Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 117
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Biomed Pharmacother ; 145: 112461, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34839253

ABSTRACT

Skin aging is accompanied by an increase in the number of senescent cells, resulting in various pathological outcomes. These include inflammation, impaired barrier function, and susceptibility to skin disorders such as cancer. Kaempferia parviflora (Thai black ginger), a medicinal plant native to Thailand, has been shown to counteract inflammation, cancer, and senescence. This study demonstrates that polymethoxyflavones (5,7-dimethoxyflavone, 5,7,4'-trimethoxyflavone, and 3,5,7,3',4'-pentamethoxyflavone) purified from K. parviflora rhizomes suppressed cellular senescence, reactive oxygen species, and the senescence-associated secretory phenotype in primary human dermal fibroblasts. In addition, they increased tropocollagen synthesis and alleviated free radical-induced cellular and mitochondrial damage. Moreover, the compounds mitigated chronological aging in a human ex vivo skin model by attenuating senescence and restoring expression of essential components of the extracellular matrix, including collagen type I, fibrillin-1, and hyaluronic acid. Finally, we report that polymethoxyflavones enhanced epidermal thickness and epidermal-dermal stability, while blocking age-related inflammation in skin explants. Our findings support the use of polymethoxyflavones from K. parviflora as natural anti-aging agents, highlighting their potential as active ingredients in cosmeceutical and nutraceutical products.


Subject(s)
Collagen Type I/metabolism , Extracellular Matrix , Flavonoids/pharmacology , Hyaluronic Acid/metabolism , Skin Aging , Skin , Zingiberaceae , Cell Line , Extracellular Matrix/drug effects , Extracellular Matrix/physiology , Fibrillin-1/metabolism , Fibroblasts/metabolism , Flavones/pharmacology , Geroscience , Humans , Rhizome , Skin/drug effects , Skin/metabolism , Skin Aging/drug effects , Skin Aging/physiology , Thailand
2.
Int J Mol Sci ; 22(2)2021 Jan 12.
Article in English | MEDLINE | ID: mdl-33445782

ABSTRACT

Intervertebral disc (IVD) degeneration is a major risk factor of low back pain. It is defined by a progressive loss of the IVD structure and functionality, leading to severe impairments with restricted treatment options due to the highly demanding mechanical exposure of the IVD. Degenerative changes in the IVD usually increase with age but at an accelerated rate in some individuals. To understand the initiation and progression of this disease, it is crucial to identify key top-down and bottom-up regulations' processes, across the cell, tissue, and organ levels, in health and disease. Owing to unremitting investigation of experimental research, the comprehension of detailed cell signaling pathways and their effect on matrix turnover significantly rose. Likewise, in silico research substantially contributed to a holistic understanding of spatiotemporal effects and complex, multifactorial interactions within the IVD. Together with important achievements in the research of biomaterials, manifold promising approaches for regenerative treatment options were presented over the last years. This review provides an integrative analysis of the current knowledge about (1) the multiscale function and regulation of the IVD in health and disease, (2) the possible regenerative strategies, and (3) the in silico models that shall eventually support the development of advanced therapies.


Subject(s)
Intervertebral Disc Degeneration/physiopathology , Intervertebral Disc/physiopathology , Animals , Computer Simulation , Extracellular Matrix/physiology , Humans , Signal Transduction/physiology , Tissue Engineering/methods
3.
Am J Physiol Cell Physiol ; 319(4): C611-C623, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32667866

ABSTRACT

The primary biomechanical driver of pathological glaucomatous cupping remains unknown. Finite element modeling indicates that stress and strain play key roles. In this article, primarily a review, we utilize known biomechanical data and currently unpublished results from our lab to propose a three-stage, tissue stiffness-based model to explain glaucomatous cupping occurring at variable levels of translaminar pressure (TLP). In stage 1, a short-term increase in TLP gradient induces a transient increase in lamina cribrosa (LC) strain. Beyond a critical level of strain, the tissue stiffness rises steeply provoking cellular responses via integrin-mediated mechanotransduction. This early mechanoprotective cellular contraction reduces strain, which reduces tissue stiffness by return of the posteriorly deflected LC to baseline. In stage 2 a prolonged period of TLP increase elicits extracellular matrix (ECM) production leading to fibrosis, increasing baseline tissue stiffness and strain and diminishing the contractile ability/ability to return to the baseline LC position. This is supported by our three-dimensional collagen contraction assays, which show significantly reduced capacity to contract in glaucoma compared with normal LC cells. Second, 15% cyclic strain in LC cells over 24 h elicits a typical increase in ECM profibrotic genes in normal LC cells but a highly blunted response in glaucoma LC cells. Stage 3 is characterized by persistent fibrosis causing further stiffening and inducing a feed-forward ECM production cycle. Repeated cycles of increased strain and stiffness with profibrotic ECM deposition prevent optic nerve head (ONH) recoil from the new deflected position. This incremental maladaptive modeling leads to pathological ONH cupping.


