Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 312
Filter
1.
Hypertension ; 81(6): 1308-1319, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38563153

ABSTRACT

BACKGROUND: Abnormalities of resistance arteries may play essential roles in the pathophysiology of aging and hypertension. Deficiency of the vascular extracellular matrix protein MFAP4 (microfibrillar-associated protein 4) has previously been observed as protective against aberrant arterial remodeling. We hypothesized that MFAP4-deficiency would reduce age- and hypertension-dependent arterial changes in extracellular matrix composition and stiffening. METHODS: Mesenteric arteries were isolated from old (20-23 months) littermate Mfap4+/+ and Mfap4-/- mice, and 2-photon excitation microscopy imaging was used to quantify elastin and collagen volumes and dimensions in the vascular wall. Ten-week-old littermate Mfap4+/+ and Mfap4-/- mice were subjected to 20 days of continuous Ang II (angiotensin II) infusion and hypertension was monitored using invasive blood pressure measurements. Arterial stiffness, responses to vascular constrictors, and myogenic tone were monitored using wire- or pressure-myography. Collagen contents were assessed by Western blotting. RESULTS: MFAP4-deficiency significantly increased collagen volume and elastin fragmentation in aged mesenteric arteries without affecting arterial stiffness. MFAP4-deficient mice exhibited reduced diastolic pressure in Ang II-induced hypertension. There was no significant effect of MFAP4-deficiency on mesenteric artery structural remodeling or myogenic tone, although collagen content in mesenteric arteries was tendentially increased in hypertensive Mfap4+/+ mice relative to Mfap4-/- mice. Increased efficacy of vasoconstrictors (phenylephrine, thromboxane) and reduced stiffness were observed in Ang II-treated Mfap4-/- mouse mesenteric arteries in ex vivo myography recordings. CONCLUSIONS: MFAP4-deficiency reduces the elastin/collagen ratio in the aging resistance artery without affecting arterial stiffness. In contrast, MFAP4-deficiency reduces the stiffness of resistance arteries and ameliorates Ang II-induced hypertension.


Subject(s)
Aging , Angiotensin II , Hypertension , Mesenteric Arteries , Vascular Resistance , Vascular Stiffness , Animals , Hypertension/physiopathology , Hypertension/metabolism , Hypertension/genetics , Mice , Mesenteric Arteries/physiopathology , Mesenteric Arteries/drug effects , Mesenteric Arteries/metabolism , Vascular Stiffness/physiology , Vascular Stiffness/drug effects , Vascular Resistance/physiology , Aging/physiology , Angiotensin II/pharmacology , Elastin/metabolism , Blood Pressure/physiology , Extracellular Matrix Proteins/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/deficiency , Mice, Knockout , Disease Models, Animal , Male , Collagen/metabolism
2.
Proc Inst Mech Eng H ; 236(8): 1106-1117, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35778813

ABSTRACT

Matrix Gla protein (MGP) is mostly known to be a calcification inhibitor, as its absence leads to ectopic calcification of different tissues such as cartilage or arteries. MGP deficiency also leads to low bone mass and delayed bone growth. In the present contribution, we investigate the effect of MGP deficiency on the structural and material mechanical bone properties by focusing on the elastic response of femurs undergoing three-points bending. To this aim, biomechanical tests are performed on femurs issued from Mgp-deficient mice at 14, 21, 28, and 35 days of postnatal life and compared to healthy control femurs. µCT acquisitions enable to reconstruct bone geometries and are used to construct subject-specific finite element models avoiding some of the reported limitations concerning the use of beam-like assumptions for small bone samples. Our results indicate that MGP deficiency may be associated to differences in both structural and material properties of femurs during early stages of development. MGP deficiency appears to be related to a decrease in bone dimensions, compensated by higher material properties resulting in similar structural bone properties at P35. The search for a unique density-elasticity relationship based on calibrated bone mineral density (BMD) indicates that MGP deficiency may affect bone tissue in several ways, that may not be represented uniquely from the quantification of BMD. Despite of its limitation to elastic response, the present preliminary study reports for the very first time the mechanical skeletal properties of Mgp-deficient mice at early stages of development.


Subject(s)
Calcium-Binding Proteins , Extracellular Matrix Proteins , Femur , Animals , Calcium-Binding Proteins/deficiency , Calcium-Binding Proteins/genetics , Cartilage/metabolism , Extracellular Matrix Proteins/deficiency , Extracellular Matrix Proteins/genetics , Femur/diagnostic imaging , Femur/physiopathology , Mice , Matrix Gla Protein
3.
Int J Mol Sci ; 22(21)2021 Oct 26.
Article in English | MEDLINE | ID: mdl-34768980

ABSTRACT

Several studies have shown that type IV fibrocytes, located in the spiral ligament, degenerate first after noise exposure. Interestingly, this is the region where Coch expression is most abundant. As it is suggested that cochlin plays a role in our innate immune system, our goal is to investigate hearing thresholds and inner ear inflammation after noise exposure in Coch knockout (Coch-/-) mice compared to Coch wildtype (Coch+/+) mice. Animals were randomly allocated to a noise exposure group and a control group. Vestibular and auditory testing was performed at 48 h and one week after noise exposure. Whole mount staining and cryosectioning of the cochlea was performed in order to investigate hair cells, spiral ganglion neurons, inner ear inflammation, Coch expression and fibrocyte degeneration. Hearing assessment revealed that Coch+/+ mice had significantly larger threshold shifts than Coch-/- mice after noise exposure. We were unable to identify any differences in hair cells, neurons, fibrocytes and influx of macrophages in the inner ear between both groups. Interestingly, Coch expression was significantly lower in the group exposed to noise. Our results indicate that the absence of Coch has a protective influence on hearing thresholds after noise exposure, but this is not related to reduced inner ear inflammation in the knockout.


