Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Int J Mol Sci ; 22(22)2021 Nov 17.
Article in English | MEDLINE | ID: mdl-34830288

ABSTRACT

Considerable evidence accumulated over the past decade supports that telocytes (TCs)/CD34+ stromal cells represent an exclusive type of interstitial cells identifiable by transmission electron microscopy (TEM) or immunohistochemistry in various organs of the human body, including the skin. By means of their characteristic cellular extensions (telopodes), dermal TCs are arranged in networks intermingled with a multitude of neighboring cells and, hence, they are thought to contribute to skin homeostasis through both intercellular contacts and releasing extracellular vesicles. In this context, fibrotic skin lesions from patients with systemic sclerosis (SSc, scleroderma) appear to be characterized by a disruption of the dermal network of TCs, which has been ascribed to either cell degenerative processes or possible transformation into profibrotic myofibroblasts. In the present study, we utilized the well-established mouse model of bleomycin-induced scleroderma to gain further insights into the TC alterations found in cutaneous fibrosis. CD34 immunofluorescence revealed a severe impairment in the dermal network of TCs/CD34+ stromal cells in bleomycin-treated mice. CD31/CD34 double immunofluorescence confirmed that CD31-/CD34+ TC counts were greatly reduced in the skin of bleomycin-treated mice compared with control mice. Ultrastructural signs of TC injury were detected in the skin of bleomycin-treated mice by TEM. The analyses of skin samples from mice treated with bleomycin for different times by either TEM or double immunostaining and immunoblotting for the CD34/α-SMA antigens collectively suggested that, although a few TCs may transition to α-SMA+ myofibroblasts in the early disease stage, most of these cells rather undergo degeneration, and then are lost. Taken together, our data demonstrate that TC changes in the skin of bleomycin-treated mice mimic very closely those observed in human SSc skin, which makes this experimental model a suitable tool to (i) unravel the pathological mechanisms underlying TC damage and (ii) clarify the possible contribution of the TC loss to the development/progression of dermal fibrosis. In perspective, these findings may have important implications in the field of skin regenerative medicine.


Subject(s)
Antigens, CD34/metabolism , Bleomycin/adverse effects , Scleroderma, Systemic/chemically induced , Scleroderma, Systemic/metabolism , Skin/pathology , Telocytes/metabolism , Actins/metabolism , Animals , Cell Count , Disease Models, Animal , Fibrosis , Fluorescent Antibody Technique/methods , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission/methods , Myofibroblasts/metabolism , Myofibroblasts/ultrastructure , Skin/ultrastructure , Telocytes/ultrastructure
2.
Exp Eye Res ; 204: 108447, 2021 03.
Article in English | MEDLINE | ID: mdl-33465394

ABSTRACT

We previously found that epigallocatechin-3-gallate (EGCG) could inhibit the myofibroblast transformation of human Tenon's fibroblasts, however, the underlying mechanism remained unclear. We therefore investigated whether the autophagic regulation involved in the anti-fibrotic function of EGCG. The fibroblasts were subjected to transforming growth factor beta-1 (TGF-ß1) induction followed by EGCG treatments. The autophagic flux was examined by transmission electron microscopy and autophagic flux analysis. The levels of autophagy-related proteins (LC3ß and p62) and alpha-smooth muscle actin (α-SMA) were measured by Western blot and immunofluorescence. Results showed that TGF-ß1 partially inhibited the autophagic function of Tenon's fibroblasts. But this inhibition effect was rescued by LY2157299, a TGF-ßR1 selective inhibitor. Compared with the cells treated with TGF-ß1 alone, EGCG treatments increased the amount of autophagosomes and autolysosomes, evaluated the ratio of LC3-II to LC3-I and decreased p62 level. Our results indicated that EGCG could recover the activity of autophagy in the TGF-ß1-treated cells. Moreover, treatments with EGCG significantly decreased the α-SMA expression. Taken together, these findings revealed that autophagic regulation involved in the action of EGCG against TGF-ß1-induced transformation of Tenon's fibroblasts. Through increasing intracellular autophagy, EGCG could be a potential anti-fibrotic reagent for preventing subconjunctival fibrosis after glaucoma filtration surgery.


Subject(s)
Antioxidants/pharmacology , Autophagy/drug effects , Catechin/analogs & derivatives , Myofibroblasts/drug effects , Tenon Capsule/drug effects , Transforming Growth Factor beta1/pharmacology , Actins/metabolism , Adenoviridae/genetics , Blotting, Western , Catechin/pharmacology , Cell Transdifferentiation/drug effects , Cells, Cultured , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Microtubule-Associated Proteins/metabolism , Myofibroblasts/metabolism , Myofibroblasts/ultrastructure , Sequestosome-1 Protein/metabolism , Tenon Capsule/metabolism , Tenon Capsule/ultrastructure , Transfection , Transforming Growth Factor beta1/antagonists & inhibitors
3.
Biomed Pharmacother ; 134: 111135, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33352448

