Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters

Publication year range
1.
Biophys J ; 122(16): 3219-3237, 2023 08 22.
Article in English | MEDLINE | ID: mdl-37415335

ABSTRACT

Collagen is a key structural component of multicellular organisms and is arranged in a highly organized manner. In structural tissues such as tendons, collagen forms bundles of parallel fibers between cells, which appear within a 24-h window between embryonic day 13.5 (E13.5) and E14.5 during mouse embryonic development. Current models assume that the organized structure of collagen requires direct cellular control, whereby cells actively lay down collagen fibrils from cell surfaces. However, such models appear incompatible with the time and length scales of fibril formation. We propose a phase-transition model to account for the rapid development of ordered fibrils in embryonic tendon, reducing reliance on active cellular processes. We develop phase-field crystal simulations of collagen fibrillogenesis in domains derived from electron micrographs of inter-cellular spaces in embryonic tendon and compare results qualitatively and quantitatively to observed patterns of fibril formation. To test the prediction of this phase-transition model that free protomeric collagen should exist in the inter-cellular spaces before the formation of observable fibrils, we use laser-capture microdissection, coupled with mass spectrometry, which demonstrates steadily increasing free collagen in inter-cellular spaces up to E13.5, followed by a rapid reduction of free collagen that coincides with the appearance of less-soluble collagen fibrils. The model and measurements together provide evidence for extracellular self-assembly of collagen fibrils in embryonic mouse tendon, supporting an additional mechanism for rapid collagen fibril formation during embryonic development.


Subject(s)
Embryonic Development , Extracellular Matrix , Animals , Mice , Extracellular Matrix/metabolism , Collagen/metabolism , Cell Membrane , Tendons/chemistry , Tendons/metabolism
2.
J Physiol ; 2023 Feb 21.
Article in English | MEDLINE | ID: mdl-36810732

ABSTRACT

Overuse injury in tendon tissue (tendinopathy) is a frequent and costly musculoskeletal disorder and represents a major clinical problem with unsolved pathogenesis. Studies in mice have demonstrated that circadian clock-controlled genes are vital for protein homeostasis and important in the development of tendinopathy. We performed RNA sequencing, collagen content and ultrastructural analyses on human tendon biopsies obtained 12 h apart in healthy individuals to establish whether human tendon is a peripheral clock tissue and we performed RNA sequencing on patients with chronic tendinopathy to examine the expression of circadian clock genes in tendinopathic tissues. We found time-dependent expression of 280 RNAs including 11 conserved circadian clock genes in healthy tendons and markedly fewer (23) differential RNAs with chronic tendinopathy. Further, the expression of COL1A1 and COL1A2 was reduced at night but was not circadian rhythmic in synchronised human tenocyte cultures. In conclusion, day-to-night changes in gene expression in healthy human patellar tendons indicate a conserved circadian clock as well as the existence of a night reduction in collagen I expression. KEY POINTS: Tendinopathy is a major clinical problem with unsolved pathogenesis. Previous work in mice has shown that a robust circadian rhythm is required for collagen homeostasis in tendons. The use of circadian medicine in the diagnosis and treatment of tendinopathy has been stifled by the lack of studies on human tissue. Here, we establish that the expression of circadian clock genes in human tendons is time dependent, and now we have data to corroborate that circadian output is reduced in diseased tendon tissues. We consider our findings to be of significance in advancing the use of the tendon circadian clock as a therapeutic target or preclinical biomarker for tendinopathy.

3.
PLoS Pathog ; 17(9): e1009840, 2021 09.
Article in English | MEDLINE | ID: mdl-34499689

ABSTRACT

COVID-19 vaccines based on the Spike protein of SARS-CoV-2 have been developed that appear to be largely successful in stopping infection. However, therapeutics that can help manage the disease are still required until immunity has been achieved globally. The identification of repurposed drugs that stop SARS-CoV-2 replication could have enormous utility in stemming the disease. Here, using a nano-luciferase tagged version of the virus (SARS-CoV-2-ΔOrf7a-NLuc) to quantitate viral load, we evaluated a range of human cell types for their ability to be infected and support replication of the virus, and performed a screen of 1971 FDA-approved drugs. Hepatocytes, kidney glomerulus, and proximal tubule cells were particularly effective in supporting SARS-CoV-2 replication, which is in-line with reported proteinuria and liver damage in patients with COVID-19. Using the nano-luciferase as a measure of virus replication we identified 35 drugs that reduced replication in Vero cells and human hepatocytes when treated prior to SARS-CoV-2 infection and found amodiaquine, atovaquone, bedaquiline, ebastine, LY2835219, manidipine, panobinostat, and vitamin D3 to be effective in slowing SARS-CoV-2 replication in human cells when used to treat infected cells. In conclusion, our study has identified strong candidates for drug repurposing, which could prove powerful additions to the treatment of COVID.