Subject(s)
Aging/metabolism , Extracellular Matrix Proteins/deficiency , Hearing Loss, Noise-Induced/metabolism , Animals , Cochlea/metabolism , Ear, Inner/metabolism , Hair Cells, Auditory/metabolism , Hearing/physiology , Inflammation/metabolism , Macrophages/metabolism , Mice , Neurons/metabolism , Noise/adverse effects
4.
J Am Soc Nephrol ; 32(11): 2777-2794, 2021 11.
Article in English | MEDLINE | ID: mdl-34716242

ABSTRACT

BACKGROUND: Autoantibodies binding to podocyte antigens cause idiopathic membranous glomerulonephritis (iMGN). However, it remains elusive how autoantibodies reach the subepithelial space because the glomerular filtration barrier (GFB) is size selective and almost impermeable for antibodies. METHODS: Kidney biopsies from patients with iMGN, cell culture, zebrafish, and mouse models were used to investigate the role of nephronectin (NPNT) regulating microRNAs (miRs) for the GFB. RESULTS: Glomerular endothelial cell (GEC)-derived miR-192-5p and podocyte-derived miR-378a-3p are upregulated in urine and glomeruli of patients with iMGN, whereas glomerular NPNT is reduced. Overexpression of miR-192-5p and morpholino-mediated npnt knockdown induced edema, proteinuria, and podocyte effacement similar to podocyte-derived miR-378a-3p in zebrafish. Structural changes of the glomerular basement membrane (GBM) with increased lucidity, splitting, and lamellation, especially of the lamina rara interna, similar to ultrastructural findings seen in advanced stages of iMGN, were found. IgG-size nanoparticles accumulated in lucidity areas of the lamina rara interna and lamina densa of the GBM in npnt-knockdown zebrafish models. Loss of slit diaphragm proteins and severe structural impairment of the GBM were further confirmed in podocyte-specific Npnt knockout mice. GECs downregulate podocyte NPNT by transfer of miR-192-5p-containing exosomes in a paracrine manner. CONCLUSIONS: Podocyte NPNT is important for proper glomerular filter function and GBM structure and is regulated by GEC-derived miR-192-5p and podocyte-derived miR-378a-3p. We hypothesize that loss of NPNT in the GBM is an important part of the initial pathophysiology of iMGN and enables autoantigenicity of podocyte antigens and subepithelial immune complex deposition in iMGN.


Subject(s)
Endothelial Cells/metabolism , Extracellular Matrix Proteins/biosynthesis , Glomerular Basement Membrane/metabolism , Glomerular Basement Membrane/physiopathology , Glomerulonephritis, Membranous/genetics , Kidney Glomerulus/metabolism , MicroRNAs/physiology , Animals , Antigen-Antibody Complex/analysis , Autoantigens/genetics , Autoantigens/immunology , Cells, Cultured , Coculture Techniques , Exosomes/metabolism , Extracellular Matrix Proteins/deficiency , Extracellular Matrix Proteins/physiology , Gene Expression Regulation , Gene Targeting , Glomerular Basement Membrane/immunology , Glomerular Basement Membrane/ultrastructure , Glomerulonephritis, Membranous/immunology , Glomerulonephritis, Membranous/metabolism , Glomerulonephritis, Membranous/physiopathology , Gold Sodium Thiosulfate , Humans , Metal Nanoparticles , Mice , MicroRNAs/biosynthesis , MicroRNAs/genetics , MicroRNAs/urine , Paracrine Communication , Permeability , Podocytes/immunology , Podocytes/metabolism , Proteinuria/etiology , Transfection , Zebrafish , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics
5.
Tissue Cell ; 73: 101652, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34560406

ABSTRACT

OBJECTIVE: Cellular senescence, associated with aging, leads to impaired tissue regeneration. We hypothesize that vaginal injury initiates cell senescence, further propagated during aging resulting in pelvic organ prolapse (POP). Our objective was to employ a mouse model of POP (Fibulin-5 knockout mice, Fbln5-/-) to determine if vaginal distention leads to cellular senescence and POP. METHODS: 6wk old females [wild-type (WT), n = 81; Fbln5-/-, n = 47)] were assigned to control vs vaginal distention, which approximated vaginal delivery. Serial POP measurements were obtained until vagina were harvested from euthanized mice at 24, 48, 72 h and 1wk. Markers of cell senescence were quantified by immunofluorescence. DNA damage was assessed with γ-H2Ax. RESULTS: WT distended mice showed decreased p53 (p = 0.0230) and γ-H2Ax (p = 0.0008) in vaginal stromal cells at 1wk compared to controls. In WT mice, SA-ß-Gal activity increased 1wk after distention (p = 0.05). In Fbln5-/- mice, p53 and γ-H2Ax did not decrease, but p16 decreased 72 h after distention (p = 0.0150). SA-ß-Gal activity also increased in Fbln5-/-, but at earlier time points and 1wk after distention (p < 0.0001). Fbln5-/- mice developed POP after distention earlier than non distended animals (p = 0.0135). CONCLUSIONS: Vaginal distention downregulates p53 and γ-H2Ax in WT mice, thereby promoting cell proliferation 1wk after injury. This was absent among Fbln5-/- distention mice suggesting they do not escape senescence. These findings indicate a failure of cellular protection from senescence in animals predisposed to POP.


