Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Circ Res ; 130(2): 213-229, 2022 01 21.
Article in English | MEDLINE | ID: mdl-34870453

ABSTRACT

BACKGROUND: Vascular calcification is a prevalent complication in chronic kidney disease and contributes to increased cardiovascular morbidity and mortality. XBP1 (X-box binding protein 1), existing as the XBP1u (unspliced XBP1) and XBP1s (spliced XBP1) forms, is a key component of the endoplasmic reticulum stress involved in vascular diseases. However, whether XBP1u participates in the development of vascular calcification remains unclear. METHODS: We aim to investigate the role of XBP1u in vascular calcification. XBP1u protein levels were reduced in high phosphate-induced calcified vascular smooth muscle cells, calcified aortas from mice with adenine diet-induced chronic renal failure, and calcified radial arteries from patients with chronic renal failure. RESULTS: Inhibition of XBP1u rather than XBP1s upregulated in the expression of the osteogenic markers Runx2 (runt-related transcription factor 2) and Msx2 (msh homeobox 2), and exacerbated high phosphate-induced vascular smooth muscle cell calcification, as verified by calcium deposition and Alizarin red S staining. In contrast, XBP1u overexpression in high phosphate-induced vascular smooth muscle cells significantly inhibited osteogenic differentiation and calcification. Consistently, smooth muscle cell-specific XBP1 deficiency in mice markedly aggravated the adenine diet- and 5/6 nephrectomy-induced vascular calcification compared with that in the control littermates. Further interactome analysis revealed that XBP1u is bound directly to ß-catenin, a key regulator of vascular calcification, via amino acid (aa) 205-230 in its C-terminal degradation domain. XBP1u interacted with ß-catenin to promote its ubiquitin-proteasomal degradation and thus inhibited ß-catenin/TCF (T-cell factor)-mediated Runx2 and Msx2 transcription. Knockdown of ß-catenin abolished the effect of XBP1u deficiency on vascular smooth muscle cell calcification, suggesting a ß-catenin-mediated mechanism. Moreover, the degradation of ß-catenin promoted by XBP1u was independent of GSK-3ß (glycogen synthase kinase 3ß)-involved destruction complex. CONCLUSIONS: Our study identified XBP1u as a novel endogenous inhibitor of vascular calcification by counteracting ß-catenin and promoting its ubiquitin-proteasomal degradation, which represents a new regulatory pathway of ß-catenin and a promising target for vascular calcification treatment.


Subject(s)
RNA Splicing , Vascular Calcification/metabolism , X-Box Binding Protein 1/metabolism , beta Catenin/metabolism , Animals , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/metabolism , HEK293 Cells , Homeodomain Proteins/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Myocytes, Smooth Muscle/metabolism , Proteolysis , Rats , Rats, Sprague-Dawley , Ubiquitination , Vascular Calcification/genetics , X-Box Binding Protein 1/genetics
2.
Arterioscler Thromb Vasc Biol ; 43(6): e172-e189, 2023 06.
Article in English | MEDLINE | ID: mdl-37128913

ABSTRACT

BACKGROUND: Thoracic aortic aneurysm and dissection (TAAD) is a highly lethal vascular disease without effective drug therapy. Whether elevated serum concentrations of uric acid are involved in TAAD development remains unclear. METHODS: Serum uric acid levels were detected in different TAAD mouse models and patients. The urate-lowering drug allopurinol was administered in the drinking water of TAAD mice. Adenine diet-induced mice were established to investigate the role of hyperuricemia in TAAD formation and RNA-sequencing of thoracic aortas from these mice was performed. RESULTS: We found serum uric acid levels were elevated in various mouse TAAD models, including mice fed a ß-aminopropionitrile diet, Marfan mice with fibrillin-1 haploinsufficiency (Fbn1C1041G/+), and ApoE-/- mice infused with Ang II (angiotensin II), as well as in patients with TAAD. Administration of urate-lowering drug allopurinol in the drinking water significantly alleviated TAAD formation in ß-aminopropionitrile-treated mice, Fbn1C1041G/+ mice, and Ang II-infused ApoE-/- mice. Moreover, an adenine diet was used to induce hyperuricemia in mice. Intriguingly, a 4-week adenine diet feeding directly induced TAAD formation characterized by increased maximal thoracic aortic diameters and severe elastin degradation, which were ameliorated by allopurinol. Unbiased RNA-sequencing in mouse thoracic aortas suggested that FcγR (Fc gamma receptor) was upregulated upon adenine diet, but reciprocally repressed by allopurinol. Mechanistically, hyperuricemia activated FcγR-mediated ERK1/2 (extracellular signal-regulated kinase 1/2) phosphorylation to induce macrophage inflammation and TAAD development, which was abrogated by allopurinol or FcγR deficiency. CONCLUSIONS: This study uncovered an important and previously unrecognized role of hyperuricemia in mediating the pathogenesis of TAAD, and uric acid-lowering drug may represent a promising therapeutic approach for TAAD.


