Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 166
Filter
1.
Int J Mol Sci ; 25(17)2024 Aug 24.
Article in English | MEDLINE | ID: mdl-39273142

ABSTRACT

Marfan syndrome (MFS) is a hereditary condition accompanied by disorders in the structural and regulatory properties of connective tissue, including elastic fibers, due to a mutation in the gene encodes for fibrillin-1 protein (FBN1 gene) and the synthesis of abnormal fibrillin-1 glycoprotein. Despite the high potential of mast cells (MCs) to remodel the extracellular matrix (ECM), their pathogenetic significance in MFS has not been considered yet. The group of patients with Marfan syndrome included two mothers and five children (three girls aged 4, 11, and 11 and two boys aged 12 and 13). Normal skin was examined in two children aged 11 and 12. Histochemical, monoplex, and multiplex immunohistochemical techniques; combined protocols of simultaneous histochemical and immunohistochemical staining (the results of staining were assessed using light, epifluorescence, and confocal microscopy); and bioinformatics algorithms for the quantitative analysis of detected targets were used to evaluate mast cells and their relationship with other cells from extracellular structures in the skin dermis. Analysis of the skin MC population in children with Marfan syndrome revealed a considerably increased number of intra-organic populations with the preservation of the specific Tryptase+Chymase+CPA3+ protease profile typical of the skin. The features of the MC histotopography phenotype in MFS consisted of closer colocalization with elastic fibers, smooth muscle cells, and fibroblasts. MCs formed many intradermal clusters that synchronized the activity of cell functions in the stromal landscape of the tissue microenvironment with the help of spatial architectonics, including the formation of cell chains and the creation of fibrous niches. In MCs, the expression of specific proteases, TGF-ß, and heparin increased, with targeted secretion of biologically active substances relative to the dermal elastic fibers, which had specific structural features in MFS, including abnormal variability in thickness along their entire length, alternating thickened and thinned areas, and uneven surface topography. This paper discusses the potential role of MCs in strain analysis (tensometry) of the tissue microenvironment in MFS. Thus, the quantitative and qualitative rearrangements of the skin MC population in MFS are aimed at altering the stromal landscape of the connective tissue. The results obtained should be taken into account when managing clinical signs of MFS manifested in other pathogenetically critical structures of internal organs, including the aorta, tendons, cartilage, and parenchymal organs.


Subject(s)
Dermis , Elastic Tissue , Marfan Syndrome , Mast Cells , Humans , Marfan Syndrome/metabolism , Marfan Syndrome/pathology , Marfan Syndrome/genetics , Mast Cells/metabolism , Mast Cells/pathology , Child , Male , Female , Elastic Tissue/metabolism , Elastic Tissue/pathology , Child, Preschool , Dermis/pathology , Dermis/metabolism , Adolescent , Fibrillin-1/metabolism , Fibrillin-1/genetics , Skin/metabolism , Skin/pathology , Extracellular Matrix/metabolism , Adipokines
2.
Int J Mol Sci ; 25(17)2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39273393

ABSTRACT

Triple-negative breast cancer (TNBC) is a subtype of breast cancer with a poor prognosis and limited treatment options. This study evaluates the prognostic value of stromal markers in TNBC, focusing on the tumor-stroma ratio (TSR) and overall stroma ratio (OSR) in whole slide images (WSI), as well as the expression of type-I collagen, type-III collagen, and fibrillin-1 on tissue microarrays (TMAs), using both visual assessment and digital image analysis (DIA). A total of 101 female TNBC patients, primarily treated with surgery between 2005 and 2016, were included. We found that high visual OSR correlates with worse overall survival (OS), advanced pN categories, lower stromal tumor-infiltrating lymphocyte count (sTIL), lower mitotic index, and patient age (p < 0.05). TSR showed significant connections to the pN category and mitotic index (p < 0.01). High expression levels of type-I collagen (>45%), type-III collagen (>30%), and fibrillin-1 (>20%) were linked to significantly worse OS (p = 0.004, p = 0.013, and p = 0.005, respectively) and progression-free survival (PFS) (p = 0.028, p = 0.025, and p = 0.002, respectively), validated at the mRNA level. Our results highlight the importance of stromal characteristics in promoting tumor progression and metastasis and that targeting extracellular matrix (ECM) components may offer novel therapeutic strategies. Furthermore, DIA can be more accurate and objective in evaluating TSR, OSR, and immunodetected stromal markers than traditional visual examination.


Subject(s)
Biomarkers, Tumor , Triple Negative Breast Neoplasms , Humans , Female , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/mortality , Prognosis , Middle Aged , Biomarkers, Tumor/metabolism , Aged , Adult , Stromal Cells/metabolism , Stromal Cells/pathology , Extracellular Matrix Proteins/metabolism , Extracellular Matrix Proteins/genetics , Fibrillin-1/metabolism , Fibrillin-1/genetics , Image Processing, Computer-Assisted/methods , Collagen Type I/metabolism , Collagen Type I/genetics , Collagen Type III/metabolism , Collagen Type III/genetics , Aged, 80 and over
3.
Int J Mol Sci ; 25(17)2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39273357