Subject(s)
Cellular Senescence , Pelvic Organ Prolapse/pathology , Vagina/pathology , Animals , Biomarkers/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Disease Models, Animal , Extracellular Matrix Proteins/deficiency , Extracellular Matrix Proteins/metabolism , Female , Mice, Knockout , Phenotype , Recombinant Proteins/metabolism , Stromal Cells/metabolism , Stromal Cells/pathology , beta-Galactosidase/metabolism
6.
J Nutr Biochem ; 98: 108868, 2021 12.
Article in English | MEDLINE | ID: mdl-34563664

ABSTRACT

Disorders in cholesterol and bile acid metabolism have been acknowledged as critical in pathogenesis of hypercholesterolemia. Coiled-coil domain containing 80 (CCDC80) has been closely linked to lipid homeostasis in mice, with its role in cholesterol metabolism yet to be fully elucidated. This study aims to uncover the regulatory mechanisms of CCDC80 in diet-induced hypercholesterolemia. We generated a CCDC80 knockout (CCDC80-/-) model in C57BL/6 mouse. The initial transcriptional and metabolic consequences of removing CCDC80 were accessed at baseline by gene expression microarrays and gas chromatography-mass spectrometry / ultra-high-performance liquid chromatography-quadrupole time-of-flight mass spectrometry, respectively. The hepatic cholesterol was investigated in both CCDC80+/+ and CCDC80-/- male mice at baseline and after feeding a high-cholesterol diet for 12 weeks. The regulatory effects of CCDC80 on gene expressions and protein masses were measured by RT-qPCR and western blot, respectively. At baseline, the KEGG pathway enrichment analysis combining metabolomics, lipidomics and transcriptomics, revealed a down-regulation of hepatic bile acid biosynthesis by CCDC80-knockout, especially for primary bile acids. In the hypercholesterolemic models, our results showed that deficiency of CCDC80 increased plasma and liver cholesterol levels, but decreased fecal neutral and acidic sterols excretion in mice. Mechanistically, we found that such effects were partly mediated by attenuating the alternative pathway of bile acid synthesis catalyzed by oxysterol 7-alpha-hydroxylase (CYP7B1). In conclusion, our results suggest CCDC80 as a novel modulator of cholesterol homeostasis in male mice. Deficiency of CCDC80 could further impair fecal sterols excretion in diet-induced hypercholesterolemia.


Subject(s)
Cholesterol/metabolism , Extracellular Matrix Proteins/metabolism , Feces/chemistry , Hypercholesterolemia/metabolism , Sterols/metabolism , Animals , Bile Acids and Salts/metabolism , Cholesterol/blood , Extracellular Matrix Proteins/deficiency , Extracellular Matrix Proteins/genetics , Gene Expression , Lipid Metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Steroid Hydroxylases/metabolism , Sterols/analysis
7.
Development ; 148(17)2021 09 01.
Article in English | MEDLINE | ID: mdl-34414407

ABSTRACT

Reelin is a large secreted glycoprotein that regulates neuronal migration, lamination and establishment of dendritic architecture in the embryonic brain. Reelin expression switches postnatally from Cajal-Retzius cells to interneurons. However, reelin function in interneuron development is still poorly understood. Here, we have investigated the role of reelin in interneuron development in the postnatal neocortex. To preclude early cortical migration defects caused by reelin deficiency, we employed a conditional reelin knockout (RelncKO) mouse to induce postnatal reelin deficiency. Induced reelin deficiency caused dendritic hypertrophy in distal dendritic segments of neuropeptide Y-positive (NPY+) and calretinin-positive (Calr+) interneurons, and in proximal dendritic segments of parvalbumin-positive (Parv+) interneurons. Chronic recombinant Reelin treatment rescued dendritic hypertrophy in Relncko interneurons. Moreover, we provide evidence that RelncKO interneuron hypertrophy is due to presynaptic GABABR dysfunction. Thus, GABABRs in RelncKO interneurons were unable to block N-type (Cav2.2) Ca2+ channels that control neurotransmitter release. Consequently, the excessive Ca2+ influx through AMPA receptors, but not NMDA receptors, caused interneuron dendritic hypertrophy. These findings suggest that reelin acts as a 'stop-growth-signal' for postnatal interneuron maturation.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Dendrites/metabolism , Extracellular Matrix Proteins/metabolism , Interneurons/cytology , Neocortex/growth & development , Nerve Tissue Proteins/metabolism , Serine Endopeptidases/metabolism , Animals , Calbindin 2/metabolism , Calcium/metabolism , Cell Adhesion Molecules, Neuronal/deficiency , Cell Adhesion Molecules, Neuronal/pharmacology , Dendrites/drug effects , Extracellular Matrix Proteins/deficiency , Extracellular Matrix Proteins/pharmacology , Hypertrophy , Interneurons/drug effects , Interneurons/metabolism , Mice , Mice, Knockout , Neocortex/cytology , Neocortex/drug effects , Neocortex/pathology , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/pharmacology , Neuropeptide Y/metabolism , Parvalbumins/metabolism , Receptors, GABA-B/metabolism , Receptors, Glutamate/metabolism , Reelin Protein , Serine Endopeptidases/deficiency , Serine Endopeptidases/pharmacology
8.
J Cardiovasc Pharmacol ; 78(5): e703-e713, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34369899