Subject(s)
COVID-19 Drug Treatment , Drug Discovery/methods , Drug Repositioning , SARS-CoV-2/drug effects , SARS-CoV-2/physiology , Animals , Biomarkers , Cell Line , Chlorocebus aethiops , Hepatocytes/virology , Humans , Luciferases/pharmacology , Nanostructures , SARS-CoV-2/genetics , Vero Cells , Virus Replication/drug effects
4.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 48(1): 24-33, 2023 Jan 28.
Article in English, Zh | MEDLINE | ID: mdl-36935174

ABSTRACT

OBJECTIVES: Hyperhomocysteinaemia (Hcy) is an independent risk factor for cardiovascular and cerebrovascular diseases. MicroRNA (miR)-18a-5p is closely related to cardiovascular diseases. This study aims to investigate the effects of miR-18a-5p on homocysteine (Hcy)-induced myocardial cells injury. METHODS: H9c2 cells were transfected with miR-18a-5p mimic/miR-18a-5p mimic negative control (NC) or combined with Hcy for intervention, and untreated cells were set as a control group. The transfection efficiency was verified by real-time RT-PCR, and cell counting kit-8 (CCK-8) assay was used to determine cell viability. Flow cytometry was used to detect apoptosis and reactive oxygen species (ROS) levels. Western blotting was performed to measure the protein levels of microtubule-associated protein 1 light chain 3 (LC3)-I, LC3-II, Beclin1, p62, Bax, Bcl-2, and Notch2. Dual luciferase reporter assay was used to detect the interaction of miR-18a-5p with Notch2. RESULTS: Compared with the control, treatment with Hcy or transfection with miR-18a-5p mimic alone, or combined treatment with Hcy and miR-18a-5p mimic/miR-18a-5p mimic NC significantly reduced the H9c2 cell viability, promoted apoptosis and ROS production, up-regulated the expressions of Bax and Beclin, down-regulated the expressions of Bcl-2, p62, and Notch2, and increased the ratio of LC3-II/LC3-I (all P<0.05). Compared with the combined intervention of miR-18a-5p mimic NC and Hcy group, the above indexes were more significantly changed in the combined intervention of miR-18a-5p mimic and Hcy group, and the difference between the 2 groups was statistically significant (all P<0.05). There is a targeted binding between Notch2 and miR-18a-5p. CONCLUSIONS: MiR-18a-5p could induce autophagy and apoptosis via increasing ROS production in cardiomyocytes, and aggravate Hcy-induced myocardial injury. Notch2 is a target of miR-18a-5p.


Subject(s)
Autophagy , MicroRNAs , Myocytes, Cardiac , Apoptosis/genetics , Autophagy/genetics , bcl-2-Associated X Protein , MicroRNAs/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Reactive Oxygen Species , Rats , Animals , Myocytes, Cardiac/drug effects , Homocysteine/adverse effects , Hyperhomocysteinemia
5.
J Clin Lab Anal ; 36(9): e24648, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36082464