ABSTRACT

Currently, the clinical treatment of preterm birth, mainly using uterine contraction inhibitors, does not fundamentally reduce the incidence of premature birth (PTB). Premature cervical ripening is an important factor in PTB. We previously found that nicotine-treated pregnant murine had significant cervical resistance to stretch and higher collagen cross-links compared to the control animals, and nicotine prolonged gestation and inhibited cervical ripening. However, the regulatory effects of nicotine on premature cervical ripening and its role in PTB remain unclear. To investigate the effects of nicotine on cervical TGF-ß1/Smad3 pathway and fibroblast-myofibroblast differentiation regulated by this pathway in PTB-like models. Intraperitoneal injection with 15 µg lipopolysaccharide (LPS) in 200 µl PBS into pregnant mice was used to induce the PTB-like model. Mice were randomly divided into four groups: control group, LPS-treated group, LPS + Nicotine co-treated group and LPS + Nicotine+α-BGT co-treated group. Pregnancy outcomes were monitored. The collagen content was assessed by Picrosirius red staining. Expressions of genes and proteins in the TGF-ß/Smad3 pathway were detected by double immunofluorescence staining and quantitative Real-time PCR (qRT-PCR). myofibroblast differentiation were investigated by double immunofluorescence staining and qRT-PCR. Ultrastructures were analyzed by conventional transmission electron microscopy. The rate of PTB and neonatal mortality at birth was significantly higher in the LPS-treated group than in the control group; collagen content also decreased remarkably; the expression of TGF-ß1 in macrophages and p-Smad3 in fibroblasts were reduced; the expression of α-smooth muscle actin (α-SMA, markers for activated fibroblasts) was down-regulated while the expression of calponin and smoothelin (markers for fibroblasts at rest) was up-regulated. Nicotine improved pregnancy outcomes and inhibited collagen degradation, activated the TGF-ß1/Smad3 pathway and promoted cervical fibroblast-myofibroblast differentiation in PTB-like mice; such effects could be reversed by α-bungarotoxin (α-BGT). Nicotine inhibited premature cervical ripening in PTB-like models in relation with up-regulating the TGF-ß/Smad3 pathway and promoting fibroblast to differentiate into myofibroblasts.


Subject(s)
Cell Differentiation/drug effects , Cervical Ripening/drug effects , Cervix Uteri/drug effects , Myofibroblasts/drug effects , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Premature Birth/prevention & control , Smad3 Protein/metabolism , Transforming Growth Factor beta1/metabolism , Actins/metabolism , Animals , Cervix Uteri/metabolism , Cervix Uteri/ultrastructure , Collagen/metabolism , Disease Models, Animal , Female , Lipopolysaccharides , Mice, Inbred C57BL , Myofibroblasts/metabolism , Myofibroblasts/ultrastructure , Phosphorylation , Pregnancy , Premature Birth/chemically induced , Premature Birth/metabolism , Premature Birth/pathology , Proteolysis , Signal Transduction
4.
Am J Physiol Renal Physiol ; 320(1): F17-F30, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33196322

ABSTRACT

Connexins (Cxs) form gap junctions for intercellular exchange of inorganic ions and messenger molecules. In the kidney, Cxs play essential roles within its compartments, but data on the precise cellular localization and cell type-related function of their isoforms are scarce. We tested whether Cx43 distribution is restricted to vascular and interstitial cells and whether medullary fibroblasts express Cx43 to coordinate profibrotic signaling. Confocal immunofluorescence techniques, ultrastructural labeling, and functional experiments in cell culture were performed. Cx43 was chiefly expressed in the vasculature but was absent from tubular epithelia. All arterial, arteriolar, and lymphatic endothelia showed continuous Cx43 signal along their borders. In the inner medulla, only the interstitium showed Cx43 signals, which were assigned to fibroblasts and their processes. Cultured Cx43-expressing medullary fibroblasts served to study the role of gap junctions in a profibrotic context. In a dye spreading assay, Cx43-sensitive diffusion of Lucifer yellow was dependent on gap junctional passage. The addition of transforming growth factor-ß1 (5 ng/mL for 48 h) activated Cx43 biosynthesis and caused Cx43-sensitive transformation of the fibroblasts into a myofibroblast phenotype. This suggested that Cx43 gap junctional channels enable the coordination of profibrotic signaling between cells of the medullary interstitium. In summary, we demonstrate the presence of Cx43-expressing gap junctions within the two major renal compartments, the vasculature and interstitium. Endothelial Cx43 likely provides functions of an earlier-defined "electrical syncytium" within the vascular wall. Additionally, Cx43 facilitates profibrotic signaling between medullary interstitial fibroblasts.


Subject(s)
Cell Differentiation , Connexin 43/metabolism , Endothelial Cells/metabolism , Fibroblasts/metabolism , Kidney Medulla/blood supply , Kidney Medulla/metabolism , Animals , Cell Communication , Cell Line , Endothelial Cells/ultrastructure , Fibroblasts/ultrastructure , Fibrosis , Gap Junctions/metabolism , Gap Junctions/ultrastructure , Humans , Kidney Medulla/ultrastructure , Male , Mice, Inbred C57BL , Myofibroblasts/metabolism , Myofibroblasts/ultrastructure , Phenotype , Rats, Wistar
5.
Cell ; 182(3): 545-562.e23, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32621799

ABSTRACT

Scar tissue size following myocardial infarction is an independent predictor of cardiovascular outcomes, yet little is known about factors regulating scar size. We demonstrate that collagen V, a minor constituent of heart scars, regulates the size of heart scars after ischemic injury. Depletion of collagen V led to a paradoxical increase in post-infarction scar size with worsening of heart function. A systems genetics approach across 100 in-bred strains of mice demonstrated that collagen V is a critical driver of postinjury heart function. We show that collagen V deficiency alters the mechanical properties of scar tissue, and altered reciprocal feedback between matrix and cells induces expression of mechanosensitive integrins that drive fibroblast activation and increase scar size. Cilengitide, an inhibitor of specific integrins, rescues the phenotype of increased post-injury scarring in collagen-V-deficient mice. These observations demonstrate that collagen V regulates scar size in an integrin-dependent manner.