ABSTRACT

ABSTRACT: Maturation of fibrillar collagen is known to play a crucial role in the pathophysiology of myocardial fibrosis. Procollagen C-proteinase enhancer 1 (PCPE1) has a key role in procollagen maturation and collagen fibril formation. The phenotype of both male and female PCPE1 knock-out mice was investigated under basal conditions to explore the potential of PCPE1 as a therapeutic target in heart failure. Global constitutive PCPE1-/- mice were generated. Serum procollagen I C-terminal propeptide, organ histology, and cutaneous wound healing were assessed in both wild type (WT) and PCPE1-/- mice. In addition, the cardiac expression of genes involved in collagen metabolism was investigated and the total and insoluble cardiac collagen contents determined. Cardiac function was evaluated by echocardiography. No differences in survival, clinical chemistry, or organ histology were observed in PCPE1-/- mice compared with WT. Serum procollagen I C-terminal propeptide was lower in PCPE1-/- mice. Cardiac mRNA expression of Bmp1, Col1a1, Col3a1, and Loxl2 was similar, whereas Tgfb and Loxl1 mRNA levels were decreased in PCPE1-/- mice compared with sex-matched WT. No modification of total or insoluble cardiac collagen content was observed between the 2 strains. Ejection fraction was slightly decreased in PCPE1-/- male mice, but not in females. Finally, wound healing was not altered in PCPE1-/- mice. PCPE1 deficiency does not trigger any major liabilities and does not affect cardiac collagen content nor its function under basal conditions. Further studies are required to evaluate its role under stressed conditions and determine its suitability as a therapeutic target for heart failure.


Subject(s)
Collagen/metabolism , Extracellular Matrix Proteins/deficiency , Myocardium/metabolism , Amino Acid Oxidoreductases/genetics , Amino Acid Oxidoreductases/metabolism , Animals , Bone Morphogenetic Protein 1/genetics , Bone Morphogenetic Protein 1/metabolism , Collagen/genetics , Collagen Type I, alpha 1 Chain/genetics , Collagen Type I, alpha 1 Chain/metabolism , Collagen Type III/genetics , Collagen Type III/metabolism , Extracellular Matrix Proteins/genetics , Female , Gene Expression Regulation , Genotype , Male , Mice, Inbred C57BL , Mice, Knockout , Peptide Fragments/blood , Phenotype , Procollagen/blood , Stroke Volume , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Ventricular Function, Left , Wound Healing
9.
Biochem Biophys Res Commun ; 571: 14-19, 2021 09 24.
Article in English | MEDLINE | ID: mdl-34298337

ABSTRACT

Restoration of hair follicle (HF) regenerative capacity is the cornerstone in tissue engineering for the loss of regenerative capacity during in vitro expansion of skin-derived precursors (SKPs). Microenvironmental cues facilitated tissue or organ regeneration offers a potential strategy to overcome this difficulty. In our previous work, plantar dermis matrix homogenate (PD) has been proved to induce sweat glands regeneration both in vivo and in vitro. Here, we found PD also restore regenerative capacity of culture impaired HF spheroids (IHFS). Further, followed by our previous iTRAQ results, the CTHRC1 was identified as a potential regulator in PD facilitated restorative effects in HF regeneration. Knockout of Cthrc1 impaired HF regenerative capacity in spheroids, decreased the diameter of HF in 28 postnatal days mice and shortened invagination of HF bud in 18 days of gestation mice. In IHFS and Cthrc1-/- spheroids, PD partially restored HF regenerative capacity while Cthrc1-/- PD (PDKO) has less or no effect. Taken together, PD is an effective microenvironmental cues for HF regenerative capacity restoration and CTHRC1 played an important role in HF regeneration.


Subject(s)
Dermis/metabolism , Extracellular Matrix Proteins/metabolism , Hair Follicle/metabolism , Animals , Extracellular Matrix Proteins/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout
10.
Elife ; 102021 05 26.
Article in English | MEDLINE | ID: mdl-34036937

ABSTRACT

Osteogenesis imperfecta (OI) is characterized by short stature, skeletal deformities, low bone mass, and motor deficits. A subset of OI patients also present with joint hypermobility; however, the role of tendon dysfunction in OI pathogenesis is largely unknown. Using the Crtap-/- mouse model of severe, recessive OI, we found that mutant Achilles and patellar tendons were thinner and weaker with increased collagen cross-links and reduced collagen fibril size at 1- and 4-months compared to wildtype. Patellar tendons from Crtap-/- mice also had altered numbers of CD146+CD200+ and CD146-CD200+ progenitor-like cells at skeletal maturity. RNA-seq analysis of Achilles and patellar tendons from 1-month Crtap-/- mice revealed dysregulation in matrix and tendon marker gene expression concomitant with predicted alterations in TGF-ß, inflammatory, and metabolic signaling. At 4-months, Crtap-/- mice showed increased αSMA, MMP2, and phospho-NFκB staining in the patellar tendon consistent with excess matrix remodeling and tissue inflammation. Finally, a series of behavioral tests showed severe motor impairments and reduced grip strength in 4-month Crtap-/- mice - a phenotype that correlates with the tendon pathology.