Subject(s)
Aortic Aneurysm, Thoracic , Aortic Dissection , Drinking Water , Hyperuricemia , Mice , Animals , Uric Acid , Aminopropionitrile/adverse effects , Allopurinol/adverse effects , Drinking Water/adverse effects , Hyperuricemia/chemically induced , Hyperuricemia/drug therapy , Receptors, IgG , Signal Transduction , Aortic Aneurysm, Thoracic/chemically induced , Aortic Aneurysm, Thoracic/genetics , Aortic Aneurysm, Thoracic/prevention & control , Aortic Dissection/chemically induced , Aortic Dissection/genetics , Aortic Dissection/prevention & control , RNA , Mice, Inbred C57BL , Disease Models, Animal
3.
Environ Res ; 245: 118074, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38160979

ABSTRACT

Exploring the effect of local government multi-objective competition on the transfer of polluting industries is of great practical significance for promoting the high-quality development in the Yangtze River Economic Belt. This paper adopted the extended shift-share analysis method to measure the scale of inter-provincial transfer of polluting industries in the Yangtze River Economic Belt from 2008 to 2020. Considering local governments' economic, innovation, talent and environmental protection competition, the paper examined the effects of local government multi-objective competition on the transfer of polluting industries in the region, and tested its spatial spillover effects. The results showed that: 1. Different competitions had different effects on the transfer of polluting industries. Economic competition intensified the transfer of polluting industries, while talent, innovation, and environmental protection competition all restrained it, among which environmental protection competition had the strongest restraining effect. 2. Compared with the transfer of polluting industries, the direction of economic competition and environmental protection competition on the transfer of industries did not change, but the degree of influence was reduced, talent competition instead promoted industrial transfer of the research region to some extent. 3. From the basin level, government competition in the upstream region more obviously intensified the transfer of polluting industries; while from the economic scale level, the restraining effect of government competition in the developed region on the transfer of polluting industries was much stronger. 4. Both innovation and environmental protection competition had positive spatial spillover effects. Therefore, it is necessary to optimize the promotion and assessment mechanism of local officials, adopt differentiated competitive constraint mechanisms in accordance with local conditions, guide local governments to transform their development concepts, promote the sharing and common use of technological innovations, and promote the orderly transfer of industries in the Yangtze River Economic Belt.


Subject(s)
Local Government , Rivers , Industry , Conservation of Natural Resources , Economic Development , China , Cities
4.
Eur Heart J ; 44(14): 1248-1261, 2023 04 07.
Article in English | MEDLINE | ID: mdl-36638776

ABSTRACT

AIMS: Whether changes in endothelial tight junctions (TJs) lead to the formation of thoracic aortic aneurysm and dissection (TAAD) and serve as an early indicator and therapeutic target remains elusive. METHODS AND RESULTS: Single-cell RNA sequencing analysis showed aberrant endothelial TJ expressions in the thoracic aortas of patients with TAAD. In a ß-aminopropionitrile (BAPN)-induced TAAD mouse model, endothelial TJ function was disrupted in the thoracic aortas at an early stage (5 and 10 days) as observed by a vascular permeability assay, while the intercellular distribution of crucial TJ components was significantly decreased by en face staining. For the non-invasive detection of endothelial TJ function, two dextrans of molecular weights 4 and 70 kDa were conjugated with the magnetic resonance imaging (MRI) contrast agent Gd-DOTA to synthesize FITC-dextran-DOTA-Gd and rhodamine B-dextran-DOTA-Gd. MRI images showed that both probes accumulated in the thoracic aortas of the BAPN-fed mice. Particularly, the mice with increased accumulated signals from 5 to 10 days developed TAAD at 14 days, whereas the mice with similar signals between the two time points did not. Furthermore, the protease-activated receptor 2 inhibitor AT-1001, which seals TJs, alleviated the BAPN-induced impairment of endothelial TJ function and expression and subsequently reduced TAAD incidence. Notably, endothelial-targeted ZO-1 conditional knockout increased TAAD incidence. Mechanistically, vascular inflammation and edema were observed in the thoracic aortas of the BAPN-fed mice, whereas these phenomena were attenuated by AT-1001. CONCLUSION: The disruption of endothelial TJ function is an early event prior to TAAD formation, herein serving as a potential indicator and a promising target for TAAD.


