induced diabetic mice (P<0.01). Conclusions The DNA damage response regulated by STING impairs endothelial function. STING signaling may be a potential therapeutic target of endothelial dysfunction caused by hyperglycemia.

3.
Sci Rep ; 13(1): 7446, 2023 05 08.
Article in English | MEDLINE | ID: mdl-37156857

ABSTRACT

Osteoarthritis (OA) is the most common joint disease associated with articular cartilage destruction. Matrix metalloproteinase-13 (MMP-13) has an essential role in OA pathogenesis by degradation of collagen II, a major component of articular cartilage. Hydrogen peroxide-inducible clone-5 (Hic-5; TGFB1I1), a transforming growth factor-ß-inducible mechanosensor, has previously been reported to promote OA pathogenesis by upregulating MMP-13 expression in mouse osteoarthritic lesions. In our current study, immunohistochemical analysis showed that Hic-5 protein expression was increased in human OA cartilage compared with normal cartilage. Functional experiments demonstrated that Hic-5 and MMP-13 expression was increased by mechanical stress, and mechanical stress-induced MMP-13 expression was suppressed by Hic-5 siRNA in human chondrocytes. Moreover, intracellular localization of Hic-5 shifted to the nucleus from focal adhesions in human chondrocytes subjected to mechanical stress, and nuclear Hic-5 increased MMP-13 gene expression. In vivo, intra-articular injection of Hic-5 siRNA decreased the Osteoarthritis Research Society International score and MMP-13 protein expression in articular cartilage of OA rats. Our findings suggest that Hic-5 regulates transcription of MMP-13 in human chondrocytes, and Hic-5 may be a novel therapeutic target for OA because OA progression was suppressed by intra-articular injection of Hic-5 siRNA in rats.


Subject(s)
Cartilage, Articular , Osteoarthritis , Animals , Humans , Mice , Rats , Cartilage, Articular/pathology , Cells, Cultured , Chondrocytes/metabolism , Cytoskeletal Proteins/metabolism , DNA-Binding Proteins/metabolism , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 13/metabolism , Osteoarthritis/metabolism , RNA, Small Interfering/metabolism , Transcription Factors/metabolism
4.
Arterioscler Thromb Vasc Biol ; 43(2): e66-e82, 2023 02.
Article in English | MEDLINE | ID: mdl-36519468

ABSTRACT

BACKGROUND: Although hypercholesterolemia reportedly counteracts lymphocyte trafficking across lymphatic vessels, the roles of lymphatic endothelial cells (LECs) in the lymphocyte regulations remain unclear. Previous studies showed that calpain-an intracellular modulatory protease-interferes with leukocyte dynamics in the blood microcirculation and is associated with hypercholesterolemic dysfunction in vascular endothelial cells. METHODS: This study investigated whether the calpain systems in LECs associate with the LEC-lymphocyte interaction under hypercholesterolemia using gene-targeted mice. RESULTS: Lipidomic analysis in hypercholesterolemic mice showed that several lysophospholipids, including lysophosphatidic acid, accumulated in the lymphatic environment. Lysophosphatidic acid enables the potentiation of calpain systems in cultured LECs, which limits their ability to stabilize regulatory T cells (Treg) without altering Th1/Th2 (T helper type1/2) subsets. This occurs via the proteolytic degradation of MEKK1 (mitogen-activated protein kinase kinase kinase 1) and the subsequent inhibition of TGF (transforming growth factor)-ß1 production in LECs. Targeting calpain systems in LECs expanded Tregs in the blood circulation and reduced aortic atherosclerosis in hypercholesterolemic mice, concomitant with the reduction of proinflammatory macrophages in the lesions. Treg expansion in the blood circulation and atheroprotection in calpain-targeted mice was prevented by the administration of TGF-ß type-I receptor inhibitor. Moreover, lysophosphatidic acid-induced calpain overactivation potentiated the IL (interleukin)-18/NF-κB (nuclear factor κB)/VCAM1 (vascular cell adhesion molecule 1) axis in LECs, thereby inhibiting lymphocyte mobility on the cells. Indeed, VCAM1 in LECs was upregulated in hypercholesterolemic mice and human cases of coronary artery disease. Neutralization of VCAM1 or targeting LEC calpain systems recovered afferent Treg transportation via lymphatic vessels in mice. CONCLUSIONS: Calpain systems in LECs have a key role in controlling Treg stability and trafficking under hypercholesterolemia.