Subject(s)
Achilles Tendon/pathology , Extracellular Matrix Proteins/deficiency , Motor Activity , Osteogenesis Imperfecta/pathology , Osteogenesis Imperfecta/physiopathology , Patellar Ligament/pathology , Achilles Tendon/metabolism , Actins/metabolism , Age Factors , Animals , Disease Models, Animal , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Extracellular Matrix Proteins/genetics , Fibrillar Collagens/genetics , Fibrillar Collagens/metabolism , Genes, Recessive , Genetic Predisposition to Disease , Hand Strength , Matrix Metalloproteinase 2/metabolism , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Molecular Chaperones/genetics , NF-kappa B/metabolism , Osteogenesis Imperfecta/genetics , Osteogenesis Imperfecta/metabolism , Patellar Ligament/metabolism , Phenotype , Phosphorylation , Physical Endurance , Stem Cells/metabolism , Stem Cells/pathology
11.
PLoS One ; 16(5): e0250429, 2021.
Article in English | MEDLINE | ID: mdl-34038418

ABSTRACT

Dentin Sialoprotein (DSP) and phosphophoryn (PP) are two most dominant non-collagenous proteins in dentin, which are the cleavage products of the DSPP (dentin sialophosphoprotein) precursor protein. The absence of the DSPP gene in DSPP knock-out (KO) mice results in characteristics that are consistent with dentinogenesis imperfecta type III in humans. Symptoms include thin dentin, bigger pulp chamber with frequent pulp exposure as well as abnormal epithelial-mesenchymal interactions, and the appearance of chondrocyte-like cells in dental pulp. To better understand how DSPP influences tooth development and dentin formation, we used a bacterial artificial chromosome transgene construct (BAC-DSPP) that contained the complete DSPP gene and promoter to generate BAC-DSPP transgenic mice directly in a mouse DSPP KO background. Two BAC-DSPP transgenic mouse strains were generated and characterized. DSPP mRNA expression in BAC-DSPP Strain A incisors was similar to that from wild-type (wt) mice. DSPP mRNA expression in BAC-DSPP Strain B animals was only 10% that of wt mice. PP protein content in Strain A incisors was 25% of that found in wt mice, which was sufficient to completely rescue the DSPP KO defect in mineral density, since microCT dentin mineral density analysis in 21-day postnatal animal molars showed essentially identical mineral density in both strain A and wt mice. Strain B mouse incisors, with 5% PP expression, only partially rescued the DSPP KO defect in mineral density, as microCT scans of 21-day postnatal animal molars indicated a reduced dentin mineral density compared to wt mice, though the mineral density was still increased over that of DSPP KO. Furthermore, our findings showed that DSPP dosage in Strain A was sufficient to rescue the DSPP KO defect in terms of epithelial-mesenchymal interactions, odontoblast lineage maintenance, along with normal dentin thickness and normal mineral density while DSPP gene dosage in Strain B only partially rescued the aforementioned DSPP KO defect.


Subject(s)
Dentin/metabolism , Extracellular Matrix Proteins/genetics , Phosphoproteins/genetics , Sialoglycoproteins/genetics , Tooth/growth & development , Animals , Chromosomes, Artificial, Bacterial/genetics , Collagen Type II , Dentin/diagnostic imaging , Dentin/pathology , Extracellular Matrix Proteins/deficiency , Extracellular Matrix Proteins/metabolism , Incisor/metabolism , Incisor/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Minerals/analysis , Phosphoproteins/deficiency , Phosphoproteins/metabolism , RNA, Messenger/metabolism , Sialoglycoproteins/deficiency , Sialoglycoproteins/metabolism , Tooth/metabolism , X-Ray Microtomography
12.
Mol Cell Neurosci ; 112: 103614, 2021 04.
Article in English | MEDLINE | ID: mdl-33845123

ABSTRACT

Homozygous Dab1 yotari mutant mice, Dab1yot (yot/yot) mice, have an autosomal recessive mutation of Dab1 and show reeler-like phenotype including histological abnormality of the cerebellum, hippocampus, and cerebral cortex. We here show abnormal hippocampal development of yot/yot mice where granule cells and pyramidal cells fail to form orderly rows but are dispersed diffusely in vague multiplicative layers. Possibly due to the positioning failure of granule cells and pyramidal cells and insufficient synaptogenesis, axons of the granule cells did not extend purposefully to connect with neighboring regions in yot/yot mice. We found that both hippocampal granule cells and pyramidal cells of yot/yot mice expressed proteins reactive with the anti-Dab1 antibody. We found that Y198- phosphorylated Dab1 of yot/yot mice was greatly decreased. Accordingly the downstream molecule, Akt was hardly phosphorylated. Especially, synapse formation was defective and the distribution of neurons was scattered in hippocampus of yot/yot mice. Some of neural cell adhesion molecules and hippocampus associated transcription factors of the neurons were expressed aberrantly, suggesting that the Reelin-Dab1 signaling pathway seemed to be importantly involved in not only neural migration as having been shown previously but also neural maturation and/or synaptogenesis of the mice. It is interesting to clarify whether the defective neural maturation is a direct consequence of the dysfunctional Dab1, or alternatively secondarily due to the Reelin-Dab1 intracellular signaling pathways.


Subject(s)
Cell Adhesion Molecules, Neuronal/physiology , Extracellular Matrix Proteins/physiology , Hippocampus/abnormalities , Mice, Mutant Strains/abnormalities , Nerve Tissue Proteins/physiology , Serine Endopeptidases/physiology , Signal Transduction/physiology , Animals , Cell Adhesion Molecules, Neuronal/deficiency , Cell Movement , Enzyme Activation , Extracellular Matrix Proteins/deficiency , Genes, Recessive , Hippocampus/embryology , Hippocampus/metabolism , Hippocampus/pathology , Homozygote , Mice , Mice, Mutant Strains/genetics , Mice, Mutant Strains/metabolism , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neural Cell Adhesion Molecules/biosynthesis , Neural Cell Adhesion Molecules/genetics , Phenotype , Phosphorylation , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/metabolism , Reelin Protein , Serine Endopeptidases/deficiency , Synapses/metabolism , Transcription Factors/biosynthesis , Transcription Factors/genetics
13.
Sci Rep ; 11(1): 4970, 2021 03 02.
Article in English | MEDLINE | ID: mdl-33654143