Subject(s)
Cicatrix/metabolism , Collagen Type V/deficiency , Collagen Type V/metabolism , Heart Injuries/metabolism , Myocardial Contraction/genetics , Myofibroblasts/metabolism , Animals , Cicatrix/genetics , Cicatrix/physiopathology , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Collagen Type III/genetics , Collagen Type III/metabolism , Collagen Type V/genetics , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Female , Fibrosis/genetics , Fibrosis/metabolism , Gene Expression Regulation/genetics , Integrins/antagonists & inhibitors , Integrins/genetics , Integrins/metabolism , Isoproterenol/pharmacology , Male , Mechanotransduction, Cellular/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Atomic Force/instrumentation , Microscopy, Electron, Transmission , Myocardial Contraction/drug effects , Myofibroblasts/cytology , Myofibroblasts/pathology , Myofibroblasts/ultrastructure , Principal Component Analysis , Proteomics , RNA-Seq , Single-Cell Analysis
6.
Proc Natl Acad Sci U S A ; 117(21): 11387-11398, 2020 05 26.
Article in English | MEDLINE | ID: mdl-32385149

ABSTRACT

Altered microarchitecture of collagen type I is a hallmark of wound healing and cancer that is commonly attributed to myofibroblasts. However, it remains unknown which effect collagen microarchitecture has on myofibroblast differentiation. Here, we combined experimental and computational approaches to investigate the hypothesis that the microarchitecture of fibrillar collagen networks mechanically regulates myofibroblast differentiation of adipose stromal cells (ASCs) independent of bulk stiffness. Collagen gels with controlled fiber thickness and pore size were microfabricated by adjusting the gelation temperature while keeping their concentration constant. Rheological characterization and simulation data indicated that networks with thicker fibers and larger pores exhibited increased strain-stiffening relative to networks with thinner fibers and smaller pores. Accordingly, ASCs cultured in scaffolds with thicker fibers were more contractile, expressed myofibroblast markers, and deposited more extended fibronectin fibers. Consistent with elevated myofibroblast differentiation, ASCs in scaffolds with thicker fibers exhibited a more proangiogenic phenotype that promoted endothelial sprouting in a contractility-dependent manner. Our findings suggest that changes of collagen microarchitecture regulate myofibroblast differentiation and fibrosis independent of collagen quantity and bulk stiffness by locally modulating cellular mechanosignaling. These findings have implications for regenerative medicine and anticancer treatments.


Subject(s)
Collagen/ultrastructure , Myofibroblasts/cytology , Stromal Cells/cytology , Adipose Tissue/cytology , Biomechanical Phenomena , Cell Differentiation , Cells, Cultured , Collagen/metabolism , Extracellular Matrix/ultrastructure , Fibronectins/metabolism , Humans , Mechanotransduction, Cellular , Myofibroblasts/metabolism , Myofibroblasts/ultrastructure , Stromal Cells/metabolism , Stromal Cells/ultrastructure
7.
Acta Histochem ; 122(4): 151536, 2020 May.
Article in English | MEDLINE | ID: mdl-32156483

ABSTRACT

The lacrimal sac (LS) empties in the nasolacrimal duct to drain the tears in the inferior nasal meatus. Different studies indicated the role of the lacrimal pump in the lacrimal drainage. Although controversial, the lacrimal pump mechanism is an extrinsic one, either active, or passive. An intrinsic contractile potential of the LS was not documented previously. We thus aimed a retrospective immunohistochemical study to test the alpha-smooth muscle actin (α-SMA) and h-caldesmon expression in the LS wall. We used archived paraffin-embedded samples of LS from ten adult patients. The α-SMA + phenotype was detected in basal epithelial cells, in subepithelial ribbons of stromal cells, in vascular smooth muscle cells, as well as in pericytes. H-caldesmon was exclusively expressed in pericytes, vascular smooth muscle cells and myoepithelial cells of the subepithelial glands. The most striking feature we found in all samples was a consistent stromal network of α-SMA+/h-caldesmon- myofibroblasts. This finding supports an intrinsic scaffold useful for the lacrimal pump.


Subject(s)
Lacrimal Apparatus/physiology , Myofibroblasts/physiology , Actins/biosynthesis , Calmodulin-Binding Proteins/biosynthesis , Epithelial Cells/metabolism , Female , Humans , Lacrimal Apparatus/anatomy & histology , Lacrimal Apparatus/cytology , Male , Middle Aged , Muscle Contraction/physiology , Myocytes, Smooth Muscle/metabolism , Myofibroblasts/ultrastructure , Pericytes/metabolism , Retrospective Studies
8.
Neurology ; 94(5): e474-e480, 2020 02 04.
Article in English | MEDLINE | ID: mdl-31892634