Subject(s)
Hypercholesterolemia , Lymphatic Vessels , Mice , Humans , Animals , Endothelial Cells/metabolism , T-Lymphocytes, Regulatory/metabolism , Calpain/metabolism , Hypercholesterolemia/complications , Hypercholesterolemia/genetics , Hypercholesterolemia/metabolism , Lymphatic Vessels/metabolism , NF-kappa B/metabolism
5.
Eur Heart J ; 42(42): 4336-4348, 2021 11 07.
Article in English | MEDLINE | ID: mdl-34226923

ABSTRACT

AIMS: Lifestyle-related diseases promote atherosclerosis, a chronic inflammatory disease; however, the molecular mechanism remains largely unknown. Endogenous DNA fragments released under over-nutrient condition provoke sterile inflammation through the recognition by DNA sensors. Here, we investigated the role of stimulator of interferon genes (STING), a cytosolic DNA sensor, in atherogenesis. METHODS AND RESULTS: Apolipoprotein E-deficient (Apoe-/-) mice fed a western-type diet (WTD), a hypercholesterolaemic mouse model, showed higher STING expression and markers for DNA damage such as γH2AX, p53, and single-stranded DNA (ssDNA) accumulation in macrophages in the aorta compared with wild-type (WT) mice. The level of cGAMP, a STING agonist, in the aorta was higher in Apoe-/- mice. Genetic deletion of Sting in Apoe-/- mice reduced atherosclerotic lesions in the aortic arch, lipid, and macrophage accumulation in plaques, and inflammatory molecule expression in the aorta compared with the control. Pharmacological blockade of STING using a specific inhibitor, C-176, ameliorated atherogenesis in Apoe-/- mice. In contrast, bone marrow-specific STING expression in Apoe-/- mice stimulated atherogenesis. Expression or deletion of STING did not affect metabolic parameters and blood pressure. In vitro studies revealed that STING activation by cGAMP or mitochondrial DNA accelerated inflammatory molecule expression (e.g. TNF-α or IFN-ß) in mouse and human macrophages. Activation of nuclear factor-κB and TANK binding kinase 1 was involved in STING-associated vascular inflammation and macrophage activation. Furthermore, human atherosclerotic lesions in the carotid arteries expressed STING and cGAMP. CONCLUSION: Stimulator of interferon genes stimulates pro-inflammatory activation of macrophages, leading to the development of atherosclerosis. Stimulator of interferon genes signalling may serve as a potential therapeutic target for atherosclerosis.


Subject(s)
Atherosclerosis , Plaque, Atherosclerotic , Animals , Atherosclerosis/genetics , DNA , Disease Models, Animal , Immunity, Innate , Inflammation , Life Style , Mice , Mice, Inbred C57BL , Mice, Knockout
6.
Sci Rep ; 10(1): 19105, 2020 11 05.
Article in English | MEDLINE | ID: mdl-33154390

ABSTRACT

Accumulated evidence suggests that activated pancreatic stellate cells (PSCs) serve as the main source of the extracellular matrix proteins accumulated under the pathological conditions leading to pancreatic fibrosis in chronic pancreatitis (CP). However, little is known about the mechanisms of PSC activation. PSCs have morphologic and functional similarities to hepatic stellate cells, which are activated by hydrogen peroxide-inducible clone-5 (Hic-5), a TGF-ß1-induced protein. In this study, we investigated whether Hic-5 activates PSCs, which promote pancreatic fibrosis development in CP. Hic-5-knockout and wild type mice were subjected to caerulein injection to induce CP. Hic-5 expression was strongly upregulated in activated PSCs from human CP tissue and from mouse pancreatic fibrosis in caerulein-induced CP. Hic-5 deficiency significantly attenuated mouse pancreatic fibrosis and PSC activation in the experimental murine CP model. Mechanistically, Hic-5 knock down significantly inhibited the TGF-ß/Smad2 signaling pathway, resulting in reduced collagen production and α-smooth muscle actin expression in the activated PSCs. Taken together, we propose Hic-5 as a potential marker of activated PSCs and a novel therapeutic target in CP treatment.