ABSTRACT

Adipose tissue fibrosis with chronic inflammation is a hallmark of obesity-related metabolic disorders, and the role of proteoglycans in developing adipose tissue fibrosis is of interest. Periodontal disease is associated with obesity; however, the underlying molecular mechanisms remain unclear. Here we investigated the roles of periodontal ligament associated protein-1 (PLAP-1)/asporin, a proteoglycan preferentially and highly expressed in the periodontal ligament, in obesity-related adipose tissue dysfunction and adipocyte differentiation. It was found that PLAP-1 is also highly expressed in white adipose tissues. Plap-1 knock-out mice counteracted obesity and alveolar bone resorption induced by a high-fat diet. Plap-1 knock-down in 3T3-L1 cells resulted in less lipid accumulation, and recombinant PLAP-1 enhanced lipid accumulation in 3T3-L1 cells. In addition, it was found that primary preadipocytes isolated from Plap-1 knock-out mice showed lesser lipid accumulation than the wild-type (WT) mice. Furthermore, the stromal vascular fraction of Plap-1 knock-out mice showed different extracellular matrix gene expression patterns compared to WT. These findings demonstrate that PLAP-1 enhances adipogenesis and could be a key molecule in understanding the association between periodontal disease and obesity-related metabolic disorders.


Subject(s)
Adipose Tissue/metabolism , Alveolar Bone Loss , Diet, High-Fat/adverse effects , Extracellular Matrix Proteins/deficiency , Metabolic Diseases , 3T3-L1 Cells , Alveolar Bone Loss/chemically induced , Alveolar Bone Loss/genetics , Alveolar Bone Loss/metabolism , Animals , Extracellular Matrix Proteins/metabolism , Metabolic Diseases/chemically induced , Metabolic Diseases/genetics , Metabolic Diseases/metabolism , Mice , Mice, Knockout
14.
Arterioscler Thromb Vasc Biol ; 41(4): 1309-1318, 2021 04.
Article in English | MEDLINE | ID: mdl-33626909
15.
Neurochem Int ; 144: 104954, 2021 03.
Article in English | MEDLINE | ID: mdl-33388358

ABSTRACT

Reelin, an extracellular matrix protein, is secreted by Cajal-Retzius cells and plays crucial roles in the development of brain structures and neuronal functions. Reductions in Reelin cause the brain dysfunctions associated with mental disorders, such as schizophrenia. A recent genome-wide copy number variation analysis of Japanese schizophrenia patients identified a novel deletion in RELN encoding Reelin. To clarify the pathophysiological role of the RELN deletion, we developed transgenic mice carrying the RELN deletion (Reln-del) and found abnormalities in their brain structures and social behavior. In the present study, we performed an in vitro analysis of Reelin expression, intracellular Reelin signaling, and the morphology of primary cultured cortical neurons from wild-type (WT) and Reln-del mice. Reelin protein levels were lower in Reln-del neurons than in WT neurons. Dab1 expression levels were significantly higher in Reln-del neurons than in WT neurons, suggesting that Reelin signaling was decreased in Reln-del neurons. Reelin was mainly expressed in γ-aminobutyric acid (GABA)-ergic inhibitory neurons, but not in parvalbumin (PV)-positive neurons. A small proportion of Ca2+/calmodulin-dependent protein kinase II α subunit (CaMKIIα)-positive excitatory neurons also expressed Reelin. In comparisons with WT neurons, significant decreases were observed in neurite lengths and branch points as well as in the number of postsynaptic density protein 95 (PSD95) immunoreactive puncta in Reln-del neurons. A disintegrin and metalloproteinase with thrombospondin motifs-3 (ADAMTS-3) is a protease that inactivates Reelin by cleavage at the N-t site. The knockdown of ADAMTS-3 by short hairpin RNAs suppressed Reelin cleavage in conditioned medium and reduced Dab1 expression, indicating that Reelin signaling was enhanced in the primary cultured cortical neurons of WT and heterozygous Reln-del. Accordingly, the inhibition of ADAMTS-3 may be a potential candidate in the clinical treatment of schizophrenia by enhancing Reelin signaling in the brain.


Subject(s)
Cell Adhesion Molecules, Neuronal/deficiency , Cerebral Cortex/metabolism , Extracellular Matrix Proteins/deficiency , Gene Deletion , Nerve Tissue Proteins/deficiency , Neurons/metabolism , Schizophrenia/metabolism , Serine Endopeptidases/deficiency , Animals , Cell Adhesion Molecules, Neuronal/biosynthesis , Cell Adhesion Molecules, Neuronal/genetics , Cells, Cultured , Cerebral Cortex/cytology , Extracellular Matrix Proteins/biosynthesis , Extracellular Matrix Proteins/genetics , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Reelin Protein , Schizophrenia/genetics , Serine Endopeptidases/biosynthesis , Serine Endopeptidases/genetics , Signal Transduction/physiology
16.
J Neurochem ; 156(5): 589-603, 2021 03.
Article in English | MEDLINE | ID: mdl-32083308

