Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.328
Filter
1.
Am J Physiol Cell Physiol ; 326(5): C1482-C1493, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38525537

ABSTRACT

Corneal fibroblasts maintain homeostasis of the corneal stroma by mediating the synthesis and degradation of extracellular collagen, and these actions are promoted by transforming growth factor-ß (TGF-ß) and interleukin-1ß (IL-1ß), respectively. The cornea is densely innervated with sensory nerve fibers that are not only responsible for sensation but also required for physiological processes such as tear secretion and wound healing. Loss or dysfunction of corneal nerves thus impairs corneal epithelial wound healing and can lead to neurotrophic keratopathy. The sensory neurotransmitter substance P (SP) promotes corneal epithelial wound healing by enhancing the stimulatory effects of growth factors and fibronectin. We have now investigated the role of SP in collagen metabolism mediated by human corneal fibroblasts in culture. Although SP alone had no effect on collagen synthesis or degradation by these cells, it promoted the stimulatory effect of TGF-ß on collagen type I synthesis without affecting that of IL-1ß on the expression of matrix metalloproteinase-1. This effect of SP on TGF-ß-induced collagen synthesis was accompanied by activation of p38 mitogen-activated protein kinase (MAPK) signaling and was attenuated by pharmacological inhibition of p38 or of the neurokinin-1 receptor. Our results thus implicate SP as a modulator of TGF-ß-induced collagen type I synthesis by human corneal fibroblasts, and they suggest that loss of this function may contribute to the development of neurotrophic keratopathy.NEW & NOTEWORTHY This study investigates the role of substance P (SP) in collagen metabolism mediated by human corneal fibroblasts in culture. We found that, although SP alone had no effect on collagen synthesis or degradation by corneal fibroblasts, it promoted the stimulatory effect of transforming growth factor-ß on collagen type I synthesis without affecting that of interleukin-1ß on the expression of matrix metalloproteinase-1.


Subject(s)
Fibroblasts , Interleukin-1beta , Substance P , Transforming Growth Factor beta , p38 Mitogen-Activated Protein Kinases , Humans , Substance P/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Transforming Growth Factor beta/metabolism , Fibroblasts/metabolism , Fibroblasts/drug effects , Cells, Cultured , Interleukin-1beta/metabolism , Collagen Type I/metabolism , Collagen Type I/biosynthesis , Receptors, Neurokinin-1/metabolism , Cornea/metabolism , Cornea/drug effects , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 1/genetics , Collagen/metabolism , Collagen/biosynthesis , Signal Transduction/drug effects , Corneal Stroma/metabolism , Corneal Stroma/drug effects , Corneal Keratocytes/metabolism , Corneal Keratocytes/drug effects
2.
J Biol Chem ; 298(9): 102246, 2022 09.
Article in English | MEDLINE | ID: mdl-35835217

ABSTRACT

Proximal tubular epithelial cells respond to transforming growth factor ß (TGFß) to synthesize collagen I (α2) during renal fibrosis. The oncoprotein DJ-1 has previously been shown to promote tumorigenesis and prevent apoptosis of dopaminergic neurons; however, its role in fibrosis signaling is unclear. Here, we show TGFß-stimulation increased expression of DJ-1, which promoted noncanonical mTORC1 and mTORC2 activities. We show DJ-1 augmented the phosphorylation/activation of PKCßII, a direct substrate of mTORC2. In addition, coimmunoprecipitation experiments revealed association of DJ-1 with Raptor and Rictor, exclusive subunits of mTORC1 and mTORC2, respectively, as well as with mTOR kinase. Interestingly, siRNAs against DJ-1 blocked TGFß-stimulated expression of collagen I (α2), while expression of DJ-1 increased expression of this protein. In addition, expression of dominant negative PKCßII and siRNAs against PKCßII significantly inhibited TGFß-induced collagen I (α2) expression. In fact, constitutively active PKCßII abrogated the effect of siRNAs against DJ-1, suggesting a role of PKCßII downstream of this oncoprotein. Moreover, we demonstrate expression of collagen I (α2) stimulated by DJ-1 and its target PKCßII is dependent on the transcription factor hypoxia-inducible factor 1α (Hif1α). Finally, we show in the renal cortex of diabetic rats that increased TGFß was associated with enhanced expression of DJ-1 and activation of mTOR and PKCßII, concomitant with increased Hif1α and collagen I (α2). Overall, we identified that DJ-1 affects TGFß-induced expression of collagen I (α2) via an mTOR-, PKCßII-, and Hif1α-dependent mechanism to regulate renal fibrosis.