ABSTRACT

Genetic factors play a significant role in the pathogenesis of mitral valve diseases, including mitral valve prolapse (MVP) and mitral valve regurgitation. Genes like Fibrillin-1 (FBN1), Filamin A (FLNA), matrix metalloproteinase 2 (MMP2), and SRY-box transcription factor 9 (SOX9) are known to influence mitral valve pathology but knowledge of the exact mechanism is far from clear. Data regarding serum parameters, transesophageal echocardiography, and genetic and histopathologic parameters were investigated in 54 patients who underwent cardiovascular surgery for mitral valve regurgitation. The possible association between Fibrillin-1, Filamin A, MMP2, and SOX9 gene expressions was checked in relationship with the parameters of systemic inflammatory response. The mRNA expression levels (RQ-relative quantification) were categorized into three distinct groups: low (RQ < 1), medium/normal (RQ = 1-2), and high (RQ > 2). Severe fibrosis of the mitral valve was reflected by high expression of FBN1 and low expression of MMP2 (p < 0.05). The myxoid degeneration level was associated with the mRNA expression level for FBN1 and a low lymphocyte-monocyte ratio was associated with an increased mRNA expression of FBN1 (p < 0.05). A high number of monocytes was associated with high values of FBN1 whereas the increase in the number of lymphocytes was associated with high levels of MMP2. In addition, we observed that the risk of severe hyalinization was enhanced by a low mRNA expression of FLNA and/or SOX9. In conclusion, a lower FLNA mRNA expression can reflect the aging process that is highlighted in mitral valve pathology as a higher risk for hyalinization, especially in males, that might be prevented by upregulation of the SOX9 gene. FBN1 and MMP2 influence the inflammation-related fibrotic degeneration of the mitral valve. Understanding the genetic base of mitral valve pathology can provide insights into disease mechanisms, risk stratification, and potential therapeutic targets.


Subject(s)
Fibrillin-1 , Filamins , Matrix Metalloproteinase 2 , Mitral Valve , SOX9 Transcription Factor , Humans , Fibrillin-1/genetics , Fibrillin-1/metabolism , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics , Filamins/metabolism , Filamins/genetics , Male , Female , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 2/genetics , Middle Aged , Mitral Valve/pathology , Mitral Valve/metabolism , Aged , Mitral Valve Prolapse/genetics , Mitral Valve Prolapse/metabolism , Mitral Valve Prolapse/pathology , Mitral Valve Insufficiency/genetics , Mitral Valve Insufficiency/metabolism , Mitral Valve Insufficiency/pathology , Adipokines
4.
Cells ; 13(15)2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39120288

ABSTRACT

Vascular smooth muscle cells (VSMCs) play a critical role in maintaining vascular integrity. VSMC dysfunction leads to numerous vascular diseases. Adenosine deaminases acting on RNA 1 (ADAR1), an RNA editing enzyme, has shown both RNA editing and non-editing functions. Global deletion of ADAR1 causes embryonic lethality, but the phenotype of homozygous ADAR1 deletion specifically in SMCs (ADAR1sm-/-) remains to be determined. By crossing ADAR1fl/fl mice with Myh11-CreERT2 mice followed by Tamoxifen induction, we found that ADAR1sm-/- leads to lethality in adult mice 14 days after the induction. Gross examination revealed extensive hemorrhage and detrimental vascular damage in different organs. Histological analyses revealed destruction of artery structural integrity with detachment of elastin laminae from VSMCs in ADAR1sm-/- aortas. Furthermore, ADAR1sm-/- resulted in severe VSMC apoptosis and mitochondrial dysfunction. RNA sequencing analyses of ADAR1sm-/- aorta segments demonstrated profound transcriptional alteration of genes impacting vascular health including a decrease in fibrillin-1 expression. More importantly, ADAR1sm-/- disrupts the elastin and fibrillin-1 interaction, a molecular event essential for artery structure. Our results indicate that ADAR1 plays a critical role in maintaining SMC survival and vascular stability and resilience.


Subject(s)
Adenosine Deaminase , Homeostasis , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Animals , Adenosine Deaminase/metabolism , Adenosine Deaminase/genetics , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Mice , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Aorta/metabolism , Aorta/pathology , Apoptosis/genetics , Fibrillin-1/genetics , Fibrillin-1/metabolism , Elastin/metabolism , Mice, Knockout , Mice, Inbred C57BL , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics
5.
Tissue Cell ; 90: 102501, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39146675

ABSTRACT

AIM: Asprosin, a protein hormone, is released by unilocular adipocytes in reaction to low blood sugar. We aimed to examine how exercise affects asprosin hormone levels and associated organs, including the liver and pancreas, in diabetes. METHODS: Twenty-one male Wistar albino rats were firstly allocated into two main groups: control (n = 7) and diabetes (n = 14). Then, the diabetes group was further separated into two subgroups: sedentary (n = 7) and exercise (n = 7). The exercise group participated in a swimming training regimen (30 min/daily, six weeks). Serum levels of asprosin and various other biochemical parameters were evaluated through commercial ELISA kits. The liver was analyzed histopathologically, and pancreatic islet cells were examined for Cas-3 immune expression. RESULTS: Asprosin and total oxidant status decreased significantly in the exercise diabetic subgroup (p < 0.05). Glucose, insulin, creatinine, IL-6, and HomaIR concentrations decreased slightly with exercise (p > 0.05). Liver tissue injury scores and Cas-3 immune expression in pancreas islet cells decreased in exercise diabetic rats. CONCLUSIONS: Reducing asprosin may lower glucose, insulin, and HOMA-IR. Physical activity decreases asprosin and total oxidative status, fostering anti-apoptosis and tissue healing in diabetes, potentially enhancing health. Monitoring asprosin levels offers insights into diabetes progression. Our findings imply that asprosin can be a therapeutic target for diabetes.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Physical Conditioning, Animal , Rats, Wistar , Animals , Male , Rats , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Liver/metabolism , Liver/pathology , Blood Glucose/metabolism , Islet Amyloid Polypeptide/metabolism , Islet Amyloid Polypeptide/blood , Fibrillin-1/metabolism
6.
J Biol Chem ; 300(7): 107445, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38844137