Subject(s)
Cytoskeletal Proteins/genetics , DNA-Binding Proteins/genetics , Fibrosis/genetics , LIM Domain Proteins/genetics , Pancreatic Stellate Cells/metabolism , Pancreatitis, Chronic/genetics , Animals , Cells, Cultured , Cytoskeletal Proteins/metabolism , DNA-Binding Proteins/metabolism , Fibrosis/metabolism , Fibrosis/pathology , Humans , LIM Domain Proteins/metabolism , Mice , Mice, Knockout , Pancreatic Stellate Cells/pathology , Pancreatitis, Chronic/metabolism , Pancreatitis, Chronic/pathology , Transforming Growth Factor beta1/metabolism , Up-Regulation
7.
Sci Rep ; 9(1): 15770, 2019 10 31.
Article in English | MEDLINE | ID: mdl-31673109

ABSTRACT

Excessive mechanical stress is a major cause of knee osteoarthritis. However, the mechanism by which the mechanical stress begets osteoarthritis development remains elusive. Hydrogen peroxide-inducible clone-5 (Hic-5; TGFß1i1), a TGF-ß inducible focal adhesion adaptor, has previously been reported as a mediator of mechanotransduction. In this study, we analyzed the in vivo function of Hic-5 in development of osteoarthritis, and found that mice lacking Hic-5 showed a significant reduction in development of osteoarthritis in the knee. Furthermore, we found reduced expression of catabolic genes, such as metalloproteinase-13 and a disintegrin and metalloproteinase with thrombospondin type 1 motif 5 in osteoarthritic lesions in mice lacking Hic-5. During osteoarthritis development, Hic-5 is detected in chondrocytes of articular cartilage. To investigate the role of Hic-5 in chondrocytes, we isolated chondrocytes from articular cartilage of wild type and Hic-5-deficient mice. In these primary cultured chondrocytes, Hic-5 deficiency resulted in suppression of catabolic gene expression induced by osteoarthritis-related cytokines such as tumor necrosis factor α and interleukin 1ß. Furthermore, Hic-5 deficiency in chondrocytes suppressed catabolic gene expression induced by mechanical stress. Revealing the regulation of chondrocyte catabolism by Hic-5 contributes to understanding the pathophysiology of osteoarthritis induced by mechanical stress.


Subject(s)
Cartilage, Articular , Chondrocytes , Cytoskeletal Proteins/deficiency , DNA-Binding Proteins/deficiency , Gene Deletion , Gene Expression Regulation , LIM Domain Proteins/deficiency , Osteoarthritis , Animals , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Chondrocytes/metabolism , Chondrocytes/pathology , Cytoskeletal Proteins/metabolism , DNA-Binding Proteins/metabolism , LIM Domain Proteins/metabolism , Mice , Mice, Knockout , Osteoarthritis/genetics , Osteoarthritis/metabolism , Osteoarthritis/pathology
9.
Elife ; 82019 09 30.
Article in English | MEDLINE | ID: mdl-31566568

ABSTRACT

The molecular mechanisms by which environmental light conditions affect cerebellar development are incompletely understood. We showed that circadian disruption by light-at-night induced Purkinje cell death through pineal allopregnanolone (ALLO) activity during early life in chicks. Light-at-night caused the loss of diurnal variation of pineal ALLO synthesis during early life and led to cerebellar Purkinje cell death, which was suppressed by a daily injection of ALLO. The loss of diurnal variation of pineal ALLO synthesis induced not only reduction in pituitary adenylate cyclase-activating polypeptide (PACAP), a neuroprotective hormone, but also transcriptional repression of the cerebellar Adcyap1 gene that produces PACAP, with subsequent Purkinje cell death. Taken together, pineal ALLO mediated the effect of light on early cerebellar development in chicks.


Subject(s)
Brain/growth & development , Circadian Rhythm , Light , Pineal Gland/physiology , Pregnanolone/metabolism , Animals , Brain/cytology , COS Cells , Cell Death , Chickens , Chlorocebus aethiops , Male , Photic Stimulation , Purkinje Cells/cytology
10.
Cell Mol Life Sci ; 76(23): 4795-4809, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31201465

ABSTRACT

Fibrillin microfibrils are ubiquitous elements of extracellular matrix assemblies that play crucial roles in regulating the bioavailability of growth factors of the transforming growth factor beta superfamily. Recently, several "a disintegrin and metalloproteinase with thrombospondin motifs" (ADAMTS) proteins were shown to regulate fibrillin microfibril function. Among them, ADAMTS17 is the causative gene of Weill-Marchesani syndrome (WMS) and Weill-Marchesani-like syndrome, of which common symptoms are ectopia lentis and short stature. ADAMTS17 has also been linked to height variation in humans; however, the molecular mechanisms whereby ADAMTS17 regulates skeletal growth remain unknown. Here, we generated Adamts17-/- mice to examine the role of Adamts17 in skeletogenesis. Adamts17-/- mice recapitulated WMS, showing shorter long bones, brachydactyly, and thick skin. The hypertrophic zone of the growth plate in Adamts17-/- mice was shortened, with enhanced fibrillin-2 deposition, suggesting increased incorporation of fibrillin-2 into microfibrils. Comprehensive gene expression analysis of growth plates using laser microdissection and RNA sequencing indicated alteration of the bone morphogenetic protein (BMP) signaling pathway after Adamts17 knockout. Consistent with this, phospho-Smad1 levels were downregulated in the hypertrophic zone of the growth plate and in Adamts17-/- primary chondrocytes. Delayed terminal differentiation of Adamts17-/- chondrocytes, observed both in primary chondrocyte and primordial metatarsal cultures, and was prevented by BMP treatment. Our data indicated that Adamts17 is involved in skeletal formation by modulating BMP-Smad1/5/8 pathway, possibly through inhibiting the incorporation of fibrillin-2 into microfibrils. Our findings will contribute to further understanding of disease mechanisms and will facilitate the development of therapeutic interventions for WMS.