ABSTRACT

OBJECTIVE: The objective of the study was to investigate the expression of LAMTOR3 in kidney renal clear cell carcinoma (KIRC) and its clinical significance. METHODS: The expression of LAMTOR3 in KIRC and its relationship with clinical features were analyzed using the UALCAN online database. The results were verified using KIRC gene chip data and clinical specimens. The prognosis of KIRC patients was analyzed with the GEPIA2 database. GO, KEGG, and GSEA analyses were conducted to analyze the possible molecular mechanism of LAMTOR3 in KIRC. Immunohistochemical (IHC) and hematoxylin and eosin (H&E) staining were used for histopathological detection. RESULTS: UALCAN database analysis showed that LAMTOR3 expression in KIRC was significantly lower than in normal kidney tissues and correlated with the clinical stage, pathological grade, and tumor genotype (p < .05). GSE53757 dataset analysis consistently showed that the expression of LAMTOR3 in KIRC was significantly lower than in normal kidney tissues (p < .01). GEPIA2 database analysis indicated that patients with low LAMTOR3 expression had poor overall and disease-free survival rates. GSEA analysis suggested that LAMTOR3 positively regulated the citrate cycle and drug metabolism cytochrome P450 and negatively regulated folate biosynthesis and olfactory transduction. The expression of LAMTOR3 in KIRC was also significantly correlated with immune cell infiltration. Finally, IHC showed that LAMTOR3 expression in the KIRC tissues was lower than in the adjacent normal tissues. CONCLUSION: LAMTOR3 expression is significantly lower in KIRC. LAMTOR3 may be a potential marker for KIRC diagnosis and therapy.


Subject(s)
Adaptor Proteins, Signal Transducing , Carcinoma, Renal Cell , Kidney Neoplasms , Adaptor Proteins, Signal Transducing/genetics , Carcinoma, Renal Cell/pathology , Humans , Kidney , Kidney Neoplasms/pathology , Prognosis
6.
Hum Mol Genet ; 28(4): 628-638, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30351356

ABSTRACT

Mutations in the collagen genes COL4A1 and COL4A2 cause Mendelian eye, kidney and cerebrovascular disease including intracerebral haemorrhage (ICH), and common collagen IV variants are a risk factor for sporadic ICH. COL4A1 and COL4A2 mutations cause endoplasmic reticulum (ER) stress and basement membrane (BM) defects, and recent data suggest an association of ER stress with ICH due to a COL4A2 mutation. However, the potential of ER stress as a therapeutic target for the multi-systemic COL4A1 pathologies remains unclear. We performed a preventative oral treatment of Col4a1 mutant mice with the chemical chaperone phenyl butyric acid (PBA), which reduced adult ICH. Importantly, treatment of adult mice with the established disease also reduced ICH. However, PBA treatment did not alter eye and kidney defects, establishing tissue-specific outcomes of targeting Col4a1-derived ER stress, and therefore this treatment may not be applicable for patients with eye and renal disease. While PBA treatment reduced ER stress and increased collagen IV incorporation into BMs, the persistence of defects in BM structure and reduced ability of the BM to withstand mechanical stress indicate that PBA may be counter-indicative for pathologies caused by matrix defects. These data establish that treatment for COL4A1 disease requires a multipronged treatment approach that restores both ER homeostasis and matrix defects. Alleviating ER stress is a valid therapeutic target for preventing and treating established adult ICH, but collagen IV patients will require stratification based on their clinical presentation and mechanism of their mutations.


Subject(s)
Cerebral Hemorrhage/drug therapy , Collagen Type IV/genetics , Molecular Targeted Therapy , Animals , Basement Membrane/drug effects , Basement Membrane/pathology , Cerebral Hemorrhage/genetics , Cerebral Hemorrhage/pathology , Disease Models, Animal , Endoplasmic Reticulum Stress/drug effects , Humans , Mice , Mutation , Phenylbutyrates/administration & dosage
7.
Proc Natl Acad Sci U S A ; 115(40): E9288-E9297, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30237286

ABSTRACT

The formation of uniaxial fibrous tissues with defined viscoelastic properties implies the existence of an orchestrated mechanical interaction between the cytoskeleton and the extracellular matrix. This study addresses the nature of this interaction. The hypothesis is that this mechanical interplay underpins the mechanical development of the tissue. In embryonic tendon tissue, an early event in the development of a mechanically robust tissue is the interaction of the pointed tips of extracellular collagen fibrils with the fibroblast plasma membrane to form stable interface structures (fibripositors). Here, we used a fibroblast-generated tissue that is structurally and mechanically matched to embryonic tendon to demonstrate homeostasis of cell-derived and external strain-derived tension over repeated cycles of strain and relaxation. A cell-derived oscillatory tension component is evident in this matrix construct. This oscillatory tension involves synchronization of individual cell forces across the construct and is induced in each strain cycle by transient relaxation and transient tensioning of the tissue. The cell-derived tension along with the oscillatory component is absent in the presence of blebbistatin, which disrupts actinomyosin force generation of the cell. The time period of this oscillation (60-90 s) is well-defined in each tissue sample and matches a primary viscoelastic relaxation time. We hypothesize that this mechanical oscillation of fibroblasts with plasma membrane anchored collagen fibrils is a key factor in mechanical sensing and feedback regulation in the formation of tensile tissues.