ABSTRACT

Fibrillin microfibrils play a critical role in the formation of elastic fibers, tissue/organ development, and cardiopulmonary function. These microfibrils not only provide structural support and flexibility to tissues, but they also regulate growth factor signaling through a plethora of microfibril-binding proteins in the extracellular space. Mutations in fibrillins are associated with human diseases affecting cardiovascular, pulmonary, skeletal, and ocular systems. Fibrillins consist of up to 47 epidermal growth factor-like repeats, of which more than half are modified by protein O-glucosyltransferase 2 (POGLUT2) and/or POGLUT3. Loss of these modifications reduces secretion of N-terminal fibrillin constructs overexpressed in vitro. Here, we investigated the role of POGLUT2 and POGLUT3 in vivo using a Poglut2/3 double knockout (DKO) mouse model. Blocking O-glucosylation caused neonatal death with skeletal, pulmonary, and eye defects reminiscent of fibrillin/elastin mutations. Proteomic analyses of DKO dermal fibroblast medium and extracellular matrix provided evidence that fibrillins were more sensitive to loss of O-glucose compared to other POGLUT2/3 substrates. This conclusion was supported by immunofluorescent analyses of late gestation DKO lungs where FBN levels were reduced and microfibrils appeared fragmented in the pulmonary arteries and veins, bronchioles, and developing saccules. Defects in fibrillin microfibrils likely contributed to impaired elastic fiber formation and histological changes observed in DKO lung blood vessels, bronchioles, and saccules. Collectively, these results highlight the importance of POGLUT2/3-mediated O-glucosylation in vivo and open the possibility that O-glucose modifications on fibrillin influence microfibril assembly and or protein interactions in the ECM environment.


Subject(s)
Fibrillins , Lung , Mice, Knockout , Animals , Mice , Animals, Newborn , Extracellular Matrix/metabolism , Extracellular Matrix/genetics , Fibrillin-1/metabolism , Fibrillin-1/genetics , Fibrillins/metabolism , Fibrillins/genetics , Glycosylation , Lung/metabolism , Lung/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism
7.
Tissue Eng Part C Methods ; 30(7): 279-288, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38943281

ABSTRACT

The synthesis and assembly of mature, organized elastic fibers remains a limitation to the clinical use of many engineered tissue replacements. There is a critical need for a more in-depth understanding of elastogenesis regulation for the advancement of methods to induce and guide production of elastic matrix structures in engineered tissues that meet the structural and functional requirements of native tissue. The dramatic increase in elastic fibers through normal pregnancy has led us to explore the potential role of mechanical stretch in combination with pregnancy levels of the steroid hormones 17ß-estradiol and progesterone on elastic fiber production by human uterine myometrial smooth muscle cells in a three-dimensional (3D) culture model. Opposed to a single strain regimen, we sought to better understand how the amplitude and frequency parameters of cyclic strain influence elastic fiber production in these myometrial tissue constructs (MTC). Mechanical stretch was applied to MTC at a range of strain amplitudes (5%, 10%, and 15% at 0.5 Hz frequency) and frequencies (0.1 Hz, 0.5 Hz, 1 Hz, and constant 0 Hz at 10% amplitude), with and without pregnancy-level hormones, for 6 days. MTC were assessed for cell proliferation, matrix elastin protein content, and expression of the main elastic fiber genes, tropoelastin (ELN) and fibrillin-1 (FBN1). Significant increases in elastin protein and ELN and FBN1 mRNA were produced from samples subjected to a 0.5 Hz, 10% strain regimen, as well as samples stretched at higher amplitude (15%, 0.5 Hz) and higher frequency (1 Hz, 10%); however, no significant effects because of third-trimester mimetic hormone treatment were determined. These results establish that a minimum level of strain is required to stimulate the synthesis of elastic fiber components in our culture model and show this response can be similarly enhanced by increasing either the amplitude or frequency parameter of applied strain. Further, our results demonstrate strain alone is sufficient to stimulate elastic fiber production and suggest hormones may not be a significant factor in regulating elastin synthesis. This 3D culture model will provide a useful tool to further investigate mechanisms underlying pregnancy-induced de novo elastic fiber synthesis and assembly by uterine smooth muscle cells.