Subject(s)
ADAMTS Proteins/physiology , Bone Morphogenetic Proteins/metabolism , Muscle, Skeletal/growth & development , Signal Transduction , ADAMTS Proteins/genetics , Animals , Bone Morphogenetic Proteins/pharmacology , Cell Differentiation/drug effects , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/metabolism , Fibrillin-2/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfibrils/metabolism , Muscle, Skeletal/pathology , Skin/physiopathology , Smad1 Protein/metabolism , Smad5 Protein/metabolism , Smad8 Protein/metabolism , Weill-Marchesani Syndrome/metabolism , Weill-Marchesani Syndrome/pathology , Weill-Marchesani Syndrome/veterinary
11.
Nat Commun ; 10(1): 1442, 2019 03 29.
Article in English | MEDLINE | ID: mdl-30926814

ABSTRACT

Exposure of articular cartilage to excessive mechanical loading is deeply involved in the pathogenesis of osteoarthritis. Here, we identify gremlin-1 as a mechanical loading-inducible factor in chondrocytes, detected at high levels in middle and deep layers of cartilage after cyclic strain or hydrostatic pressure loading. Gremlin-1 activates nuclear factor-κB signalling, leading to subsequent induction of catabolic enzymes. In mice intra-articular administration of gremlin-1 antibody or chondrocyte-specific deletion of Gremlin-1 decelerates osteoarthritis development, while intra-articular administration of recombinant gremlin-1 exacerbates this process. Furthermore, ras-related C3 botulinum toxin substrate 1 activation induced by mechanical loading enhances reactive oxygen species (ROS) production. Amongst ROS-activating transcription factors, RelA/p65 induces Gremlin-1 transcription, which antagonizes induction of anabolic genes such as Sox9, Col2a1, and Acan by bone morphogenetic proteins. Thus, gremlin-1 plays essential roles in cartilage degeneration by excessive mechanical loading.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , NF-kappa B/metabolism , Osteoarthritis/metabolism , Osteoarthritis/pathology , Signal Transduction , Anabolic Agents/pharmacology , Animals , Bone Morphogenetic Proteins/pharmacology , Chondrocytes , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Reactive Oxygen Species/metabolism , Stress, Mechanical , Weight-Bearing , rac1 GTP-Binding Protein/metabolism
12.
J Am Heart Assoc ; 8(7): e010860, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30905257

ABSTRACT

Background Toll-like receptor ( TLR ) 9 recognizes bacterial DNA , activating innate immunity, whereas it also provokes inflammation in response to fragmented DNA released from mammalian cells. We investigated whether TLR 9 contributes to the development of vascular inflammation and atherogenesis using apolipoprotein E-deficient ( Apoe -/-) mice. Methods and Results Tlr9-deficient Apoe -/- ( Tlr9 -/- Apoe -/-) mice and Apoe -/- mice on a Western-type diet received subcutaneous angiotensin II infusion (1000 ng/kg per minute) for 28 days. Angiotensin II increased the plasma level of double-stranded DNA, an endogenous ligand of TLR 9, in these mice. Genetic deletion or pharmacologic blockade of TLR 9 in angiotensin II-infused Apoe -/- mice attenuated atherogenesis in the aortic arch ( P<0.05), reduced the accumulation of lipid and macrophages in atherosclerotic plaques, and decreased RNA expression of inflammatory molecules in the aorta with no alteration of metabolic parameters. On the other hand, restoration of TLR 9 in bone marrow in Tlr9 -/- Apoe -/- mice promoted atherogenesis in the aortic arch ( P<0.05). A TLR 9 agonist markedly promoted proinflammatory activation of Apoe -/- macrophages, partially through p38 mitogen-activated protein kinase signaling. In addition, genomic DNA extracted from macrophages promoted inflammatory molecule expression more effectively in Apoe -/- macrophages than in Tlr9 -/- Apoe -/- macrophages. Furthermore, in humans, circulating double-stranded DNA in the coronary artery positively correlated with inflammatory features of coronary plaques determined by optical coherence tomography in patients with acute myocardial infarction ( P<0.05). Conclusions TLR 9 plays a pivotal role in the development of vascular inflammation and atherogenesis through proinflammatory activation of macrophages. TLR 9 may serve as a potential therapeutic target for atherosclerosis.