Subject(s)
Collagen Type I , Diabetes Mellitus, Experimental , Diabetic Nephropathies , Hypoxia-Inducible Factor 1, alpha Subunit , Kidney , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Oncogene Proteins , Protein Deglycase DJ-1 , Animals , Collagen Type I/biosynthesis , Collagen Type I/genetics , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Fibrosis , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney/pathology , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Protein Deglycase DJ-1/genetics , Protein Deglycase DJ-1/metabolism , Protein Kinase C beta/metabolism , RNA, Small Interfering/metabolism , Rats , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology
3.
Int Urogynecol J ; 33(7): 1803-1812, 2022 07.
Article in English | MEDLINE | ID: mdl-35596801

ABSTRACT

INTRODUCTION AND HYPOTHESIS: This study was aimed at identifying the difference in collagen type-1 expression in women with and without pelvic organ prolapse (POP). METHODS: A systematic review and meta-analysis was carried out women with and without pelvic organ prolapse. This meta-analysis was conducted on research articles describing the evaluation of collagen type-1 expression between patients with and without POP. The articles were obtained from PubMed, EBSCO, and ProQuest, and were published between January 2000 and June 2021. Pooled mean difference (MD) and pooled odds ratio (OR) were calculated using fixed effect models. Review Manager (RevMan 5.4) was used to analyze the data. The main outcome measures were pooled MD and pooled OR of collagen type-1 expression in patients with and without POP. RESULTS: A total of seven case-control studies were included in the meta-analysis using the effect size of the MD and two case-control studies were included in the meta-analysis using the effect size of the OR. A total of 247 POP cases and 132 non-POP cases were identified from the studies. Our study indicated that patients with POP had a lower level of collagen type-1 expression than non-POP patients (MD = -6.77; 95% CI: -8.37, -5.17, p < 0.00001). Patients with low expression of collagen type-1 in pelvic support tissue are at a more than 3 times higher risk of suffering from pelvic organ prolapse (OR = 3.23, 95% CI: 1.52 to 6.87, p = 0.002). CONCLUSION: The results of this study showed that patients with pelvic organ prolapse have lower expression of collagen type-1 than nonpelvic organ prolapse patients.


Subject(s)
Collagen Type I , Pelvic Organ Prolapse , Collagen Type I/biosynthesis , Collagen Type I/metabolism , Female , Humans , Pelvic Organ Prolapse/metabolism
4.
Tissue Cell ; 74: 101704, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34871826

ABSTRACT

As a principal matrisomal protein, collagen is involved in the regulation of the structural framework of extracellular matrix (ECM) and therefore is potentially crucial in determining the biophysical character of the ECM. It has been suggested that collagen architecture plays a role in ovarian cancer development, progression and therapeutic responses which led us to examine the collagen morphology in normal and cancerous ovarian tissue. Also, the behaviour of ovarian cancer cells cultured in four qualitatively different collagen gels was investigated. The results here provide evidence that collagen I morphology in the cancerous ovary is distinct from that in the normal ovary. Tumour-associated collagen I showed streams or channels of thick elongated collagen I fibrils. Moreover, fibril alignment was significantly more prevalent in endometrioid and clear cell cancers than other ovarian cancer subtypes. In this work, for the first-time collagen I architecture profiling (CAP) was introduced using histochemical staining, which distinguished between the collagen I morphologies of ovarian cancer subtypes. Immunohistochemical examination of ovarian normal and cancerous tissues also supported the notion that focal adhesion and Rho signalling are upregulated in ovarian cancers, especially in the high-grade serous tumours, as indicated by higher expression of p-FAK and p190RhoGEF. The results also support the concept that collagen I architecture, which might be collagen I concentration-dependent, influences proliferation in ovarian cancer cells. The study provides evidence that modification of collagen I architecture integrity is associated with ovarian cancer development and therapeutic responses.