Subject(s)
Fibrosis/physiopathology , Glaucoma/physiopathology , Optic Disk/physiology , Vascular Stiffness/physiology , Biomechanical Phenomena , Extracellular Matrix/metabolism , Extracellular Matrix/physiology , Fibrosis/therapy , Finite Element Analysis , Glaucoma/therapy , Humans , Models, Theoretical , Optic Disk/pathology
5.
J Bodyw Mov Ther ; 24(1): 138-146, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31987534

ABSTRACT

This article presents an overview of research conducted by Dr Jean-Claude Guimberteau into the architecture and spatial organization of living matter and the relationship between the cells and the extracellular matrix. His research is discussed in the context of previous and current research into fascial anatomy. Andrew Taylor Still, the founder of Osteopathy, did not have access to modern research and yet his observations are proving to be surprisingly accurate in the light of recent findings. This article sets out to highlight the relevance of his insights from a purely anatomical perspective, and to draw parallels with a new way of thinking about the architecture of the living human body that is slowly emerging. Dr Guimberteau's research shows that a force applied to the surface of the skin is transmitted deep into living tissue via a continuous bodywide multifibrillar network. It also confirms the concept of the body as a dynamic functional unit, as proposed by A.T. Still. Still also proposed that structure and function are interrelated at all levels within the living human body. There is a growing body of research to support this. Intratissular endoscopy has highlighted the importance of the quality of the mobility and adaptability of the network of collagen and elastin fibers that structures the ECM in healthy living tissue. Factors such as abnormal stiffness of collagen fibers in the ECM are thought to have adverse effects on local tissue health.


Subject(s)
Collagen/physiology , Endoscopy/methods , Extracellular Matrix/physiology , Fascia/physiology , Osteopathic Medicine , Cellular Microenvironment/physiology , Fibrillar Collagens/physiology , Human Body , Humans
6.
Brain Struct Funct ; 225(1): 45-56, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31748912

ABSTRACT

During hibernation, mammals like the 13-lined ground squirrel cycle between physiological extremes. Most of the hibernation season is spent in bouts of torpor, where body temperature, heart rate, and cerebral blood flow are all very low. However, the ground squirrels periodically enter into interbout arousals (IBAs), where physiological parameters return to non-hibernating levels. During torpor, neurons in many brain regions shrink and become electrically quiescent, but reconnect and regain activity during IBA. Previous work showed evidence of extracellular matrix (ECM) changes occurring in the hypothalamus during hibernation that could be associated with this plasticity. Here, we examined expression of a specialized ECM structure, the perineuronal net (PNN), in the forebrain of ground squirrels in torpor, IBA, and summer (non-hibernating). PNNs are known to restrict plasticity, and could be important for retaining essential connections in the brain during hibernation. We found PNNs in three regions of the hypothalamus: ventrolateral hypothalamus, paraventricular nucleus (PVN), and anterior hypothalamic area. We also found PNNs throughout the cerebral cortex, amygdala, and lateral septum. The total area covered by PNNs within the PVN was significantly higher during IBA compared to non-hibernating and torpor (P < 0.01). Additionally, the amount of PNN coverage area per Nissl-stained neuron in the PVN was significantly higher in hibernation compared to non-hibernating (P < 0.05). No other significant differences were found across seasons. The PVN is involved in food intake and homeostasis, and PNNs found here could be essential for retaining vital life functions during hibernation.