Subject(s)
Cell Membrane/metabolism , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Stress, Mechanical , Tensile Strength , Humans
8.
PLoS Genet ; 13(10): e1007068, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29084269

ABSTRACT

The coronary vasculature is an essential vessel network providing the blood supply to the heart. Disruptions in coronary blood flow contribute to cardiac disease, a major cause of premature death worldwide. The generation of treatments for cardiovascular disease will be aided by a deeper understanding of the developmental processes that underpin coronary vessel formation. From an ENU mutagenesis screen, we have isolated a mouse mutant displaying embryonic hydrocephalus and cardiac defects (EHC). Positional cloning and candidate gene analysis revealed that the EHC phenotype results from a point mutation in a splice donor site of the Myh10 gene, which encodes NMHC IIB. Complementation testing confirmed that the Myh10 mutation causes the EHC phenotype. Characterisation of the EHC cardiac defects revealed abnormalities in myocardial development, consistent with observations from previously generated NMHC IIB null mouse lines. Analysis of the EHC mutant hearts also identified defects in the formation of the coronary vasculature. We attribute the coronary vessel abnormalities to defective epicardial cell function, as the EHC epicardium displays an abnormal cell morphology, reduced capacity to undergo epithelial-mesenchymal transition (EMT), and impaired migration of epicardial-derived cells (EPDCs) into the myocardium. Our studies on the EHC mutant demonstrate a requirement for NMHC IIB in epicardial function and coronary vessel formation, highlighting the importance of this protein in cardiac development and ultimately, embryonic survival.


Subject(s)
Coronary Vessels/growth & development , Embryonic Development/genetics , Myosin Heavy Chains/genetics , Nonmuscle Myosin Type IIB/genetics , Pericardium/growth & development , Animals , Cell Differentiation/genetics , Coronary Vessels/metabolism , Embryo, Mammalian , Epithelial-Mesenchymal Transition/genetics , Humans , Hydrocephalus/genetics , Hydrocephalus/metabolism , Hydrocephalus/pathology , Mice , Mice, Knockout , Mutation , Myocardium/metabolism , Pericardium/metabolism
9.
Dev Biol ; 441(1): 4-11, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29883658

ABSTRACT

Fibrillar collagen is a major component of many tissues but has been difficult to image in vivo using transgenic approaches because of problems associated with establishing cells and organisms that generate GFP-fusion collagens that can polymerise into functional fibrils. Here we have developed and characterised GFP and mCherry collagen-I fusion zebrafish lines with basal epidermal-specific expression. We use these lines to reveal the dynamic nature of collagen-I fibril deposition beneath the developing embryonic epidermis, as well as the repair of this collagen meshwork following wounding. Transmission electron microscope studies show that these transgenic lines faithfully reproduce the collagen ultrastructure present in wild type larval skin. During skin development we show that collagen I is deposited by basal epidermal cells initially in fine filaments that are largely randomly orientated but are subsequently aligned into a cross-hatch, orthogonal sub-epithelial network by embryonic day 4. Following skin wounding, we see that sub-epidermal collagen is re-established in the denuded domain, initially as randomly orientated wisps that subsequently become bonded to the undamaged collagen and aligned in a way that recapitulates developmental deposition of sub-epidermal collagen. Crossing our GFP-collagen line against one with tdTomato marking basal epidermal cell membranes reveals how much more rapidly wound re-epithelialisation occurs compared to the re-deposition of collagen beneath the healed epidermis. By use of other tissue specific drivers it will be possible to establish zebrafish lines to enable live imaging of collagen deposition and its remodelling in various other organs in health and disease.