Subject(s)
Collagen Type I/biosynthesis , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/blood , Ovarian Neoplasms/metabolism , Cell Line, Tumor , Collagen Type I/genetics , Female , Humans , Neoplasm Proteins/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology
5.
J Mol Biol ; 434(2): 167394, 2022 01 30.
Article in English | MEDLINE | ID: mdl-34896113

ABSTRACT

Excessive synthesis of type I collagen is a hallmark of fibrotic diseases. Binding of La-related protein 6 (LARP6) to the 5' stem-loop (5'SL) of collagen mRNAs regulates their translation leading to an unnaturally elevated rate of collagen biosynthesis in fibrosis. Previous work suggested that LARP6 needs two domains to form stable complex with 5'SL RNA, the La domain and the juxtaposed RNA recognition motif (RRM), jointly called the La-module. Here we describe that La domain of LARP6 is necessary and sufficient for recognition of 5'SL in RNA sequence specific manner. A three-amino-acid motif located in the flexible loop connecting the second α-helix to the ß-sheet of the La domain, called the RNK-motif, is critical for binding. Mutation of any of these three amino acids abolishes the binding of the La domain to 5'SL. The major site of crosslinking of LARP6 to 5'SL RNA was mapped to this motif, as well. The RNK-motif is not found in other LARPs, which cannot bind 5'SL. Presence of RRM increases the stability of complex between La domain and 5'SL RNA and RRM domain does not make extensive contacts with 5'SL RNA. We propose a model in which the initial recognition of 5'SL by LARP6 is mediated by the RNK epitope and further stabilized by the RRM domain. This discovery suggests that the interaction between LARP6 and collagen mRNAs can be blocked by small molecules that target the RNK epitope and will help rational design of the LARP6 binding inhibitors as specific antifibrotic drugs.


Subject(s)
Autoantigens/chemistry , Collagen Type I/chemistry , Fibrosis/metabolism , RNA, Messenger/chemistry , Ribonucleoproteins/chemistry , Amino Acid Motifs , Autoantigens/genetics , Autoantigens/metabolism , Collagen , Collagen Type I/biosynthesis , Humans , Nucleic Acid Conformation , Pharmaceutical Preparations , Protein Binding , Protein Domains , RNA, Messenger/metabolism , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , SS-B Antigen
6.
Nat Commun ; 12(1): 7199, 2021 12 10.
Article in English | MEDLINE | ID: mdl-34893625

ABSTRACT

Type I collagen (Col1) is the most abundant protein in mammals. Col1 contributes to 90% of the total organic component of bone matrix. However, the precise cellular origin and functional contribution of Col1 in embryogenesis and bone formation remain unknown. Single-cell RNA-sequencing analysis identifies Fap+ cells and Fsp1+ cells as the major contributors of Col1 in the bone. We generate transgenic mouse models to genetically delete Col1 in various cell lineages. Complete, whole-body Col1 deletion leads to failed gastrulation and early embryonic lethality. Specific Col1 deletion in Fap+ cells causes severe skeletal defects, with hemorrhage, edema, and prenatal lethality. Specific Col1 deletion in Fsp1+ cells results in Osteogenesis Imperfecta-like phenotypes in adult mice, with spontaneous fractures and compromised bone healing. This study demonstrates specific contributions of mesenchymal cell lineages to Col1 production in organogenesis, skeletal development, and bone formation/repair, with potential insights into cell-based therapy for patients with Osteogenesis Imperfecta.


Subject(s)
Collagen Type I/biosynthesis , Embryonic Development/physiology , Fibroblasts/metabolism , Osteogenesis Imperfecta/metabolism , Osteogenesis/physiology , Animals , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Lineage , Collagen Type I/genetics , Collagen Type I, alpha 1 Chain/biosynthesis , Collagen Type I, alpha 1 Chain/genetics , Embryonic Development/genetics , Female , Femur , Fibroblasts/pathology , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , Osteogenesis/genetics , Osteogenesis Imperfecta/genetics , Osteogenesis Imperfecta/pathology , Phenotype , Pregnancy
7.
Plast Reconstr Surg ; 148(6S): 32S-38S, 2021 Dec 01.
Article in English | MEDLINE | ID: mdl-34847096

ABSTRACT

SUMMARY: Recombinant human type I collagen, identical in structure and functionality to human type I collagen, was successfully expressed and extracted from genetically modified tobacco plants. Contrarily to tissue extracted protein, rhCollagen is not immunogenic and not allergenic and has an intact triple helix structure showing superior biological functionality. A photocurable rhCollagen was developed by chemically modifying the protein to allow cross-linking under illumination at various wavelengths, maintaining the protein structural and biological functions. The use of the photocurable rhCollagen in aesthetic medicine, especially as a dermal filler and as a bioink for 3D-printed breast implant is discussed in this article.