Subject(s)
Brain/cytology , Brain/physiology , Extracellular Matrix/physiology , Hibernation/physiology , Neurons/cytology , Neurons/physiology , Sciuridae/anatomy & histology , Sciuridae/physiology , Amygdala/cytology , Amygdala/physiology , Animals , Cerebral Cortex/cytology , Cerebral Cortex/physiology , Female , Hypothalamus/cytology , Hypothalamus/physiology , Male , Neuronal Plasticity
7.
Wounds ; 31(10): 262-268, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31730505

ABSTRACT

OBJECTIVE: This retrospective study evaluates the efficacy of acellular fish skin graft for the treatment of full-thickness diabetic foot ulcers (DFUs). The primary objective is to calculate the total wound surface area (cm2) healed over a 16-week period. The secondary objective is to provide a systematic review on acellular fish skin grafts. MATERIALS AND METHODS: There were 51 patients with a total of 58 DFUs treated with an acellular fish skin graft by the principal investigator. The initial wound surface area at first application was compared with the final wound surface area to conclude the percentage of total wound healed over a 16-week treatment period. RESULTS: At 16 weeks, there was a mean reduction of wound surface area by 87.57% and 35 wounds (60.34%) fully healed. The systematic literature review included 10 fish graft articles, 3 of which specifically evaluated lower extremity ulcers. The reviewed studies supported improved wound healing with fish graft application, with benefits noted in dentistry, neurology, and wound care. CONCLUSIONS: This retrospective study further supports previous evidence that acellular fish skin graft promotes wound healing in DFUs. In particular, a rapid increase in wound healing was observed during the initial 4 weeks following graft application. This study and review of the literature indicated that fish graft encourages wound healing by enabling the wound to transition from a chronic to an acute stage of healing.


Subject(s)
Acellular Dermis , Dermatologic Surgical Procedures , Diabetic Foot/therapy , Extracellular Matrix/physiology , Fatty Acids, Omega-3/therapeutic use , Fishes , Skin , Wound Healing/physiology , Aged , Aged, 80 and over , Animals , Diabetic Foot/pathology , Female , Fish Proteins/pharmacology , Humans , Male , Middle Aged , Retrospective Studies , Treatment Outcome , Wound Healing/drug effects
8.
J Appl Oral Sci ; 27: e20180596, 2019 Sep 09.
Article in English | MEDLINE | ID: mdl-31508793

ABSTRACT

Bone development and healing processes involve a complex cascade of biological events requiring well-orchestrated synergism with bone cells, growth factors, and other trophic signaling molecules and cellular structures. Beyond health processes, MMPs play several key roles in the installation of heart and blood vessel related diseases and cancer, ranging from accelerating metastatic cells to ectopic vascular mineralization by smooth muscle cells in complementary manner. The tissue inhibitors of MMPs (TIMPs) have an important role in controlling proteolysis. Paired with the post-transcriptional efficiency of specific miRNAs, they modulate MMP performance. If druggable, these molecules are suggested to be a platform for development of "smart" medications and further clinical trials. Thus, considering the pleiotropic effect of MMPs on mammals, the purpose of this review is to update the role of those multifaceted proteases in mineralized tissues in health, such as bone, and pathophysiological disorders, such as ectopic vascular calcification and cancer.


Subject(s)
Bone Remodeling/physiology , Extracellular Matrix/physiology , Matrix Metalloproteinases/physiology , Bone Diseases/metabolism , Bone Diseases/physiopathology , Disease Progression , Humans , Matrix Metalloproteinase Inhibitors/therapeutic use , Neoplasms/metabolism , Neoplasms/physiopathology , Osteoblasts/physiology , Tissue Inhibitor of Metalloproteinases/physiology , Vascular Calcification/metabolism , Vascular Calcification/physiopathology
9.
Am J Cardiovasc Drugs ; 19(6): 541-552, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31292859

ABSTRACT

Heart failure (HF) with reduced ejection fraction (HFrEF) presents as the severest phenotype on the spectrum of HF. Although great progress has been made with respect to its treatment over the past 3 decades, morbidity and mortality remain high, posing a big burden on human health. Recent evidence suggests vitamin D has a critical role in maintaining heart health through activation of the vitamin D receptor expressed in cardiomyocytes, and vitamin D deficiency may be implicated in the pathophysiology of HFrEF through activation of the renin-angiotensin system, impaired calcium handling, exaggerated inflammation, secondary hyperparathyroidism, pro-fibrotic properties, and proatherogenic potential. Additionally, epidemiological data disclosed that vitamin D deficiency is highly prevalent in patients with HFrEF and is associated with poor clinical outcomes. However, randomized control trials of vitamin D supplementation in HF, especially in HFrEF, have shown inconsistent results. Thus, this article aims to review the epidemiology, pathophysiology, and prognostic value of vitamin D deficiency in HF, with a special focus on randomized control trials associated with vitamin D supplementation in patients with HFrEF.