ABSTRACT

OBJECTIVE: Varicella zoster virus (VZV) can spread anterogradely and infect cerebral arteries causing VZV vasculopathy and arterial ischemic stroke. In this study, we tested the hypothesis that virus-infected cerebrovascular fibroblasts undergo phenotypic changes that promote vascular remodeling and facilitate virus transmission in an in vitro model of VZV vasculopathy. The aims of this project were therefore to examine the changes that virus-infected human brain adventitial vascular fibroblasts (HBVAFs) undergo in an in vitro model of VZV vasculopathy and to identify disease biomarkers relating to VZV-related vasculopathy. METHODS: HBVAFs were infected with VZV, and their ability to migrate, proliferate, transdifferentiate, and interact with endothelial cells was studied with flow cytometry. Microparticles (MPs) released from these cells were isolated and imaged with transmission electron microscopy, and their protein content was analyzed with mass spectrometry. Circulating MP profiles were also studied in children with VZV and non-VZV vasculopathy and compared with controls. RESULTS: VZV-infected HBVAFs transdifferentiated into myofibroblasts with enhanced proliferative and migratory capacity. Interaction of VZV-infected HBVAFs with endothelial cells resulted in endothelial dysfunction. These effects were, in part, mediated by the release of MPs from VZV-infected HBVAFs. These MPs contained VZV virions that could transmit VZV to neighboring cells, highlighting a novel model of VZV cell-to-cell viral dissemination. MPs positive for VZV were significantly higher in children with VZV-related vasculopathy compared to children with non-VZV vasculopathy (p = 0.01) and controls (p = 0.007). CONCLUSIONS: VZV-infected HBVAFs promote vascular remodeling and facilitate virus transmission. These effects were mediated by the release of apoptotic MPs that could transmit VZV infection to neighboring cells through a Trojan horse means of productive viral infection. VZV+ MPs may represent a disease biomarker worthy of further study.


Subject(s)
Cell Movement , Cell Proliferation , Cell Transdifferentiation , Cell-Derived Microparticles/virology , Cerebrovascular Disorders/virology , Fibroblasts/virology , Myofibroblasts/virology , Vascular Remodeling , Adolescent , Adventitia , Cell-Derived Microparticles/ultrastructure , Cerebral Arteries , Cerebrovascular Disorders/physiopathology , Child , Child, Preschool , Endothelial Cells , Female , Fibroblasts/physiology , Fibroblasts/ultrastructure , Herpesvirus 3, Human , Humans , In Vitro Techniques , Male , Mass Spectrometry , Microscopy, Electron, Transmission , Myofibroblasts/physiology , Myofibroblasts/ultrastructure , Stroke/physiopathology , Stroke/virology , Virus Cultivation
9.
Biosci Rep ; 40(1)2020 01 31.
Article in English | MEDLINE | ID: mdl-31894858

ABSTRACT

Hypertrophic scars (HTSs) are common and cause functional and psychological morbidity. UVA1 (340-400 nm) phototherapy has been previously shown to be effective in the treatment of localized scleroderma, systemic sclerosis, and POEMS syndrome with minimal side effects, all of which are presented as collagen fibrils hyperplasia that is common with scarring in skin histology. In the present study, we aimed to investigate the impact of UVA1 on the protein expression of TGF-ß signal pathway and myofibroblasts in a rabbit model of cutaneous scarring. Full-thickness skin wounds (2 cm × 5 cm in diameter) were made in New Zealand white rabbits to establish the hypertrophic scarring model. New Zealand white rabbits were divided into two treatment groups (n=30 wounds per group with an equal number of controls): medium-dose of UVA1 phototherapy group: 60 J/cm2; high-dose of UVA1 phototherapy group: 110 J/cm2. Left ears were used for treatment and the right ones were used for control. Treatment was administered five times weekly for 6 weeks. Treated and untreated control wounds were harvested at various time points and examined by histologic examination, immunohistochemical assessment, and ultrastructural evaluation. The results showed that UVA1 phototherapy caused a significant reduction in dermal thickness by histological features, whereas the scar index was descended significantly in both medium- and high-dose UVA1 groups compared with the control group. Examination of immunohistochemistry also revealed a marked suppression of tissue growth factor-ß (TGF-ß) (both medium- and high-dose), α smooth muscle actin (α-SMA) (only high-dose), and tissue inhibitor of metalloproteinase 1 (TIMP-1) (only high-dose), and apparent increase in matrix metalloproteinases (MMP-1) (both medium- and high-dose) compared with the control. The ultrastructural evaluation showed the collagen fibers' diameter had shrunk, and that fibroblastic cytoplasm was not affluent and in a quiescent stage. These findings of the present study suggested that administration of UVA1 irradiation is effective to improve the experimental HTS model and raises a possibility of the therapeutic approach of UVA1 in the scar. Although not directly examined in the present study, MMP inhibition is hypothesized to be responsible for this effect. However, early UVA1 treatment could not prevent the formation of scar model.


Subject(s)
Cicatrix, Hypertrophic/radiotherapy , Myofibroblasts/radiation effects , Skin/radiation effects , Ultraviolet Therapy , Actins/metabolism , Animals , Cicatrix, Hypertrophic/metabolism , Cicatrix, Hypertrophic/pathology , Collagen/metabolism , Disease Models, Animal , Female , Matrix Metalloproteinase 1/metabolism , Myofibroblasts/metabolism , Myofibroblasts/ultrastructure , Rabbits , Skin/metabolism , Skin/ultrastructure , Tissue Inhibitor of Metalloproteinase-1/metabolism , Transforming Growth Factor beta/metabolism
10.
Med Mol Morphol ; 53(2): 86-93, 2020 Jun.
Article in English | MEDLINE | ID: mdl-31559505

ABSTRACT

Fibroblasts and myofibroblasts have been known to be present in both ruptured and intact human anterior cruciate ligament (ACL), and although their relevant histology and immunochemistry have been studied in the past, ultrastructural features of these cells are largely lacking. Therefore, we aim to characterise the ultrastructural details of these cells with the help of transmission electron microscopy (TEM) and to study the changes and their significance with duration of injury. Samples from 60 ruptured human ACL undergoing surgery were obtained and categorised according to duration of injury and observed under TEM with main focus on the following ultrastructural features: cellular morphology, presence of rough endoplasmic reticulum, Golgi apparatus, lamina, myofilaments, and presence of myofibroblasts. These features were further correlated with the duration of injury and association, if any, determined using appropriate statistical analysis. A total of 54 male and 6 female patients with mean duration of the injury of 23.01 ± 26.09 weeks (2-108 weeks) were included in the study and categorised into five groups based on duration of injury as follows: I (< 6 weeks), II (7-12 weeks), III (13-20 weeks), IV (21-50 weeks) and V (> 50 weeks). There was a significant association between the above-mentioned ultrastructural features and the duration of injury (p < 0.05) except for the presence of ovoid fibroblast cells (p = 0.53). Furthermore, number of myofibroblasts and cells with Golgi apparatus and rough endoplasmic reticulum was seen to peak at 13-20 weeks following injury. We describe ultrastructural features of fibroblast of different morphology along with myofibroblasts in the ligaments following injury, the changes in which might have a potential bearing on ligament healing.