Subject(s)
Elastin , Myometrium , Stress, Mechanical , Female , Humans , Pregnancy , Adipokines , Cell Culture Techniques, Three Dimensional/methods , Cells, Cultured , Elastin/metabolism , Elastin/biosynthesis , Estradiol/biosynthesis , Estradiol/pharmacology , Estradiol/metabolism , Fibrillin-1/metabolism , Fibrillins/metabolism , Microfilament Proteins/metabolism , Microfilament Proteins/genetics , Models, Biological , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/cytology , Myometrium/metabolism , Myometrium/cytology , Tissue Engineering/methods , Tropoelastin/metabolism
8.
Antioxid Redox Signal ; 41(7-9): 488-504, 2024 09.
Article in English | MEDLINE | ID: mdl-38814824

ABSTRACT

Aims: Asprosin, a newly discovered hormone, is linked to insulin resistance. This study shows the roles of asprosin in vascular smooth muscle cell (VSMC) proliferation, migration, oxidative stress, and neointima formation of vascular injury. Methods: Mouse aortic VSMCs were cultured, and platelet-derived growth factor-BB (PDGF-BB) was used to induce oxidative stress, proliferation, and migration in VSMCs. Vascular injury was induced by repeatedly moving a guidewire in the lumen of the carotid artery in mice. Results: Asprosin overexpression promoted VSMC oxidative stress, proliferation, and migration, which were attenuated by toll-like receptor 4 (TLR4) knockdown, antioxidant (N-Acetylcysteine, NAC), NADPH oxidase 1 (NOX1) inhibitor ML171, or NOX2 inhibitor GSK2795039. Asprosin overexpression increased NOX1/2 expressions, whereas asprosin knockdown increased heme oxygenase-1 (HO-1) and NADPH quinone oxidoreductase-1 (NQO-1) expressions. Asprosin inhibited nuclear factor E2-related factor 2 (Nrf2) nuclear translocation. Nrf2 activator sulforaphane increased HO-1 and NQO-1 expressions and prevented asprosin-induced NOX1/2 upregulation, oxidative stress, proliferation, and migration. Exogenous asprosin protein had similar roles to asprosin overexpression. PDGF-BB increased asprosin expressions. PDGF-BB-induced oxidative stress, proliferation, and migration were enhanced by Nrf2 inhibitor ML385 but attenuated by asprosin knockdown. Vascular injury increased asprosin expression. Local asprosin knockdown in the injured carotid artery promoted HO-1 and NQO-1 expressions but attenuated the NOX1 and NOX2 upregulation, oxidative stress, neointima formation, and vascular remodeling in mice. Innovation and Conclusion: Asprosin promotes oxidative stress, proliferation, and migration of VSMCs via TLR4-Nrf2-mediated redox imbalance. Inhibition of asprosin expression attenuates VSMC proliferation and migration, oxidative stress, and neointima formation in the injured artery. Asprosin might be a promising therapeutic target for vascular injury. Antioxid. Redox Signal. 41, 488-504.


Subject(s)
Cell Movement , Cell Proliferation , Fibrillin-1 , Muscle, Smooth, Vascular , Neointima , Oxidative Stress , Vascular System Injuries , Animals , Oxidative Stress/drug effects , Neointima/metabolism , Neointima/pathology , Mice , Fibrillin-1/metabolism , Fibrillin-1/genetics , Cell Proliferation/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/cytology , Cell Movement/drug effects , Vascular System Injuries/metabolism , Vascular System Injuries/pathology , Myocytes, Smooth Muscle/metabolism , NF-E2-Related Factor 2/metabolism , Male , Toll-Like Receptor 4/metabolism , Disease Models, Animal
10.
J Hypertens ; 42(8): 1427-1439, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38690935

ABSTRACT

OBJECTIVE: Proliferation and migration of vascular smooth muscle cells (VSMCs) contribute to vascular remodeling. Asprosin, a newly discovered protein hormone, is involved in metabolic diseases. Little is known about the roles of asprosin in cardiovascular diseases. This study focused on the role and mechanism of asprosin on VSMC proliferation and migration, and vascular remodeling in a rat model of hypertension. METHODS AND RESULTS: VSMCs were obtained from the aortic media of 8-week-old male Wistar-Kyoto rats (WKY) and spontaneously hypertensive rats (SHR). Asprosin was upregulated in the VSMCs of SHR. For in vitro studies, asprosin promoted VSMC proliferation and migration of WKY and SHR, and increased Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX) activity, NOX1/2/4 protein expressions and superoxide production. Knockdown of asprosin inhibited the proliferation, migration, NOX activity, NOX1/2 expressions and superoxide production in the VSMCs of SHR. The roles of asprosin in promoting VSMC proliferation and migration were not affected by hydrogen peroxide scavenger, but attenuated by superoxide scavenger, selective NOX1 or NOX2 inhibitor. Toll-like receptor 4 (TLR4) was upregulated in SHR, TLR4 knockdown inhibited asprosin overexpression-induced proliferation, migration and oxidative stress in VSMCs of WKY and SHR. Asprosin was upregulated in arteries of SHR, and knockdown of asprosin in vivo not only attenuated oxidative stress and vascular remodeling in aorta and mesentery artery, but also caused a subsequent persistent antihypertensive effect in SHR. CONCLUSIONS: Asprosin promotes VSMC proliferation and migration via NOX-mediated superoxide production. Inhibition of endogenous asprosin expression attenuates VSMC proliferation and migration, and vascular remodeling of SHR.