Subject(s)
Heart Failure/epidemiology , Heart Failure/physiopathology , Vitamin D Deficiency/epidemiology , Vitamin D Deficiency/physiopathology , Atrial Fibrillation/epidemiology , Atrial Fibrillation/physiopathology , Calcium/metabolism , Dietary Supplements , Extracellular Matrix/physiology , Fibrosis/epidemiology , Fibrosis/physiopathology , Humans , Hyperparathyroidism, Secondary/epidemiology , Hyperparathyroidism, Secondary/physiopathology , Inflammation Mediators/metabolism , Myocytes, Cardiac/metabolism , Prognosis , Renin-Angiotensin System/physiology , Severity of Illness Index , Stroke Volume/physiology , Vitamin D/metabolism , Vitamin D/therapeutic use , Vitamin D Deficiency/drug therapy
10.
PLoS One ; 14(1): e0204197, 2019.
Article in English | MEDLINE | ID: mdl-30608923

ABSTRACT

Bone remodeling involves the coordinated actions of osteoclasts, which resorb the calcified bony matrix, and osteoblasts, which refill erosion pits created by osteoclasts to restore skeletal integrity and adapt to changes in mechanical load. Osteoblasts are derived from pluripotent mesenchymal stem cell precursors, which undergo differentiation under the influence of a host of local and environmental cues. To characterize the autocrine/paracrine signaling networks associated with osteoblast maturation and function, we performed gene network analysis using complementary "agnostic" DNA microarray and "targeted" NanoString nCounter datasets derived from murine MC3T3-E1 cells induced to undergo synchronized osteoblastic differentiation in vitro. Pairwise datasets representing changes in gene expression associated with growth arrest (day 2 to 5 in culture), differentiation (day 5 to 10 in culture), and osteoblast maturation (day 10 to 28 in culture) were analyzed using Ingenuity Systems Pathways Analysis to generate predictions about signaling pathway activity based on the temporal sequence of changes in target gene expression. Our data indicate that some pathways involved in osteoblast differentiation, e.g. Wnt/ß-catenin signaling, are most active early in the process, while others, e.g. TGFß/BMP, cytokine/JAK-STAT and TNFα/RANKL signaling, increase in activity as differentiation progresses. Collectively, these pathways contribute to the sequential expression of genes involved in the synthesis and mineralization of extracellular matrix. These results provide insight into the temporal coordination and complex interplay between signaling networks controlling gene expression during osteoblast differentiation. A more complete understanding of these processes may aid the discovery of novel methods to promote osteoblast development for the treatment of conditions characterized by low bone mineral density.


Subject(s)
Cell Differentiation/genetics , Osteoblasts/physiology , Osteogenesis/genetics , Signal Transduction/genetics , Transcriptome/physiology , 3T3 Cells , Animals , Autocrine Communication/genetics , Bone Density/physiology , Bone Remodeling/genetics , Datasets as Topic , Extracellular Matrix/physiology , Gene Expression Profiling , Gene Regulatory Networks/physiology , Mice , Oligonucleotide Array Sequence Analysis , Paracrine Communication/genetics
11.
Cartilage ; 10(2): 157-172, 2019 04.
Article in English | MEDLINE | ID: mdl-28933195

ABSTRACT

OBJECTIVE: Hyaline cartilage degenerative pathologies induce morphologic and biomechanical changes resulting in cartilage tissue damage. In pursuit of therapeutic options, electrical and mechanical stimulation have been proposed for improving tissue engineering approaches for cartilage repair. The purpose of this review was to highlight the effect of electrical stimulation and mechanical stimuli in chondrocyte behavior. DESIGN: Different information sources and the MEDLINE database were systematically revised to summarize the different contributions for the past 40 years. RESULTS: It has been shown that electric stimulation may increase cell proliferation and stimulate the synthesis of molecules associated with the extracellular matrix of the articular cartilage, such as collagen type II, aggrecan and glycosaminoglycans, while mechanical loads trigger anabolic and catabolic responses in chondrocytes. CONCLUSION: The biophysical stimuli can increase cell proliferation and stimulate molecules associated with hyaline cartilage extracellular matrix maintenance.