Subject(s)
Atherosclerosis/genetics , Cell-Free Nucleic Acids/immunology , Inflammation/immunology , Macrophages/immunology , Plaque, Atherosclerotic/genetics , Toll-Like Receptor 9/genetics , Aged , Angiotensin II/toxicity , Animals , Aorta, Thoracic/pathology , Atherosclerosis/chemically induced , Atherosclerosis/immunology , Atherosclerosis/pathology , Bone Marrow Transplantation , Cell-Free Nucleic Acids/blood , Chemokine CCL2/genetics , Chemokine CCL2/immunology , Female , Humans , In Vitro Techniques , Inflammation/genetics , Lipids , Macrophages/pathology , Macrophages, Peritoneal/immunology , Male , Mice , Mice, Knockout , Mice, Knockout, ApoE , Microscopy, Electron , Myocardial Infarction/blood , Myocardial Infarction/therapy , Percutaneous Coronary Intervention , Plaque, Atherosclerotic/immunology , Plaque, Atherosclerotic/pathology , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptor 9/antagonists & inhibitors , Toll-Like Receptor 9/immunology , Tomography, Optical Coherence , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Vasoconstrictor Agents/toxicity
13.
FASEB J ; 33(2): 2037-2046, 2019 02.
Article in English | MEDLINE | ID: mdl-30199285

ABSTRACT

The transformation of fibroblasts to myofibroblasts plays a major role in fibrogenic responses during dermal wound healing. We show a contribution of calpain systems (intracellular regulatory protease systems) in vascular endothelial cells (ECs) to myofibroblast differentiation in wound sites. Dermal wound healing experiments in mice found that calpastatin (an endogenous inhibitor of calpains) is enriched in preexisting vessels but not in newly formed capillaries. Transgenic overexpression of calpastatin in ECs delayed wound healing in mice as well as reducing the keratinocyte layer, extracellular matrix deposition, and myofibroblast accumulation in wound sites. EC and leukocyte markers, however, remain unchanged. Calpastatin overexpression reduced the expression of genes encoding platelet-derived growth factor-B and PDGF receptor-ß (PDGFR-ß). Topical application of platelet-derived growth factor-BB-containing ointment to wounds accelerated healing in control mice, but calpastatin overexpression prevented this acceleration. In cultured human dermal fibroblasts, α-smooth muscle actin and PDGFR-ß were up-regulated by coculturing with ECs, but this action was inhibited by suppression of EC calpain activity. EC-driven transformation of mouse dermal fibroblasts was also suppressed by calpastatin overexpression in ECs. These results suggest that endothelial calpain systems influence PDGFR-ß signaling in fibroblasts, EC-driven myofibroblast differentiation, and subsequent fibrogenic responses in wounds.-Miyazaki, T., Haraguchi, S., Kim-Kaneyama, J.-R., Miyazaki, A. Endothelial calpain systems orchestrate myofibroblast differentiation during wound healing.


Subject(s)
Calpain/biosynthesis , Cell Differentiation , Dermis/metabolism , Endothelial Cells/enzymology , Myofibroblasts/metabolism , Wound Healing , Animals , Calcium-Binding Proteins/biosynthesis , Calcium-Binding Proteins/genetics , Calpain/genetics , Coculture Techniques , Dermis/pathology , Endothelial Cells/pathology , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Keratinocytes/metabolism , Keratinocytes/pathology , Mice , Mice, Transgenic , Myofibroblasts/pathology , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism
14.
Sci Rep ; 8(1): 2167, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29391479