Subject(s)
Animals, Genetically Modified , Collagen Type I , Green Fluorescent Proteins , Optical Imaging/methods , Recombinant Fusion Proteins , Skin , Zebrafish , Animals , Animals, Genetically Modified/embryology , Animals, Genetically Modified/genetics , Collagen Type I/biosynthesis , Collagen Type I/genetics , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Skin/cytology , Skin/embryology , Zebrafish/embryology , Zebrafish/genetics
10.
Development ; 143(21): 3933-3943, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27621060

ABSTRACT

Recently, blood vessels have been implicated in the morphogenesis of various organs. The vasculature is also known to be essential for endochondral bone development, yet the underlying mechanism has remained elusive. We show that a unique composition of blood vessels facilitates the role of the endothelium in bone mineralization and morphogenesis. Immunostaining and electron microscopy showed that the endothelium in developing bones lacks basement membrane, which normally isolates the blood vessel from its surroundings. Further analysis revealed the presence of collagen type I on the endothelial wall of these vessels. Because collagen type I is the main component of the osteoid, we hypothesized that the bone vasculature guides the formation of the collagenous template and consequently of the mature bone. Indeed, some of the bone vessels were found to undergo mineralization. Moreover, the vascular pattern at each embryonic stage prefigured the mineral distribution pattern observed one day later. Finally, perturbation of vascular patterning by overexpressing Vegf in osteoblasts resulted in abnormal bone morphology, supporting a role for blood vessels in bone morphogenesis. These data reveal the unique composition of the endothelium in developing bones and indicate that vascular patterning plays a role in determining bone shape by forming a template for deposition of bone matrix.


Subject(s)
Blood Vessels/embryology , Bone Development/physiology , Collagen Type I/metabolism , Endothelium/metabolism , Morphogenesis/physiology , Animals , Blood Vessels/physiology , Body Patterning/physiology , Bone Matrix/embryology , Bone Matrix/metabolism , Bone and Bones/embryology , Bone and Bones/metabolism , Calcification, Physiologic/physiology , Embryo, Mammalian , Endothelium/blood supply , Female , Mice , Mice, Transgenic , Osteoblasts/physiology , Pregnancy
11.
Pak J Pharm Sci ; 32(5(Special)): 2433-2436, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31894030

ABSTRACT

This study was designed to compare the efficacy of two different racemic antihypertensive drugs on elderly patients with hypertension and their effects on vascular endothelial function and atherosclerosis. A total of 84 elderly hypertensive patients were randomly divided into control and treatment group with 42 patients in each group. The control group was treated with 2.5mg levamlodipine while the treatment group was given 5mg amlodipine. Total effective rate of the treatment group was 90.5%, higher than the control group, that was 71.4% (P<0.05). The time for recovery of related indicators like blood pressure, the total duration of medication were significantly (P<0.05) shorter in the treatment group. Only 1 case of adverse drug reaction was found in the treatment group while 6 cases in control group. Compared to the control group, the treatment group had massive improvement in fingertip pulse volume, flow-mediated dilation of the brachial arteries and endothelin-1 level, carotid intima-media thickness, plaque length & thickness, and blood pressure after the administration. The rate of satisfaction with the in treatment group was 95.3%, higher than that the control group, which was 78.6%. The study concluded that in elderly patients with hypertension, the treatment with 5mg amlodipine enhanced curative effect, fully improved endothelial function & arteriosclerosis and reduced adverse reactions thereby shortening treatment time.


Subject(s)
Amlodipine/therapeutic use , Atherosclerosis/drug therapy , Endothelium, Vascular/drug effects , Hypertension/drug therapy , Niacin/analogs & derivatives , Aged , Aged, 80 and over , Antihypertensive Agents/therapeutic use , Female , Humans , Male , Middle Aged , Niacin/therapeutic use , Vasodilator Agents/therapeutic use
12.
Hum Mol Genet ; 23(2): 283-92, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24001601

ABSTRACT

Haemorrhagic stroke accounts for ∼20% of stroke cases and porencephaly is a clinical consequence of perinatal cerebral haemorrhaging. Here, we report the identification of a novel dominant G702D mutation in the collagen domain of COL4A2 (collagen IV alpha chain 2) in a family displaying porencephaly with reduced penetrance. COL4A2 is the obligatory protein partner of COL4A1 but in contrast to most COL4A1 mutations, the COL4A2 mutation does not lead to eye or kidney disease. Analysis of dermal biopsies from a patient and his unaffected father, who also carries the mutation, revealed that both display basement membrane (BM) defects. Intriguingly, defective collagen IV incorporation into the dermal BM was observed in the patient only and was associated with endoplasmic reticulum (ER) retention of COL4A2 in primary dermal fibroblasts. This intracellular accumulation led to ER stress, unfolded protein response activation, reduced cell proliferation and increased apoptosis. Interestingly, the absence of ER retention of COL4A2 and ER stress in cells from the unaffected father indicate that accumulation and/or clearance of mutant COL4A2 from the ER may be a critical modifier for disease development. Our analysis also revealed that mutant collagen IV is degraded via the proteasome. Importantly, treatment of patient cells with a chemical chaperone decreased intracellular COL4A2 levels, ER stress and apoptosis, demonstrating that reducing intracellular collagen accumulation can ameliorate the cellular phenotype of COL4A2 mutations. Importantly, these data highlight that manipulation of chaperone levels, intracellular collagen accumulation and ER stress are potential therapeutic options for collagen IV diseases including haemorrhagic stroke.