Subject(s)
Cartilage, Articular/cytology , Chondrocytes/physiology , Hyaline Cartilage/cytology , Osteoarthritis/physiopathology , Physical Stimulation/methods , Aggrecans/physiology , Animals , Cartilage, Articular/physiopathology , Cell Proliferation/physiology , Collagen Type II/physiology , Electric Stimulation/methods , Electric Stimulation Therapy/methods , Extracellular Matrix/physiology , Glycosaminoglycans/physiology , Humans , Hyaline Cartilage/physiopathology , Tissue Engineering/methods
12.
Biotechnol J ; 14(3): e1700763, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30052320

ABSTRACT

Lysyl oxidase (LOX)-mediated collagen crosslinking can regulate osteoblastic phenotype and enhance mechanical properties of tissues, both areas of interest in bone tissue engineering. The objective of this study is to investigate the effect of lysyl oxidase-like 2 (LOXL2) on osteogenic differentiation of mesenchymal stem cells (MSCs) cultured in perfusion bioreactors, enzymatic collagen crosslink formation in the extracellular matrix (ECM), and mechanical properties of engineered bone grafts. Exogenous LOXL2 to MSCs seeded in composite scaffolds under perfusion culture for up to 28 days is administered. Constructs treated with LOXL2 appear brown in color and possess greater DNA content and osteogenic potential measured by a twofold increase in bone sialoprotein gene expression. Collagen expression of LOXL2-treated scaffolds is lower than untreated controls. Functional outputs such as calcium deposition, osteocalcin expression, and compressive modulus are unaffected by LOXL2 supplementation. Excitingly, LOXL2-treated constructs contain 1.8- and 1.4-times more pyridinoline (PYD) crosslinks per mole of collagen and per wet weight, respectively, than untreated constructs. Despite these increases, compressive moduli of LOXL2-treated constructs are similar to untreated constructs over the 28-day culture duration. This is the first report of LOXL2 application to engineered, three-dimensional bony constructs. The results suggest a potentially new strategy for engineering osteogenic grafts with a mature ECM by modulating crosslink formation.


Subject(s)
Amino Acid Oxidoreductases/metabolism , Collagen/metabolism , Osteogenesis/physiology , Amino Acids/metabolism , Cell Culture Techniques/methods , Cell Differentiation/physiology , Cells, Cultured , Extracellular Matrix/metabolism , Extracellular Matrix/physiology , Humans , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/physiology , Tissue Engineering/methods , Tissue Scaffolds
13.
J. appl. oral sci ; J. appl. oral sci;27: e20180596, 2019. graf
Article in English | LILACS, BBO - Dentistry | ID: biblio-1019968

ABSTRACT

Abstract Bone development and healing processes involve a complex cascade of biological events requiring well-orchestrated synergism with bone cells, growth factors, and other trophic signaling molecules and cellular structures. Beyond health processes, MMPs play several key roles in the installation of heart and blood vessel related diseases and cancer, ranging from accelerating metastatic cells to ectopic vascular mineralization by smooth muscle cells in complementary manner. The tissue inhibitors of MMPs (TIMPs) have an important role in controlling proteolysis. Paired with the post-transcriptional efficiency of specific miRNAs, they modulate MMP performance. If druggable, these molecules are suggested to be a platform for development of "smart" medications and further clinical trials. Thus, considering the pleiotropic effect of MMPs on mammals, the purpose of this review is to update the role of those multifaceted proteases in mineralized tissues in health, such as bone, and pathophysiological disorders, such as ectopic vascular calcification and cancer.


Subject(s)
Humans , Bone Remodeling/physiology , Matrix Metalloproteinases/physiology , Extracellular Matrix/physiology , Osteoblasts/physiology , Bone Diseases/physiopathology , Bone Diseases/metabolism , Disease Progression , Tissue Inhibitor of Metalloproteinases/physiology , Vascular Calcification/physiopathology , Vascular Calcification/metabolism , Matrix Metalloproteinase Inhibitors/therapeutic use , Neoplasms/physiopathology , Neoplasms/metabolism
14.
JCI Insight ; 3(21)2018 11 02.
Article in English | MEDLINE | ID: mdl-30385720

ABSTRACT

In this study we evaluated the role of hyaluronan (HA) in reactive adipogenesis, a local expansion of preadipocytes that provides host defense by release of antimicrobial peptides. We observed that HA accumulated during maturation of adipocytes in vitro and was associated with increased expression of preadipocyte factor 1, zinc finger protein 423, and early B cell factor 1. Although HA is normally abundant in the extracellular matrix, a further increase in HA staining occurred in mice at sites of reactive adipogenesis following injury of colon by dextran sodium sulfate or injury of skin from infection with Staphylococcus aureus. HA also abundantly accumulated around adipocytes seen in the colons of patients with inflammatory bowel disease. This HA was necessary for adipocyte maturation because digestion of HA by administration of soluble hyaluronidase or transgenic expression of hyaluronidase 1 inhibited adipogenesis in vitro and in vivo. Furthermore, hyaluronidase also suppressed inflammation of both skin and colon and decreased antimicrobial peptide expression by developing preadipocytes. This resulted in increased bacterial transit across the epithelial barrier despite decreased tissue injury from inflammation. These observations suggest HA plays an important role in reactive adipogenesis and host defense after injury.