ABSTRACT

Reelin is a protein that is best known for its role in controlling neuronal layer formation in the developing cortex. Here, we studied its role for post-natal cortical network function, which is poorly explored. To preclude early cortical migration defects caused by Reelin deficiency, we used a conditional Reelin knock-out (RelncKO ) mouse, and induced Reelin deficiency post-natally. Induced Reelin deficiency caused hyperexcitability of the neocortical network in vitro and ex vivo. Blocking Reelin binding to its receptors ApoER2 and VLDLR resulted in a similar effect. Hyperexcitability in RelncKO organotypic slice cultures could be rescued by co-culture with wild-type organotypic slice cultures. Moreover, the GABAB receptor (GABAB R) agonist baclofen failed to activate and the antagonist CGP35348 failed to block GABAB Rs in RelncKO mice. Immunolabeling of RelncKO cortical slices revealed a reduction in GABAB R1 and GABAB R2 surface expression at the plasma membrane and western blot of RelncKO cortical tissue revealed decreased phosphorylation of the GABAB R2 subunit at serine 892 and increased phosphorylation at serine 783, reflecting receptor deactivation and proteolysis. These data show a role of Reelin in controlling early network activity, by modulating GABAB R function. Cover Image for this issue: https://doi.org/10.1111/jnc.15054.


Subject(s)
Cell Adhesion Molecules, Neuronal/deficiency , Extracellular Matrix Proteins/deficiency , Neocortex/metabolism , Nerve Tissue Proteins/deficiency , Receptors, GABA-B/physiology , Serine Endopeptidases/deficiency , Signal Transduction/physiology , Animals , Animals, Newborn , Cell Adhesion Molecules, Neuronal/genetics , Extracellular Matrix Proteins/genetics , Female , GABA-B Receptor Agonists/pharmacology , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Organ Culture Techniques , Reelin Protein , Serine Endopeptidases/genetics , Signal Transduction/drug effects
17.
Nature ; 588(7839): 705-711, 2020 12.
Article in English | MEDLINE | ID: mdl-33299187

ABSTRACT

Recent studies have suggested that lymphatics help to restore heart function after cardiac injury1-6. Here we report that lymphatics promote cardiac growth, repair and cardioprotection in mice. We show that a lymphoangiocrine signal produced by lymphatic endothelial cells (LECs) controls the proliferation and survival of cardiomyocytes during heart development, improves neonatal cardiac regeneration and is cardioprotective after myocardial infarction. Embryos that lack LECs develop smaller hearts as a consequence of reduced cardiomyocyte proliferation and increased cardiomyocyte apoptosis. Culturing primary mouse cardiomyocytes in LEC-conditioned medium increases cardiomyocyte proliferation and survival, which indicates that LECs produce lymphoangiocrine signals that control cardiomyocyte homeostasis. Characterization of the LEC secretome identified the extracellular protein reelin (RELN) as a key component of this process. Moreover, we report that LEC-specific Reln-null mouse embryos develop smaller hearts, that RELN is required for efficient heart repair and function after neonatal myocardial infarction, and that cardiac delivery of RELN using collagen patches improves heart function in adult mice after myocardial infarction by a cardioprotective effect. These results highlight a lymphoangiocrine role of LECs during cardiac development and injury response, and identify RELN as an important mediator of this function.


Subject(s)
Heart/embryology , Lymphatic System/cytology , Lymphatic System/metabolism , Myocardium/cytology , Myocytes, Cardiac/cytology , Regeneration , Signal Transduction , Animals , Animals, Newborn , Apoptosis , Cell Adhesion Molecules, Neuronal/deficiency , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Cell Proliferation , Cell Survival , Cells, Cultured , Endothelial Cells/metabolism , Extracellular Matrix Proteins/deficiency , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Female , Humans , Integrin beta1/metabolism , Mice , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocytes, Cardiac/metabolism , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Organ Size , Organogenesis , Reelin Protein , Serine Endopeptidases/deficiency , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism
18.
J Dermatol Sci ; 100(2): 99-109, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33046330

ABSTRACT

BACKGROUND: Lichen sclerosus (LS) is an acquired inflammatory mucocutaneous disease affecting the anogenital area, characterized histologically by hyalinosis and thickened vessel walls in the dermis. The presence of serum autoantibodies against extracellular matrix protein 1 (ECM1) in LS patients may suggest its involvement in disease pathogenesis. OBJECTIVE: To examine if reduced ECM1 production by dermal fibroblasts contributes to the pathogenic features of LS. METHODS: Gene expression in ECM1 knockdown human dermal fibroblasts was analyzed by cDNA microarray. Functional enrichment for genes involved in cellular functions was conducted. Protein expression was analyzed by ELISA and confocal laser scanning microscopy using LS skin. RESULTS: Microarray analysis identified 3035 differentially expressed genes in ECM1 knockdown cells, wherein 1471 were upregulated genes related exclusively to cell adhesion, proliferation, apoptosis, intracellular signaling, and extracellular matrix organization. Further narrowing with criteria specific for localization and function of ECM1 identified 48 upregulated genes identified to have structural, fibrogenic, and carcinogenic properties. Of these, laminin-332 and collagen-IV displayed altered immunolabeling within the basement membrane zone (BMZ) and dermal vessels in LS skin, similar to that of collagen-VII, which exhibited unchanged transcription levels in ECM1-knockdown fibroblasts. Collagen-VII bound to recombinant ECM1 in a solid-phase immunoassay and colocalized with ECM1 in the skin BMZ. Further, ECM1-knockdown fibroblasts exhibited a marked delay in cell migration and gel contraction. CONCLUSION: In the absence of ECM1 expression in fibroblasts there is selective dysregulation and disassembly of structural and extracellular matrix molecules, which may result in microstructural abnormalities reminiscent of LS.