ABSTRACT

The COS-7 (CV-1 in Origin with SV40 genes) cells are known as non-steroidogenic cells because they are derived from kidney cells and the kidney is defined as a non-steroidogenic organ. Therefore, COS-7 cells are used for transfection experiments to analyze the actions of functional molecules including steroids. However, a preliminary study suggested that COS-7 cells metabolize [3H]testosterone to [3H]androstenedione. These results suggest that COS-7 cells are able to metabolize steroids. Therefore, the present study investigated the expression of steroidogenic enzymes and the metabolism of steroids in COS-7 cells. RT-PCR analyses demonstrated the expressions of several kinds of steroidogenic enzymes, such as cytochrome P450 side-chain cleavage enzyme, 3ß-hydroxysteroid dehydrogenase/Δ5-Δ4 isomerase, cytochrome P450 7α-hydroxylase, cytochrome P450 17α-hydroxylase/17,20-lyase, 17ß-hydroxysteroid dehydrogenase, 5α-reductase, cytochrome P450 21-hydroxylase, cytochrome P450 11ß-hydroxylase, and cytochrome P450 aromatase in COS-7 cells. In addition, steroidogenic enzymes 3ß-HSD, P4507α, 5α-reductase, P450c17, P450c21, P450c11ß, and 17ß-HSD actively metabolized various steroids in cultured COS-7 cells. Finally, we demonstrated that 17ß-HSD activity toward androstenedione formation was greater than other steroidogenic enzyme activities. Our results provide new evidence that COS-7 cells express a series of steroidogenic enzyme mRNAs and actively metabolize a variety of steroids.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Metabolic Networks and Pathways , Steroid 17-alpha-Hydroxylase/metabolism , Steroids/metabolism , Animals , COS Cells , Chlorocebus aethiops , Cytochrome P-450 Enzyme System/genetics , Steroid 17-alpha-Hydroxylase/genetics
15.
Oncogene ; 37(9): 1205-1219, 2018 03.
Article in English | MEDLINE | ID: mdl-29242607

ABSTRACT

Carcinoma-associated fibroblasts (CAFs) influence tumor initiation, progression, and metastasis within the tumor-associated stroma. This suggests that CAFs would be a potential target for tumor therapy. Here we found that Hydrogen peroxide-inducible clone-5 (Hic-5), also named transforming growth factor beta-1-induced transcript 1 protein (Tgfb1i1), was strongly induced in CAFs found in human colorectal cancer. To investigate the role of Hic-5 in CAFs, we isolated CAFs and the control counterpart normal fibroblasts (NFs) from human colorectal cancer and non-cancerous regions, respectively. Hic-5 was highly expressed in isolated human CAFs and strongly induced in NFs in culture by the supernatant from cultured colorectal cancer cells as well as cytokines such as TGF-ß, IL-1ß and stromal cell-derived factor 1 (SDF-1/CXCL12). Furthermore, tumor growth was inhibited in a co-culture assay with Hic-5 knockdown fibroblasts compared with control fibroblasts. To clarify the function and significance of Hic-5 in colorectal cancer in vivo, we utilized a mouse model of azoxymethane (AOM)-induced colorectal cancer using Hic-5-deficient mice. Lack of Hic-5 in CAFs completely prevented AOM-induced colorectal cancer development in the colon tissues of mice. Mechanistic investigation revealed that Hic-5 promoted the expression of lysyl oxidase and collagen I in human control counterpart fibroblasts. Taken together, these results demonstrate that Hic-5 in CAFs is responsible for orchestrating or generating a tumor-promoting stroma.


Subject(s)
Cancer-Associated Fibroblasts/pathology , Carcinogenesis/pathology , Colorectal Neoplasms/pathology , Cytoskeletal Proteins/physiology , DNA-Binding Proteins/physiology , Fibroblasts/pathology , Intracellular Signaling Peptides and Proteins/metabolism , LIM Domain Proteins/metabolism , LIM Domain Proteins/physiology , Stromal Cells/pathology , Animals , Apoptosis , Azoxymethane/toxicity , Biomarkers, Tumor , Cancer-Associated Fibroblasts/metabolism , Carcinogenesis/metabolism , Cell Proliferation , Coculture Techniques , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/metabolism , Fibroblasts/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , LIM Domain Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Prognosis , Protein-Lysine 6-Oxidase , Signal Transduction , Stromal Cells/metabolism , Tumor Cells, Cultured
16.
Elife ; 62017 01 31.
Article in English | MEDLINE | ID: mdl-28139974

ABSTRACT

The visual responses of vertebrates are sensitive to the overall composition of retinal interneurons including amacrine cells, which tune the activity of the retinal circuitry. The expression of Paired-homeobox 6 (PAX6) is regulated by multiple cis-DNA elements including the intronic α-enhancer, which is active in GABAergic amacrine cell subsets. Here, we report that the transforming growth factor ß1-induced transcript 1 protein (Tgfb1i1) interacts with the LIM domain transcription factors Lhx3 and Isl1 to inhibit the α-enhancer in the post-natal mouse retina. Tgfb1i1-/- mice show elevated α-enhancer activity leading to overproduction of Pax6ΔPD isoform that supports the GABAergic amacrine cell fate maintenance. Consequently, the Tgfb1i1-/- mouse retinas show a sustained light response, which becomes more transient in mice with the auto-stimulation-defective Pax6ΔPBS/ΔPBS mutation. Together, we show the antagonistic regulation of the α-enhancer activity by Pax6 and the LIM protein complex is necessary for the establishment of an inner retinal circuitry, which controls visual adaptation.