Subject(s)
Adipocytes/metabolism , Adipogenesis/drug effects , Colon/drug effects , Hyaluronic Acid/adverse effects , Hyaluronoglucosaminidase/metabolism , Skin/drug effects , Adjuvants, Immunologic/adverse effects , Animals , Calcium-Binding Proteins , Colon/injuries , Colon/metabolism , Colon/pathology , DNA-Binding Proteins , Extracellular Matrix/enzymology , Extracellular Matrix/physiology , Humans , Hyaluronoglucosaminidase/adverse effects , Inflammation/immunology , Inflammation/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic/genetics , Skin/injuries , Skin/metabolism , Skin/pathology , Trans-Activators , Transcription Factors
15.
J Neurochem ; 147(5): 626-646, 2018 12.
Article in English | MEDLINE | ID: mdl-30326149

ABSTRACT

Visual information is detected by the retina and transmitted into the brain by retinal ganglion cells. In rodents, the visual thalamus is a major recipient of retinal ganglion cells axons and is divided into three functionally distinct nuclei: the dorsal lateral geniculate nucleus (dLGN), ventral LGN (vLGN), and intergeniculate leaflet. Despite being densely innervated by retinal input, each nucleus in rodent visual thalamus possesses diverse molecular profiles which underpin their unique circuitry and cytoarchitecture. Here, we combined large-scale unbiased proteomic and transcriptomic analyses to elucidate the molecular expression profiles of the developing mouse dLGN and vLGN. We identified several extracellular matrix proteins as differentially expressed in these regions, particularly constituent molecules of perineuronal nets (PNNs). Remarkably, we discovered at least two types of molecularly distinct Aggrecan-rich PNN populations in vLGN, exhibiting non-overlapping spatial, temporal, and cell-type specific expression patterns. The mechanisms responsible for the formation of these two populations of PNNs also differ as the formation of Cat315+ PNNs (but not WFA+ PNNs) required input from the retina. This study is first to suggest that cell type- and molecularly specific supramolecular assemblies of extracellular matrix may play important roles in the circuitry associated with the subcortical visual system and in the processing of visual information. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/. Cover Image for this issue: doi: 10.1111/jnc.14203.


Subject(s)
Nerve Net/metabolism , Thalamus/metabolism , Vision, Ocular/physiology , Animals , Extracellular Matrix/metabolism , Extracellular Matrix/physiology , Extracellular Matrix Proteins/metabolism , Geniculate Bodies/metabolism , Immunohistochemistry , Mice , Mice, Inbred C57BL , Nerve Net/growth & development , Proteomics , Real-Time Polymerase Chain Reaction , Retina/physiology , Retinal Ganglion Cells/physiology , Thalamus/growth & development , Visual Perception/physiology
16.
Adv Exp Med Biol ; 1098: 131-150, 2018.
Article in English | MEDLINE | ID: mdl-30238369

ABSTRACT

The cardiac extracellular matrix (cECM) is comprised of proteins and polysaccharides secreted by cardiac cell types, which provide structural and biochemical support to cardiovascular tissue. The roles of cECM proteins and the associated family of cell surface receptor, integrins, have been explored in vivo via the generation of knockout experimental animal models. However, the complexity of tissues makes it difficult to isolate the effects of individual cECM proteins on a particular cell process or disease state. The desire to further dissect the role of cECM has led to the development of a variety of in vitro model systems, which are now being used not only for basic studies but also for testing drug efficacy and toxicity and for generating therapeutic scaffolds. These systems began with 2D coatings of cECM derived from tissue and have developed to include recombinant ECM proteins, ECM fragments, and ECM mimics. Most recently 3D model systems have emerged, made possible by several developing technologies including, and most notably, 3D bioprinting. This chapter will attempt to track the evolution of our understanding of the relationship between cECM and cell behavior from in vivo model to in vitro control systems. We end the chapter with a summary of how basic studies such as these have informed the use of cECM as a direct therapy.