Subject(s)
Collagen Type I/biosynthesis , Esthetics , Nicotiana/chemistry , Plant Extracts/chemistry , Recombinant Proteins/biosynthesis , Breast Implants , Dermal Fillers/therapeutic use , Humans , Printing, Three-Dimensional , Prosthesis Design , Skin Aging/drug effects
8.
Int J Mol Sci ; 22(22)2021 Nov 17.
Article in English | MEDLINE | ID: mdl-34830270

ABSTRACT

The extracellular matrix of the bladder consists mostly of type I and III collagen, which are required during loading. During bladder injury, there is an accumulation of collagen that impairs bladder function. Little is known about the genes that regulate production of collagens in the bladder. We demonstrate that the transcription factor Odd-skipped related 1 (Osr1) is expressed in the bladder mesenchyme and epithelium at the onset of development. As development proceeds, Osr1 is mainly expressed in mesenchymal progenitors and their derivatives. We hypothesized that Osr1 regulates mesenchymal cell differentiation and production of collagens in the bladder. To test this hypothesis, we examined newborn and adult mice heterozygous for Osr1, Osr1+/-. The bladders of newborn Osr1+/- mice had a decrease in collagen I by western blot analysis and a global decrease in collagens using Sirius red staining. There was also a decrease in the cellularity of the lamina propria, where most collagen is synthesized. This was not due to decreased proliferation or increased apoptosis in this cell population. Surprisingly, the bladders of adult Osr1+/- mice had an increase in collagen that was associated with abnormal bladder function; they also had a decrease in bladder capacity and voided more frequently. The results suggest that Osr1 is important for the differentiation of mesenchymal cells that give rise to collagen-producing cells.


Subject(s)
Collagen Type I/biosynthesis , Mesenchymal Stem Cells/metabolism , Signal Transduction/genetics , Transcription Factors/metabolism , Urinary Bladder/metabolism , Animals , Animals, Newborn , Cell Differentiation/genetics , Cell Proliferation/genetics , Cells, Cultured , Extracellular Matrix/metabolism , Gene Expression Regulation, Developmental , Mesenchymal Stem Cells/cytology , Mesoderm/metabolism , Mice , Mice, Transgenic , Mucous Membrane/cytology , Mucous Membrane/metabolism , Organogenesis/genetics , Transcription Factors/genetics
9.
Int J Mol Sci ; 22(9)2021 May 05.
Article in English | MEDLINE | ID: mdl-34063118

ABSTRACT

Mesenchymal stem cells (MSCs) have been widely used in therapeutic applications for many decades. However, more and more evidence suggests that factors such as the site of origin and pre-implantation treatment have a crucial impact on the result. This study investigates the role of freshly isolated MSCs in the lacrimal gland after allogeneic transplantation. For this purpose, MSCs from transgenic GFP mice were isolated and transplanted into allogeneic and syngeneic recipients. While the syngeneic MSCs maintained a spherical shape, allogeneic MSCs engrafted into the tissue as spindle-shaped cells in the interstitial stroma. Furthermore, the MSCs produced collagen type I in more than 85% to 95% of the detected GFP+ MSCs in the recipients of both models, supposedly contributing to pathogenic fibrosis in allogeneic recipients compared to syngeneic models. These findings indicate that allogeneic MSCs act completely differently from syngeneic MSCs, highlighting the importance of understanding the exact mechanisms behind MSCs.


Subject(s)
Bone Marrow Transplantation , Collagen Type I/biosynthesis , Mesenchymal Stem Cells/metabolism , Animals , Lacrimal Apparatus/cytology , Mesenchymal Stem Cells/cytology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Transplantation, Homologous , Transplantation, Isogeneic
10.
Int J Mol Sci ; 22(11)2021 May 23.
Article in English | MEDLINE | ID: mdl-34071003

ABSTRACT

Superoxide dismutase (SOD) is a major antioxidant enzyme for superoxide removal, and cytoplasmic SOD (SOD1) is expressed as a predominant isoform in all cells. We previously reported that renal SOD1 deficiency accelerates the progression of diabetic nephropathy (DN) via increasing renal oxidative stress. To evaluate whether the degree of SOD1 expression determines regeneration capacity and sarcopenic phenotypes of skeletal muscles under incipient and advanced DN conditions, we investigated the alterations of SOD1 expression, oxidative stress marker, inflammation, fibrosis, and regeneration capacity in cardiotoxin (CTX)-injured tibialis anterior (TA) muscles of two Akita diabetic mouse models with different susceptibility to DN, DN-resistant C57BL/6-Ins2Akita and DN-prone KK/Ta-Ins2Akita mice. Here, we report that KK/Ta-Ins2Akita mice, but not C57BL/6-Ins2Akita mice, exhibit delayed muscle regeneration after CTX injection, as demonstrated by the finding indicating significantly smaller average cross-sectional areas of regenerating TA muscle myofibers relative to KK/Ta-wild-type mice. Furthermore, we observed markedly reduced SOD1 expression in CTX-injected TA muscles of KK/Ta-Ins2Akita mice, but not C57BL/6-Ins2Akita mice, along with increased inflammatory cell infiltration, prominent fibrosis and superoxide overproduction. Our study provides the first evidence that SOD1 reduction and the following superoxide overproduction delay skeletal muscle regeneration through induction of overt inflammation and fibrosis in a mouse model of progressive DN.