Subject(s)
Anterior Cruciate Ligament Injuries/pathology , Anterior Cruciate Ligament/ultrastructure , Tibia/ultrastructure , Adolescent , Adult , Anterior Cruciate Ligament/cytology , Anterior Cruciate Ligament/pathology , Anterior Cruciate Ligament/surgery , Anterior Cruciate Ligament Injuries/surgery , Arthroscopy , Endoplasmic Reticulum, Rough/ultrastructure , Female , Golgi Apparatus/ultrastructure , Humans , Male , Microscopy, Electron, Transmission , Myofibroblasts/cytology , Myofibroblasts/ultrastructure , Prospective Studies , Tibia/cytology , Tibia/pathology , Tibia/surgery , Time Factors , Young Adult
11.
Gene Expr ; 20(1): 25-37, 2020 06 12.
Article in English | MEDLINE | ID: mdl-31757226

ABSTRACT

Hepatic stellate cells (HSC) are critical effector cells of liver fibrosis. In the injured liver, HSC differentiate into a myofibrobastic phenotype. A critical feature distinguishing myofibroblastic from quiescent HSC is cytoskeletal reorganization. Soluble NSF attachment receptor (SNARE) proteins are important in trafficking of newly synthesized proteins to the plasma membrane for release into the extracellular environment. The goals of this project were to determine the expression of specific SNARE proteins in myofibroblastic HSC and to test whether their alteration changed the HSC phenotype in vitro and progression of liver fibrosis in vivo. We found that HSC lack the t-SNARE protein, SNAP-25, but express a homologous protein, SNAP-23. Downregulation of SNAP-23 in HSC induced reduction in polymerization and disorganization of the actin cytoskeleton associated with loss of cell movement. In contrast, reduction in SNAP-23 in mice by monogenic deletion delayed but did not prevent progression of liver fibrosis to cirrhosis. Taken together, these findings suggest that SNAP-23 is an important regular of actin dynamics in myofibroblastic HSC, but that the role of SNAP-23 in the progression of liver fibrosis in vivo is unclear.


Subject(s)
Actin Cytoskeleton/ultrastructure , Hepatic Stellate Cells/ultrastructure , Myofibroblasts/ultrastructure , Qb-SNARE Proteins/deficiency , Qc-SNARE Proteins/deficiency , Actin Cytoskeleton/chemistry , Actin Depolymerizing Factors/biosynthesis , Actins/analysis , Animals , Carbon Tetrachloride/toxicity , Cell Line , Cell Movement , Cell Separation , Gene Knockdown Techniques , Hepatic Stellate Cells/metabolism , Humans , Liver/cytology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/pathology , Mice , Qb-SNARE Proteins/antagonists & inhibitors , Qb-SNARE Proteins/genetics , Qb-SNARE Proteins/physiology , Qc-SNARE Proteins/antagonists & inhibitors , Qc-SNARE Proteins/genetics , Qc-SNARE Proteins/physiology , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Signal Transduction , Stress Fibers/chemistry , Stress Fibers/ultrastructure , Wound Healing , rho-Associated Kinases/physiology
12.
Oral Dis ; 25(1): 174-181, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30168888

ABSTRACT

OBJECTIVE: To immunohistochemically characterize a group of oral myofibroblastic lesions (MLs) and to evaluate the ultrastructural features of myofibroblasts. MATERIAL AND METHODS: Using a tissue microarray technique (TMA), cases of myofibroma (MF), of nodular fasciitis (NF), of desmoplastic fibroma (DF), and of myofibroblastic sarcoma (MS) from the Universidad Autónoma Metropolitana Xochimilco, and a Private Oral Pathology Service in Mexico City were stained with antibodies against alpha-smooth muscle actin (α-SMA), H-caldesmon, vimentin, desmin, ß-catenin, CD34, anaplastic lymphoma protein kinase (ALK-1), and Ki-67. RESULTS: Nineteen of the 22 MF cases, 2/5 of the NF cases, 1/10 of the DF cases, and 1/2 of the MS cases were positive for α-SMA. 1/2 of the MS cases were positive for desmin; 6/10 of the DF cases were positive for ß-catenin, and 2 of the MF cases were positive for ALK-1. All of the MLs were positive for vimentin and negative for H-caldesmon and CD-34. The Ki-67 labeling index in all of the 8/22 MF, 3/5 NF, and 2/2 MS cases was ≥10%. For all of the MLs evaluated, ultrastructural analysis revealed spindle-shaped cells containing endoplasmic reticulum and peripheral actin filament bundles. CONCLUSION: In certain myofibroblastic lesions, the use of auxiliary techniques (such as immunohistochemistry) can be critical for differential diagnosis.