Subject(s)
Cell Movement , Cell Proliferation , Hypertension , Muscle, Smooth, Vascular , Rats, Inbred SHR , Rats, Inbred WKY , Signal Transduction , Superoxides , Vascular Remodeling , Animals , Male , Superoxides/metabolism , Rats , Hypertension/metabolism , Hypertension/physiopathology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , NADPH Oxidases/metabolism , Peptide Hormones/metabolism , Fibrillin-1/metabolism , Toll-Like Receptor 4/metabolism
11.
Nat Commun ; 15(1): 4015, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38740766

ABSTRACT

Microfibril-associated glycoprotein 4 (MFAP4) is a 36-kDa extracellular matrix glycoprotein with critical roles in organ fibrosis, chronic obstructive pulmonary disease, and cardiovascular disorders, including aortic aneurysms. MFAP4 multimerises and interacts with elastogenic proteins, including fibrillin-1 and tropoelastin, and with cells via integrins. Structural details of MFAP4 and its potential interfaces for these interactions are unknown. Here, we present a cryo-electron microscopy structure of human MFAP4. In the presence of calcium, MFAP4 assembles as an octamer, where two sets of homodimers constitute the top and bottom halves of each octamer. Each homodimer is linked together by an intermolecular disulphide bond. A C34S missense mutation prevents disulphide-bond formation between monomers but does not prevent octamer assembly. The atomic model, built into the 3.55 Å cryo-EM map, suggests that salt-bridge interactions mediate homodimer assembly, while non-polar residues form the interface between octamer halves. In the absence of calcium, an MFAP4 octamer dissociates into two tetramers. Binding studies with fibrillin-1, tropoelastin, LTBP4, and small fibulins show that MFAP4 has multiple surfaces for protein-protein interactions, most of which depend upon MFAP4 octamer assembly. The C34S mutation does not affect these protein interactions or cell interactions. MFAP4 assemblies with fibrillin-1 abrogate MFAP4 interactions with cells.


Subject(s)
Cryoelectron Microscopy , Extracellular Matrix Proteins , Fibrillin-1 , Microfibrils , Tropoelastin , Humans , Adipokines , Calcium/metabolism , Extracellular Matrix Proteins/metabolism , Extracellular Matrix Proteins/chemistry , Extracellular Matrix Proteins/genetics , Fibrillin-1/metabolism , Fibrillin-1/genetics , Fibrillin-1/chemistry , Glycoproteins , HEK293 Cells , Microfibrils/metabolism , Microfibrils/chemistry , Microfibrils/ultrastructure , Models, Molecular , Mutation, Missense , Protein Binding , Protein Multimerization , Tropoelastin/metabolism , Tropoelastin/chemistry , Tropoelastin/genetics
12.
Sci Rep ; 14(1): 11313, 2024 05 17.
Article in English | MEDLINE | ID: mdl-38760452

ABSTRACT

Physical activity promotes various metabolic benefits by balancing pro and anti-inflammatory adipokines. Recent studies suggest that asprosin might be involved in progression of metabolic syndrome (MetS), however, the underlying mechanisms have not been understood yet. This study aimed to evaluate the effects of high-intensity interval training (HIIT), moderate-intensity continuous training (MICT), and further detraining on MetS indices, insulin resistance, serum and the liver levels of asprosin, and AMP-activated protein kinase (AMPK) pathway in menopause-induced MetS model of rats. A total of 64 Wistar rats were used in this study and divided into eight groups: Sham1, OVX1 (ovariectomized), Sham2, OVX2, OVX + HIIT, OVX + MICT, OVX + HIIT + Det (detraining), and OVX + MICT + Det. Animals performed the protocols, and then serum concentrations of asprosin, TNF-α, insulin, fasting blood glucose, and lipid profiles (TC, LDL, TG, and HDL) were assessed. Additionally, the liver expression of asprosin, AMPK, and P-AMPK was measured by western blotting. Both HIIT and MICT caused a significant decrease in weight, waist circumference, BMI (P = 0.001), and serum levels of glucose, insulin, asprosin (P = 0.001), triglyceride, total cholesterol, low-density lipoprotein (LDL), and TNF-α (P = 0.001), but an increase in the liver AMPK, P-AMPK, and P-AMPK/AMPK (P = 0.001), compared with OVX2 noexercised group. MICT was superior to HIIT in reducing serum asprosin, TNF-a, TG, LDL (P = 0.001), insulin, fasting blood glucose, HOMA-IR, and QUEKI index (P = 0.001), but an increase in the liver AMPK, and p-AMPK (P = 0.001). Although after two months of de-training almost all indices returned to the pre exercise values (P < 0.05). The findings suggest that MICT effectively alleviates MetS induced by menopause, at least partly through the activation of liver signaling of P-AMPK and the reduction of asprosin and TNF-α. These results have practical implications for the development of exercise interventions targeting MetS in menopausal individuals, emphasizing the potential benefits of MICT in mitigating MetS-related complications.


Subject(s)
AMP-Activated Protein Kinases , Disease Models, Animal , Fibrillin-1 , Metabolic Syndrome , Physical Conditioning, Animal , Rats, Wistar , Signal Transduction , Animals , Fibrillin-1/metabolism , Metabolic Syndrome/metabolism , Metabolic Syndrome/therapy , Rats , Female , AMP-Activated Protein Kinases/metabolism , High-Intensity Interval Training/methods , Liver/metabolism , Insulin Resistance , Blood Glucose/metabolism , Insulin/blood , Insulin/metabolism , Peptide Fragments/blood , Peptide Fragments/metabolism
13.
Int J Biol Macromol ; 268(Pt 2): 131868, 2024 May.
Article in English | MEDLINE | ID: mdl-38677690