Subject(s)
Aortic Aneurysm, Thoracic , Aortic Dissection , Mice , Animals , Aminopropionitrile/adverse effects , Tight Junctions/metabolism , Tight Junctions/pathology , Signal Transduction , Aortic Aneurysm, Thoracic/prevention & control
5.
Am J Physiol Cell Physiol ; 317(4): C762-C775, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31365297

ABSTRACT

Compelling evidence indicates that epigenetic regulations orchestrate dynamic macrophage polarization. N6-methyladenosine (m6A) methylation is the most abundant epigenetic modification of mammalian mRNA, but its role in macrophage polarization is still completely unknown. Here, we show that the m6A-catalytic enzyme methyltransferase like 3 (METTL3) is specifically upregulated following the M1 polarization of mouse macrophages. Furthermore, METTL3 knockdown through siRNA transfection markedly inhibited M1, but enhanced M2, macrophage polarization. Conversely, its overexpression via plasmid transfection greatly facilitated M1, but attenuated M2, macrophage polarization. Further methylated RNA immunoprecipitation and in vitro m6A methylation assays suggested that METTL3 directly methylates mRNA encoding signal transducer and activator of transcription 1 (STAT1), a master transcription factor controlling M1 macrophage polarization, at its coding sequence and 3'-untranslated regions. In addition, METTL3-mediated STAT1 mRNA methylation significantly increased mRNA stability and subsequently upregulated STAT1 expression. In conclusion, METTL3 drives M1 macrophage polarization by directly methylating STAT1 mRNA, potentially serving as an anti-inflammatory target.


Subject(s)
Adenosine/analogs & derivatives , Anti-Inflammatory Agents/pharmacology , Macrophages/drug effects , Methyltransferases/drug effects , Adenosine/pharmacology , Animals , Gene Expression Regulation/drug effects , Macrophage Activation/drug effects , Macrophages/metabolism , Male , Methylation/drug effects , Mice, Inbred C57BL , RNA, Messenger/metabolism , STAT1 Transcription Factor/drug effects
6.
Arterioscler Thromb Vasc Biol ; 38(7): 1616-1631, 2018 07.
Article in English | MEDLINE | ID: mdl-29853563

ABSTRACT

OBJECTIVE: Chemokine-mediated neutrophil recruitment contributes to the pathogenesis of abdominal aortic aneurysm (AAA) and may serve as a promising therapeutic target. FAM3D (family with sequence similarity 3, member D) is a recently identified novel chemokine. Here, we aimed to explore the role of FAM3D in neutrophil recruitment and AAA development. APPROACH AND RESULTS: FAM3D was markedly upregulated in human AAA tissues, as well as both elastase- and CaPO4-induced mouse aneurysmal aortas. FAM3D deficiency significantly attenuated the development of AAA in both mouse models. Flow cytometry analysis indicated that FAM3D-/- mice exhibited decreased neutrophil infiltration in the aorta during the early stage of AAA formation compared with their wild-type littermates. Moreover, application of FAM3D-neutralizing antibody 6D7 through intraperitoneal injection markedly ameliorated elastase-induced AAA formation and neutrophil infiltration. Further, in vitro coculture experiments with FAM3D-neutralizing antibody 6D7 and in vivo intravital microscopic analysis indicated that endothelial cell-derived FAM3D induced neutrophil recruitment. Mechanistically, FAM3D upregulated and activated Mac-1 (macrophage-1 antigen) in neutrophils, whereas inhibition of FPR1 (formyl peptide receptor 1) or FPR2 significantly blocked FAM3D-induced Mac-1 activation, indicating that the effect of FAM3D was dependent on both FPRs. Moreover, specific inhibitors of FPR signaling related to Gi protein or ß-arrestin inhibited FAM3D-activated Mac-1 in vitro, whereas FAM3D deficiency decreased the activation of both FPR-Gi protein and ß-arrestin signaling in neutrophils in vivo. CONCLUSIONS: FAM3D, as a dual agonist of FPR1 and FPR2, induced Mac-1-mediated neutrophil recruitment and aggravated AAA development through FPR-related Gi protein and ß-arrestin signaling.


Subject(s)
Aorta, Abdominal/metabolism , Aortic Aneurysm, Abdominal/prevention & control , Cytokines/deficiency , Neutrophil Infiltration , Neutrophils/metabolism , Animals , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/pathology , Cells, Cultured , Coculture Techniques , Cytokines/genetics , Disease Models, Animal , Humans , Leukocyte Rolling , Macrophage-1 Antigen/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout, ApoE , Receptors, Formyl Peptide/metabolism , Signal Transduction , beta-Arrestins/metabolism
7.
Pancreatology ; 17(2): 247-254, 2017.
Article in English | MEDLINE | ID: mdl-28131523