Subject(s)
Diabetic Nephropathies/complications , Muscle, Skeletal/drug effects , Nerve Regeneration/drug effects , Sarcopenia/etiology , Superoxide Dismutase-1/drug effects , Animals , Cardiotoxins/toxicity , Collagen Type I/biosynthesis , Collagen Type I/genetics , Collagen Type I, alpha 1 Chain , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/genetics , Diabetic Nephropathies/enzymology , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Disease Progression , Enzyme Induction/drug effects , Fibrosis , Gene Expression Regulation, Enzymologic , Genetic Predisposition to Disease , Glomerular Mesangium/pathology , Inflammation , Insulin/deficiency , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Muscle, Skeletal/enzymology , Muscle, Skeletal/pathology , Muscle, Skeletal/physiology , Oxidative Stress/drug effects , Superoxide Dismutase-1/biosynthesis , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/physiology , Superoxides/metabolism
11.
Biochim Biophys Acta Mol Cell Res ; 1868(10): 119083, 2021 09.
Article in English | MEDLINE | ID: mdl-34147561

ABSTRACT

Airway wall remodeling, a main pathology of asthma was linked to vitamin-D deficiency and protein arginine methyltransferase-1 (PRMT1) expression in sub-epithelial cell layers. Calcitriol reduced remodeling in asthma model, but its mode of action is unclear. This study assessed the effect of calcitriol on PRMT1-dependent fibroblast remodeling in human lung fibroblasts, and allergen-induced asthma in E3-rats. Fibroblasts were activated with thymic stromal lymphopoietin (TLSP); asthma was induced by ovalbumin inhalation in rats. The airway structure was assessed by immunohistology. Protein expression in fibroblasts and activation of the mitogen activated protein kinases were detected by Western-blotting. Transcription factor activation was determined by luciferase reporter assay. PRMT1 action was blocked by siRNA and PRMT-inhibition. Ovalbumin upregulated the expression of TSLP, PRMT1, matrix metallopro-teinase-1 (MMP1), interleukin-25, and collagen type-I in sub-epithelial fibroblasts. In isolated fibroblasts, TSLP induced the same proteins, which were blocked by inhibition of Erk1/2 and p38. TLSP induced PRMT1 through activation of signal transducer and activator of transcription-3. PRMT1 inhibition reduced collagen type-I expression and suppressed MMP1. In fibroblasts, calcitriol supplementation over 12 days prevented TSLP-induced remodeling by blocking the PRMT1 levels. Interestingly, short-term calcitriol treatment had no such effect. The data support the beneficial role of calcitriol in asthma therapy.


Subject(s)
Collagen Type I/biosynthesis , Cytokines/metabolism , Fibroblasts/metabolism , Lung/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , STAT3 Transcription Factor/metabolism , Animals , Calcitriol/pharmacology , Cell Line , Fibroblasts/drug effects , Humans , Lung/drug effects , Rats
12.
Am J Physiol Lung Cell Mol Physiol ; 321(2): L377-L391, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34105356