Subject(s)
Cytoskeletal Proteins/metabolism , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic , Gene Expression Regulation , LIM Domain Proteins/metabolism , LIM-Homeodomain Proteins/metabolism , PAX6 Transcription Factor/metabolism , Retina/physiology , Transcription Factors/metabolism , Adaptation, Ocular , Animals , Mice , Mice, Knockout
17.
J Atheroscler Thromb ; 24(1): 1-13, 2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27476665

ABSTRACT

NADPH oxidases (NOX) are enzymes that catalyze the production of reactive oxygen species (ROS). Four species of NOX catalytic homologs (NOX1, NOX2, NOX4, and NOX5) are reportedly expressed in vascular tissues. The pro-atherogenic roles of NOX1, NOX2, and their organizer protein p47ph°x were manifested, and it was noted that the hydrogen peroxide-generating enzyme NOX4 possesses atheroprotective effects. Loss of NOX1 or p47ph°x appears to ameliorate murine aortic dissection and subsequent aneurysmal diseases; in contrast, the ablation of NOX2 exacerbates the aneurysmal diseases. It is possible that the loss of NOX2 activates inflammatory cascades in macrophages in the lesions. Roles of NOX5 in vascular functions are currently undetermined, owing to the absence of this enzyme in rodents and the limitation of the experimental procedure. Thus, it is possible that the NOX family of enzymes exhibits heterogeneity in the atherosclerotic diseases. In this aspect, subtype-selective NOX inhibitor may be promising when NOX systems serve as a molecular target for atherosclerotic and aneurysmal diseases.


Subject(s)
Aneurysm/enzymology , Aneurysm/physiopathology , Atherosclerosis/enzymology , Atherosclerosis/physiopathology , NADPH Oxidases/metabolism , Animals , Humans
18.
J Clin Invest ; 126(9): 3417-32, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27525442

ABSTRACT

Macrophages contribute to the development of atherosclerosis through pinocytotic deposition of native LDL-derived cholesterol in macrophages in the vascular wall. Inhibiting macrophage-mediated lipid deposition may have protective effects in atheroprone vasculature, and identifying mechanisms that potentiate this process may inform potential therapeutic interventions for atherosclerosis. Here, we report that dysregulation of exon junction complex-driven (EJC-driven) mRNA splicing confers hyperpinocytosis to macrophages during atherogenesis. Mechanistically, we determined that inflammatory cytokines induce an unconventional nonproteolytic calpain, calpain-6 (CAPN6), which associates with the essential EJC-loading factor CWC22 in the cytoplasm. This association disturbs the nuclear localization of CWC22, thereby suppressing the splicing of target genes, including those related to Rac1 signaling. CAPN6 deficiency in LDL receptor-deficient mice restored CWC22/EJC/Rac1 signaling, reduced pinocytotic deposition of native LDL in macrophages, and attenuated macrophage recruitment into the lesions, generating an atheroprotective phenotype in the aorta. In macrophages, the induction of CAPN6 in the atheroma interior limited macrophage movements, resulting in a decline in cell clearance from the lesions. Consistent with this finding, we observed that myeloid CAPN6 contributed to atherogenesis in a murine model of bone marrow transplantation. Furthermore, macrophages from advanced human atheromas exhibited increased CAPN6 induction and impaired CWC22 nuclear localization. Together, these results indicate that CAPN6 promotes atherogenicity in inflamed macrophages by disturbing CWC22/EJC systems.


Subject(s)
Atherosclerosis/pathology , Calpain/physiology , Macrophages/physiology , Microtubule-Associated Proteins/physiology , RNA Precursors , RNA Splicing , Adult , Aged , Aged, 80 and over , Animals , Aorta/metabolism , Atherosclerosis/genetics , Bone Marrow Transplantation , Calpain/genetics , Cell Nucleus/metabolism , Cytokines/metabolism , Female , Gene Expression Regulation , History, Ancient , Humans , Inflammation , Lipoproteins, LDL/metabolism , Male , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/genetics , Middle Aged , Monocytes/cytology , Neuropeptides/metabolism , Phenotype , Pinocytosis , Plaque, Atherosclerotic/metabolism , Signal Transduction , rac1 GTP-Binding Protein/metabolism
19.
J Nutr Biochem ; 35: 66-73, 2016 09.
Article in English | MEDLINE | ID: mdl-27416363