ABSTRACT

Phenotype transformation of vascular smooth muscle cells (VSMCs) plays an important role in the development of atherosclerosis. Asprosin is a newly discovered adipokine, which is critical in regulating metabolism. However, the relationship between asprosin and phenotype transformation of VSMCs in atherosclerosis remains unclear. The aim of this study is to investigate whether asprosin affects the progression of atherosclerosis by inducing phenotype transformation of VSMCs. We established an atherosclerosis model in ApoE-/- mice and administered asprosin recombinant protein and asprosin antibody to mice. Knocking down asprosin was also as an intervention. Interestingly, we found a correlation between asprosin levels and atherosclerosis. Asprosin promoted plaque formation and phenotype transformation of VSMCs. While, AspKD or asprosin antibody reduced the plaque lesion and suppressed vascular stiffness in ApoE-/- mice. Mechanistically, asprosin induced phenotype transformation of MOVAs by binding to GPR54, leading to Gαq/11 recruitment and activation of the PLC-PKC-ERK1/2-STAT3 signaling pathway. Si GPR54 or GPR54 antagonist partially inhibited the action of asprosin in MOVAs. Mutant GPR54-(267, 307) residue cancelled the binding of asprosin and GPR54. In summary, this study confirmed asprosin activated GPR54/Gαq/11-dependent ERK1/2-STAT3 signaling pathway, thereby promoting VSMCs phenotype transformation and aggravating atherosclerosis, thus providing a new target for the treatment of atherosclerosis.


Subject(s)
Atherosclerosis , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Phenotype , Animals , Atherosclerosis/metabolism , Atherosclerosis/pathology , Atherosclerosis/genetics , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Mice , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Fibrillin-1/metabolism , Fibrillin-1/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Male , Signal Transduction , Disease Models, Animal , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Humans , Extracellular Matrix Proteins/metabolism , Extracellular Matrix Proteins/genetics , Mice, Knockout
14.
J Endocrinol Invest ; 47(8): 1839-1850, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38568373

ABSTRACT

BACKGROUND AND PURPOSE: Asprosin was discovered as a new endocrine hormone originating from fibrillin-1 cleavage that plays a crucial role in various metabolic-related diseases, such as obesity, nonalcoholic fatty liver disease (NAFLD), diabetes, polycystic ovary syndrome (PCOS), and cardiovascular diseases. The purpose of this review is to describe the recent advancements of asprosin. METHOD: Narrative review. RESULT: This comprehensive review explores its tissue-specific functions, focusing on white adipose tissue, liver, hypothalamus, testis, ovary, heart, pancreas, skeletal muscle, and kidney. CONCLUSION: Asprosin is a multifaceted protein with tissue-specific roles in various physiological and pathological processes. Further research is needed to fully understand the mechanisms and potential of asprosin as a therapeutic target. These insights could provide new directions for treatments targeting metabolic-related diseases.


Subject(s)
Fibrillin-1 , Metabolic Diseases , Humans , Fibrillin-1/metabolism , Metabolic Diseases/metabolism , Animals , Extracellular Matrix Proteins/metabolism , Adipokines
15.
Arterioscler Thromb Vasc Biol ; 44(7): 1540-1554, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38660802

ABSTRACT

BACKGROUND: Myxomatous valve disease (MVD) is the most common cause of mitral regurgitation, leading to impaired cardiac function and heart failure. MVD in a mouse model of Marfan syndrome includes valve leaflet thickening and progressive valve degeneration. However, the underlying mechanisms by which the disease progresses remain undefined. METHODS: Mice with Fibrillin 1 gene variant Fbn1C1039G/+ recapitulate histopathologic features of Marfan syndrome, and Wnt (Wingless-related integration site) signaling activity was detected in TCF/Lef-lacZ (T-cell factor/lymphoid enhancer factor-ß-galactosidase) reporter mice. Single-cell RNA sequencing was performed from mitral valves of wild-type and Fbn1C1039G/+ mice at 1 month of age. Inhibition of Wnt signaling was achieved by conditional induction of the secreted Wnt inhibitor Dkk1 (Dickkopf-1) expression in periostin-expressing valve interstitial cells of Periostin-Cre; tetO-Dkk1; R26rtTA; TCF/Lef-lacZ; Fbn1C1039G/+ mice. Dietary doxycycline was administered for 1 month beginning with MVD initiation (1-month-old) or MVD progression (2-month-old). Histological evaluation and immunofluorescence for ECM (extracellular matrix) and immune cells were performed. RESULTS: Wnt signaling is activated early in mitral valve disease progression, before immune cell infiltration in Fbn1C1039G/+ mice. Single-cell transcriptomics revealed similar mitral valve cell heterogeneity between wild-type and Fbn1C1039G/+ mice at 1 month of age. Wnt pathway genes were predominantly expressed in valve interstitial cells and valve endothelial cells of Fbn1C1039G/+ mice. Inhibition of Wnt signaling in Fbn1C1039G/+ mice at 1 month of age prevented the initiation of MVD as indicated by improved ECM remodeling and reduced valve leaflet thickness with decreased infiltrating macrophages. However, later, Wnt inhibition starting at 2 months did not prevent the progression of MVD. CONCLUSIONS: Wnt signaling is involved in the initiation of mitral valve abnormalities and inflammation but is not responsible for later-stage valve disease progression once it has been initiated. Thus, Wnt signaling contributes to MVD progression in a time-dependent manner and provides a promising therapeutic target for the early treatment of congenital MVD in Marfan syndrome.