ABSTRACT

Genome-wide association studies have shown that a gene variant in the Family with sequence similarity 13, member A (FAM13A) is strongly associated with reduced lung function and the appearance of respiratory symptoms in patients with chronic obstructive pulmonary disease (COPD). A key player in smoking-induced tissue injury and airway remodeling is the transforming growth factor-ß1 (TGF-ß1). To determine the role of FAM13A in TGF-ß1 signaling, FAM13A-/- airway epithelial cells were generated using CRISPR-Cas9, whereas overexpression of FAM13A was achieved using lipid nanoparticles. Wild-type (WT) and FAM13A-/- cells were treated with TGF-ß1, followed by gene and/or protein expression analyses. FAM13A-/- cells augmented TGF-ß1-induced increase in collagen type 1 (COL1A1), matrix metalloproteinase 2 (MMP2), expression compared with WT cells. This effect was mediated by an increase in ß-catenin (CTNNB1) expression in FAM13A-/- cells compared with WT cells after TGF-ß1 treatment. FAM13A overexpression was partially protective from TGF-ß1-induced COL1A1 expression. Finally, we showed that airway epithelial-specific FAM13A protein expression is significantly increased in patients with severe COPD compared with control nonsmokers, and negatively correlated with lung function. In contrast, ß-catenin (CTNNB1), which has previously been linked to be regulated by FAM13A, is decreased in the airway epithelium of smokers with COPD compared with non-COPD subjects. Together, our data showed that FAM13A may be protective from TGF-ß1-induced fibrotic response in the airway epithelium via sequestering CTNNB1 from its regulation on downstream targets. Therapeutic increase in FAM13A expression in the airway epithelium of smokers at risk for COPD, and those with mild COPD, may reduce the extent of airway tissue remodeling.


Subject(s)
Airway Remodeling , GTPase-Activating Proteins/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Respiratory Mucosa/metabolism , Smoking/metabolism , Transforming Growth Factor beta1/metabolism , Adult , Aged , Cell Line , Collagen Type I/biosynthesis , Collagen Type I/genetics , Collagen Type I, alpha 1 Chain , Female , GTPase-Activating Proteins/genetics , Gene Expression Regulation , Humans , Male , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 2/genetics , Middle Aged , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/pathology , Respiratory Mucosa/pathology , Smoking/genetics , Smoking/pathology , Transforming Growth Factor beta1/genetics , beta Catenin/biosynthesis , beta Catenin/genetics
13.
Hum Genet ; 140(8): 1121-1141, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34169326

ABSTRACT

Osteogenesis Imperfecta (OI) comprises a heterogeneous group of patients who share bone fragility and deformities as the main characteristics, albeit with different degrees of severity. Phenotypic variation also exists in other connective tissue aspects of the disease, complicating disease classification and disease course prediction. Although collagen type I defects are long established as the primary cause of the bone pathology, we are still far from comprehending the complete mechanism. In the last years, the advent of next generation sequencing has triggered the discovery of many new genetic causes for OI, helping to draw its molecular landscape. It has become clear that, in addition to collagen type I genes, OI can be caused by multiple proteins connected to different parts of collagen biosynthesis. The production of collagen entails a complex process, starting from the production of the collagen Iα1 and collagen Iα2 chains in the endoplasmic reticulum, during and after which procollagen is subjected to a plethora of posttranslational modifications by chaperones. After reaching the Golgi organelle, procollagen is destined to the extracellular matrix where it forms collagen fibrils. Recently discovered mutations in components of the retrograde transport of chaperones highlight its emerging role as critical contributor of OI development. This review offers an overview of collagen regulation in the context of recent gene discoveries, emphasizing the significance of transport disruptions in the OI mechanism. We aim to motivate exploration of skeletal fragility in OI from the perspective of these pathways to identify regulatory points which can hint to therapeutic targets.


Subject(s)
Bone and Bones/metabolism , Collagen Type I/biosynthesis , Osteoblasts/metabolism , Osteogenesis Imperfecta/metabolism , Procollagen/biosynthesis , Protein Processing, Post-Translational , Bone and Bones/pathology , Collagen Type I/genetics , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Golgi Apparatus/genetics , Golgi Apparatus/metabolism , High-Throughput Nucleotide Sequencing , Humans , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Mutation , Osteoblasts/pathology , Osteogenesis Imperfecta/genetics , Osteogenesis Imperfecta/pathology , Procollagen/genetics , Protein Biosynthesis , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Protein Transport , Severity of Illness Index
15.
Eur J Pharmacol ; 900: 174045, 2021 Jun 05.
Article in English | MEDLINE | ID: mdl-33745956