ABSTRACT

Oxidized cholesterols (oxycholesterols) in food have been recognized as strong atherogenic components, but their tissue distributions and roles in cardiovascular diseases remain unclear. To investigate whether accumulation of oxycholesterols is linked to cardiac morphology and function, and whether reduction of oxycholesterols can improve cardiac performance, domestic male swine were randomized to a control diet (C), high caloric diet (HCD) or HCD+Ezetimibe, an inhibitor of intestinal cholesterol absorption, group (HCD+E) and evaluated for: (1) distribution of oxycholesterol components in serum and tissues, (2) levels of oxycholesterol-related enzymes, (3) paracardial and epicardial coronary fat thickness, and (4) cardiac performance. Ezetimibe treatment for 8weeks attenuated increases in oxycholesterols in the HCD group almost completely in liver, but reduced only levels of 4ß-hydroxycholesterol in left ventricular (LV) myocardium. Ezetimibe treatment altered the expression of genes for cholesterol and fatty acid metabolism and decreased the expression of CYP3A46, which catabolizes cholesterol to 4ß-hydroxycholesterol, strongly in liver. An increase in epicardial fat thickness and impaired cardiac performance in the HCD group were improved by ezetimibe treatment, and the improvement was closely related to the reduction in levels of 4ß-hydroxycholesterol in LV myocardium. In conclusion, an increase in oxycholesterols in the HCD group was closely related to cardiac hypertrophy and dysfunction, as well as an increase in epicardial fat thickness. Ezetimibe may directly reduce oxycholesterol in liver and LV myocardium, and improve cardiac morphology and function.


Subject(s)
Adipose Tissue, White/drug effects , Adiposity/drug effects , Anticholesteremic Agents/therapeutic use , Cholesterol/analogs & derivatives , Heart Diseases/etiology , Heart/drug effects , Lipoproteins, LDL/adverse effects , Adipose Tissue, White/pathology , Animals , Cardiomegaly/etiology , Cardiomegaly/prevention & control , Cholesterol/adverse effects , Cholesterol/blood , Cholesterol/metabolism , Ezetimibe/therapeutic use , Gastrointestinal Agents/therapeutic use , Gene Expression Regulation, Enzymologic/drug effects , Heart/physiopathology , Heart Diseases/metabolism , Heart Diseases/pathology , Heart Diseases/physiopathology , Hydroxycholesterols/antagonists & inhibitors , Hydroxycholesterols/blood , Hydroxycholesterols/metabolism , Hyperphagia/physiopathology , Intestinal Absorption/drug effects , Lipoproteins, LDL/blood , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Myocardium/enzymology , Myocardium/metabolism , Myocardium/pathology , Oxidation-Reduction , Pericardium , Random Allocation , Sus scrofa
20.
J Cell Biol ; 213(5): 585-99, 2016 06 06.
Article in English | MEDLINE | ID: mdl-27269065

ABSTRACT

Invadosomes are acto-adhesive structures able to both bind the extracellular matrix (ECM) and digest it. Paxillin family members-paxillin, Hic-5, and leupaxin-are implicated in mechanosensing and turnover of adhesion sites, but the contribution of each paxillin family protein to invadosome activities is unclear. We use genetic approaches to show that paxillin and Hic-5 have both redundant and distinctive functions in invadosome formation. The essential function of paxillin-like activity is based on the coordinated activity of LD motifs and LIM domains, which support invadosome assembly and morphology, respectively. However, paxillin preferentially regulates invadosome assembly, whereas Hic-5 regulates the coupling between ECM degradation and acto-adhesive functions. Mass spectrometry analysis revealed new partners that are important for paxillin and Hic-5 specificities: paxillin regulates the acto-adhesive machinery through janus kinase 1 (JAK1), whereas Hic-5 controls ECM degradation via IQGAP1. Integrating the redundancy and specificities of paxillin and Hic-5 in a functional complex provides insights into the coupling between the acto-adhesive and ECM-degradative machineries in invadosomes.


Subject(s)
Cytoskeletal Proteins/metabolism , DNA-Binding Proteins/metabolism , Extracellular Matrix/metabolism , LIM Domain Proteins/metabolism , Paxillin/metabolism , Podosomes/metabolism , Amino Acid Motifs , Animals , Cell Adhesion , Janus Kinase 1/metabolism , Mice , Models, Biological , Paxillin/chemistry , Protein Binding , Protein Domains , Structure-Activity Relationship , ras GTPase-Activating Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...