ABSTRACT

BACKGROUND: Clinical evidence indicates that hepatic abnormalities in patients with chronic pancreatitis are not uncommon. Here we aimed to study the possible association between liver and pancreatic damage in a recently described experimental mouse model of CP. METHODS: The severity of the damage to pancreas, liver and other organs was assessed by biochemical markers and histopathology. The methods applied included Hematoxylin Eosin staining, electron microscope examination, biochemical measurements, RT-PCR, ELISA, and the correlations among some of the parameters contributing to these changes were statistically analyzed. RESULTS: The hepatic aberrations were mainly represented by mild infiltration of inflammatory cells in portal triad and congestion of central vein of liver, and the main features of drug-induced hepatotoxicity could not be observed. Severe fibrosis of pancreatic tissue was noticed in experimental group, and the existence of multiple organ injuries was also seen under the microscope. Hepatic pathologic scores were positively correlated with those from the corresponding pancreatic specimens (r = 0.72, P < 0.01). TGF-ß1 protein levels significantly elevated both in the test pancreas and liver (P < 0.05) and these values were positively correlated (r = 0.86, P < 0.01). The level of interleukin-1ß was increased in the serum and tissue of the liver. In addition, cardiac troponin (Tn-I) level not only significantly increased in myocardial homogenates (P < 0.05) but also was positively correlated with the corresponding pathologic score of the liver (r = 0.88, P < 0.01). CONCLUSION: The liver aberrations might be associated with L-arginine induced chronic pancreatitis.


Subject(s)
Arginine/toxicity , Liver Diseases/etiology , Liver/metabolism , Pancreatitis, Chronic/chemically induced , Pancreatitis, Chronic/complications , Animals , Enzyme-Linked Immunosorbent Assay , Immunochemistry/methods , Kidney/physiology , Liver Diseases/pathology , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
8.
Cell Rep Med ; 4(6): 101072, 2023 06 20.
Article in English | MEDLINE | ID: mdl-37301198

ABSTRACT

Current antihypertensive options still incompletely control blood pressure, suggesting the existence of uncovered pathogenic mechanisms. Here, whether cytokine-like protein family with sequence similarity 3, member D (FAM3D) is involved in hypertension etiology is evaluated. A case-control study exhibits that FAM3D is elevated in patients with hypertension, with a positive association with odds of hypertension. FAM3D deficiency significantly ameliorates angiotensin II (AngII)-induced hypertension in mice. Mechanistically, FAM3D directly causes endothelial nitric oxide synthase (eNOS) uncoupling and impairs endothelium-dependent vasorelaxation, whereas 2,4-diamino-6-hydroxypyrimidine to induce eNOS uncoupling abolishes the protective effect of FAM3D deficiency against AngII-induced hypertension. Furthermore, antagonism of formyl peptide receptor 1 (FPR1) and FPR2 or the suppression of oxidative stress blunts FAM3D-induced eNOS uncoupling. Translationally, targeting endothelial FAM3D by adeno-associated virus or intraperitoneal injection of FAM3D-neutralizing antibodies markedly ameliorates AngII- or deoxycorticosterone acetate (DOCA)-salt-induced hypertension. Conclusively, FAM3D causes eNOS uncoupling through FPR1- and FPR2-mediated oxidative stress, thereby exacerbating the development of hypertension. FAM3D may be a potential therapeutic target for hypertension.


Subject(s)
Cytokines , Hypertension , Animals , Mice , Blood Pressure/physiology , Case-Control Studies , Cytokines/metabolism , Hypertension/drug therapy , Hypertension/metabolism , Vasodilation/physiology , Humans
9.
Article in English | MEDLINE | ID: mdl-36030000

ABSTRACT

Abdominal aortic aneurysm (AAA) is a permanent dilatation of the abdominal aorta and is highly lethal. The main purpose of the current study is to search for noninvasive medical therapies for AAA, for which there is currently no effective drug therapy. Network medicine represents a cutting-edge technology, as analysis and modeling of disease networks can provide critical clues regarding the etiology of specific diseases and which therapeutics may be effective. Here, we proposed a novel algorithm to quantify disease relations based on a large accumulated microRNA-disease association dataset and then built a disease network covering 15 disease classes and 304 diseases. Analysis revealed some patterns for these diseases. For instance, diseases tended to be clustered and coherent in the network. Surprisingly, we found that AAA showed the strongest similarity with rheumatoid arthritis and systemic lupus erythematosus, both of which are autoimmune diseases, suggesting that AAA could be one type of autoimmune disease in etiology. Based on this observation, we further hypothesized that drugs for autoimmune disease could be repurposed for the prevention and therapy of AAA. Finally, animal experiments confirmed that methotrexate, a drug for autoimmune disease, was able to alleviated the formation and development of AAA.

SELECTION OF CITATIONS
SEARCH DETAIL