ABSTRACT

The ubiquitin-proteasome system (UPS) plays an essential role in cellular homeostasis and myocardial function. Ubiquitin carboxy-terminal hydrolase 1 (UCHL1) is involved in cardiac remodeling, but its underlying mechanisms are largely unknown. Here, we observed that the UCHL1 was significantly up-regulated in angiotensin II-infused heart and primary cardiac fibroblast (CF). Systemic administration of the UCHL1 inhibitor LDN57444 significantly ameliorated cardiac fibrosis and improved cardiac function induced by angiotensin II. Also, LDN57444 inhibited CF cell proliferation as well as attenuated collagen I, and CTGF gene expression in the presence of Ang II. Mechanistically, UCHL1 promotes angiotensin II-induced fibrotic responses by way of activating nuclear factor kappa B (NF-κB) signaling. Moreover, suppression of the NF-κB pathway interfered with UCHL1 overexpression-mediated fibrotic responses. Besides, the chromatin immunoprecipitation assay demonstrated that NF-κB can bind to the UCHL1 promoter and trigger its transcription in cardiac fibroblasts. These findings suggest that UCHL1 positively regulates cardiac fibrosis by modulating NF-κB signaling pathway and identify UCHL1 could be a new treatment strategy for cardiac fibrosis.


Subject(s)
Fibroblasts/drug effects , Myocardium/pathology , NF-kappa B/drug effects , Signal Transduction/drug effects , Ubiquitin Thiolesterase/antagonists & inhibitors , Angiotensin II/pharmacology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Animals, Newborn , Cell Proliferation/drug effects , Collagen Type I/antagonists & inhibitors , Collagen Type I/biosynthesis , Connective Tissue Growth Factor/antagonists & inhibitors , Fibrosis/prevention & control , Mice , Rats , Rats, Sprague-Dawley
16.
Histochem Cell Biol ; 155(4): 503-512, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33398434

ABSTRACT

Hypertension leads to structural remodeling of cerebral blood vessels, which has been implicated in the pathophysiology of cerebrovascular diseases. The remodeling and progression of arteriolosclerosis under hypertension involve fibrosis along with the production of type I collagen around cerebral arterioles. However, the source and regulatory mechanisms of this collagen production remain elusive. In this study, we examined if perivascular macrophages (PVMs) are involved in collagen production around cerebral small vessels in hypertensive SHRSP/Izm rats. Immunoreactivity for type I collagen around cerebral small vessels in 12-week-old hypertensive rats tended to higher than those in 4-week-old hypertensive and 12-week-old control rats. In ultrastructural analyses using transmission electron microscopy, the substantial deposition of collagen fibers could be observed in the intercellular spaces around PVMs near the arterioles of rats with prolonged hypertension. In situ hybridization analyses revealed that cells positive for mRNA of Col1a1, which comprises type I collagen, were observed near cerebral small vessels. The Col1a1-positive cells around cerebral small vessels were colocalized with immunoreactivity for CD206, a marker for PVMs, but not with those for glial fibrillary acidic protein or desmin, markers for other perivascular cells such as astrocytes and vascular smooth muscle cells. These results demonstrated that enhanced production of type I collagen is observed around cerebral small vessels in rats with prolonged hypertension and Col1a1 is expressed by PVMs, and support the concept that PVMs are involved in collagen production and vascular fibrosis under hypertensive conditions.


Subject(s)
Cerebral Arteries/metabolism , Collagen Type I/biosynthesis , Hypertension/metabolism , Macrophages/metabolism , Animals , Disease Models, Animal , Male , Rats , Rats, Inbred SHR , Rats, Inbred WKY
17.
Med Sci Monit ; 27: e927577, 2021 Jan 02.
Article in English | MEDLINE | ID: mdl-33386384

ABSTRACT

BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a serious irreversible lung disease. The mechanism of immune checkpoint in idiopathic pulmonary fibrosis is still unknown. MATERIAL AND METHODS First, the expression levels of PD-1/PD-L1 on the surface of CD4+ T cells and the proportion of Treg cells in IPF or controls were detected by flow cytometry. Then, expression of TGF-ß in blood samples was detected with ELISA. Moreover, a co-culture system was composed of fibroblasts stimulated by TGF-ß and CD4+ T cells from healthy people. The proportions of Treg cells and PD-1 in the co-culture system were detected. In addition, we detected the proportion of Treg cells and the level of collagen-1 after adding PD-1 or PD-L1 protein antibody blocker to the co-culture system. RESULTS Flow cytometry revealed the upregulated expression of PD-1/PD-L1 in CD4+ T cells of IPF patients. PD-1 appears to inhibit the differentiation of CD4+ T cells into Treg cells. Co-culture of myofibroblasts and CD4+ T cells induced the generation of collagen-1 and reduced the proliferation of CD4+ T cells. When PD-1 was blocked, the inhibition of Treg cell differentiation was reversed, accompanied by decreased collagen-1 production. CONCLUSIONS This work identified the molecular mechanism of PD-1 in patients with IPF. It may provide a new perspective on the therapeutic effect of PD-1.