Subject(s)
Extracellular Matrix Proteins/deficiency , Gene Expression Regulation/immunology , Lichen Sclerosus et Atrophicus/genetics , Adult , Aged , Aged, 80 and over , Cells, Cultured , Extracellular Matrix/immunology , Extracellular Matrix/pathology , Extracellular Matrix Proteins/genetics , Female , Fibroblasts/cytology , Fibroblasts/immunology , Fibroblasts/pathology , Gene Expression Profiling , Gene Knockdown Techniques , Humans , Lichen Sclerosus et Atrophicus/immunology , Lichen Sclerosus et Atrophicus/pathology , Middle Aged , Oligonucleotide Array Sequence Analysis , Primary Cell Culture , RNA Interference , Skin/cytology , Skin/immunology , Skin/pathology
19.
J Histochem Cytochem ; 68(10): 703-718, 2020 10.
Article in English | MEDLINE | ID: mdl-32921220

ABSTRACT

Dentin sialophosphoprotein (DSPP), which expresses and synthesizes in odontoblasts of dental pulp, is a critical protein for normal teeth mineralization. Originally, DSPP was identified as a dentin-specific protein. In 2010, DSPP was also found in femoral head cartilage, and it is still unclear what roles DSPP play in femoral head cartilage formation, growth, and maintenance. To reveal biological functions of DSPP in the femoral head cartilage, we examined Dspp null mice compared with wild-type (WT) mice to observe DSPP expression as well as localization in WT mice and to uncover differences of femoral head cartilage, bone morphology, and structure between these two kinds of mice. Expression data demonstrated that DSPP had heterogeneous fragments, expressed in each layer of femoral head cartilage and subchondral bone of WT mice. Dspp null mice exhibited a significant reduction in the thickness of femoral head cartilage, with decreases in the amount of proliferating cartilage cells and increases in apoptotic cells. In addition, the subchondral bone mineralization decreased, and the expressions of vessel markers (vascular endothelial growth factor [VEGF] and CD31), osteoblast markers (Osterix and dentin matrix protein 1 [DMP1]), osteocyte marker (sclerostin [SOST]), and osteoclast marker (tartrate-resistant acid phosphatase [TRAP]) were remarkably altered. These indicate that DSPP deletion can affect the proliferation of cartilage cells in the femoral head cartilage and endochondral ossification in subchondral bone. Our data clearly demonstrate that DSPP plays essential roles in the femoral head cartilage growth and maintenance and subchondral biomineralization.


Subject(s)
Calcification, Physiologic , Cartilage/metabolism , Extracellular Matrix Proteins/metabolism , Femur Head/metabolism , Phosphoproteins/metabolism , Sialoglycoproteins/metabolism , Animals , Cartilage/cytology , Cell Proliferation , Extracellular Matrix Proteins/deficiency , Extracellular Matrix Proteins/isolation & purification , Femur Head/cytology , Mice , Mice, Knockout , Phosphoproteins/deficiency , Phosphoproteins/isolation & purification , Sialoglycoproteins/deficiency , Sialoglycoproteins/isolation & purification
20.
J Mol Med (Berl) ; 98(11): 1639-1656, 2020 11.
Article in English | MEDLINE | ID: mdl-32964303

ABSTRACT

Fibulin-3 (F3) is an extracellular matrix glycoprotein found in basement membranes across the body. An autosomal dominant R345W mutation in F3 causes a macular dystrophy resembling dry age-related macular degeneration (AMD), whereas genetic removal of wild-type (WT) F3 protects mice from sub-retinal pigment epithelium (RPE) deposit formation. These observations suggest that F3 is a protein which can regulate pathogenic sub-RPE deposit formation in the eye. Yet the precise role of WT F3 within the eye is still largely unknown. We found that F3 is expressed throughout the mouse eye (cornea, trabecular meshwork (TM) ring, neural retina, RPE/choroid, and optic nerve). We next performed a thorough structural and functional characterization of each of these tissues in WT and homozygous (F3-/-) knockout mice. The corneal stroma in F3-/- mice progressively thins beginning at 2 months, and the development of corneal opacity and vascularization starts at 9 months, which worsens with age. However, in all other tissues (TM, neural retina, RPE, and optic nerve), gross structural anatomy and functionality were similar across WT and F3-/- mice when evaluated using SD-OCT, histological analyses, electron microscopy, scotopic electroretinogram, optokinetic response, and axonal anterograde transport. The lack of noticeable retinal abnormalities in F3-/- mice was confirmed in a human patient with biallelic loss-of-function mutations in F3. These data suggest that (i) F3 is important for maintaining the structural integrity of the cornea, (ii) absence of F3 does not affect the structure or function of any other ocular tissue in which it is expressed, and (iii) targeted silencing of F3 in the retina and/or RPE will likely be well-tolerated, serving as a safe therapeutic strategy for reducing sub-RPE deposit formation in disease. KEY MESSAGES: • Fibulins are expressed throughout the body at varying levels. • Fibulin-3 has a tissue-specific pattern of expression within the eye. • Lack of fibulin-3 leads to structural deformities in the cornea. • The retina and RPE remain structurally and functionally healthy in the absence of fibulin-3 in both mice and humans.


Subject(s)
Cornea/metabolism , Extracellular Matrix Proteins/deficiency , Retina/metabolism , Animals , Biomarkers , Cornea/pathology , Disease Susceptibility , Gene Expression , Genotype , Humans , Macular Degeneration/etiology , Macular Degeneration/metabolism , Macular Degeneration/pathology , Mice , Mice, Knockout , Mutation , Organ Specificity/genetics , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...