Subject(s)
Fibroma/diagnosis , Fibroma/pathology , Mouth/pathology , Myofibroblasts/pathology , Adolescent , Adult , Aged , Child , Child, Preschool , Female , Humans , Immunohistochemistry , Infant , Male , Mexico , Middle Aged , Myofibroblasts/ultrastructure , Tissue Array Analysis , Young Adult
13.
Sci Rep ; 8(1): 14780, 2018 10 03.
Article in English | MEDLINE | ID: mdl-30283023

ABSTRACT

Telocytes are a recently described stromal cell type widely distributed in various organs including the female and male reproductive systems. This study was aimed to investigate for the first time the existence, distribution and characteristics of telocytes in normal human testis by an integrated morphological approach (immunohistochemistry, immunofluorescence and transmission electron microscopy). We found that telocytes displaying typical long and moniliform prolongations and coexpressing CD34 and PDGFRα formed networks in the outer layer of peritubular tissue and around Leydig cells and vessels in the intertubular stroma. Testicular telocytes were immunophenotypically negative for CD31, c-kit/CD117 as well as α-SMA, thus making them clearly distinguishable from myoid cells/myofibroblasts located in the inner layer of peritubular tissue. Transmission electron microscopy confirmed the presence of cells ultrastructurally identifiable as telocytes (i.e. cells with telopodes alternating podomers and podoms) in the aforementioned locations. Intercellular contacts between neighboring telocytes and telopodes were observed throughout the testicular stromal compartment. Telopodes intimately surrounded and often established close contacts with peritubular myoid cells/myofibroblasts, Leydig cells and vessels. Extracellular vesicles were also frequently detected near telopodes. In summary, we demonstrated that telocytes are a previously neglected stromal component of human testis with potential implications in tissue homeostasis deserving further investigation.


Subject(s)
Leydig Cells/ultrastructure , Telocytes/ultrastructure , Telopodes/ultrastructure , Testis/ultrastructure , Antigens, CD34/genetics , Connective Tissue/metabolism , Connective Tissue/ultrastructure , Gene Expression Regulation , Humans , Immunohistochemistry , Immunophenotyping , Leydig Cells/metabolism , Male , Microscopy, Electron, Transmission , Myofibroblasts/metabolism , Myofibroblasts/ultrastructure , Receptor, Platelet-Derived Growth Factor alpha/genetics , Stromal Cells/metabolism , Stromal Cells/ultrastructure , Telocytes/metabolism , Telopodes/metabolism , Testis/metabolism
14.
Clin Sci (Lond) ; 132(21): 2299-2322, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30293967

ABSTRACT

Autophagy has been identified as a cellular process of bulk degradation of cytoplasmic components and its persistent activation is critically involved in the renal damage induced by ureteral obstruction. However, the role and underlying mechanisms of autophagy in hyperuricemic nephropathy (HN) remain unknown. In the present study, we observed that inhibition of autophagy by 3-methyladenine (3-MA) abolished uric acid-induced differentiation of renal fibroblasts to myofibroblasts and activation of transforming growth factor-ß1 (TGF-ß1), epidermal growth factor receptor (EGFR), and Wnt signaling pathways in cultured renal interstitial fibroblasts. Treatment with 3-MA also abrogated the development of HN in vivo as evidenced by improving renal function, preserving renal tissue architecture, reducing the number of autophagic vacuoles, and decreasing microalbuminuria. Moreover, 3-MA was effective in attenuating renal deposition of extracellular matrix (ECM) proteins and expression of α-smooth muscle actin (α-SMA) and reducing renal epithelial cells arrested at the G2/M phase of cell cycle. Injury to the kidney resulted in increased expression of TGF-ß1 and TGFß receptor I, phosphorylation of Smad3 and TGF-ß-activated kinase 1 (TAK1), and activation of multiple cell signaling pathways associated with renal fibrogenesis, including Wnt, Notch, EGFR, and nuclear factor-κB (NF-κB). 3-MA treatment remarkably inhibited all these responses. In addition, 3-MA effectively suppressed infiltration of macrophages and lymphocytes as well as release of multiple profibrogenic cytokines/chemokines in the injured kidney. Collectively, these findings indicate that hyperuricemia-induced autophagy is critically involved in the activation of renal fibroblasts and development of renal fibrosis and suggest that inhibition of autophagy may represent a potential therapeutic strategy for HN.


Subject(s)
Adenine/analogs & derivatives , Autophagy/drug effects , Glomerulonephritis/prevention & control , Hyperuricemia/drug therapy , Kidney/drug effects , Myofibroblasts/drug effects , Adenine/pharmacology , Animals , Autophagy-Related Proteins/metabolism , Cell Line , Disease Models, Animal , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Fibrosis , G2 Phase Cell Cycle Checkpoints/drug effects , Glomerulonephritis/metabolism , Glomerulonephritis/pathology , Hyperuricemia/metabolism , Hyperuricemia/pathology , Kidney/metabolism , Kidney/ultrastructure , Male , Myofibroblasts/metabolism , Myofibroblasts/ultrastructure , Oxidative Stress/drug effects , Phenotype , Phosphorylation , Rats , Rats, Sprague-Dawley , Wnt Signaling Pathway/drug effects
15.
Microsc Res Tech ; 81(1): 46-57, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29024123