Subject(s)
Disease Models, Animal , Disease Progression , Fibrillin-1 , Mitral Valve , Wnt Signaling Pathway , Animals , Fibrillin-1/genetics , Fibrillin-1/metabolism , Mitral Valve/metabolism , Mitral Valve/pathology , Mitral Valve/drug effects , Mice , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Mice, Transgenic , Marfan Syndrome/genetics , Marfan Syndrome/complications , Marfan Syndrome/metabolism , Marfan Syndrome/pathology , Mitral Valve Insufficiency/pathology , Mitral Valve Insufficiency/metabolism , Mitral Valve Insufficiency/prevention & control , Mitral Valve Insufficiency/genetics , Mice, Inbred C57BL , Inflammation/metabolism , Inflammation/pathology , Inflammation/prevention & control , Inflammation/genetics , Male , Female , Cell Adhesion Molecules , Adipokines
16.
J Dermatol ; 51(6): 816-826, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38470170

ABSTRACT

Ultraviolet (UV)-induced skin photoaging is caused by qualitative and quantitative degradation of dermal extracellular matrix components such as collagen and elastic fibers. Elastic fibers are important for maintaining cutaneous elasticity, despite their small amount in the skin. Previously, microfibril-associated protein 4 (MFAP-4), which is downregulated in photoaging dermis, has been found to be essential for elastic fiber formation by interaction with both fibrillin-1 and elastin, which are core components of elastic fiber. In addition, enhanced cutaneous MFAP-4 expression in a human skin-xenografted murine photoaging model protects against UV-induced photodamage accompanied by the prevention of elastic fiber degradation and aggravated elasticity. We therefore hypothesized that the upregulation of MFAP-4 in dermal fibroblasts may more efficiently accelerate elastic fiber formation. We screened botanical extracts for MFAP-4 expression-promoting activity in normal human dermal fibroblasts (NHDFs). We found that rosemary extract markedly promotes early microfibril formation and mature elastic fiber formation along with a significant upregulation of not only MFAP-4 but also fibrillin-1 and elastin in NHDFs. Furthermore, rosmarinic acid, which is abundant in rosemary extract, accelerated elastic fiber formation via upregulation of transforming growth factor ß-1. This was achieved by the induction of cAMP response element-binding protein phosphorylation, demonstrating that rosmarinic acid represents one of the active ingredients in rosemary extract. Based on the findings in this study, we conclude that rosemary extract and rosmarinic acid represent promising materials that exert a preventive or ameliorative effect on skin photoaging by accelerating elastic fiber formation.


Subject(s)
Cinnamates , Depsides , Elastic Tissue , Elastin , Fibrillin-1 , Fibroblasts , Plant Extracts , Rosmarinic Acid , Skin Aging , Humans , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/radiation effects , Depsides/pharmacology , Fibrillin-1/metabolism , Cinnamates/pharmacology , Plant Extracts/pharmacology , Elastin/metabolism , Elastic Tissue/drug effects , Elastic Tissue/metabolism , Skin Aging/drug effects , Skin Aging/radiation effects , Cells, Cultured , Rosmarinus/chemistry , Up-Regulation/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Skin/drug effects , Skin/radiation effects , Skin/cytology , Skin/pathology , Skin/metabolism , Dermis/cytology , Dermis/drug effects , Dermis/metabolism , Ultraviolet Rays/adverse effects , Extracellular Matrix Proteins/metabolism , Adipokines
17.
Int J Mol Sci ; 25(5)2024 Feb 25.
Article in English | MEDLINE | ID: mdl-38473909

ABSTRACT

Mutations of the FBN1 gene lead to Marfan syndrome (MFS), which is an autosomal dominant connective tissue disorder featured by thoracic aortic aneurysm risk. There is currently no effective treatment for MFS. Here, we studied the role of mitochondrial dysfunction in the phenotypic transformation of human smooth muscle cells (SMCs) and whether a mitochondrial boosting strategy can be a potential treatment. We knocked down FBN1 in SMCs to create an MFS cell model and used rotenone to induce mitochondrial dysfunction. Furthermore, we incubated the shFBN1 SMCs with Coenzyme Q10 (CoQ10) to assess whether restoring mitochondrial function can reverse the phenotypic transformation. The results showed that shFBN1 SMCs had decreased TFAM (mitochondrial transcription factor A), mtDNA levels and mitochondrial mass, lost their contractile capacity and had increased synthetic phenotype markers. Inhibiting the mitochondrial function of SMCs can decrease the expression of contractile markers and increase the expression of synthetic genes. Imposing mitochondrial stress causes a double-hit effect on the TFAM level, oxidative phosphorylation and phenotypic transformation of FBN1-knockdown SMCs while restoring mitochondrial metabolism with CoQ10 can rapidly reverse the synthetic phenotype. Our results suggest that mitochondria function is a potential therapeutic target for the phenotypic transformation of SMCs in MFS.