Subject(s)
Basement Membrane/physiopathology , Brain Diseases/drug therapy , Collagen Type IV/deficiency , Hemiplegia/drug therapy , Intracranial Hemorrhages/drug therapy , Phenylbutyrates/pharmacology , Stroke/drug therapy , Apoptosis/drug effects , Basement Membrane/drug effects , Brain Diseases/genetics , Brain Diseases/pathology , Cell Proliferation/drug effects , Cells, Cultured , Collagen Type IV/genetics , Collagen Type IV/metabolism , Endoplasmic Reticulum Stress/drug effects , Female , Hemiplegia/genetics , Hemiplegia/pathology , Humans , Intracranial Hemorrhages/complications , Intracranial Hemorrhages/genetics , Male , Mutation , Phenotype , Porencephaly , Proteasome Endopeptidase Complex/metabolism , Stroke/complications , Stroke/genetics
13.
Proc Natl Acad Sci U S A ; 110(49): E4743-52, 2013 Dec 03.
Article in English | MEDLINE | ID: mdl-24248360

ABSTRACT

Collagen fibrils can exceed thousands of microns in length and are therefore the longest, largest, and most size-pleomorphic protein polymers in vertebrates; thus, knowing how cells transport collagen fibrils is essential for a more complete understanding of protein transport and its role in tissue morphogenesis. Here, we identified newly formed collagen fibrils being transported at the surface of embryonic tendon cells in vivo by using serial block face-scanning electron microscopy of the cell-matrix interface. Newly formed fibrils ranged in length from ~1 to ~30 µm. The shortest (1-10 µm) occurred in intracellular fibricarriers; the longest (~30 µm) occurred in plasma membrane fibripositors. Fibrils and fibripositors were reduced in numbers when collagen secretion was blocked. ImmunoEM showed the absence of lysosomal-associated membrane protein 2 on fibricarriers and fibripositors and there was no effect of leupeptin on fibricarrier or fibripositor number and size, suggesting that fibricarriers and fibripositors are not part of a fibril degradation pathway. Blebbistatin decreased fibricarrier number and increased fibripositor length; thus, nonmuscle myosin II (NMII) powers the transport of these compartments. Inhibition of dynamin-dependent endocytosis with dynasore blocked fibricarrier formation and caused accumulation of fibrils in fibripositors. Data from fluid-phase HRP electron tomography showed that fibricarriers could originate at the plasma membrane. We propose that NMII-powered transport of newly formed collagen fibrils at the plasma membrane is fundamental to the development of collagen fibril-rich tissues. A NMII-dependent cell-force model is presented as the basis for the creation and dynamics of fibripositor structures.


Subject(s)
Cell Membrane/metabolism , Collagen/metabolism , Myosin Type II/metabolism , Actomyosin/metabolism , Amino Acids, Dicarboxylic , Animals , Biological Transport , Chick Embryo , Collagen/biosynthesis , Collagen/physiology , Collagen/ultrastructure , Extracellular Matrix/metabolism , Microscopy, Electron, Scanning , Microscopy, Immunoelectron , Prolyl-Hydroxylase Inhibitors/pharmacology
14.
J Anat ; 224(5): 548-55, 2014 May.
Article in English | MEDLINE | ID: mdl-24571576