Subject(s)
B7-H1 Antigen/metabolism , Idiopathic Pulmonary Fibrosis/immunology , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes, Regulatory/immunology , Aged , Cell Differentiation , Cell Proliferation , Collagen Type I/biosynthesis , Female , Humans , Male , Myofibroblasts/pathology , Signal Transduction , Transforming Growth Factor beta/metabolism
18.
Front Immunol ; 12: 816509, 2021.
Article in English | MEDLINE | ID: mdl-35126373

ABSTRACT

Fibrosis is a prominent feature of chronic allograft rejection, caused by an excessive production of matrix proteins, including collagen-1. Several cell types produce collagen-1, including mesenchymal fibroblasts and cells of hematopoietic origin. Here, we sought to determine whether tissue-resident donor-derived cells or allograft-infiltrating recipient-derived cells are responsible for allograft fibrosis, and whether hematopoietic cells contribute to collagen production. A fully MHC-mismatched mouse heterotopic heart transplantation model was used, with transient depletion of CD4+ T cells to prevent acute rejection. Collagen-1 was selectively knocked out in recipients or donors. In addition, collagen-1 was specifically deleted in hematopoietic cells. Tissue-resident macrophages were depleted using anti-CSF1R antibody. Allograft fibrosis and inflammation were quantified 20 days post-transplantation. Selective collagen-1 knock-out in recipients or donors showed that tissue-resident cells from donor hearts, but not infiltrating recipient-derived cells, are responsible for production of collagen-1 in allografts. Cell-type-specific knock-out experiments showed that hematopoietic tissue-resident cells in donor hearts substantially contributed to graft fibrosis. Tissue resident macrophages, however, were not responsible for collagen-production, as their deletion worsened allograft fibrosis. Donor-derived cells including those of hematopoietic origin determine allograft fibrosis, making them attractive targets for organ preconditioning to improve long-term transplantation outcomes.


Subject(s)
Collagen Type I/biosynthesis , Graft Rejection/etiology , Graft Rejection/metabolism , Heart Transplantation/adverse effects , Tissue Donors , Animals , Biomarkers , Chronic Disease , Collagen Type I/immunology , Disease Models, Animal , Disease Susceptibility , Graft Rejection/diagnosis , Heart Transplantation/methods , Immunophenotyping , Mice , Mice, Transgenic , Transplantation, Homologous
19.
Blood ; 137(7): 994-999, 2021 02 18.
Article in English | MEDLINE | ID: mdl-32915956
20.
Br J Nutr ; 126(3): 321-336, 2021 08 14.
Article in English | MEDLINE | ID: mdl-32718370

ABSTRACT

In the current research, a 60-d experiment was conducted with the purpose of exploring the impacts of methionine (Met) on growth performance, muscle nutritive deposition, muscle fibre growth and type I collagen synthesis as well as the related signalling pathway. Six diets (iso-nitrogenous) differing in Met concentrations (2·54, 4·85, 7·43, 10·12, 12·40 and 15·11 g/kg diets) were fed to 540 grass carp (178·47 (SD 0·36) g). Results showed (P < 0·05) that compared with Met deficiency, optimal level of dietary Met (1) increased feed intake, feed efficiency, specific growth rate and percentage weight gain (PWG); (2) increased fish muscle protein, lipid and free amino acid contents and improved fish muscle fatty acid profile as well as increased protein content in part associated with the target of rapamycin complex 1 (TORC1)/S6K1 signalling pathway; (3) increased the frequency distribution of muscle fibre with >50 µm of diameter; (4) increased type I collagen synthesis partly related to the transforming growth factor-ß1/Smads and CK2/TORC1 signalling pathways. In conclusion, dietary Met improved muscle growth, which might be due to the regulation of muscle nutritive deposition, muscle fibre growth and type I collagen synthesis-related signal molecules. Finally, according to PWG and muscle collagen content, the Met requirements for on-growing grass carp (178-626 g) were estimated to be 9·56 g/kg diet (33·26 g/kg protein of diet) and 9·28 g/kg diet (32·29 g/kg of dietary protein), respectively.


Subject(s)
Carps , Collagen Type I/biosynthesis , Methionine/administration & dosage , Muscle Fibers, Skeletal/physiology , Animal Feed/analysis , Animal Nutritional Physiological Phenomena , Animals , Carps/growth & development , Diet/veterinary , Dietary Supplements , Mechanistic Target of Rapamycin Complex 1 , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...