Subject(s)
Extracellular Matrix , Myocardium/ultrastructure , Regenerative Medicine/methods , Tissue Engineering/methods , Tissue Scaffolds , Animals , Biopolymers/chemistry , Cell Growth Processes , Cell Transplantation/methods , Drug Evaluation, Preclinical/methods , Extracellular Matrix/chemistry , Extracellular Matrix/physiology , Extracellular Matrix/ultrastructure , Extracellular Matrix Proteins/deficiency , Extracellular Matrix Proteins/physiology , Extracellular Matrix Proteins/therapeutic use , Humans , In Vitro Techniques , Mice , Mice, Knockout , Myocytes, Cardiac/cytology , Printing, Three-Dimensional , Recombinant Proteins/therapeutic use
17.
Mol Ther ; 26(7): 1735-1745, 2018 07 05.
Article in English | MEDLINE | ID: mdl-29843956

ABSTRACT

Effective vascular regeneration could provide therapeutic benefit for multiple pathologies, especially in chronic peripheral artery disease (PAD) and myocardial ischemia. The hypoxia inducible factors (HIFs) mediate the cellular transcriptional response to hypoxia and regulate multiple processes that are required for angiogenesis to ultimately restore perfusion and oxygen supply. In endothelial cells, both HIF1α and HIF2α are known to contribute to this role; however, the extent and individual roles of each of these HIFα remain unclear. To characterize the individual roles of HIFα, we sequenced the transcriptional outputs of stabilized forms of HIF1α and HIF2α, where they regulated 701 and 1,454 genes, respectively. HIF1α transcription primarily regulated metabolic reprogramming, whereas HIF2α exerted a larger role in regulating angiogenic extracellular signaling, guidance cues, and extracellular matrix remodeling factors. Furthermore, HIF2α almost exclusively regulated a large and diverse subset of transcription factors and coregulators that contribute to its diverse roles in hypoxia. Further understanding of how HIFs regulate cellular processes in hypoxia and angiogenesis could offer new avenues to modulate physiological angiogenesis to enhance revascularisation in ischemic conditions and other pathologies.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Transcription, Genetic/genetics , Cell Line , Extracellular Matrix/genetics , Extracellular Matrix/physiology , Gene Expression Regulation/genetics , Human Umbilical Vein Endothelial Cells , Humans , Hypoxia/genetics , Hypoxia/physiopathology , Neovascularization, Physiologic/genetics , Neovascularization, Physiologic/physiology , Transcription Factors/genetics
18.
Osteoarthritis Cartilage ; 25(10): 1698-1707, 2017 10.
Article in English | MEDLINE | ID: mdl-28647469

ABSTRACT

OBJECTIVE: Disruptions of extracellular matrix (ECM) homeostasis are key events in the pathogenesis of osteoarthritis (OA). MicroRNA-140 (miRNA-140) is expressed specifically in cartilage and regulates ECM-degrading enzymes. Our objective in this study was to determine if intra-articular injection of miRNA-140 can attenuate OA progression in rats. DESIGN: miRNA-140 levels in human normal and OA cartilage derived chondrocytes and synovial fluid were assessed by polymerase chain reaction (PCR). After primary human chondrocytes were transfected with miRNA-140 mimic or inhibitor, PCR and western blotting were performed to quantify Collagen II, MMP-13, and ADAMTS-5 expression. An OA model was induced surgically in rats, and subsequently treated with one single intra-articular injection of miRNA-140 agomir. At 4, 8, and 12 weeks after surgery, OA progression were evaluated macroscopically, histologically, and immunohistochemically in these rats. RESULTS: miRNA-140 levels were significantly reduced in human OA cartilage derived chondrocytes and synovial fluid compared with normal chondrocytes and synovial fluid. Overexpressing miRNA-140 in primary human chondrocytes promoted Collagen II expression and inhibited MMP-13 and ADAMTS-5 expression. miRNA-140 levels in rat cartilage were significantly higher in the miRNA-140 agomir group than in the control group. Moreover, behavioural scores, chondrocyte numbers, cartilage thickness and Collagen II expression levels in cartilage were significantly higher, while pathological scores and MMP-13 and ADAMTS-5 expression levels were significantly lower in the miRNA-140 agomir group than in the control group. CONCLUSION: Intra-articular injection of miRNA-140 can alleviate OA progression by modulating ECM homeostasis in rats, and may have potential as a new therapy for OA.