ABSTRACT

Renal fibrosis is characterized by glomerulosclerosis and tubulointerstitial fibrosis and its pathogenesis is associated with the activity of mesenchymal cells (fibroblasts), being essentially characterized by a process of excessive accumulation resulting from the deposition of extracellular matrix components. The aim of this study was to characterize the morphological presentation of chronic and fibrotic lesions in the glomerular, tubular, interstitial, and vascular compartments in feline CKD, as well as the possible participation of myofibroblasts in renal fibrotic processes in this species. Cat kidneys were collected and processed according to the conventional techniques for light microscopy, circular polarization, immunohistochemistry, and electron microscopy. Fibrotic alterations were present in all compartments analyzed. The main findings in the glomerular compartment were different degrees of glomerular sclerosis, synechia formation, Bowman's capsule calcification, in addition to glomerular basement membrane thickening and pericapsular fibrosis. The tubulointerstitial compartment had intense tubular degeneration and the immunostaining in tubular cells for mesenchymal cell markers demonstrated the possibility of mesenchymal epithelial transition and consequent involvement of myofibroblasts in the development of interstitial tubule damage. Infiltration of inflammatory cells, added to vessel thickening and fibrosis, demonstrated the severity and role of inflammation in the development and perpetuation of damage. Thus, we may conclude that fibrotic lesions play a relevant role in feline CKD and the mechanism of perpetuation of these lesions need further elucidation regarding the origin and participation of myofibroblasts and consequent mesenchymal epithelial transition in this species.


Subject(s)
Cat Diseases/pathology , Kidney/pathology , Renal Insufficiency, Chronic/veterinary , Actins/ultrastructure , Animals , Cats , Collagen/ultrastructure , Extracellular Matrix/ultrastructure , Female , Fibroblasts/ultrastructure , Fibrosis/veterinary , Immunohistochemistry/methods , Immunohistochemistry/veterinary , Inflammation/veterinary , Kidney/ultrastructure , Kidney Glomerulus/pathology , Kidney Glomerulus/ultrastructure , Male , Microscopy/methods , Microscopy/veterinary , Microscopy, Confocal/veterinary , Microscopy, Electron/veterinary , Microscopy, Polarization/veterinary , Myofibroblasts/ultrastructure , Renal Insufficiency, Chronic/pathology
16.
Pancreas ; 46(9): 1141-1151, 2017 10.
Article in English | MEDLINE | ID: mdl-28902784

ABSTRACT

OBJECTIVES: The origin of collagen-producing myofibroblasts in pancreatic fibrosis is still controversial. Pancreatic stellate cells (PSCs), which have been recognized as the pancreatic counterparts of hepatic stellate cells (HSCs), are thought to play an important role in the development of pancreatic fibrosis. However, sources of myofibroblasts other than PSCs may exist because extensive studies of liver fibrosis have uncovered myofibroblasts that did not originate from HSCs. This study aimed to characterize myofibroblasts in an experimental pancreatic fibrosis model in mice. METHODS: We used transgenic mice expressing green fluorescent protein via the collagen type I α1 promoter and induced pancreatic fibrosis with repetitive injections of cerulein. RESULTS: Collagen-producing cells that are negative for glial fibrillary acidic protein (ie, not derived from PSCs) exist in the pancreas. Pancreatic stellate cells had different characteristics from those of HSCs in a very small possession of vitamin A using mass spectrometry and a low expression of lecithin retinol acyltransferase. The microstructure of PSCs was entirely different from that of HSCs using flow cytometry and electron microscopy. CONCLUSIONS: Our study showed that characteristics of PSCs are different from those of HSCs, and myofibroblasts in the pancreas might be derived not only from PSCs but also from other fibrogenic cells.


Subject(s)
Collagen/biosynthesis , Hepatic Stellate Cells/metabolism , Pancreas/metabolism , Pancreatic Stellate Cells/metabolism , Animals , Cells, Cultured , Collagen/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/ultrastructure , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron , Microscopy, Fluorescence , Myofibroblasts/cytology , Myofibroblasts/metabolism , Myofibroblasts/ultrastructure , Pancreas/cytology , Pancreatic Stellate Cells/cytology , Pancreatic Stellate Cells/ultrastructure
17.
Autophagy ; 13(8): 1420-1434, 2017 Aug 03.
Article in English | MEDLINE | ID: mdl-28613983

ABSTRACT

Accumulation of profibrotic myofibroblasts is involved in the process of fibrosis development during idiopathic pulmonary fibrosis (IPF) pathogenesis. TGFB (transforming growth factor ß) is one of the major profibrotic cytokines for myofibroblast differentiation and NOX4 (NADPH oxidase 4) has an essential role in TGFB-mediated cell signaling. Azithromycin (AZM), a second-generation antibacterial macrolide, has a pleiotropic effect on cellular processes including proteostasis. Hence, we hypothesized that AZM may regulate NOX4 levels by modulating proteostasis machineries, resulting in inhibition of TGFB-associated lung fibrosis development. Human lung fibroblasts (LF) were used to evaluate TGFB-induced myofibroblast differentiation. With respect to NOX4 regulation via proteostasis, assays for macroautophagy/autophagy, the unfolded protein response (UPR), and proteasome activity were performed. The potential anti-fibrotic property of AZM was examined by using bleomycin (BLM)-induced lung fibrosis mouse models. TGFB-induced NOX4 and myofibroblast differentiation were clearly inhibited by AZM treatment in LF. AZM-mediated NOX4 reduction was restored by treatment with MG132, a proteasome inhibitor. AZM inhibited autophagy and enhanced the UPR. Autophagy inhibition by AZM was linked to ubiquitination of NOX4 via increased protein levels of STUB1 (STIP1 homology and U-box containing protein 1), an E3 ubiquitin ligase. An increased UPR by AZM was associated with enhanced proteasome activity. AZM suppressed lung fibrosis development induced by BLM with concomitantly reduced NOX4 protein levels and enhanced proteasome activation. These results suggest that AZM suppresses NOX4 by promoting proteasomal degradation, resulting in inhibition of TGFB-induced myofibroblast differentiation and lung fibrosis development. AZM may be a candidate for the treatment of the fibrotic lung disease IPF.