Subject(s)
Marfan Syndrome , Mitochondrial Diseases , Ubiquinone/analogs & derivatives , Humans , Marfan Syndrome/genetics , Phenotype , Myocytes, Smooth Muscle/metabolism , Mitochondrial Diseases/metabolism , Fibrillin-1/metabolism , Adipokines/metabolism
18.
Environ Sci Pollut Res Int ; 31(13): 19674-19686, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38363509

ABSTRACT

Evidence around the relationship between air pollution and the development of diabetes mellitus (DM) remains limited and inconsistent. To investigate the potential mediation effect of asprosin on the association between fine particulate matter (PM2.5), tropospheric ozone (O3) and blood glucose homeostasis. A case-control study was conducted on a total of 320 individuals aged over 60 years, including both diabetic and non-diabetic individuals, from six communities in Taiyuan, China, from July to September 2021. Generalized linear models (GLMs) suggested that short-term exposure to PM2.5 was associated with elevated fasting blood glucose (FBG), insulin resistance index (HOMA-IR), as well as reduced pancreatic ß-cell function index (HOMA-ß), and short-term exposure to O3 was associated with increased FBG and decreased HOMA-ß in the total population and elderly diabetic patients. Mediation analysis showed that asprosin played a mediating role in the relationship of PM2.5 and O3 with FBG, with mediating ratios of 10.2% and 18.4%, respectively. Our study provides emerging evidence supporting that asprosin mediates the short-term effects of exposure to PM2.5 and O3 on elevated FBG levels in an elderly population. Additionally, the elderly who are diabetic, over 70 years, and BMI over 24 kg/m2 are more vulnerable to air pollutants and need additional protection to reduce their exposure to air pollution.


Subject(s)
Air Pollutants , Air Pollution , Diabetes Mellitus , Fibrillin-1 , Aged , Humans , Middle Aged , Air Pollutants/adverse effects , Air Pollution/adverse effects , Blood Glucose/metabolism , Case-Control Studies , China/epidemiology , Diabetes Mellitus/metabolism , Environmental Exposure/analysis , Particulate Matter/analysis , Fibrillin-1/metabolism , Adipokines/metabolism
19.
Sci Rep ; 14(1): 3517, 2024 02 12.
Article in English | MEDLINE | ID: mdl-38347040

ABSTRACT

Aqueous humor (AH) and blood levels of transforming growth factor ß (TGFß) are elevated in idiopathic primary open angle glaucoma (POAG) representing a disease biomarker of unclear status and function. Tsk mice display a POAG phenotype and harbor a mutation of fibrillin-1, an important regulator of TGFß bioavailability. AH TGFß2 was higher in Tsk than wild-type (WT) mice (by 34%; p = 0.002; ELISA); similarly, AH TGFß2 was higher in human POAG than controls (2.7-fold; p = 0.00005). As in POAG, TGFß1 was elevated in Tsk serum (p = 0.01). Fibrillin-1 was detected in AH from POAG subjects and Tsk mice where both had similar levels relative to controls (p = 0.45). 350 kDa immunoblot bands representing WT full-length fibrillin-1 were present in human and mouse AH. A 418 kDa band representing mutant full-length fibrillin-1 was present only in Tsk mice. Lower molecular weight fibrillin-1 antibody-reactive bands were present in similar patterns in humans and mice. Certain bands (130 and 32 kDa) were elevated only in human POAG and Tsk mice (p ≤ 0.04 relative to controls) indicating discrete isoforms relevant to disease. In addition to sharing a phenotype, Tsk mice and human POAG subjects had common TGFß and fibrillin-1 features in AH and also blood that are pertinent to understanding glaucoma pathogenesis.


Subject(s)
Aqueous Humor , Glaucoma, Open-Angle , Animals , Humans , Mice , Aqueous Humor/metabolism , Fibrillin-1/genetics , Fibrillin-1/metabolism , Phenotype , Transforming Growth Factor beta/metabolism
20.
Mol Biotechnol ; 66(5): 1266-1278, 2024 May.
Article in English | MEDLINE | ID: mdl-38206528

ABSTRACT

The objective of this study was to investigate the mechanism of curcumin in diabetic foot ulcer (DFU) wound healing. A DFU rat model was established, and fibroblasts were cultured in a high-glucose (HG) environment to create a cell model. Various techniques, including Western blot, RT‒qPCR, flow cytometry, Transwell, cell scratch test and H&E staining, were employed to measure the levels of relevant genes and proteins, as well as to assess cell proliferation, apoptosis, migration, and pathological changes. The results showed that miR-152-3p was overexpressed in DFU patients, while FBN1 was underexpressed. Curcumin was found to inhibit fibroblast apoptosis, promote proliferation, migration, and angiogenesis in DFU rats, and accelerate wound healing in DFU rats. In addition, overexpression of miR-152-3p weakened the therapeutic effect of curcumin, while overexpression of FBN1 reversed the effects of the miR-152-3p mimic. Further investigations into the underlying mechanisms revealed that curcumin expedited wound healing in DFU rats by restoring the FBN1/TGF-ß pathway through the inhibition of miR-152-3p. In conclusion, curcumin can suppress the activity of miR-152-3p, which, in turn, leads to the rejuvenation of the FBN1/TGF-ß pathway and accelerates DFU wound healing.


Subject(s)
Curcumin , Diabetic Foot , MicroRNAs , Signal Transduction , Wound Healing , Animals , Female , Humans , Male , Rats , Adipokines , Apoptosis/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Curcumin/pharmacology , Diabetic Foot/metabolism , Diabetic Foot/genetics , Diabetic Foot/drug therapy , Diabetic Foot/pathology , Disease Models, Animal , Fibrillin-1/genetics , Fibrillin-1/metabolism , Fibroblasts/metabolism , Fibroblasts/drug effects , MicroRNAs/genetics , MicroRNAs/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/genetics , Wound Healing/drug effects , Wound Healing/genetics
SELECTION OF CITATIONS
SEARCH DETAIL