Subject(s)
Arthritis, Experimental/drug therapy , Extracellular Matrix/drug effects , MicroRNAs/administration & dosage , Osteoarthritis/drug therapy , ADAMTS5 Protein/biosynthesis , ADAMTS5 Protein/genetics , Adult , Aged , Aged, 80 and over , Animals , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cells, Cultured , Chondrocytes/metabolism , Collagen Type II/biosynthesis , Collagen Type II/genetics , Disease Progression , Drug Evaluation, Preclinical/methods , Extracellular Matrix/physiology , Gene Expression Regulation/physiology , Homeostasis/drug effects , Humans , Injections, Intra-Articular , Male , Matrix Metalloproteinase 13/biosynthesis , Matrix Metalloproteinase 13/genetics , MicroRNAs/metabolism , MicroRNAs/pharmacology , MicroRNAs/therapeutic use , Middle Aged , Osteoarthritis/metabolism , Osteoarthritis/pathology , Rats, Sprague-Dawley , Synovial Fluid/metabolism , Young Adult
19.
J Long Term Eff Med Implants ; 27(2-4): 199-231, 2017.
Article in English | MEDLINE | ID: mdl-29773040

ABSTRACT

Tissue formation within the body, as part of a development or repair process, is a complex event in which cell populations self-assemble into functional units. There is intense academic, medical, and commercial interest in finding methods of replicating these events outside the body. This interest has accelerated with the demonstration of the engineering of skin and cartilage tissue in the laboratory and there is now worldwide activity in the in vitro regeneration of tissues including nerve, liver, bone, heart valves, blood vessels, bladder, and kidney. Approaches to tissue engineering center on the need to provide signals to cell populations to promote cell proliferation and differentiation. This review considers recent advances in methods of providing these signals to cells using examples of progress in the engineering of complex tissues.


Subject(s)
Tissue Engineering/methods , Animals , Cell Communication/physiology , Cell Proliferation/physiology , Electric Stimulation Therapy , Extracellular Matrix/physiology , Humans , In Vitro Techniques , Intercellular Signaling Peptides and Proteins/pharmacology , Regeneration , Tissue Engineering/trends , Tissue Scaffolds
20.
Am J Clin Dermatol ; 17(3): 201-23, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26894654

ABSTRACT

Since their earliest description, keloids and hypertrophic scars have beleaguered patients and clinicians alike. These scars can be aesthetically disfiguring, functionally debilitating, emotionally distressing, and psychologically damaging, culminating in a significant burden for patients. Our current understanding of keloid pathophysiology has grown and continues to advance while molecular biology, genetics, and technology provide ever-deepening insight into the nature of wound healing and the pathologic perturbations thereof. Greater understanding will lead to the development and application of refined therapeutic modalities. This article provides an overview of our current understanding of keloids, highlighting clinical characteristics and diagnostic criteria while providing a comprehensive summary of the many therapeutic modalities available. The proposed mechanism, application, adverse events, and reported efficacy of each modality is evaluated, and current recommendations are summarized.


Subject(s)
Cicatrix, Hypertrophic , Fibroblasts/physiology , Keloid , Wound Healing/physiology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/adverse effects , Adjuvants, Immunologic/therapeutic use , Aminoquinolines/administration & dosage , Aminoquinolines/adverse effects , Aminoquinolines/therapeutic use , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/adverse effects , Anti-Inflammatory Agents/therapeutic use , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/adverse effects , Antibiotics, Antineoplastic/therapeutic use , Antimetabolites/administration & dosage , Antimetabolites/adverse effects , Antimetabolites/therapeutic use , Bleomycin/administration & dosage , Bleomycin/adverse effects , Bleomycin/therapeutic use , Cell Proliferation , Cicatrix, Hypertrophic/etiology , Cicatrix, Hypertrophic/pathology , Cicatrix, Hypertrophic/therapy , Clinical Trials as Topic , Collagen/metabolism , Combined Modality Therapy/methods , Cryotherapy/methods , Extracellular Matrix/physiology , Fluorouracil/administration & dosage , Fluorouracil/adverse effects , Fluorouracil/therapeutic use , Glucocorticoids/administration & dosage , Glucocorticoids/adverse effects , Glucocorticoids/therapeutic use , Humans , Imiquimod , Inflammation/metabolism , Keloid/etiology , Keloid/pathology , Keloid/therapy , Laser Therapy/methods
SELECTION OF CITATIONS
SEARCH DETAIL