Subject(s)
Azithromycin/pharmacology , Cell Differentiation/drug effects , Lung/pathology , Myofibroblasts/pathology , NADPH Oxidase 4/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis/drug effects , Animals , Bleomycin , Disease Models, Animal , Fibrosis , Humans , Idiopathic Pulmonary Fibrosis/enzymology , Idiopathic Pulmonary Fibrosis/pathology , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Models, Biological , Myofibroblasts/drug effects , Myofibroblasts/enzymology , Myofibroblasts/ultrastructure , Reactive Oxygen Species/metabolism , Transforming Growth Factor beta1/pharmacology , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/drug effects , Unfolded Protein Response/drug effects
18.
Adv Exp Med Biol ; 913: 403-424, 2016.
Article in English | MEDLINE | ID: mdl-27796902

ABSTRACT

Currently, the exact role of telocytes within fascial structures is unknown. The morphology, distribution and behaviour of fascial telocytes as well as the mutual relationship between telocytes and other cellular fascia constituents should be definitely a subject of further studies. It will contribute to better understanding of the role of the fascial system in health and diseases, may shed light on the regeneration potential of these tissues and may help to find targets for future treatments for locomotor disorders, including fascial diseases. Last but not least, confirmation of the presence of telocytes within fascia may contribute to optimise the use of fascia as a graft material.


Subject(s)
Back Muscles/ultrastructure , Fascia Lata/ultrastructure , Telocytes/ultrastructure , Back Muscles/metabolism , Cell Communication , Connective Tissue/metabolism , Connective Tissue/ultrastructure , Endothelial Cells/metabolism , Endothelial Cells/ultrastructure , Fascia Lata/metabolism , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Humans , Mast Cells/metabolism , Mast Cells/ultrastructure , Microscopy, Electron, Transmission , Myofibroblasts/metabolism , Myofibroblasts/ultrastructure , Neurons/metabolism , Neurons/ultrastructure , Regeneration/physiology , Telocytes/metabolism
19.
Ultrastruct Pathol ; 40(5): 276-87, 2016.
Article in English | MEDLINE | ID: mdl-27409252

ABSTRACT

The present work deals with the simultaneous ultrastructure and triple immunofluorescence study of the three main hepatic fibrogenic cells, hepatic stellate cell, myofibroblast (MF), and fibroblast, in a group of hepatitis C virus (HCV) RNA positive patients, as their exact interrelation behavior in vivo with the progress of hepatic fibrosis is still inadequate. In this study, for the first time, cells having the morphological characteristic of MF and not bone marrow fibrocytes were revealed in liver portal vessels. This necessitates the reevaluation of the available knowledge concerning bone marrow fibrocyte. Also, the distribution, cellular interrelations, and the fate of MF were highlighted.


Subject(s)
Fibroblasts/ultrastructure , Hepatic Stellate Cells/ultrastructure , Hepatitis C/pathology , Liver Cirrhosis/pathology , Myofibroblasts/ultrastructure , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Hepatitis C/complications , Humans , Hyaluronic Acid , Liver Cirrhosis/virology , Microscopy, Confocal , Microscopy, Electron, Transmission
20.
Circ J ; 80(9): 1971-9, 2016 Aug 25.
Article in English | MEDLINE | ID: mdl-27396441

ABSTRACT

BACKGROUND: Inflammatory responses, especially by CD4(+)T cells activated by dendritic cells, are known to be important in the pathophysiology of cardiac repair after myocardial infarction (MI). Although co-stimulatory signals through B7 (CD80/86) and CD28 are necessary for CD4(+)T cell activation and survival, the roles of these signals in cardiac repair after MI are still unclear. METHODS AND RESULTS: C57BL/6 (Control) mice and CD28 knockout (CD28KO) mice were subjected to left coronary artery permanent ligation. The ratio of death by cardiac rupture within 5 days after MI was significantly higher in CD28KO mice compared with Control mice. Although there were no significant differences in the infarct size between the 2 groups, left ventricular end-diastolic and end-systolic diameters were significantly increased, and fractional shortening was significantly decreased in CD28KO mice compared with Control mice. Electron microscopic observation revealed that the extent of extracellular collagen fiber was significantly decreased in CD28KO mice compared with Control mice. The number of α-smooth muscle actin-positive myofibroblasts was significantly decreased, and matrix metalloproteinase-9 activity and the mRNA expression of interleukin-1ß were significantly increased in CD28KO mice compared with Control mice. CONCLUSIONS: Deletion of CD28 co-stimulatory signals exacerbates left ventricular remodeling and increases cardiac rupture after MI through prolongation of the inflammatory period and reduction of collagen fiber in the infarct scars. (Circ J 2016; 80: 1971-1979).


Subject(s)
CD28 Antigens/deficiency , Gene Deletion , Heart Rupture, Post-Infarction/metabolism , Myocardial Infarction/metabolism , Signal Transduction , Ventricular Remodeling , Animals , CD28 Antigens/metabolism , Gene Expression Regulation , Heart Rupture, Post-Infarction/genetics , Heart Rupture, Post-Infarction/pathology , Heart Rupture, Post-Infarction/physiopathology , Interleukin-1beta/biosynthesis , Interleukin-1beta/genetics , Male , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase 9/genetics , Mice , Mice, Knockout , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myofibroblasts/metabolism , Myofibroblasts/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...