ABSTRACT

Achilles tendinopathies display focal tissue thickening with pain and ultrasonography changes. Whilst complete rupture might be expected to induce changes in tissue organization and protein composition, little is known about the consequences of non-rupture-associated tendinopathies, especially with regards to changes in the content of collagen type I and III (the major collagens in tendon), and changes in tendon fibroblast (tenocyte) shape and organization of the extracellular matrix (ECM). To gain new insights, we took biopsies from the tendinopathic region and flanking healthy region of Achilles tendons of six individuals with clinically diagnosed tendinopathy who had no evidence of cholesterol, uric acid and amyloid accumulation. Biochemical analyses of collagen III/I ratio were performed on all six individuals, and electron microscope analysis using transmission electron microscopy and serial block face-scanning electron microscopy were made on two individuals. In the tendinopathic regions, compared with the flanking healthy tissue, we observed: (i) an increase in the ratio of collagen III : I proteins; (ii) buckling of the collagen fascicles in the ECM; (iii) buckling of tenocytes and their nuclei; and (iv) an increase in the ratio of small-diameter : large-diameter collagen fibrils. In summary, load-induced non-rupture tendinopathy in humans is associated with localized biochemical changes, a shift from large- to small-diameter fibrils, buckling of the tendon ECM, and buckling of the cells and their nuclei.


Subject(s)
Achilles Tendon/ultrastructure , Collagen Type III/ultrastructure , Tendinopathy/pathology , Achilles Tendon/cytology , Adult , Extracellular Matrix/pathology , Humans , Imaging, Three-Dimensional , Microscopy, Electron , Middle Aged , Stress, Mechanical
15.
Matrix Biol Plus ; 21: 100142, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38328801

ABSTRACT

Tendons maintain mechanical function throughout postnatal development whilst undergoing significant microstructural changes. We present a study of postnatal tendon growth and characterise the major changes in collagen fibril architecture in mouse tail tendon from birth to eight weeks by analysing the geometries of cross-sectional transmission electron microscopy images. This study finds that a bimodal distribution of fibril diameters emerges from a unimodal distribution of narrow fibrils as early as the eighth day postnatal, and three distinct fibril populations are visible at around 14 days. The tendons in this study do not show evidence of precise hexagonal packing, even at birth, and the spaces between the fibrils remain constant throughout development. The fibril number in the tissue stabilises around day 28, and the fibril area fraction stabilises around day 26. This study gives coarse-grained insight into the transition periods in early tendon development.

17.
bioRxiv ; 2024 May 10.
Article in English | MEDLINE | ID: mdl-38766096

ABSTRACT

Collagen fibrils are the primary supporting scaffold of vertebrate tissues but how they are assembled is unclear. Here, using CRISPR-tagging of type I collagen and SILAC labelling, we elucidate the cellular mechanism for the spatiotemporal assembly of collagen fibrils, in cultured fibroblasts. Our findings reveal multifaceted trafficking of collagen, including constitutive secretion, intracellular pooling, and plasma membrane-directed fibrillogenesis. Notably, we differentiate the processes of collagen secretion and fibril assembly and identify the crucial involvement of endocytosis in regulating fibril formation. By employing Col1a1 knockout fibroblasts we demonstrate the incorporation of exogenous collagen into nucleation sites at the plasma membrane through these recycling mechanisms. Our study sheds light on the assembly process and its regulation in health and disease. Mass spectrometry data are available via ProteomeXchange with identifier PXD036794.

18.
Biochem J ; 441(2): 707-17, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-21967573

ABSTRACT

Proteolytic cleavage of procollagen I to collagen I is essential for the formation of collagen fibrils in the extracellular matrix of vertebrate tissues. Procollagen is cleaved by the procollagen N- and C-proteinases, which remove the respective N- and C-propeptides from procollagen. Procollagen processing is initiated within the secretory pathway in tendon fibroblasts, which are adept in assembling an ordered extracellular matrix of collagen fibrils in vivo. It was thought that intracellular processing was restricted to the TGN (trans-Golgi network). In the present study, brefeldin A treatment of tendon explant cultures showed that N-proteinase activity is present in the resulting fused ER (endoplasmic reticulum)-Golgi compartment, but that C-proteinase activity is restricted to the TGN in embryonic chick tendon fibroblasts. In late embryonic and postnatal rat tail and postnatal mouse tail tendon, C-proteinase activity was detected in TGN and pre-TGN compartments. Preventing activation of the procollagen N- and C-proteinases with the furin inhibitor Dec-RVKR-CMK (decanoyl-Arg-Val-Lys-Arg-chloromethylketone) indicated that only a fraction of intracellular procollagen cleavage was mediated by newly activated proteinases. In conclusion, the N-propeptides are removed earlier in the secretory pathway than the C-propeptides. The removal of the C-propeptides in post-Golgi compartments most probably indicates preparation of collagen molecules for fibril formation at the cell-matrix interface.


Subject(s)
Bone Morphogenetic Protein 1/metabolism , Collagen Type I/metabolism , Collagen/biosynthesis , Procollagen N-Endopeptidase/metabolism , Secretory Pathway/physiology , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Brefeldin A , Chick Embryo , Enzyme Activation/drug effects , Fibroblasts/enzymology , Mice , Rats , Tendons/drug effects , Tendons/embryology , trans-Golgi Network/enzymology
19.
Matrix Biol ; 124: 8-22, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37913834

ABSTRACT

The circadian clock in tendon regulates the daily rhythmic synthesis of collagen-I and the appearance and disappearance of small-diameter collagen fibrils in the extracellular matrix. How the fibrils are assembled and removed is not fully understood. Here, we first showed that the collagenase, membrane type I-matrix metalloproteinase (MT1-MMP, encoded by Mmp14), is regulated by the circadian clock in postnatal mouse tendon. Next, we generated tamoxifen-induced Col1a2-Cre-ERT2::Mmp14 KO mice (Mmp14 conditional knockout (CKO)). The CKO mice developed hind limb dorsiflexion and thickened tendons, which accumulated narrow-diameter collagen fibrils causing ultrastructural disorganization. Mass spectrometry of control tendons identified 1195 proteins of which 212 showed time-dependent abundance. In Mmp14 CKO mice 19 proteins had reversed temporal abundance and 176 proteins lost time dependency. Among these, the collagen crosslinking enzymes lysyl oxidase-like 1 (LOXL1) and lysyl hydroxylase 1 (LH1; encoded by Plod2) were elevated and had lost time-dependent regulation. High-pressure chromatography confirmed elevated levels of hydroxylysine aldehyde (pyridinoline) crosslinking of collagen in CKO tendons. As a result, collagen-I was refractory to extraction. We also showed that CRISPR-Cas9 deletion of Mmp14 from cultured fibroblasts resulted in loss of circadian clock rhythmicity of period 2 (PER2), and recombinant MT1-MMP was highly effective at cleaving soluble collagen-I but less effective at cleaving collagen pre-assembled into fibrils. In conclusion, our study shows that circadian clock-regulated Mmp14 controls the rhythmic synthesis of small diameter collagen fibrils, regulates collagen crosslinking, and its absence disrupts the circadian clock and matrisome in tendon fibroblasts.


Subject(s)
Collagen , Matrix Metalloproteinase 14 , Animals , Mice , Circadian Rhythm , Collagen/metabolism , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Homeostasis , Matrix Metalloproteinase 14/genetics , Matrix Metalloproteinase 14/metabolism
20.
Dev Dyn ; 240(11): 2520-8, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22012594

ABSTRACT

A distinctive feature of embryonic tendon development is the steady increase in collagen fibril diameter and associated improvement of tissue mechanical properties. A potential mechanical stimulus for these changes is slow stretching of the tendon during limb growth. Testing this hypothesis in vivo is complicated by the presence of other developmental processes including muscle development and innervation. Here we used a cell culture tendon-like construct to determine if slow stretch can explain the increases in fibril diameter and mechanical properties that are observed in vivo. Non-stretched constructs had an ultrastructural appearance and mechanical properties similar to those of early embryonic tendon. However, slowly stretching during 4 days in culture increased collagen fibril diameter, fibril packing volume, and mechanical stiffness, and thereby mimicked embryonic development. 3D EM showed cells with improved longitudinal alignment and elongated nuclei, which raises the hypothesis that nuclear deformation could be a novel mechanism during tendon development.


Subject(s)
Biomimetics/methods , Embryonic Development/physiology , Stress, Mechanical , Tendons/physiology , Tissue Expansion/methods , Animals , Biomechanical Phenomena/physiology , Cell Culture Techniques , Cells, Cultured , Chick Embryo , Tendons/cytology , Tensile Strength/physiology , Tissue Engineering/methods , Tissue Expansion/instrumentation , Toe Joint/cytology , Toe Joint/embryology , Toes/embryology
SELECTION OF CITATIONS
SEARCH DETAIL